Article Text
Abstract
Background The potency of T cell-mediated responses is a determinant of immunotherapy effectiveness in treating malignancies; however, the clinical efficacy of T-cell therapies has been limited in hepatocellular carcinoma (HCC) owing to the extensive immunosuppressive microenvironment.
Objective Here, we aimed to investigate the key genes contributing to immune escape in HCC and raise a new therapeutic strategy for remoulding the HCC microenvironment.
Design The genome-wide in vivo clustered regularly interspaced short palindromic repeats (CRISPR) screen library was conducted to identify the key genes associated with immune tolerance. Single-cell RNA-seq (scRNA-seq), flow cytometry, HCC mouse models, chromatin immunoprecipitation and coimmunoprecipitation were used to explore the function and mechanism of adenylate cyclase 7 (ADCY7) in HCC immune surveillance.
Results Here, a genome-wide in vivo CRISPR screen identified a novel immune modulator-ADCY7. The transmembrane protein ADCY7 undergoes subcellular translocation via caveolae-mediated endocytosis and then translocates to the nucleus with the help of leucine-rich repeat-containing protein 59 (LRRC59) and karyopherin subunit beta 1 (KPNB1). In the nucleus, it functions as a transcription cofactor of CCAAT/enhancer binding protein alpha (CEBPA) to induce CCL5 transcription, thereby increasing CD8+ T cell infiltration to restrain HCC progression. Furthermore, ADCY7 can be secreted as exosomes and enter neighbouring tumour cells to promote CCL5 induction. Exosomes with high ADCY7 levels promote intratumoural infiltration of CD8+ T cells and suppress HCC tumour growth.
Conclusion We delineate the unconventional function and subcellular location of ADCY7, highlighting its pivotal role in T cell-mediated immunity in HCC and its potential as a promising treatment target.
- IMMUNE RESPONSE
- HEPATOCELLULAR CARCINOMA
Data availability statement
Data are available on reasonable request. All data relevant to the study are included in the article or uploaded as online supplemental information.
This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/.
Statistics from Altmetric.com
WHAT IS ALREADY KNOWN ON THIS TOPIC
Immunotherapy, especially T-cell therapies, demonstrates excellent clinical efficacy across various tumours and holds great promise for clinical applications.
The extensive immunosuppressive microenvironment prevalent in hepatocellular carcinoma (HCC) has constrained the effectiveness of T-cell therapies.
WHAT THIS STUDY ADDS
Adenylate cyclase 7 (ADCY7), a transmembrane protein belonging to the adenylate cyclase family, induces CD8+ T cells infiltration and cytotoxicity to retard HCC progression.
Mechanistically, ADCY7 translocates into the nucleus and serves as a transcriptional cofactor for CEBPA, binding to the promoter of CCL5 to promote its transcription, thereby facilitating CD8+ T cell infiltration.
ADCY7 sheds from the plasma membrane through caveolae-mediated endocytosis and then translocates to the nucleus with the help of leucine-rich repeat-containing protein 59 (LRRC59) and karyopherin subunit beta 1 (KPNB1).
Exosomal ADCY7 reshapes the immunosuppressive microenvironment and inhibits HCC progression.
HOW THIS STUDY MIGHT AFFECT RESEARCH, PRACTICE OR POLICY
ADCY7 could be a viable therapeutic target for HCC patients by regulating T cell-mediated immune response.
Introduction
Hepatocellular carcinoma (HCC) is the third-leading cause of cancer-related death worldwide, and its incidence and mortality rates are increasing.1 Although innovative and efficient therapeutic strategies and advances in surgical techniques and precision medicine have been developed,2–6 the majority of HCC patients are ineligible for curative radical resection, and the prognosis is still unsatisfactory; this is ascribable to the late stage on diagnosis, early recurrence, metastasis and drug resistance.7
A growing amount of studies have highlighted the strong efficacy of immunotherapy in halting the spread of a range of human cancers by harnessing the immune system.8–12 Numerous regimens targeting inhibitory receptors such as programmed death-1 (PD-1), programmed death-ligand 1 (PD-L1) and cytotoxic T-lymphocyte associated antigen-4 (CTLA-4) have been developed and/or approved as first-line or second-line medications in the clinic, either as single agents or in combination with traditional therapeutics.13 14 Regretfully, only a fraction of HCC patients have been shown to benefit clearly from these immune checkpoint inhibitors (ICIs). Successful immunotherapy is contingent on T cell-mediated antitumour immunity, whereas the major barrier is the paucity of T cell infiltration and gradual dysfunction of antitumour T cells in the HCC tumour microenvironment (TME), resulting in a thorough immunosuppressive microenvironment.15 These findings emphasise the need for novel targets to improve the immune microenvironment to enhance immunotherapy efficacy in HCC patients.
Genome-wide, targeted loss-of-function pooled screens via the clustered regularly interspaced short palindromic repeats (CRISPR)-associated nuclease Cas9 (CRISPR-Cas9) system are powerful reverse-genetics tools capable of inducing complete gene knockout in cells.16 Due to the advantages of greater efficacy, ease of candidate identification and minimal off-target effects, managing genome-wide CRISPR screening techniques in oncology is gaining popularity, enabling comprehensive exploration of the mechanisms driving malignancy and potential drug targets.17–19 Genome-scale CRISPR screens have been successfully employed in vitro and/or in vivo to find common essential genes and other cancer type-dependent survival genes in diverse types of tumours.20–23 Furthermore, in conjunction with antitumour immune cells and existing treatments, such as CD8+ T cells, sorafenib and PD-1 antibodies, CRISPR screens have enabled the identification of key genes that indicate resistance or sensitivity to these treatments; thus, this approach has great significance in clinical decision-making for the treatment of cancers.24–29 However, an unbiased in vivo CRISPR screen to assess the immune response of HCC has not yet been carried out. Here, we leveraged the exceptional advantages of this technique to explore the crux of directing the T cell-mediated immune response in HCC to open the door for the astonishing efficacy of immunotherapy. Through systematic analysis, we identified the transmembrane protein ADCY7 as a robust regulator of T cell-mediated immunity and a predictor of the ICI response. Mechanistically, we revealed the previously undescribed subcellular nuclear localisation of ADCY7 and the role of ADCY7 in inducing CCL5 expression by acting as a transcription cofactor. In addition, ADCY7 can be secreted in the form of exosomes, and exosome-transmitted ADCY7 promotes CCL5 transcription in HCC cells to facilitate CD8+ T cell infiltration and tumour inhibition. Our findings highlight ADCY7 as a regulator of the T cell-mediated immune response in HCC and suggest that ADCY7 may be a promising target for HCC treatment.
Materials and methods
Additional materials and methods are included in online supplemental material and online supplemental tables.
Supplemental material
Supplemental material
Results
Identification of putative regulators of T cell-mediated antitumour immunity via a genome-wide in vivo CRISPR screen
To decipher potential genes that regulate T cell-mediated immunity in HCC, genome-wide in vivo CRISPR knockout screens were performed in three mouse models under different immune selective pressures, including immunocompetent (wild-type (WT)), immunocompromised (Rag1null (T-cell-deficient) and severe combined immunodeficient (NSG)) models (figure 1A). The mouse GeCKO v2 library, a pooled genome-wide CRISPR knockout library containing 130 209 single-guide RNAs (sgRNAs) targeting 20 611 genes, was introduced into Hepa1-6 cells at a low multiplicity of infection (<0.3). After puromycin screening, the infected Hepa1-6 cells were subcutaneously inoculated into the aforementioned mice (figure 1A). To determine the abundance of single-guide RNAs (sgRNAs) isolated from xenografts, the MAGeCKFlute algorithm30 was used to calculate β scores (an indicator of fold change between the Rag1null, NSG and WT groups) after PCR amplification of the genomic DNA.
With a Δβ score±SD (Δβ score signifies β score of the Rag1null or NSG group minus β score of the WT group), |β score of the WT group|>2 and |β score of the Rag1null or NSG group|<1 as the criteria, four regions with significant enrichment of sgRNAs were identified (figure 1B). Among them, genes enrichment in regions 2 and 4 indicated that gene knockout may affect tumour cell proliferative ability, while those enrichment in regions 1 and 3 signified sensitivity or resistance to immune pressure, respectively. Because sufficient genes are needed for candidates selection, we focused on region 3 (figure 1B, online supplemental tables 1 and 2). As shown by the detailed workflow of candidate selection in figure 1C,D, we obtained 521 common genes between the two regions three of which indicated that their deletion would confer resistance to T cell-mediated immunity. Subsequently, the following criteria were performed to narrow down these genes: difference of absolute β scores between the Rag1null and NSG groups<0.2 (147 genes), which further supported the possibility that the candidates were involved in modulating adaptive immune system-mediated responses rather than affecting tumour cell viability (online supplemental table 3). After filtering, 118 genes homologous between human and mouse species remained (figure 1D, online supplemental table 4). We next used the GEO-combat cohort (a pooled cohort combining 5 HCC GEO datasets generated via the R package ‘sva’, online supplemental figure 1A,B) to calculate the correlations between these genes and T cell-mediated antitumour response scores (seven gene sets that have been identified as indicators of elevated T-cell infiltration and antitumour response) (online supplemental table 5).31–34
By integrating the above results, two genes (ADCY7 and Fc fragment of IgE receptor 1 g, FCER1G) that were strongly correlated with T cell-mediated immunity (Pearson r>0.5 and p<0.05) were preliminarily selected (figure 1E,F, online supplemental table 6). Subsequent analysis demonstrated that ADCY7 and FCER1G were highly expressed in HCC patients who were sensitive to ICIs treatment (figure 1G). Although both were downregulated in tumour tissues, only ADCY7 was associated with a better prognosis in HCC patients in the GEO-combat and TCGA-LIHC>Ex datasets (downloaded and processed through the UCSC Xena database, https://xenabrowser.net/datapages/) (figure 1H–J). More importantly, to our knowledge, the involvement of ADCY7 in immunoregulation has not yet been well defined. Therefore, we selected this gene for further exploration. The proteomics data from the CPTAC database and western blot results from paired tumour and adjacent non-tumour tissues as well as different HCC cell lines, revealed lower ADCY7 protein levels in HCC tissues and cell lines (online supplemental figure 1C,D). Intriguingly, IHC staining showed that ADCY7 expression was relatively low in both normal liver tissues and HCC tissues but was high in cirrhosis tissues and adjacent non-tumour ones (online supplemental figure 1E). Furthermore, our HCC tissue microarray (HCC-TMA) cohort corroborated that ADCY7 protein expression was downregulated in HCC tissues compared with adjacent ones (figure 1K). The ADCY7 protein levels in HCC tissues vary greatly among individuals and patients with high ADCY7 had a better prognosis, underscoring the remarkable heterogeneity of ADCY7 expression in HCC (figure 1L). Moreover, univariate and multivariate Cox proportional hazards regression analyses indicated that ADCY7 was an independent favourable factor for HCC prognosis (online supplemental figure 1F, figure 1M). Collectively, ADCY7 may play an important role in HCC development.
Single-cell transcriptome analysis confirmed the association of CD8+ T-cell infiltration with ADCY7 expression in HCC
To systematically explore the association between ADCY7 expression and T-cell infiltration in the HCC microenvironment, single-cell RNA-seq (scRNA-seq) data from 10 primary HCC tissues (GSE149614) from the GEO database were downloaded and analysed. Following quality control and batch effect removal, 34 204 single cells were obtained and partitioned into seven subclusters (malignant cells, myeloid cells, T cells, endothelial cells, mesenchymal cells, epithelial cells and B cells) (online supplemental figure 2A.B). Based on the median value of ADCY7 mRNA expression in malignant cells, HCC samples were categorised into high-ADCY7 and low-ADCY7 groups (online supplemental figure 2C). Subsequently, OR analysis, which has been employed as a distribution preference indicator (an OR>1.5 denotes the distribution preference of this tissue, while an OR<0.5 signifies the opposite), highlighted the enrichment of T cells in the high-ADCY7 expression group (online supplemental figure 2D). T cells were further grouped into 10 subclusters (CD8-C1-IFNG, CD8-C2-FCGR3A, CD8-C3-MKI67, CD8-C4-PDCD1, MAIT, CD4 Tn, CD4-C1-TIGIT, CD4-C2-FOXP3, CD4-C3-S100A4 and NKT cells) according to their specific markers, with the CD8-C1-IFNG and CD4 Tn clusters emerging as two primary populations (online supplemental figure 2E,F).
We then assessed the enrichment of publicly available gene sets for naïve, cytotoxic, exhausted and regulatory T cells in the subclusters and derived a transcriptional score model using the AddModuleScore.35 Among the CD8+ T cells, the CD8-C1-IFNG and CD8-C2-FCGR3A cells had high cytotoxicity scores, while the CD8-C4-PDCD1 cells had the highest exhaustion score (online supplemental figure 2G). Among the CD4+ T cells, CD4 Tn cells had a high cytotoxicity score, while CD4-C1-TIGIT and CD4-C2-FOXP3 cells had high exhaustion and Treg scores (online supplemental figure 2H). OR analysis indicated that cytotoxic subclusters, such as CD8-C1-IFNG, CD8-C2-FCGR3A and CD4 Tn, were preferentially distributed in the high-ADCY7 subgroup, while immunosuppressive subclusters, such as CD8-C4-PDCD1 and CD4-C2-FOXP3, were preferentially distributed in the low-ADCY7 subgroup (online supplemental figure 2I,J). On the other hand, we investigated interactions between malignant cells and T cells via CellChat. Multiple T-cell subsets preferentially interacted with the high-ADCY7 subgroup compared with the low-ADCY7 subgroup (online supplemental figure 2K). Analysis of the ligand and receptor pairs revealed that of all the T-cell clusters, IFNG-(IFNGR1+IFNGR2) occurred only between the CD8+ T subsets and malignant cells in the high-ADCY7 subgroup; CD8-C1-IFNG and CD8-C2-FCGR3A were the main sources of IFNG; and malignant cells with high ADCY7 expression were the main cells receiving the signals (online supplemental figure 2L–O). Collectively, these data support the idea that ADCY7 is closely associated with the infiltration of cytotoxic CD8+ T cells.
Re-expression of ADCY7 promotes the CD8+ T cell-mediated antitumour response in HCC
Having established the potential association of ADCY7 with CD8+ T cells, we performed in vitro and in vivo experiments to further verify the function of ADCY7 in the T cell-mediated antitumour response during HCC development. Notably, ADCY7 re-expression had no significant effect on the proliferative capacity of HCC cells in vitro or tumour growth in immunocompromised NSG mice (online supplemental figure 3A–C). Moreover, tumours from the control and ADCY7 re-expression groups showed equivalent levels of Ki-67 and cleaved-caspase three staining (online supplemental figure 3D,E). Similar results were obtained in T cell-deficient Rag1null mice (online supplemental figure 3F–I). However, we noticed that ADCY7 re-expression drastically suppressed tumour growth compared with control group in immunocompetent C57BL/6J mice (figure 2A–C). The flow cytometry (FCM) results revealed increased infiltration of CD3+ T, CD8+ T and IFN-γ+CD8+ T cells but no significant changes in CD4+ T and NK cells in tumours from the ADCY7 re-expression group (figure 2D,E), which was consistent with the IHC results (figure 2F,G).
Furthermore, in our own HCC TMA cohort, we observed significant infiltration of CD3+ T and CD8+ T cells in patients with high ADCY7 expression (figure 2H,I). Remarkably, the T-cell migration assay revealed that the human HCC cell lines, HCCLM3 and HepG2, attracted more CD8+ T cells after ADCY7 re-expression (figure 2J–L). These findings rule out the possibility that ADCY7 mainly regulates adaptive immunity rather than HCC cell proliferation to suppress HCC progression.
ADCY7 induces CCL5 expression to attract CD8+ T cells in HCC
To explore the mechanism by which ADCY7 regulates CD8+ T cells recruitment, KEGG and GO analyses were performed on the differentially expressed genes (|fold change|>1.5 fold or <0.585 fold, p<0.05) between HCC patients with high and low ADCY7 mRNA levels in the GEO-combat cohort. The results showed that ADCY7 was positively associated with the chemokine signalling pathway and T-cell activation (figure 3A, online supplemental figure 4A,B). Furthermore, gene set enrichment analysis indicated significant upregulation of the chemokine signalling pathway in the high ADCY7 group (figure 3B). To elucidate such chemokines governing CD8+ T cells recruitment in ADCY7 re-expression HCC cells, the human chemokine antibody array was conducted using supernatants of HepG2 and HCCLM3 cells. Among them, chemokine (C-C motif) ligand 5 (CCL5), CCL26, (C-X-C motif) ligand 12 (CXCL12) and Midkine (MDK) were upregulated in ADCY7 high group (figure 3C, online supplemental figure 4C). Subsequent T cell migration analysis showed that silencing CCL5, rather than the other three chemokines, could obviously attenuate the CD8+ T cells attraction induced by ADCY7 re-expression (online supplemental figure 4D–G). Meanwhile, multiple prior articles have also reported its indispensable role in T-cell infiltration in diverse types of tumours.36–38 Therefore, CCL5 was focused for further exploration and we verified the upregulation of CCL5 mRNA and protein expression after ADCY7 re-expression in HCC cell lines (figure 3D,E).
To clarify the role of CCL5 in ADCY7-mediated antitumour immunity, we exposed ADCY7 re-expressing HCC cells to lentiviruses containing either a control or a CCL5-targeted short hairpin RNA (shRNA). CCL5-sh3 was chosen for subsequent experiments due to its best knockdown efficacy (online supplemental figure 4H). Similar results of T cell migration were obtained in control and ADCY7 re-expression HCC cell lines with CCL5 knockdown using shRNA (figure 3F). An in vivo xenograft model in C57BL/6J mice demonstrated that restoring ADCY7 expression failed to halt tumour growth in the context of CCL5 knockdown (figure 3G,H). IHC results showed a significant reduction in CD3+ T and CD8+ T-cell infiltration in ADCY7&sh-CCL5 group, suggesting that CCL5 is needed for the ADCY7-mediated immune response in HCC (online supplemental figure 4I,J). Consistently, the orthotopic HCC model corroborated that ADCY7 significantly inhibited tumour development, whereas knockdown of CCL5 was capable of mitigating this suppression effect (figure 3I,J). FCM analysis showed a higher infiltration of CD3+ T, CD8+ T cells and IFN-γ+CD8+ T cells in ADCY7 re-expression group (figure 3K,L). IHC results further ascertained these findings (figure 3M,N). Subsequently, we looked more closely at the function of ADCY7 in CD34+ humanised mice with a relatively intact human immune system.39 As anticipated, re-expression of ADCY7 in human HCCLM3 cells decreased tumour volume, whereas these effects could be counteracted by CCL5 knockdown (online supplemental figure 4K). Taken together, these results indicate that CCL5 is a principal molecule involved in the ADCY7-mediated antitumour response in HCC.
Nuclear ADCY7 functions as a transcription cofactor to induce CCL5 expression
We then attempted to determine how ADCY7 induces CCL5 expression. ADCY7 is a transmembrane protein that catalyses the conversion of ATP to cyclic adenosine 3′,5′-monophosphate (cAMP) to activate specific signalling pathways.40 Therefore, we first speculated that the ability of ADCY7 to retard HCC progression was dependent on its enzyme activity. Intriguingly, no differences in intracellular cAMP levels or expression of key downstream proteins (CREB, p-CREB, AMPK or p-AMPK) were observed between ADCY7 re-expressing and control cells (online supplemental figure 5A,B). Then we used forskolin, a direct activator of adenylate cyclases (ACs) to activate its enzyme activity; however, a comparable or slightly reduced CCL5 protein expression was observed between ADCY7 re-expressing and control cells (online supplemental figure 5A and C). These indicated that CCL5 elevation is induced by other ADCY7-related mechanisms rather than its enzyme activity in HCC.
Unexpectedly, we found that the ADCY7 protein was distributed in the nucleus in some HCC tissues (online supplemental figure 5D), which was not previously reported. Western blot analysis and confocal microscopy confirmed that ADCY7 was distributed in the nuclei of HCC cells (figure 4A,B, online supplemental figure 5E). This made us consider that ADCY7 might regulate CCL5 expression by binding to its promoter sequence. To verify this, we extracted the promoter sequence of the CCL5 gene (2000 bp) and designed 11 primers every 200 bp from the transcription start site (figure 4C). Chromatin immunoprecipitation assays of ADCY7 re-expressing HCCLM3 cells revealed that ADCY7 was able to bind to the CCL5 promoter sequence, mainly at the region −1100 to−900 bp from the transcription start site (figure 4D). However, ADCY7 cannot bind to the DNA sequence predicted by the iDRBP_MMC and DNAbinder databases, suggesting that ADCY7 indirectly regulates CCL5 transcription. After searching and selecting potential transcription factors in online public database, CCAAT/enhancer-binding protein alpha (CEBPA) and MYB were obtained as the potential transcription factors of CCL5 (figure 4E). qPCR showed that CEBPA silencing significantly reduced CCL5 mRNA expression compared with MYB knockdown (figure 4F,G). Moreover, coimmunoprecipitation (co-IP) and confocal microscopy demonstrated an interaction and colocalisation between ADCY7 and CEBPA, in contrast to MYB (figure 4H, online supplemental figure 5F–H). ADCY7 re-expression in HCC cells had no apparent effect on CEBPA expression but significantly enhanced the capacity of CEBPA to bind to the CCL5 promoter (figure 4I,J). We also found that CEBPA silencing obviously attenuated the ADCY7-induced increase in CCL5 transcription and protein expression (figure 4K,L). Furthermore, a series truncated plasmids of ADCY7 were constructed to verify the key domains for its nuclear translocation and interaction with CEBPA (figure 4M). As a result, all truncations were still able to translocate into the nucleus and induced CCL5 mRNA expression except the N-tail deleted ADCY7 mutant (figure 4N,O). Co-IP results further confirmed the interaction of N-tail with CEBPA (figure 4P). These results together demonstrated that nuclear ADCY7 through the N-tail assisted the transcription factor CEBPA in promoting CCL5 transcription.
Caveolae-mediated endocytosis of ADCY7 and translocation to the nucleus require its interaction with LRRC59 and KPNB1
The above-described function of ADCY7 encouraged us to explore the mechanism of its nuclear translocation. In terms of transmembrane proteins, the first step of translocation is shedding from the membrane into the cytoplasm. Clathrin-dependent or caveolae-mediated endocytosis is a crucial route by which proteins, such as PD-L1 and EGFR, enter the cytoplasm.41 42 Fortunately, we discovered that though its N-tail, ADCY7 can interact with caveolin-1 (CAV-1), an important component that results in the formation of caveolae (figure 5A, online supplemental figure 6A). Confocal microscopy also confirmed the colocalisation of the two proteins in HCC cells (figure 5B, online supplemental figure 6B). Moreover, filipin III,43 an inhibitor of caveolae-mediated endocytosis that had no obvious effect on ADCY7 and CCL5 expression, could elevate the plasma localisation of ADCY7 and diminish its nuclear distribution along with CCL5 mRNA and protein reduction in ADCY7 re-expressing cells (figure 5C, online supplemental figure 6C,D). Meanwhile, several studies have shown a lack of or low expression of CAV-1 in Hepa1-6 and HepG2 cells,44 45 which appears to contradict our results that ADCY7 facilitates its protein translocation by binding to CAV-1. However, we observed that CAV-1 levels were elevated in the ADCY7 re-expression group. Moreover, concurrent results were observed with CAV-1 knockdown and filipin III treatment, indicating that caveolae-mediated endocytosis might signify an initial step in ADCY7 translocation (figure 5D, online supplemental figure 6E–H).
Subsequently, we determined how ADCY7 reached the nucleus from the cytoplasm. Our above results indicated that the N-tail of ADCY7 was important for ADCY7 nuclear translocation (figure 4N), however, its N-tail domain does not contain potential nuclear location sequence (NLS) predicted by the NLStradamus website tool (http://www.moseslab.csb.utoronto.ca/NLStradamus/),46 suggesting that ADCY7 might enter the nucleus by other means. Some proteins without an NLS can enter the nucleus with the assistance of importin alpha/beta subunits, which may bind to other molecules containing NLSs and then form compounds to move into the nucleus.47–49 On the basis of our mass spectrum analysis, we found that ADCY7 might interact with the endoplasmic reticulum (ER) protein, leucine-rich repeat containing 59 (LRRC59), which has been reported to play an important role in protein nuclear translocation in cooperation with karyopherin subunit beta 1 (KPNB1), a member of the importin beta family (online supplemental figure 6I-J). As expected, our confocal microscopy experiments revealed the colocalisation of ADCY7 with LRRC59 and KPNB1 (figure 5E,F, online supplemental figure 6K). Co-IP further confirmed that ADCY7 interacted with LRRC59 and KPNB1 via the N-tail (figure 5G, online supplemental figure 6A). Additionally, when LRRC59 was silenced, the interaction between ADCY7 and KPNB1 was significantly decreased and the distribution of ADCY7 in the nucleus was also reduced (figure 5H–J). Moreover, to confirm the above findings, we used importazole (IPZ), a selective inhibitor known to block importin beta-mediated nuclear translocation, to assess its effect on the subcellular distribution of ADCY7.50 After treatment with IPZ at a concentration near the IC50 in HCC cells (online supplemental figure 6L), the total expression of ADCY7 remained stable, while nuclear ADCY7 distribution was obviously decreased (figure 5K–M). Furthermore, such effects of filipin III on CCL5 expression were also observed in HCC cell lines under LRRC59 silence and IPZ treatment, respectively (online supplemental figure 6M–P). Collectively, these results indicate that ADCY7 is taken into the nucleus via caveolae-mediated endocytosis and LRRC59 and KPNB1.
Exosomal ADCY7 suppresses HCC progression by attracting CD8+ T cells
To improve the clinical utility of our findings, we investigated whether ADCY7 is present in exosomes because membrane proteins that are released into the cytoplasm can be packaged into exosomes and secreted.42 According to the public database Vesiclepedia, ADCY7 has been detected in extracellular vesicles (EVs) from some human cancer cell lines.51 To test this phenomenon in HCC cells, we first purified and validated exosomes from the culture supernatant of HCC cells by transmission electron microscopy and analysed the exosomal markers (figure 6A,B). Western blotting and LC-MS/MS confirmed the existence of ADCY7 protein in exosomes derived from the ADCY7 re-expressing HCCLM3 cells (figure 6B,C). To investigate the function of exosomal ADCY7 in vitro, HCCLM3 cells were co-cultured with exosomes from control and ADCY7 re-expressing cells, respectively. Confocal microscopy revealed that exosomal ADCY7 moved to HCCLM3 cells and correspondingly upregulated CCL5 expression (figure 6D). Consistently, ADCY7 was also detected in exosomes from ADCY7 re-expressing Hepa1-6 and HepG2 cells (figure 6E, online supplemental figure 7A,B). Furthermore, in C57BL/6J mice bearing subcutaneous xenografted Hepa1-6 cells, intratumoural injection of exosomes from the ADCY7 re-expressing Hepa1-6 cells significantly inhibited tumour growth (figure 6F–H). FCM and IHC analysis revealed relatively high infiltration of CD3+ T cells (online supplemental figure 7C–F). Importantly, the proportion of CD8+ T cells and IFN-γ+CD8+ T cells was elevated in the tumours treated with ADCY7-expressing exosomes (figure 6I,J). Multiplex IHC confirmed the presence of exosomal ADCY7 and increased CCL5 expression in tumour cells (figure 6K). Taken together, these results suggest that exosomal ADCY7 can suppress HCC progression by reshaping the immune microenvironment.
Discussion
Harnessing the immune system against tumours, including HCC, has been identified as an effective therapeutic approach. However, a tremendous barrier to HCC treatment is the comprehensive immunosuppressive microenvironment,52 which confers a dismal outcome for HCC patients and needs finding out the significant regulatory mechanisms, as altering the state of immunosuppression is a desirable treatment strategy. In this study, we identified a new key gene (ADCY7) involved in adaptive immune regulation via an in vivo genome-wide CRISPR screen. ADCY7 is a transmembrane protein belonging to the AC family, whose main function is to generate the second messenger cAMP from ATP. To date, one study has demonstrated that ADCY7 exerts an oncogenic effect on leukaemia by inducing c-MYC expression,53 and sporadic studies have briefly characterised the function of ADCY7 in immune surveillance.54 Here, we discovered that ADCY7 was downregulated in HCC and correlated with poor prognosis. Moreover, its expression was also low in normal liver tissues compared with cirrhosis tissues and adjacent non-tumour ones at both mRNA and protein levels. This expression pattern leads us to speculate that ADCY7 levels might be linked to inflammation, given the inflammatory nature of chronic liver diseases. Meanwhile, a prior study has delineated the unique characteristics of adjacent non-tumour tissues, representing an intermediate state between healthy and tumour tissues. It is proposed that chronic inflammation, through the secretion of proinflammatory signals, induces specific alterations in gene expression, thereby modulating the surrounding microenvironment and establishing a distinct tissue composition conducive to tumour progression or suppression.55 Accordingly, ADCY7 might be one of these specifically elevated genes, with its expression being inducible under chronic inflammation rather than constitutive in liver tissues. However, its reduced expression in HCC may be attributed to high promoter methylation, highlighting the need for further detailed investigation.
Subsequent scRNA-seq analysis and multiple experiments demonstrated that ADCY7 high expression was not only associated with more CD8+ T cells infiltration but had a closer relationship with various CD8+ T cytotoxic subclusters, underscoring the potential significant value of ADCY7 in regulating HCC TME. Notably, we identified an unexpected nuclear location and function of ADCY7. Nuclear ADCY7 served as the transcriptional cofactor of CEBPA, inducing CCL5 expression and then attracting a considerable number of CD8+ T cells to impede HCC development. However, despite evidence that CCL5 could attract CD4+ T cells and Treg cells,56 57 no significant association between ADCY7 expression and CD4+ T cells has been observed in our study, which was inconsistent with previous reports.58 59 This discrepancy might stem from variations in receptor expression. Literature indicates that CCR5, the receptor with the highest affinity for CCL5, is the major receptor on CD8+ T cells, while most CD4+ T cell subsets mainly express CCR4 and CXCR3.60 Moreover, ADCY7 might affect CD4+ T cell infiltration through other potential mechanisms, requiring further exploration. Additionally, ADCY7 can also be packaged into exosomes, and exosomal ADCY7 enters tumour cells with low ADCY7 expression to induce CCL5 transcription (figure 6L), highlighting the pivotal role of ADCY7 in reshaping the TME.
Previous studies have reported that the localisation of many proteins can shift in tumours with corresponding consequences. Transmembrane proteins are generally shed into the cytoplasm via clathrin-mediated/caveolae-mediated endocytosis or alternative mechanisms, and then move to a special subcellular location to perform their corresponding functions. For example, the notable membrane protein PD-L1 enters the cytoplasm through clathrin-mediated endocytosis and then moves to the nucleus to exert its function with the help of the cytoskeletal protein vimentin and the nuclear transport protein KPNB1.42 Exogenous fibroblast growth factor 1 (FGF1) moves into cells’ nucleus by interacting with leucine-rich repeat containing 59 (LRRC59), an ER-bound protein that assisted proteins nuclear translocation,61 where FGF1 exerts its growth regulatory activity.62 Moreover, a recent study revealed that TSPAN8, a membrane protein, can translocate into the nucleus after binding to cholesterol and subsequently interact with STAT3 to induce the transcription of downstream oncogenes and promote breast cancer progression.63 We here discovered successful ADCY7 nuclear translocation initiated with caveolae-mediated endocytosis followed by interacting with LRRC59 and KPNB1. But rather than promoting tumour development, nuclear ADCY7 induced CCL5 expression by enhancing the ability of CEBPA to bind to the CCL5 gene promoter, which in turn attracted considerable CD8+ T cells to impede HCC progression. Additionally, the nuclear location of ADCY7 is also observed in certain para-tumourous hepatocytes but not in liver tissues of healthy individuals. However, ADCY7 is still predominantly localised to the plasma membrane and cytoplasm in those non-tumour tissues. This may be attributed to relatively low expression levels of those molecules (such as CAV-1 and KPNB1),64 65 which guide the nuclear translocation of ADCY7 or other mechanisms waiting for exploration.
Exosomes, as small EV ranging in size from 50 to 160 nm, are significant components of cell–cell communication and regulate recipient cells’ physiological activities.66 67 Growing evidence has proven the importance of exosomes in tumour development and clinical treatment and exosomes have been seen as a promising new tool of immunotherapy to combat cancer in a cell-free system.68–70 Moreover, it is well known that membrane proteins entering the cell mediated by endocytosis could be secreted by exosomes.42 71 In this study, we confirmed that ADCY7 was present in exosomes isolated from HCC cells re-expressing ADCY7 and exosomal ADCY7 facilitated CD8+ T cells infiltration and showed strong antitumour immunity in vivo, suggesting great potential for clinical use of ADCY7 in the future.
In summary, we identified a novel gene, ADCY7, as an immune regulator of HCC progression by means of an in vivo genome-wide CRISPR screen. ADCY7 penetrates the nucleus and assists CEBPA in binding to the CCL5 gene promoter to recruit CD8+ T cells to HCC. On the other hand, ADCY7 is packaged into exosomes to boost CD8+ T-cell infiltration by enhancing CCL5 transcription. Collectively, our findings highlight that ADCY7 is a viable therapeutic target for the clinical treatment of HCC patients.
Data availability statement
Data are available on reasonable request. All data relevant to the study are included in the article or uploaded as online supplemental information.
Ethics statements
Patient consent for publication
Ethics approval
This study involves human participants and was approved by the Ethical Committee of Eastern Hepatobiliary Surgery Hospital (No. EHBHKY2018-1-001). Participants gave informed consent to participate in the study before taking part.
Acknowledgments
We are grateful to patients and doctors in Eastern Hepatobiliary Hospital for HCC tissues collection.
References
Supplementary materials
Supplementary Data
This web only file has been produced by the BMJ Publishing Group from an electronic file supplied by the author(s) and has not been edited for content.
Footnotes
Contributors JC, YJ and MH are joint first authors. HW and JF conceived and designed the experiments; JC, YJ, MH and CL performed the experiments and bioinformatics analysis; EL, YZ, XW, ZM, MG and YS collected the human specimens; JF, JC, YJ and MH analysed the data; JF, JC and YJ wrote the manuscript. All authors have read, revised and approved the final manuscript. JF and HW are responsible for the overall content as the guarantor.
Funding This study is supported by the National Research Program of China (2022YFC3400902), the National Natural Science Foundation of China (81988101, 82073411, 82203209), Shanghai Discipline Leaders Program (2022XD058), Shanghai Sailing Program (21YF1458500), China Postdoctoral Science Foundation (2022M720786).
Competing interests None declared.
Provenance and peer review Not commissioned; externally peer reviewed.
Supplemental material This content has been supplied by the author(s). It has not been vetted by BMJ Publishing Group Limited (BMJ) and may not have been peer-reviewed. Any opinions or recommendations discussed are solely those of the author(s) and are not endorsed by BMJ. BMJ disclaims all liability and responsibility arising from any reliance placed on the content. Where the content includes any translated material, BMJ does not warrant the accuracy and reliability of the translations (including but not limited to local regulations, clinical guidelines, terminology, drug names and drug dosages), and is not responsible for any error and/or omissions arising from translation and adaptation or otherwise.