Introduction
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a type II transmembrane protein belonging to the tumor necrosis factor (TNF) superfamily that can be cleaved by cysteine protease to obtain a soluble ligand [
[1]- Liabakk N-B
- Sundan A
- Torp S
- Aukrust P
- Frøland SS
- Espevik T.
Development, characterization and use of monoclonal antibodies against sTRAIL: measurement of sTRAIL by ELISA.
]. It is physiologically expressed by various cells of both the innate and adaptive immune system, including macrophages, T lymphocytes, dendritic cells and natural killer (NK) and natural killer T cells, and it mediates immunoregulatory and immune-effector functions by binding to its functional death receptors, TRAIL-R1/DR4 and TRAIL-R2/DR5, expressed on the target cell surface, with the consequent activation of apoptotic signaling pathways [
[2]- Falschlehner C
- Schaefer U
- Walczak H.
Following TRAIL's path in the immune system.
,
[3]- Song K
- Chen Y
- Göke R
- Wilmen A
- Seidel C
- Göke A
- et al.
Tumor necrosis factor–related apoptosis-inducing ligand (TRAIL) is an inhibitor of autoimmune inflammation and cell cycle progression.
]. In particular, endogenous TRAIL acts as a modulator of events involving the immune system, such as the maintenance of physiological homeostasis and the response to viral infections and autoimmune diseases [
[4]- Beyer Baukloh
- Stoyanova Kamphues
- Sattler Kotsch
Interactions of tumor necrosis factor–related apoptosis-inducing ligand (TRAIL) with the immune system: implications for inflammation and cancer.
]. It has been reported that TRAIL induces deletion of developing autoreactive T and B lymphocytes in central tolerance mechanisms, thereby preserving T-cell homeostasis [
[5]- Bossi F
- Bernardi S
- Zauli G
- Secchiero P
- Fabris B.
TRAIL modulates the immune system and protects against the development of diabetes.
]; accelerates neutrophils apoptosis, inducing the resolution of inflammation [
[6]- Kamohara H
- Matsuyama W
- Shimozato O
- Abe K
- Galligan C
- Hashimoto S-I
- et al.
Regulation of tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) and TRAIL receptor expression in human neutrophils.
] as well as inhibits T helper 1 and 2 (Th1 and Th2) cells [
[7]- Ikeda T
- Hirata S
- Fukushima S
- Matsunaga Y
- Ito T
- Uchino M
- et al.
Dual effects of TRAIL in suppression of autoimmunity: the inhibition of Th1 cells and the promotion of regulatory T cells.
]. TRAIL has a protective role against autoimmune diseases such as autoimmune encephalomyelitis, rheumatoid arthritis, and type I diabetes, preserving peripheral tolerance through the killing of autoreactive T lymphocytes, inhibition of their activation and promotion of T regulatory cell proliferation [
[8]- Rossin A
- Miloro G
- Hueber A-O.
TRAIL and FasL functions in cancer and autoimmune diseases: towards an increasing complexity.
]. In addition, TRAIL expressed by immune cells plays a fundamental role in immunosurveillance against tumors and metastasis, acting as a tumor suppressor [
[9]- Cretney E
- Takeda K
- Yagita H
- Glaccum M
- Peschon JJ
- Smyth MJ.
Increased susceptibility to tumor initiation and metastasis in TNF-related apoptosis-inducing ligand-deficient mice.
]. In this context, TRAIL can exert its effect directly by inducing apoptosis in tumor cells or indirectly by promoting cytokine secretion, including interleukin (IL)-8, protein urokinase and matrix metalloproteinase-7 and -9, by cancer cells stimulating immune system regulation [
[4]- Beyer Baukloh
- Stoyanova Kamphues
- Sattler Kotsch
Interactions of tumor necrosis factor–related apoptosis-inducing ligand (TRAIL) with the immune system: implications for inflammation and cancer.
]. In particular, TRAIL is a relevant part in the apoptotic activity of NK against tumor cells: NK-cell activation is followed by TRAIL expression with a proapoptotic antitumor effect even against disseminated cancer cells [
[10]Mechanisms of natural killer cell-mediated cellular cytotoxicity.
]. Moreover, monocytes, neutrophils and immune effector cytotoxic dendritic cells and T cells contribute to TRAIL-mediated apoptosis in cancer cells [
[11]- de Looff M
- de Jong S
- Kruyt FAE.
Multiple interactions between cancer cells and the tumor microenvironment modulate TRAIL signaling: implications for TRAIL receptor targeted therapy.
].
Based on the anticancer activity of TRAIL-expressing immune effector cells, the recombinant human (rh) TRAIL molecule has attracted significant attention as an antitumor compound because of its ability to selectively trigger apoptotic signaling in tumor cells expressing functional TRAIL receptors while sparing healthy tissues. However, because of the suboptimal pharmacokinetics of rhTRAIL, the therapeutic response in patients has been lower than expected, and its clinical use has been limited [
[12]- Lemke J
- von Karstedt S
- Zinngrebe J
- Walczak H.
Getting TRAIL back on track for cancer therapy.
]. Given this scenario, different research groups explored various delivery strategies to ameliorate the bioavailability of TRAIL, including the use of genetically modified mesenchymal stromal cells (MSCs) engineered to express this proapoptotic protein. The capacity of MSCs to be manipulated by viral vectors encoding for therapeutic molecules along with their tropism for tumor sites makes them an efficient vehicle for TRAIL delivery in both preclinical and clinical settings [
[13]Stem cell-based therapies for cancer treatment: separating hope from hype.
,
[14]- Hammer K
- Kazcorowski A
- Liu L
- Behr M
- Schemmer P
- Herr I
- et al.
Engineered adenoviruses combine enhanced oncolysis with improved virus production by mesenchymal stromal carrier cells: oncolytic adenoviruses for MSC delivery to pancreatic cancer.
]. In the past few years, our group has developed an anticancer gene therapy approach based on adipose-derived (AD) MSC to express TRAIL against several cancer types [
15- Grisendi G
- Bussolari R
- Cafarelli L
- Petak I
- Rasini V
- Veronesi E
- et al.
Adipose-derived mesenchymal stem cells as stable source of tumor necrosis factor–related apoptosis-inducing ligand delivery for cancer therapy.
,
16- Grisendi G
- Spano C
- D'souza N
- Rasini V
- Veronesi E
- Prapa M
- et al.
Mesenchymal progenitors expressing TRAIL induce apoptosis in sarcomas.
,
17- Golinelli G
- Grisendi G
- Spano C
- Dominici M.
Surrounding pancreatic adenocarcinoma by killer mesenchymal stromal/stem cells.
,
18- Spano C
- Grisendi G
- Golinelli G
- Rossignoli F
- Prapa M
- Bestagno M
- et al.
Soluble TRAIL armed human MSC as gene therapy for pancreatic cancer.
,
19- Rossignoli F
- Spano C
- Grisendi G
- Foppiani EM
- Golinelli G
- Mastrolia I
- et al.
MSC-delivered soluble TRAIL and paclitaxel as novel combinatory treatment for pancreatic adenocarcinoma.
,
20- Golinelli G
- Grisendi G
- Prapa M
- Bestagno M
- Spano C
- Rossignoli F
- et al.
Targeting GD2-positive glioblastoma by chimeric antigen receptor empowered mesenchymal progenitors.
,
21- Golinelli G
- Grisendi G
- Dall'Ora M
- Casari G
- Spano C
- Talami R
- et al.
Anti-GD2 CAR MSCs against metastatic Ewing's sarcoma.
]. AD-MSC delivering a secreted TRAIL variant as a soluble ligand (sTRAIL) have been generated, and these cells are characterized by a greater capacity to form multimeric complexes. This increased polymerization capacity confers sTRAIL superior cytotoxic activity against different tumor cell lines compared with the wild-type form [
18- Spano C
- Grisendi G
- Golinelli G
- Rossignoli F
- Prapa M
- Bestagno M
- et al.
Soluble TRAIL armed human MSC as gene therapy for pancreatic cancer.
,
19- Rossignoli F
- Spano C
- Grisendi G
- Foppiani EM
- Golinelli G
- Mastrolia I
- et al.
MSC-delivered soluble TRAIL and paclitaxel as novel combinatory treatment for pancreatic adenocarcinoma.
,
20- Golinelli G
- Grisendi G
- Prapa M
- Bestagno M
- Spano C
- Rossignoli F
- et al.
Targeting GD2-positive glioblastoma by chimeric antigen receptor empowered mesenchymal progenitors.
,
21- Golinelli G
- Grisendi G
- Dall'Ora M
- Casari G
- Spano C
- Talami R
- et al.
Anti-GD2 CAR MSCs against metastatic Ewing's sarcoma.
]. Considering the multiple and critical roles played by TRAIL in the immune system, both in physiological and pathological conditions, we aimed to determine the impact of the stronger cytotoxic activity by the novel sTRAIL form together with the capability of MSC to regulate immune cell activity by immunomodulatory mechanisms [
[22]Proteomic techniques for characterisation of mesenchymal stem cell secretome.
] on white blood cells (WBCs). In this regard, we investigated the viability, metabolic activity and cytokine release of WBCs to collect exploratory
in vitro data to inform the clinical safety of this MSC-based TRAIL delivery therapeutic approach.
Methods
Isolation and in vitro culture of WBCs
Human peripheral blood mononuclear cells (PBMCs) and human polymorphonuclear cells (PMNs) were separated by density gradient centrifugation (Lymphoprep; Alere Technologies AS, Oslo, Norway) from the peripheral blood of healthy donors (n = 3) as approved by the institutional review board of University Hospital of Modena, Modena, Italy.
Monocytes culture
PBMCs were plated in RPMI 1640 (Gibco-Life Technologies, Grand Island, NY, USA) with 0.5% heat-inactivated defined fetal bovine serum (FBS; HyClone Laboratories, UT, USA) and 1% glutamine (200 mmol/L; Euroclone, Pero, MI, Italy) for 1 h to isolate monocytes. Non-adherent cells were then removed and monocytes were cultivated for 24 h in RPMI 1640 supplemented with 10% heat-inactivated defined FBS and 1% glutamine and then used for fluorescence-activated cell sorting (FACS) analysis and cell viability assays.
Polymorphonuclear culture
PMNs, isolated after gradient density separation, were immediately tested by FACS analysis or plated in RPMI 1640 with 10% heat-inactivated defined FBS and 1% glutamine for cell viability assays.
Lymphocytes culture
PBMCs were plated in RPMI 1640 with 1% heat-inactivated defined FBS, 1% glutamine and 1% penicillin–streptomycin (10 000 units penicillin and 10 mg/mL streptomycin in 0.9% sodium chloride; Sigma-Aldrich, St Louis, MO, USA) for 2 h to obtain lymphocytes. Non-adherent cells were collected and pre-stimulated for 48 h in RPMI 1640 supplemented with 10% heat-inactivated defined FBS, 1% glutamine, 1% penicillin–streptomycin, 500 UI/mL rh interleukin-2 (Proleukin; Clinigen Healthcare Ltd, Staffordshire, UK) and 7 μg/mL phytohemagglutinin (Sigma-Aldrich) at a concentration of 1 × 106 cells/mL. After 48 h, isolated cells were cultured in RPMI 1640 with 10% heat-inactivated defined FBS, 1% glutamine, 1% penicillin–streptomycin and 500 UI/mL rh interleukin-2 and used for FACS analysis, cell viability and metabolic activity assays.
FACS analyses
Monocytes were stained using fluorescein isothiocyanate (FITC)-conjugated anti-CD45 (BD Biosciences, San Jose, CA, USA), phycoerythrin (PE)-conjugated anti-CD14 (BD Biosciences), peridinin–chlorophyll–protein–conjugated anti-CD3 (BD Biosciences) and BD Via Probe Cell Viability Solution (7-aminoactinomycin D [7-AAD]; BD Pharmingen, San Diego, CA, USA). PMNs were stained using FITC-conjugated anti-CD45, allophycocyanin (APC)-conjugated anti-CD10 (BD Biosciences) and BD Via Probe Cell Viability Solution (7-AAD). In vitro–expanded T lymphocytes were stained using FITC-conjugated anti-CD45, peridinin–chlorophyll–protein–conjugated anti-CD3, APC-conjugated anti-CD4 (BD Biosciences), PE-conjugated anti-CD8 (BD Biosciences) and BD Via Probe Cell Viability Solution (7-AAD). To assess TRAIL receptor expression, monocytes, PMNs and T lymphocytes were tested for PE-conjugated anti-DR4 (BioLegend, San Diego, CA, USA), APC-conjugated anti-DR5 (BioLegend), PE-conjugated anti-DcR1 (BioLegend) and APC-conjugated anti-DcR2 (R&D Systems, Minneapolis, MN, USA). Isotype control antibodies were used for all cell types and antigens analyzed. All samples were acquired using the BD FACSAria III (BD, Franklin Lakes, NJ, USA) flow cytometer and analyzed using the BD FACSDiva software.
Cell viability assays
Monocytes, PMNs and T cells were tested in a dose–response assay for sTRAIL cytotoxicity using the CellTiter-Glo Luminescent Cell Viability assay (Promega, Madison, WI, USA). In detail, cells were seeded in a 96-well plate at a density of 200 000 cells/well in cell culture medium (without IL-2 for T lymphocytes). Supernatants conditioned by AD-MSC secreting sTRAIL (AD-MSC sTRAIL) or AD-MSC EMPTY as the control were produced as previously reported [
[18]- Spano C
- Grisendi G
- Golinelli G
- Rossignoli F
- Prapa M
- Bestagno M
- et al.
Soluble TRAIL armed human MSC as gene therapy for pancreatic cancer.
]. The next day, the cell medium was replaced with the supernatants collected from AD-MSC sTRAIL, containing increasing doses of sTRAIL (from 50 to 5000 pg/mL). WBCs were also treated with increasing volumes of supernatants collected from AD-MSC EMPTY, corresponding to the volumes of supernatant from AD-MSC sTRAIL tested. After 24 h, treated cells were incubated with the CellTiter-Glo reagent for 10 min according to the manufacturer's instructions. The cell viability rate was quantified by measuring the bioluminescence signal using the GloMax Discover microplate reader (Promega). To assess the impact of direct cell-to-cell contact with AD-MSC sTRAIL on monocyte and T-lymphocyte death, primary immune cells were cultured over a layer of AD-MSC sTRAIL, which were previously allowed to adhere to 12-well plates for 24 h, at different effector:target (E:T) ratios (1:30, 1:15, 1:10, 1:1, and 10:1; E = AD-MSC, T = T cells) in RPMI 1640 with 10% heat-inactivated defined FBS and 1% glutamine (and 1% penicillin–streptomycin for T lymphocytes). Monocytes or T cells alone (CTL) and co-cultured with AD-MSC EMPTY at a 10:1 E:T ratio were used as controls. Monocyte and T-lymphocyte death rate was evaluated by the supravital propidium iodide (PI; Sigma-Aldrich) assay after 24 h of co-culture. To summarize, non-permeabilized cells were stained using 50 µg/mL PI for 30 min [
[23]- Zamai L
- Canonico B
- Luchetti F
- Ferri P
- Melloni E
- Guidotti L
- et al.
Supravital exposure to propidium iodide identifies apoptosis on adherent cells.
] and primary immune cell death was quantified by FACS, gating on PI-positive cells.
Metabolic activity assay
The metabolic activity of T lymphocytes treated by sTRAIL was evaluated using the alamarBlue assay (Invitrogen Corporation, Carlsbad, CA, USA). T lymphocytes were incubated with increasing doses of sTRAIL (from 50 to 5000 pg/mL) following the same aforementioned protocol for the cell viability assay. After 24, 48 and 72 h of treatment, T lymphocytes were incubated with 10% alamarBlue for 4 h at 37°C. T-cell metabolic activity was quantified by measuring the fluorescence signal (λex = 530-560 and λem = 590 nm) using the GloMax Discover microplate reader.
Multiplex enzyme-linked immunosorbent assay (ELISA)
IL-10, tumor necrosis factor alpha (TNFα), interferon gamma (IFNγ), vascular endothelial growth factor A (VEGF-A) and IL-6 levels were measured in supernatants collected from the co-culture of T cells and AD-MSC sTRAIL (or EMPTY as the control) at different E:T ratios for 24, 48 and 72 h. The evaluation was done by Labospace Srl using a magnetic Luminex assay (multiplex ELISA; R&D Systems). Data were reported as fg/mL per T cell for cytokines secreted by T lymphocytes, and fg/mL per AD-MSC for cytokines secreted by AD-MSC.
Statistical analysis
Continuous variables are expressed as absolute or percentage frequencies and are described as mean ± standard error. A multiple comparison of the observed data is obtained through the one-way ANOVA followed by Tukey's test as post hoc test. Significant results were considered when P ˂ 0.05.
Discussion
For years, the potential use of TRAIL as an anticancer agent has aroused great interest in oncology research. The tumor-selective activity of TRAIL, which triggers the apoptotic signaling cascade preferentially in cancer cells with little-to-no effect in normal cells (e.g., lung fibroblasts, skeletal muscle cells, epidermal keratinocytes and melanocytes) [
[12]- Lemke J
- von Karstedt S
- Zinngrebe J
- Walczak H.
Getting TRAIL back on track for cancer therapy.
], could be of value to the clinical use of this molecule. Nevertheless, the issues concerning its poor pharmacokinetics and biodistribution have considerably limited its use in oncologic patients [
[12]- Lemke J
- von Karstedt S
- Zinngrebe J
- Walczak H.
Getting TRAIL back on track for cancer therapy.
,
[25]- Moniri MR
- Sun X-Y
- Rayat J
- Dai D
- Ao Z
- He Z
- et al.
TRAIL-engineered pancreas-derived mesenchymal stem cells: characterization and cytotoxic effects on pancreatic cancer cells.
]. For this reason, many technological improvements have been made to ameliorate TRAIL delivery and effectiveness against solid tumors, including the use of gene-modified MSC as a cellular vehicle [
[14]- Hammer K
- Kazcorowski A
- Liu L
- Behr M
- Schemmer P
- Herr I
- et al.
Engineered adenoviruses combine enhanced oncolysis with improved virus production by mesenchymal stromal carrier cells: oncolytic adenoviruses for MSC delivery to pancreatic cancer.
,
[15]- Grisendi G
- Bussolari R
- Cafarelli L
- Petak I
- Rasini V
- Veronesi E
- et al.
Adipose-derived mesenchymal stem cells as stable source of tumor necrosis factor–related apoptosis-inducing ligand delivery for cancer therapy.
]. Thanks to their ability to be attracted to tumor sites and their resistance to the proapoptotic stimuli mediated by TRAIL, MSCs represent a good cellular carrier for the delivery of this antitumor molecule in a clinical scenario. Recently, we have developed an anticancer cell therapy approach based on gene-modified AD-MSC expressing a novel secreted variant of TRAIL characterized by the presence of an isoleucine zipper domain that confers the capacity to form more stable multimeric complexes of TRAIL, thus resulting in greater cytotoxic activity on tumor cells compared with the recombinant human molecule [
[18]- Spano C
- Grisendi G
- Golinelli G
- Rossignoli F
- Prapa M
- Bestagno M
- et al.
Soluble TRAIL armed human MSC as gene therapy for pancreatic cancer.
]. The improvement in the protein structure and the MSC-based delivery may represent the needed strategies to enhance the use of TRAIL in a clinical setting.
According to that, the assessment of the immunological safety of AD-MSC sTRAIL is crucial considering both the immunomodulatory activity displayed by MSC on immune cells and the role played by TRAIL in the regulation of innate and adaptive immune responses [
[2]- Falschlehner C
- Schaefer U
- Walczak H.
Following TRAIL's path in the immune system.
,
[22]Proteomic techniques for characterisation of mesenchymal stem cell secretome.
]. Indeed, TRAIL can be expressed by immune cells based on their activation status, with the upregulation of TRAIL on both innate and adaptive immune cells, such as monocytes, neutrophils, T and B lymphocytes and NK cells, when stimulated with type I IFN. In addition to TRAIL expression, activated immune cells also can express decoy TRAIL receptors, thereby creating resistance to TRAIL-mediated apoptotic signaling [
[26]- Cardoso Alves L
- Corazza N
- Micheau O
- Krebs P
The multifaceted role of TRAIL signaling in cancer and immunity.
]. TRAIL has been referred to as a guardian against pathogen infections, autoimmunity and cancer onset and progression, playing a crucial role in immunosurveillance mechanisms [
[4]- Beyer Baukloh
- Stoyanova Kamphues
- Sattler Kotsch
Interactions of tumor necrosis factor–related apoptosis-inducing ligand (TRAIL) with the immune system: implications for inflammation and cancer.
]. Moreover, it has been reported that TRAIL expressed by neutrophils, CD8+ T lymphocytes and NK cells promotes the elimination of pathogen-infected cells expressing functional TRAIL receptors, thus containing infections [
[26]- Cardoso Alves L
- Corazza N
- Micheau O
- Krebs P
The multifaceted role of TRAIL signaling in cancer and immunity.
]. In autoimmunity settings, TRAIL inhibits the activation of autoreactive T cells but promotes T regulatory proliferation in diabetes, rheumatoid arthritis, and autoimmune encephalomyelitis [
[8]- Rossin A
- Miloro G
- Hueber A-O.
TRAIL and FasL functions in cancer and autoimmune diseases: towards an increasing complexity.
]. Moreover, monocytes stimulated with IFNγ, CD4+ T cells and NK cells expressing TRAIL can recognize and then neutralize transformed cells, inhibiting tumor growth and preventing tumor metastases within the immunosurveillance process [
[27]- Griffith TS
- Wiley SR
- Kubin MZ
- Sedger LM
- Maliszewski CR
- Fanger NA.
Monocyte-mediated tumoricidal activity via the tumor necrosis factor–related cytokine, TRAIL.
].
For the first time, we also have evaluated the impact of a novel form of sTRAIL released by AD-MSC on the viability, metabolic activity and cytokine release of WBCs. At first, we evaluated by flow cytometry the expression of both functional and decoy TRAIL receptors responsible for inducing or inhibiting TRAIL signaling, respectively, on freshly isolated purified blood leukocytes from healthy donors. As reported by Liguori
et al. [
[28]- Liguori M
- Buracchi C
- Pasqualini F
- Bergomas F
- Pesce S
- Sironi M
- et al.
Functional TRAIL receptors in monocytes and tumor-associated macrophages: A possible targeting pathway in the tumor microenvironment.
], we observed heterogeneous expression of TRAIL receptors on resting WBCs: DR5 was mainly expressed on monocytes, whereas the decoy receptors (DcR1 and DcR2) were essentially expressed on neutrophils. On the contrary, resting T cells revealed weak expression of all TRAIL receptors.
Since our MSC-based cell product releases multimeric sTRAIL complexes into the cell culture medium [
[18]- Spano C
- Grisendi G
- Golinelli G
- Rossignoli F
- Prapa M
- Bestagno M
- et al.
Soluble TRAIL armed human MSC as gene therapy for pancreatic cancer.
], we first investigated the biological impact of supernatant collected from AD-MSC sTRAIL on primary resting leukocytes. For all considered immune cell subtypes, we did not observe any toxicity even at the greatest tested sTRAIL concentration (5000 pg/mL). Particularly for monocytes, despite the high expression of DR5, we did not observe any cytotoxic effect mediated by sTRAIL but rather we registered an unexpected significative increase of cell viability directly related to the sTRAIL concentrations. If on the one hand the mechanisms of TRAIL resistance in transformed cells are largely investigated, since they represent a fundamental key to improve the anticancer potential of TRAIL based therapies, on the other hand the cellular processes leading TRAIL resistance in non-transformed cells are not so explored. Along with the expression of decoy receptors, it has been demonstrated recently that non-transformed cells, including monocyte from healthy donors, escape TRAIL-mediated apoptosis by the activation of multiple and redundant antiapoptotic molecular mechanisms, including the up-regulation of c-FLIP, IAP and Bcl-2, which prevents the activation of apoptotic signals [
29- Olingy CE
- Dinh HQ
- Hedrick CC.
Monocyte heterogeneity and functions in cancer.
,
30- Patysheva M
- Frolova A
- Larionova I
- Afanas'ev S
- Tarasova A
- Cherdyntseva N
- Kzhyshkowska J.
Monocyte programming by cancer therapy.
,
31- van Dijk M
- Halpin-McCormick A
- Sessler T
- Samali A
- Szegezdi E.
Resistance to TRAIL in non-transformed cells is due to multiple redundant pathways.
,
32- Zhang XD
- Nguyen T
- Thomas WD
- Sanders JE
- Hersey P.
Mechanisms of resistance of normal cells to TRAIL induced apoptosis vary between different cell types.
]
. The removal of one of these inhibitors is not sufficient to sensitize non-transformed cells to TRAIL and only the simultaneous loss of two or more of these proteins (i.e., cellular FLICE-like inhibitory protein [cFLIP] and XIAP expression or loss of cFLIP and antiapoptotic Bcl-2 protein-function) could induce TRAIL sensitivity [
[31]- van Dijk M
- Halpin-McCormick A
- Sessler T
- Samali A
- Szegezdi E.
Resistance to TRAIL in non-transformed cells is due to multiple redundant pathways.
]. Surprisingly, we registered a relevant and significant increase in cell viability for PMNs after exposure to CM collected from engineered AD-MSC. This effect was visible in cells treated with supernatant collected from both AD-MSC sTRAIL and AD-MSC EMPTY, suggesting that soluble trophic factors and cytokines secreted by MSC within the supernatant, such as granulocyte-macrophage colony-stimulating factor, IL-6 and IL-8, could support PMN's
in vitro performance [
[32]- Zhang XD
- Nguyen T
- Thomas WD
- Sanders JE
- Hersey P.
Mechanisms of resistance of normal cells to TRAIL induced apoptosis vary between different cell types.
,
[33]Multipotent mesenchymal stromal cells and the innate immune system.
]. In addition, no significant variations were revealed in the viability of T cells treated with CM from AD-MSC sTRAIL compared with the control. It has been reported that resting peripheral T cells are resistant to TRAIL-mediated apoptosis, in contrast to T lymphocytes activated by antigenic stimulation or within autoreactive processes, as a mechanism to prevent potentially dangerous autoimmune damage [
7- Ikeda T
- Hirata S
- Fukushima S
- Matsunaga Y
- Ito T
- Uchino M
- et al.
Dual effects of TRAIL in suppression of autoimmunity: the inhibition of Th1 cells and the promotion of regulatory T cells.
,
34- Joel MDM
- Yuan J
- Wang J
- Yan Y
- Qian H
- Zhang X
- et al.
MSC: immunoregulatory effects, roles on neutrophils and evolving clinical potentials.
,
35- Martínez-Lorenzo MJ
- Alava MA
- Gamen S
- Kim KJ
- Chuntharapai A
- Piñeiro A
- et al.
Involvement of APO2 ligand/TRAIL in activation-induced death of Jurkat and human peripheral blood T cells.
]. This result is not unexpected, considering both the low expression of DR4 and DR5 revealed by FACS analysis and the data reported by Clancy
et al. [
[36]- Clancy L
- Mruk K
- Archer K
- Woelfel M
- Mongkolsapaya J
- Screaton G
- et al.
Preligand assembly domain-mediated ligand-independent association between TRAIL receptor 4 (TR4) and TR2 regulates TRAIL-induced apoptosis.
] indicating that in T lymphocytes, decoy receptors mediate resistance to TRAIL-induced apoptosis by creating mixed functional-decoy receptor complexes that do not efficiently trigger TRAIL signaling. The health status of T cells also was assessed by evaluating their metabolic activity after exposure to increasing doses of sTRAIL released by AD-MSC in CM at different time points (24, 48 and 72 hours). A significant but slight reduction in T-cell metabolism was observed at 24 h with 50–2500 pg/mL sTRAIL, but this trend was completely reversed with both CM from AD-MSC sTRAIL with the greatest sTRAIL concentration (containing 5000 pg/mL of sTRAIL) and from AD-MSC EMPTY and at later time points (48 and 72 h). This suggests that, similar to what was observed for T-cell proliferation, lymphocyte metabolism may be more influenced by trophic factors secreted by MSC than by the toxic impact of sTRAIL. More, these data suggest that non-activated T cells would be resistant to sTRAIL released by engineered AD-MSC thanks to the high expression of TRAIL-mediated apoptosis inhibitors, such as cFLIP, as reported for the wild-type form of TRAIL [
36- Clancy L
- Mruk K
- Archer K
- Woelfel M
- Mongkolsapaya J
- Screaton G
- et al.
Preligand assembly domain-mediated ligand-independent association between TRAIL receptor 4 (TR4) and TR2 regulates TRAIL-induced apoptosis.
,
37Activated human NK and CD8+ T cells express both TNF-related apoptosis-inducing ligand (TRAIL) and TRAIL receptors but are resistant to TRAIL-mediated cytotoxicity.
,
38- Wendling U
- Walczak H
- Dörr J
- Jaboci C
- Weller M
- Krammer PH
- et al.
Expression of TRAIL receptors in human autoreactive and foreign antigen-specific T cells.
].
To further investigate whether the cellular crosstalk between MSC and WBCs could modify TRAIL sensitivity in leucocytes, we established co-culture experiments. Monocyte viability was confirmed as not being influenced by cell-to-cell interactions with engineered AD-MSC secreting sTRAIL despite the high expression of DR5 registered by FACS analysis. On T lymphocytes, we instead observed a slight increase in cell death at greater E:T doses (E:T ratios of 1:1 and 10:1) compared with CTL and co-culture with AD-MSC EMPTY. This modest (lower than 20%) but significant reduction in T-cell viability registered at the 10:1 E:T ratio is likely due to sTRAIL secreted by AD-MSC, since it was not observed in co-culture with AD-MSC EMPTY. In addition, TRAIL-mediated apoptosis did not selectively affect T-cell subtypes, since no significant variations in CD4+ versus CD8+ cell ratios were observed in co-culture with gene-modified AD-MSC and lymphocytes alone. The reason for the fact that the same amount of sTRAIL is not having any cytotoxic impact on T cells when used as CM from AD-MSC sTRAIL versus coculture may be in the presence of AD-MSC cytokines able to sensitize T cells to TRAIL apoptosis during cell-to-cell interaction. In this sense, it has been reported that IFNγ and TNFα enhance TRAIL sensitivity on target cells [
39- Lünemann JD
- Waiczies S
- Ehrlich S
- Wendling U
- Seeger B
- Kamradt T
- et al.
Death ligand TRAIL induces no apoptosis but inhibits activation of human (auto)antigen-specific T cells.
,
40- Park S-Y
- Billiar TR
- Seol D-W.
IFN-γ inhibition of TRAIL-induced IAP-2 upregulation, a possible mechanism of IFN-γ-enhanced TRAIL-induced apoptosis.
,
41- Park S-Y
- Seol J-W
- Lee Y-J
- Cho J-H
- Kang H-S
- Kim I-S
- et al.
IFN-γ enhances TRAIL-induced apoptosis through IRF-1: IFN-γ enhances TRAIL-induced apoptosis.
]. Therefore, cytokine crosstalk accounting for MSC immunomodulatory properties was investigated in co-cultures of T cells and AD-MSC sTRAIL. The levels of the IL-10, TNFα and IFNγ cytokines released by T cells in co-culture with AD-MSC (both sTRAIL and EMPTY) were massively increased at the 10:1 E:T ratio, suggesting that the secretion of these molecules could be indirectly stimulated by MSC, as already reported [
[42]- Liu F
- Hu X
- Zimmerman M
- Waller JL
- Wu P
- Hayes-Jordan A
- et al.
TNFα cooperates with IFN-γ to repress Bcl-xL expression to sensitize metastatic colon carcinoma cells to TRAIL-mediated apoptosis.
]. Therefore, the relevant increase in TNFα and IFNγ amounts registered in the culture medium at the greatest E:T ratios may be responsible for the increased sensitivity of lymphocytes to the apoptotic impact of TRAIL, justifying the enhanced T-cell death after direct exposure to AD-MSC sTRAIL. Similarly, cytokines secreted by MSC, such as VEGF-A and IL-6, also were evaluated. VEGF-A is a key promoter of blood vessel growth and vascular permeability as well as an inhibitor of lymphocyte activation, thus stimulating tumor neoangiogenesis, progression and metastasis [
[43]Secretion of immunoregulatory cytokines by mesenchymal stem cells.
]. We observed the most relevant increase in both VEGF-A and IL-6 secretion per AD-MSC at the 1:30 E:T ratio after 72 h of co-culture with T lymphocytes, which is probably related to the release of TNFα and IFNγ by T cells, as shown by Nagineni
et al. [
44- Marti LC
- Pavon L
- Severino P
- Sibov T
- Guilhen D
- Moreira-Filho CA.
Vascular endothelial growth factor-A enhances indoleamine 2,3-dioxygenase expression by dendritic cells and subsequently impacts lymphocyte proliferation.
,
45- Najar M
- Rouas R
- Raicevic G
- Boufker HI
- Lewalle P
- Meuleman N
- et al.
Mesenchymal stromal cells promote or suppress the proliferation of T lymphocytes from cord blood and peripheral blood: the importance of low cell ratio and role of interleukin-6.
,
46- Nagineni CN
- William A
- Cherukuri A
- Samuel W
- Hooks JJ
- Detrick B.
Inflammatory cytokines regulate secretion of VEGF and chemokines by human conjunctival fibroblasts: role in dysfunctional tear syndrome.
]. Moreover, it is known that IL-6 (alone or in combination with TNFα) can induce VEGF-A secretion by AD-MSC through the ERK, JNK and PI3K pathways [
[46]- Nagineni CN
- William A
- Cherukuri A
- Samuel W
- Hooks JJ
- Detrick B.
Inflammatory cytokines regulate secretion of VEGF and chemokines by human conjunctival fibroblasts: role in dysfunctional tear syndrome.
]. Therefore, we highlighted the cytokine crosstalk between T cells and AD-MSC sTRAIL in co-culture. In particular, AD-MSC induced IL-10, TNFα and IFNγ secretion by T lymphocytes and TNFα and IFNγ, in turn, promoted VEGF-A and IL-6 release by AD-MSC [
[47]- Herrmann JL
- Weil BR
- Abarbanell AM
- Wang Y
- Poynter JA
- Manukyan MC
- et al.
IL-6 and TGF-α costimulate mesenchymal stem cell vascular endothelial growth factor production by ERK-, JNK-, and PI3K-mediated mechanisms.
].
To conclude, these data demonstrate that healthy WBCs are refractory to the proapoptotic effect displayed by sTRAIL, confirming an immunological safety and a clinical feasibility of an anticancer gene therapy strategy based on AD-MSC expressing sTRAIL.
Author Contributions
Conception and design of the study: GC, MDa, GG and MD. Acquisition of data: GC, MDa, VM, GG, MP, CC, GN, MCS, AM and AD. Analysis and interpretation of data: GC, MDa, GG, MP and CC. Drafting or revising the manuscript: GC, MDa, GG and MD. All authors have approved the final article.
Acknowledgments
sTRAIL vector has been generously made available by Rigenerand Srl, now EVOTEC (Modena) srl.
Article info
Publication history
Published online: April 01, 2023
Accepted:
February 15,
2023
Received:
September 1,
2022
Copyright
© 2023 International Society for Cell & Gene Therapy. Published by Elsevier Inc.