Summary
Goal: Though the function of round RNAs (circRNAs) in tumor development and immune regulation is well-known, the particular circRNA molecules that mediate immune responses after radiotherapy (RT) and the underlying mechanisms haven’t been recognized.
Strategies: Cytometry with time-of-flight (CyTOF) was used to research blood samples from sufferers with liver most cancers exhibiting abscopal results (AEs) after stereotactic physique radiotherapy (SBRT) to quantify the variety of dendritic cells (DCs) and CD8+ T cells and interferon-beta (IFN-β) degree. circTMEM56 and IFN-β ranges had been measured in 76 sufferers with liver most cancers utilizing qPCR and ELISA. Immunohistochemistry validated circTMEM56 and CD141 staining in tissues. The interplay between circTMEM56, miR-136-5p, and STING, in addition to the influence on anti-tumor immunity, was verified utilizing circTMEM56-specific probes, dual-luciferase exercise assays, proteomics evaluation, and western blot evaluation.
Outcomes: The function of circTMEM56 in enhancing anti-tumor immunity and response to RT in hepatocellular carcinoma (HCC) was decided. Greater circTMEM56 ranges had been linked to an improved RT response and higher medical outcomes in sufferers with HCC. circTMEM56 enhanced cGAS-STING signaling, elevated the variety of tumor-infiltrating CD8+ T cells, and elevated the serum IFN-β ranges. Furthermore, circTMEM56 administration considerably boosted the response to RT in tumors with low circTMEM56 expression.
Conclusions: Excessive circTMEM56 expression in HCC modulates the distant results of HCC RT by activating the cGAS-STING pathway to reshape the tumor microenvironment. This research gives a brand new method to enhance RT efficacy for HCC.
key phrases
Introduction
Liver most cancers has the third highest cancer-related mortality charge in response to the 2022 international most cancers statistics1. The commonest kind of liver most cancers is hepatocellular carcinoma (HCC), which is normally identified at a complicated stage and is difficult to deal with2. Furthermore, postoperative recurrence charges following radical surgical resection stay excessive, making locoregional and systemic therapies key elements in HCC administration. Radiotherapy (RT) is a dependable and efficient therapy for HCC, offering glorious native management and changing unresectable tumors into resectable tumors, thus enhancing therapeutic outcomes. Latest advances in novel RT know-how, together with image-guided radiotherapy (IGRT), intensity-modulated radiotherapy (IMRT), and stereotactic physique radiotherapy (SBRT), have considerably expanded the scope of RT. Historically seen solely as a cytotoxic therapy, RT has advanced to profoundly affect the tumor immunoenvironment. For instance, the abscopal impact (AE) demonstrates the potential of immunomodulators to reinforce the therapeutic worth of RT, giving rise to a brand new area of research often known as immunoradiotherapy3. Regardless of the numerous enchancment in abscopal response charges achieved by combining RT with immunotherapeutic brokers, the underlying mechanisms haven’t been established.
Round RNAs (circRNAs) are considered merchandise of splicing in mammalian cells, the place 1000’s of circRNAs are related to numerous organic capabilities, together with microRNA (miRNA) sponging4. Because of this, circRNAs are dysregulated in a number of illnesses, together with most cancers5. A number of genes, similar to circTRIM33-12 and circMET, have been implicated in HCC development6. circRNAs are identified to be steady and enriched in exosomes. Proof means that exosome circRNA dysregulation impacts exosome-mediated intracellular communication, particularly throughout tumor improvement. For instance, HCC cells are immunosuppressed by exosome circUHRH1, whereas non-small cell lung most cancers (NSCLC) cells are dysregulated by exosome circUSP7 and resist anti-PD-1 remedy7. Though circRNAs have an essential function within the therapeutic efficacy of RT, the influence of circRNAs on RT effectiveness shouldn’t be identified. Additional analysis is required to make clear how circRNAs affect the therapeutic outcomes of RT in HCC.
Cells in malignant tumors survive immune assaults by mechanisms, similar to lack of immunogenicity and immunosuppression8. Immune evasion is a trademark of most cancers cells. The interaction between tumor cells and the immune system has an important function throughout superior most cancers levels9. Moreover, immune checkpoint blockade (ICB) therapies have been proven to enhance the prognosis of most cancers by successfully focusing on and eliminating most cancers cells10. Anti-PD-1 antibody, for instance, is accepted as second-line remedy for superior HCC11. Nevertheless, anti-PD-1 therapy achieves full or partial responses in solely 17%–18% of sufferers with superior HCC and the quite a few opposed occasions underscore the necessity for a deeper understanding of HCC-related immunosuppression12.
Pre-RT specimens and blood samples from six sufferers with metastatic HCC had been analyzed. Three sufferers reported AEs, whereas three had tumor development. These instances had been chosen for circRNA sequencing to establish differentially expressed circRNAs to raised perceive how RT impacts the tumor immunoenvironment by figuring out crucial circRNAs. In the end, the present research aimed to evaluate the efficacy of RT and suggest a novel mixed therapy technique for enhancing RT outcomes in sufferers with HCC.
Supplies and strategies
Samples
From January 2016 to December 2020 recent puncture specimens had been collected from sufferers with HCC earlier than surgical procedure or RT. Blood samples had been collected utilizing EDTA tubes and all specimens had been promptly saved at −80°C. Our earlier research outlined the creation of tumor microarrays and subsequent follow-up protocols13. Moral approval was obtained from the Institutional Evaluate Board of Zhongshan Hospital [Fudan University, Shanghai, China (Approval no. ZS20240204)].
Cell strains and transfection
Human HCC cell strains (HepG2, Hep3B, Huh7, PLC/PRF/5, HCCLM3, and MHCC97H) and mouse HCC cell strains (Hepa 1–6) had been sourced from the Chinese language Academy of Science Cell Financial institution (Shanghai, China). Cell strains had been cultured in DMEM or RPMI-1640 medium supplemented with 10% fetal bovine serum and 1% antibiotics at 37°C in a humidified incubator with 5% CO2.
Shanghai Genomeditech Firm (Shanghai, China) offered the vectors. HCCLM3 cells had been transfected with circTMEM56 shRNA lentiviral and management vectors. circTMEM56 cDNA vectors and the corresponding management vectors had been transfected into Hep3B and Hep1-6 cell strains, which had been additionally constructed by Shanghai Genomeditech Firm. The circTMEM56 shRNA goal sequences had been 5′-GTGAATCAGCGGTTGAAGAAA-3′ (shRNA1), 5′-GTTTGTGAATCAGCGGTTGAA-3′ (shRNA2), and 5′-GAATCAGCGGTTGAAGAAATA-3′ (shRNA3). The STING shRNA goal sequences had been 5′-CAGCATTACAACAACCTGCTA-3′ (shRNA1), 5′-CTGCTCGTAGCGCCGGGCCTT-3′ (shRNA2), and 5′-GACCGGTGACCATGCTGGCAT-3′ (shRNA3). Antibiotic-resistant transfected cells had been chosen by including puromycin to the tradition medium for 7 d.
RNA-seq and evaluation
A Ribo-Zero equipment (Illumina, San Diego, CA, USA) was used to deplete whole RNA of rRNA. An Illumina TruSeq RNA Pattern Prep equipment (Illumina, San Diego, CA, USA) was used to organize cDNA libraries from whole RNA. Sequencing was carried out utilizing an Illumina HiSeq2500 platform by GeneWiz (Suzhou, Jiangsu, China). CIRI software program (model 2; BGI, Shenzhen, Guangdong, China) was used to establish circRNAs with the sequencing reads aligned to the GRCh38 reference genome. Rely information and differentially expressed circRNAs (DE circRNAs) had been analyzed in a earlier research13.
In situ hybridization
Slides had been deparaffinized, rehydrated, digested with pepsin, and subsequently dehydrated earlier than hybridization with 50-nm locked nucleic acid (LNA)-modified DIG-labeled probes particular to circTMEM56. The slides had been washed 3 times after hybridization, soaked in blocking buffer, and incubated with alkaline phosphatase-conjugated anti-DIG Fab fragments. The nuclei had been stained with nuclear quick crimson.
SILAC
Cells had been cultured and labeled utilizing SILAC kits (Invitrogen, Carlsbad, CA, USA). After harvesting an equal variety of cells from each teams had been mixed for crude protein extraction. The peptides had been analyzed utilizing nano-HPLC-MS/MS utilizing a Q Exactive mass spectrometer (Thermo Fisher Scientific, metropolis, state, nation). Acquired MS/MS information had been processed with MaxQuant software program (Shanghai Institute of Biochemistry and Cell Biology, Shanghai, China) utilizing the Andromeda search engine (v.1.3.0.5). The reverse decoy database and protein sequences of widespread contaminants had been concatenated utilizing the UniProtKB mouse database (43,233 sequences).
Mice tumor problem and RT therapy
HCC cells had been used to create subcutaneous or in situ xenograft mouse fashions. A radiation dose (10 Gy) was administered to evaluate the response to RT14. Subcutaneous xenograft progress was monitored each 3 d, whereas in situ xenograft quantity was measured on day 25 after injection. Six mice per group had been sacrificed on day 25 after inoculation.
Actual-time quantitative PCR (qRT-PCR), immunohistochemistry (IHC), and immunoblot (IB) analyses
SYBR Inexperienced Actual-time PCR Grasp Combine (Yeasen, Shanghai, China) was used to extract RNA from tissues or cell strains. The PCR primers had been particular for circTMEM56 (ahead: 5′-TGCTGGCATACATTGGGAAT-3′, reverse: 5′-GCTGAAAGGTGAAAAAGCTGA-3′), GADPH (ahead: 5′-GGTATGACAACGAATTTGGC-3′, reverse: 5′-GAGCACAGGGTACTTTATTG-3′), TMEM173 (ahead: 5′-TTTGCCATGT CACAGGATGC-3′, reverse: 5′-ATGAGGCGGCAGTTATTTCG-3′), and cGAS (ahead: 5′-TGGTGGGAAGAGTGGTGATTTC-3′, reverse: 5′-TGCATTCCAATGGCAGAAGC-3′). IHC evaluation was carried out following established protocols, as outlined in our earlier research, utilizing qualitative standards for staining depth. The built-in optical density (IOD) values had been quantified utilizing Picture Professional Plus software program (Media Cybernetics, Rockville, MD, USA) based mostly on our earlier analysis findings. IB evaluation was carried out15 and relative protein expression was decided utilizing ImageJ software program.
Cell migration, matrigel invasion, and cell proliferation assays
Cell migration and Matrigel invasion assays had been carried out as reported in earlier research. For the cell proliferation assay, 2 × 103 cells had been seeded into 96-well plates and monitored at designated time intervals. Cell Counting Equipment-8 [CCK-8] (Yeasen, Shanghai, China) was used to find out the optical density (OD).
Immunofluorescent staining and movement cytometry assays
Immunofluorescence staining was carried out on HCC cells as beforehand described. movement Cytometric evaluation was used to find out apoptosis charges. The cells had been stained with Annexin V-FITC/PI. Cells situated within the lower-right quadrant had been categorised as Annexin V-positive and recognized as early apoptotic cells, whereas cells within the upper-right quadrant, which had been constructive for each Annexin V and PI, had been recognized as late apoptotic cells. The resistance of those cells to apoptosis was assessed in each quadrants.
Tumor progress in vivo
4-week-old nude mice had been obtained from the Shanghai Institute of Materials Drugs (Shanghai, China) and housed in a pathogen-free animal facility. A complete of 5 × 106 HCC cells had been injected into nude mice to induce tumor progress. Tumor progress was monitored each 5 d post-injection till day 32 by measuring tumors bi-dimensionally, as described in our earlier research. Lung tissue sections had been stained with hematoxylin and eosin (H&E) and the metastasis charge was decided utilizing established strategies. All of the mouse experiments carried out at Zhongshan Hospital had been accepted by the Fudan College Animal Ethics Committee (Approval no. ZS-Y2022-323).
Twin luciferase reporter assay
Mutant luciferase reporter vectors had been constructed utilizing a mutagenesis equipment (Qiagen, Hilden, Germany) in response to the producer’s directions. The plasmid was transiently transfected into 293T cells, which had been lysed and picked up after 48 h. The samples had been centrifuged at 8,944–20,124 × g for 3–5 min to acquire the supernatants.
Luciferase was detected in response to the instrument tips with a measurement period of 10 s and an interval of two s. The assay concerned gently mixing the pattern with Firefly Luciferase Assay reagent (Beyotime Biotechnology, Shanghai, China) two-to-three instances, adopted by measuring relative mild items (RLUs) utilizing cell lysis buffer as a clean management. This process was repeated utilizing Renilla Luciferase Assay reagent (Beyotime Biotechnology, Shanghai, China) and the RLU values had been calculated as a measure of reporter gene activation.
Statistical evaluation
Quantitative information evaluation was carried out utilizing SPSS 21.0 (SPSS, Inc., metropolis, state, nation). The 2 teams had been in contrast utilizing a Pupil’s t-test. The Pearson correlation coefficient was used to evaluate the affiliation between the 2 proteins, whereas the Kaplan-Meier methodology and log-rank take a look at had been used to research the general survival and cumulative recurrence charges. Statistical significance was set at a P < 0.05. Pupil’s t-test was used to find out statistical significance until in any other case specified. Quantification information are introduced because the imply worth ± SEM or in box-and-whisker plots with field dimensions encompassing the 25th-75th percentiles. Horizontal bars characterize the median and error bars characterize the minimal and most values.
Outcomes
circTMEM56 downregulation is related to a poor RT response and prognosis in sufferers with HCC
AEs describe the phenomenon during which RT at one location causes tumor regression in non-irradiated areas. In line with Response Analysis Standards in Stable Tumors 1.1, three sufferers who developed AEs after SBRT had been enrolled between January 2016 and December 2020. Blood samples from these sufferers had been analyzed by cytometry utilizing time-of-flight (CyTOF), which revealed a better variety of dendritic cells (DCs) and CD8+ T cells (Figure 1A). Sufferers with AEs had a better interferon-beta (IFN-β) degree and variety of DCs, indicating that RT-induced AEs are associated to systemic anti-tumor immunity (Figure 1B–D). The circRNA profiles from tumors and blood samples of sufferers with and with out AEs following SBRT (Figure 1E) recognized 89 DE circRNAs (log2|FC| > 1, q-value < 0.05; Figure 1E, proper panel). By overlapping the 2 circRNA profiles, circRNA hsa_circ_0005720 (circTMEM56), derived from TMEM56, was proven to be probably the most upregulated circRNA in each tumor tissues and blood samples from sufferers with AEs (Figure 1F). qPCR and Sanger sequencing had been carried out to establish the loop construction of circTEME56 utilizing particular primers (Figure 1G, H). The circTEME56 degree in HCC tissues and blood samples from these sufferers was decided and proven to be larger in sufferers with AEs in comparison with sufferers with out AEs (Figure 1I–K). circTEME56 expression was additional investigated utilizing puncture samples from 30 sufferers with HCC receiving SBRT, which was proven to be low normally (Figure 1L).
Downregulation of circTMEM56 is related to a poor radiotherapy (RT) response and prognosis in sufferers with liver most cancers. (A) t-SNE diagram of CD45+ main tumor-infiltrating immune cell varieties in sufferers with and with out abscopal results (AEs). (B) CD8+ T cell degree and variety of dendritic cells (DCs) within the blood of sufferers with and with out AEs. (C) IFN-β degree within the blood of sufferers with and with out AEs. (D) Peripheral blood CD141+ DCs (cDC1 subset) ranges in AE-positive versus AE-negative affected person cohorts. (E) Warmth maps of circRNA expression in tumor tissues of liver most cancers sufferers with and with out AEs after RT. (F) Differentially expressed circRNAs recognized in tumor tissues and plasma exosomes of sufferers with and with out AEs. (G) qPCR outcomes exhibiting circTMEM56 transcribed utilizing primers from cDNA. (H) Schematic diagram of circTMEM56. Sanger sequencing revealed back-splicing websites. (I) qPCR outcomes exhibiting considerably larger circTMEM56 expression in hepatocellular carcinoma (HCC) tissues of sufferers with AEs in comparison with sufferers with out AEs. (J) qPCR outcomes exhibiting considerably larger circTMEM56 expression within the blood of sufferers with AEs in comparison with sufferers with out AEs. (Okay) In situ hybridization outcomes indicating considerably larger expression of circTMEM56 in HCC tissues of sufferers with AEs in comparison with sufferers with out AEs. (L) Variations in circTEME56 expression in 30 HCC tissues and adjoining tissues. (M) In situ hybridization outcomes exhibiting considerably decrease circTMEM56 expression in HCC tissues in comparison with para-cancer tissues. (N) Kaplan-Meier evaluation of total survival (OS) and progression-free survival (PFS) in 209 HCC sufferers was carried out based mostly on circTEME56 expression. *P < 0.05; **P < 0.01; ***P < 0.001.
As well as, circTEME56 expression in HCC samples was decided. Semi-quantitative in situ hybridization microarray evaluation confirmed considerably lowered circTEME56 expression in HCC tissues (Figure 1M). Kaplan-Meier evaluation confirmed that HCC sufferers with decrease circTEME56 expression (above the median) had considerably shorter survival (P = 0.0052) and better recurrence charges after RT in comparison with sufferers with larger circTEME56 expression (Figure 1N). Primarily based on these information, low circTMEM56 expression contributed to HCC development by modulating RT response.
Serum exosome circTMEM56 degree correlates with the RT impact in sufferers with HCC
Round RNAs have just lately been proven to be extremely concentrated in exosomes, exhibiting exceptional stability and abundance. Exosomes are naturally present in numerous cells, the place exosomes mediate cell-to-cell communication and take part in a number of tumor improvement processes. We beforehand confirmed that circTEME56 is upregulated in sufferers with AEs in comparison with sufferers with out AEs. Herein the exosome ranges of circTEME56 in these sufferers was additional examined (Figure 2A, B) and confirmed to be elevated in sufferers with AEs (Figure 2B). As well as, the exosome ranges of circTMEM56 in blood after RT was decided in 76 sufferers with HCC. The circTMEM56 degree was proven to be excessive in sufferers with a partial response (PR) and full response (CR), average in sufferers with steady illness (SD), and low in sufferers with progressive illness (PD) (Figure 2C). Moreover, blood samples from sufferers with a PR and CR had a better IFN-β degree and better variety of DCs in comparison with sufferers with PD (Figure 2D, E). The exosome circTMEM56 degree was positively related to the IFN-β degree and variety of standard kind 1 dendritic cells (cDC1s) (Figure 2F, G). Tumor tissues from sufferers with a PR and CR confirmed larger DC infiltration following SBRT than tissues from sufferers with PD and SD with a corresponding improve within the IFN-β degree and variety of CD141+ cells within the blood (Figure 2H). Moreover, the connection between the blood exosome circTMEM56 degree and response to RT was decided in 60 sufferers. A constructive correlation existed between the blood exosome circTMEM56 and IFN-β ranges in sufferers with RT (Figure 2I), suggesting that the exosome circTMEM56 degree is positively correlated with the response to RT in sufferers with HCC.
Exosome circTMEM56 from hepatocellular carcinoma (HCC) cells is related to the radiotherapy (RT) impact. (A) Consultant picture of exosomes extracted from affected person blood utilizing ultracentrifugation. Exosome markers are detected on exosomes enriched in affected person blood. (B) circTMEM56 expression in affected person blood with an abscopal impact (AE) is considerably larger than sufferers with out AEs. (C) circTMEM56 ranges within the blood of 76 sufferers with HCC after RT analyzed by qPCR. (D) Interferon-beta (IFN-β) ranges within the blood of 76 sufferers with HCC after RT analyzed by ELISA. (E) Variety of cDC1s within the blood of 76 sufferers with HCC after RT. (F) Correlation evaluation exhibiting a constructive relationship between circTMEM56 and the IFN-β degree. (G) Correlation evaluation exhibiting a constructive relationship between circTMEM56 and the variety of cDC1s. (H) Consultant photographs of circTMEM56 and CD141 staining in HCC. (I) Correlation evaluation exhibiting a constructive relationship between circTMEM56 and the IFN-β degree in 209 sufferers with HCC. ***P < 0.001.
Elevated circTMEM56 amplifies the RT-induced anti-tumor immune response by way of regulated kind I IFN (IFN-I) manufacturing in DCs
Elevated circTMEM56 ranges could enhance the radiation response and efficacy by regulating IFN-I induction in DCs. Step one was to find out the endogenous circTMEM56 ranges in seven HCC cell strains. Most HCC cell strains, particularly HCC cell strains with excessive metastatic potential, exhibited decrease circTMEM56 ranges in comparison with the HepG2 cell line. Subsequent, steady HepG2 and HCCLM3 cells had been generated with circTMEM56 knockout or overexpression (Figure 3A, B). Along with medical efficacy, RT induces DNA harm, releasing double-stranded DNA (dsDNA), which results in tumor cell dying and enhances native and distant tumor management by way of most cancers cell-extrinsic mechanisms, such because the augmentation of tumor-specific immunity. Graded RT doses (0, 2, 4, 8, and 10 Gy) elevated the dsDNA focus within the supernatant of HCCLM3 cells in a dose-dependent method (Figure 3C, D). Moreover, IFN-β and dsDNA launch in HCCLM3 cells following RT weren’t influenced by circTMEM56 expression (Figure 3E, F).
circTMEM56 will increase the manufacturing of kind I IFN (IFN-I) from dendritic cells (DCs) and mediates an anti-tumor immune response. (A) circTMEM56 degree measured by qPCR in 7 completely different hepatocellular carcinoma (HCC) cell strains. (B) Efficacy of circTMEM56 overexpression and interference analyzed by qRT-PCR. (C) dsDNA content material within the cell supernatant following completely different radiation doses. (D) Extracellular dsDNA content material detected by immunofluorescence after completely different radiation doses. (E) Results of circTMEM56 overexpression and knockdown on dsDNA launch from hepatocellular carcinoma cells after radiation. (F) Results of circTMEM56 overexpression and knockdown on IFNB1 mRNA ranges in hepatocellular carcinoma cells after radiation. (G) IFNB1 mRNA ranges in DCs after co-culturing with irradiated liver most cancers cells. (H) Variety of DCs after co-culturing with irradiated liver most cancers cells. (I) IFN-β focus within the supernatant after co-culturing of DCs with irradiated hepatocellular carcinoma cells. (J) Modifications in IFNB1 mRNA ranges in DCs after co-culturing with irradiated HCCs with or with out GW4869. (Okay) Variety of DCs after co-culturing with irradiated hepatocellular carcinoma cells with or with out GW4869. ***P < 0.001.
HCC cells had been co-cultured with completely different circTMEM56 and DCs remoted from wholesome human blood utilizing microbeads to find out how circTMEM56 prompts DCs and produces IFN-β. IFN-β and cDC1 cells had been elevated because of circTMEM56 overexpression, an impact that was inhibited by GW4869. This discovering means that RT-induced anti-tumor immune responses and IFN-I manufacturing are enhanced in HCC cells expressing circTMEM56 (Figure 3G–J).
circTMEM56 enhances RT-induced anti-tumor immune responses in HCC by the miR-136-5p/STING1 axis
Provided that circRNAs operate as miRNA sponges, whether or not circTMEM56 sponges goal particular miRNAs to modulate HCC development was investigated16. circTMEM5-interacting miRNAs had been remoted utilizing DCs and circTMEM56-specific probes and qRT-PCR was used to detect 385 potential miRNAs (Starbase v3.0). circTMEM56 particularly enriched miR-136-5p in comparison with the unfavourable management (Figure 4A). A dual-luciferase reporter assay was carried out in HEK293T cells to confirm that miR-136-5p is sponged by circTMEM56. Wild kind (WT) and mutant circTMEM56, missing miR-136-5p binding websites, had been cloned within the luciferase reporter vector, pLG3 (Figure 4B). The miR-136-5p mimic considerably lowered luciferase exercise in WT-circTMEM56 however not in mutant-circTMEM56 (Figure 4C). Moreover, biotinylated miR-136-5p mimics considerably enriched circTMEM56 throughout pulldown assays in comparison with unfavourable controls (Figure 4D). Furthermore, fluorescence in situ hybridization (FISH) analyses of most cancers cells demonstrated that circTMEM56 co-localizes with miR-136-5p (Figure 4E). The influence of circTMEM56 overexpression on the proteome was decided utilizing liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based quantitative proteomic evaluation with SILAC to substantiate the miR-136-5p targets. Practically 7,000 annotated proteins had been recognized in DC-circTMEM56 and -control cells. Amongst these 7,000 proteins, 435 and 501 had been up- and down-regulated, respectively, within the DC-circTMEM56 cells (Figure 4F). Purposeful evaluation utilizing GO and KEGG indicated that the differentially expressed proteins in DC-circTMEM56 cells had been primarily related to the immune response, chemotaxis, the inflammatory response, cell adhesion, angiogenesis, constructive regulation of cell migration, and the PI3K-Akt, the TNF, and chemokine signaling pathways (Figure 4G). Western blot evaluation confirmed that circTMEM56 expression will increase the STING protein and associated cytokines ranges, which was in keeping with the quantitative proteomic evaluation (Figure 4H). Impartial transcriptomic and proteomic analyses revealed that circTMEM56 impacts the cGAS-STING pathway in DCs.
circTMEM56 amplifies radiotherapy (RT)-induced anti-tumor immune response by the miR-136-5p/STING1 axis in dendritic cells (DCs). (A) RIP was carried out for circRNA in DCs utilizing a circTMEM56 probe and a unfavourable management (NC) probe. (B) Putative binding websites of miR-136-5p with respect to circTMEM56 predicated by way of StarBase v3.0. (C) Luciferase exercise of pLG3-circTMEM56 in 293T cells after co-transfection with miR-136-5p. (D) circTMEM56 ranges within the streptavidin-captured fractions of the 293T cell lysates after transfection with biotinylated miR-136-5p or the NC. (E) FISH evaluation in hepatocellular carcinoma (HCC) exhibiting co-localization of circTMEM56 with miR-136-5p within the cytoplasm. (F) A graph exhibiting overlap of differentiated proteins. (G) GO and KEGG analyses had been carried out. (H) Ranges of STING and CXCL10 protein expression in DCs with various ranges of circTMEM56 expression detected by western blot evaluation. (I) Putative binding web site of miR-136-5p on STING by way of StarBase v3.0. (J) Luciferase exercise of pLG3-STING in 293T cells co-transfected with miR-136-5p. (Okay) STING mRNA ranges in DCs expressed by completely different miR-136-5p or circTMEM56. (L) Interferon-β (IFN-β) ranges in DC supernatant detected after co-culturing with irradiated HCC cells with completely different ranges of miR-136-5p or circTMEM56 expression. (M) IFN-β ranges in DCs supernatant after co-culturing with irradiated HCC cells and ranging ranges of STING expression. ***P < 0.001.
STING could also be a human goal of miR-136-5p based mostly on RNA-seq outcomes and on-line prediction (Figure 4I). Initially, the potential binding websites between STING and miR-136-5p had been recognized. Subsequent dual-luciferase reporter assays demonstrated a lower in luciferase exercise in cells transfected with WT miR-136-5p in comparison with cells transfected with mutant miR-136-5p (Figure 4J). To higher perceive miR-136-5p capabilities in circTMEM56-induced signaling, lentiviral vectors expressing miR-136-5p and miR-136-5p inhibitors had been constructed and steady DCs had been established. STING and IFN-β expression had been upregulated in DC-shmiR-136-5p and DC-circTMEM56 (Figure 4K, L). Moreover, corresponding adjustments in IFN-β expression had been famous (Figure 4M). Provided that circTMEM56 overexpression compromised the immune microenvironment in DCs by way of the miR-136-5p/STING1 axis, we concluded that circTMEM56 overexpression promotes HCC development.
Elevated exosome circTMEM56 particularly targets DCs to facilitate the cGAS-STING pathway in HCC
The innate immune sensor, cGAS-STING, has a crucial function in tumors and immune cells and could be activated in tumor cells, DCs, and macrophages to control numerous levels of the cancer-immunity cycle. The function of those cells in RT-induced anti-tumor immune responses in HCC was investigated. Measurement of miR-136-5p ranges in THP-1 cells, macrophages, and DCs confirmed that DCs expressed larger miR-136-5p ranges in comparison with THP-1 cells and macrophages (Figure 5A). Moreover, HCCLM3-circTMEM56 and HepG2-shcircTMEM56 cells modulated STING expression in DCs, however not THP-1 cells or macrophages (Figure 5B). When co-cultured with CD8+ T cells, IFN-β expression in THP-1 and M2 macrophages remained unchanged underneath HCCLM3-circTMEM56 and HepG2-shcircTMEM56 cell therapy (Figure 5C, D), indicating that circTMEM56 influenced the expression of STING in DCs. The cGAS-STING pathway reached most activation in DCs expressing circTMEM56 at 72 h, as demonstrated by the dose-response experiments (Figure 5E). By measuring the impact of circTMEM56 on the cGAS-STING pathway in DCs, p-STING and p-IRF3 ranges had been proven to be larger after therapy with the supernatant from HCCLM3-circTMEM56 cells following RT in comparison with the supernatant from HCCLM3-control cells after RT. The rise in miR-136-5p expression was suppressed or counteracted by STING overexpression (Figure 5F–H), as confirmed by multiplex immunofluorescence (mIF) in HCC tissues post-RT (Figure 5I). These information indicated that circTEMM56 overexpression particularly influences the cGAS-STING pathway.
Elevated exosome circTMEM56 particularly targets dendritic cells (DCs) and promotes activation of cGAS-STING pathway. (A) Relative ranges of miR-136-5p in THP-1, macrophages, and cDC1 detected by qPCR. (B) Ranges of STING expression in THP-1, M2 macrophages, and cDC1 cultured in conditioned medium and detected by qPCR. (C) Interferon-β (IFN-β) ranges within the supernatant of THP-1 cells after co-culturing with irradiated liver most cancers cells and the variety of CD8+ T cells detected after co-culturing with CD8+ T cells. (D) IFN-β ranges within the supernatant of M2 macrophages after co-culturing with irradiated liver most cancers cells and the variety of CD8+ T cells detected after co-culturing with CD8+ T cells. (E) Activation of STING detected by western blot at completely different radiation doses and time factors. (F) Western blot evaluation of STING pathway activation in DCs cultured in several conditioned media. (G) Western blot evaluation of STING pathway activation in DCs cultured with completely different miR-136-5p ranges in several conditioned media. (H) Western blot evaluation to detect STING pathway activation in DCs with completely different ranges of STING expression cultured in several conditioned media. (I) Consultant photographs of CD8, CD141, and p-STING staining in HCC tissues. ***P < 0.001.
Exosome circTMEM56 improves RT-induced anti-tumor immunity in HCC
Primarily based on these findings it was predicted that circTMEM56 may enhance the tumor response to RT in mice with HCC. Hepa1-6-circTMEM56 and Hepa1-6-control in situ tumor fashions had been constructed in TMEME173 and cGASWT, in addition to TMEM173KO and cGASKO mice, and subsequently handled with RT (8 GyX3). Tumor progress was considerably delayed in Hepa1-6-circTMEM56 tumors in comparison with Hepa1-6-control tumors in TMEM173 and cGASWT mice. Nevertheless, Hepa1-6-circTMEM56 in situ tumors exhibited progress charges like Hep1-6-control in situ tumors in TMEM173 and cGASWT mice however in contrast to the delayed progress noticed in TMEM173KO and cGASKO mice (Figure 6A, B). The administration of exosome circTMEM56 delayed the expansion of in situ Hepa1-6-control tumors in Teme173 and cGASWT mice however this impact was not famous in STING-deficient (TMEM173KO) and cGASKO mice (Mb21d1–/–). Importantly, a major improve within the variety of DCs and CD8+ T cells was noticed in Hepa1-6-circTMEM56 tumors and Hepa1-6-control tissues handled with exosome circTMEM56 derived from Teme173 and cGASWT mice following RT in comparison with the corresponding management teams (Figure 6C, D).
Exosome circTMEM56 improves the radiotherapy (RT)-induced anti-tumor immunity efficacy in vivo. (A) Consultant CT photographs of the stomach of mice with corresponding quantification. (B) Consultant photographs of CD8 and CD11c staining in hepatocellular carcinoma (HCC). (C) Consultant CT photographs of the stomach of mice with corresponding quantification. (D) Consultant photographs of CD8 and CD11c staining in HCC. (E) Consultant images of the Hepa1-6 subcutaneous tumor mannequin, imaged utilizing the IVIS Imaging System. *P < 0.05; ***P < 0.001.
A subcutaneous tumor mannequin was established by injecting Hepa1-6-circTMEM56 and Hepa1-6-control each 2 d, adopted by RT on the beforehand injected tumor. When evaluating Hepa1-6-circTMEM56 with Hep1-6-control, non-irradiated tumor lesions steadily regressed within the teams handled with exosome circTMEM56 (Figure 6E). In conclusion, circTMEM56 enhanced anti-tumor immunity in DCs by activating the cGAS-STING pathway.
Dialogue
Though the function of RT in modulating the tumor microenvironment has been acknowledged for years, our understanding of the molecular mechanisms by which RT influences innate and adaptive immune responses continues to evolve. The present research revealed that sufferers with HCC who exhibited AEs after RT had elevated circTMEM56 ranges in tissues and serum in comparison with RT-resistant sufferers. We advise that low circTMEM56 ranges are related to longer total survival and decrease recurrence charges in sufferers with HCC. The outcomes of the present research revealed that elevated circTMEM56 expression enhances cGAS-STING signaling by the miR-136-5p/STING axis throughout tumor cell-to-DC communication (Figure 7). In line with the findings herein, elevated circTMEM56 augmented the variety of tumor-infiltrating CD8+ T cells and serum IFN-β degree in transgenic mice with HCC. Administering exosome circTMEM56 considerably improved the response to RT in tumors with low circTMEM56 ranges, highlighting the medical significance. These findings recommend that utilizing exosome circTMEM56 may function a novel adjuvant to standard RT by resetting the tumor immunomicroenvironment.
Working mannequin: circTMEM56 regulates cGAS-STING signaling pathway in DCs. miR-136-5p within the cytoplasm exerts an inhibitory impact on the STING activation earlier than radiation (left panel). Beneath regular situations STING is activated when DCs detect dsDNA launched by tumor cells. As soon as activated STING strikes from the endoplasmic reticulum to the Golgi equipment, the place STING interacts with TBK1 on the Golgi membrane. Nevertheless, the inhibitory motion of miR-136-5p impairs STING exercise and prevents STING translocation and interplay with TBK1, thereby disrupting IRF3 phosphorylation and resulting in suppression of downstream IFN signaling. Consequently, the STING signaling pathway stays in a comparatively inactive state, limiting immune activation and impairing IFN-mediated responses. Radiation induces the upregulation of circTMEM56 within the cytoplasm (proper panel), which acts as a ceRNA to sponge miR-136-5p, thereby decreasing the inhibitory results on STING signaling. Consequently, activation of STING results in enhanced recruitment of TBK1 and phosphorylation of IRF3. Phosphorylated IRF3 translocates into the nucleus, the place phosphorylated IRF3 binds to particular promoter areas of DNA and promotes the expression of IFN. This radiation-induced enhancement of STING activation suggests a possible mechanism for enhancing anti-tumor immunity by regulation of circRNA-miRNA interactions. circTMEM56, round RNA TMEM56; ER, endoplasmic reticulum; IFN, interferon; IRF3, interferon regulatory issue 3; miR-136-5p, microRNA-136-5p; STING, Stimulator of Interferon Genes; TBK1, TANK-binding kinase 1.
The cGAS-STING pathway has an essential function in regulating immune responses17. Via cGAS-STING-IFN-mediated immune activation, RT stimulates anti-tumor immunity18–20. The tumor microenvironment displays extremely advanced cGAS-STING signaling activation and influences completely different cell varieties, similar to most cancers cells, DCs, and macrophages. Activating the cGAS-STING pathway in tumor cells can act as a barrier to the early development of most cancers by selling IFN-I expression and different pro-inflammatory genes. Moreover, this pathway is strongly related to triggering mobile senescence in most cancers cells, which contributes to tumor-suppressive results21. The findings herein revealed that exosome circTMEM56 selectively elevated STING expression in DCs with out affecting HCC and macrophages, which was attributed to low STING ranges in HCC and lowered miR-136-3p in macrophages. Primarily based on the statement that cGAS-STING activation facilitates tumor development in mice22–24 and drives liver irritation in macrophages, the consequences of exosome circTMEM56 on HCC xenografts had been confirmed in a number of mouse fashions, together with transgenic fashions. The findings herein present novel proof for the function of cGAS-STING signaling in RT-induced immune responses and recommend a therapeutic technique for enhancing anti-tumor immunity. Nevertheless, additional investigation is required to establish the exact mechanisms by which the cGAS-STING pathway enhances RT efficacy in tumor cells.
Intercellular communication depends closely on exosomes, which act as messengers carrying bioactive molecules25. The current discovery of circRNAs in exosomes has attracted important consideration as a result of enrichment and stability. Aberrant circRNA expression has been implicated in numerous illnesses, significantly most cancers. For instance, exosome circUHRF1 induces NK cell dysfunction and contributes to immunosuppression and resistance to anti-PD-1 immunotherapy in HCC. Moreover, by the miR-934/SHP2 axis, the exosome circuit reshapes the tumor microenvironment in non-small cell lung most cancers by selling CD8+ T-cell dysfunction. The event of novel therapeutic and diagnostic methods using exosomes for most cancers therapy has been facilitated by research on exosome traits throughout tumor pathogenesis. Bioengineered exosomes, which create an optimum microenvironment for immunomodulatory elements, have proven nice potential as most cancers immunotherapies. Bioengineered exosomes successfully activate levels of the most cancers immunity cycle to induce robust cancer-specific immune responses. By co-culturing DCs with circTMEM56, larger ranges of exosome circTMEM56 had been proven to considerably increase DC proliferation and activation, enhancing IFN-1β manufacturing. Notably, the administration of exosome circTMEM56 improved RT efficacy by boosting anti-tumor immunity. The outcomes of the present research steered that exosome circTMEM56 could also be an efficient adjuvant for RT in sufferers with HCC. The supply of circTMEM56 by way of exosomes represents a complicated analysis area that capitalizes on the biocompatibility, stability, and focusing on capabilities of exosomes. Methods, similar to parental cell transfection, direct loading, or electroporation, may very well be used to include circTMEM56 into exosomes. Nonetheless, some persistent challenges embody exosome purification, upkeep of circTMEM56 stability and exercise, and analysis of circTMEM56 security and efficacy. With advances in know-how, extra environment friendly and safe supply methodologies are anticipated to emerge, thereby facilitating the medical software of circTMEM56.
Along with figuring out the molecular mechanism by which circTMEM56 enhances the radiotherapy-induced tumor immunomicroenvironment, the present research gives a novel and efficient adjuvant for HCC radiotherapy in a particular subgroup of sufferers. A theoretical foundation is offered for growing mixture therapies to additional enhance the survival charge of sufferers with HCC and scale back recurrences. A preliminary medical trial involving 34 sufferers with superior HCC was carried out to find out the function of circTMEM56 in RT mixed with anti-PD-1 remedy. The outcomes indicated that larger ranges of circTMEM56 expression had been considerably related to elevated response charges and improved total survival, suggesting that circTMEM56 could function a possible biomarker for predicting therapeutic outcomes in HCC sufferers. Nevertheless, given the restricted pattern measurement, these findings warrant additional validation by massive scale corollary research (Figure S1).
Conclusions
The findings of the present research confirmed that circTMEM56 enhances RT efficacy in HCC by serving as a aggressive endogenous RNA to sponge miR-136-5p, thereby assuaging suppression of STING and activating the cGAS-STING signaling pathway. Elevated circTMEM56 ranges correlated with elevated tumor-infiltrating CD8+ T cells, elevated serum IFN-β ranges, and improved medical outcomes in HCC sufferers. Exosome circTMEM56 particularly targets DCs to amplify cGAS-STING-driven anti-tumor immunity, highlighting the potential of exosome circTMEM56 as a therapeutic adjuvant to enhance the RT response. These findings underscore the function of circTMEM56 in reshaping the immunomicroenvironment and suggest a novel technique for optimizing RT efficacy in HCC. Additional validation by large-scale medical trials is important to translate these insights into actionable therapeutic interventions.
Battle of curiosity assertion
No potential conflicts of curiosity are disclosed.
Writer contributions
Conceived and designed the evaluation: Shisuo Du, Keai wu, Chao Gao.
Collected the info: Junjie Cheng, Shilin Lin, Aying Ma, Zhaochong Zeng.
Contributed information or evaluation instruments: Li Yuan, Kunchao Li, Yiming Zheng.
Carried out the evaluation: Li Yuan, Yue Wang, Junjie Cheng.
Wrote the paper: Li Yuan, Chao Gao, Yue Wang.
Information availability assertion
The datasets used and/or analyzed within the present research can be found from the corresponding writer upon affordable request.
- Acquired November 22, 2024.
- Accepted February 26, 2025.
- Copyright: © 2025, The Authors