Next Article in Journal
The Safety and Clinical Validity of Endoscopic Submucosal Dissection for Early Gastric Cancer in Patients Aged More Than 85 Years
Next Article in Special Issue
Tumor Infiltrating Lymphocyte (TIL) Therapy for Solid Tumor Treatment: Progressions and Challenges
Previous Article in Journal
Physical Fitness and Frailty in Males after Allogeneic Hematopoietic Stem Cell Transplantation in Childhood: A Long-Term Follow-Up Study
Previous Article in Special Issue
Exploring Real World Outcomes with Nivolumab Plus Ipilimumab in Patients with Metastatic Extra-Pulmonary Neuroendocrine Carcinoma (EP-NEC)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Store-Operated Ca2+ Entry Is Up-Regulated in Tumour-Infiltrating Lymphocytes from Metastatic Colorectal Cancer Patients

1
Laboratory of General Physiology, Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy
2
Department of Biology, College of Science, Salahaddin University, Erbil 44001, Iraq
3
Department of Sciences Clinic-Surgical, Diagnostic and Pediatric, University of Pavia, 27100 Pavia, Italy
4
Pediatric Clinic, Foundation IRCCS Policlinico San Matteo, 27100 Pavia, Italy
5
Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 27100 Novara, Italy
6
UOSD Cell Factory, Pediatric Hematology/Oncology, Foundation IRCCS Policlinico San Matteo, 27100 Pavia, Italy
7
Department of Molecular Medicine, Human Physiology Unit, University of Pavia, 27100 Pavia, Italy
8
Filippo Conca and Konstantinos Lefkimmiatis, Foundation for Advanced Biomedical Research, Veneto Institute of Molecular Medicine, 35129 Padua, Italy
9
Internal Medicine and Medical Therapy, University of Pavia, 27100 Pavia, Italy
10
Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2022, 14(14), 3312; https://doi.org/10.3390/cancers14143312
Submission received: 10 June 2022 / Revised: 3 July 2022 / Accepted: 4 July 2022 / Published: 7 July 2022
(This article belongs to the Special Issue Advances in Cancer Immunotherapy)

Abstract

:

Simple Summary

Store-operated Ca2+ entry (SOCE) has long been known to regulate the differentiation and effector functions of T cells as well as to be instrumental to the ability of cytotoxic T lymphocytes to target cancer cells. Currently, no information is available regarding the expression and function of SOCE in tumour-infiltrating lymphocytes (TILs) that have been expanded in vitro for adoptive cell therapy (ACT). This study provides the first evidence that SOCE is up-regulated in ex vivo-expanded TILs from metastatic colorectal cancer (mCRC) patients. The up-regulation of SOCE mainly depends on diacylglycerol kinase (DGK), which prevents the protein kinase C-dependent inhibition of Ca2+ entry in normal T cells. Of note, the pharmacological blockade of SOCE with the selective inhibitor, BTP-2, during target cell killing significantly increases cytotoxic activity at low TIL density, i.e., when TILs-mediated cancer cell death is rarer. This study, albeit preliminary, could lay the foundation to propose an alternative strategy to effect ACT. It has been shown that ex vivo-expanded TILs did not improve the disease-free survival rate in mCRC patients. Our results strongly suggest that pre-treating autologous TILs with a SOCE or DGK inhibitor before being infused into the patient could improve their cytotoxic activity against cancer cells.

Abstract

(1) Background: Store-operated Ca2+ entry (SOCE) drives the cytotoxic activity of cytotoxic T lymphocytes (CTLs) against cancer cells. However, SOCE can be enhanced in cancer cells due to an increase in the expression and/or function of its underlying molecular components, i.e., STIM1 and Orai1. Herein, we evaluated the SOCE expression and function in tumour-infiltrating lymphocytes (TILs) from metastatic colorectal cancer (mCRC) patients. (2) Methods: Functional studies were conducted in TILs expanded ex vivo from CRC liver metastases. Peripheral blood T cells from healthy donors (hPBTs) and mCRC patients (cPBTs) were used as controls. (3) Results: SOCE amplitude is enhanced in TILs compared to hPBTs and cPBTs, but the STIM1 protein is only up-regulated in TILs. Pharmacological manipulation showed that the increase in SOCE mainly depends on tonic modulation by diacylglycerol kinase, which prevents the protein kinase C-dependent inhibition of SOCE activity. The larger SOCE caused a stronger Ca2+ response to T-cell receptor stimulation by autologous mCRC cells. Reducing Ca2+ influx with BTP-2 during target cell killing significantly increases cytotoxic activity at low target:effector ratios. (4) Conclusions: SOCE is enhanced in ex vivo-expanded TILs deriving from mCRC patients but decreasing Ca2+ influx with BTP-2 increases cytotoxic activity at a low TIL density.

Graphical Abstract

1. Introduction

Colorectal carcinoma (CRC) represents the third most common cause of malignancy-associated mortality, which is mainly due to the development of liver metastases [1]. Surgical resection represents the only potentially curative treatment in combination with adjuvant or perioperative chemotherapy [1]. Unfortunately, metastatic recurrence affects ≈80% of the patients [1], the majority of whom succumb to disease progression [2]. Therefore, the design of a more effective strategy to fully eradicate liver metastases is a compelling need for metastatic CRC (mCRC) patients.
Adoptive cell therapy (ACT) represents a promising approach to achieve complete and durable cancer eradication by the transfer of ex vivo-expanded cells reactive against tumour [3,4]. Studies on metastatic melanoma demonstrated that autologous tumour-infiltrating lymphocytes (TILs), ex vivo-expanded in the presence of interleukin-2 (IL-2) and then reinoculated into the patients, may be exploited to induce a long-lasting (up to 3 years) disease regression [5,6]. The ability of TILs to induce a complete response and promote tumour regression was recently reported also for cervical carcinoma [7]. CD8+ cytotoxic T lymphocytes (CTLs), which are induced to recognise specific tumour-associated antigens (TAAs), have also been shown to be effective against types of solid tumours [8,9,10,11,12]. The short-term expansion of naturally occurring tumour-reactive CD8+ TILs from the liver metastases of CRC patients has recently been described by two independent groups [13,14]. However, it is still unclear whether these TILs could be employed to effectively target metastatic CRC (mCRC) [15]. Unravelling whether the intracellular signalling pathways that are recruited downstream of T-cell receptors (TCR) are deranged is mandatory to fully exploit the therapeutic potential of expanded TILs against cancer cells.
An increase in intracellular Ca2+ concentration ([Ca2+]i) is of paramount importance to promote the differentiation and effector functions of T cells by driving processes that are key to the immune response, including proliferation, differentiation, cytokine secretion and cytotoxicity [16,17]. TCR engagement results in the activation of phospholipase C-1γ1 (PLCγ1) that cleaves phosphatidylinositol-4,5-bisphosphate (PIP2) into inositol-1,4,5-trisphosphate (InsP3) and diacylglycerol (DAG). InsP3 binds to and gates the ionotropic InsP3 receptors (InsP3Rs), which mediate Ca2+ release from the endoplasmic reticulum (ER), the largest endogenous Ca2+ reservoir in T cells. The following reduction in ER Ca2+ concentration ([Ca2+]ER) is detected by Stromal Interaction Molecule 1 (STIM1), which serves as sensor of [Ca2+]ER and is thereafter prompted to translocate from perinuclear to peripheral ER cisternae, where STIM1 traps and activates the Ca2+-selective Orai1 channels at the plasma membrane [16,18,19]. The following influx of Ca2+, known as store-operated Ca2+ entry (SOCE), induces the nuclear translocation of the Ca2+-dependent nuclear factor of activated T cells c2 (NFATc2 or NFAT1), cytokine production and cytotoxicity [16,17,20]. Therefore, ex vivo-expanded TILs are predicted to impinge on a functional SOCE to be therapeutically effective against cancer cells. In accord, the pharmacological or genetic deletion of SOCE impairs the cytotoxic activity of human [21,22] and murine [23] CTLs, thereby favouring the engraftment of cancer cells and promoting tumour growth [23]. Surprisingly, although neoplastic transformation may induce a dramatic derangement in the expression and/or function of the SOCE machinery [24,25,26,27,28,29,30], it is still unknown whether a functional SOCE is expressed in TILs that are generated for ACT. Herein, we addressed this compelling issue by focusing on ex vivo-expanded TILs isolated from the liver metastases of mCRC patients. Peripheral blood T (PBT) cells from healthy donors (hPBTs) and mCRC patients (cPBTs) were used as controls, as recently suggested [31,32].

2. Materials and Methods

2.1. Patients and Donors

This study was approved by the Institutional Review Board of the Foundation IRCCS Policlinico San Matteo (Protocol Number 20190069408). After signing an informed consent form, 10 patients (>18 years) affected by mCRC who had undergone surgery to remove liver metastases were enrolled (Table S1). All procedures were performed according to the guidelines prescribed for the treatment of CRC neoplasia, and no patient was subjected to unnecessary invasive procedures. Peripheral blood cells were derived from the same mCRC patients (cPBT) at the time of surgery or from the buffy coats of 10 healthy donors (hPBT). Buffy-coat units were released by the Blood Bank of SIMT, Policlinico San Matteo, Pavia, according to Article 8 of the Legislative Decree 2, November 2015, of the Italian Department of Health, with blood donor-specific informed consent implemented by the Blood Bank Conference of Regione Lombardia.

2.2. Isolation of TILs and Tumour Cells

Tumour specimens (liver metastases) from 10 patients (Table S1) were processed as previously described [9] with the GentleMACS Dissociator (Miltenyi Biotec, Bologna, Italy, cat# 130-093-235) after being treated with the Tumor Dissociation Kit (Miltenyi Biotech, cat# 130-095-929). After this procedure, the tumour cells were filtered (Cell Strainers, Falcon, Corning Stone, Staffordshire, England, cat# 352350) to remove clusters and to obtain a single-cell suspension before magnetic separation. The cells were then checked for viability by trypan blue dye exclusion and resuspended in buffer containing phosphate-buffered saline (PBS, Euroclone, Pero, Italy, cat# ECB4004L) pH 7.2, 0.5% bovine serum albumin (BSA, Kedrion Biopharma, Barga, Italy) for the isolation of CD45-positive cells, using human CD45 microbeads (cat# 130-118-780) and an LS Column (cat# 130-042-401) (Miltenyi Biotec), according to the manufacturers’ instructions. Negative and positive cells were collected, centrifuged and checked for viability and recovery.

2.3. Expansion of mCRC Cells

Negative cells, which contain tumour cells, were plated in 12-well plates at a concentration of 0.5–1 × 106 cells/mL of CellGro SCGM (Cell Genix, Freiburg, Germany, cat# 20802-0500), supplemented with 20% foetal bovine serum (FBS) (Euroclone, cat# ECS0180D) and 0.1% gentamicin (Gibco, Life Technologies Limited, Paisley, UK, cat# 15750-037), and cultured in 25 cm2 tissue flasks (Corning Stone, Staffordshire, UK) at 37 °C and 5% CO2. Viable tumour cells attached to the flask within 12–24 h. At the first medium change, the cells were put into a fresh flask. The cells at 75% to 100% confluence were detached with 0.25% trypsin and 0.02% EDTA (Euroclone) in a calcium/magnesium-free balanced solution. The culture medium was changed twice a week and cellular homogeneity was evaluated microscopically every 24–48 h. Tumour cells derived from early passages (3–5) cultures were cryopreserved in 90% FBS and 10% DMSO (Alchimia, Ponte San Nicolò, Italy, cat# CRN 001-00) and stored in liquid nitrogen for further experiments. To confirm the neoplastic origin of the cultured cells, the cells underwent morphological and immunocytochemical analysis, as previously described [9,26]. We were able to isolate and grow tumour cells from all 10 patients enrolled in this study.

2.4. TIL Expansion and Characterisation

We designed a methodological approach to optimise TIL expansion from mCRC samples after the isolation of lymphoid cells using CD45 TIL Microbeads (Miltenyi Biotec). CD45+ cells were rapidly expanded for 14 days at 37 °C in six-well plates (Corning Stone) in the CellGRO medium in the presence of OKT3 (30 ng/mL) (Miltenyi Biotech, cat# 170-076-124), IL-15 (10 ng/mL) (Miltenyi Biotech, cat# 170-076-114), IL-2 (3000 U) (Novartis, Basilea, Switzerland, cat# AIC 02713110/M) and irradiated (50 Gy) feeder cells. On days 7 and 10, the cells were monitored, counted and split with medium containing IL-2 and IL-15; on day 14, the cells were checked for viability and then cryopreserved for further characterisation. Based upon our preliminary experiments aimed to optimise the expansion method, we documented that a 14-day culture represents the best timepoint for the expansion rate and vitality of recovered TILs; further expansion might reduce the number of viable cells.
The TILs were analysed by immunostaining with specific monoclonal antibodies, followed by analysis using flow cytometry (BD FACS Canto II, Beston, Dickinson and Company, BD Biosciences, Franklin Lakes, NJ, USA), as previously described [9]. The following conjugated monoclonal antibodies were used: anti-CD45, anti-CD3 (cat# IM2467), anti-CD4 (cat# 651849), anti-CD8 (cat# 641400) and anti-CD56 (cat# 345810) (Beckman Coulter, Brea, CA, USA & BD Biosciences). The cytotoxic capacity of the TILs was evaluated in a 5 h cytotoxicity assay against the colorectal carcinoma SW480 cell line (obtained from ATCC, Sesto San Giovanni, Milan, Italy) and against autologous mCRC cells labelled with Chromium-51 (51Cr). Briefly, SW 480 cells were labelled overnight (ON) in 200 µL with 20 µL of 51Cr, whereas adherent, non-confluent mCRC cells were washed and then labelled ON with 20 µL of 51Cr. The day after, the SW 480 cells were washed, while the mCRC cells were detached and washed. The SW480 and mCRC cells were then added to TILs at varying effector:target (E:T) ratios. After 5 h incubation at 37 °C, supernatants were harvested for the quantification of chromium release. As a control, we employed donor-derived anti-leukaemia CTL generated and expanded using a methodological approach that includes leukaemia-specific stimulations followed by a rapid expansion using the same protocol employed for TILs [33].

2.5. Total RNA Extraction and Real-Time PCR

Total mRNA was extracted from 0.5 × 106 cells using a SPLIT RNA Extraction Kit (Lexogen, Austria, cat# 008.48) according to the manufacturer’s instructions, as described in [34]. cDNA was synthesised from 0.5–1 µg of total RNA using a SensiFAST cDNA Synthesis Kit (BioLine, London, UK, Cat. BIO-65054). Real-Time PCR was performed using iTaq qPCR master mix according to the manufacturer’s instructions (Bio-Rad Laboratories, Hercules, CA, USA; cat# 1725124) on an SFX96 Real-Time system (Bio-Rad Laboratories). An S18 ribosomal subunit was used to normalise the raw data. The data are calculated for each sample as 2^-(Ct(GOI) − Ct(S18)) and expressed as mean ± standard deviation from three independent cultures, each run in triplicate. The primers’ sequences are provided in Table S2.

2.6. Immunoblotting

The cells were lysed with 100 µL of lysis buffer (50 mM Tris-HCl, pH = 7.4), sodium dodecyl sulphate (SDS) 0.5%, 5 mM EDTA, 10 µL of protease inhibitors cocktail (PIC) (Millipore, cat# 539133) and phosphatase inhibitor cocktail (ThermoFisher Scientific, Waltham, MA, USA; cat# 78428) and collected in a 1.5 mL tube. The lysates were boiled at 96 °C for 5 min and then quantified with a QuantiPro BCA Assay Kit (Sigma-Aldrich, Milan, Italy; cat# SLBF3463). Then, 10 µg (for STIM1) and 20 µg (for ORAI1) of proteins were mixed with the right amount of Laemmli Sample Buffer 4X (Bio-Rad Laboratories), and boiled. The samples were then loaded onto a 12% polyacrylamide–sodium dodecyl sulphate gel for SDS-PAGE. The proteins were transferred onto nitrocellulose membrane, using Mini Transfer Packs with Trans-Blot® Turbo TM (Bio-Rad Laboratories) according to manufacturer’s instructions (Bio-Rad Laboratories). The membranes were blocked in 5% skimmed milk (Sigma, cat# 70166) for 45 min at room temperature. Subsequently, the membranes were incubated with indicated primary antibody, overnight, at 4 °C. The primary antibodies used are listed in Table S3; anti-β-Actin was used to normalise the protein loading.

2.7. Solutions

The physiological salt solution (PSS) had the following composition: 150 mM NaCl, 6 mM KCl, 1.5 mM CaCl2, 1 mM MgCl2, 10 mM glucose, 10 mM HEPES. In Ca2+-free solution (0Ca2+), Ca2+ was replaced with 2 mM NaCl and 0.5 mM EGTA was added. The solutions were titrated to pH 7.4 with NaOH. The osmolality of the extracellular solution, as measured with an osmometer (Wescor 5500), was 300–310 mmol/kg.

2.8. [Ca2+]i Measurements

The T cells were loaded with 2 µM Fura-2 acetoxymethyl ester (Fura-2/AM; 1 mM stock in dimethyl sulfoxide) in PSS for 30 min at 37 °C, as previously shown for mCRC cells [26]. After washing in PSS, the coverslip was fixed to the bottom of a Petri dish and the cells observed by an upright epifluorescence Axiolab microscope (Carl Zeiss, Oberkochen, Germany), usually equipped with a Zeiss ×40 Achroplan objective (water-immersion, 2.0 mm working distance, 0.9 numerical aperture). The T cells were excited alternately at 340 and 380 nm, and the emitted light was detected at 510 nm. A first neutral density filter (1 or 0.3 optical density) reduced the overall intensity of the excitation light, and a second neutral density filter (optical density = 0.3) was coupled to the 380 nm filter to approach the intensity of the 340 nm light. A round diaphragm was used to increase the contrast. The excitation filters were mounted on a filter wheel (Lambda 10, Sutter Instrument, Novato, CA, USA). Custom software, working in the LINUX environment, was used to drive the camera (Extended-ISIS Camera, Photonic Science, Millham, UK) and the filter wheel, and to measure and plot online the fluorescence from 10 up to 40 rectangular “regions of interest” (ROI). Each ROI was identified by a number. Since the cell borders were not clearly identifiable, an ROI may not include the whole cell or may include part of an adjacent cell. Adjacent ROIs never superimposed. [Ca2+]i was monitored by measuring, for each ROI, the ratio of the mean fluorescence emitted at 510 nm when exciting alternately at 340 and 380 nm (shortly termed “ratio”). An increase in [Ca2+]i causes an increase in the ratio [26]. Ratio measurements were performed and plotted online every 3 s. The experiments were performed at room temperature (22 °C). Resting [Ca2+]i in the three cell types was monitored by using the Grynkiewicz method, as described in [35].

2.9. Statistics

All of the Ca2+ imaging data were collected from T cells deriving from three donors. Tumour-derived T cells, i.e., cPBTs and TILs, isolated from all 10 patients were used throughout the study. Therefore, cPBTs and TILs expanded from three distinct donors could be used in different experimental conditions. Likewise, the hPBTs isolated from three healthy donors were used for Ca2+ imaging experiments. Pooled data are given as mean ± SE, while the number of cells analysed is indicated between parentheses. Statistical significance (p < 0.05) was evaluated using the Student’s t-test for unpaired observations and the one-way ANOVA test followed by the post hoc Bonferroni test, as appropriate.

2.10. Chemicals

Fura-2/AM was obtained from Invitrogen (Life Technologies; cat# F1221). BTP-2 (cat# 203890) and myo-inositol 1,4,5-trisphosphate hexakis (butyryloxymethyl) (InsP3BM) (cat# 3-1-145) were obtained from Calbiochem (Merk Millipore). All other chemicals were of analytical grade and were obtained from Sigma-Aldrich: cyclopiazonic acid (CPA) (cat# C1530), ATP (cat# A2383), caffeine (cat# C0750), R59949 (cat# D5794), ritanserin (cat# R103), phorbol 12-myristate 13-acetate (PMA) (cat# 79346), Gö-6983 (cat# G1918), U73122 (Cat# U6756, 2-Aminoethyl diphenylborinate (2-APB) (cat# D9754) and ionomycin (cat# I0634).

3. Results

3.1. Characterisation of Ex Vivo-Expanded TILs

The cultures of the TILs obtained from the 10 patients enrolled in this study were evaluated after 14 days for surface antigens and potency, in order to confirm that the cells expanded from the tumour samples were T cells specifically directed against cancer cells. The great majority of the TILs were CD3+ cells (>96%), containing both CD3+/CD4+ and CD3+/CD8+ cells (mean 60%, SD 18% and mean 34%, SD 22%, respectively) (Figure 1A). Negligible percentages of CD3−/CD56+ (less than 5%) cells were documented (Figure 1A). The dot blots of two representative patients are shown in Figure S1. The potency of the TILs was evaluated against both the SW480 cells lines and autologous mCRC cells obtained from all 10 patients (Figure 1D). The TIL potency was evaluated in a 5-h cytotoxicity assay based on 51Cr release by the target cells. All TILs displayed high levels of cytotoxicity against SW480 cells (mean: 56% and SD 7% at effector:target (E:T) ratio of 50:1), which were maintained even at lower E:T ratios. The TILs were also able to efficiently kill autologous tumour cells as well (mean: 41% and SD 8% at E:T ratio of 50:1), despite being slightly lower compared to the immortalised cell line, which is also maintained even at lower E:T ratios. These data documented that the cytotoxic activity was not due to CD3−/CD56+ NK cells, which were almost completely absent from the expanded TILs, as shown in Figure 1A, confirming that the TILs were specifically directed against mCRC cells, both in the autologous and immortalised cell line (Figure 1B).

3.2. SOCE Is Larger in Ex Vivo-Expanded TILs

The remodelling of the Ca2+ handling machinery in TILs, compared to hPBTs and cPBTs, was evaluated in cells loaded with the Ca2+-sensitive fluorophore Fura-2/AM. The resting [Ca2+]i was significantly (p < 0.001) higher in the TILs than in the cPBTs and hPBTs (Figure S2). The “Ca2+ add-back” protocol was then exploited to simultaneously evaluate ER Ca2+ release and SOCE in the three cell types. As described elsewhere [23,26], the cells were challenged with CPA (30 µM), a selective inhibitor of Sarco-Endoplasmic Reticulum Ca2+-ATPase (SERCA) activity, in the absence of extracellular Ca2+ (0Ca2+) to induce ER Ca2+ release and thereby promote ER Ca2+ depletion. Extracellular Ca2+ was restored upon recovery to the baseline to selectively measure the SOCE. Careful analysis of the Ca2+ tracings (Figure 2A) revealed that the amplitude of CPA-induced ER Ca2+ release was as follows: cPBTs > TILs > hPBTs (Figure 2B). However, the CPA-evoked SOCE was significantly (p < 0.001) larger in the TILs than in cPBTs, which, in turn, exhibited a larger SOCE than hPBTs (Figure 2B). In keeping with these observations, when the T cells were challenged with CPA (30 µM) in the presence of extracellular Ca2+ (1.5 mM), the amplitude of the Ca2+ peak was as follows: TILs >> cPBTs > hPBTs (Figure S3). Pre-incubation with BTP-2 (20 µM), which selectively inhibits Orai1 in hPBTs [36] and CTLs [23], significantly (p < 0.001) reduced CPA-evoked SOCE in the TILs (Figure 2C,D). Therefore, Orai1 channels also mediate SOCE in these cells. These pieces of evidence indicate that ex vivo-expanded TILs undergo a remodelling of the Ca2+-handling machinery that leads to SOCE hyperactivation.

3.3. InsP3Rs- and RyRs-Mediated Ca2+ Release Result in Enhanced SOCE in Ex Vivo-Expanded TILs

Ca2+ release from the ER is mediated by InsP3Rs and ryanodine receptors (RyRs), which can both result in SOCE activation in hPBTs [16,17]. The InsP3-induced intracellular Ca2+ mobilisation was first evaluated by stimulating the cells with the InsP3-synthesising autacoid, ATP (100 µM), as shown in other types of cancer cells [26,38]. Preliminary experiments confirmed that the ATP-induced intracellular Ca2+ mobilisation was mediated by InsP3Rs in hPBTs (Figure S4). The ATP-induced ER Ca2+ release did not differ among the three different cell types (Figure 3A,B). However, SOCE amplitude in the TILs and cPBTs was similar and significantly (p < 0.001) higher than hPBTs (Figure 3A,B). When the T cells were challenged with ATP (100 µM) in the presence of extracellular Ca2+ (1.5 mM), the amplitude of the Ca2+ peak was as follows: TILs = cPBTs > hPBTs (Figure S5).
To circumvent the Gq/11 protein–PLCβ-InsP3-signalling pathway recruited downstream of purinergic P2Y receptors, InsP3Rs were directly activated with a membrane-permeant esterified form of InsP3, known as InsP3BM [38]. The two components (ER Ca2+ release and SOCE) of the Ca2+ response to InsP3BM (1 µM) displayed a similar trend as the one described for ATP: no statistically relevant difference in ER Ca2+ release among the three cell types, while SOCE amplitude was as follows: TILs > cPBTs > hPBTs (Figure 3C,D).
The RyRs-mediated Ca2+ signals were compared by stimulating the cells with caffeine (5 mM), a selective RyR agonist [39]. The caffeine-evoked ER Ca2+ release did not differ between the hPBTs and TILs, but it was significantly (p < 0.001) larger in cPBTs (Figure 3E,F). However, the caffeine-evoked SOCE in the cPBTs and TILs was still significantly (p < 0.001) larger than in the hPBTs (Figure 3E,F).
Overall, these findings demonstrate that SOCE amplitude is always enhanced in ex vivo-expanded TILs, whatever the stimulus inducing ER Ca2+ depletion (CPA, ATP, InsP3BM and caffeine), although the distinct ER Ca2+ sub-compartments (InsP3Rs vs. RyRs) underwent different patterns of remodelling in TILs vs. cPBTs (i.e., selective increase in RyRs-mediated ER Ca2+ release in cPBTs).

3.4. Molecular Characterisation of the Ca2+ Toolkit Reveals the Up-Regulation of STIM1 Protein in Ex Vivo-Expanded TILs

In order to gain further insights into the molecular mechanisms underlying the remodelling of the Ca2+-handling machinery in the TILs, we first performed a thorough qRT-PCR analysis of the mRNAs harvested from the three different cell types. Figure 4A shows that InsP3R1, RyR1, STIM1 and Orai1 were the predominant isoforms expressed in hPBTs, in agreement with previous observations [18,19,41,42,43,44]. Subsequent transcriptomic analysis, which is presented in Figure S6, revealed that InsP3R1, but not InsP3R2 and InsP3R3, was significantly (p < 0.001) downregulated in TILs compared to hPBTs and cPBTs. RyR1 was less expressed in cPBTs and TILs compared to hPBTs, while RyR2 was similarly expressed in the three cell types (Figure S6). Likewise, there were no significant changes in the expression levels of the transcripts encoding for SERCA2B (Figure S6), the major SERCA isoforms in T cells [17]. The comparison of ΔCt values of the mRNAs obtained through qRT-PCR did not reveal any significant difference in the expression levels of Orai1 and Orai3, as shown in Figure S6. Conversely, the levels of STIM1 mRNA were as follows: hPBTs > cPBTs > TILs (Figure S6). These transcriptomic data were somehow contrasting with the evidence reported above that the SOCE amplitude increases during the progression from hPBTs to cPBTs and then TILs. Therefore, we carried out a Western blot analysis of the relative changes in the expression of STIM1 and Orai1 proteins, which affect SOCE in hPBTs [18,19,41,43]. The immunoblots revealed a major band of ≈90 kDa for STIM1 (Figure 4B) and of ≈33 kDa for Orai1 (Figure 4B), as expected by their molecular weight [26,43]. The STIM1 protein was modestly, but significantly, up-regulated in TILs compared to hPBTs and cPBTs (Figure 4C). Conversely, there was no difference in the expression levels of the Orai1 protein (Figure 4C). These findings suggest that STIM1 up-regulation could enhance SOCE in TILs, but not in cPBTs, in which the protein expression of the ER Ca2+ sensor is not increased. Thus, an additional mechanism could regulate SOCE amplitude in cPBTs and, possibly, in ex vivo-expanded TILs.

3.5. Enhanced SOCE in TILs and cPBTs Is Maintained by Diacylglycerol Kinase (DGK) Activity

DGK limits diacylglycerol (DAG) signalling in hPBTs by converting DAG into phosphatidic acid. Interestingly, DGK function and/or expression are up-regulated in TILs, thereby attenuating T-cell cytotoxicity against cancer cells [45,46]. Moreover, SOCE can be inhibited by protein kinase C (PKC) [47,48], which is a downstream target of DAG [45]. Therefore, the pharmacological blockade of DGK activity is predicted to reinstate PKC-mediated inhibition and reduce the SOCE amplitude in neoplastic T cells. Figure 5A shows that CPA-evoked SOCE is not altered by two established DGK inhibitors, i.e., ritanserin (40 µM) and DGKiII (5 µM; also known as R59949) [45,46], in hPBTs. However, SOCE was significantly (p < 0.001) increased by inhibiting the basal PKC activity with Gö-6983 (10 µM) (Figure 5A), whereas it was significantly (p < 0.001) reduced by directly activating PKC with PMA (10 µM) (Figure 5A). The statistical analysis of these data is reported in Figure 5B. Overall, these preliminary findings revealed that PKC is able to inhibit SOCE in hPBTs, but that its activity is unlikely to be limited by basal DGK activity. Conversely, ritanserin (40 µM) and R59949 (5 µM) dramatically reduced CPA-evoked SOCE in both cPBTs (Figure 5C) and TILs (Figure 5E). Interestingly, Gö-6983 (10 µM) did not remarkably affect SOCE either in cPBTs (Figure 5C) or in TILs (Figure 5E), whereas PMA (10 µM) exerted a significant (p < 0.001) inhibitory effect in cPBTs (Figure 5C) and in TILs (Figure 5E). The statistical analysis of these data is reported in Figure 5D,F. Overall, these data strongly suggest that SOCE amplitude in TILs and cPBTs is larger than in hPBTs due to the DGK-positive input that dampens the basal inhibition operated by PKC.

3.6. TCR Stimulation Induced a Larger Increase in [Ca2+]i in TILs

In order to assess the impact of the increase in SOCE on TCR stimulation, the different types of T cells were challenged with soluble antibodies direct against CD3, which transduces antigen recognition by the variable component of TCR into an increase in [Ca2+]i [21,41,49]. One µg/mL anti-CD3 monoclonal antibody (mAB) triggered a stronger oscillatory Ca2+ response in TILs than in cPBTs and hPBTs (Figure 6A–C). Although there was no significant difference in the percentage of oscillating cells (Figure 6D), the area under the curve (AUC) corresponding to the intracellular Ca2+ signals evoked by TCR stimulation (Figure 6E) and the oscillation frequency (Figure 6F) were significantly larger in the TILs than in the hPBTs and cPBTs. The pharmacological blockade of Orai1 with BTP-2 (20 µM) dampened the spiking Ca2+ response (Figure 6G) and significantly (p < 0.001) hindered the AUC of the Ca2+ signal (Figure 6H) evoked in TILs by 1 µg/mL anti-CD3 antibody.
The “Ca2+ add-back” was then exploited to further assess the role of SOCE in the stronger Ca2+ response to anti-CD3 stimulation in ex vivo-expanded TILs. Statistical analysis of the Ca2+ tracings (Figure 7A) revealed that the anti-CD3 antibody evoked a similar endogenous Ca2+ response in the TILs to the hPBTs, while the intracellular Ca2+ mobilisation was surprisingly smaller in the cPBTs (Figure 7B). Conversely, SOCE was remarkably larger in the TILs than in the hPBTs and cPBTs (Figure 7B). Overall, these findings demonstrate that a larger SOCE results in a stronger oscillatory Ca2+ response to anti-CD3 stimulation in ex vivo-expanded TILs.

3.7. SOCE Tunes the Amplitude of the Ca2+ Response during Target Cell Killing

TCR is physiologically stimulated by the formation of the immune synapse between cPBTs or TILs with the target cancer cell to induce an increase in [Ca2+]i that supports their cytotoxic activity [21,31,50,51]. T cells were adhered to glass substrates, loaded with Fura-2/AM and mCRC cancer cells were then dropped onto them. The preliminary experiments conducted with the SW480 mCRC cell line revealed that 1 × 104 cells/mL was the most suitable cell density to evoke robust intracellular Ca2+ oscillations in the TILs (Figure S7). When the adherent cells were challenged with autologous (i.e., deriving from the same patient) mCRC cells, we observed three main intracellular Ca2+ signatures: (1) 46.08% (106/230) of responding cells presented intracellular Ca2+ oscillations that persist for about 40 min from the beginning of the stimulation (termed “oscillatory pattern”) (Figure 8A); (2) 34.7% (80/230) of the responding cells displayed two slow, intracellular Ca2+ waves that again lasted for about 40 min (termed “slow pattern”) (Figure 8B); and (3) 19.1% (44/230) of responding cells exhibited a fast Ca2+ transient followed by a slow increase in [Ca2+]i that ultimately achieved a plateau phase (therefore termed “plateau pattern”) (Figure 8C), which declined to resting Ca2+ levels upon washout of the mCRC cells. When the same experiment was repeated in the same day using the same batches of TILs and mCRC cells, we observed that the pharmacological blockade of SOCE with BTP-2 (20 µM, 10 min) induced all of the TILs to display the “oscillatory pattern” (141/141 responding cells; 100%) (Figure 8D), thereby abolishing the “slow” and “plateau” Ca2+ signatures. The AUC corresponding to the intracellular Ca2+ signals evoked by TCR stimulation confirmed that BTP-2 inhibited the amount of Ca2+ liberated during the immune response (Figure 8E), whereas it increased oscillation frequency (Figure 8F), which reflects the disappearance of the “slow” and “plateau” Ca2+ waveforms. In a parallel set of experiments, the cytotoxic activity was measured in the absence (Ctrl) and presence of BTP-2 (20 µM) using the same batches of cells that were employed for the Ca2+ imaging recordings. Surprisingly, the average cytotoxic activity against autologous mCRC cells at each E:T ratio was increased by pre-treating the ex vivo-expanded TILs with BTP-2 (20 µM) (Figure 8G), with the increase becoming statistically significant (p < 0.05) at low E:T ratios (Figure 8H). Overall, these findings demonstrate that reducing Ca2+ influx with BTP-2 during target cell killing significantly increases the cytotoxic activity at a low TIL density, i.e., when TILs-mediated cancer cell death is rarer.

4. Discussion

SOCE is a crucial signalling pathway to stimulate T cells against cancer cells [22,23,52]. Therefore, unravelling whether the SOCE machinery is deranged and/or whether it is uncoupled from the downstream Ca2+-dependent cytotoxic response against cancer cells is of paramount importance to help improve the therapeutic outcome of ACT via autologous TILs. Herein, we demonstrated, for the first time, that SOCE is up-regulated in TILs and cPBTs deriving from mCRC patients compared to hPBTs, mainly due to tonic DGK-dependent modulation. Surprisingly, the increase in SOCE amplitude is associated with a reduction in TILs’ cytotoxic activity against autologous mCRC cells compared to that of CTLs. This unexpected finding could explain their limited therapeutic efficacy in the patients [53].
Neoplastic transformation is associated with a dramatic remodelling of the Ca2+-handling machinery [24,25], which does not only involve tumour cells, but also other cellular constituents of the tumour microenvironment [54]. Notably, SOCE is strongly downregulated in fresh TILs from a number of tumours due to the inhibitory effect of programmed death-1 (PD-1) [31,32], but SOCE amplitude quickly recovers after tumour dissociation [31]. Nevertheless, no information is currently available regarding the molecular remodelling and the role of SOCE in TILs that have been expanded in culture to be reinoculated within the patient. This gap in the knowledge is quite surprising, as intracellular Ca2+ dynamics, particularly SOCE activation, are crucial for CTLs to mount the immune response against cancer cells upon T antigen presentation by the major histocompatibility complex [22,23]. In accord, STIM1- and Orai1-deficient subjects displayed a significant propensity to develop human herpesvirus-8-dependent Kaposi sarcoma [55] and Epstein–Barr virus-associated B-cell lymphoma [56]. Herein, we adopted a protocol that involves restoring Ca2+ to the extracellular solution after depletion of the ER Ca2+ store with the specific SERCA inhibitor, CPA, to separately evaluate ER Ca2+ release and SOCE activation [26,38]. By doing so, we found that SOCE was larger in TILs than in hPBTs and cPBTs, which is the most likely explanation for their larger resting [Ca2+]i [17]. Enhanced SOCE could reflect larger ER Ca2+ depletion and/or enhanced STIM/Orai protein expression. However, the ER Ca2+-releasing ability of TILs, as monitored by CPA-evoked intracellular Ca2+ mobilisation, was not increased compared to cPBTs and hPBTs. In T cells, SOCE can be elicited by ER Ca2+ depletion following intraluminal Ca2+ efflux through either InsP3Rs or RyRs [16,17]. However, ER Ca2+ mobilisation via InsP3Rs (physiologically activated with ATP or directly engaged with InsP3-BM) and RyRs (pharmacologically stimulated with caffeine) was never enhanced in TILs, although SOCE was always more up-regulated compared to hPBTs. The lack of a clear relationship between the extent of the ER Ca2+ mobilisation and SOCE activation reported in the present investigation is consistent with the long-standing notion that SOCE is not tightly coupled to store emptying [57] and that discrete ER subdomains can effectively stimulate SOCE upon direct stimulation of InsP3Rs and RyRs in hPBTs [41]. Overall, these preliminary findings demonstrated that in in vitro-expanded TILs: (1) SOCE is operative, (2) can be engaged by InsP3Rs- and RyRs-mediated depletion of the ER Ca2+ store, and (3) is up-regulated compared to hPBTs. Therefore, it would be tempting to conclude that the molecular mechanism responsible for SOCE up-regulation in TILs is the same as in cPBTs, a fraction of which is recruited to liver metastases and is likely to represent the main cellular population expanded in vitro. This hypothesis was supported by the notion that the SOCE, activated upon the physiological depletion of the ER Ca2+ pool via InsP3Rs and RyRs, is also up-regulated in cPBTs compared to hPBTs.
Surprisingly, the molecular analysis of STIM and Orai expression by qRT-PCR and immunoblotting revealed that STIM1 protein is only increased in TILs, and not in cPBTs, compared to hPBTs. Furthermore, there was no difference in the protein levels of Orai1 among the three cell types. Notably, there was a discrepancy between the reduction in STIM1 transcripts and the increase in the STIM1 proteins observed in TILs compared to hPBTs. Similarly, recent work carried out on patient-derived mCRC cells showed that the mRNA encoding for STIM1 is equally expressed in primary and metastatic CRC cells, whereas the STIM1 protein was remarkably up-regulated in the latter [26]. Moreover, the Villalobos group reported that the Orai1 protein is significantly up-regulated in the HT29 CRC cell line compared to non-neoplastic cells, although there is no difference in its transcript expression levels between the two cell types [37]. The same study also clearly demonstrated that, although STIM2 is highly expressed at the transcript level, it is almost absent at the protein level in HT29 cells [37]. The protein expression of the multiple components of the Ca2+-handling machinery could be finely regulated by controlling mRNA stability [58]. For instance, SERCA2A mRNA presents a longer half-life in left ventricular myocytes with respect to stomach smooth muscle, thereby causing a 100-fold increase in SERCA2A protein expression in the former cells [59]. An alternative, but not mutually exclusive, hypothesis involves a higher rate of STIM1 protein production in TILs compared to hPBTs (and cPBTs) [60].
These findings suggested that SOCE up-regulation could also reflect differences in the regulatory mechanisms between cPBTs (and, possibly, TILs) and hPBTs. It has been recognised that enhanced DGK expression/activity in TILs attenuates tumour dismantling by limiting DAG-dependent pathways in favour of Ca2+-mediated signalling [45,46]. DGK phosphorylates DAG into phosphatidic acid, thereby interfering with the recruitment of DAG-dependent effectors, including PKC [45,46]. Notably, PKC may inhibit SOCE by phosphorylating Orai1 at NH2-terminal Ser-27 and Ser-30 residues [47,48]. Thus, we reasoned that the pharmacological blockade of DGK could selectively reduce SOCE by rescuing the PKC-mediated inhibition of Orai1 in TILs and cPBTs. Ten DGK isoforms have been detected and categorised into five subtypes in mammals: I (α, β, γ), II (δ, η, κ), III (ε), V (ζ and ι) and V (θ) [47,48]. Of these, type I DGKα may inhibit T-cell proliferation and function in the tumour microenvironment [45,61]. In agreement with our working hypothesis, R59949, which inhibits type I DGKs, and ritanserin, a selective blocker of DGKα, dramatically reduced CPA-evoked SOCE in both in vitro-expanded TILs and cPBTs, but not hPBTs. Conversely, blocking basal PKC activity with Gö-6983 strongly enhanced SOCE in hPBTs, but not in CTLs and TILs. These findings indicate that PKC exerts a weak inhibition of SOCE in TILs and cPBTs and that this is likely to reflect an increase in DGKα expression/activity that reduces DAG levels and DAG-dependent signalling. In agreement with this hypothesis, the direct stimulation of PKC with PMA decreased SOCE amplitude in all cell types. This finding shows that PKC can still inhibit SOCE in TILs and cPBTs, but the basal production of its physiological ligand, i.e., DAG, is maintained under the threshold to effectively reduce the SOCE amplitude. Altogether, our results strongly suggest that DGKα enables SOCE to be hyperactivated in TILs and cPBTs by preventing DAG-dependent PKC-mediated inhibition. Similarly, the PKC-dependent modulation of Orai1 activity has been reported in invasive melanoma cells [47] and in rat basophilic leukaemia cells [62].
SOCE supports the increase in [Ca2+]i that arises in both CTLs and TILs following TCR stimulation at the immune synapse to support cancer cell killing [21,31,50,51]. Preliminary experiments revealed that the increase in SOCE led to a stronger oscillatory Ca2+ signal in TILs than in cPBTs, following stimulation with an anti-CD3 antibody, which is routinely employed to assess the strength of the specific T-cell response to TCR engagement [17,31,49]. However, when ex vivo-expanded TILs were stimulated with autologous mCRC cells, they displayed multiple Ca2+ signatures, the most frequent of which (i.e., “slow pattern” and “plateau pattern”, occurring in 53.8% of the cells) present a sustained increase in [Ca2+]i. The pharmacological blockade of SOCE with BTP-2 significantly increased the percentage of TILs displaying intracellular Ca2+ oscillations (from 46.08% to 100%) and significantly reduced the amount of total Ca2+ introduced into the cytosol during the immune response. Notably, pre-treating the TILs with BTP-2 improved their cytotoxic activity against the mCRC cells at each E:T ratio, and this increase became statistically relevant at a lower E:T ratio, suggesting that this effect was more evident when the TILs-mediated lysis was low. This feature is quite relevant, since, in non-haematological solid cancers, TILs often fail to kill target cells during 1:1 conjugation [63]. Our results are consistent with the recent report that a reduction in SOCE enhances CTL and natural killer cell (NK) cytotoxicity against cancer cells [21]. In accord, this and other studies showed that exaggerated Ca2+ entry in CTLs or NK can interfere with multiple steps of the immune response, e.g., migration and release of lytic granules, while it can enhance proliferation and apoptosis [21,52]. The mechanism(s) whereby exaggerated Ca2+ entry in TILs during mCRC cell killing reduces cytotoxic activity, e.g., increased apoptotic rate, is (are) under current investigation.

5. Conclusions

We provide the first evidence that SOCE is up-regulated in ex vivo-expanded TILs from mCRC patients. Extracellular Ca2+ entry is mainly enhanced by DGK activity, which prevents SOCE inhibition by PKC. The pharmacological blockade of SOCE with BTP-2 enhances TILs’ ability to kill autologous mCRC cells at low-density ratios. This preliminary evidence might suggest an alternative pharmacological strategy to improve the therapeutic outcome of ACT in mCRC patients.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers14143312/s1, Figure S1: Resting [Ca2+]i is higher in TILs compared to hPBTs and cPBTs; Figure S2: Evidence that InsP3Rs mediate ATP-evoked intracellular Ca2+ release in hPBTs; Figure S3: Differential expression of the main component of the Ca2+-handling machinery in hPBTs, cPBTs and TILs. Figure S4: Intracellular Ca2+ oscillations elicited by SW480 cells in TILs. Figure S5: The Ca2+ response to ATP is larger in TILs than in as compared to hPBTs and cPBTs. Figure S6: Differential expression of the main component of the Ca2+-handling machinery in hPBTs, cPBTs, and TILs. Figure S7: Intracellular Ca2+ oscillations elicited by SW480 cells in TILs. Figure S8: Whole Western blot showing all bands depicted in Figure 4B. Table S1: Clinical characteristics of the patients. Table S2: List of oligonucleotide primers used for real-time PCR. Table S3: List of primary antibodies used to characterise the SOCE machinery.

Author Contributions

Investigation, formal analysis, and data curation: P.F., A.R. and L.T.; investigation: M.T., A.M., F.C. and S.N.; supervision: K.L., P.P. and D.L.; writing—review and editing: K.L., P.P. and D.L.; conceptualization, data curation, writing—original draft preparation, project administration and funding acquisition: D.M. and F.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Italian Ministry of Health Grants RF-2010-2316319 (D.M.) and Program “Ricerca Corrente” of the IRCCS Foundation Policlinico San Matteo: RC/08059815B (D.M.); Italian Ministry of Education, University and Research (MIUR): Dipartimenti di Eccellenza Program (2018–2022)—Dept. of Biology and Biotechnology “L. Spallanzani”, University of Pavia (F.M.); Fondo Ricerca Giovani from the University of Pavia (F.M.).

Institutional Review Board Statement

This study was approved by the Institutional Review Board of the Foundation IRCCS Policlinico San Matteo (Protocol Number 20190069408). Peripheral blood cells were derived from mCRC patients at the time of surgery or from buffy coats of healthy donors. Buffy-coat units were released by the Blood Bank of SIMT, Foundation IRCCS Policlinico San Matteo, Pavia, according to Article 8 of the Legislative Decree 2, November 2015, of the Italian Department of Health with blood donor-specific informed consent implemented by the Blood Bank Conference of Regione Lombardia.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Data supporting the reported results can be obtained upon reasonable request to the authors.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Newhook, T.E.; Vauthey, J.N. Colorectal liver metastases: State-of-the-art management and surgical approaches. Langenbecks Arch. Surg. 2022, 1–14. [Google Scholar] [CrossRef] [PubMed]
  2. Pedrosa, L.; Esposito, F.; Thomson, T.M.; Maurel, J. The Tumor Microenvironment in Colorectal Cancer Therapy. Cancers 2019, 11, 1172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Yang, J.C.; Rosenberg, S.A. Adoptive T-Cell Therapy for Cancer. Adv. Immunol. 2016, 130, 279–294. [Google Scholar] [CrossRef] [PubMed]
  4. Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef] [PubMed]
  5. Rosenberg, S.A.; Yang, J.C.; Sherry, R.M.; Kammula, U.S.; Hughes, M.S.; Phan, G.Q.; Citrin, D.E.; Restifo, N.P.; Robbins, P.F.; Wunderlich, J.R.; et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin. Cancer. Res. 2011, 17, 4550–4557. [Google Scholar] [CrossRef] [Green Version]
  6. Rosenberg, S.A.; Yannelli, J.R.; Yang, J.C.; Topalian, S.L.; Schwartzentruber, D.J.; Weber, J.S.; Parkinson, D.R.; Seipp, C.A.; Einhorn, J.H.; White, D.E. Treatment of patients with metastatic melanoma with autologous tumor-infiltrating lymphocytes and interleukin 2. J. Natl. Cancer Inst. 1994, 86, 1159–1166. [Google Scholar] [CrossRef]
  7. Stevanovic, S.; Helman, S.R.; Wunderlich, J.R.; Langhan, M.M.; Doran, S.L.; Kwong, M.L.M.; Somerville, R.P.T.; Klebanoff, C.A.; Kammula, U.S.; Sherry, R.M.; et al. A Phase II Study of Tumor-infiltrating Lymphocyte Therapy for Human Papillomavirus-associated Epithelial Cancers. Clin. Cancer. Res. 2019, 25, 1486–1493. [Google Scholar] [CrossRef]
  8. Montagna, D.; Turin, I.; Schiavo, R.; Montini, E.; Zaffaroni, N.; Villa, R.; Secondino, S.; Schiavetto, I.; Caliogna, L.; Locatelli, F.; et al. Feasibility and safety of adoptive immunotherapy with ex vivo-generated autologous, cytotoxic T lymphocytes in patients with solid tumor. Cytotherapy 2012, 14, 80–90. [Google Scholar] [CrossRef]
  9. Turin, I.; Pedrazzoli, P.; Tullio, C.; Montini, E.; La Grotteria, M.C.; Schiavo, R.; Perotti, C.; Locatelli, F.; Carretto, E.; Maccario, R.; et al. GMP production of anti-tumor cytotoxic T-cell lines for adoptive T-cell therapy in patients with solid neoplasia. Cytotherapy 2007, 9, 499–507. [Google Scholar] [CrossRef]
  10. Yee, C.; Thompson, J.A.; Byrd, D.; Riddell, S.R.; Roche, P.; Celis, E.; Greenberg, P.D. Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: In vivo persistence, migration, and antitumor effect of transferred T cells. Proc. Natl. Acad. Sci. USA 2002, 99, 16168–16173. [Google Scholar] [CrossRef] [Green Version]
  11. Mackensen, A.; Meidenbauer, N.; Vogl, S.; Laumer, M.; Berger, J.; Andreesen, R. Phase I study of adoptive T-cell therapy using antigen-specific CD8+ T cells for the treatment of patients with metastatic melanoma. J. Clin. Oncol. 2006, 24, 5060–5069. [Google Scholar] [CrossRef] [Green Version]
  12. Wang, H.; Liu, P.; Mi, J.; Chen, J.; Liu, Y. A Preliminary Study of Adoptive T-cell Transfer Therapy for Patients With Non-Small-cell Lung Adenocarcinoma With Brain Metastasis: A Case Report of 3 Patients. Clin. Lung Cancer 2020, 21, e270–e273. [Google Scholar] [CrossRef]
  13. Turcotte, S.; Gros, A.; Tran, E.; Lee, C.C.; Wunderlich, J.R.; Robbins, P.F.; Rosenberg, S.A. Tumor-reactive CD8+ T cells in metastatic gastrointestinal cancer refractory to chemotherapy. Clin. Cancer. Res. 2014, 20, 331–343. [Google Scholar] [CrossRef] [Green Version]
  14. Kroemer, M.; Turco, C.; Spehner, L.; Viot, J.; Idirene, I.; Bouard, A.; Renaude, E.; Deschamps, M.; Godet, Y.; Adotevi, O.; et al. Investigation of the prognostic value of CD4 T cell subsets expanded from tumor-infiltrating lymphocytes of colorectal cancer liver metastases. J. Immunother. Cancer 2020, 8. [Google Scholar] [CrossRef]
  15. Mandriani, B.; Pelle, E.; Pezzicoli, G.; Strosberg, J.; Abate-Daga, D.; Guarini, A.; Cives, M.; Porta, C. Adoptive T-cell immunotherapy in digestive tract malignancies: Current challenges and future perspectives. Cancer Treat. Rev. 2021, 100, 102288. [Google Scholar] [CrossRef]
  16. Vaeth, M.; Kahlfuss, S.; Feske, S. CRAC Channels and Calcium Signaling in T Cell-Mediated Immunity. Trends Immunol. 2020, 41, 878–901. [Google Scholar] [CrossRef]
  17. Trebak, M.; Kinet, J.P. Calcium signalling in T cells. Nat. Rev. Immunol. 2019, 19, 154–169. [Google Scholar] [CrossRef]
  18. Feske, S.; Gwack, Y.; Prakriya, M.; Srikanth, S.; Puppel, S.H.; Tanasa, B.; Hogan, P.G.; Lewis, R.S.; Daly, M.; Rao, A. A mutation in Orai1 causes immune deficiency by abrogating CRAC channel function. Nature 2006, 441, 179–185. [Google Scholar] [CrossRef]
  19. Oh-Hora, M.; Yamashita, M.; Hogan, P.G.; Sharma, S.; Lamperti, E.; Chung, W.; Prakriya, M.; Feske, S.; Rao, A. Dual functions for the endoplasmic reticulum calcium sensors STIM1 and STIM2 in T cell activation and tolerance. Nat. Immunol. 2008, 9, 432–443. [Google Scholar] [CrossRef] [Green Version]
  20. Feske, S.; Skolnik, E.Y.; Prakriya, M. Ion channels and transporters in lymphocyte function and immunity. Nat. Rev. Immunol. 2012, 12, 532–547. [Google Scholar] [CrossRef] [Green Version]
  21. Zhou, X.; Friedmann, K.S.; Lyrmann, H.; Zhou, Y.; Schoppmeyer, R.; Knorck, A.; Mang, S.; Hoxha, C.; Angenendt, A.; Backes, C.S.; et al. A calcium optimum for cytotoxic T lymphocyte and natural killer cell cytotoxicity. J. Physiol. 2018, 596, 2681–2698. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Maul-Pavicic, A.; Chiang, S.C.; Rensing-Ehl, A.; Jessen, B.; Fauriat, C.; Wood, S.M.; Sjoqvist, S.; Hufnagel, M.; Schulze, I.; Bass, T.; et al. ORAI1-mediated calcium influx is required for human cytotoxic lymphocyte degranulation and target cell lysis. Proc. Natl. Acad. Sci. USA 2011, 108, 3324–3329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Weidinger, C.; Shaw, P.J.; Feske, S. STIM1 and STIM2-mediated Ca2+ influx regulates antitumour immunity by CD8+ T cells. EMBO Mol. Med. 2013, 5, 1311–1321. [Google Scholar] [CrossRef] [PubMed]
  24. Moccia, F.; Zuccolo, E.; Poletto, V.; Turin, I.; Guerra, G.; Pedrazzoli, P.; Rosti, V.; Porta, C.; Montagna, D. Targeting Stim and Orai Proteins as an Alternative Approach in Anticancer Therapy. Curr. Med. Chem. 2016, 23, 3450–3480. [Google Scholar] [CrossRef]
  25. Chalmers, S.B.; Monteith, G.R. ORAI channels and cancer. Cell Calcium 2018, 74, 160–167. [Google Scholar] [CrossRef] [Green Version]
  26. Zuccolo, E.; Laforenza, U.; Ferulli, F.; Pellavio, G.; Scarpellino, G.; Tanzi, M.; Turin, I.; Faris, P.; Lucariello, A.; Maestri, M.; et al. Stim and Orai mediate constitutive Ca2+ entry and control endoplasmic reticulum Ca2+ refilling in primary cultures of colorectal carcinoma cells. Oncotarget 2018, 9, 31098–31119. [Google Scholar] [CrossRef] [Green Version]
  27. Jardin, I.; Lopez, J.J.; Salido, G.M.; Rosado, J.A. Store-Operated Ca2+ Entry in Breast Cancer Cells: Remodeling and Functional Role. Int. J. Mol. Sci. 2018, 19, 4053. [Google Scholar] [CrossRef] [Green Version]
  28. Jardin, I.; Rosado, J.A. STIM and calcium channel complexes in cancer. Biochim. Biophys. Acta 2016, 1863, 1418–1426. [Google Scholar] [CrossRef]
  29. Shapovalov, G.; Gordienko, D.; Prevarskaya, N. Store operated calcium channels in cancer progression. Int. Rev. Cell Mol. Biol. 2021, 363, 123–168. [Google Scholar] [CrossRef]
  30. Dragoni, S.; Turin, I.; Laforenza, U.; Potenza, D.M.; Bottino, C.; Glasnov, T.N.; Prestia, M.; Ferulli, F.; Saitta, A.; Mosca, A.; et al. Store-operated Ca2+ entry does not control proliferation in primary cultures of human metastatic renal cellular carcinoma. BioMed Res. Int. 2014, 2014, 739494. [Google Scholar] [CrossRef] [Green Version]
  31. Wang, S.F.; Fouquet, S.; Chapon, M.; Salmon, H.; Regnier, F.; Labroquere, K.; Badoual, C.; Damotte, D.; Validire, P.; Maubec, E.; et al. Early T cell signalling is reversibly altered in PD-1+ T lymphocytes infiltrating human tumors. PLoS ONE 2011, 6, e17621. [Google Scholar] [CrossRef] [PubMed]
  32. Chimote, A.A.; Hajdu, P.; Sfyris, A.M.; Gleich, B.N.; Wise-Draper, T.; Casper, K.A.; Conforti, L. Kv1.3 Channels Mark Functionally Competent CD8+ Tumor-Infiltrating Lymphocytes in Head and Neck Cancer. Cancer Res. 2017, 77, 53–61. [Google Scholar] [CrossRef] [Green Version]
  33. Daudt, L.; Maccario, R.; Locatelli, F.; Turin, I.; Silla, L.; Montini, E.; Percivalle, E.; Giugliani, R.; Avanzini, M.A.; Moretta, A.; et al. Interleukin-15 favors the expansion of central memory CD8+ T cells in ex vivo generated, antileukemia human cytotoxic T lymphocyte lines. J. Immunother. 2008, 31, 385–393. [Google Scholar] [CrossRef]
  34. Poletto, V.; Dragoni, S.; Lim, D.; Biggiogera, M.; Aronica, A.; Cinelli, M.; De Luca, A.; Rosti, V.; Porta, C.; Guerra, G.; et al. Endoplasmic Reticulum Ca2+ Handling and Apoptotic Resistance in Tumor-Derived Endothelial Colony Forming Cells. J. Cell. Biochem. 2016, 117, 2260–2271. [Google Scholar] [CrossRef]
  35. Astesana, V.; Faris, P.; Ferrari, B.; Siciliani, S.; Lim, D.; Biggiogera, M.; De Pascali, S.A.; Fanizzi, F.P.; Roda, E.; Moccia, F.; et al. [Pt(O,O′-acac)(gamma-acac)(DMS)]: Alternative Strategies to Overcome Cisplatin-Induced Side Effects and Resistance in T98G Glioma Cells. Cell. Mol. Neurobiol. 2020, 41, 563–587. [Google Scholar] [CrossRef]
  36. Zitt, C.; Strauss, B.; Schwarz, E.C.; Spaeth, N.; Rast, G.; Hatzelmann, A.; Hoth, M. Potent inhibition of Ca2+ release-activated Ca2+ channels and T-lymphocyte activation by the pyrazole derivative BTP2. J. Biol. Chem. 2004, 279, 12427–12437. [Google Scholar] [CrossRef] [Green Version]
  37. Sobradillo, D.; Hernandez-Morales, M.; Ubierna, D.; Moyer, M.P.; Nunez, L.; Villalobos, C. A reciprocal shift in transient receptor potential channel 1 (TRPC1) and stromal interaction molecule 2 (STIM2) contributes to Ca2+ remodeling and cancer hallmarks in colorectal carcinoma cells. J. Biol. Chem. 2014, 289, 28765–28782. [Google Scholar] [CrossRef] [Green Version]
  38. Pierro, C.; Cook, S.J.; Foets, T.C.; Bootman, M.D.; Roderick, H.L. Oncogenic K-Ras suppresses IP3-dependent Ca2+ release through remodelling of the isoform composition of IP3Rs and ER luminal Ca2+ levels in colorectal cancer cell lines. J. Cell Sci. 2014, 127, 1607–1619. [Google Scholar] [CrossRef] [Green Version]
  39. Thakur, P.; Dadsetan, S.; Fomina, A.F. Bidirectional coupling between ryanodine receptors and Ca2+ release-activated Ca2+ (CRAC) channel machinery sustains store-operated Ca2+ entry in human T lymphocytes. J. Biol. Chem. 2012, 287, 37233–37244. [Google Scholar] [CrossRef] [Green Version]
  40. Rusko, J.; Wang, X.; van Breemen, C. Regenerative caffeine-induced responses in native rabbit aortic endothelial cells. Br. J. Pharmacol. 1995, 115, 811–821. [Google Scholar] [CrossRef] [Green Version]
  41. Diercks, B.P.; Werner, R.; Weidemuller, P.; Czarniak, F.; Hernandez, L.; Lehmann, C.; Rosche, A.; Kruger, A.; Kaufmann, U.; Vaeth, M.; et al. ORAI1, STIM1/2, and RYR1 shape subsecond Ca2+ microdomains upon T cell activation. Sci. Signal. 2018, 11, eaat0358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. McCarl, C.A.; Picard, C.; Khalil, S.; Kawasaki, T.; Rother, J.; Papolos, A.; Kutok, J.; Hivroz, C.; Ledeist, F.; Plogmann, K.; et al. ORAI1 deficiency and lack of store-operated Ca2+ entry cause immunodeficiency, myopathy, and ectodermal dysplasia. J. Allergy Clin. Immunol. 2009, 124, 1311–1318.e1317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Chou, J.; Badran, Y.R.; Yee, C.S.K.; Bainter, W.; Ohsumi, T.K.; Al-Hammadi, S.; Pai, S.Y.; Feske, S.; Geha, R.S. A novel mutation in ORAI1 presenting with combined immunodeficiency and residual T-cell function. J. Allergy Clin. Immunol. 2015, 136, 479–482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Jayaraman, T.; Ondriasova, E.; Ondrias, K.; Harnick, D.J.; Marks, A.R. The inositol 1,4,5-trisphosphate receptor is essential for T-cell receptor signaling. Proc. Natl. Acad. Sci. USA 1995, 92, 6007–6011. [Google Scholar] [CrossRef] [Green Version]
  45. Arranz-Nicolas, J.; Merida, I. Biological regulation of diacylglycerol kinases in normal and neoplastic tissues: New opportunities for cancer immunotherapy. Adv. Biol. Regul. 2020, 75, 100663. [Google Scholar] [CrossRef]
  46. Sim, J.A.; Kim, J.; Yang, D. Beyond Lipid Signaling: Pleiotropic Effects of Diacylglycerol Kinases in Cellular Signaling. Int. J. Mol. Sci. 2020, 21, 6861. [Google Scholar] [CrossRef]
  47. Hooper, R.; Zhang, X.; Webster, M.; Go, C.; Kedra, J.; Marchbank, K.; Gill, D.L.; Weeraratna, A.T.; Trebak, M.; Soboloff, J. Novel Protein Kinase C-Mediated Control of Orai1 Function in Invasive Melanoma. Mol. Cell. Biol. 2015, 35, 2790–2798. [Google Scholar] [CrossRef] [Green Version]
  48. Kawasaki, T.; Ueyama, T.; Lange, I.; Feske, S.; Saito, N. Protein kinase C-induced phosphorylation of Orai1 regulates the intracellular Ca2+ level via the store-operated Ca2+ channel. J. Biol. Chem. 2010, 285, 25720–25730. [Google Scholar] [CrossRef] [Green Version]
  49. Hess, S.D.; Oortgiesen, M.; Cahalan, M.D. Calcium oscillations in human T and natural killer cells depend upon membrane potential and calcium influx. J. Immunol. 1993, 150, 2620–2633. [Google Scholar]
  50. Pores-Fernando, A.T.; Zweifach, A. Calcium influx and signaling in cytotoxic T-lymphocyte lytic granule exocytosis. Immunol. Rev. 2009, 231, 160–173. [Google Scholar] [CrossRef]
  51. Lyubchenko, T.A.; Wurth, G.A.; Zweifach, A. Role of calcium influx in cytotoxic T lymphocyte lytic granule exocytosis during target cell killing. Immunity 2001, 15, 847–859. [Google Scholar] [CrossRef] [Green Version]
  52. Kaschek, L.; Zophel, S.; Knorck, A.; Hoth, M. A calcium optimum for cytotoxic T lymphocyte and natural killer cell cytotoxicity. Semin. Cell Dev. Biol. 2021, 115, 10–18. [Google Scholar] [CrossRef]
  53. Gardini, A.; Ercolani, G.; Riccobon, A.; Ravaioli, M.; Ridolfi, L.; Flamini, E.; Ridolfi, R.; Grazi, G.L.; Cavallari, A.; Amadori, D. Adjuvant, adoptive immunotherapy with tumor infiltrating lymphocytes plus interleukin-2 after radical hepatic resection for colorectal liver metastases: 5-year analysis. J. Surg. Oncol. 2004, 87, 46–52. [Google Scholar] [CrossRef]
  54. Moccia, F. Endothelial Ca2+ Signaling and the Resistance to Anticancer Treatments: Partners in Crime. Int. J. Mol. Sci. 2018, 19, 217. [Google Scholar] [CrossRef] [Green Version]
  55. Byun, M.; Abhyankar, A.; Lelarge, V.; Plancoulaine, S.; Palanduz, A.; Telhan, L.; Boisson, B.; Picard, C.; Dewell, S.; Zhao, C.; et al. Whole-exome sequencing-based discovery of STIM1 deficiency in a child with fatal classic Kaposi sarcoma. J. Exp. Med. 2010, 207, 2307–2312. [Google Scholar] [CrossRef] [Green Version]
  56. Fuchs, S.; Rensing-Ehl, A.; Speckmann, C.; Bengsch, B.; Schmitt-Graeff, A.; Bondzio, I.; Maul-Pavicic, A.; Bass, T.; Vraetz, T.; Strahm, B.; et al. Antiviral and regulatory T cell immunity in a patient with stromal interaction molecule 1 deficiency. J. Immunol. 2012, 188, 1523–1533. [Google Scholar] [CrossRef] [Green Version]
  57. Parekh, A.B.; Fleig, A.; Penner, R. The store-operated calcium current I(CRAC): Nonlinear activation by InsP3 and dissociation from calcium release. Cell 1997, 89, 973–980. [Google Scholar] [CrossRef] [Green Version]
  58. Misquitta, C.M.; Chen, T.; Grover, A.K. Control of protein expression through mRNA stability in calcium signalling. Cell Calcium 2006, 40, 329–346. [Google Scholar] [CrossRef]
  59. Khan, I.; Spencer, G.G.; Samson, S.E.; Crine, P.; Boileau, G.; Grover, A.K. Abundance of sarcoplasmic reticulum calcium pump isoforms in stomach and cardiac muscles. Biochem. J. 1990, 268, 415–419. [Google Scholar] [CrossRef] [Green Version]
  60. Clemens, M.J. Targets and mechanisms for the regulation of translation in malignant transformation. Oncogene 2004, 23, 3180–3188. [Google Scholar] [CrossRef] [Green Version]
  61. Zhou, P.; Shaffer, D.R.; Alvarez Arias, D.A.; Nakazaki, Y.; Pos, W.; Torres, A.J.; Cremasco, V.; Dougan, S.K.; Cowley, G.S.; Elpek, K.; et al. In vivo discovery of immunotherapy targets in the tumour microenvironment. Nature 2014, 506, 52–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Parekh, A.B.; Penner, R. Depletion-activated calcium current is inhibited by protein kinase in RBL-2H3 cells. Proc. Natl. Acad. Sci. USA 1995, 92, 7907–7911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Breart, B.; Lemaitre, F.; Celli, S.; Bousso, P. Two-photon imaging of intratumoral CD8+ T cell cytotoxic activity during adoptive T cell therapy in mice. J. Clin. Investig. 2008, 118, 1390–1397. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Characteristics of expanded TILs. Immunophenotype and potency were evaluated in TIL derived from 10 patients after 14 days’ expansion. (A) Analysis of surface antigen expression. Results are expressed as the mean and SD % receptor surface expression of CD3+, CD3+/CD4+, CD3+/CD8+ and CD3−/CD56+ cell populations. (B) Cytotoxic activity of TILs was tested in a 5-h cytotoxicity assay against the autologous mCRC cells (black column) and SW480 cell lines (grey column) at various E:T ratios ranging from 50:1 to 12:1. Means and SD of percentages of specific lysis are reported.
Figure 1. Characteristics of expanded TILs. Immunophenotype and potency were evaluated in TIL derived from 10 patients after 14 days’ expansion. (A) Analysis of surface antigen expression. Results are expressed as the mean and SD % receptor surface expression of CD3+, CD3+/CD4+, CD3+/CD8+ and CD3−/CD56+ cell populations. (B) Cytotoxic activity of TILs was tested in a 5-h cytotoxicity assay against the autologous mCRC cells (black column) and SW480 cell lines (grey column) at various E:T ratios ranging from 50:1 to 12:1. Means and SD of percentages of specific lysis are reported.
Cancers 14 03312 g001
Figure 2. SOCE is enhanced in TILs. (A) The ER Ca2+ pool was depleted by challenging the cells with CPA (30 µM), a selective inhibitor of SERCA activity, in the absence of external Ca2+ (0Ca2+). Under 0Ca2+ conditions, CPA evoked a transient increase in [Ca2+]i that was due to passive ER Ca2+ efflux through leakage channels, followed by Ca2+ sequestration by multiple mechanisms, such as mitochondria, Na+/Ca2+ exchanger (NCX) and plasma-membrane Ca2+-ATPase (PMCA) [26,37,38]. Subsequently, SOCE was evaluated by replenishing Ca2+ to the perfusate (1.5 mM). (B) Mean ± SE of the amplitude of CPA-induced intracellular Ca2+ release and CPA-induced SOCE in hPBTs, cPBTs and TILs. One-way ANOVA followed by the post hoc Bonferroni test: * p < 0.05 and *** p < 0.001. (C) Pre-incubating TILs with BTP-2 (20 µm, 20 min), a selective Orai1 inhibitor, strongly reduced SOCE. BTP-2 was added in the continuous presence of extracellular Ca2+ after the recovery to the baseline of CPA-evoked intracellular Ca2+ mobilisation. (D) Mean ± SE of SOCE amplitude in the absence (Ctrl) and presence of BTP-2. Student’s t-test: *** p < 0.001. In panels (B,D), the numbers within or above the histogram bars indicate the number of responding cells over the total number of analysed cells.
Figure 2. SOCE is enhanced in TILs. (A) The ER Ca2+ pool was depleted by challenging the cells with CPA (30 µM), a selective inhibitor of SERCA activity, in the absence of external Ca2+ (0Ca2+). Under 0Ca2+ conditions, CPA evoked a transient increase in [Ca2+]i that was due to passive ER Ca2+ efflux through leakage channels, followed by Ca2+ sequestration by multiple mechanisms, such as mitochondria, Na+/Ca2+ exchanger (NCX) and plasma-membrane Ca2+-ATPase (PMCA) [26,37,38]. Subsequently, SOCE was evaluated by replenishing Ca2+ to the perfusate (1.5 mM). (B) Mean ± SE of the amplitude of CPA-induced intracellular Ca2+ release and CPA-induced SOCE in hPBTs, cPBTs and TILs. One-way ANOVA followed by the post hoc Bonferroni test: * p < 0.05 and *** p < 0.001. (C) Pre-incubating TILs with BTP-2 (20 µm, 20 min), a selective Orai1 inhibitor, strongly reduced SOCE. BTP-2 was added in the continuous presence of extracellular Ca2+ after the recovery to the baseline of CPA-evoked intracellular Ca2+ mobilisation. (D) Mean ± SE of SOCE amplitude in the absence (Ctrl) and presence of BTP-2. Student’s t-test: *** p < 0.001. In panels (B,D), the numbers within or above the histogram bars indicate the number of responding cells over the total number of analysed cells.
Cancers 14 03312 g002
Figure 3. SOCE induced by ER Ca2+ depletion through InsP3Rs and RyRs is enhanced in TILs. (A) The ER Ca2+ pool was depleted by challenging the cells with the InsP3-producing autacoid, ATP (100 µM) in the absence of external Ca2+ (0Ca2+). Under 0Ca2+ conditions, ATP evoked a transient increase in [Ca2+]i that rapidly returned to the baseline upon clearance of cytosolic Ca2+ by mitochondria, SERCA, NCX and PMCA [26,37,38]. Subsequently, SOCE was evaluated by replenishing Ca2+ (1.5 mM) to the perfusate in the absence of the agonist, which was removed 100 sec before re-addition of extracellular Ca2+. (B) Mean ± SE of the amplitude of ATP-induced intracellular Ca2+ release and SOCE in hPBTs, cPBTs and TILs. One-way ANOVA followed by the post hoc Bonferroni test: *** p < 0.001. (C) The ER Ca2+ pool was depleted by challenging the cells with InsP3BM (1 µM) in the absence of external Ca2+ (0Ca2+). Under 0Ca2+ conditions, InsP3BM evoked a transient increase in [Ca2+]i that rapidly returned to the baseline upon clearance of cytosolic Ca2+ by mitochondria, SERCA, NCX and PMCA [38]. Subsequently, SOCE was evaluated by replenishing Ca2+ to the perfusate (1.5 mM) in the absence of the agonist, which was removed 100 sec before re-addition of extracellular Ca2+. (D) Mean ± SE of the amplitude of InsP3BM-induced intracellular Ca2+ release and SOCE in hPBTs, cPBTs and TILs. One-way ANOVA followed by the post hoc Bonferroni test: ** p < 0.01 and *** p < 0.001. (E) The ER Ca2+ pool was depleted by challenging the cells with caffeine (5 mM), a selective RyR agonist, in the absence of external Ca2+ (0Ca2+). Under 0Ca2+ conditions, caffeine evoked a transient increase in [Ca2+]i that rapidly returned to the baseline upon clearance of cytosolic Ca2+ by mitochondria, SERCA, NCX and PMCA [40]. Subsequently, SOCE was evaluated by replenishing Ca2+ to the perfusate (1.5 mM) in the absence of the agonist, which was removed 100 sec before re-addition of extracellular Ca2+. (F) Mean ± SE of the amplitude of caffeine-induced intracellular Ca2+ release and SOCE in hPBTs, cPBTs and TILs. One-way ANOVA followed by the post hoc Bonferroni test: * p < 0.05 and *** p < 0.001. In panels (B,D,F), the numbers within or above the histogram bars indicate the number of responding cells over the total number of analysed cells.
Figure 3. SOCE induced by ER Ca2+ depletion through InsP3Rs and RyRs is enhanced in TILs. (A) The ER Ca2+ pool was depleted by challenging the cells with the InsP3-producing autacoid, ATP (100 µM) in the absence of external Ca2+ (0Ca2+). Under 0Ca2+ conditions, ATP evoked a transient increase in [Ca2+]i that rapidly returned to the baseline upon clearance of cytosolic Ca2+ by mitochondria, SERCA, NCX and PMCA [26,37,38]. Subsequently, SOCE was evaluated by replenishing Ca2+ (1.5 mM) to the perfusate in the absence of the agonist, which was removed 100 sec before re-addition of extracellular Ca2+. (B) Mean ± SE of the amplitude of ATP-induced intracellular Ca2+ release and SOCE in hPBTs, cPBTs and TILs. One-way ANOVA followed by the post hoc Bonferroni test: *** p < 0.001. (C) The ER Ca2+ pool was depleted by challenging the cells with InsP3BM (1 µM) in the absence of external Ca2+ (0Ca2+). Under 0Ca2+ conditions, InsP3BM evoked a transient increase in [Ca2+]i that rapidly returned to the baseline upon clearance of cytosolic Ca2+ by mitochondria, SERCA, NCX and PMCA [38]. Subsequently, SOCE was evaluated by replenishing Ca2+ to the perfusate (1.5 mM) in the absence of the agonist, which was removed 100 sec before re-addition of extracellular Ca2+. (D) Mean ± SE of the amplitude of InsP3BM-induced intracellular Ca2+ release and SOCE in hPBTs, cPBTs and TILs. One-way ANOVA followed by the post hoc Bonferroni test: ** p < 0.01 and *** p < 0.001. (E) The ER Ca2+ pool was depleted by challenging the cells with caffeine (5 mM), a selective RyR agonist, in the absence of external Ca2+ (0Ca2+). Under 0Ca2+ conditions, caffeine evoked a transient increase in [Ca2+]i that rapidly returned to the baseline upon clearance of cytosolic Ca2+ by mitochondria, SERCA, NCX and PMCA [40]. Subsequently, SOCE was evaluated by replenishing Ca2+ to the perfusate (1.5 mM) in the absence of the agonist, which was removed 100 sec before re-addition of extracellular Ca2+. (F) Mean ± SE of the amplitude of caffeine-induced intracellular Ca2+ release and SOCE in hPBTs, cPBTs and TILs. One-way ANOVA followed by the post hoc Bonferroni test: * p < 0.05 and *** p < 0.001. In panels (B,D,F), the numbers within or above the histogram bars indicate the number of responding cells over the total number of analysed cells.
Cancers 14 03312 g003
Figure 4. Molecular characterisation of the Ca2+-handling machinery in mCRC-derived T cells. (A) Transcriptomic profile of InsP3R, RyR, STIM, and Orai isoforms expressed in hPBTs. Data are calculated as 2ΔCt(GOI-S18) and presented as mean ± SE from seven biological replicates from three donors for each condition (hPBTs, cPBTs and TILs). One-way ANOVA followed by the post hoc Bonferroni test: *** p < 0.001. (B) Expression of STIM1 and Orai1 proteins in hPBTs, cPBTs and TILs. Major bands of the expected molecular weight for each protein were observed. (C) Mean ± SE of the densitometric analysis of three different experiments, each carried out on a different donor. STIM1 and Orai1 protein bands were derived from the same experiment; thus, actin bands are common to the two proteins studied. One-way ANOVA followed by the post hoc Bonferroni test: * p < 0.05 and ** p < 0.01. Whole Western blot showing all bands in Figure S8.
Figure 4. Molecular characterisation of the Ca2+-handling machinery in mCRC-derived T cells. (A) Transcriptomic profile of InsP3R, RyR, STIM, and Orai isoforms expressed in hPBTs. Data are calculated as 2ΔCt(GOI-S18) and presented as mean ± SE from seven biological replicates from three donors for each condition (hPBTs, cPBTs and TILs). One-way ANOVA followed by the post hoc Bonferroni test: *** p < 0.001. (B) Expression of STIM1 and Orai1 proteins in hPBTs, cPBTs and TILs. Major bands of the expected molecular weight for each protein were observed. (C) Mean ± SE of the densitometric analysis of three different experiments, each carried out on a different donor. STIM1 and Orai1 protein bands were derived from the same experiment; thus, actin bands are common to the two proteins studied. One-way ANOVA followed by the post hoc Bonferroni test: * p < 0.05 and ** p < 0.01. Whole Western blot showing all bands in Figure S8.
Cancers 14 03312 g004
Figure 5. DGK regulates SOCE amplitude in TILs. (A) SOCE was activated by challenging hPBTs with CPA (30 µM) for 20 min under 0Ca2+ conditions (not shown) followed by restitution of extracellular Ca2+ to the perfusate (1.5 mM). SOCE was measured in the absence (Ctrl) and in the presence of the following drugs: ritanserin (40 µM, 10 min; Rit) or R59949 (5 µM, 10 min; R59), which inhibit DGK activity; Gö-6983 (10 µM, 10 min; Gö-6), which blocks PKC; and PMA (10 µM, 10 min), which stimulates PKC. CPA-evoked SOCE was reduced by PMA, enhanced by Gö-6983, and not affected by ritanserin or R59949. (B) Mean ± SE of SOCE amplitude in hPBTs under the designated treatments. One-way ANOVA followed by the post hoc Bonferroni test: *** p < 0.001. (C) SOCE was activated by challenging cPBTs with CPA (30 µM) for 20 min under 0Ca2+ conditions (not shown) followed by restitution of extracellular Ca2+ to the perfusate (1.5 mM). SOCE was measured in the absence (Ctrl) and in the presence of the following drugs: ritanserin (40 µM, 10 min; Rit) or R59949 (5 µM, 10 min, R59), which inhibit DGK activity; Gö-6983 (10 µM, 10 min; Gö-6), which blocks PKC; and PMA (10 µM, 10 min), which stimulates PKC. CPA-evoked SOCE was reduced by PMA, ritanserin or R59949 and not affected by Gö-6983. (D) Mean ± SE of SOCE amplitude in cPBTs under the designated treatments. One-way ANOVA followed by the post hoc Bonferroni test: *** p < 0.001. (E) SOCE was activated by challenging TILs with CPA (30 µM) for 20 min under 0Ca2+ conditions (not shown) followed by restitution of extracellular Ca2+ to the perfusate (1.5 mM). SOCE was measured in the absence (Ctrl) and in the presence of the following drugs: ritanserin (40 µM, 10 min; Rit) or R59949 (5 µM, 10 min; R59), which inhibit DGK activity; Gö-6983 (10 µM, 10 min; Gö-6), which blocks PKC; and PMA (10 µM, 10 min), which stimulates PKC. CPA-evoked SOCE was reduced by PMA, ritanserin or R59949 and not affected by Gö-6983. (F) Mean ± SE of SOCE amplitude under the designated treatments. One-way ANOVA followed by the post hoc Bonferroni test: *** p < 0.001. In panels (B,D,F), the numbers within or above the histogram bars indicate the number of responding cells over the total number of analysed cells.
Figure 5. DGK regulates SOCE amplitude in TILs. (A) SOCE was activated by challenging hPBTs with CPA (30 µM) for 20 min under 0Ca2+ conditions (not shown) followed by restitution of extracellular Ca2+ to the perfusate (1.5 mM). SOCE was measured in the absence (Ctrl) and in the presence of the following drugs: ritanserin (40 µM, 10 min; Rit) or R59949 (5 µM, 10 min; R59), which inhibit DGK activity; Gö-6983 (10 µM, 10 min; Gö-6), which blocks PKC; and PMA (10 µM, 10 min), which stimulates PKC. CPA-evoked SOCE was reduced by PMA, enhanced by Gö-6983, and not affected by ritanserin or R59949. (B) Mean ± SE of SOCE amplitude in hPBTs under the designated treatments. One-way ANOVA followed by the post hoc Bonferroni test: *** p < 0.001. (C) SOCE was activated by challenging cPBTs with CPA (30 µM) for 20 min under 0Ca2+ conditions (not shown) followed by restitution of extracellular Ca2+ to the perfusate (1.5 mM). SOCE was measured in the absence (Ctrl) and in the presence of the following drugs: ritanserin (40 µM, 10 min; Rit) or R59949 (5 µM, 10 min, R59), which inhibit DGK activity; Gö-6983 (10 µM, 10 min; Gö-6), which blocks PKC; and PMA (10 µM, 10 min), which stimulates PKC. CPA-evoked SOCE was reduced by PMA, ritanserin or R59949 and not affected by Gö-6983. (D) Mean ± SE of SOCE amplitude in cPBTs under the designated treatments. One-way ANOVA followed by the post hoc Bonferroni test: *** p < 0.001. (E) SOCE was activated by challenging TILs with CPA (30 µM) for 20 min under 0Ca2+ conditions (not shown) followed by restitution of extracellular Ca2+ to the perfusate (1.5 mM). SOCE was measured in the absence (Ctrl) and in the presence of the following drugs: ritanserin (40 µM, 10 min; Rit) or R59949 (5 µM, 10 min; R59), which inhibit DGK activity; Gö-6983 (10 µM, 10 min; Gö-6), which blocks PKC; and PMA (10 µM, 10 min), which stimulates PKC. CPA-evoked SOCE was reduced by PMA, ritanserin or R59949 and not affected by Gö-6983. (F) Mean ± SE of SOCE amplitude under the designated treatments. One-way ANOVA followed by the post hoc Bonferroni test: *** p < 0.001. In panels (B,D,F), the numbers within or above the histogram bars indicate the number of responding cells over the total number of analysed cells.
Cancers 14 03312 g005
Figure 6. TCR stimulation by an anti-CD3 mAB evoked a stronger oscillatory Ca2+ response in TILs than in hPBTs and cPBTs. (A) Intracellular Ca2+ oscillations evoked in hPBTs by an anti-CD3 mAB (1 µg/mL). (B) Intracellular Ca2+ oscillations evoked in cPBTs by an anti-CD3 mAB (1 µg/mL). (C) Intracellular Ca2+ oscillations evoked in TILs by an anti-CD3 mAB (1 µg/mL). (D) Mean ± SE of the percentage of hPBTs, cPBTs and TILs responding to the anti-CD3 mAB stimulation. (E) Mean ± SE of the AUC of the spiking Ca2+ signal arising in the three cell types upon stimulation with the anti-CD3 mAB. One-way ANOVA followed by the post hoc Bonferroni test: *** p < 0.001. (F) Mean ± SE of oscillation frequency in the three cell types displaying the spiking Ca2+ response (i.e., more than 1 Ca2+ transient) to the anti-CD3 mAB. One-way ANOVA followed by the post hoc Bonferroni test: *** p < 0.001. (G) Pre-treating TILs with BTP-2 (20 µm, 20 min), which selectively inhibits SOCE by blocking extracellular Ca2+ influx through Orai1, dampens the intracellular Ca2+ oscillations evoked by the anti-CD3 mAB (1 µg/mL). (H) Mean ± SE of the AUC of the spiking Ca2+ signal arising in TILs challenged with the anti-CD3 mAB in the absence (Ctrl) and presence of BTP-2. Student’s t-test: *** p < 0.001. In panels (B,D,F,H), the numbers within or above the histogram bars indicate the number of responding cells over the total number of analysed cells.
Figure 6. TCR stimulation by an anti-CD3 mAB evoked a stronger oscillatory Ca2+ response in TILs than in hPBTs and cPBTs. (A) Intracellular Ca2+ oscillations evoked in hPBTs by an anti-CD3 mAB (1 µg/mL). (B) Intracellular Ca2+ oscillations evoked in cPBTs by an anti-CD3 mAB (1 µg/mL). (C) Intracellular Ca2+ oscillations evoked in TILs by an anti-CD3 mAB (1 µg/mL). (D) Mean ± SE of the percentage of hPBTs, cPBTs and TILs responding to the anti-CD3 mAB stimulation. (E) Mean ± SE of the AUC of the spiking Ca2+ signal arising in the three cell types upon stimulation with the anti-CD3 mAB. One-way ANOVA followed by the post hoc Bonferroni test: *** p < 0.001. (F) Mean ± SE of oscillation frequency in the three cell types displaying the spiking Ca2+ response (i.e., more than 1 Ca2+ transient) to the anti-CD3 mAB. One-way ANOVA followed by the post hoc Bonferroni test: *** p < 0.001. (G) Pre-treating TILs with BTP-2 (20 µm, 20 min), which selectively inhibits SOCE by blocking extracellular Ca2+ influx through Orai1, dampens the intracellular Ca2+ oscillations evoked by the anti-CD3 mAB (1 µg/mL). (H) Mean ± SE of the AUC of the spiking Ca2+ signal arising in TILs challenged with the anti-CD3 mAB in the absence (Ctrl) and presence of BTP-2. Student’s t-test: *** p < 0.001. In panels (B,D,F,H), the numbers within or above the histogram bars indicate the number of responding cells over the total number of analysed cells.
Cancers 14 03312 g006
Figure 7. SOCE evoked by an anti-CD3 mAB is larger in TILs compared to hPBTs and cPBTs. (A) Intracellular Ca2+ mobilisation and SOCE activation in response to exogenous administration of an anti-CD3 mAB (1 µg/mL) in hPBTs, cPBTs and TILs. The anti-CD3 mAB was administered under 0Ca2+ conditions to evaluate endogenous Ca2+ release. Extracellular Ca2+ was returned to the perfusate (1.5 mM) after the recovery of the initiate Ca2+ transient to the baseline to measure SOCE. (B) Mean ± SE of the amplitude of ER Ca2+ mobilisation and SOCE evoked by the anti-CD3 mAB in the three cell types. One-way ANOVA followed by the post hoc Bonferroni test: *** p < 0.001. The numbers within or above the histogram bars indicate the number of responding cells over the total number of analysed cells.
Figure 7. SOCE evoked by an anti-CD3 mAB is larger in TILs compared to hPBTs and cPBTs. (A) Intracellular Ca2+ mobilisation and SOCE activation in response to exogenous administration of an anti-CD3 mAB (1 µg/mL) in hPBTs, cPBTs and TILs. The anti-CD3 mAB was administered under 0Ca2+ conditions to evaluate endogenous Ca2+ release. Extracellular Ca2+ was returned to the perfusate (1.5 mM) after the recovery of the initiate Ca2+ transient to the baseline to measure SOCE. (B) Mean ± SE of the amplitude of ER Ca2+ mobilisation and SOCE evoked by the anti-CD3 mAB in the three cell types. One-way ANOVA followed by the post hoc Bonferroni test: *** p < 0.001. The numbers within or above the histogram bars indicate the number of responding cells over the total number of analysed cells.
Cancers 14 03312 g007
Figure 8. Reducing SOCE with BTP-2 increases TIL cytotoxicity against autologous mCRC cells. (AC): Three distinct patterns of intracellular Ca2+ signals evoked in TILs plated on 8 mm glass coverslips (4 × 105 cells/mL) by the contact with autologous mCRC cells (1 × 104 cells/mL): (A), intracellular Ca2+ oscillations (“oscillatory pattern”); (B), two slow, intracellular Ca2+ waves (“slow pattern”); and (C), fast Ca2+ transient followed by a slow increase in [Ca2+]i that ultimately achieved a plateau phase (“plateau pattern”). (D) Pre-treating TILs with BTP-2 (20 µm, 20 min) to selectively inhibit SOCE dampened the Ca2+ response to TCR engagement by autologous mCRC cells (1 × 104 cells/mL) and suppressed the “slow” and “plateau” patterns. (E) Mean ± SE of the AUC of the spiking Ca2+ signal arising in TILs challenged with autologous mCRC cells in the absence (Ctrl) and presence of BTP-2. Student’s t-test: *** p < 0.001. (F) Mean ± SE of oscillation frequency in TILs exposed to autologous mCRC cells in the absence (Ctrl) and presence of BTP-2. Student’s t-test: *** p < 0.001. (G) Δ of the % increase in specific lysis at each E:T ratio. (H) Cytotoxic activity of TIL derived from five patients was tested through a 5 h cytotoxicity assay against autologous mCRC cells in medium alone (Ctrl) or in the presence of BTP-2 at various E:T ratios. Data are presented as mean ± SE from three separate experiments. Student’s t-test: * p < 0.05. In panels (E,F), the numbers within or above the histogram bars indicate the number of responding cells over the total number of analysed cells.
Figure 8. Reducing SOCE with BTP-2 increases TIL cytotoxicity against autologous mCRC cells. (AC): Three distinct patterns of intracellular Ca2+ signals evoked in TILs plated on 8 mm glass coverslips (4 × 105 cells/mL) by the contact with autologous mCRC cells (1 × 104 cells/mL): (A), intracellular Ca2+ oscillations (“oscillatory pattern”); (B), two slow, intracellular Ca2+ waves (“slow pattern”); and (C), fast Ca2+ transient followed by a slow increase in [Ca2+]i that ultimately achieved a plateau phase (“plateau pattern”). (D) Pre-treating TILs with BTP-2 (20 µm, 20 min) to selectively inhibit SOCE dampened the Ca2+ response to TCR engagement by autologous mCRC cells (1 × 104 cells/mL) and suppressed the “slow” and “plateau” patterns. (E) Mean ± SE of the AUC of the spiking Ca2+ signal arising in TILs challenged with autologous mCRC cells in the absence (Ctrl) and presence of BTP-2. Student’s t-test: *** p < 0.001. (F) Mean ± SE of oscillation frequency in TILs exposed to autologous mCRC cells in the absence (Ctrl) and presence of BTP-2. Student’s t-test: *** p < 0.001. (G) Δ of the % increase in specific lysis at each E:T ratio. (H) Cytotoxic activity of TIL derived from five patients was tested through a 5 h cytotoxicity assay against autologous mCRC cells in medium alone (Ctrl) or in the presence of BTP-2 at various E:T ratios. Data are presented as mean ± SE from three separate experiments. Student’s t-test: * p < 0.05. In panels (E,F), the numbers within or above the histogram bars indicate the number of responding cells over the total number of analysed cells.
Cancers 14 03312 g008
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Faris, P.; Rumolo, A.; Tapella, L.; Tanzi, M.; Metallo, A.; Conca, F.; Negri, S.; Lefkimmiatis, K.; Pedrazzoli, P.; Lim, D.; et al. Store-Operated Ca2+ Entry Is Up-Regulated in Tumour-Infiltrating Lymphocytes from Metastatic Colorectal Cancer Patients. Cancers 2022, 14, 3312. https://doi.org/10.3390/cancers14143312

AMA Style

Faris P, Rumolo A, Tapella L, Tanzi M, Metallo A, Conca F, Negri S, Lefkimmiatis K, Pedrazzoli P, Lim D, et al. Store-Operated Ca2+ Entry Is Up-Regulated in Tumour-Infiltrating Lymphocytes from Metastatic Colorectal Cancer Patients. Cancers. 2022; 14(14):3312. https://doi.org/10.3390/cancers14143312

Chicago/Turabian Style

Faris, Pawan, Agnese Rumolo, Laura Tapella, Matteo Tanzi, Alessia Metallo, Filippo Conca, Sharon Negri, Konstantinos Lefkimmiatis, Paolo Pedrazzoli, Dmitry Lim, and et al. 2022. "Store-Operated Ca2+ Entry Is Up-Regulated in Tumour-Infiltrating Lymphocytes from Metastatic Colorectal Cancer Patients" Cancers 14, no. 14: 3312. https://doi.org/10.3390/cancers14143312

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop