1,556
Views
74
CrossRef citations to date
0
Altmetric
Research Article

Effector CD8+ T cell IFN-γ production and cytotoxicity are enhanced by mild hyperthermia

, , &
Pages 9-18 | Received 01 Jul 2011, Accepted 17 Aug 2011, Published online: 11 Jan 2012

Abstract

Purpose: Clinical trials combining hyperthermia with radiation and/or chemotherapy for cancer treatment have resulted in improved overall survival and control of local recurrences. The contribution of thermally enhanced anti-immune function in these effects is of considerable interest, but not understood; studies on the fundamental effects of elevated temperature on immune effector cells are needed. The goal of this study is to investigate the potential of mild hyperthermia to impact tumour antigen-specific (Ag) effector CD8+ T cell functions.

Method: Pmel-1 Ag-specific CD8+ T cells were exposed to mild hyperthermia and tested for changes in IFN-γ production and cytotoxicity. Additionally, overall plasma membrane organisation and the phosphorylation of signalling proteins were also investigated following heat treatment.

Results: Exposing effector Pmel-1-specific CD8+ T cells to mild hyperthermia (39.5°C) resulted in significantly enhanced Ag-specific IFN-γ production and tumour target cell killing compared to that seen using lower temperatures (33° and 37°C). Further, inhibition of protein synthesis during hyperthermia did not reduce subsequent Ag-induced IFN-γ production by CD8+ T cells. Correlated with these effects, we observed a distinct clustering of GM1+ lipid microdomains at the plasma membrane and enhanced phosphorylation of LAT and PKCθ which may be related to an observed enhancement of Ag-specific effector CD8+ T cell IFN-γ gene transcription following mild hyperthermia. However, mitogen-mediated production of IFN-γ, which bypasses T cell receptor activation with antigen, was not enhanced.

Conclusions: Antigen-dependent effector T cell activity is enhanced following mild hyperthermia. These effects could potentially occur in patients being treated with thermal therapies. These data also provide support for the use of thermal therapy as an adjuvant for immunotherapies to improve CD8+ effector cell function.

Introduction

Hyperthermia, used to elevate the temperature of tumours to between 40° and 45°C, is proving to be an important adjuvant in cancer therapy when combined with chemo- and/or radiation therapy. More specifically, hyperthermia has been shown to improve clinical responses to radiation in several clinical trials in patients with breast Citation1, Citation2, head and neck Citation3, Citation4, melanoma Citation5, cervical Citation6, Citation7, and brain Citation8, Citation9 cancers and improves response to chemo-radiation therapy in patients with soft tissue sarcoma Citation10. Despite the growing documentation of the positive impact of hyperthermia as an adjuvant in cancer treatment, its use is still quite rare in comparison to other therapies. In part, this is due to a relative lack of new clinical trials that could document its effectiveness in various applications. However, the increased use of hyperthermia in cancer treatment may also be limited by a still-incomplete understanding of the precise mechanisms or cellular events by which temperature manipulation affects the tumour microenvironment or the efficacy of radiation or chemotherapy. Without more documentation of the complete range of cellular and/or molecular effects of temperature elevation, it is difficult to design the most effective clinical protocols.

Recently, there has been a growing appreciation for that fact that, due to physiological heat dissipating processes, the hyperthermic temperatures achieved in large regions of the tumour and surrounding area may be as mild as 39–40°C. Because this temperature range is not considered cytotoxic (at least not in treatment periods used in the clinic) the possibility exists that thermally sensitive activation of various cellular processes could be contributing to the beneficial effects seen with the addition of hyperthermia. One particularly attractive hypothesis is that the function of cells important for the anti-tumour immune system may be sensitive to thermal signals, possibly because of a natural, long-conserved sensitivity to elevated temperatures during fever. Based on a growing literature, it is apparent that the effect of hyperthermia on anti-tumour immunity is quite complex impacting various arms of the innate and adaptive immune response Citation11, Citation12. Excitingly, very recent studies in a clinically relevant animal model have demonstrated that adding hyperthermia to chemotherapy results in a curative therapy that is immunologically mediated Citation13. It has been widely reported that tumour infiltrating lymphocytes are essential for the inhibition of tumour growth and improved prognosis in various cancer types Citation14–19. Moreover, the specific immune cells located within tumours have been shown to be a favourable predictor of survival among colorectal cancer patients, even more accurate than histology Citation20. In particular, many of these observations indicate that the infiltration of CD8+ T cells into the tumour microenvironment is an immune mechanism benefiting patient survival. The effect of hyperthermia on effector CD8+ T cells is unknown in terms of antigen-specific and T cell-dependent events. However, our lab has shown that hyperthermia enhances the rate of antigen-dependent contact hypersensitivity through a T cell-mediated response Citation21. This increased rate of hypersensitivity leads to an increased infiltration of lymphocytes accompanied by swelling at the site of antigen contact and blood vessel dilation. Further, our recent studies have indicated that increasing temperature enhances the early activation and differentiation of naïve CD8+ T cells into effector cells [22]. Moreover, work by others Citation23 has shown that the differentiation of CD4+ T cells into TH1 cells is accompanied by increased interferon gamma (IFN-γ) production and subsequent promotion of CD8+ T cell-mediated adaptive immune response. Hyperthermia trials have shown an effect on serum cytokine levels Citation24, but whether thermal treatment can directly influence IFN-γ production is also not known. However, since effector, but not naïve CD8+ T cells are able to directly kill tumour cells, it would be very important to determine whether hyperthermia could enhance the functions of this differentiated subset.

Here, we address the question of whether mild heat treatment can enhance tumour antigen-specific, effector CD8+ T cell functions, and in particular, their cytotoxicity and IFN-γ production. We also examined the effects of mild heating on the signalling pathway known to be engaged when CD8+ T cells are activated. Finally, we show that temperature-induced effects on CD8+ T cell function are acting through events and signals mediated by the TCR in the plasma membrane. Collectively our data suggest that fever-range temperature can enhance important CD8+ T cell functions and thereby may help to promote tumour control and improve long-term survival in patients receiving hyperthermia as part of their therapy.

Methods

Animals and cell lines

C57BL/6 mice were obtained from National Cancer Institute (Bethesda, MA). B6.Cg-Thy1a/Cy Tg (TcraTcrb)8Rest/J (Pmel-1) mice were obtained from Jackson Laboratories (Bar Harbor, ME, USA). Mice were maintained in specific pathogen-free facilities and were treated in accordance with the guidelines established by the Animal Care and Use Committee at Roswell Park Cancer Institute (Buffalo, NY). B16.F10 melanoma cells and EL-4 lymphoma cells were obtained from the American Type Culture Collection (ATCC, Manassas, VA). Cells were cultured in 10% FBS, l0 mM L-glutamine, and 100 µg/mL penicillin/streptomycin in RPMI 1640 (Gibco, Grand Island, NY).

Antibodies, peptide, and reagents

PE-conjugated anti-CD8 mAb (53–6.7), FITC-conjugated Thy1.1 mAb (OX-7), FITC-conjugated CD8 mAb (Ly-2), and PE-Cy5-conjugated CD8 (53–6.7) were purchased from BD Pharmingen (San Diego, CA). p-LAT was purchased from Millipore and p-PKCθ, and β-actin antibodies were purchased from Cell Signaling Technology (Boston, MA) for western blot analysis. gp10022–33 peptide was synthesised and purchased from JPT peptide (Berlin, Germany). FITC-cholera toxin B (CTxB) was purchased from Sigma (St Louis, MO). Recombinant human IL-2 was purchased from R&D Systems (Minneapolis, MN).

In vitro generation of Pmel-1 effector CD8+ T cells

Pmel-1 splenocytes were cultured with 0.1 µg/mL of gp10025–33 peptide and 30 IU of rhIL-2 (R&D Systems) in 10% FBS, 2mM l0mM L-glutamine, and 100 µg/mL penicillin/streptomycin in RPMI (Gibco) for 5 days at 37°C. Cells were harvested and run over a Ficoll gradient. Effector CD8+ T cell phenotype was confirmed by FACS analysis.

51Cr release assay for Ag-specific target cell killing

Target cells were labelled with 100–150 µCi of Na51CrO4 (51Cr) (PerkinElmer, Waltham, MA) at 37°C for 1 h in FBS. After washing targets three times with 5% FBS in RPMI 1640 they were plated at 104 51Cr-labelled cells/well and mixed with effector Pmel-1 CD8+ T cells (incubated at indicated temperatures) in round bottom microtiter plates at indicated E:T ratios in triplicate. After 4 h incubation at 37°C, plates were centrifuged for 5 min at 500 × g, and 50 µL of supernatant was collected and counted in a MicroBeta TriLux liquid scintillation counter (PerkinElmer). Percent-specific lysis was determined as (experimental release − spontaneous release)/(maximal release − spontaneous release) × 100.

ELISA

Supernatants were collected 18 h after stimulating CD8+ T cells with C57BL/6 Ag-pulsed splenocytes. IFN-γ levels were measured in a sandwich ELISA using anti-IFN-γ capture mAb and biotin conjugated anti-IFN-γ mAb (XMG1.2) purchased from BD Pharmingen.

Cell staining

To examine GM1 clustering, cells were adhered onto Alcian blue-coated cover-slips, washed with PBS, and fixed with 4% paraformaldehyde. Cells were stained with FITC-CTxB, visualised, and quantified by fluorescent microscopy.

Western blot analysis

Cells were collected by centrifugation and lysed in lysis buffer (20 mM Tris (ph 7.5), 120 mM NaCl, 100 mM NaF, 0.5% Nonionic P40) containing protease inhibitors (10 mM sodium pyrophosphate (NaPPi), 4 mM phenylmethylsulphonyl fluoride (PMSF), 1 µg/mL Lleupeptin, 2 mM benzamidine, and 10 µg/mL aprotinin) and phosphatase inhibitors (200 µM sodium vanadate and 50 mM b-glycerophosphate) for 30 min on ice. Lysates were centrifuged at 12,000 × g for 10 min at 4°C and the supernatants were collected. Protein concentration in the supernatants was determined using the Pierce BCA protein assay kit. Lysates were boiled for 5 min in the presence of 1 × SDS-PAGE loading buffer and electrophoresced on a 10% SDS-polyacrylamide gel for 1.5 h at 100 V. The proteins were transferred to nitrocellulose and probed with indicated antibodies. The membrane was washed three times for 5 min with 0.1% Tween 20 and 1 × TBS. Detection was performed with the appropriate secondary Abs conjugated with HRP followed by ECL detection reagent (Pierce).

Quantitative RT-PCR analysis

Total RNA was prepared from CD8+ T cells stimulated with Ag-pulsed splenocytes using RNeasy kit (Qiagen, Valencia, CA). cDNA was synthesised using oligo-dT primers and SuperScript® polymerase (Invitrogen, Grand Island, NY). Quantitative real-time PCR was performed in an Applied Biosystems (Carlsbad, CA) 7900HT Fast Real-Time PCR system using SYBR Green PCR kit from Applied Biosystems and specific primers to amplify 200 bp fragments from different genes analysed. A threshold was set in the linear part of the amplification curve and the number of cycles needed to reach it was calculated for each gene. Normalisation was performed using primers to glyceraldehyde 3-phosphate dehydrogenase (GAPDH).

Statistical analysis

Results are expressed as mean ± standard deviation. Student's two-tailed t test was used for comparing experimental groups with p value < 0.05 considered significant.

Results

Ag-specific effector CD8+ T cell IFN-α production is temperature sensitive

Previous work has shown that non-antigen-specific T cell function has been enhanced with hyperthermia Citation25, Citation26. Moreover, our earlier work has demonstrated that elevated temperatures can enhance the differentiation of naïve CD8+ T cells into effector cells [22]. However, whether increased temperature affects Ag-specific, effector CD8+ T cell function is still unclear. Thus, we tested whether different physiological temperatures could regulate Ag-specific effector CD8+ T cell function, using antigen to achieve activation, rather than non-specific mitogens or crosslinking antibodies against surface receptors. Effector CD8+ T cells were generated from Pmel-1 splenocytes by stimulating cells for 5 days with 0.1 µg/mL gp10025–33 peptide and 30 IU rIL-2. Effector CD8+ T cells were then incubated at 33°, 37°, or 39.5°C for 6 h and re-stimulated with gp10023-33 peptide-pulsed C57BL/6 splenocytes for 18 h. First we examined production of IFN-γ because of the importance of this molecule in up-regulating MHC class I expression on tumour cells, allowing them to be recognised by the immune system. We found that Ag-specific production of IFN-γ by CD8+ T cells is temperature sensitive with the highest production of IFN-γ observed with incubation at 39.5°C (). Cells pre-incubated at 37°C had higher IFN-γ production than those incubated at 33°C. Re-stimulation of the CD8+ T cells with cognate peptide is still required, as temperature alone does not result in production of IFN-γ. The temperature sensitive effect on IFN-γ production by CD8+ T cells is enhanced with a greater duration of heating (). Pre-incubation at 39.5°C for 6 h resulted in the highest level of IFN-γ production by effector CD8+ T cells. Consistent with previously published results Citation27, Citation28, maximum effect on other immune cell subsets occurs by heating at 39.5°C for 6 h. We next determined the range of Ag concentrations at which thermal enhancement of IFN-γ production could be observed. We incubated effector CD8+ T cells at 33°, 37°, and 39.5°C for 6 h and then stimulated them with splenocytes pulsed with varying concentrations of gp10025–33 peptide. The effect of higher temperatures on enhancing IFN-γ production is maximal at a peptide concentration of 0.1 to 0.01 µg/mL. This enhancement is lost at higher peptide concentrations where IFN-γ production by cells incubated at 33° or 37°C is comparable to production by cells incubated at 39.5°C (). Thus, if Ag stimulation is optimal, exposure to elevated temperature does not further enhance IFN-γ. When CD8+ T cells are stimulated at sub-optimal Ag concentrations, higher temperatures can enhance CD8+ T cell IFN-γ production.

Figure 1. Ag-specific effector CD8+ T cell IFN-γ production is temperature sensitive. Effector Pmel-1 CD8+ T cells were generated by pulsing Pmel-1 splenocytes with 0.1 µg/mL gp10025-33 peptide (at 37°C) and 30 IU of rIL-2 for 5 days. (A) Effector Pmel-1 CD8+ T cells were incubated at 33°, 37°, and 39.5°C for 6 h and re-stimulated with C57BL/6 splenocytes pulsed with 0.1 µg/mL gp10025-33 peptide. Supernatants were removed after 18 h of co-incubation at 37°C and analysed for IFN-γ by ELISA. (B) Effector Pmel-1 CD8+ T cells were incubated at 33°, 37°, or 39.5°C for 2 to 6 h and activated with pulsed C57BL/6 splenocytes. Supernatants were analysed for IFN-γ by ELISA 18 h after pulsing. (C) Effector Pmel-1 CD8+ T cells were incubated at 33°, 37°, or 39.5°C for 6 h and stimulated with varying concentrations of gp10025-33 peptide pulsed C57BL/6 splenocytes. Results are reported as the mean ± SD. These results are representative of two independent experiments (p < 0.05, #vs 37°C and *vs 33°C).

Figure 1. Ag-specific effector CD8+ T cell IFN-γ production is temperature sensitive. Effector Pmel-1 CD8+ T cells were generated by pulsing Pmel-1 splenocytes with 0.1 µg/mL gp10025-33 peptide (at 37°C) and 30 IU of rIL-2 for 5 days. (A) Effector Pmel-1 CD8+ T cells were incubated at 33°, 37°, and 39.5°C for 6 h and re-stimulated with C57BL/6 splenocytes pulsed with 0.1 µg/mL gp10025-33 peptide. Supernatants were removed after 18 h of co-incubation at 37°C and analysed for IFN-γ by ELISA. (B) Effector Pmel-1 CD8+ T cells were incubated at 33°, 37°, or 39.5°C for 2 to 6 h and activated with pulsed C57BL/6 splenocytes. Supernatants were analysed for IFN-γ by ELISA 18 h after pulsing. (C) Effector Pmel-1 CD8+ T cells were incubated at 33°, 37°, or 39.5°C for 6 h and stimulated with varying concentrations of gp10025-33 peptide pulsed C57BL/6 splenocytes. Results are reported as the mean ± SD. These results are representative of two independent experiments (p < 0.05, #vs 37°C and *vs 33°C).

Ag-specific effector CD8+ T cell cytotoxicity is temperature sensitive

Another important anti-tumour effector function of CD8+ T cells is cytotoxicity. A previous study has shown that Fas ligand-specific killing is enhanced with hyperthermia Citation29; however, little is known about how Ag-specific T cell cytotoxicity is impacted by hyperthermia. To test Ag-specific CD8+ T cell cytotoxicity, effector Pmel-1 CD8+ T cells were generated in vitro by activating with C57BL/6 splenocytes pulsed with peptide for 5 days, and then effector cells were incubated at 33°, 37°, and 39.5°C for 6 h and then co-incubated with B16.F10 (gp100 expressing) or EL-4 (gp100 negative) 51Cr labelled tumour cells. Similar to the effect of temperature on IFN-γ production, effector Pmel-1 CD8+ T cells incubated at higher temperatures displayed an enhanced cytotoxicity against B16.F10 cells (). Cytotoxicity was antigen-specific as no lysis was observed when effector CD8+ T cells were co-incubated with chromium labelled EL-4 cells (). These results demonstrate that antigen-specific CD8+ T cell effector responses are highly dependent on temperature. These observations suggest an important role for mildly elevated temperature (i.e. during a fever or hyperthermia) in enhancing a cellular mediated T cell response (IFN-γ production and cytotoxicity) to antigen.

Figure 2. Mild hyperthermia enhances Ag-specific CD8+ cytotoxicity. Effector Pmel-1 CD8+ T cells were incubated at 33°, 37°, or 39.5°C for 6 h and then co-incubated with Cr51 labelled EL-4 (gp100 negative) or B16.F10 (gp100 positive) target cells for 4 h at 37°C. Supernatant was collected and percentage lysis was determined by chromium release by lysed target cells. Results are reported as the mean ± SD. These results are representative of two independent experiments.

Figure 2. Mild hyperthermia enhances Ag-specific CD8+ cytotoxicity. Effector Pmel-1 CD8+ T cells were incubated at 33°, 37°, or 39.5°C for 6 h and then co-incubated with Cr51 labelled EL-4 (gp100 negative) or B16.F10 (gp100 positive) target cells for 4 h at 37°C. Supernatant was collected and percentage lysis was determined by chromium release by lysed target cells. Results are reported as the mean ± SD. These results are representative of two independent experiments.

Temperature-dependent IFN-γ production does not depend on synthesis of new protein but does induce GM-1 clustering

To investigate the mechanisms by which increased temperature could affect IFN-γ production in effector CD8+ T cells, we first asked whether additional protein synthesis occurs during heating which could be mediating the thermal regulation of IFN-γ production when the CD8+ T cells are activated with Ag-pulsed splenocytes. CD8+ T cells were incubated with 10 µm cycloheximide (to block protein synthesis) during incubation at 33°, 37°, and 39.5°C and then washed three times with pre-warmed media and subsequently stimulated with Ag-pulsed splenocytes from C57BL/6 mice. After incubation for 18 h at 37°C supernatants were collected and analysed by ELISA for IFN-γ. Overall, there is a slight decrease in IFN-γ production compared to DMSO controls as some cycloheximide may have remained in culture after subsequent washes. However, cells incubated at 39.5°C, even when co-cultured with cycloheximide during the heating process, still resulted in more IFN-γ secretion compared to cells incubated at 33° and 37°C (). Thus, additional protein synthesis is not required for the temperature-dependent effect on Ag-specific IFN-γ production by CD8+ T cells.

Figure 3. Mild hyperthermia does not affect new protein synthesis but induces GM1 clustering in the plasma membrane. (A) Effector Pmel-1 CD8+ T cells were incubated at 33°, 37°, or 39.5°C for 6 h in the presence of 10 µm cycloheximide then stimulated with gp10025–33 peptide pulsed C57BL/6 splenocytes for 18 h. Supernatants were analysed for IFN-γ production by ELISA. (B) Effector cells were incubated at 33°, 37°, 39.5°C for 6 h and adhered onto alcian blue coated coverslips, fixed with paraformaldehyde. Cells were stained with FITC-CTxB, visualised for clustering by fluorescent microscopy, and (C) quantified. Results are reported as the mean ± SD. These results are representative of two independent experiments (p < 0.05, #vs 37°C and *vs 33°C).

Figure 3. Mild hyperthermia does not affect new protein synthesis but induces GM1 clustering in the plasma membrane. (A) Effector Pmel-1 CD8+ T cells were incubated at 33°, 37°, or 39.5°C for 6 h in the presence of 10 µm cycloheximide then stimulated with gp10025–33 peptide pulsed C57BL/6 splenocytes for 18 h. Supernatants were analysed for IFN-γ production by ELISA. (B) Effector cells were incubated at 33°, 37°, 39.5°C for 6 h and adhered onto alcian blue coated coverslips, fixed with paraformaldehyde. Cells were stained with FITC-CTxB, visualised for clustering by fluorescent microscopy, and (C) quantified. Results are reported as the mean ± SD. These results are representative of two independent experiments (p < 0.05, #vs 37°C and *vs 33°C).

Recent work has shown that incubating naïve CD8+ T cells at 39.5°C for 6 h induces the clustering of GM1+ cholesterol-dependent (CD)-microdomains in the plasma membrane Citation22. We asked whether effector CD8+ T cells were similarly impacted, thus changing the signalling required for IFN-γ production. Effector CD8+ T cells were incubated at 33°, 37°, and 39.5°C for 6 h and GM1 was stained using CTxB to look at possible alterations in GM1 organisation in the plasma membrane. GM1 proteins appeared either diffuse or clustered depending on thermal treatment (). Staining patterns were quantified by fluorescent microscopy, and we observed a higher percentage of effector CD8+ T cells with GM1 clustering at 39.5°C compared to cells incubated at 33° or 37°C (). This data suggests that temperature-induced changes at the plasma membrane may be associated with the enhanced Ag-specific CD8+ effector function.

Increased temperature does not affect non-antigen-specific CD8+ signalling that is independent of the TCR

Since elevated temperatures impact CD8+ T cell functions through altering the distribution of CD-microdomains, we hypothesised that CD8+ T cell activation through a stimulation signal bypassing the cellular membrane would not be influenced by temperature changes. Treatment with mitogen and ionophore (PMA and ionomycin) bypasses the need for TCR engagement on the membrane. We did not observe differences in IFN-γ production between any of the groups when the effector CD8+ T cells were incubated at 33°, 37°, 39.5°C for 6 h and then stimulated with PMA and ionomycin (). This indicates that the temperature-dependent effect on CD8+ T cell function is acting through events and signals involving the plasma membrane.

Figure 4. Mild hyperthermia does not affect effector CD8+ T cell activation when TCR signalling is bypassed. Effector Pmel-1 CD8+ T cells were incubated at 33°, 37°, 39.5°C for 6 h and activated with 0.5 µg/mL of ionomyocin and varying concentrations of PMA for 18 h at 37°C. Supernatants were collected and analysed for IFN-γ by ELISA. Results are reported as the mean ± SD. These results are representative of two independent experiments.

Figure 4. Mild hyperthermia does not affect effector CD8+ T cell activation when TCR signalling is bypassed. Effector Pmel-1 CD8+ T cells were incubated at 33°, 37°, 39.5°C for 6 h and activated with 0.5 µg/mL of ionomyocin and varying concentrations of PMA for 18 h at 37°C. Supernatants were collected and analysed for IFN-γ by ELISA. Results are reported as the mean ± SD. These results are representative of two independent experiments.

Elevated physiological temperatures enhance CD8+ T cell TCR signalling

We next investigated how temperature could affect early signalling events following ligation of TCR during an Ag-specific CD8+ T cell response. Phosphorylation of PKCθ and LAT plays an important role in mediating TCR signalling Citation30–32 and previous work from our lab has shown that following hyperthermia, PKC reorganisation and activity is enhanced in T cells Citation33. Therefore we asked how the phosphorylation of PKCθ and LAT might differ in antigen-stimulated Pmel-1 CD8+ T cells following pre-treatment with different temperatures. The effector Pmel-1 CD8+ T cells were incubated at 33°, 37°, and 39.5°C for 6 h and then activated for 0–30 min at 37°C (). Lysates were analysed by western blot for PKCθ and LAT and densitometry was used to quantify the relative phosphorylated protein levels compared to control β-actin expression. The phosphorylation of LAT and PKCθ were enhanced after 5 min of Ag-stimulation when pre-incubated at 39.5°C compared to a delayed phosphorylation when cells were pre-incubated at 33° or 37°C prior to Ag-stimulation (B and 5D). These results suggest that TCR signalling is enhanced in a temperature-dependent fashion and that higher temperatures increase the rate of activation. At later time points, the phosphorylation of LAT and PKC in the 37° and 39.5°C groups begins to decrease but remains higher in the 33°C pre-incubated cells. This suggests that signalling may not be occurring efficiently at lower temperatures (33°C).

Figure 5. Effector CD8+ T cell Ag-specific signalling is enhanced by mild hyperthermia. Cells were incubated at 33°, 37°, or 39.5°C for 6 h and stimulated with 0.1 µg/mL gp10025–33 peptide pulsed C57BL/6 splenocytes at 37°C. Stimulation was varied between 0–30 min. (A–D) Expression of phosphorylated LAT and PKCθ were determined by western blot. (A, C) Western blots. (B, D) Densitometry was performed using total β-actin levels as background controls. These results are representative of two independent experiments.

Figure 5. Effector CD8+ T cell Ag-specific signalling is enhanced by mild hyperthermia. Cells were incubated at 33°, 37°, or 39.5°C for 6 h and stimulated with 0.1 µg/mL gp10025–33 peptide pulsed C57BL/6 splenocytes at 37°C. Stimulation was varied between 0–30 min. (A–D) Expression of phosphorylated LAT and PKCθ were determined by western blot. (A, C) Western blots. (B, D) Densitometry was performed using total β-actin levels as background controls. These results are representative of two independent experiments.

Ag-specific effector CD8+ T cell IFN-γ gene transcription is regulated by temperature

To test whether these signalling effects resulted in enhanced IFN-γ gene transcription, CD8+ T cells were similarly incubated at 33°, 37°, and 39.5°C for 6 h and re-stimulated with Ag-pulsed C57BL/6 splenocytes for 0 to 2 h after which RNA was isolated. Ag-specific IFN-γ mRNA expression is regulated by temperature with higher-fold message levels in cells incubated at 39.5°C (). Cells pre-incubated at 33°C showed a slower rate of IFN-γ transcription than cells incubated at 37° and 39.5°C temperatures. This demonstrates that mild hyperthermia can result in changes in genes downstream of TCR signalling, thus resulting in the enhanced IFN-γ production observed at higher temperatures ().

Figure 6. Effector CD8+ T cell IFN-γ transcription is regulated by temperature. Effector CD8+ T cells were incubated at 33°, 37°, 39.5°C for 6 h and stimulated with 0.1 µg/mL gp10025–33 peptide pulsed C57BL/6 splenocytes for 0–2 h at 37°C. RNA was isolated and cDNA was synthesised. mRNA levels were assessed by real-time PCR. All message levels are relative to GAPDH controls and experimental gene expression is relative to cells activated with null peptide. Results are reported as the mean ± SD. These results are representative of two independent experiments (p < 0.05, #vs 37°C and *vs 33°C).

Figure 6. Effector CD8+ T cell IFN-γ transcription is regulated by temperature. Effector CD8+ T cells were incubated at 33°, 37°, 39.5°C for 6 h and stimulated with 0.1 µg/mL gp10025–33 peptide pulsed C57BL/6 splenocytes for 0–2 h at 37°C. RNA was isolated and cDNA was synthesised. mRNA levels were assessed by real-time PCR. All message levels are relative to GAPDH controls and experimental gene expression is relative to cells activated with null peptide. Results are reported as the mean ± SD. These results are representative of two independent experiments (p < 0.05, #vs 37°C and *vs 33°C).

Discussion

New strategies to enhance the activity of CD8+ T cells are critical for achieving long-term protection from cancer recurrence. T cell infiltration of tumours has been documented to be very important in overall survival, but substantial evidence exists that a suppressive microenvironment within the tumour limits the activation and function of CD8+ T cells. Indeed, many different factors and cell types within the tumour exert suppressive forces against a CD8+ T cell anti-tumour response. For example, factors such as IL-6, VEGF, MMPs, GM-CSF, IL-10 and many other cytokines and chemokines can be found within the tumour microenvironment and are responsible for the suppression of an anti-tumour immune response Citation34–36.

These factors and others promote the accumulation of cells including myeloid-derived suppressor cells (MDSCs), which suppress CD8+ T cell activity by secreting ROS, depleting arginine, and decreasing TCR-associated signalling proteins Citation37–39. MDSCs also secrete anti-inflammatory cytokines including IL-10 and TGF-β which promote differentiation of T regulatory (Treg) cells that also inhibit CD8+ T cell anti-tumour activity. A recent study suggested that infiltrating DCs can suppress CD8+ T cell function by inhibiting T cell L-arginine metabolism in a spontaneous mammary tumour model leading to decreased T cell expansion and IFN-γ production, ultimately resulting in anergy Citation40.

Since multiple factors and cell types within the tumour microenvironment can suppress T cell activity, new therapies or adjuvants that could help to increase the activation and function of these cells could overcome this suppression would be important in generating an improved anti-tumour response. In this report we document the ability of mild temperature elevation to significantly enhance activities (IFN-γ production and cytotoxicity) that would be important in tumour control, and we have associated these effects with changes in the organisation of the plasma membrane and downstream signalling pathways, alterations which may be involved in the underlying mechanisms by which elevated temperature affects cellular function. Earlier studies in our laboratory have reported that heating mice in vivo enhances PKC activity and results in the aggregation of PKC and the cytoskeletal proteins spectrin and vimentin Citation27, Citation41. We have shown in this study that elevated temperature results in enhanced downstream signalling, with greater levels of phosphorylated PKCθ and LAT. Subsequent enhancement in IFN-γ gene transcription was observed when effector CD8+ T cells were incubated at higher temperatures and that these changes are also associated temporally with a thermally induced reorganisation of membrane domains.

It will now be important to determine whether these thermally enhanced CD8+ T cell functions could play a role in controlling tumour growth in vivo. Previous studies have demonstrated general enhancement of immune activity against tumours following mild hyperthermia, but these studies have not studied a tumour antigen-dependent response. For example, in mice, fever-range hyperthermia has been shown to enhance anti-tumour immunity through both the innate and adaptive immune systems Citation42. Hyperthermia has also been shown to significantly enhance efficiency of heat shock protein vaccines Citation43. When combined with intratumoural DC injection, hyperthermia induces DC migration to the tumour draining lymph nodes and enhances the priming of CTLs in animal melanoma models Citation44. Clinically, Guo et al. have shown that patients with advanced melanoma treated three times a week with local hyperthermia followed by intratumoural injections of immature DCs experienced significantly longer time to tumour progression (p < 0.05) Citation45. Furthermore, this study demonstrated that DC vaccination in combination with hyperthermia resulted in an increased infiltration of activated CD8+ cells into the tumour site accompanied by decreased infiltration of immune suppressive Treg cells, possibly creating an environment for improved tumour control Citation45. Recently, long-term anti-tumour immunity was achieved in murine models; treatment with oxaliplatin chemotherapy followed 24 h later by 6 h whole body hyperthermia was able to cure all primary and metastatic tumours in 50% of MTLn3 tumour-bearing rats Citation13.

We have shown that mild hyperthermia increases the clustering of GM-1+ regions within the plasma membrane (). These GM-1 regions contain important signalling molecules, such as TCR, Lck, LAT and many other proteins important in recognising antigen presented by an APC or target cell Citation46. Formation of the immunological synapse between an APC and target cells requires the aggregation and localisation of these signalling enriched GM-1 membrane domains to occur at this synapse Citation47. The hyperthermia-induced increase in GM-1 clustering could potentially pre-condition effector T cells and allow them to react faster to antigen presentation and initiate killing mechanisms than at lower temperatures.

In summary, this study presents novel information describing the role of hyperthermia in improving tumour antigen-specific, effector CD8+ T cell function. Our studies were preformed in Pmel-1 transgenic mice which carry a rearranged TCR specific for gp100 Citation48. While most prior work has been on non-specific antigen-dependent immune responses, the gp100 protein is expressed by most melanoma cells, making this an important model for future in vivo work on the antigen-specific immune response. Furthermore, the fundamental observations made here may expedite the design and implementation of new clinical protocols utilising hyperthermia as an adjuvant immunotherapy.

Acknowledgements

We thank members of the Department of Flow and Image Cytometry Core Facility as well as the Roswell Park Cancer Institute Animal Resource Facility. We also thank Jeanne Prendergast for her assistance in the lab and Bonnie Hylander for her help in reviewing this manuscript.

Declaration of interest: This work was supported by grants NIH R01 CA135368-01A1, R01 CA071599-11, 2 T32 CA085183 Immunology Department Training Grant, and the Roswell Park Cancer Institute Core grant CA16056. The authors alone are responsible for the content and writing of the paper.

References

  • Vernon CC, Hand JW, Field SB, Machin D, Whaley JB, van der Zee J, et al. Radiotherapy with or without hyperthermia in the treatment of superficial localized breast cancer: Results from five randomized controlled trials. International Collaborative Hyperthermia Group. Int J Radiat Oncol Biol Phys 1996; 35: 731–744
  • Jones EL, Oleson JR, Prosnitz LR, Samulski TV, Vujaskovic Z, Yu D, et al. Randomized trial of hyperthermia and radiation for superficial tumors. J Clin Oncol 2005; 23: 3079–3085
  • Datta NR, Bose AK, Kapoor HK, Gupta S. Head and neck cancers: Results of thermoradiotherapy versus radiotherapy. Int J Hyperthermia 1990; 6: 479–486
  • Valdagni R, Amichetti M. Report of long-term follow-up in a randomized trial comparing radiation therapy and radiation therapy plus hyperthermia to metastatic lymph nodes in stage IV head and neck patients. Int J Radiat Oncol Biol Phys 1994; 28: 163–169
  • Overgaard J, Gonzalez Gonzalez D, Hulshof MC, Arcangeli G, Dahl O, et al. Randomised trial of hyperthermia as adjuvant to radiotherapy for recurrent or metastatic malignant melanoma. European Society for Hyperthermic Oncology. Lancet 1995; 345: 540–543
  • Van der Zee J, Van Rhoon GC, Broekmeyer-Reurink MP, Kuijs AE, Van Den Berg AP, Treurniet-Donker D, et al. Clinical experience with local hyperthermia in Rotterdam. Adv Exp Med Biol 1990; 267: 383–386
  • Harima Y, Nagata K, Harima K, Ostapenko VV, Tanaka Y, Sawada S. A randomized clinical trial of radiation therapy versus thermoradiotherapy in stage IIIB cervical carcinoma. Int J Hyperthermia 2001; 17: 97–105
  • Sneed PK, Stauffer PR, McDermott MW, Diederich CJ, Lamborn KR, Prados MD, et al. Survival benefit of hyperthermia in a prospective randomized trial of brachytherapy boost ± hyperthermia for glioblastoma multiforme. Int J Radiat Oncol Biol Phys 1998; 40: 287–295
  • Falk MH, Issels RD. Hyperthermia in oncology. Int J Hyperthermia 2001; 17: 1–18
  • Issels RD, Lindner LH, Verweij J, Wust P, Reichardt P, Schem BC, 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
  • Baronzio G, Gramaglia A, Fiorentini G. Hyperthermia and immunity. A brief overview. In Vivo 2006; 20: 689–695
  • Peer AJ, Grimm MJ, Zynda ER, Repasky EA. Diverse immune mechanisms may contribute to the survival benefit seen in cancer patients receiving hyperthermia. Immunol Res 2010; 46: 137–154
  • Rowe RW, Strebel FR, Proett JM, Deng W, Chan D, He G, et al. Fever-range whole body thermotherapy combined with oxaliplatin: A curative regimen in a pre-clinical breast cancer model. Int J Hyperthermia 2010; 26: 565–576
  • Naito Y, Saito K, Shiiba K, Ohuchi A, Saigenji K, Nagura H, et al. CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer. Cancer Res 1998; 58: 3491–3494
  • Diederichsen AC, Hjelmborg JB, Christensen PB, Zeuthen J, Fenger C. Prognostic value of the CD4+/CD8+ ratio of tumour infiltrating lymphocytes in colorectal cancer and HLA-DR expression on tumour cells. Cancer Immunol Immunother 2003; 52: 423–428
  • Fukunaga A, Miyamoto M, Cho Y, Murakami S, Kawarada Y, Oshikiri T, et al. CD8+ tumor-infiltrating lymphocytes together with CD4+ tumor-infiltrating lymphocytes and dendritic cells improve the prognosis of patients with pancreatic adenocarcinoma. Pancreas 2004; 28: e26–31
  • Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F, et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci USA 2005; 102: 18538–18543
  • Piersma SJ, Jordanova ES, van Poelgeest MI, Kwappenberg KM, van der Hulst JM, Drijfhout JW, et al. High number of intraepithelial CD8+ tumor-infiltrating lymphocytes is associated with the absence of lymph node metastases in patients with large early-stage cervical cancer. Cancer Res 2007; 67: 354–361
  • Yamada N, Oizumi S, Kikuchi E, Shinagawa N, Konishi-Sakakibara J, Ishimine A, et al. CD8+ tumor-infiltrating lymphocytes predict favorable prognosis in malignant pleural mesothelioma after resection. Cancer Immunol Immunother 2010; 59: 1543–1549
  • Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pages C, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 2006; 313: 1960–1964
  • 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
  • Mace TA, Zhong L, Kilpatrick C, Zynda E, Lee C-T, Capitano M, et al. Differentiation of CD8+ T cells into effector cells is enhanced by physiological range hyperthermia. J Leukocyte Biol 2011
  • Szabo SJ, Sullivan BM, Peng SL, Glimcher LH. Molecular mechanisms regulating Th1 immune responses. Annu Rev Immunol 2003; 21: 713–758
  • Atanackovic D, Nierhaus A, Neumeier M, Hossfeld DK. Hegewisch-Becker S. 41.8°C whole body hyperthermia as an adjunct to chemotherapy induces prolonged T cell activation in patients with various malignant diseases. Cancer Immunol Immunother 2002; 51: 603–613
  • Hanson DF. Fever and the immune response. The effects of physiological temperatures on primary murine splenic T-cell responses in vitro. J Immunol 1993; 151: 436–448
  • Smith JB, Knowlton RP, Agarwal SS. Human lymphocyte responses are enhanced by culture at 40°C. J Immunol 1978; 121: 691–694
  • 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
  • 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
  • Cippitelli M, Fionda C, Di Bona D, Piccoli M, Frati L, Santoni A. Hyperthermia enhances CD95-ligand gene expression in T lymphocytes. J Immunol 2005; 174: 223–232
  • Shim EK, Jung SH, Lee JR. Role of two adaptor molecules SLP-76 and LAT in the PI3K signaling pathway in activated T cells. J Immunol 2011; 186: 2926–2935
  • Huang SC, Tsai HF, Tzeng HT, Liao HJ, Hsu PN. Lipid raft assembly and Lck recruitment in TRAIL costimulation mediates NF-kappaB activation and T cell proliferation. J Immunol 2011; 186: 931–939
  • Ma JS, Haydar TF, Radoja S. Protein kinase C delta localizes to secretory lysosomes in CD8+ CTL and directly mediates TCR signals leading to granule exocytosis-mediated cytotoxicity. J Immunol 2008; 181: 4716–4722
  • Wang XY, Ostberg JR, Repasky EA. Effect of fever-like whole-body hyperthermia on lymphocyte spectrin distribution, protein kinase C activity, and uropod formation. J Immunol 1999; 162: 3378–3387
  • Bonecchi R, Locati M, Mantovani A. Chemokines and cancer: A fatal attraction. Cancer Cell 2011; 19: 434–435
  • Keeley EC, Mehrad B, Strieter RM. CXC chemokines in cancer angiogenesis and metastases. Adv Cancer Res 2010; 106: 91–111
  • Grivennikov SI, Karin M. Inflammatory cytokines in cancer: Tumour necrosis factor and interleukin 6 take the stage. Ann Rheum Dis 2011; 70: S104–108
  • Bronte V, Serafini P, Mazzoni A, Segal DM, Zanovello P. L-arginine metabolism in myeloid cells controls T-lymphocyte functions. Trends Immunol 2003; 24: 302–306
  • Kusmartsev S, Nefedova Y, Yoder D, Gabrilovich DI. Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J Immunol 2004; 172: 989–999
  • Ezernitchi AV, Vaknin I, Cohen-Daniel L, Levy O, Manaster E, Halabi A, et al. TCR zeta down-regulation under chronic inflammation is mediated by myeloid suppressor cells differentially distributed between various lymphatic organs. J Immunol 2006; 177: 4763–4772
  • Norian LA, Rodriguez PC, O'Mara LA, Zabaleta J, Ochoa AC, Cella M, et al. Tumor-infiltrating regulatory dendritic cells inhibit CD8+ T cell function via L-arginine metabolism. Cancer Res 2009; 69: 3086–3094
  • Di YP, Repasky E, Laszlo A, Calderwood S, Subjeck J. Hsp70 translocates into a cytoplasmic aggregate during lymphocyte activation. J Cell Physiol 1995; 165: 228–238
  • Burd R, Dziedzic TS, Xu Y, Caligiuri MA, Subjeck JR, Repasky EA. 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
  • Wang XY, Kazim L, Repasky EA, Subjeck JR. Characterization of heat shock protein 110 and glucose-regulated protein 170 as cancer vaccines and the effect of fever-range hyperthermia on vaccine activity. J Immunol 2001; 166: 490–497
  • Tanaka K, Ito A, Kobayashi T, Kawamura T, Shimada S, Matsumoto K, et al. Intratumoral injection of immature dendritic cells enhances antitumor effect of hyperthermia using magnetic nanoparticles. Int J Cancer 2005; 116: 624–633
  • Guo J, Zhu J, Sheng X, Wang X, Qu L, Han Y, 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
  • Patel VP, Moran M, Low TA, Miceli MC. A molecular framework for two-step T cell signaling: Lck Src homology 3 mutations discriminate distinctly regulated lipid raft reorganization events. J Immunol 2001; 166: 754–764
  • Huse M, Quann EJ, Davis MM. Shouts, whispers and the kiss of death: Directional secretion in T cells. Nat Immunol 2008; 9: 1105–1111
  • Overwijk WW, Theoret MR, Finkelstein SE, Surman DR, de Jong LA, Vyth-Dreese FA, et al. Tumor regression and autoimmunity after reversal of a functionally tolerant state of self-reactive CD8+ T cells. J Exp Med 2003; 198: 569–580

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.