Selective Targeting of Protein Kinase C (PKC)-θ Nuclear Translocation Reduces Mesenchymal Gene Signatures and Reinvigorates Dysfunctional CD8+ T Cells in Immunotherapy-Resistant and Metastatic Cancers
<p>nPKC-θ signatures are enriched in CTCs and metastatic tissues and are associated with poor patient survival in immunotherapy-resistant disease. (<b>A</b>) Immunohistofluorescence analysis of PKC-θ expression in CD4<sup>+</sup> and CD8<sup>+</sup> T cells isolated from healthy donor liquid biopsies. Bar/dot plots show the Fn/c (nuclear to cytoplasmic ratio) of PKC-θ phosphorylated at threonine 568 (PKC-θ-Thr568p). A score below 1 indicates cytoplasmic bias. Data are from three separate patients, <span class="html-italic">n</span> ≥ 20 cells per patient. Representative images are shown for each dataset. PKC-θ-Thr568p (red); CD8/CD4 (purple), and DAPI (cyan) were used to visualize expression and nuclei; scale bar represents 10 µM. (<b>B</b>) Immunohistofluorescence analysis of PKC-θ expression in human MCF-7 inducible model (MCF-7-IM) and MDA-MB-231 breast cancer cells. Human MCF-7 epithelial cells (MCF-7epi) were activated with PMA to induce EMT and generate MCF-7 mesenchymal-like (MCF-7mes) breast cancer cells. Bar graphs show the mean nuclear fluorescence intensity (NFI), cytoplasmic fluorescence intensity (CFI), and Fn/c for PKC-θ-Thr568p, <span class="html-italic">n</span> ≥ 20 cells per group. Representative images are shown for each dataset. PKC-θ-Thr568p (green) and DAPI (blue) were used to visualize nuclei. Scale bar represents 10 µM. (<b>C</b>) Contrast-enhanced CT scans of tumors in CR and PD metastatic melanoma patients at baseline and 12 weeks after treatment with immunotherapy (nivolumab). Red circles indicate tumor lesions. Tumor lesions are reduced in CR compared with baseline, while PD shows increased tumor burden. (<b>D</b>) Dot plot quantification of PKC-θ-Thr568p, CSV, and ABCB5 fluorescence intensity in circulating tumor cells (CTCs) isolated from immunotherapy-responsive (CR, partial response (PR)) or resistant (stable disease (SD), PD) melanoma patients defined using RECIST 1.1 criteria. The Fn/c for PKC-θ-Thr568p, mean CFI for CSV, and mean TFI for ABCB5 were quantified using ASI digital pathology. Representative images are shown for each cohort (six patients were profiled per cohort, <span class="html-italic">n</span> ≥ 20 cells per group); scale bar represents 10 µM. (<b>E</b>) Dot plot quantification of PKC-θ-Thr568p, CSV, and ABCB5 fluorescence in FFPE sections of primary melanomas from patients (<span class="html-italic">n</span> = 18 patients) with CR, SD, or PD. The Fn/c for PKC-θ-Thr568p, mean CFI for CSV, and mean TFI for ABCB5 were quantified by ASI digital pathology (<span class="html-italic">n</span> ≥ 40 cells per patient sample, four samples per patient). Representative images for each dataset are shown, scale bar represents 30 µM. (<b>F</b>) Dot plot quantification of PKC-θ-Thr568p, CSV, and ABCB5 fluorescence intensity in FFPE sections from breast cancer brain metastases (<span class="html-italic">n</span> = 30 patients) and primary breast cancer biopsies (<span class="html-italic">n</span> = 15 patients). The Fn/c for PKC-θ-Thr568p, mean CFI for CSV, and mean TFI for ABCB5 were quantified by ASI digital pathology (<span class="html-italic">n</span> > 40 cells per patient sample). Representative images for each dataset are shown (top); scale bar represents 30 µM. (<b>G</b>) Percent change in tumor lesion from baseline for a single patient with metastatic melanoma (Patient D) who was resistant to first-line treatment with pembrolizumab and displayed PD (baseline, 12 weeks) as defined by RECIST 1.1 criteria but subsequently responded to second-line nivolumab and ipilimumab to show a CR (24 weeks, 36 weeks). (<b>H</b>) CT scan showing overall tumor burden in Patient D at baseline, SD, and PR (partial response). (<b>I</b>) Mesenchymal protein expression (PKC-θ-Thr568p, CSV, and ABCB5) was profiled in CTCs isolated from Patient D at 0 (baseline) and 12-, 24-, and 36-weeks post-immunotherapy. The Fn/c for PKC-θ-Thr568p, mean CFI for CSV, and mean TFI for ABCB5 were determined by ASI Digital Pathology. Representative images for each dataset are shown (<span class="html-italic">n</span> ≥ 20 cells per group); scale bar represents 10 µM. (<b>J</b>) Metastatic melanoma patients (<span class="html-italic">n</span> = 18 patients) were scored for the Fn/c of PKC-θ from four liquid biopsies over 12 months, with Fn/c categorized as <3 or ≥3 (Fn/c >1 indicates nuclear bias, whereas <1 indicates cytoplasmic bias). These patients were tracked for an additional two years (total 36 months), and their survival data are plotted as Fn/c <3 or ≥3. Statistical significance is denoted by ns (not significant), * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.005, *** <span class="html-italic">p</span> ≤ 0.0005, and **** <span class="html-italic">p</span> ≤ 0.0001.</p> "> Figure 2
<p>PKC-θ shows high affinity binding to the Impα/β1 heterodimer and dependence on Impα/β for nuclear accumulation in vitro. (<b>A</b>) Affymetrix microarrays previously in MCF-7 cells treated with PMA and FACS sorted into CD44<sup>high</sup> and CD24<sup>low</sup> cancer stem cells (CSC) and non-stem cancer cells (NS), respectively were used to profile the mRNA expression of importins [<a href="#B6-cancers-14-01596" class="html-bibr">6</a>]. Graphs plot the mRNA expression of expressed importins in NS and CSC. (<b>B</b>) The strength of binding of recombinant purified His6-PKC-θ to increasing concentrations of the indicated Imps was determined using an AlphaScreen binding assay. (<b>C</b>) Nuclear import of fluorescently labelled PKC-θ (DTAF-PKC-θ) was reconstituted in vitro in mechanically perforated HTC cells in the presence (+) or absence (−) of exogenous cytosol and an ATP regeneration system. CLSM images were acquired periodically for measurement of accumulation of DTAF-PKC-θ (green panels) into intact nuclei. Nuclear integrity was confirmed by the exclusion of Texas red-labelled 70 kDa dextran (TR70; red panels). Antibodies targeting Imps (anti-Impα2, anti-Impβ1, and anti-Impα4 or a combination of anti-Impα2 and anti-Impβ1) were also included, as indicated. Images are shown at 20 min time points. (<b>D</b>) Image analysis was performed on the photomicrographs, such as those shown in C, using ImageJ. The nuclear to cytoplasmic fluorescence ratio (Fn/c) was calculated for the indicated samples at each time point. Curve fits were determined in GraphPad Prism using an exponential one-phase association equation. Results are for the mean Fn/c ± SEM (<span class="html-italic">n</span> > 10). <span class="html-italic">p</span>-values were determined for each time point compared to the + cytosol sample using the <span class="html-italic">t</span>-test with Welch’s correction. *** <span class="html-italic">p</span> < 0.0001. (<b>E</b>) The % maximal Fn/c was determined from graphs such as those shown in D for each sample. Results represent the mean ± SEM (<span class="html-italic">n</span> = 3). ** <span class="html-italic">p</span> < 0.01, *** <span class="html-italic">p</span> < 0.001 compared with the no addition sample (−). (F) Nuclear import of PKC-θ was reconstituted as per (<b>C</b>) in the presence of vehicle or either the PKC-θ inhibitor C27 or the Imp α/β1-dependent nuclear transport inhibitor ivermectin. CLSM images of perforated nuclei at a 20 min time point to examine the nuclear accumulation of DTAF- PKC-θ (green panels) using TR70 (red channel) to monitor nuclear integrity. (<b>G</b>) Maximal % Fn/c for each sample was determined as per (<b>E</b>). *** <span class="html-italic">p</span> < 0.001 compared to no addition (−).</p> "> Figure 3
<p>A novel PKC-θ peptide inhibitor specifically inhibits nuclear translocation of PKC-θ without affecting PKC-θ catalytic activity. (<b>A</b>) Schematic of PKC-θ depicting its domains including the canonical nuclear localization sequence (NLS) and SPT motifs. Red bars indicate location of peptide inhibitor sequences and blue bars indicate the sequence used to create PKC-θ plasmids. Graphical and schematic representation showing that PKC-θ can be targeted therapeutically. The full-length PKC-θ WT gene sequence and its mutants were used to transfect MCF-7 cells, and the localization of expressed PKC-θ was studied by confocal laser scanning microscopy. Fn/c values for each construct are shown, with significant differences between datasets indicated (<span class="html-italic">n</span> > 15 for each dataset). (<b>B</b>) Structure-guided design of the peptide inhibitor targeting the NLS region of PKC-θ. Left, PyMOL-generated PKC-θ based on the previously determined crystal structure (PDB 2ED) showing the region responsible for nuclear localization in red and blue. Right, structure of the nuclear import adapter IMPα (in ribbon format) bound to a cargo (in stick format) at the major binding site [<a href="#B50-cancers-14-01596" class="html-bibr">50</a>], highlighting the strategy for inhibiting nuclear localization of PKC-θ. (<b>C</b>) The nuclear localization of PKC-θ and other PKCs (PKC-β2, PKC-β1, PKC-δ, and PKC-α) were examined in the MCF-7 inducible model (IM). Representative images are displayed and the Fn/c shown. Scale bar represents 5 µm. (<b>D</b>) p65, p53, and Rb protein expression was examined in mesenchymal MCF-7 cells stimulated with PKC-θ activators PMA and TGF-β. MCF-7 cells were pretreated for 24 h with vehicle or 25 µM nPKC-θi2. Representative immunofluorescence images and Fn/c plots for MCF-7 cells treated with nPKC-θi2 are shown: − represents stimulated control; + represents stimulated samples pretreated with nPKC-θi2. Fn/c was assessed for each target protein (<span class="html-italic">n</span> ≥ 20 cells per group). The Mann–Whitney test was used to determine statistical significance. ns (not significant), <span class="html-italic">p</span> > 0.05; * <span class="html-italic">p</span> ≤ 0.05; ** <span class="html-italic">p</span> ≤ 0.01; *** <span class="html-italic">p</span> ≤0.001; **** <span class="html-italic">p</span> ≤ 0.0001. (<b>E</b>) Various concentrations of nPKC-θi2 and C27 inhibit the co-expression of PKC-θ with p65 or H2Bs32 in MDA-MB-231 cells. nPKC-θi2 concentrations: C1 = 12.5 µM, C2 = 25 µM, C3 = 50 µM, C4 = 100 µM. C27 concentrations: C1 = 1.875 µM, C2 = 3.75 µM, C3 = 7.5 µM, C4 = 15 µM. ASI digital pathology system microscopy was performed on MDA-MB-231 metastatic cancer cells probed with antibodies targeting PKC-θ and H2Bs32p with DAPI. >500 cells per group were scanned to profile the % positive population of MDA-MB-231 cells. Graphs show the % PKC-θ<sup>+</sup>H2Bs32<sup>+</sup> population change with increasing concentrations of C27 (purple line) or nPKC-θi2 (green line). (<b>F</b>) Inhibition of recombinant PKC-θ activity (%) by C27 or nPKC-θi2 relative to untreated control using a PKC activity kit (Enzo Life Sciences).</p> "> Figure 4
<p>nPKC-θi2 targets CSC signatures and mesenchymal pathways in metastatic and resistant cancer cell lines. (<b>A</b>) WST proliferation assay in melanoma (RPMI-7951, SK-MEL-3), breast cancer (MDA-MB-231), and immunotherapy-resistant (4T1, 4T1 brain clone, and B16F10) cancer cell lines. Cells were treated with nPKC-θi2 for 72 h before addition of WST reagent. Absorbance was measured at 450 nm. (<b>B</b>) Scratch wound assay in MDA-MB-231 breast cancer cells treated with vehicle, C27 (7.5 µM), or nPKC-θi2 (25 µM). Wound healing images were acquired by real-time imaging using the IncuCyte Zoom live cell analysis system every 6 h for 24 h. Relative wound density (%) was analyzed using IncuCyte Zoom software. One-way ANOVA was used to determine statistical significance. ns (not significant) <span class="html-italic">p</span> > 0.05; * <span class="html-italic">p</span> ≤ 0.05; ** <span class="html-italic">p</span> ≤ 0.01; *** <span class="html-italic">p</span> ≤0.001; **** <span class="html-italic">p</span> ≤ 0.0001. (<b>C</b>) RNA isolated from MDA-MB-231 murine tumors treated with nPKC-θi2 was analyzed using the NanoString pan-cancer panel. * indicates direct PKC-θ binding targets determined by overlaying NanoString data with ChIP sequencing data from PKC-θ enriched MDA-MB-231 samples. (<b>D</b>) Dot plot quantification of PKC-θ-Thr568p, CSV, and ALDH1A fluorescence intensity in the 4T1 TNBC brain cancer clone treated with vehicle, C27 (5 µM), or nPKC-θi2 (25 µM). The Fn/c for PKC-θ-Thr568p, mean CFI for CSV, and mean NFI for ALDH1A were determined by immunohistofluorescence analysis. <span class="html-italic">n</span> ≥ 20 cells per group. (<b>E</b>) Dot plot quantification of PKC-θ-Thr568p in MDA-MB-231 brain cancer clone cells treated with vehicle, C27 (5 µM) or nPKC-θi2 (25 µM). The Fn/c for PKC-θ-Thr568p was determined by immunohistofluorescence analysis. The Mann–Whitney test was used to determine statistical significance. ns (not significant) <span class="html-italic">p</span> > 0.05; * <span class="html-italic">p</span> ≤ 0.05; ** <span class="html-italic">p</span> ≤ 0.01; *** <span class="html-italic">p</span> ≤ 0.001; **** <span class="html-italic">p</span> ≤ 0.0001. (<b>F</b>) FACS plot of % CD44<sup>hi</sup>/CD24<sup>lo</sup> CSC inhibition in mesenchymal-like MCF-7 cells activated with PMA and TGF-β and MDA-MB-231 breast cancer cells treated with nPKC-θi2 1 µM (C1), 5 µM (C2), 25 µM (C3), 50 µM (C4), and 100 (C5) µM relative to their respective untreated control cells.</p> "> Figure 5
<p>Impact of the novel PKC-θ inhibitor on tumors in a TNBC xenograft model and CTCs from melanoma patients. (<b>A</b>) Tumor volume in MDA-MB-231 mouse-bearing tumors treated with vehicle control, docetaxel (4 mg/kg), nPKC-θi2 (40 mg/kg), or both (docetaxel given 3 times, 1 week apart and nPKC-θi2 given daily for 5 weeks). Tumor volumes were measured daily for each mouse. Each data point represents a single mouse (<span class="html-italic">n</span> = 4 mice per group). (<b>B</b>) Percent CD44<sup>hi</sup>/CD24<sup>lo</sup> CSC cells in total tumor in the MDA-MB-231 mouse model. (<b>C</b>) Immunofluorescence microscopy of tumor cells from MDA-MB-231 TNBC mice treated with nPKC-θi2 in combination with docetaxel showing that nPKC-θi2 inhibits the fluorescence intensity of PKC-θ and key stem cell niche markers CD133, ALDH1A, and ABCB5 and mesenchymal marker CSV. (<b>D</b>) CTCs were isolated from melanoma patient liquid biopsies (CR = complete response, PR = partial response, PD = progressive disease) and were pre-clinically treated with either vehicle control or nPKC-θi2. Samples were fixed and immunofluorescence microscopy performed on these cells with primary antibodies targeting CSV, PKC-θ, and ABCB5. Representative images for each dataset are shown. Graph represents the TCFI values for CSV, NFI for PKC-θ, and TFI for ABCB5 measured using ImageJ to select the nucleus minus background (<span class="html-italic">n</span> ≥ 20 cells/sample). (<b>E</b>) Heatmap of tumor transcriptomes using all significant genes together with a Venn diagram comparison of genes induced by docetaxel/combination therapy or inhibited by docetaxel/combination therapy relative to vehicle control and the overlap between these groups. (<b>F</b>) Heat map of enriched pathways in gene sets induced relative to vehicle control with comparison of geneset pathways induced by docetaxel (DOC) or docetaxel and nPKC-θi2 (COM). Statistical significance is denoted by ns (not significant), * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.005, *** <span class="html-italic">p</span> ≤ 0.0005, and **** <span class="html-italic">p</span> ≤ 0.0001.</p> "> Figure 6
<p>PKC-θ is enriched in the nuclei of dysfunctional CD8+ T cells isolated from stage IV metastatic cancers. (<b>A</b>) Quantification of PKC-θ-Thr568p Fn/c in CD8+ T cells or CSV+ CTCs isolated from immunotherapy-responsive (complete response, CR; partial response PR) or resistant (PD, progressive disease) melanoma patients defined using RECIST 1.1 criteria. The Fn/c for PKC-θ-Thr568p was quantified by ASI digital pathology and ImageJ-Fiji (<span class="html-italic">n</span> ≥ 20 cells per group). (<b>B</b>) Quantification of PKC-θ-Thr568p Fn/c in CD8+ T cells or CSV+ CTCs isolated from healthy donors (HD), stage IV metastatic breast cancer patients (Mets), or stage IV breast cancer patients with brain metastases (Brain Mets). The Fn/c for PKC-θ-Thr568p was quantified by ASI digital pathology and ImageJ-Fiji (<span class="html-italic">n</span> ≥ 40 cells per group). (<b>C</b>) The amino acid sequence of ZEB1 indicating the top eight peptides for peptide phosphorylation by PKC-θ. The top peptide sequences are displayed in a table with the mean signal intensity (2 SD above the mean was considered a positive phosphorylation event). (<b>D</b>) Top peptides positive for phosphorylation and their overlap with the ZEB1 amino acid sequence as well as the structure of ZEB1, adapted from [<a href="#B3-cancers-14-01596" class="html-bibr">3</a>]. (<b>E</b>) Duolink<sup>®</sup> proximity ligation assay (PLA) for PKC-θ and ZEB1 in CD8+PD1+ T cells isolated from immunotherapy-resistant or responder melanoma patients. Representative images are shown for PKC-θ/ZEB1, scale bar represents 10 µM. Graphs represent the PLA signal intensity of the Duolink<sup>®</sup> assay; data represent <span class="html-italic">n</span> ≥ 100 cells/sample. Graphs plot the percentage of PLA signal positive cells out of total cells for (A). Data represent <span class="html-italic">n</span> ≥ 100 cells/sample. (<b>F</b>) FFPE sections from primary breast cancers (<span class="html-italic">n</span> = 6 patients, >500 cells counted per patient) or breast cancer brain metastases (<span class="html-italic">n</span> = 20 patients, >500 cells counted per patient) were processed for high-resolution microscopy using the BondRX platform. FFPE sections were fixed and immunofluorescence microscopy performed probing with primary antibodies targeting CD8, PKC-θ (T53p), and ZEB1 with DAPI. Plots represent the % population of CD8+ T cells positive for PKC-θ and ZEB1 out of total CD8+ T cells. Example images are shown with 20 µM scale bar. (<b>G</b>) CD8+ cells were isolated from melanoma patient liquid biopsies (responder = complete response (CR) or resistant, where primary = primary resistance, secondary = secondary resistance, PD = progression of disease) and stimulated with phorbol 12-myristate 13-acetate (PMA) and calcium ionophore (CI) and pre-clinically screened with either vehicle control or nPKC-i2. Samples were then fixed and immunofluorescence microscopy performed with primary antibodies targeting ZEB1, PKC-θ, and CD8. Representative images for each dataset are shown in <a href="#app1-cancers-14-01596" class="html-app">Figure S5E</a>. Graph represents the mean TNFI for PKC-θ and ZEB1 measured using ImageJ to select the nucleus minus background (<span class="html-italic">n</span> > 20 cells/sample). (<b>H</b>) Plot profiles for each cohort for ZEB1 and PKC-θ are also depicted (red = ZEB1, green = PKC-θ) with the Pearson correlation coefficient (PCC) used to quantify colocalization between fluorophore-tagged proteins indicated and plotted. −1 = inverse of colocalization; 0 = no colocalization; +1 = perfect colocalization. Statistical significance is denoted by ns (not significant), ** <span class="html-italic">p</span> ≤ 0.005 and **** <span class="html-italic">p</span> ≤ 0.0001.</p> "> Figure 7
<p>nPKC-θi2 disrupts the nuclear ZEB1/PKC-θ complex and induces cytokine production in CD8+ T cells. (<b>A</b>) Graphs depicting the % inhibition or induction based on protein expression were also plotted for each protein target relative to untreated sample. (<b>B</b>) Percent of PKC-θ+/ZEB1+/CD8+ T cells in samples isolated from melanoma patients responsive (PR/CR) or primary/secondary resistant (PD) to immunotherapy. CD8+ T cells were treated with nPKC-θi2 before activation ex vivo with PMA/ionomycin. (<b>C</b>) Gene expression of key effector cytokines IL2, IFNG, and TNFA in PBMCs isolated from resistant and responder patients either treated with vehicle control or nPKC-θi2 before activation ex vivo with PMA/ionomycin. (<b>D</b>) Protein expression of TNF-α and IFN-γ in CD8+ T cells isolated from primary or secondary resistant or responder patient liquid biopsies and treated with nPKC-θi2. Graphs show the % CD8+ increase in expression of TNF-α or IFN-γ in CD8+ T cells stimulated with PMA/ionomycin in addition to treatment with nPKC-θi2. One-way ANOVA was used to compare groups, where ns (not significant), **** <span class="html-italic">p</span> < 0.0001, *** <span class="html-italic">p</span> < 0.001, ** <span class="html-italic">p</span> < 0.01, and * <span class="html-italic">p</span> < 0.05.</p> ">
Abstract
:Simple Summary
Abstract
1. Introduction
2. Materials and Methods
2.1. Cell Culture
2.2. MCF-7 Inducible Model (MCF-7-IM)
2.3. Reconstitution of Nuclear Import
2.4. AlphaScreen® Binding Assay
2.5. CD8+ and CTC Enrichment from Blood
2.6. Cell and Tissue Processing and Immunofluorescence Microscopy
2.7. Applied Spectral Imaging (ASI) Digital Pathology
2.8. Proximity Ligation Assay (PLA)
2.9. Nuclear to Cytoplasmic Fluorescence Ratio (Fn/c) Analysis
2.10. PKC-θ Activity Assay
2.11. WST-1 Cell Viability Assay
2.12. IncuCyte® Scratch Wound Assay
2.13. In Vivo Mouse Xenograft Model
2.14. Flow Cytometry
2.15. RNA Sequencing
2.16. RNA-Seq Bioinformatics
2.17. Synthesis of Inhibitor Peptides
2.18. PBMC RNA Extraction and Quantitative Reverse Transcription PCR (qRT-PCR)
2.19. NanoString nCounter Assay
2.20. Peptide Microarrays
2.21. Quantification and Statistical Analysis
3. Results
3.1. nPKC-θ Is Enriched in Immunotherapy-Resistant CTCs and Is Associated with Poor Patient Survival in Immunotherapy-Resistant Metastatic Disease
3.2. ATP Competitive Catalytic PKC-θ Inhibitors Do Not Target Its Nuclear Import
3.3. A Novel PKC-θ Peptide Inhibitor Specifically Inhibits Nuclear Translocation of PKC-θ while Preserving PKC-θ Catalytic Activity
3.4. nPKC-θ Induces CSC Signatures and Mesenchymal Pathways in Metastatic and Resistant Cancer Cell Lines
3.5. Inhibition of nPKC-θ Reduces Mesenchymal Signatures in Primary Tumors and CTCs In Vivo
3.6. Targeting nPKC-θ Reduces the Chemotherapy-Induced Mesenchymal Signature on Tumor Cell Transcriptome
3.7. PKC-θ Is Enriched in the Nuclei of CD8+ T Cells Isolated from Stage IV Metastatic Cancers
3.8. PKC-θ Forms a Repressive Complex with ZEB1 in the Nuclei of CD8+ T Cells Isolated from Metastatic Cancer Patients
3.9. Inhibition of nPKC-θ Inhibits Dysfunctional ZEB1/PKC-θ Nuclear Complex and Induces Cytokine Expression in CD8+ T Cells
4. Discussion
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Dhanasekaran, N.; Premkumar Reddy, E. Signaling by Dual Specificity Kinases. Oncogene 1998, 17, 1447–1455. [Google Scholar] [CrossRef] [Green Version]
- Sutcliffe, E.L.; Li, J.; Zafar, A.; Hardy, K.; Ghildyal, R.; McCuaig, R.; Norris, N.C.; Lim, P.S.; Milburn, P.J.; Casarotto, M.G.; et al. Chromatinized Protein Kinase CΘ: Can It Escape the Clutches of NF-ΚB? Front. Immunol. 2012, 3, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Misheva, M.; Kaur, G.; Ngoei, K.R.W.; Yeap, Y.Y.; Ng, I.H.W.; Wagstaff, K.M.; Ng, D.C.H.; Jans, D.A.; Bogoyevitch, M.A. Intracellular Mobility and Nuclear Trafficking of the Stress-Activated Kinase JNK1 Are Impeded by Hyperosmotic Stress. Biochim. Biophys. Acta Mol. Cell Res. 2014, 1843, 253–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sutcliffe, E.L.; Bunting, K.L.; He, Y.Q.; Li, J.; Phetsouphanh, C.; Seddiki, N.; Zafar, A.; Hindmarsh, E.J.; Parish, C.R.; Kelleher, A.D.; et al. Chromatin-Associated Protein Kinase C-θ Regulates an Inducible Gene Expression Program and MicroRNAs in Human T Lymphocytes. Mol. Cell 2011, 41, 704–719. [Google Scholar] [CrossRef]
- Li, J.; Hardy, K.; Phetsouphanh, C.; Tu, W.J.; Sutcliffe, E.L.; McCuaig, R.; Sutton, C.R.; Zafar, A.; Mee Ling Munier, C.; Zaunders, J.J.; et al. Nuclear PKC-θ Facilitates Rapid Transcriptional Responses in Human Memory CD4+T Cells through P65 and H2B Phosphorylation. J. Cell Sci. 2016, 129, 2448–2461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zafar, A.; Wu, F.; Hardy, K.; Li, J.; Tu, W.J.; McCuaig, R.; Harris, J.; Khanna, K.K.; Attema, J.; Gregory, P.A.; et al. Chromatinized Protein Kinase C-θ Directly Regulates Inducible Genes in Epithelial to Mesenchymal Transition and Breast Cancer Stem Cells. Mol. Cell. Biol. 2014, 34, 2961–2980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boulding, T.; McCuaig, R.D.; Tan, A.; Hardy, K.; Wu, F.; Dunn, J.; Kalimutho, M.; Sutton, C.R.; Forwood, J.K.; Bert, A.G.; et al. LSD1 Activation Promotes Inducible EMT Programs and Modulates the Tumour Microenvironment in Breast Cancer. Sci. Rep. 2018, 8, 73. [Google Scholar] [CrossRef] [PubMed]
- Bi, K.; Tanaka, Y.; Coudronniere, N.; Sugie, K.; Hong, S.; Stipdonk, M.J. van; Altman, A. Antigen-Induced Translocation of PKC-Theta to Membrane Rafts Is Required for T Cell Activation. Nat. Immunol. 2001, 2, 556–563. [Google Scholar] [CrossRef]
- Monks, C.R.; Kupfer, H.; Tamir, I.; Barlow, A.; Kupfer, A. Selective Modulation of Protein Kinase C-Theta during T-Cell Activation. Nature 1997, 385, 83–86. [Google Scholar] [CrossRef]
- Brezar, V.; Tu, W.J.; Seddiki, N. PKC-Theta in Regulatory and Effector T-Cell Functions. Front. Immunol. 2015, 6, 530. [Google Scholar] [CrossRef] [Green Version]
- Isakov, N.; Altman, A. PKC-Theta-Mediated Signal Delivery from the TCR/CD28 Surface Receptors. Front. Immunol. 2012, 3, 2643–2649. [Google Scholar] [CrossRef] [Green Version]
- Zanin-Zhorov, A.; Dustin, M.L.; Blazar, B.R. PKC-θ Function at the Immunological Synapse: Prospects for Therapeutic Targeting. Trends Immunol. 2011, 32, 358–363. [Google Scholar] [CrossRef] [Green Version]
- Coudronniere, N.; Villalba, M.; Englund, N.; Altman, A. NF-ΚB Activation Induced by T Cell Receptor/CD28 Costimulation Is Mediated by Protein Kinase C-θ. Proc. Natl. Acad. Sci. USA 2000, 97, 3394–3399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, Z.; Arendt, C.W.; Ellmeier, W.; Schaeffer, E.M.; Sunshine, M.J.; Gandhl, L.; Annes, J.; Petrzilka, D.; Kupfer, A.; Schwartzberg, P.L.; et al. PKC-θ Is Required for TCR-Induced NF-ΚB Activation in Mature but Not Immature T Lymphocytes. Nature 2000, 404, 402–407. [Google Scholar] [CrossRef]
- Graff, J.R.; McNulty, A.M.; Hanna, K.R.; Konicek, B.W.; Lynch, R.L.; Bailey, S.N.; Banks, C.; Capen, A.; Goode, R.; Lewis, J.E.; et al. The Protein Kinase Cβ-Selective Inhibitor, Enzastaurin (LY317615.HCl), Suppresses Signaling through the AKT Pathway, Induces Apoptosis, and Suppresses Growth of Human Colon Cancer and Glioblastoma Xenografts. Cancer Res. 2005, 65, 7462–7469. [Google Scholar] [CrossRef] [Green Version]
- Haarberg, K.M.K.; Li, J.; Heinrichs, J.; Wang, D.; Liu, C.; Bronk, C.C.; Kaosaard, K.; Owyang, A.M.; Holland, S.; Masuda, E.; et al. Pharmacologic Inhibition of PKCa & PKCu Prevents GVHD While Preserving GVL Activity in Mice. Blood 2013, 122, 2500–2511. [Google Scholar] [CrossRef] [Green Version]
- Jourdan, E.; Leblond, V.; Maisonneuve, H.; Benhadji, K.A.; Hossain, A.M.; Nguyen, T.S.; Wooldridge, J.E.; Moreau, P. A Multicenter Phase II Study of Single-Agent Enzastaurin in Previously Treated Multiple Myeloma. Leuk. Lymphoma 2014, 55, 2013–2017. [Google Scholar] [CrossRef]
- Kovarik, J.M.; Stitah, S.; Slade, A.; Vitaliti, A.; Straube, F.; Grenet, O.; Winter, S.; Sfikas, N.; Seiberling, M. Sotrastaurin and Cyclosporine Drug Interaction Study in Healthy Subjects. Biopharm. Drug Dispos. 2010, 31, 331–339. [Google Scholar] [CrossRef]
- Matz, M.; Weber, U.; Mashreghi, M.F.; Lorkowski, C.; Ladhoff, J.; Kramer, S.; Neumayer, H.H.; Budde, K. Effects of the New Immunosuppressive Agent AEB071 on Human Immune Cells. Nephrol. Dial. Transplant. 2010, 25, 2159–2167. [Google Scholar] [CrossRef] [Green Version]
- Jimenez, J.-M.; Boyall, D.; Brenchley, G.; Collier, P.N.; Davis, J.C.; Damien, F.; Keily, S.B.; Henderson, J.; Miller, A.; Pierard, F.; et al. Design and Optimization of Selective Protein Kinase C θ (PKCθ) Inhibitors for the Treatment of Autoimmune Diseases. J. Med. Chem. 2013, 56, 1799–1810. [Google Scholar] [CrossRef]
- Xia, A.; Zhang, Y.; Xu, J.; Yin, T.; Lu, X.J. T Cell Dysfunction in Cancer Immunity and Immunotherapy. Front. Immunol. 2019, 10, 1719. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Z.; Liu, S.; Zhang, B.; Qiao, L.; Zhang, Y.; Zhang, Y. T Cell Dysfunction and Exhaustion in Cancer. Front. Cell Dev. Biol. 2020, 8, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Anderson, A.C.; Joller, N.; Kuchroo, V.K. Lag-3, Tim-3, and TIGIT: Co-Inhibitory Receptors with Specialized Functions in Immune Regulation. Immunity 2016, 44, 989–1004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thommen, D.S.; Schumacher, T.N. T Cell Dysfunction in Cancer. Cancer Cell 2018, 33, 547–562. [Google Scholar] [CrossRef] [Green Version]
- Pauken, K.E.; Wherry, E.J. Overcoming T Cell Exhaustion in Infection and Cancer Kristen. Trends Immunol. 2015, 36, 265–276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schnell, A.; Bod, L.; Madi, A.; Kuchroo, V.K. The Yin and Yang of Co-Inhibitory Receptors: Toward Anti-Tumor Immunity without Autoimmunity. Cell Res. 2020, 30, 285–299. [Google Scholar] [CrossRef] [Green Version]
- Rowe, C.L.; Wagstaff, K.M.; Oksayan, S.; Glover, D.J.; Jans, D.; Moseley, G.W. Nuclear Trafficking of the Rabies Virus Interferon Antagonist P-Protein Is Regulated by an Importin-Binding Nuclear Localization Sequence in the C-Terminal Domain. PLoS ONE 2016, 11, e0150477. [Google Scholar] [CrossRef] [Green Version]
- Wagstaff, K.M.; Headey, S.; Telwatte, S.; Tyssen, D.; Hearps, A.C.; Thomas, D.R.; Tachedjian, G.; Jans, D.A. Molecular Dissection of an Inhibitor Targeting the HIV Integrase Dependent Preintegration Complex Nuclear Import. Cell. Microbiol. 2018, 21, e12953. [Google Scholar] [CrossRef] [Green Version]
- Wagstaff, K.M.; Jans, D.A. Intramolecular Masking of Nuclear Localization Signals: Analysis of Importin Binding Using a Novel AlphaScreen-Based Method. Anal. Biochem. 2006, 348, 49–56. [Google Scholar] [CrossRef]
- Wagstaff, K.M.; Rawlinson, S.M.; Hearps, A.C.; Jans, D.A. An AlphaScreen®-Based Assay for High-Throughput Screening for Specific Inhibitors of Nuclear Import. J. Biomol. Screen. 2011, 16, 192–200. [Google Scholar] [CrossRef] [Green Version]
- Wagstaff, K.M.; Glover, D.J.; Tremethick, D.J.; Jans, D.A. Histone-Mediated Transduction as an Efficient Means for Gene Delivery. Mol. Ther. 2007, 15, 721–731. [Google Scholar] [PubMed]
- Eisenhauer, E.A.; Therasse, P.; Bogaerts, J.; Schwartz, L.H.; Sargent, D.; Ford, R.; Dancey, J.; Arbuck, S.; Gwyther, S.; Mooney, M.; et al. New Response Evaluation Criteria in Solid Tumours: Revised RECIST Guideline (Version 1.1). Eur. J. Cancer 2009, 45, 228–247. [Google Scholar] [CrossRef] [PubMed]
- Schwartz, L.H.; Litière, S.; De Vries, E.; Ford, R.; Mandrekar, S.; Shankar, L.; Bogaerts, J.; Chen, A.; Hayes, W.; Hodi, F.S.; et al. RECIST 1.1—Update and Clarification: From the RECIST Committee. Eur. J. Cancer 2016, 62, 132–137. [Google Scholar] [CrossRef] [PubMed]
- Tu, W.J.; McCuaig, R.D.; Melino, M.; Rawle, D.J.; Le, T.T.; Yan, K.; Suhrbier, A.; Johnston, R.L.; Koufariotis, L.T.; Waddell, N.; et al. Targeting Novel LSD1-Dependent ACE2 Demethylation Domains Inhibits SARS-CoV-2 Replication. Cell Discov. 2021, 7, 37. [Google Scholar] [CrossRef]
- Tu, W.J.; McCuaig, R.D.; Tan, A.H.Y.; Hardy, K.; Seddiki, N.; Ali, S.; Dahlstrom, J.E.; Bean, E.G.; Dunn, J.; Forwood, J.; et al. Targeting Nuclear LSD1 to Reprogram Cancer Cells and Reinvigorate Exhausted T Cells via a Novel LSD1-EOMES Switch. Front. Immunol. 2020, 11, 1228. [Google Scholar] [CrossRef]
- Wu, F.; McCuaig, R.D.; Sutton, C.R.; Tan, A.H.Y.; Jeelall, Y.; Bean, E.G.; Dai, J.; Prasanna, T.; Batham, J.; Malik, L.; et al. Nuclear-Biased DUSP6 Expression Is Associated with Cancer Spreading Including Brain Metastasis in Triple-Negative Breast Cancer. Int. J. Mol. Sci. 2019, 20, 3080. [Google Scholar] [CrossRef] [Green Version]
- Schindelin, J.; Arganda-Carreras, I.; Frise, E.; Kaynig, V.; Longair, M.; Pietzsch, T.; Preibisch, S.; Rueden, C.; Saalfeld, S.; Schmid, B.; et al. Fiji: An Open-Source Platform for Biological-Image Analysis. Nat. Methods 2012, 9, 676–682. [Google Scholar] [CrossRef] [Green Version]
- Bolger, A.M.; Lohse, M.; Usadel, B. Trimmomatic: A Flexible Trimmer for Illumina Sequence Data. Bioinformatics 2014, 30, 2114–2120. [Google Scholar] [CrossRef] [Green Version]
- Lassmann, T.; Hayashizaki, Y.; Daub, C.O. TagDust—A Program to Eliminate Artifacts from next Generation Sequencing Data. Bioinformatics 2009, 25, 2839–2840. [Google Scholar] [CrossRef] [Green Version]
- Kim, D.; Langmead, B.; Salzberg1, S.L. HISAT: A Fast Spliced Aligner with Low Memory Requirements Daehwan HHS Public Access. Nat. Methods 2015, 12, 357–360. [Google Scholar] [CrossRef] [Green Version]
- Frazee, A.C.; Pertea, G.; Jaffe, A.E.; Langmead, B.; Salzberg, S.L.; Leek, J.T. Ballgown Bridges the Gap between Transcriptome Assembly and Expression Analysis. Nat. Biotechnol. 2015, 33, 243–246. [Google Scholar] [CrossRef] [Green Version]
- Robinson, M.D.; McCarthy, D.J.; Smyth, G.K. EdgeR: A Bioconductor Package for Differential Expression Analysis of Digital Gene Expression Data. Bioinformatics 2009, 26, 139–140. [Google Scholar] [CrossRef] [Green Version]
- Heinz, S.; Benner, C.; Spann, N.; Bertolino, E.; Lin, Y.C.; Laslo, P.; Cheng, J.X.; Murre, C.; Singh, H.; Glass, C.K. Simple Combinations of Lineage-Determining Transcription Factors Prime Cis-Regulatory Elements Required for Macrophage and B Cell Identities. Mol. Cell 2010, 38, 576–589. [Google Scholar] [CrossRef] [Green Version]
- Enseñat-Waser, R.; Martin, F.; Barahona, F.; Vazquez, J.; Soria, B.; Reig, J.A. Direct Visualization by Confocal Fluorescent Microscopy of the Permeation of Myristoylated Peptides through the Cell Membrane. IUBMB Life 2002, 54, 33–36. [Google Scholar] [CrossRef]
- Liu, Y.; Graham, C.; Li, A.; Fisher, R.J.; Shaw, S. Phosphorylation of the Protein Kinase C-Theta Activation Loop and Hydrophobic Motif Regulates Its Kinase Activity, but Only Activation Loop Phosphorylation Is Critical to in Vivo Nuclear-Factor-ΚB Induction. Biochem. J. 2002, 361, 255–265. [Google Scholar] [CrossRef] [Green Version]
- Fillmore, C.M.; Kuperwasser, C. Human Breast Cancer Cell Lines Contain Stem-like Cells That Self-Renew, Give Rise to Phenotypically Diverse Progeny and Survive Chemotherapy. Breast Cancer Res. 2008, 10, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Yang, S.N.; Atkinson, S.C.; Wang, C.; Lee, A.; Bogoyevitch, M.A.; Borg, N.A.; Jans, D.A. The Broad Spectrum Antiviral Ivermectin Targets the Host Nuclear Transport Importin α/Β1 Heterodimer. Antivir. Res. 2020, 177, 104760. [Google Scholar] [CrossRef]
- Martin, A.J.; Jans, D.A. Antivirals That Target the Host IMPα/Β1-Virus Interface. Biochem. Soc. Trans. 2021, 49, 281–295. [Google Scholar] [CrossRef]
- DeVries, T.A.; Neville, M.C.; Reyland, M.E. Nuclear Import of PKCδ Is Required for Apoptosis: Identification of a Novel Nuclear Import Sequence. EMBO J. 2002, 21, 6050–6060. [Google Scholar] [CrossRef] [Green Version]
- Smith, K.; Himiari, Z.; Tsimbalyuk, S.; Forwood, J. Structural Basis for Importin-α Binding of the Human Immunodeficiency Virus Tat. Sci. Rep. 2017, 7, 1–11. [Google Scholar] [CrossRef] [Green Version]
- Kosugi, S.; Hasebe, M.; Entani, T.; Takayama, S.; Tomita, M.; Yanagawa, H. Design of Peptide Inhibitors for the Importin α/β Nuclear Import Pathway by Activity-Based Profiling. Chem. Biol. 2008, 15, 940–949. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martelli, A.M.; Faenza, I.; Billi, A.M.; Fala, F.; Coco, L.; Manzoli, L. Nuclear Protein Kinase C Isoforms: Key Players in Multiple Cell Functions? Histol. Histopathol. 2003, 18, 1301–1312. [Google Scholar] [PubMed]
- Belguise, K.; Milord, S.; Galtier, F.; Moquet-Torcy, G.; Piechaczyk, M.; Chalbos, D. The PKCθ Pathway Participates in the Aberrant Accumulation of Fra-1 Protein in Invasive ER-Negative Breast Cancer Cells. Oncogene 2012, 31, 4889–4897. [Google Scholar] [CrossRef] [Green Version]
- Byerly, J.; Halstead-Nussloch, G.; Ito, K.; Katsyv, I.; Irie, H.Y. PRKCQ Promotes Oncogenic Growth and Anoikis Resistance of a Subset of Triple-Negative Breast Cancer Cells. Breast Cancer Res. 2016, 18, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bild, A.H.; Yao, G.; Chang, J.T.; Wang, Q.; Potti, A.; Chasse, D.; Joshi, M.B.; Harpole, D.; Lancaster, J.M.; Berchuck, A.; et al. Oncogenic Pathway Signatures in Human Cancers as a Guide to Targeted Therapies. Nature 2006, 439, 353–357. [Google Scholar] [CrossRef]
- Charafe-Jauffret, E.; Ginestier, C.; Monville, F.; Finetti, P.; Adélaïde, J.; Cervera, N.; Fekairi, S.; Xerri, L.; Jacquemier, J.; Birnbaum, D.; et al. Gene Expression Profiling of Breast Cell Lines Identifies Potential New Basal Markers. Oncogene 2006, 25, 2273–2284. [Google Scholar] [CrossRef] [Green Version]
- Moserle, L.; Indraccolo, S.; Ghisi, M.; Frasson, C.; Fortunato, E.; Canevari, S.; Miotti, S.; Tosello, V.; Zamarchi, R.; Corradin, A.; et al. The Side Population of Ovarian Cancer Cells Is a Primary Target of IFN-α Antitumor Effects. Cancer Res. 2008, 68, 5658–5668. [Google Scholar] [CrossRef] [Green Version]
- Anastassiou, D. Universality of a Mesenchymal Transition Signature in Invasive Solid Cancers. Nat. Preced. 2012, 2–6. [Google Scholar] [CrossRef]
- Huang, A.C.; Postow, M.A.; Orlowski, R.J.; Mick, R.; Bengsch, B.; Manne, S.; Xu, W.; Harmon, S.; Giles, J.R.; Wenz, B.; et al. T-Cell Invigoration to Tumor Burden Ration Associated with Anti-PD-1 Response. Nature 2017, 545, 60–65. [Google Scholar] [CrossRef] [Green Version]
- Chen, L.; Gibbons, D.L.; Goswami, S.; Cortez, M.A.; Ahn, Y.H.; Byers, L.A.; Zhang, X.; Yi, X.; Dwyer, D.; Lin, W.; et al. Metastasis Is Regulated via MicroRNA-200/ZEB1 Axis Control of Tumour Cell PD-L1 Expression and Intratumoral Immunosuppression. Nat. Commun. 2014, 5, 611–629. [Google Scholar] [CrossRef]
- Guo, Y.; Lu, X.; Chen, Y.; Rendon, B.; Mitchell, R.A.; Cuatrecasas, M.; Cortés, M.; Postigo, A.; Liu, Y.; Dean, D.C. Zeb1 Induces Immune Checkpoints to Form an Immunosuppressive Envelope around Invading Cancer Cells. Sci. Adv. 2021, 7, eabd7455. [Google Scholar] [CrossRef]
- Guan, T.; Dominguez, C.X.; Amezquita, R.A.; Laidlaw, B.J.; Cheng, J.; Henao-Mejia, J.; Williams, A.; Flavell, R.A.; Lu, J.; Kaech, S.M. ZEB1, ZEB2, and the MiR-200 Family Form a Counterregulatory Network to Regulate CD8+ T Cell Fates. J. Exp. 2018, 215, 1153–1168. [Google Scholar] [CrossRef]
- Drápela, S.; Bouchal, J.; Jolly, M.K.; Culig, Z.; Souček, K. ZEB1: A Critical Regulator of Cell Plasticity, DNA Damage Response, and Therapy Resistance. Front. Mol. Biosci. 2020, 7, 36. [Google Scholar] [CrossRef]
- Belguise, K.; Sonenshein, G.E. PKCθ Promotes C-Rel-Driven Mammary Tumorigenesis in Mice and Humans by Repressing Estrogen Receptor α Synthesis. J. Clin. Investig. 2007, 117, 4009–4021. [Google Scholar] [CrossRef] [Green Version]
- Chu, P.-Y.; Chung-Heng Hsu, N.; Tai, H.-C.; Yeh, C.-M.; Lin, S.-H.; Hou, M.-F.; Yeh, K.-T. High Nuclear Protein Kinase C Theta Expression May Correlate with Disease Recurrence and Poor Survival in Oral Squamous Cell Carcinoma. Hum. Pathol. 2012, 43, 276–281. [Google Scholar] [CrossRef]
- Ou, W.B.; Zhu, M.J.; Demetri, G.D.; Fletcher, C.D.M.; Fletcher, J.A. Protein Kinase C-θ Regulates KIT Expression and Proliferation in Gastrointestinal Stromal Tumors. Oncogene 2008, 27, 5624–5634. [Google Scholar] [CrossRef] [Green Version]
- Byerly, J.H.; Port, E.R.; Irie, H.Y. PRKCQ Inhibition Enhances Chemosensitivity of Triple-Negative Breast Cancer by Regulating Bim. Breast Cancer Res. 2020, 22, 1–10. [Google Scholar] [CrossRef] [PubMed]
- Dunn, J.; Rao, S. Epigenetics and Immunotherapy: The Current State of Play. Mol. Immunol. 2017, 87, 227–239. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Dunn, J.; McCuaig, R.D.; Tan, A.H.Y.; Tu, W.J.; Wu, F.; Wagstaff, K.M.; Zafar, A.; Ali, S.; Diwakar, H.; Dahlstrom, J.E.; et al. Selective Targeting of Protein Kinase C (PKC)-θ Nuclear Translocation Reduces Mesenchymal Gene Signatures and Reinvigorates Dysfunctional CD8+ T Cells in Immunotherapy-Resistant and Metastatic Cancers. Cancers 2022, 14, 1596. https://doi.org/10.3390/cancers14061596
Dunn J, McCuaig RD, Tan AHY, Tu WJ, Wu F, Wagstaff KM, Zafar A, Ali S, Diwakar H, Dahlstrom JE, et al. Selective Targeting of Protein Kinase C (PKC)-θ Nuclear Translocation Reduces Mesenchymal Gene Signatures and Reinvigorates Dysfunctional CD8+ T Cells in Immunotherapy-Resistant and Metastatic Cancers. Cancers. 2022; 14(6):1596. https://doi.org/10.3390/cancers14061596
Chicago/Turabian StyleDunn, Jenny, Robert D. McCuaig, Abel H. Y. Tan, Wen Juan Tu, Fan Wu, Kylie M. Wagstaff, Anjum Zafar, Sayed Ali, Himanshu Diwakar, Jane E. Dahlstrom, and et al. 2022. "Selective Targeting of Protein Kinase C (PKC)-θ Nuclear Translocation Reduces Mesenchymal Gene Signatures and Reinvigorates Dysfunctional CD8+ T Cells in Immunotherapy-Resistant and Metastatic Cancers" Cancers 14, no. 6: 1596. https://doi.org/10.3390/cancers14061596
APA StyleDunn, J., McCuaig, R. D., Tan, A. H. Y., Tu, W. J., Wu, F., Wagstaff, K. M., Zafar, A., Ali, S., Diwakar, H., Dahlstrom, J. E., Bean, E. G., Forwood, J. K., Tsimbalyuk, S., Cross, E. M., Hardy, K., Bain, A. L., Ahern, E., Dolcetti, R., Mazzieri, R., ... Rao, S. (2022). Selective Targeting of Protein Kinase C (PKC)-θ Nuclear Translocation Reduces Mesenchymal Gene Signatures and Reinvigorates Dysfunctional CD8+ T Cells in Immunotherapy-Resistant and Metastatic Cancers. Cancers, 14(6), 1596. https://doi.org/10.3390/cancers14061596