Abstract
Introduction: Hodgkin lymphoma (HL) is deficient in major histocompatibility complex class I, rendering it susceptible to antitumoral immunity by natural killer (NK) cells. Despite the functional impairment of PD-1+ NK cells in HL, the underlying mechanisms of NK cell dysfunction remain unclear. Methods: This study involved 14 HL patients and SNK10/KHYG-1 cell lines to assess NK cell activation against cancer cells. Activation was measured through transcript (PCR) and protein expression (flow cytometry). Regulatory mechanisms associated with IRE1α activation were validated through knockdown and luciferase reporter assays. Results: Our findings reveal a novel role for IRE1α-endonuclease in fine-tuning NK cell effector functions by orchestrating the XBP1s/microRNA-34a-5p/PD-1 axis. When NK cells encounter cancer cells, IRE1α endonuclease activates the decay of microRNA-34a-5p, resulting in increased expression of XBP1s and PD-1. IRE1α-endonuclease activation enhances NK cell functions while promoting PD-1 expression. In turn, PD-1 is directly regulated by microRNA-34a-5p, which binds to the 3′UTR of PD-1 transcript to repress PD-1 protein on the NK cell surface. Importantly, IRE1α-pathway activation is impaired in NK cells from HL patients. Conclusion: The IRE1α endonuclease emerges as a key player, simultaneously regulating the XBP1s/microRNA-34a-5p/PD-1 axis in NK cells, a process disrupted in HL. Targeting the IRE1α-pathway holds promise as a therapeutic strategy to optimize NK cell functions in Hodgkin lymphoma treatments.
Plain Language Summary
Natural killer (NK) cells play a critical role in our innate immune defense against tumors without prior exposure to cancer cells and, therefore, make an attractive instrument to harness antitumoral immunity as novel immunotherapies. Hodgkin lymphoma (HL) typically lacks major histocompatibility complex class I which is a requirement for an antigen-specific recognition of effector T cells but not for the NK cell antitumoral function. NK cells are dysfunctional in HL patients mainly due to upregulation of immune checkpoint PD-1 expression that inhibits immune cells effector functions. However, the mechanisms underpinning PD-1 expression and NK cell dysfunction remain unknown. In this study, we uncovered the mechanism by which immune checkpoint PD-1 expression is upregulated upon activation of the endonuclease IRE1α to rapidly change the abundance of key regulatory molecules including a small noncoding regulatory RNA (microRNA-34a) and the transcription factor XBP1. The IRE1α endonuclease plays a dual role in regulating the XBP1/miRNA-34a axis and PD-1 expression within NK cells, which is disrupted in patients with HL. IRE1α pathway in NK cells represents a potential target to optimize NK cell immune functions in future HL treatments.
Introduction
It is established that PD-1+ natural killer (NK) cells are functionally impaired in classical Hodgkin lymphoma (HL) [1]. However, NK cell dysfunction remains poorly characterized in this disease. NK cells are an important subset of innate effector cells that possess the unique ability to lyse malignant cells without previous sensitization [2, 3]. Until recently, attention on immune effector cell function in classical HL has largely focused on cytotoxic T lymphocytes (CTL), in part because relative to CTL the proportion of NK cells within malignant lymph nodes are very modest. However, whereas only a small fraction of CTL will express the relevant T-cell receptor capable of recognizing a malignant cell in an antigen-specific manner, the majority of NK cells are able to exert antitumoral cytotoxicity. Furthermore, their cytotoxic capabilities allow them to kill tumor cells even at relatively low ratios [4]. For these reasons, NK cells have recently attracted attention for their potential use as immune-based therapies [5].
A well-characterized immune-evasion strategy in HL includes copy number gain of Janus kinase signaling and Epstein-Barr virus-driven gene amplification of programmed cell death protein ligand 1 (PD-L1) and PD-L2 ligands in the malignant Hodgkin Reed-Sternberg (HRS) cells [6, 7]. Interaction between PD-L1/PD-L2 and their cognate receptor PD-1 on immune effector cells has been shown to dampen antitumor activity [8]. Although blockade of the PD-1 axis with anti-PD-1 antibodies has proven clinical utility in relapsed/refractory HL [9, 10], relapses frequently occur, indicating the need to mobilize additional immune effector mechanisms. Even though HL diseased lymph nodes contain few NK cells, several lines of evidence suggest that HL is a highly attractive disease to harness peripheral NK cells to the lymphoid circulation. In the missing-self hypothesis, NK cells preferentially lyse cells with reduced or lost expression of major histocompatibility complex class I (MHC-I) molecules while sparing cells with intact MHC-I expression [11]. Notably, the HRS cells in HL have frequently lost MHC-I on their cell surface [12]. It is known that NK cells are dysregulated in HL, and that PD-1 blockade partially abrogates NK cell dysfunction [1] and appears to synergize with bi-specific NK cell engagers [13]. Among relapsed/refractory patients with HL, the number of circulating NK cells is associated with response to PD-1 blockade [14], which is associated with a peripheral immune signature that includes an abundance of activated NK cells [15]. Intriguingly, in responsive patients with HL receiving PD-1 blockade, shortly after treatment the numbers of peripheral blood circulating NK cells are reduced, potentially consistent with their migration to the lymphoid circulation [16]. Furthermore, although the transcriptional mechanisms that regulate PD-1 expression in NK cells are of great importance and interest, they are not yet well understood. Elucidating these mechanisms may enable novel NK cell-driven immunotherapeutic strategies to be pursued.
Inositol-requiring protein 1α (IRE1α) is a serine/threonine-proteinase/endoribonuclease stress sensor that operates within the unfolded protein response system. Under endoplasmic reticulum (ER) stress, IRE1α undergoes oligomerization and subsequent autophosphorylation that triggers its endonuclease activity. The latter enables processing of its principal substrate, X-Box Binding Protein 1 (XBP1) mRNA, via excision of a 26-nucleotide intron and generation of the active form, the transcription factor XBP1-spliced (XBP1s) [17]. The IRE1α pathway is a highly evolutionarily conserved cell signaling pathway with multiple roles in both innate and adaptive immune cells including plasma cells, dendritic cells, eosinophils, macrophages, T cells, and NK cells [18‒25]. Within these immune cell subsets, the functional role of the IRE1α pathway is highly cell context-specific.
Additionally, it has been established in cell-free systems that the IRE1α endonuclease recognizes a consensus sequence (CUGCAG) motif that enables it to cleave selected microRNAs (i.e., miR-17, miR-34a, miR-96, and miR-125b) during ER stress [26, 27]. MicroRNAs (miRNAs) function in part by targeting the 3′ untranslated regions (3′UTRs) of messenger RNAs (mRNAs) for degradation and/or translational repression to “fine-tune” expression of various proteins. Of particular interest within an immune context is miR-34a-5p, which binds to the 3′UTR of PD-L1 in the malignant cells of various solid malignancies and blood cancers [28‒30]. The regulation of the miR-34a-5p/PD-L1 axis by diverse stimuli such as the p53 pathway and Epstein-Barr virus within a range of malignant cell types strongly implicates it as an important regulatory hub that can be manipulated for therapeutic benefit [28‒31].
Interestingly, miR-34a-5p is also expressed by a range of immune effector cells including dendritic cells, macrophages, mast cells, B cells, and T cells [32‒35]. Importantly, miR-34a-5p is a key regulator of multiple inhibitory/activating receptors and chemokines in NK cells [36]. Recently, it has been shown that the IRE1α pathway mediates NK cell responses toward viral infections and murine melanoma tumor models in vivo, as well as cytotoxic function and proliferation of human NK cells from healthy blood donors [24, 25]. To our knowledge, there are minimal data on the functional role of the IRE1α pathway in primary NK cells (pNK cells) from patients with human malignancies, and the role of a potential IRE1α-miR-34a-5p axis in NK cells remains unexplored. In the current study, we sought to investigate the role of the IRE1α pathway on effector function and PD-1 expression in NK cells of patients with HL. Results point to IRE1α endonuclease playing a dual role in regulating the XBP1/miRNA-34a axis and PD-1 expression within NK cells, which is disrupted in patients with HL.
Methods
NK Cell Stimulation Using Cell Lines
NK cell lines used were SNK10 (chronic active Epstein-Barr virus) and KHYG-1 (NK cell leukemia). Target cell lines were HDLM2 (HL) from a patient with poor-risk (elderly, advanced stage disease), KMH2 (HL) (chemo-refractory disease), and K562 (HLA-deficient erythroleukemia). Effector:Target (E:T) ratios used in experimental assays were 1:1. NK cell lines were typically stimulated for 2 h (K562) or 8 h (KMH2 and HDLM2). All RT-qPCR assays to measure gene expression related to activated NK cells were performed with NK cells following isolation from the cocultures with target cancer cells (NK cell isolation kit, Miltenyi Biotec).
IRE1α-Pathway Activation and Inhibition Assays
The IRE1α pathway was activated by treatment with thapsigargin (TG) and was inhibited by using small-molecule inhibitors such as 8-formyl-7-hydroxy-4-methylcoumarin (4µ8c) or 6-bromo-2-hydroxy-3-methoxybenzaldehyde (6-bromo). Knockdown of IRE1α expression was performed using Dicer-substrate short-interfering RNA (Integrated DNA technologies) as outlined in the online supplementary material (for all online suppl. material, see https://doi.org/10.1159/000536044).
Identification and Validation of miRNA Target Gene Regulation
Putative target sites in the 3′UTR region of XBP1 and PD-1 were predicted for IRE1α-regulated miRNAs (miR-17, miR-34a, miR-96, and miR-125b) using miRanda algorithm. Mature miRNA sequences for all four miRNAs were downloaded from the miRBase database (Release 22) [38], and the XBP1 and PD-1 transcript sequences were retrieved from GENCODE (Release 21 Reference Human Genome GRCh38) [39]. Only predicted target sites with stringent parameters (free energy ≤−18 and miRanda score ≥140) were considered for further experimental validations (online suppl. Table S1). Functional validation of miRNA binding sites including the dual luciferase assay and transfection of miRNA mimics is outlined in more details in the online supplementary material.
Identification and Validation of XBP1 Target Sites in PD-1 Gene
The computational prediction of XBP1 binding sites was performed on regulatory regions of the PDCD1 (PD-1) gene found to be active in human NK cells (ENCODE; www.encodeproject.org/experiments/ENCSR808HWS/). We used MEME motif discovery program [40] for a de novo discovery of XBP1 binding sites on sequences enriched in regions experimentally determined by XBP1-ChIP assay and RNA-seq (as described elsewhere [41] and datasets available at ArrayExpress: E-MTAB-6327, E-MTAB-6894, and E-MATB-7104). We identified overrepresented DNA patterns (or motifs) in the XBP1 binding regions of the top 500 most upregulated genes in activated TH2 (high levels of XBP1s) versus naïve TH2 (889 peaks) and the top 500 most downregulated genes in activated TH2 versus naïve TH2 (595 peaks). Two putative XBP1 motifs were found: one canonical motif similar to the one described in JASPAR database (MA0844) and a novel variant XBP1 motif not yet described in any available database or publication. TAMO package [42] was used for the identification of putative XBP1 binding sites in the promoter regions of PD-1 gene. Only putative binding sites at least 70% similarity to canonical or variant XBP1 motifs were selected.
A luciferase reporter system (pGL3 basic vector, Promega) was used to validate the direct interaction of XBP1s and the predicted promoter regions of PD-1 as well as the individual XBP1s binding sites identified within PD-1 promoter regions. More details of methods are described in the supplemental material.
Patient Samples
Pre-therapy blood samples from 14 patients with HL were collected before therapy (median age 37 years, range 16–73; F:M ratio 9:5; 6 stage I-II, 6 stage III-IV, 2 stage unknown; 10 nodular sclerosing, 1 lymphocyte rich, 3 classical HL otherwise not classified); and from 10 healthy participants (median age 37 years, range 25–48, F:M ratio 5:5). Peripheral blood mononuclear cells were isolated and cryopreserved, as described previously [43]. This study conformed to the Declaration of Helsinki, and informed consent was provided by all participants in accordance with participating hospitals/research institute Human Research Ethics Committee guidelines.
NK Cell Functional Assays
pNK cells were isolated from cryopreserved peripheral blood mononuclear cells and expanded as previously described [1]. Flow cytometry, NK cell migration, NK cell immune synapse (NKIS) formation, and cytotoxicity are outlined in the supplemental material. For PD-1 blockade experiments, NK cells were cultured for 72 h with 10 µg/mL of pembrolizumab™ (or IgG4 control), prior to the addition of targets. At the completion of the culture, the KHYG-1 cells or pNK cells were collected and incubated with an equal number of targets. Assays were performed in triplicate. More details are provided in the online supplementary material.
Statistical Analysis
All statistical analysis was done using GraphPad Prism 7.00 (GraphPad Software). Comparison of measurements was performed using paired 2-tailed t tests (except when stated otherwise in figure legends). Only p < 0.05 was considered significant.
Results
Activation of the IRE1α Endonuclease in NK Cells Reduces Levels of miR-34a-5p
To confirm that the IRE1α pathway can be activated in NK cells in response to blood cancer cell lines, we coincubated the NK cell-line SNK10 with the HRS cell-lines HDLM2 (MHC-I deficient) or KMH2 (MHC-I intact), and the NK cell-sensitive target leukemia cell line K562 (MHC-I and II deficient). Splicing of the transcription factor XBP1 to its active form XBP1s was used as the functional readout to indicate IRE1α-endonuclease activation. The mRNA expression of XBP1s in K562 activated SNK10 cells increased by at least 2-fold in comparison to unstimulated cells (shown in Fig. 1a). Similarly, a significant increase in XBP1s protein expression was observed in stimulated SNK10 cells (shown in Fig. 1b; online suppl. Fig. S1). However, the increase of XBP1s protein expression in SNK10 cells stimulated with both HL cell lines (KMH2 and HDLM2) was far more pronounced (up to ∼5–6-fold) than with K562 leukemia cells (∼2-fold; shown in Fig. 1b; online suppl. Fig. S1).
IRE1α pathway downregulates miR-34a-5p in NK cells following activation by HRS and leukemia cells. a IRE1α-pathway activation was confirmed by qRT-PCR analysis of the XBP1 spliced isoform (XBP1s) upon SNK10 NK cell co-incubation for at least 2 h with leukemia (K562) or HL (HDLM2 and KMH2). The bars show changes in the expression of XBP1s in SNK10 cells activated by target cells, relative to resting SNK10 cells (shown by the dotted line; B2M is used as endogenous control, unstim condition n = 4; other n = 3). b The expression of XBP1s protein is also upregulated in NK cells stimulated with different target cells (relative to unstimulated NK cells). Graph shows the mean ± SEM. FACS data of % XBP1s + SNK10 cells (unstim condition n = 6, other n = 3). c qRT-PCR analysis confirms that miR-34a-5p expression is downregulated in SNK10 cells stimulated with all three cancer cell lines (RNAU6 used as endogenous control and SNK10 unstimulated as reference condition). A one-tailed paired t test was used for statistical analysis (n = 3). d HEK293T cells treated with the ER stressor TG, to activate IRE1α pathway, significantly increased XBP1s mRNA while suppressing miR-34a-5p expression, showing that this mechanism is conserved beyond immune cells. Unpaired t test two-tailed used for statistical analysis (n = 4).
IRE1α pathway downregulates miR-34a-5p in NK cells following activation by HRS and leukemia cells. a IRE1α-pathway activation was confirmed by qRT-PCR analysis of the XBP1 spliced isoform (XBP1s) upon SNK10 NK cell co-incubation for at least 2 h with leukemia (K562) or HL (HDLM2 and KMH2). The bars show changes in the expression of XBP1s in SNK10 cells activated by target cells, relative to resting SNK10 cells (shown by the dotted line; B2M is used as endogenous control, unstim condition n = 4; other n = 3). b The expression of XBP1s protein is also upregulated in NK cells stimulated with different target cells (relative to unstimulated NK cells). Graph shows the mean ± SEM. FACS data of % XBP1s + SNK10 cells (unstim condition n = 6, other n = 3). c qRT-PCR analysis confirms that miR-34a-5p expression is downregulated in SNK10 cells stimulated with all three cancer cell lines (RNAU6 used as endogenous control and SNK10 unstimulated as reference condition). A one-tailed paired t test was used for statistical analysis (n = 3). d HEK293T cells treated with the ER stressor TG, to activate IRE1α pathway, significantly increased XBP1s mRNA while suppressing miR-34a-5p expression, showing that this mechanism is conserved beyond immune cells. Unpaired t test two-tailed used for statistical analysis (n = 4).
In addition to activating XBP1s splicing, it has been shown in cell-free systems that the IRE1α pathway also plays a role in the degradation by cleavage of a subset of CUGCAG motif-containing miRNAs including miR-34a [27]. Next, we sought to establish if degradation of miR-34a is operative within NK cells. First, we measured miR-34a-5p expression by qRT-PCR in SNK10 cells stimulated with these three different target cell lines: K562, KMH2, and HDLM2. This showed significant downregulation of miR-34a-5p in SNK10 cells after stimulation with all three targets cell lines (shown in Fig. 1c). To confirm the mechanism of IRE1α-mediated regulation of XBP1s and miR-34a-5p is conserved in cells other than NK cells, we tested IRE1α activation in HEK293T cells using a well-known ER stress inducer TG. Activation of the IRE1α pathway in HEK293T cells by TG downregulated miR-34a-5p expression, whilst XBP1s isoform was upregulated (shown in Fig. 1d). These results strongly suggest that IRE1α endonuclease activity plays a dual role in the control of expression of XBP1s and miR-34a-5p within NK cells.
miR-34a-5p Targets the 3′UTRs of XBP1 and PD-1 mRNAs to Suppress Their Expression
Although miR-34a-5p is known to regulate multiple inhibitory/activating receptors and chemokines in NK cells [36], its potential role in regulating PD-1 expression in NK cells remains unknown. Similarly, although it has also been shown that miR-34a-5p can target XBP1 in neuronal cells [44], the role of this miRNA in NK cells is unexplored. Therefore, to examine this further, we looked for putative binding sites for the miRNAs in the 3′UTR of XBP1 and PD-1 mRNA. Next, we performed a computational analysis to identify putative target sites for all four miRNAs known to be regulated by IRE1α (miR-17, miR-34a, miR-96, and miR-125b) [27], in the 3′UTR region of XBP1 and PD-1 mRNAs. Adopting a stringent criterion for miRNA target site prediction (miRanda score ≥140 and free energy <−18 kcal/mol), we found that only miR-34a-5p had putative target sites for the 3′UTR of XBP1 and PD-1 mRNAs (online suppl. Table S1). Therefore, efforts were focused on validating binding of miR-34a to XBP1 (two target sites) and PD-1 (one target site) in NK cells.
To validate the three putative target sites of miR-34a-5p in the 3′UTR of XBP1 and PD-1 transcripts, we performed a dual luciferase reporter assay [45]. We tested the regulatory effect of miR-34a-5p mimics on the expression of Renilla Luciferase (Rluc) fused with the predicted target site found in the 3′UTR of XBP1 (shown in Fig. 2a, b) or PD-1 (shown in Fig. 2c) using HEK293T cells to host the dual luciferase expression vector. For this assay, Caenorhabditis elegans miR-67-5p (cel-miR-67-5p), with no putative binding sites found in XBP1 and PD-1 3′UTRs, was used as a negative control to confirm the specific repression of Rluc (carrying candidate target sites) by miR-34a-5p (shown in Fig. 2a–c). We then performed a rescue assay by quantifying the expression of Rluc carrying the miR-34a-5p target sites after treatment with miR-34a-5p mimics and rescue with miR-34a-5p antagomir (anti-miR-34a-5p) compared to negative control antagomir (anti-miR NC, a nontargeting negative control for antagomir assays that do not recognize any sequences in human transcriptomes including miR-34a-5p sequence). As expected, normalized Rluc signal was significantly reduced by the presence of miR-34a-5p mimics (plus anti-miR NC; shown in online suppl. Fig. S2) when compared to the rescue condition (plus anti-miR-34a-5p; shown in online suppl. Fig. S2). In addition, we performed a biotinylated miR-34a-5p/mRNA pull-down assay to confirm a significant enrichment of XBP1 and PD-1 mRNAs (shown in Fig. 2d).
miR-34a-5p directly binds to the 3′UTR of XBP1 and PD-1 transcripts to suppress their expression. miR-34a-5p binding sites, site 1 (a) and site 2 (b) predicted in the 3′UTR of XBP1 and (c) PD-1, were experimentally validated by the dual-luciferase reporter assay. HEK293T cells co-transfected with miRNA mimics and plasmid construct containing predicted miR-34a-5p binding site showed a significant reduction in the luciferase signal with miR-34a-5p mimics compared to negative control (cel-miR-67-5p) (n = 4). d XBP1 and PD-1 mRNAs were significantly enriched in KHYG-1 cell mRNAs pull-down assay using biotinylated miR-34a-5p mimics. SDHA gene (nontarget) was used as negative control (n = 4). e PD-1 and XBP1 mRNA expression in KHYG-1 cell line was significantly decreased upon miR-34a-5p mimics treatment compared to negative control cel-miR-67-5. A one-tailed paired t test was used for statistical analysis (n = 3). f qRT-PCR analysis of miR-34a-5p and PD-1 mRNA in FACS-sorted PD-1high KHYG-1 cells (in contrast to PD-1low KHYG-1 cells) suggests a negative correlation between miR-34a-5p and PD-1 expression (18SrRNA and GAPDH used as endogenous controls for miR-34a-5p and PD-1, respectively). Unpaired t test two-tailed used for statistical analysis (n = 3).
miR-34a-5p directly binds to the 3′UTR of XBP1 and PD-1 transcripts to suppress their expression. miR-34a-5p binding sites, site 1 (a) and site 2 (b) predicted in the 3′UTR of XBP1 and (c) PD-1, were experimentally validated by the dual-luciferase reporter assay. HEK293T cells co-transfected with miRNA mimics and plasmid construct containing predicted miR-34a-5p binding site showed a significant reduction in the luciferase signal with miR-34a-5p mimics compared to negative control (cel-miR-67-5p) (n = 4). d XBP1 and PD-1 mRNAs were significantly enriched in KHYG-1 cell mRNAs pull-down assay using biotinylated miR-34a-5p mimics. SDHA gene (nontarget) was used as negative control (n = 4). e PD-1 and XBP1 mRNA expression in KHYG-1 cell line was significantly decreased upon miR-34a-5p mimics treatment compared to negative control cel-miR-67-5. A one-tailed paired t test was used for statistical analysis (n = 3). f qRT-PCR analysis of miR-34a-5p and PD-1 mRNA in FACS-sorted PD-1high KHYG-1 cells (in contrast to PD-1low KHYG-1 cells) suggests a negative correlation between miR-34a-5p and PD-1 expression (18SrRNA and GAPDH used as endogenous controls for miR-34a-5p and PD-1, respectively). Unpaired t test two-tailed used for statistical analysis (n = 3).
To further confirm the regulatory effect of miR-34a-5p on XBP1 and PD-1 expression, we treated the PD-1 expressing NK cell-line KHYG-1 with miR-34a-5p mimics. There was a significant knockdown of XBP1 and PD-1 mRNA in miR-34a-5p mimics-treated KHYG-1 cells compared to negative control treatment (cel-miR-67-5p; shown in Fig. 2e). These results confirm that miR-34a-5p can suppress both XBP1 and PD-1 mRNAs in NK cells. To further confirm the association between miR-34a-5p and PD-1, we assessed the correlative association between miR-34a-5p and PD-1 in vitro by using the PD-1 expressing NK cell-line KHYG-1. PD-1high and PD-1low KHYG-1 cells were sorted with fluorescent-activated cell sorter. Relative quantities of miR-34a-5p and PD-1 transcripts in the two KHYG-1 populations were assessed by qRT-PCR. In agreement with our hypothesis of PD-1 repression by miR-34a-5p, we observed a significant downregulation of miR-34a-5p expression in PD-1high KHYG-1 cells when compared to PD-1low KHYG-1 cells (shown in Fig. 2f).
The IRE1α Pathway Plays a Key Role in the Downregulation of miR-34a-5p and Upregulation of PD-1 Expression in NK Cells
A time course experiment for IRE1α blockade using DsiIRE1 (Dicer-substrate RNA interference designed to knockdown IRE1α expression) assay showed a significant increase in miR-34a expression in NK cells that is inversely proportional to IRE1α expression in keeping with an IRE1α-negative regulation of miR-34a-5p. For this experiment, we again used the PD-1 expressing NK cell-line KHYG-1 to interrogate the role of IRE1α in the regulation of miR-34a and PD-1. First, we confirmed the inhibitory effect of DsiIRE1 on IRE1α mRNA with maximum knockdown of IRE1α mRNA observed at 12 h post-DsiIRE1 treatment (shown in Fig. 3a), although IRE1α knockdown lasted for 24 h post treatment. As expected, the two arms of miR-34a, miR34a-5p and miR-34a-3p, showed an inverted association with IRE1α mRNA expression levels (shown in Fig. 3b, c). The expression of both mature sequences of miR-34a hairpin (miR-34a-5p and miR-34a-3p) peaked at 12 h and remained significantly upregulated until 24 h post DsiIRE1 treatment. In contrast, the cells with IRE1α inhibited by DsiIRE1 demonstrated significantly reduced PD-1 mRNA expression at 12 h post treatment, which remained downregulated until 24 h post treatment (shown in Fig. 3d). Therefore, we further validated the effect of IRE1α on PD-1 protein expression using TG (IRE1α activator) and DsiIRE1 (IRE1α inhibitor) treatments in the KHYG-1 cells. Our experiments confirmed a significant reduction in PD-1 protein expression in the cells treated with DsiIRE1 at 12 h and more so at 24 h (shown in Fig. 3e). On the other hand, IRE1α activation by TG treatment showed a stimulating effect on PD-1 protein expression and this effect was significant even after 24 h in the KHYG-1 cells treated with DsiIRE1 compared to negative control DsiNC (shown in Fig. 3f). Furthermore, we also tested if IRE1α knockdown affects XBP1s expression. We found that XBP1s expression is significantly downregulated at 12 h post-DsiIRE1 treatment (followed by 2 h of TG activation; online suppl. Fig. S3A, B) when IRE1α is silenced. Moreover, XBP1s expression is positively correlated with IRE1α transcription (Pearson’s correlation coefficient = 0.98, p = 0.02; Fig. S3C), providing additional evidence that IRE1α transcription is directly associated with XBP1s splicing output.
IRE1α suppresses miR-34a expression and increases PD-1 expression in NK cells. a Time course experiment for IRE1α blockade by DsiIRE1 in KHYG-1 cells. The highest significant reduction in IRE1α mRNA was observed after 12 h of DsiIRE1 treatment compared to negative control DsiRNA (DsiNC) treatment in KHYG-1 cells (n = 3). The expression of both miR-34a arms: miR34a-5p (b) and miR-34a-3p (c) was significantly increased in the KHYG-1 cells treated with DsiIRE1. Each miR-34a arm showed an opposite direction to IRE1α mRNA (n = 3). d PD-1 mRNA altered in the same direction as IRE1α mRNA. PD-1 mRNA significantly reduced at 12 h post DsiIRE1 treatment in KHYG-1 cells and this reduction persisted up to 24 h (n = 3). e IRE1α blockade significantly reduced PD-1 protein expression in KHYG-1 cells after 12 h of DsiIRE1 treatment and this effect lasted up to 24 h. f IRE1α activation by TG treatment significantly increased PD-1 expression in KHYG-1 cells when pretreated with DsiNC but not when IRE1α was downregulated by DsiIRE1. This effect was present in the cells up to 24 h after TG treatment. n = 3 for each experiment. DsiRNA, Dicer-substrate short-interfering RNA.
IRE1α suppresses miR-34a expression and increases PD-1 expression in NK cells. a Time course experiment for IRE1α blockade by DsiIRE1 in KHYG-1 cells. The highest significant reduction in IRE1α mRNA was observed after 12 h of DsiIRE1 treatment compared to negative control DsiRNA (DsiNC) treatment in KHYG-1 cells (n = 3). The expression of both miR-34a arms: miR34a-5p (b) and miR-34a-3p (c) was significantly increased in the KHYG-1 cells treated with DsiIRE1. Each miR-34a arm showed an opposite direction to IRE1α mRNA (n = 3). d PD-1 mRNA altered in the same direction as IRE1α mRNA. PD-1 mRNA significantly reduced at 12 h post DsiIRE1 treatment in KHYG-1 cells and this reduction persisted up to 24 h (n = 3). e IRE1α blockade significantly reduced PD-1 protein expression in KHYG-1 cells after 12 h of DsiIRE1 treatment and this effect lasted up to 24 h. f IRE1α activation by TG treatment significantly increased PD-1 expression in KHYG-1 cells when pretreated with DsiNC but not when IRE1α was downregulated by DsiIRE1. This effect was present in the cells up to 24 h after TG treatment. n = 3 for each experiment. DsiRNA, Dicer-substrate short-interfering RNA.
Activation of the IRE1α Pathway Upregulates PD-1 Transcription via XBP1s Direct Binding to Target Sites in PD-1 Promoter Regions
We then investigated whether XBP1s putatively binds to the promoter and/or enhancer regions of PD-1 gene. Putative XBP1 target sites were identified using the canonical XBP1 binding motif as described in the JASPAR database (MA0844.1) [46] and a novel variant XBP1 motif that we found in a de novo discovery cohort using XBP1-ChIP assay data previously published [41] (shown in Fig. 4a; see Methods for details). The canonical XBP1 binding motif reported in JASPAR database is derived from published collections of experimentally defined transcription factor binding sites, whereas the novel variant XBP1 motif is not reported anywhere in the literature. Next, similar occurrences (at least 70% of similarity) of these two alternative XBP1 motifs were searched in active regulatory regions of PD-1 previously determined by ATAC-seq (Assay for Transposase-Accessible Chromatin using sequencing) in primary human NK cells (ENCODE; www.encodeproject.org/experiments/ENCSR808HWS/). Two canonical XBP1 target sites were identified in downstream regions of PD-1 gene, while several putative sites were found for the new variant XBP1 motif distributed across the upstream, intronic, and downstream regions (shown in Fig. 4b).
XBP1s regulates PD-1 transcription via target sites in PD-1 promoter regions. a Two putative XBP1 motifs discovered in regulatory regions bound to XBP1 previously published [41]. We identified an XBP1 motif similar to the canonical motif as described in the JASPAR database (MA0844) and a novel variant motif not previously published. b Genomic organization of PD-1 gene showing the location of putative XBP1 target sites. Boxes labeled upstream, intronic, and downstream indicate regions selected for experimental validations. Tracks at the bottom of gene diagram depict ATAC-seq data for human NK cells. Red arrows indicate the position of canonical XBP1 target sites and green arrows indicate novel variant XBP1 target sites. Dark grey boxes represent exons and light grey boxes the 3′UTRs. c Multiple XBP1s binding sites were predicted within the upstream, intronic, and downstream regions of PD-1. PGL3 vector containing these putative regulatory sequences in the promoter region of a firefly luciferase gene was transfected into HEK293T cells and then treated with TG for activation of XBP1s. PGL3 vectors containing upstream and downstream regulatory regions showed a significant increase in firefly luciferase expression upon TG treatment compared to DMSO control. d Four different binding sites with novel variant XBP1s binding sites (S1, S2, S3, and S4) were cloned into PGL3 vector promoter region. Three binding sites (S1, S2, and S4) showed a significant increase in firefly expression upon upregulation of XBP1s (by TG treatment) compared to DMSO treatment as negative control. e The binding sites with canonical XBP1s motif (S5 and S6) did not elicit upregulation of firefly luciferase upon XBP1s activation (by TG treatment). A one-tailed paired t test (n = 3) was used for statistical analysis.
XBP1s regulates PD-1 transcription via target sites in PD-1 promoter regions. a Two putative XBP1 motifs discovered in regulatory regions bound to XBP1 previously published [41]. We identified an XBP1 motif similar to the canonical motif as described in the JASPAR database (MA0844) and a novel variant motif not previously published. b Genomic organization of PD-1 gene showing the location of putative XBP1 target sites. Boxes labeled upstream, intronic, and downstream indicate regions selected for experimental validations. Tracks at the bottom of gene diagram depict ATAC-seq data for human NK cells. Red arrows indicate the position of canonical XBP1 target sites and green arrows indicate novel variant XBP1 target sites. Dark grey boxes represent exons and light grey boxes the 3′UTRs. c Multiple XBP1s binding sites were predicted within the upstream, intronic, and downstream regions of PD-1. PGL3 vector containing these putative regulatory sequences in the promoter region of a firefly luciferase gene was transfected into HEK293T cells and then treated with TG for activation of XBP1s. PGL3 vectors containing upstream and downstream regulatory regions showed a significant increase in firefly luciferase expression upon TG treatment compared to DMSO control. d Four different binding sites with novel variant XBP1s binding sites (S1, S2, S3, and S4) were cloned into PGL3 vector promoter region. Three binding sites (S1, S2, and S4) showed a significant increase in firefly expression upon upregulation of XBP1s (by TG treatment) compared to DMSO treatment as negative control. e The binding sites with canonical XBP1s motif (S5 and S6) did not elicit upregulation of firefly luciferase upon XBP1s activation (by TG treatment). A one-tailed paired t test (n = 3) was used for statistical analysis.
We selected three putative regulatory regions, upstream, intronic, and downstream, of the PD-1 gene where chromatin is accessible in human NK cells and putative XBP1 binding sites were predicted (shown in Fig. 4b). All three regulatory regions were amplified from genomic DNA extracted from KHYG-1 cells and cloned into the promoter of a luciferase gene in a PGL3 vector. HEK293T cells were transfected with PGL3 vectors containing the upstream (810 bp), intronic (437 bp), or downstream (280bp) regions. Transfected HEK293T cells were then treated with TG (positive control for XBP1s activation) or DMSO (negative control in which XBP1s is not expressed) and the luciferase activity measured using a luciferase reporter assay (compared to native PGL3 vector carrying basic promoter as background control). The results indicate that XBP1s can significantly promote the expression of luciferase gene carrying the upstream and downstream regions containing XBP1s binding sites found in active chromatin regions nearby PD-1 gene (shown in Fig. 4c).
We then tested a few selected putative XBP1s binding sites (highest similarity to XBP1s motifs) identified within each regulatory region for further validation. The most abundant XBP1s binding site found in these regions was the variant XBP1s motif (shown in Fig. 4b) with the canonical XBP1s motif found only at the downstream region and a more distal accessible region further downstream of PD-1 gene (shown in Fig. 4b). Specific putative XBP1s binding sites found in these regulatory regions were cloned into PGL3 vector. The luciferase expression measured in KHYG-1 cells (DMSO or TG treated) transfected with PGL3 vectors carrying candidate XBP1s binding sites revealed that the variant XBP1s binding sites (S1, S2, and S4) can enhance XBP1s-driven expression of luciferase gene (shown in Fig. 4d), while the canonical XBP1 binding sites (S5 and S6) were not able to elicit any significant change in luciferase expression (shown in Fig. 4e).
The IRE1α Pathway Is Impaired and Associated with Reduced Effector Function in Primary NK Cells of Patients with Hodgkin Lymphoma
To investigate the translational relevance of our findings, our study was extended to pNK cells obtained from patients with HL, a blood cancer in which NK cells expressing PD-1 are known to be functionally impaired [1]. We tested ex vivo nonexpanded pNK cells taken from peripheral blood from patients with HL prior to therapy, to establish the base levels of ex vivo IRE1α-endonuclease activity. XBP1s expression was quantified by flow cytometry in HRS-stimulated pNK cells from patients with HL and compared to age-matched healthy participants. Mononuclear CD3−CD14−CD19−CD56+ pNK cells were enumerated and classified into conventional pNK cell subsets based on CD56 and CD16 expression: CD56brightCD16−ve and CD56dimCD16+. Following HRS stimulation, XBP1s expression was reduced in both CD56brightCD16−ve (shown in Fig. 5a) and CD56dimCD16+ subsets (shown in Fig. 5b) in HL patients, indicating that IRE1α-endonuclease activity (required to effectively process the functional XBP1s isoform) is reduced in pNK cells from cancer patients.
IRE1α pathway is impaired in pNK cells from patients with HL and PD-1 blockade partially restores the IRE1α pathway and pNK cell effector functions. The effect of HRS cells on IRE1α-pathway activation was tested in pre-therapy pNK cells (pNK cells) from patients with HL compared to age-matched healthy participants. PBMCs were incubated for 4 h in the presence of HRS cells (KMH2) admixed at 1:1 ratio. a XBP1s expression in the CD56brightCD16−ve NK cell subset was quantified by flow cytometry data and found to be downregulated in pre-therapy pNK cells from patients with HL (compared to pNK cells from healthy participants). b The absolute numbers of XBP1s+ CD56dim CD16+ NK cell subset was quantified by flow cytometry showing downregulation in pre-therapy pNK cell from patients. Mann-Whitney test (n = 14) used for statistical analysis. c–i The effector functions of NK cells from patients with HL were reduced by the IRE1α inhibitor (4μ8c 60 μm) when compared to control (DMSO and IgG4 isotype), after stimulation with HDLM2 cells for 6 h. PD-1 blockade (α-PD-1) partially restored pNK cell functions. Cytokines (c, d) IFNγ and (e, f) TNFα were assessed by FACS in CD56bright CD16−ve and CD56dim CD16+ cells and were significantly reduced after IRE1α inhibition. PD-1 blockade reversed IFNγ release reduced by IRE1α inhibition in CD56bright CD16−ve subset of NK cells. TNFα release reduced by IRE1α inhibition was reversed by PD-1 blockade in both CD56bright CD16−ve and CD56dim CD16+ NK cell subsets. A paired t test was used for statistical analysis. At the end of a 2-week expansion, NK cells from patients with HL were assessed. g, h Cells were stained with CD107a antibody to assess the level of degranulation in NK cell subsets. IRE1α blockade inhibited degranulation; however, PD-1 blockade did not reverse IRE1α blockade-mediated impairment of NK cell degranulation in (g) CD56brightCD16ve- or (h) CD56dim CD16+ subset of NK cells. i IRE1α blockade inhibited lysis of HRS target cells, but no significant difference in cytotoxicity was observed upon PD-1 blockade. Statistical differences between experimental groups were evaluated by Student’s t test (n = 3). j NKIS formation between NK cell and HRS cells that is impaired by IRE1α blockade is restored after PD-1 blockade with increased F-actin polymerization at the NKIS. Statistical differences between experimental groups were evaluated by unpaired t tests. Histograms show the mean ± SD from 3 independent experiments with 30 conjugates analyzed per experiment. k Representative images of NKIS formation between NK cell (DAPI, blue) and HRS cells (APC, red) showing the expanded NK cells from patients with HL stimulated with HDLM2 for 6 h in the presence of IRE1α inhibitor, PD-1 blockade (α-PD-1) + 4μ8c. Arrows indicate actin accumulation (green) at the NK cell/HRS synapse site. In the control assays, HRS target cells and pNK cells were co-incubated with immunoglobulin (Ig) G4 isotype control. Original magnification ×63. PBMCs, peripheral blood mononuclear cells.
IRE1α pathway is impaired in pNK cells from patients with HL and PD-1 blockade partially restores the IRE1α pathway and pNK cell effector functions. The effect of HRS cells on IRE1α-pathway activation was tested in pre-therapy pNK cells (pNK cells) from patients with HL compared to age-matched healthy participants. PBMCs were incubated for 4 h in the presence of HRS cells (KMH2) admixed at 1:1 ratio. a XBP1s expression in the CD56brightCD16−ve NK cell subset was quantified by flow cytometry data and found to be downregulated in pre-therapy pNK cells from patients with HL (compared to pNK cells from healthy participants). b The absolute numbers of XBP1s+ CD56dim CD16+ NK cell subset was quantified by flow cytometry showing downregulation in pre-therapy pNK cell from patients. Mann-Whitney test (n = 14) used for statistical analysis. c–i The effector functions of NK cells from patients with HL were reduced by the IRE1α inhibitor (4μ8c 60 μm) when compared to control (DMSO and IgG4 isotype), after stimulation with HDLM2 cells for 6 h. PD-1 blockade (α-PD-1) partially restored pNK cell functions. Cytokines (c, d) IFNγ and (e, f) TNFα were assessed by FACS in CD56bright CD16−ve and CD56dim CD16+ cells and were significantly reduced after IRE1α inhibition. PD-1 blockade reversed IFNγ release reduced by IRE1α inhibition in CD56bright CD16−ve subset of NK cells. TNFα release reduced by IRE1α inhibition was reversed by PD-1 blockade in both CD56bright CD16−ve and CD56dim CD16+ NK cell subsets. A paired t test was used for statistical analysis. At the end of a 2-week expansion, NK cells from patients with HL were assessed. g, h Cells were stained with CD107a antibody to assess the level of degranulation in NK cell subsets. IRE1α blockade inhibited degranulation; however, PD-1 blockade did not reverse IRE1α blockade-mediated impairment of NK cell degranulation in (g) CD56brightCD16ve- or (h) CD56dim CD16+ subset of NK cells. i IRE1α blockade inhibited lysis of HRS target cells, but no significant difference in cytotoxicity was observed upon PD-1 blockade. Statistical differences between experimental groups were evaluated by Student’s t test (n = 3). j NKIS formation between NK cell and HRS cells that is impaired by IRE1α blockade is restored after PD-1 blockade with increased F-actin polymerization at the NKIS. Statistical differences between experimental groups were evaluated by unpaired t tests. Histograms show the mean ± SD from 3 independent experiments with 30 conjugates analyzed per experiment. k Representative images of NKIS formation between NK cell (DAPI, blue) and HRS cells (APC, red) showing the expanded NK cells from patients with HL stimulated with HDLM2 for 6 h in the presence of IRE1α inhibitor, PD-1 blockade (α-PD-1) + 4μ8c. Arrows indicate actin accumulation (green) at the NK cell/HRS synapse site. In the control assays, HRS target cells and pNK cells were co-incubated with immunoglobulin (Ig) G4 isotype control. Original magnification ×63. PBMCs, peripheral blood mononuclear cells.
Next, we evaluated if there is still a remnant role for the impaired IRE1α pathway in the regulation of pNK cell effector functions in HL. As the numbers of intratumoral pNK cells in patients with HL are limited, circulating pNK cells from pre-therapy blood samples in patients with HL were expanded in vitro. Then the functional impact of IRE1α blockade (using IRE1α-inhibitor 4μ8c) was tested in the presence of HDLM2 target cells. This showed a reduction of IFNγ (shown in Fig. 5c, d) and TNFα cytokines release (shown in Fig. 5e, f), CD107a degranulation (shown in Fig. 5g, h), HRS target cell killing (shown in Fig. 5i), and F-actin accumulation at the NKIS (shown in Fig. 5j, k) in pNK cells from patients with HL.
These results strongly suggest that IRE1α pathway still retains a residual activity and thus it may be possible to restore a “healthy” homeostasis. As our previous study demonstrates that pNK cell effector functions are significantly improved by the immunotherapy medication Pembrolizumab (a monoclonal antibody designed to target and block PD-1) [1], we sought to determine whether PD-1 blockade restores NK cell effector functions that are reduced due to impairment of IRE1α pathway. Cytokine secretion (IFNγ, TNFα), degranulation (CD107a), HRS target cell killing, and F-actin accumulation at the NKIS were tested in pNK cells from patients with HL. We tested the effect of Pembrolizumab (α-PD-1) on pNK cell functions upon IRE1α inhibition (shown in Fig. 5c–k). PD-1 blockade restored both IFNγ (shown in Fig. 5c, d) and TNFα (shown in Fig. 5e, f) cytokines release in pNK cells (except for IFNγ in CD56dimCD16+). However, PD-1 blockade did not restore degranulation (shown in Fig. 5g, h) and cytotoxicity (shown in Fig. 5i) functions, despite a significant increase in immune synapse formation when PD-1 is blocked (shown in Fig. 5j, k), in pNK cells from patients with HL. Moreover, we observed that pNK cells from patients treated with Pembrolizumab alone have restored IFNγ release (shown in online suppl. Fig. S4A, B) and immune synapse (shown in online suppl. Fig. S4H, I), but none of the other effector functions were enhanced (shown in online suppl. Fig. S4), suggesting that the improvement in NK cell function induced by PD-1 blockade is only partially by rescuing IRE1α-pathway impairment.
In confirmatory experiments, we also measured cytokine secretion, degranulation, and cytotoxicity upon IRE1α blockade on the SNK10 cell line stimulated with K562 and HDLM2 target cells. The results showed a significant reduction in all effector functions including IFNγ release (shown in online suppl. Fig. S5A, B), TNFα release (shown in online suppl. Fig. S5C, D), CD107a degranulation (shown in online suppl. Fig. S5E), and lysis of target cells (shown in online suppl. Fig. S5F), confirming that the IRE1α pathway has an important role in NK cell effector function. Moreover, IRE1α blockade efficiently inhibits XBP1 splicing in NK cells when exposed to all three tested target cells (K562, KMH2, and HDLM2; shown in online suppl. Fig. S5G), further confirming the IRE1α-pathway involvement in XBP1 splicing in NK cells. We also tested if the IRE1α pathway has a role in NK cell movement and synapse formation. SNK10 cell motility was assessed by live cell imaging (shown in online suppl. Fig. S5H). IRE1α-blocked NK cells traveled shorter distances and at lower velocity (shown in online suppl. Fig. S5H).
As a recent study has reported that NK cells can acquire functional PD-1 transferred from cancer cells via trogocytosis [47], we also investigated whether the PD-1 expression detected in the cellular membrane of NK cells used in our assays could have been acquired from malignant HRS cells due to trogocytosis in HL instead of an endogenous expression in NK cells. First, PD-1 expression was tested in a range of HL cell lines and none of the tested lines expressed PD-1 mRNA or protein (shown in online suppl. Fig. S6A–C). Then, we tested the expression of PD-1 and CD30 (a marker of HRS cells) on pNK cells in pre-therapy blood samples in patients with HL. The results demonstrate that pNK cells express PD-1 but not CD30 (shown in online suppl. Fig. S6D), supporting the notion that PD-1 expression on pNK cells is not due to trogocytosis but an endogenous expression in pNK cells.
miR-34a-5p Is Negatively Correlated to PD-1 in NK Cells from Patients with Hodgkin Lymphoma
Having established that the IRE1α pathway is impaired in the pNK cells of patients with HL, we next compared the native expression of miR-34a-5p and PD-1 in pNK cells from 9 healthy individuals and 11 patients with HL. Our results with NK cell lines indicate that miR-34a-5p negatively regulates PD-1 expression; therefore, an inverse relationship is expected to emerge between miR-34a-5p and PD-1 expression in pNK cells. We used a PrimeFlow RNA assay to simultaneously measure expression of miR-34a-5p and PD-1 protein by flow cytometry. With the specific oligonucleotide target probe, the assay hybridizes and amplifies miRNA molecules at single-cell resolution without the requirement for RNA extraction and quantification by conventional qRT-PCR. We observed two distinct clusters based on miR-34a-5p and PD-1 expression in pNK cells from patients with HL (cluster 1: CD56+CD3−vePD-1+miR-34a-5plow and cluster 2: CD56+CD3−vePD-1+miR-34a-5phigh; shown in Fig. 6a). We then tested the two subgroups for correlation between miR-34a-5p and PD-1 expression with the CD56+CD3−vePD-1+miR-34a-5phigh subgroup, showing a significant negative correlation (Pearson’s correlation coefficient R = −0.92; p = 0.009; shown in Fig. 6a), while the CD56+CD3−vePD-1+miR-34a-5plow subgroup showed a negative correlation; however, this did not reach statistical significance (Pearson’s correlation coefficient R = −0.83; p = 0.08; shown in Fig. 6a). No significant negative correlation was found in healthy subjects with detectable levels of PD-1 expression (Pearson’s correlation coefficient R = 0.18; p = 0.73; data not shown). This inverted relationship confirms our observations in NK cell lines in which miR-34a-5p is identified as a key inhibitory regulator of PD-1 expression.
Inverse correlation between miR-34a-5p and PD-1 expression in pNK cells from patients with HL. a Simultaneous quantification of miR-34a-5p and PD-1 protein in ex vivo pNK cells from HL patients (n = 11) showed a significant negative correlation between miR-34a-5p and PD-1 protein expression levels in two subgroups CD56+ CD3−ve PD-1+miR-34a-5plow (cluster 1) and CD56+ CD3−ve PD-1+miR-34a-5phigh (cluster 2). Size of the dots indicates combined miR-34a-5p and PD-1 protein expression levels. CD56+CD3-vePD-1+miR-34a-5phigh subgroup showed a significant negative correlation (p = 0.009), whereas the negative correlation in the CD56+CD3-vePD-1+miR-34a-5plow subgroup did not reach significance (p = 0.08). K-means clustering was used to group samples based on miR-34a-5p and PD-1 expression. Pearson’s correlation test was used for statistical analysis. b miR-34a-5p expression in patients with HL from cluster 1 is significantly downregulated compared to healthy individuals (n = 10), whereas patients with HL from cluster 2 have a marginally significant increase in the miR-34a-5p expression level.
Inverse correlation between miR-34a-5p and PD-1 expression in pNK cells from patients with HL. a Simultaneous quantification of miR-34a-5p and PD-1 protein in ex vivo pNK cells from HL patients (n = 11) showed a significant negative correlation between miR-34a-5p and PD-1 protein expression levels in two subgroups CD56+ CD3−ve PD-1+miR-34a-5plow (cluster 1) and CD56+ CD3−ve PD-1+miR-34a-5phigh (cluster 2). Size of the dots indicates combined miR-34a-5p and PD-1 protein expression levels. CD56+CD3-vePD-1+miR-34a-5phigh subgroup showed a significant negative correlation (p = 0.009), whereas the negative correlation in the CD56+CD3-vePD-1+miR-34a-5plow subgroup did not reach significance (p = 0.08). K-means clustering was used to group samples based on miR-34a-5p and PD-1 expression. Pearson’s correlation test was used for statistical analysis. b miR-34a-5p expression in patients with HL from cluster 1 is significantly downregulated compared to healthy individuals (n = 10), whereas patients with HL from cluster 2 have a marginally significant increase in the miR-34a-5p expression level.
Intriguingly, we also found miR-34a-5p expression is reduced in pNK cell cluster 1 (CD56+CD3−vePD-1+miR-34a-5plow subgroup) compared to healthy individuals but not in cluster 2 (CD56+CD3−vePD-1+miR-34a-5phigh, shown in Fig. 6b). Arguably, the impaired IRE1α pathway observed in pNK cells from patients with HL may also impact the regulation of miR-34a-5p expression and consequently PD-1 expression. However, a more detailed understanding of the mechanistic nature of IRE1α-pathway impairment in cancer will require further studies with an appropriate design to address these specific clinical questions.
Discussion
Herein we uncovered a new mechanistic association between the miRNA miR-34a-5p, transcription factor XBP1, and immune checkpoint PD-1 upon IRE1α-pathway activation to modulate human NK cell effector function and tolerance in HL. Activation of NK cells involves the IRE1α pathway, which triggers a rapid change in XBP1s-mediated gene regulation. This is amplified by an IRE1α endonuclease-mediated reduction of miR-34a-5p. Notably, miR-34a-5p inhibits XBP1 and PD-1 expression. XBP1s also directly promotes PD-1 expression that is further enhanced by the IRE1α-mediated reduction of miR-34a-5p, thus fine-tuning NK cell effector function. However, the IRE1α pathway is impaired in NK cells within patients with HL, resulting in a disruption of the XBP1/miR-34a axis and of PD-1 expression within NKcells of patients with HL. The mechanisms by which HL cells influence NK cells would be an interesting area of future research.
Several studies indicate that the NK cell/tumor cell PD-1/PD-L1 axis is involved in immune evasion in blood cancers including HL [1, 5, 48]; however, the molecular mechanisms that fine-tune PD-1 expression are unknown. The findings from the current study indicate that the IRE1α pathway regulates PD-1 in HL. Upon initial contact with HRS cells, the IRE1α pathway is induced resulting in upregulation of NK cell effector function. However, under sustained immune activation, PD-1 expression is upregulated, promoting immune tolerance (or exhaustion). Arguably, this “negative feedback” serves as a regulatory mechanism to adjust to chronic NK cell activation imposed by cancer-induced stress. The second outcome of IRE1α pathway activation is to fine-tune PD-1 expression by removing the inhibitory miR-34a-5p that in turn increases PD-1 expression (shown in Fig. 7). Chronic activation of the IRE1α pathway downregulates miR-34a-5p, which increases PD-1 expression and its inhibitory effects in NK cells. In line with these findings, miR-34a-5p and PD-1 were inversely correlated in pNK cells of patients with HL, implicating the IRE1α/miR-34a-5p/PD-1 axis as a novel mechanism in NK cell linking immune cell activation and tolerance through IRE1α and immune checkpoint PD-1.
Proposed mechanism for the dual role of IRE1α pathway in regulating the XBP1/miR-34a axis to increase PD-1 expression. a When IRE1α is inactive, the XBP1 transcript is not spliced (XBP1u), which lacks a transcription factor domain and is rapidly degraded by proteasome activity. In IRE1α inactive state, the miR-34a biogenesis is not disrupted by IRE1α endonuclease activity and functional miR-34a-5p is produced to suppress XBP1u expression, further increasing XBP1u decay by RNA interference. The expression of miR-34a-5p also plays a role to maintain PD-1 expression under minimal thresholds. b Upon IRE1α activation, miR-34a-5p is degraded by IRE1α endonuclease by cleavage of the primary miR-34a hairpin (via CUGCAG motif); through a similar mechanism a 26-nt intron is removed from XBP1 mRNA encoding an effective transcription factor (XBP1s) that changes the transcription of several downstream genes involved in immune response. c A chronic IRE1α activation elicited by cancer cells increases PD-1 expression, which is now released from miRNA suppression and can be effectively translated into functional PD-1 protein.
Proposed mechanism for the dual role of IRE1α pathway in regulating the XBP1/miR-34a axis to increase PD-1 expression. a When IRE1α is inactive, the XBP1 transcript is not spliced (XBP1u), which lacks a transcription factor domain and is rapidly degraded by proteasome activity. In IRE1α inactive state, the miR-34a biogenesis is not disrupted by IRE1α endonuclease activity and functional miR-34a-5p is produced to suppress XBP1u expression, further increasing XBP1u decay by RNA interference. The expression of miR-34a-5p also plays a role to maintain PD-1 expression under minimal thresholds. b Upon IRE1α activation, miR-34a-5p is degraded by IRE1α endonuclease by cleavage of the primary miR-34a hairpin (via CUGCAG motif); through a similar mechanism a 26-nt intron is removed from XBP1 mRNA encoding an effective transcription factor (XBP1s) that changes the transcription of several downstream genes involved in immune response. c A chronic IRE1α activation elicited by cancer cells increases PD-1 expression, which is now released from miRNA suppression and can be effectively translated into functional PD-1 protein.
Recent studies have demonstrated IRE1α as a “double-edged sword” that co-opts its endonuclease activity to regulate the activation of XBP1s while simultaneously repressing a subset of miRNAs [27]. IRE1α-endonuclease activation causes a rapid decay of CUGCAG motif-containing miRNAs (i.e., miR-17, miR-34a, miR-96, and miR-125b) and hence strongly increases the expression of several genes affecting multiple cellular pathways [27, 42]. The current study is the first to provide experimental evidence that IRE1α endonuclease works as a dual regulatory switch to enhance PD-1 expression in the NK cells of patients with HL, through simultaneous activation of XBP1s and repression of miR-34a-5p. Interestingly, neither PD-1 mRNA or protein was expressed across HRS cell lines, nor was the HRS marker CD30 observed in pNK cells in patients with HL, indicating that the expression of PD-1 in the NK cells of patients cannot be explained by trogocytosis as previous published [47]. The IRE1α/miR-34a/PD-1 axis may be one factor to explain some contradictory findings regarding PD-1 expression on NK cells [1, 5, 49, 50].
Notably, miR-34a-5p has been previously found to be a master regulator of tumor suppression [34], and to enhance the repression of PD-L1 in acute myeloid leukemia and lung cancer [28, 29]. Therefore, the IRE1α/miR-34a regulatory circuitry in the PD-1/PD-L1 immune checkpoint axis may partially explain the immune exhaustion commonly observed in other blood and solid cancers [34]. Notably, T-cells are abundant within the tumor microenvironment (TME), but only a few possess the appropriate T-cell receptor to recognize antigens expressed on HRS cells. In contrast, NK cells are not as abundant in the TME and they do not induce lysis through the recognition of specific antigens. However, unlike T-cells, NK cells tend to target cells with reduced or lost expression of MHC-class I. This indicates that the impairment of NK cell function is an important mechanism of immune evasion in HL. We also found that PD-1 blockade only partially reversed NK cell dysfunction and neither degranulation nor cytotoxic function was restored by PD-1 blockade after IRE1α-pathway inhibition. This indicates that alternative strategies are required to restore NK cell mediated anti-HL immunity. Hence, our findings support the current growing interest in small RNA-based therapies to manipulate immune checkpoints in immune cells including NK cells to improve cancer immune therapies in blood cancers [51].
In conclusion, a hitherto undescribed NK cell dual regulatory axis is outlined, by which the IRE1α pathway fine-tunes NK cell effector activity through an interconnected activation of the transcription factor XBP1s and inhibition of miR-34a-5p to modulate PD-1 expression. The IRE1α pathway is impaired in NK cells within patients with HL, resulting in disruption of the dual role IRE1α pathway plays in regulating the XBP1/miR-34a axis and PD-1 expression within NK cells. IRE1α endonuclease in NK cells represents a potential target to optimize NK cell immunotherapeutic strategies in future HL treatments.
Acknowledgments
The work utilized the flow cytometry core facility at the Translational Research Institute, Brisbane, QLD, Australia.
Statement of Ethics
This study protocol was reviewed and approved by Metro South Health Human Research Ethics Committee (HREC/07/QPAH/035). Written informed consent was obtained for all participants in this study in accordance with Human Research Ethics Committee guidelines.
Conflict of Interest Statement
The authors declare no competing nonfinancial interests but the following competing financial interests: Maher K. Gandhi receives research funding from Janssen and Beigene and has received honoraria from Novartis.
Funding Sources
This work was supported by the Leukaemia Foundation and the Mater Foundation (M.K.G.), Princess Alexandra Hospital Foundation (M.K.G. and A.S.C.), and University of Queensland PhD scholarships for international students (G.T. and K.B.). The Translational Research Institute is supported by the Australian Government.
Author Contributions
A.S.C., M.K.G., S.M., J.N., K.B., G.T., and M.K.G. conceived and designed the study; M.K.G. provided study materials and/or patients; A.S.C., K.B., G.T., M.B.S., S.M., J.N., L.M.L., F.V., Q.C., S.C.L., A.P., and J.G. collected and assembled data; A.S.C., K.B., G.T., M.K.G., S.M., and J.N. provided data analysis and interpretation; A.S.C. and M.K.G. provided financial support; and all authors undertook manuscript writing and final approval and are accountable for all aspects of the work.
Additional Information
Karolina Bednarska and Gayathri Thillaiyampalam contributed equally to this work, and Maher K. Gandhi and Alexandre S. Cristino shared senior authorship.
Data Availability Statement
All data generated or analyzed during this study are included in this article and its online supplementary material files. Further inquiries can be directed to the corresponding authors.