Abstract Background The aberrant upregulation of PVT1 in gastric cancer (GC) has emerged as a critical molecular marker, while its mechanism of remodeling the tumor microenvironment (TME) through metabolic regulation remains unclear. This study aims to unveil the novel mechanism by which PVT1 regulates the fate determination of tumor-associated neutrophils (TANs) via the calcium signaling pathway. Methods Clinical analysis of 90 GC cases revealed a correlation between PVT1 expression and neutrophil infiltration in tumor tissues. Mechanistically, multi-omics integration of transcriptomics and 4D-label-free quantitative proteomics identified PVT1-associated pathways driving GC progression. Functional validation through in vitro and in vivo models demonstrated PVT1’s oncogenic role, while transcriptomic profiling further decoded neutrophil activation mechanisms regulated by PVT1. Results The expression of PVT1 was significantly elevated in GC tissues compared to adjacent non-tumor tissues (p < 0.001), with 73.3% of cases showing an increased proportion of PVT1-positive cells. High PVT1 expression negatively correlated with the infiltration of neutrophils (r = − 0.3554, p = 0.0012), and patients in the PVT1 + CD66b- subgroup exhibited the poorest prognosis, characterized by a median survival of 20.5 months and a mortality rate of 72.5%. Silencing PVT1 upregulated calcium signaling pathway-related genes (RYR2 and RYR3) in GC cells, accompanied by a marked increase in intracellular Ca^2⁺ concentration (p < 0.01). Proteomic analysis revealed that PVT1 knockdown significantly enhanced the secretion of synaptopodin (SYNPO), which demonstrated co-expression and physical interactions with calcium channel proteins RYR2/RYR3. PVT1 knockdown markedly suppressed GC cell proliferation, migration, and invasion (p < 0.05) while elevating the expression of γ-H2AX (p < 0.05). In xenograft models, tumors with PVT1 knockdown displayed reduced volume and decreased Ki67 expression (p < 0.01). Conditioned medium from PVT1-knockdown GC cells promoted neutrophil activation, delayed apoptosis, and amplified intracellular Ca^2⁺ signaling. Exogenous SYNPO supplementation mimicked these effects and attenuated neutrophil replicative senescence by modulating p53 pathway-associated genes (TP53, CHEK1, and SERPINE1). Conclusions Overall, this study provides the first evidence that PVT1 establishes a metabolic checkpoint by inducing calcium overload, thereby regulating both the senescence process and functional phenotype of neutrophils, which provides a novel metabolic intervention target for GC immunotherapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-025-06912-6. Keywords: Gastric cancer, PVT1, Tumor-associated neutrophils, Calcium signaling pathway, Replicative senescence Introduction Gastric cancer (GC) remains one of the most prevalent malignancies, ranking fifth in incidence and fourth as cause of cancer death globally [[40]1]. Although chemotherapy has been the primary treatment for advanced GC for many years, we are now entering an era of more targeted drugs targeting abnormally expressed genes and signaling pathways [[41]2–[42]5]. Efficient cancer management heavily relies on the identification and investigation of key mediators involved in tumorigenesis. With the advent of whole-transcriptome sequencing (WTS) and computational sciences, profound insights into non-coding RNAs have been gained [[43]6]. Abnormally expressed lncRNAs might be potential molecular biomarkers for GC diagnosis. Plasmacytoma variant translocation 1 (PVT1), which is located on chromosome 8q24, was found to be amplified in double minutes of GC cells and overexpressed in GC tissues [[44]7, [45]8]. The level of PVT1 in tissues or in serum can help to assess the risk of lymphatic metastasis in GC patients [[46]9, [47]10]. PVT1 could also regulate the viability, invasion and cell cycle progression of GC cells as a ceRNA [[48]11]. Besides, overexpressed PVT1 could enhance the 5-fluorouracil resistance and anti-apoptosis of GC cells [[49]12]. These findings that elucidate the regulatory mechanisms of PVT1 may provide novel insights for the therapeutics of tumors. However, the exact mechanism of PVT1 in GC still needs to be elucidated. Cancer-related inflammation serves as the primary driver of tumor initiation and progression, facilitating cancer cells to evade immune surveillance and infiltration of diverse immune cells in tumor microenvironment (TME) has been demonstrated to play a pivotal role in tumorigenesis [[50]13, [51]14]. Tumor-associated neutrophils (TANs) are defined as neutrophils that have infiltrated the TME and constitute a significant component of the inflammatory milieu within tumors. The presence of neutrophil infiltration in tumorigenesis is widely recognized as a crucial factor governing tumor progression, angiogenesis, and modulation of immune responses [[52]15, [53]16]. TANs exhibit diverse associations, either promoting or inhibiting the progression of various cancers [[54]17]. The study suggests that neutrophils may undergo phenotypic transformation in response to specific cytokines or metabolites within the TME, thereby directly influencing on tumor cell proliferation and metastasis [[55]18]. The occurrence and prognosis of GC are intricately associated with the infiltration of immune cells within the TME. An increase in infiltrated immune cells is indicative lower incidence of lymph node metastasis and overall survival for GC patients [[56]19]. Notably, elevated levels of neutrophil infiltration have been observed in the cancerous tissues of individuals diagnosed with GC, accompanied by an extended lifespan of neutrophils activated by GC cells [[57]20]. Senescence-targeting may ultimately affect neutrophil states. In turn, the identification of specific senescence-associated neutrophil subsets may open a new avenue of treatments that limit senescence and cancer by targeting neutrophils, without affecting neutrophilic response against pathogens [[58]21]. An increasing number of studies have unveiled the involvement of abnormal gene expression in GC in regulating neutrophil infiltration [[59]22–[60]24]. Studies indicate that, when constructing a risk prediction model for GC based on TCGA dataset, PVT1 was identified as a crucial gene for predicting immune cell infiltration (T cells, monocytes, and neutrophils) and patient prognosis in GC [[61]25]. Therefore, gaining a comprehensive understanding of the regulatory role of neutrophils within the TME in GC and exploring the association between PVT1 and neutrophils could offer novel insights for therapeutic interventions and prognostic assessments in GC. Materials and methods Gastric cancer tissues chip of patients, blood in healthy population The tissue chip consisted of 90 cases of human gastric cancer tissues and their corresponding adjacent tissues, totaling 180 sites (number: HStmA180Su11, lot number: XT17-035), obtained from Shanghai Outdo Biotech Co., Ltd. (Shanghai, China). The chip was accompanied by matching HE staining information. Among the samples, there were 32 patient fatalities and 58 survivors. The peripheral blood samples were obtained from 10 healthy subjects at the Second Affiliated Hospital of Harbin Medical University following routine examination, collected in EDTA-K3 anticoagulant tubes. This study received ethical approval from the Medical Ethics Committee of the Second Affiliated Hospital of Harbin Medical University prior to commencement (Approval number: YJSKY-2022-517). Cell culture, transfection and treatment The human gastric mucosal epithelial cell line GES-1 and the human gastric cancer cell lines HGC-27, AGS, NCI-N87 and MKN-45 were maintained in accordance with a previous report [[62]26]. GC cell line SNU-16 was purchased from American Type Culture Collection (ATCC, Manassas, VA, USA) and cultured in RPMI-1640 (Gibco BRL, Waltham, MA, USA) with 10% fetal bovine serum at 5% CO[2] in a 37 ℃ incubator. The genotypically validated of cell lines were authenticated by Beijing Microread Genetics Co., Ltd. (Beijing, China) using short tandem repeat (16-STR) analysis. The antisense oligonucleotide (ASO) for PVT1 and negative control (NC) were purchased from Guangzhou RiboBio Co., Ltd. (lnc6180719010843-1-5, Guangzhou, China). Cells were seeded in six-well plates, and transfected with PVT1-ASO or NC using riboFECTTMCP Transfection kit (C10511-1, RiboBio, Guangzhou, China) according to the instructions. The transfection efficiency was measured by real-time PCR at 48 h and 72 h after transfection. For tumorigenicity assay in vivo, lentiviral particles for PVT1-shRNA(Catalog #: LPP-CS-SH3617-LVRU6GP-a-500) and for negative control (Catalog #: LPP-CSHCTR001-LVRU6GP-500) were purchased and infected to HGC-27 cells using the Lentiviral granulosa Cell Infection Experiment User's Manual from GeneCopoeia, Inc. Recombinant Human TNF-α was purchased from Beyotime Biotechnology (p5318, Shanghai, China) and recombinant Human SYNPO protein was purchased from Zeye Biotechnology (ZY685Hu01P, Shanghai, China). Isolation, extraction, and identification of neutrophils from healthy individuals The isolation of peripheral blood neutrophils was conducted following the protocol from the human peripheral blood neutrophil isolation kit (Solarbio, P9040, Beijing, China). Briefly, 4 mL of reagent A was layered with 2 mL of reagent C in a 15 mL tube, followed by overlaying 5 mL of anticoagulated blood. The mixture was centrifuged at 2500 rpm for 30 min at room temperature to separate the mononuclear cell layer (upper) and neutrophil layer (lower). Neutrophils were aspirated, washed with 10 mL washing solution (1200 rpm for 10 min), lysed with 2 mL lysis buffer for 10 min, neutralized with 5 mL washing solution, centrifuged, and resuspended in culture medium. After isolation, cell viability was assessed using the Trypan Blue staining kit (Beyotime, C0011, Shanghai, China) and an automated cell counter (LUNA-II™, Logos Biosystems, Shanghai, China). Neutrophil purity was determined by Wright’s stain solution (Solarbio, G1040, Beijing, China), followed by observation and photography under an optical microscope (ZEISS Axio Vert.A1, Germany). Dual hybridization of fluorescence in situ hybridization (FISH) and Immunofluorescence (IF) To account for inter-individual variability in GC patients, the percentage of positive cells was used to evaluate the expression levels of PVT1 and neutrophil infiltration in both tumor tissues and adjacent tissues obtained from the same GC patient. The tissues were deparaffinized and subjected to antigen retrieval according to standard protocols. Subsequently, the samples were digested with proteinase K, pre-hybridized, and hybridized with a PVT1 fluorescent probe (1:67 dilution, Genepharma) combined with the RNA FISH SA-Biotin amplification system kit (Genepharma). The tissues area was sealed at room temperature, followed by the addition of the anti-CD66b primary antibody (1:500, Abcam, ab300122). After overnight hybridization at 4 ℃, incubation with FITC-labeled goat anti-rabbit secondary antibody (1:500, Immunoway, RS0004) was performed. Finally, the DAPI dye solution was added. The tissues were examined using a fluorescence microscope (NIKON, YS100, Japan) and images were acquired. CD66b-positive cells emitted green fluorescence, PVT1 produced red fluorescence, and DAPI-stained nuclei showed blue fluorescence. Whole transcriptome sequencing (WTS) WTS was conducted by Shanghai Outdo Biotech Co., Ltd. (Shanghai, China). Briefly, the total RNA was extracted using the RNeasy Mini Kit (Qiagen, 74106, Germany). Sequencing libraries were generated using VAHTS^™ Stranded mRNA-seq Library Prep Kit for Illumina (Vazyme, NR612, Nanjing, China) according to the manufacturer’s instructions. The sequencing was performed on Illumina NovaSeq 6000 platform (Illumina, San Diego, CA, USA). Raw data were processed through Seqtk. In this step, the raw data were cleaned by removing adapter sequences, contaminant reads, and low-quality bases. Additionally, the Q30, and GC content were calculated to estimate the quality of clean reads. Next, the clean reads were aligned to the reference genome (GRCh38) using HISAT2. The Read Counts of transcripts and ncRNAs were calculated by String Tie. And the expression of mRNA and lncRNA was normalized to FPKM. FPKM refers to the number of fragments per kilobase length from a gene per million fragments mapped considering the effect of both sequencing depth and gene length on fragments count. The sequencing depth (in total reads) of WTS is summarized in Table S1. 4D-Label-free quantitative proteome analysis by LC–MS/MS LC–MS/MS was conducted by Shanghai Outdo Biotech Co., Ltd. (Shanghai, China). It comprised protein extraction and peptide hydrolysis, as well as LC–MS/MS data collection. The samples were lysed by SDT (4% (w/v) SDS, 100 mM Tris–HCl pH 7.6, 0.1 M DTT). Each sample was separated using Brucker nanoElute, a ultra performance liquid chromatography (UPLC) liquid phase system with a nanoliter flow rate. Samples separated by nanoflow ultra-high performance liquid chromatography (nanoUHPLC) were analyzed by data-dependent acquisition (DDA) mass spectrometry using timsTOF Pro mass spectrometer (Bruker). The detection mode was positive ion mode, and the ion source voltage was set to 1.5 kv. Time of flight (TOF) was used for detection and analysis of MS and MS/MS. The mass spectrum scanning range was set to 100–1700 m/z. Parallel accumulation aerial fragmentation (PASEF) mode was used for data acquisition. Real time quantitative polymerase chain reaction (Real-time PCR) Total RNA were extracted from cells using the SevenFast^™ Total RNA Extraction Kit (SEVEN, SM130, Beijing, China), and first-strand cDNA was synthesized using the All-in-one First Strand cDNA Synthesis Kit (SEVEN, SM131, Beijing, China) according to the manufacturer’s protocol. The cDNA was subjected to real-time PCR in SLAN-96P real-time PCR system (Hongshitech, Shanghai, China) using 2 × SYBR Green qPCR Master Mix (SEVEN, SM133, Beijing, China) with fluorescence detection. Genes expression levels were normalized to β-actin by the 2^−△△Ct method. The sequence of primers are listed in Table S2. Western blot analysis Total protein was extracted from the GC cells using RIPA buffer (Beyotime, P0013B). The protein samples were separated by sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE), and then transferred onto polyvinylidene difluoride (PVDF) membranes. The membranes were blocked with 5% nonfat milk for 2 h at room temperature and then hybridized with primary antibodies: anti-Synaptopodin (1:1,000; Affinity, DF12173), anti-MMP9 (1:1,000; Beyotime, AF5234) and anti-β-actin (1:10,000; Affinity, AF7018) overnight at 4 °C, followed by incubation with a horseradish peroxidase HRP-conjugated goat anti-rabbit secondary antibody (1:10,000; ZSGB-BIO, ZB-5301). Blots were imaged using chemiluminescence imaging system (ChemiScope 6300, CLiNX). CCK-8 assay and cell viability analysis Cells were seeded into 96-well plates at a density of 2000 cells per well. Cell proliferation was monitored daily over 5 consecutive days, while viability was assessed at 16 h or 24 h using the Cell Counting Kit-8 (SEVEN, SC119, Beijing, China) according to the manufacturer’s protocol. Optical density (OD) was measured at 450 nm using a microplate reader (BIO-RAD, iMark, Japan). Each experiment included six biological replicates, and data are expressed as mean ± standard deviation (SD). graphic file with name d33e590.gif Colony formation assay Cells were seeded in 6-well plates at a density of 200 cells per well. Following 10 days of culture, the medium was removed, and cells were washed twice with phosphate-buffered saline (PBS). Subsequently, cells were fixed with methanol for 20 min at room temperature, followed by PBS washing and staining with Giemsa solution (Beyotime, C0133, Shanghai, China) for 30 min. The assay was performed in three independent replicates. Colonies were imaged and quantified with ImageJ software. Wound healing assay and transwell migration assay Cell migration ability was evaluated through wound healing and transwell assays. Wound healing assay Cells were seeded in 6-well plates and cultured until reaching 90% confluency. A uniform scratch was created using a sterile 200 μL pipette tip. After washing with PBS to remove detached cells, cells were maintained in medium containing 2% FBS (to suppress proliferation). Wound closure was monitored at 0 h and 24 h using an inverted microscope (ZEISS, Axio Vert.A1, Germany). Transwell migration assay Transwell chambers (8 μm pore size, Corning, NY, USA) were placed in a 24-well plate. The upper chamber was loaded with 200 μL serum-free medium containing 3 × 10^4 cells, while the lower chamber contained 800 μL medium with 10% FBS as a chemoattractant. After 24 h incubation, non-migrated cells on the upper membrane surface were removed with a cotton swab. Migrated cells on the lower surface were fixed with 4% methanol for 20 min, stained with Giemsa solution (Beyotime, C0133, China) for 30 min, and imaged under an optical microscope (ZEISS, Axio Vert.A1, Germany). IF, apoptosis/necrosis, and Fluo-4 calcium assays IF Cells were cultured in 6-well plates. DNA damage was assessed using the DNA Damage Assay Kit (Beyotime, C2035S) according to the manufacturer’s protocol. For protein detection, cells were fixed with 4% paraformaldehyde, permeabilized with 0.1% Triton X-100, and blocked with 5% BSA. Subsequently, cells were incubated with anti-γ-H2AX antibody (Beyotime, C2035S-4), anti-SYNPO (1:500, Affinity, DF12173), anti-SERPINE1 (1:200, Immunoway, YT3569), anti-TP53 (1:200, Immunoway, YT3528) and anti-CHEK1 (1:200, Immunoway, YT0902) overnight at 4 °C. After washing, cells were incubated with either Alexa Fluor 488-conjugated anti-rabbit IgG (Beyotime, C2035S-5) or Goat Anti Rabbit IgG (H + L) (AbFluor 594) (1:200, Immunoway, RS3611) for 1 h at room temperature. Cells were counterstained with DAPI (Beyotime, C1002) or Phalloidin (YEASEN, 40774ES03) and imaged using a fluorescence microscope (NIKON, YS100, Japan). Apoptosis and necrosis assay Cell apoptosis and death was analyzed using the Apoptosis and Necrosis Assay Kit (Beyotime, C1056). Briefly, cells were stained with Hoechst 33,342 (10 μg/mL) and propidium iodide (PI) (5 μg/mL) in staining buffer for 20 min at 37 °C. After PBS washing, apoptotic (Hoechst-positive) and necrotic (PI-positive) cells were quantified under a fluorescence microscope (NIKON, YS100, Japan). Fluo-4 calcium assay Intracellular calcium levels were measured with the Fluo-4 Calcium Assay Kit (Beyotime, S1061S). Cells were loaded with Fluo-4 AM in staining solution at 37 °C for 30 min. Green fluorescence signals (Ex/Em = 490/525 nm) were captured using a fluorescence microscope (NIKON, YS100, Japan). Data were normalized to baseline fluorescence intensity. Tumorigenicity assay in vivo Female BALB/c Nude mice aged 4–6 weeks were purchased from Beijing Vital River Laboratory Animal Technology Co., Ltd. (Beijing, China) and were housed under pathogen-free conditions. Mice were randomly divided into NC groups (n = 5) and experimental groups (n = 5). Subcutaneous injections of HGC-27 cells (1 × 10^7) with or without stable PVT1 silencing were administered to establish xenografts. Tumor growth was monitored for 4 weeks before mice were sacrificed. Mice were sacrificed by cervical dislocation and tumor tissues were obtained for pathological examination. Paraffin sections of tumors were prepared by conventional methods, and the tumor tissues were observed and examined via hematoxylin and eosin (HE) staining. Ki67 expression was assessed by immunohistochemistry using an anti-Ki67 antibody (1:100, Affinity, AF0198) in tumor tissues among different groups. All animal studies were performed in accordance with the ethical principles of animal protection and welfare, and approved by the Ethics Committee of the Second Affiliated Hospital of Harbin Medical University (Ethical review approval number: YJSDW2022-252). Statistical analysis Data are expressed as mean ± SD and analyzed using GraphPad Prism software (v10.1.2). Comparisons between two groups were performed using the Wilcoxon rank-sum test or Student’s t-test. A p < 0.05 was considered statistically significant unless otherwise stated. Sequence data visualization was generated by [63]https://www.bioinformatics.com.cn (last accessed on 20 Feb 2023), an online platform for data analysis and visualization. Results PVT1 expression negatively correlates with neutrophil infiltration and predicts poor prognosis in gastric cancer The correlation between PVT1 and neutrophil infiltration was observed in gastric cancer (GC). The somatic copy number alterations (SCNA) analysis module of the TIMER database was utilized to compare neutrophil infiltration levels in GC tissues with different PVT1 SCNA statuses. Paradoxically, high amplification, arm-level gain and deletion of PVT1 all seem to be associated with the decreased neutrophil infiltration compared to the diploid/normal status (Fig. [64]1A). Therefore, RNA FISH for PVT1 and IF for CD66b (Neutrophils marker) were performed in 90 GC cases and paired adjacent tissues to investigate the differential expression of PVT1 and neutrophil infiltration (Fig. S1A). Notably, CD66b exhibited robust specificity for neutrophils in GC tissues (Fig. S1B). The findings demonstrated a significant increase in the proportion of PVT1-positive cells in GC tissues compared to non-adjacent cancerous tissues (Fig. [65]1B, p < 0.001), and 66 (73.3%) of the patients exhibited a higher abundance of PVT1-positive cells in GC tissues compared to the adjacent tissues (Fig. [66]1D). In contrast, 53 (61.1%) of the patients showed fewer CD66b-positive cells in GC tissues compared to adjacent tissues, while 3 (3.3%) exhibited deficient expression of CD66b in tissues (Fig. [67]1E). However, no significant difference in CD66b expression was observed between GC and non-adjacent cancerous tissues (Fig. [68]1C, p > 0.05). These findings suggest a decrease in the number of neutrophil infiltrates in GC tissues exhibiting high expression levels of PVT1 (Fig. S1C). Fig. 1. [69]Fig. 1 [70]Open in a new tab The expression of PVT1 demonstrated an inverse correlation with neutrophil infiltration in GC. A Box plots are presented to show the distributions of neutrophil infiltration levels for each copy number status of PVT1 in GC. The infiltration level for each SCNA category is compared with the normal using a two-sided wilcoxon rank-sum test. B Positive cells of PVT1 in tissues. C Positive cells of CD66b in tissues. D The positive cell rate of PVT1 in 90 GC tissues. E The positive cell rate of CD66b in 90 GC tissues. F The proportion in combinations of PVT1 expression and neutrophil infiltration. The cases of GC with a higher number of PVT1 positive cells in cancer tissues compared to adjacent tissues were designated as PVT1 +, while those with a lower number were designated as PVT1-. Similarly, GC cases with a higher number of CD66b-positive cells in cancer tissues compared to adjacent tissues were labeled as CD66b +, while those with lower or no expression were labeled as CD66b-. G The representative cases in combinations of PVT1 expression and neutrophil infiltration. H The proportion of survival and death in groups. I The mean survival time in groups. J The fraction survival proportion in groups. K Correlation analysis was performed on the PVT1 + CD66b- group. (ns no significance, **p < 0.01, and ***p < 0.001) Four PVT1/CD66b expression patterns were identified: PVT1 + CD66b + (26 cases, 28.9%), PVT1 + CD66b- (40 cases, 44.4%), PVT1-CD66b + (8 cases, 8.9%), and PVT1-CD66b- (16 cases, 17.8%). Notably, the combination of PVT1 + CD66b- exhibited the highest proportion among all observed patterns in GC tissues (Fig. [71]1F and G). Besides, of the 40 patients in PVT1 + CD66b- group, 11 survived and 29 died, accounting for 72.5% of deaths (Fig. [72]1H), the median survival time was 20.5 months (Fig. [73]1I). This suggests that GC patients with high PVT1 expression and low neutrophil infiltration have a poor prognosis (Fig. [74]1J). The Pearson correlation analysis revealed a negative correlation between PVT1 expression and neutrophil infiltration in PVT1 + CD66b- group (r = − 0.3554, p = 0.0012) (Fig. [75]1K). The present findings further validate the inverse correlation between PVT1 expression and neutrophil infiltration in GC. Whole transcriptome sequencing revealed activation of the calcium signaling pathway following PVT1 silencing in gastric cancer cells To investigate PVT1’s regulatory mechanism in GC, we first evaluated PVT1 expression levels across GC cell lines and selected HGC-27 for further experiments (Fig. S2A, p < 0.001). PVT1 knockdown in HGC-27 cells (Fig. S2B, p < 0.001) enabled identification of differential expressed genes (DEGs) through whole transcriptome sequencing (WTS) between negative control (NC) and PVT1-knockdown groups. The quality control of sequencing data is shown in Fig. S3A-3B. Differential expression analysis revealed 15 PVT1 transcript variants, with 11 downregulated (PVT1-229, PVT1-233, PVT1-234, PVT1-239, PVT1-258, PVT1-273, PVT1-287, PVT1-304, PVT1-329, PVT1-333, PVT1-367) were shown in Fig. [76]2A, Fig. S3C. The overall distribution of DEGs were visually shown in the volcano map (Fig. [77]2B). After knockdown of PVT1 in GC cells, the expression of histone 3′ UTR stem-loop region was increased (Fig. S3D). The peak chromosome distribution map has exhibited the distribution of DEGs on each chromosome and changes in abundance of tRNA expression (Fig. S3E-3F). A total of 1,126 DEGs (968 upregulated, 158 downregulated;|log2FC|> 2.0, p < 0.05) were visualized via clustering heatmap (Fig. [78]2C). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses highlighted enrichment in calcium channel complexes (Fig. [79]2D, Fig. S3G-3H), and calcium signaling pathway (Fig. [80]2E). Fluo-4 AM assays confirmed increased intracellular Ca^2⁺ concentration in PVT1-ASO-treated cells (Fig. [81]2F, p < 0.01). Transcriptome sequencing further demonstrated calcium pathway activation through upregulated RYR2 and RYR3 expression (Fig. [82]2G), validated by PCR in PVT1-knockdown cells (Fig. [83]2H, p < 0.05). Fig. 2. [84]Fig. 2 [85]Open in a new tab Activation of calcium signaling pathway in GC cells. A Differential expression of PVT1 transcripts variants. B The overall distribution of DEGs in the volcano map. C The clustering heatmap visualization of DEGs. D GO and E KEGG pathway enrichment analyses of DEGs. F The intracellular Ca^2+ concentration in HGC-27 cells (NC vs PVT1-ASO) measured by fluorescence intensity of Fluo-4 AM calcium indicator. G Upregulation of RYR2 and RYR3 in calcium signaling pathway revealed by transcriptome sequencing. H Expression levels of RYR2 and RYR3 validated by real-time PCR. (ns no significance, *p < 0.05, **p < 0.01, and ***p < 0.001) To further explore the contribution of PVT1 knockdown to the phenotypic plasticity of GC cells, we performed in vitro and in vivo experiments. Cell proliferation (Fig. [86]3A) and colony formation (Fig. [87]3B, p < 0.05) were both significantly decreased after PVT1 knockdown in HGC-27 cells. Transwell and wound healing assays revealed that the migration (Fig. [88]3C, p < 0.05) and invasion (Fig. [89]3D, p < 0.01) capabilities of HGC-27 cells were markedly impaired following PVT1 knockdown. MMP9 expression was also downregulated upon PVT1 silencing (Fig. [90]3E, p < 0.01). Genomic stability assessment via γ-H2AX assay showed substantially increased nuclear green fluorescence intensity in PVT1-knockdown cells (Fig. [91]3F, p < 0.05). In subcutaneous xenograft models established using PVT1-shRNA-transfected HGC-27 cells (Fig. S2C), tumor volume in the PVT1-shRNA group was significantly smaller than the NC group (Fig. [92]3G), with immunohistochemical analysis showing reduced Ki67 expression in tumor tissues (Fig. [93]3H, p < 0.01). These findings demonstrate that PVT1 knockdown critically regulates multiple malignant phenotypes of GC cells. Fig. 3. [94]Fig. 3 [95]Open in a new tab PVT1 silencing modulates phenotypic plasticity in GC cells. A Proliferation capacity of HGC-27 cells post-PVT1-ASO transfection assessed by CCK-8 assay. B Clonogenic ability of PVT1-knockdown HGC-27 cells. C Transwell and D wound healing assays evaluating migratory capacity. E Western blot analysis of MMP9 expression pre- and post-PVT1 knockdown. F Genomic stability assessment via γ-H2AX foci formation assay, the average number of γ-H2AX foci per cell was obtained by counting in 100 cells. G Subcutaneous xenograft model (n = 5/group) showing tumor growth inhibition in PVT1-ASO group, with HE staining confirming malignancy (100 ×), and tumor volume were calculated as volume = 0.5 × a × b^2 mm^3, a: length, b: width. H Ki67 expression levels across groups quantified by immunohistochemistry (100 ×). (ns no significance, *p < 0.05, **p < 0.01, and ***p < 0.001) Proteomic profiling revealed that SYNPO secretion may be a key activator of calcium signaling in gastric cancer cells 4D-label-free quantitative proteomic analyses were performed via LC–MS/MS on cell culture supernatants from NC and PVT1-knockdown GC cells. We detected 18,470 proteins and obtained 188,030 peptide spectra, with 9,122 proteins (92,250 unique peptides) identified in the NC group and 9,348 proteins (95,780 unique peptides) in the PVT1-ASO group (Fig. S4A-4D). The overall distribution of differentially expressed proteins (DEPs) were shown in the volcano plot (Fig. [96]4A), of which 460 DEPs were identified with p < 0.05. Further, 46 DEPs with|log2FC|> 1.0 and p < 0.05 (37 DEPs up-regulated and 9 DEPs down-regulated) were identified and visualized in the hierarchical clustering heat map (Fig. [97]4B). InterProScan domain prediction showed significant enrichment of C2H2-type zinc fingers and EGF-like domains (Fig. [98]4C). GO analysis demonstrated DEP enrichment in clathrin-associated vesicular structures (Fig. [99]4D), while KEGG pathway analysis highlighted involvement in endoplasmic reticulum protein processing and endocrine-regulated calcium reabsorption (Fig. [100]4E). Fig. 4. [101]Fig. 4 [102]Open in a new tab 4D-Label-free quantitative proteome analysis in GC cells. A Volcano plot showing the overall distribution of DEPs. B Hierarchical clustering of identified DEPs. C Top 20 statistically enriched protein domain classifications. D GO and E KEGG enrichment analyses of DEPs. F Venn diagram of transcriptomic-proteomic DEP intersections. G Expression-level comparison of overlapping DEPs. H PVT1-SYNPO correlation analysis. I Western blot analysis of SYNPO expression in GC cells. J GENEMANIA-based interaction network (SYNPO, RYR2, RYR3) showing co-expression, physical interactions, and correlations ([103]http://genemania.org/search/homo-sapiens/). (*p < 0.05) Correlation analysis between DEGs and DEPs, based on mRNA-protein translational relationships, identified six genes (SYNPO, MBTPS1, APOOL, ENO2, THBS3, CAPN7) (Fig. [104]4F). SYNPO exhibited significant upregulation at both transcriptional and protein levels (Fig. [105]4G). Furthermore, correlation analysis showed that SYNPO seems to be negatively correlated with PVT1 expression in GC cells (r = − 0.216, p < 0.001) (Fig. [106]4H), which was consisted with the elevated SYNPO expression in PVT1 knockdown GC cells (F[107]ig. [108]4I, Fig. S4E). Notably, SYNPO showed positive correlations with calcium channel proteins RYR2 (r = 0.412, p < 0.001) and RYR3 (r = 0.481, p < 0.001). GeneMANIA database analysis ([109]http://genemania.org/) further indicated direct/indirect interactions between SYNPO and RYR2/RYR3 through co-expression networks and physical binding (Fig. [110]4J). These findings collectively suggest that SYNPO upregulation may trigger calcium signaling activation in GC cells. Whole transcriptome sequencing revealed gene expression profiles in neutrophils activated by PVT1-knockdown gastric cancer cells To investigate the negative correlation between PVT1 expression and neutrophil infiltration in GC tissues, we developed an in vitro tumor microenvironment (TME) model. Peripheral blood neutrophils were isolated from healthy donors (Fig. S1D-1F) and co-cultured with 48 h conditioned medium (CM) from HGC-27 GC cells (with/without PVT1 knockdown). The results demonstrated that neutrophils co-cultured with GC CM for 16 h exhibited aggregation. A subset of cells transitioned from round/oval morphology to a polarized phenotype with surface irregularities and pseudopodia formation (Fig. [111]5A). Cell viability assays showed prolonged survival of neutrophils exposed to CM from PVT1-knockdown GC cells compared to control CM (Fig. [112]5B, p < 0.05). Combined apoptosis/necrosis assays further confirmed reduced cell death in the PVT1-knockdown group (Fig. [113]5C). Hoechst staining detected neutrophil extracellular traps (NETs) in CM-treated neutrophils, with TNF-α as a positive control (Fig. [114]5D). These findings suggest PVT1 knockdown in GC cells enhances neutrophil survival and activation through secreted factors. Fig. 5. [115]Fig. 5 [116]Open in a new tab PVT1 knockdown in GC cells enhances neutrophil activation via secreted factors. A Neutrophil morphology before and after CM induction. B Viability comparison of neutrophils exposed to CM from NC vs PVT1-knockdown HGC-27 cells. C Apoptosis/necrosis staining of CM-induced neutrophils. D NETs formation assay with TNF-α positive control. (*p < 0.05) To investigate the mechanism of PVT1-mediated neutrophil activation, we performed WTS on healthy donor neutrophils (n = 3) exposed to CM (Fig. S5A-5C). Sequencing identified 16,024 DEGs, with volcano plot visualization showing distinct expression patterns (Fig. [117]6A).Using stringent thresholds (p < 0.05,|log2FC|> 2), we identified 714 DEGs (384 upregulated, 330 downregulated) associated with neutrophil activation, as demonstrated by hierarchical clustering (Fig. [118]6B). GO enrichment analysis revealed three key functional categories: (1) Biological processes: cell adhesion mediated by integrin, regulation of chromatin silencing, chromatin assembly, replicative senescence and negative regulation of chromatin silencing. (2) Cellular components: microtubule end, sarcoplasmic reticulum, microtubule plus − end and sarcoplasmic reticulum membrane. (3) Molecular functions: sodium antiporter activity and calcium: cation antiporter activity (Fig. [119]6C). KEGG pathway analysis identified two mechanistically relevant pathways for further investigation: (1) Calcium signaling pathway (aligned with cation transport functions in GO). (2) p53 signaling pathway (consistent with senescence regulation in GO) (Fig. [120]6D). These pathways were prioritized for further investigation based on mechanistic relevance. Fig. 6. [121]Fig. 6 [122]Open in a new tab Transcriptomic profiling of neutrophils induced by GC CM. A Volcano plot displaying the overall distribution of DEGs. B Hierarchical clustering of DEGs. C GO enrichment analysis (biological processes, cellular components, molecular functions). D KEGG pathway enrichment analysis Suppression of PVT1 in gastric cancer cells enhances neutrophil calcium signaling and delays replicative senescence The analysis showed that the DEGs in the expression profile of neutrophils involved in the calcium signaling pathway, covering the regulation of promoting or inhibiting the increase the concentration of cytoplasmic Ca^2+ (Fig. [123]7A). The cells of each group were stained with Fluo-4 AM, revealing an increased cytoplasmic calcium signal in neutrophils subsequent to co-culturing with PVT1-knockdown GC cells. (Fig. [124]7B). Fig. 7. [125]Fig. 7 [126]Open in a new tab Mechanistic validation of calcium and p53 signaling in neutrophils. A Regulatory node mapping in calcium signaling pathway. B Intracellular Ca^2⁺ levels measured by Fluo-4 AM fluorescence intensity. C Dose-dependent Ca^2⁺ response to SYNPO. D SYNPO internalization (red) and F-actin (green) via immunofluorescence. E Venn diagram of p53 pathway-senescence gene intersection. F Transcriptome sequencing analysis of TP53, CHEK1, and SERPINE1 RNA levels. G Immunofluorescence of TP53, CHEK1, and SERPINE1 protein expression Given the observed increase in SYNPO secretion from PVT1-knockdown GC cells, we validated its role using recombinant human SYNPO protein. Neutrophils exposed to 1.0 ng/mL SYNPO showed maximal calcium signal enhancement (Fig. [127]7C). Immunofluorescence confirmed SYNPO internalization by neutrophils, with signal intensity correlating with exogenous SYNPO concentration (Fig. [128]7D). These findings suggest that SYNPO upregulation in PVT1-knockdown GC CM may regulate neutrophil calcium signaling. Intersection analysis identified TP53, CHEK1, and SERPINE1 through overlapping replicative senescence genes and p53 pathway DEGs (Fig. [129]7E). Notably, the expression levels of p53 and CHEK1 were found to be decreased, while the expression level of SERPINE1 was observed to increase (Fig. [130]7F). Consistent with the transcriptomic sequencing results, immunofluorescence analysis also revealed a significant decrease in TP53 and CHEK1 expression levels in neutrophils from both the co-cultured group consisting of PVT1-knockdown GC cells and the SYNPO-supplemented group, while there was an observed increase in SERPINE1 expression (Fig. [131]7G). Meanwhile, clinical correlation analysis revealed a significant negative association (r = − 0.307, p < 0.001) between SYNPO expression and CHEK1 expression, as well as a positive correlation (r = 0.296, p < 0.001) with SERPINE1 expression in GC tissues (Fig. S5D-5F). The findings of this study suggest that co-culture with PVT1-knockdown GC cells delays neutrophil replicative senescence, and the increased presence of SYNPO in the supernatant of PVT1-knockdown GC cells may be serves as one of the regulatory factors implicated in this process. Discussion While anti-angiogenesis and immunotherapy have advanced GC treatment [[132]27], tumor heterogeneity and dynamic TME remodeling continue to impede therapeutic efficacy [[133]13, [134]28]. Recent investigations reveal various TME remodeling modulators. For instance, CD4⁺ T cells can reshape the TME following oncogene inactivation [[135]29], while carcinogenic EGFR signals remodel the TME and trigger immune escape mechanisms [[136]30]. Similar to other tumors, GC occurrence and prognosis correlate strongly with immune cell infiltration. Elevated immune cell infiltration levels correlate with increased lymph node metastasis and reduced overall survival rates in GC [[137]19]. TANs, referring to neutrophil infiltration into the TME, represent a pivotal component of the tumor inflammatory milieu [[138]31]. Studies demonstrate that RAI14 gene and Notch receptor expression in GC correlate with neutrophil recruitment in tissues, indicating unfavorable prognosis in GC patients [[139]23, [140]24]. Neutrophils constitute a prominent population of infiltrating immune cells within the TME of GC [[141]20]. To elucidate the correlation between PVT1 expression and neutrophil infiltration in GC tissues, we selected CD66b (a specific neutrophil marker [[142]32]) and employed IF combined with FISH. In 90 GC tissue samples exhibiting PVT1 and CD66b co-detection, we observed diverse PVT1 expression patterns with neutrophil infiltration. Intriguingly, the PVT1⁺CD66b⁻ subgroup accounted for the highest proportion (44.4%). Moreover, GC patients with high PVT1 expression and low neutrophil infiltration displayed poor prognosis. Our findings revealed a negative correlation between PVT1 expression levels and neutrophil infiltration density. Mechanistically, PVT1 silencing induced cytoplasmic calcium overload. Our study suggests that this phenomenon may result from SYNPO upregulation, which further activates endoplasmic reticulum (ER) Ca^2⁺ release channels (RYR2 and RYR3), thereby promoting Ca^2⁺ efflux. These findings indicate that PVT1 knockdown in GC cells disrupts calcium homeostasis. As a ubiquitous signaling molecule, intracellular Ca^2⁺ controls fundamental cellular processes including proliferation, differentiation, and cell death [[143]33, [144]34]. Consequently, ER Ca^2⁺ homeostasis maintenance is critical for cellular survival [[145]35]. Notably, calcium overload can induce immunogenic cell death through tumor-associated antigen release while simultaneously promoting calreticulin exposure an "eat me" signal that enhances immunotherapy via Ca^2⁺-mediated immune cell recruitment [[146]36, [147]37]. Intriguingly, recent preclinical models utilizing CaCl₂/CaCO₃ mixtures demonstrated remarkable anti-tumor efficacy through calcium overload induction [[148]38]. Importantly, this study also noted that modulating cellular calcium homeostasis could synergize with calcium signaling-targeting drugs to enhance therapeutic outcomes [[149]38]. Therefore, we speculate that PVT1 may play an important role in the alleviation of calcium overload alleviation during GC progression. As expected, PVT1 knockdown in HGC-27 cells significantly inhibited malignant phenotypes in vitro and suppressed tumor growth in vivo. Surprisingly, reduced PVT1 expression markedly elevated γH2AX (a DNA damage biomarker) nuclear levels in GC cells, indicating enhanced genomic instability (GI) [[150]39]. Increasing evidence showed that point mutations or genome rearrangements caused by defects in genome maintenance, can cause neo-antigens presentation and thus be recognized by immune cells, making tumor cells susceptible to recognition by the immune system [[151]40]. This suggests that PVT1 knockdown in GC cells may promote immune cell infiltration. Subsequently, we conducted in vitro observations to assess neutrophil activation under PVT1-regulated TME conditions through co-culture systems. After 16 h co-culture with GC cells culture supernatants, both experimental groups exhibited significant neutrophil morphological changes. Notably, neutrophils exposed to PVT1-knockdown GC cell supernatants showed reduced apoptosis/necrosis rates and extended survival duration. Studies have demonstrated that the half-life of neutrophils in the systemic circulation of healthy individuals is approximately 7 h [[152]41], while cancer patients may exhibit a prolonged half-life of up to 17 h [[153]42]. These results indicate successful in vitro replication of PVT1-regulated TME, with the PVT1-deficient GC microenvironment showing enhanced neutrophil survival capacity. GO and KEGG pathway enrichment analyses revealed that DEGs in activated neutrophils were predominantly enriched in calcium signaling pathways and cellular senescence-related pathways. Intracellular calcium homeostasis is mechanistically linked to cellular senescence. For instance, EDTA-mediated calcium chelation inhibits DNA synthesis in phytohemagglutinin (PHA)-stimulated lymphocytes, suggesting cytoplasmic calcium influx regulates proliferation. Senescent WI-38 fibroblasts exhibit either intracellular Ca^2⁺ store release or reduced extracellular inflow, implying diminished cytoplasmic Ca^2⁺ levels may suppress proliferation and accelerate aging [[154]43, [155]44]. In this study, we observed co-culture with PVT1-knockdown GC cells supernatants augmented cytoplasmic Ca^2⁺ signaling in neutrophils, indicating a potential enhancement in intracellular Ca^2+ flux, augmented release of calcium stores, or delayed expulsion of Ca^2+ to compensate for cytoplasmic calcium deficiency during cellular senescence, which is consistent with the phenotype associated with aging of neutrophils. By integrating the findings of quantitative proteomic analysis, our results suggest that increased expression of SYNPO protein in PVT1-knocked down GC cells and subsequent phagocytosis by neutrophils may potentially act as regulatory factors mediating enhanced cytoplasmic Ca^2+ signaling in neutrophils. Cell senescence represents a prolonged and irreversible state of the cell cycle arrest [[156]45]. Under normal physiological conditions, proliferating cells undergo follow regular cell cycle progression [[157]46]. It has been reported that the aging process in neutrophils may impact their immunosuppressive effect within tumors [[158]47], and potentially compromise their ability to release NETs [[159]48]. Further analysis of the expression of TP53, CHEK1, and SERPINE1 in the senescence pathway revealed a gradual decrease in p53 and CHEK1 expression in neutrophils co-cultured with supernatant and SYNPO protein derived from knockdown PVT1 GC cells. Conversely, there was an observed increase in SERPINE1 expression. The P53, which encodes a transcriptional regulatory protein, which encodes as a DNA transcription regulator to induce G1 phase cell cycle arrest for DNA damage repair [[160]49]. Specifically, D40p53, D133p53α, and p53β isoforms are primarily involved in cellular senescence during its early stages [[161]50]. CHEK1 can induce a delay in the cell cycle process, allowing sufficient time for DNA damage repair [[162]51]. SERPINE1 belongs to the serine protease inhibitor family [[163]52] and has recently been discovered to exhibit pro-angiogenic, proliferative, migratory, and anti-apoptotic activities in tumors [[164]53]. To summarize, neutrophils co-cultured with culture supernatant and SYNPO protein derived from PVT1-knocked down GC cells showed postponed senescence. In conclusion, this study elucidates the dual oncogenic mechanisms of PVT1 in GC progression (Fig. [165]8): (1) Remodel the TME by suppressing neutrophil infiltration/activation, and (2) disrupt genomic stability and induce calcium overload via PVT1 knockdown to establish a metabolic checkpoint. We provide the first evidence that PVT1 orchestrates calcium homeostasis to regulate both neutrophil senescence and functional polarization, with SYNPO emerging as a critical downstream mediator of PVT1-driven calcium signaling in immune-stromal crosstalk. The discovery of this PVT1-centered metabolic-immune axis not only reveals a novel therapeutic target for GC immunotherapy but also offers innovative strategies to overcome TME-mediated therapeutic resistance by simultaneously addressing calcium dysregulation and neutrophil dysfunction. Fig. 8. [166]Fig. 8 [167]Open in a new tab Possible underlying mechanism of PVT1 knockdown in GC cells. This figure was created with Figdraw (ID:ASOTT28c1d) Supplementary Information [168]Supplementary material 1. ^(9.2MB, zip) [169]Supplementary material 2. ^(23.7KB, zip) Acknowledgements