1. Introduction
Colorectal cancer (CRC) is the second leading cause of cancer-related death in humans [
1]. In some patients diagnosed with advanced CRC, the 5-year survival rate is only 14% [
2]. Compared with conventional chemotherapy and targeted therapy, immunotherapy has changed the treatment prospects for patients with various solid tumors and has become the standard regimen in CRC [
3]. Programmed cell death 1 (PD-1) antibodies, serving as immune checkpoint inhibitors (ICIs), have demonstrated notable therapeutic efficacy in patients exhibiting high microsatellite instability (MSI-H), a condition present in approximately 15% of all CRC cases [
4]. Regrettably, the predominant subset of CRC patients characterized by microsatellite stability (MSS) typically shows a lack of response to ICI therapy. In 2018, only 12.5% of patients with cancer who received treatment with ICIs in the United States had an objective response [
5]. This may be due to inter-patient differences in the tumor microenvironment (TME) and differences in tumor cell clonality, which reflects the highly regulated and complex nature of the immune system [
6,
7]. Therefore, understanding the inhibitory mechanisms of immunotherapy in TME will facilitate the development of personalized immunotherapy regimens, thereby improving the efficacy of tumor immunotherapy.
Metabolic disorder is not only the result of carcinogenic transformation but also one of the main driving factors in cancer development. Fumarate hydratase (FH) is an important metabolic enzyme in the tricarboxylic acid (TCA) cycle, and the loss of FH can act as a cancer driver [
8]. In addition, FH plays a protective role in maintaining the interaction between interferon and cytokines in macrophages [
9]. This indicates that there may be a relation between FH expression and the efficacy of tumor immunotherapy; however, the specific mechanism remains unclear. We found that the expression of FH was lower in some of the CRC patients. Moreover, through RNA-seq, we identified that protein invertase subtilisin/kexin 9 (PCSK9) may be a downstream effector of FH. Importantly, PCSK9 can reduce the expression of major histocompatibility complex (MHC) I protein on the surface of tumor cells, weaken T-cell receptor circulation and signal transmission, and inhibit the tumor infiltration and anti-tumor activity of CD8+ T cells [
10,
11].
Our results showed that low FH expression was related to increased Ras-related nucleoprotein (RAN)-mediated nuclear translocation of the PCSK9 transcription factor, SREBF1/2, which promoted the expression of PCSK9, thus weakening the monotherapy effect of a PD-1 antibody. Moreover, PCSK9 inhibition combined with PD-1 antibody therapy enhanced the effect of tumor immunotherapy. Therefore, we propose that FH may regulate sensitivity to treatment with PD-1 antibodies in patients with CRC and that patients with CRC with low FH expression may benefit from combined treatment with PCSK9 inhibitors and PD-1 antibodies. In addition, targeting key molecular entities in tumors can help maximize therapeutic efficacy [
12,
13], and FH has the potential in this regard.
2. Materials and Methods
2.1. Data Mining and Bioinformatics Analysis
The Cancer Genome Atlas (TCGA), UCSC Xena platform (UCSC Xena), Gene Set Enrichment Analysis (GSEA), and ChIP-Atlas were used for data mining and bioinformatics analysis (please refer to
Supplementary Methods for details).
2.2. Clinical Specimen
We collected postoperative cancer tissue and adjacent normal tissue from 12 patients with CRC who were treated at the Union Hospital Affiliated with Tongji Medical College of Huazhong University of Science and Technology. Postoperative pathological specimens from six rectal cancer patients were sensitive to neoadjuvant radiotherapy and chemotherapy combined with PD-1 antibodies (defined as tumor shrinkage of ≥40%), and six patients were insensitive (defined as tumor shrinkage < 40%). All procedures in this study were approved by the Ethics Committee of Union Medical College, Affiliated with Tongji Medical College, Huazhong University of Science and Technology (2022IEC-094). Prior to this study, all patients provided written informed consent. CRC tissue microarrays (HCol-Ade060CS-01,60 cases) were purchased from Shanghai Xinchao Biotechnology Co., Ltd. (Shanghai, China). The research methods used conform to the standards stipulated in the Helsinki Declaration.
2.3. Cell Line and Culture
A normal colon epithelial cell line (HCoEpiC), several human colon cancer cell lines (HCT15, LoVo, HCT116, SW620, SW480), and a mouse colon cancer cell line (MC38) were purchased from Type Culture Collection Cell Bank, Chinese Academy of Sciences (Shanghai, China). The authenticity of all cell lines was confirmed by short tandem repeat (STR) DNA analysis. HCT15 was cultured in RPMI-1640 medium (Gibco, Cat No. 11875093, Grand Island, NY, USA). LoVo was cultured in an F-12K medium (Gibco, Cat No. 21127022, Grand Island, NY, USA). HCT116 was cultured in McCoy’s 5A medium (Gibco, Cat No. 16600082, Grand Island, NY, USA). HCoEpiCS, SW620, SW480, and MC38 were cultured in high glucose DMEM medium (Gibco, Cat No. C11995500BT, Grand Island, NY, USA). All cells were cultured in 10% FBS (CELLiGENT, Cat No. CG0430B, Hamilton, New Zealand) and a 1% mixed solution of penicillin and streptomycin (Gibco, Cat No. 15140122, Toronto, ON, Canada). Cell lines were cultured in a saturated humidity incubator at 37 °C and 5% CO2.
2.4. Cell Transfection and RNA Interference Based on Lentivirus
An FH overexpression plasmid and empty vector (pcDNA3.1) was purchased from OriGene Company (Cat No. RC200614, Rockville, MD, USA). Lipofectamine 2000 (Invitrogen, Cat No. 11668019, Waltham, MA, USA) was used to transfect plasmids. FH-specific lentivirus (shRAN) was purchased from Gene-Chem (Shanghai, China). At 16 h post-transfection, the medium was changed, and 48 h later, the cell lines were screened for knockdown of the FH gene using 4 μg/mL puromycin (Bioroxx, Cat No. 1299MG025, Nordrhein-Westfalen, Germany) or 100 μg/mL Hygromycin B (BioFroxx, Cat No. 1366ML010, Nordrhein-Westfalen, Germany). The effects of plasmid transfection and lentivirus infection were verified by Western blot or RT-qPCR. The sequences of all shRNA constructs are listed in
Table S1.
2.5. Western Blot Analysis and Co-Immunoprecipitation (Co-IP)
Cell or tissue proteins were collected, and total proteins were extracted using RIPA buffer (Thermo Scientific, Cat No. 89900, Waltham, MA, USA) containing protease inhibitors (Bimake, Cat No. B14001, Houston, TX, USA). Cytoplasmic protein and nuclear proteins were extracted using a Nuclear and Cytoplasmic Protein Extraction Kit (Beyotime, Cat No. P0028, Shanghai, China). A BCA protein concentration determination kit (Beyotime, Cat No. P0012, Shanghai, China) was used to determine the protein concentration. Proteins were loaded onto SDS/PAGE gels for electrophoresis and transferred to PVDF membranes (Millipore, Cat No. IPVH00010, Burlington, MA, USA). Following overnight incubation with the primary antibody, membranes were incubated with the respective secondary antibody and detected by enhanced chemiluminescence (ECL). ImageJ (Version: 1.50G) software was used for protein quantitative analysis. The normalization with beta actin has been done in new clean blots (in general), and in a few cases in the same blot.
For co-immunoprecipitation, cells were lysed with IP lysis buffer (Servicebio, Cat No. G2038-100ML, Wuhan, China) containing phosphorylase inhibitor (Servicebio, Cat No. G2007-1ML, Wuhan, China) and PMSF (Servicebio, Cat No. G2008-1ML, Wuhan, China). The cell lysate was centrifuged at 12,000 rpm for 25 min. Next, the supernatant, corresponding antibody, and protein A/G agarose beads (Santa Cruz, Cat No. sc-2003, Santa Cruz, CA, USA) were incubated overnight at 4 °C on a shaker. The immune complex was washed with IP lysis buffer mixed with loading buffer, and immunoblotting was carried out. The antibodies used are listed in
Table S3.
2.6. Quantitative Real-Time PCR Analysis
Total RNA was extracted with TRIzol (Takara, Cat No. T9108, Shiga, Japan) according to the manufacturer’s instructions. cDNA was synthesized by reverse transcription with HiScript II Q RT SuperMix for qPCR kit (Vazyme, Cat No. R222-01, Nanjing, China), and RT-qPCR was performed with Chamq Universal Sybr QCPR Master Mix kit (Vazyme, Cat No. Q711-02, Nanjing, China). β-Actin was used as a reference, and the calculation formula is 2
−ΔΔCT. The primers used are listed in
Table S3.
2.7. Transwell Invasion Assay
The Transwell invasion assays were conducted using Transwell cell culture chambers (Corning, Cat No. 3422, Corning, NY, USA) with a diameter of 6.5 mm and a pore size of 8 μm. A total volume of 300 µL serum-free Opti-MEM (Gibco, Cat No. 31985070, Grand Island, NE, USA) was seeded into the upper chamber coated with Matrigel (BD Bioscience, Cat No. 356234, Beijing, China) at a concentration of 50 μL/cm2. Simultaneously, 600 µL medium containing 10% FBS was added to the lower chamber. After incubation for 24 h, cells were fixed with 4% paraformaldehyde (Servicebio, Cat No. G1101, Wuhan, China), followed by staining with a 1% crystal violet solution. Images were captured under an inverted microscope after wiping the upper surface of the chamber with sterile cotton swabs. Finally, the cells that had migrated through polycarbonate (PC) membranes were analyzed using Image J software (Version: 1.50G).
2.8. Colony-Forming Assay
1 × 103 HCT116 or SW620 cells were seeded into 6-well tissue culture plates following different treatments. Following a 14-day culture, the cells were fixed and stained using a 1% crystal violet solution, and visible colonies were observed through counting.
2.9. CCK8 Assay
The CCK8 assay was performed according to the manufacturer’s instructions. Briefly, a Cell Proliferation Assay Cocktail Kit (Abbkine, Cat No. KTD103-CN, Wuhan, China) was used to analyze cell proliferation. A total of 5 × 103 cells/well containing 100 μL of medium were seeded in 96-well plates. The cells were incubated in a humidified incubator at 37 °C with 5% CO2 for 24, 48, 72, 96, or 120 h, and 10 μL CCK-8 solution was added at the corresponding time points and cultured in the incubator for another 2 h. Wells without cells were used as blank controls, and absorbance at 450 nm was measured using a microplate reader (PerkinElmer EnSpire, SG, Pleasanton, CA, USA).
2.10. Flow Cytometry Analysis
In total, 100 μL of MC38 cell suspension (approximately 2 × 106 cells) corresponding to the shFh1Control or shFh1 was inoculated subcutaneously into C57BL/6 mice. Tumors were collected on day 21 after inoculation, minced and incubated in DNase I (BioFroxx, 0.2 mg/mL, Cat No. 1121MG010, Einhausen, Germany), collagenase I (BioFroxx, 1 mg/mL, Cat No. 1904MG100, Einhausen, Germany) and dispase II (Sigma-Aldrich, 2 mg/mL, Cat No. D4693, Burligton, MA, USA) for 60 min at 37 °C in a constant temperature shaker. After termination of digestion, the cells were filtered through a 70 μm cell filter to obtain a single-cell suspension. Cells were incubated with antibodies targeting cell surface antigens for 30 min at 4 °C in the dark using the following antibodies: CD45 antibody (BD, FITC, Cat No. 553080, Franklin Lake, NJ, USA), CD3 antibody (BioLegend, PE/Cyanine7, Cat No. 100219, San Diego, CA, USA), CD4 antibody (BioLegend, APC/Cyanine7, Cat No. 100525, San Diego, CA, USA), CD8a (BioLegend, PerCP/Cyanine5.5, Cat No. 100733, San Diego, CA, USA) and PD-L1 antibody (BioLegend, Biotin, Cat No. 124305, San Diego, CA, USA). Cells were fixed/permeabilized with 1× Foxp3 Fix/Perm Buffer (Invitrogen, Cat No. 2518973, Carlsbad, CA, USA) and incubated with Foxp3 antibody (Invitrogen, Cat No. 2518973, California, CA, USA) or isotype control IgG1 antibody (BioLegend, PE, Cat No. 400139, San Diego, CA, USA) in 1× Foxp3 Perm Buffer (Invitrogen, Cat No. 2518973, Carlsbad, CA, USA) for 30 min at room temperature in the dark. Finally, analyses were performed using a BD FACSCanto II flow cytometer. FlowJo (V10) was used to analyze the data.
2.11. Immunohistochemistry (IHC)
Human or mouse subcutaneous tumor model CRC tissue specimens were fixed with 4% paraformaldehyde and embedded in paraffin to prepare 4-μm sections. The prepared tissue sections or tissue microarrays were decaffinized and rinsed with water. The tissue antigens were repaired with citric acid repair solution (PH 6.0), and endogenous enzymes were blocked with 3% H
2O
2. After blocking with serum from the same source as the secondary antibody, the primary antibody was incubated overnight at 4 °C and the secondary antibody at 37 °C for 1 h. Diamino-benzidine (DAB) color solution (Servicebio, Cat No. G1212-200T, Wuhan, China) was used for color development, followed by counterstaining with hematoxylin, dehydration, and sealing. Images were obtained after microscopic observation. The primary antibodies used are listed in
Table S3. The pathologist scored the staining using a double-blind method. The IHC score was determined as the percentage of positive cells multiplied by the intensity of staining (0–3).
2.12. Animal Studies
Mice were provided by Hunan Westlake Jingda Laboratory Animal Co., Ltd. (Changsha, China), and the animal experimental protocol was approved by the Animal Ethics Committee of Huazhong University of Science and Technology and conducted accordingly (S3563). Mice were randomly assigned to the indicated groups. Tumor volumes were calculated according to the formula (L × W2)/2. HCT116 cells were infected with shControl, shFH, shPCSK9, or shFH+PCSK9 lentivirus after stable screening. Nude mice were subcutaneously inoculated with 100 μL of HCT116 cell suspension corresponding to approximately 2 × 106 cells to establish a subcutaneous tumor model. Mouse MC38 cell lines infected with shControl and shFh1 lentiviruses were stably screened. C57BL/6 mice were subcutaneously inoculated with 100 μL of MC38 cell suspension to establish a C57BL/6 mouse subcutaneous tumor model. PD-1 antibody (Bioxcell, Cat No. BP0146, Lebanon, NH, USA) and isotype IgG antibody (Bioxcell, Cat No. BE0089, Lebanon, NH, USA) against MC38 subcutaneous tumor models were administered by intraperitoneal injection at a dose of 5 mg/kg every 3 days. A PCSK9 inhibitor (Selleck, Cat No. 489415-96-5, Houston, TX, USA) and DMSO were administered subcutaneously at a dose of 4 mg/kg/day.
2.13. RNA Sequencing and Analysis
Total RNA was extracted from cells using TRIzol (Takara, Cat No. T9108, Japan). RNA sequencing (RNA-Seq) was performed by Novogene (Beijing, China). RNA integrity was verified using an Agilent 2100 bioanalyzer (Agilent Technologies, Santa Clara, CA, USA). Double-ended library sequencing on Illumina HiSeq 2500 was performed with the mRNA-seq sample preparation kit (Illumina, San Duego, CA, USA) according to the instructions provided by the manufacturer. Next, the sequencing data were analyzed using the Illumina data analysis pipeline. To minimize experimental bias, all samples were assigned to lane assignments and performed in a blinded fashion.
2.14. Chromatin Immunoprecipitation (ChIP) and ChIP-qPCR
Follow the manufacturer’s instructions. ChIP assays were performed using the SimpleChIP Enzymatic Chromatin IP Kit (Cell Signaling Technologies, Danvers, MA, USA, Cat No. 9003). After washing and purification, the DNA was analyzed by qPCR. Specific antibodies are shown in
Table S3, and primer sequences are shown in
Table S2.
2.15. Immunofluorescence Staining
Cells were inoculated in a 12-well plate and fixed with 4% paraformaldehyde (Servicebio, Cat No. G1101, Wuhan, China) for 20 min. After washing, cells were sealed with 5% bovine serum albumin (Sigma-Aldrich, Cat No. A1933, USA) for 1 h, followed by incubation overnight with primary antibody at 4 °C (see
Table S3 for specific antibodies). Next, cells were washed three times and incubated with CoraLite 594-Conjugated Goat Anti-Rabbit IgG (H+L) (Protein Tech, Cat No. SA00013-4, 1:100 dilution, Wuhan, China) at room temperature for 1 h. The nuclei were stained with DAPI (Beyotime, Cat No. C1006, Shanghai, China) and incubated at room temperature for 10 min. Anti-fluorescence quenching sealing solution (Beyotime, Cat No. P0126, Shanghai, China) was used to seal the film for 5 min. Images were taken under a fluorescence microscope (Olympus IX73, Tokyo, Japan).
2.16. Statistical Analysis
GraphPad Prism 9.4 software and R 4.3.0 software were used for statistical analysis; experimental data are expressed as mean ± SD, and at least three independent experiments were repeated in all cases. Unpaired two-tailed Student’s t-test or Mann-Whitney U test was used to compare the differences between the two groups and a two-way ANOVA test was used to compare differences between the two groups in the presence of time factors. Differences were considered statistically significant when p-values were less than 0.05. Survival curves were plotted by Kaplan-Meier analysis and assessed by log-rank test (* p < 0.05, ** p < 0.01, *** p < 0.001; NS indicates no statistical difference).
4. Discussion
Immunosuppressive regulation of the tumor microenvironment is a promising therapeutic approach for the treatment of various malignant tumors [
24]. However, due to the poor infiltration of cytotoxic T lymphocytes (CTL), the effect of immunotherapy in CRC tumors with pMMR and, to a lesser extent, dMMR is limited [
5,
6]. Our findings provide new insights for improving the effect of CRC immunotherapy. We observed that low FH expression was related to the insensitivity of immunotherapy in patients with CRC. Mechanistically, decreased FH expression promotes Ran-mediated SREBF1/2 nuclear translocation, leading to increased PCSK9 expression, which in turn leads to decreased CD8+ T cell clonal expansion and ultimately weakens the effect of PD-1 antibodies monotherapy. Therefore, FH may be a promising biomarker for PD-1 antibody treatment in CRC.
Metabolic alterations involved in tumorigenesis have multifaceted effects, not only relating to the nutritional status of cells but also influencing cell behavior, including the regulation of signaling and mechanics [
25]. As a key metabolic enzyme in the TCA cycle, FH loss caused by mutation or transcriptional inhibition is related to the occurrence of a variety of cancers [
26]. Dysregulation of energy homeostasis resulting from FH loss leads to the activation of key oncogenic pathways and transcriptional programs, such as those regulated by HIF, mTOR, and PI3K [
16]. Furthermore, depletion of FH and subsequent accumulation of FA can induce EMT, thereby promoting aggressive tumor characteristics [
18]. Recent studies have demonstrated that dysfunction of the FH gene may adversely impact relapse-free survival and overall survival rates in patients with CRC [
15]. Our findings reveal a significant downregulation of FH expression in CRC tissues compared with normal tissues. Moreover, low expression of FH is positively related to poor prognosis in patients with CRC. Additionally, FH plays a pivotal role in CRC cell proliferation and invasion. Low expression of FH impedes CD8+ T-cell clonal expansion and diminishes the therapeutic efficacy of PD-1 antibodies. These results underscore the crucial involvement of FH in CRC and its potential as a promising biomarker for therapeutic intervention.
Our findings indicate that PCSK9 is a downstream target of FH, and FH can negatively regulate the expression of PCSK9 in CRC. Previous studies have demonstrated that the deletion or pharmacological inhibition of PCSK9 in tumor cells can enhance the anti-tumor activity of CD8+ T cells, subsequently impeding tumor progression [
11]. Simultaneously, PCSK9 plays a crucial role in regulating MHC I levels on cell surfaces, influencing immune infiltration within tumors, and responding to immune checkpoint therapy [
10]. Our findings corroborate previous research and provide additional evidence for the involvement of PCSK9 in immune microenvironment modulation and immunotherapy. These results reveal novel regulatory mechanisms of PCSK9 from the perspective of metabolic reprogramming and lay a solid foundation for future interventional applications of PCSK9 as an immunotherapeutic target.
The nuclear translocation of SREBF1/2 mediated by Ran protein is one of the transcription regulation mechanisms that affect the expression of PCSK9 [
21]. Both in vitro and in vivo transcriptional levels are regulated by SREBPs, with SREBP-2 being the primary regulator responsible for sterol-dependent PCSK9 expression in vivo [
19,
20]. Furthermore, ran proteins facilitate the nuclear translocation of PCSK9. Ran GTPases belong to the Ras superfamily and govern nucleoplasmic transport through nuclear pore complexes while also regulating microtubule polymerization and mitotic spindle formation to control cell cycle progression [
23,
27]. Disruption of Ran expression is implicated across various stages of cancer development, from carcinogenesis to metastasis [
28,
29]. Our findings suggest that SREBF1/2 acts as a transcription factor for PCSK9. Additionally, FH binds to Ran and influences SREBF1/2 nuclear import, resulting in decreased PCSK9 expression. Conversely, knockdown of FH enhances SREBF1/2 nuclear import and upregulates PCSK9 expression. In addition, there is no doubt that the process of CRC progression and treatment resistance is complicated. More detailed relevant mechanisms of FH are indeed worthy of further exploration in the future.
Despite remarkable advancements in immunotherapy, a subset of patients with CRC fail to respond due to limited infiltration of T lymphocytes [
30]. Consequently, there is growing interest in developing rational combinations that can attract T lymphocytes into these tumors. Preclinical and clinical studies have demonstrated efficacy combined with radiation, high-dose IL-2, and IL-10 through the expansion of CD8+ T cells against melanoma and renal cell carcinoma [
31,
32]. However, this therapeutic effect relies on continuous administration of high doses of drugs, which lead to toxicity concerns and hamper the development of tumor immunotherapy combinations. Our findings reveal that combining a PCSK9 inhibitor with PD-1 antibodies promotes CD8+ T cell clonal expansion and significantly enhances tumor immunotherapy in an FH-low expression mouse model of CRC. Considering these results, our findings hold potential for future clinical applications. It is undeniable that the evidence for clinical validation of our findings is limited. Therefore, these findings need to be verified in a larger cohort in the future. Further prospective recruitment research samples were evaluated.