International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
With an aging population and the continuation of organized cancer screening programs, gastric cancer (GC) has shown a consistent trend of increasing morbidity and mortality (1,2). GC has a significant demographic disparity, with male prevalence rates nearly twice that of females (1,3). Geographical variations are also evident, with disproportionately high incidence rates in East Asia (particularly Japan and Mongolia) and Eastern Europe, which together account for 87% of GC cases worldwide (1,3). GC has thus become a global public health challenge and even surgically treated patients, such as those with gastric stump cancer, still face a considerable disease burden and poor outcomes (4). The low detection rate of early-stage GC can be attributed to the lack of specific clinical symptoms (5). GC screening can detect precancerous lesions and early-stage GC in asymptomatic patients, reducing mortality and improving treatment efficacy (6). The majority (>70%) of patients are diagnosed at advanced stages, mainly due to the absence of clinical symptoms during early pathogenesis (7). This delay in diagnosis contributes to a poor prognosis and the clinical management of GC presents obvious challenges. Despite the availability of chemotherapy and radiotherapy, treatment outcomes for advanced gastrointestinal malignancies, such as pancreatic and GC, remain unsatisfactory (8). Patients diagnosed at a late stage have a poor prognosis without targeted treatment (2). Traditional Chinese Medicine (TCM)-derived polysaccharides, such as Rhizoma Coptidis polysaccharides and Anemarrhena asphodeloides polysaccharides, have been explored as adjuncts in GC management (9,10). To address these clinical challenges, there is a pressing need to improve our understanding of the pathogenesis of GC. Therefore, it is necessary to elucidate the mechanisms associated with the pathogenesis and progression of GC and to explore potential targets for effective treatment. Such discoveries are essential for the development of more effective treatment strategies and improved survival outcomes.
Mitochondria are vital for cellular homeostasis concerning energy production, calcium regulation and apoptosis. Their dysfunction has been widely reported to be associated with neurodegenerative diseases, diabetes and chronic pulmonary conditions (11-14). Mitophagy, the selective degradation of defective mitochondria, is essential for maintaining cellular homeostasis and influencing various cancer-related pathways. In various types of cancer, including bladder and colorectal, mitophagy has been linked to survival and therapy resistance. The influence of mitophagy on tumor microenvironments and immune responses has the potential to facilitate tumor growth (15-17). Conversely, the progression of breast cancer is inhibited by Urolithin A, which activates transcription factor EB (TFEB)-mediated mitophagy in tumor macrophages (18). Esophageal cancer was associated with a reduction in mitophagy, which has been linked to an increased potential for metastasis (19). These results indicate that mitophagy inhibition may facilitate tumor spread. Consequently, the manipulation of mitophagy pathways may present a novel therapeutic strategy with the potential to enhance treatment outcomes in cancer management.
The Ras homolog family member T1 (RHOT1) gene encodes a mitochondrial GTPase that plays a crucial role in various processes within mitochondria, including mitochondrial transport, mitochondrial calcium buffering and mitophagy. Its role in disease pathogenesis, particularly neurodegenerative disorders and metabolic conditions, has been well-documented. For instance, the dysfunction of RHOT1 has been demonstrated to disrupt Parkin-mediated mitophagy, leading to the accumulation of damaged mitochondria and exacerbating neuronal loss in Parkinson's disease (20). Additionally, the dysfunction of RHOT1 has been observed to impede calcium handling and mitochondrial transport, thereby contributing to metabolic dysregulation in diabetes (11). RHOT1 plays a pivotal role in maintaining mitochondrial quality control and has been identified as essential for preserving mitochondrial integrity (20). In GC, increasing research has identified mitochondrial dysfunction as a pivotal factor driving tumor progression, therapy resistance and metabolic reprogramming (21-24). Peng et al (25) investigated the correlation between the expression of RHOT1 and the clinicopathological features of tumor-node-metastasis staging and lymph node metastasis. RHOT1 has been thought to serve as a biomarker for GC. However, to date, no studies have reported the specific role of RHOT1 in the regulation of mitophagy in GC. Its conserved function in mitochondrial quality control suggests that aberrant RHOT1 activity may influence tumors by regulating mitophagy. Consequently, the investigation of RHOT1 in GC cells has the potential to reveal novel associations between mitochondrial dynamics and oncogenesis, thus providing potential therapeutic targets for diseases associated with mitochondrial dysfunction.
The present study hypothesized that RHOT1 may affect the behavior of GC cells through mitophagy. It aimed to investigate the role of RHOT1 in regulating mitochondrial quality control, energy metabolism and mitophagy-related signaling in GC. Furthermore, its effect on the proliferation, invasion and migration of GC cells in vitro was examined. These findings may provide a theoretical basis for considering RHOT1 as a potential alternative therapeutic target for GC.
The cell lines used were HGC-27, MKN-45, AGS, SNU-1 and GES-1 (human gastric cancer cell lines and a human gastric epithelial cell line, respectively) obtained from Wuhan Servicebio Technology Co., Ltd. (cell batch: IM-H084202303). They were cultured in Roswell Park Memorial Institute (RPMI) 1640 medium (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% fetal bovine serum (FBS; Clark Bioscience), 100 U/ml penicillin and 100 μg/ml streptomycin and maintained at 37°C in a humidified atmosphere with 5% CO2.
RNA Easy Fast Tissue/Cell Kit (Tiangen Biotech Co., Ltd.) was prepared for total RNA extraction according to the manufacturer's instructions under RNase-free conditions. Total RNA was extracted from GC cell lines (HGC-27, MKN-45, AGS, SNU-1) and GES-1 after transfection or treatment, depending on the experimental group. PrimeScript RT Master Mix Kit (Takara Biotechnology Co., Ltd.) was used for reverse transcription and the SYBR Green Master Mix Kit (Takara Biotechnology Co., Ltd.) was employed to detect the relative expression of mRNAs. The reaction conditions were performed under the following conditions: 95°C for 2 min, 95°C for 15 sec, 60°C for 1 min and 72°C for 30 sec for 40 cycles. Primer sequences are presented in Table I. All primers were designed based on Homo sapiens RefSeq/NCBI sequences and targeted exon-exon junctions to ensure specificity for mRNA. GAPDH was used to normalize the gene being tested. All primers were synthesized by Sangon Biotech Co., Ltd. and validated by melt curve analysis and agarose gel electrophoresis to confirm specificity. Each sample was run in triplicate using the 7500 Fast (Applied Biosystems; Thermo Fisher Scientific, Inc.). Gene expression results were expressed as fold changes relative to GAPDH using the 2-ΔΔCq method (26).
Target-specific siRNA (si-RHOT1) (Table II) and a negative control siRNA (si-NC) were transfected into HGC-27 cells cultured in six-well plates. Cells were transfected with 20 μM siRNA using Lipo 6000 transfection reagent (Beyotime Institute of Biotechnology) and then cultured for 48 h at 37°C. The silencing efficiency was evaluated by qPCR. The si-NC used in the present study was a non-targeting control siRNA purchased from Shanghai GenePharma Co., Ltd. and its sequence was not disclosed by the manufacturer.
Cell counting kit 8 (CCK-8; MedChemExpress) was used for the detection of proliferation of HGC-27 cells. Cells (~2×103) were seeded in 100 μl of medium in a 96-well plate and incubated at 37°C. CCK-8 reagent (10 μl) was added to each well at 0, 24, 48, 72 and 96 h. Incubation was performed at 37°C for 2 h. A microplate reader (Molecular Devices, LLC) was used to read OD values at 450 nm. Blank control wells containing medium and CCK-8 reagent without cells were included and their absorbance values were subtracted from sample readings to eliminate background interference. Cell proliferation results were expressed as OD values at 450 nm, normalized to control wells.
A wound-healing assay was used to evaluate the migration ability of HGC-27 cells. Cells were cultured at 3×105/ml per well in a 12-well plate. After 6 h of incubation, 1 ml of cell medium was added to the 12-well plate. A 20 μl pipette tip was used to create a wound and PBS was used to wash the slide 2-3 times. Then serum-free medium was prepared and the images were captured under the microscope at 0, 24 and 48 h. The experiment was performed three times.
For the Transwell migration assay, 200 μl of cell suspension was transferred to a Transwell upper chamber (Corning, Inc.) and 500 μl of medium with 20% FBS was transferred to a Transwell lower chamber and incubated with 5% CO2 in a 37°C incubator for 48 h. Finally, 50 μl of the diluted cell suspension was added to the upper chamber of the Transwell (Corning, Inc.) and incubated at 37°C for 1 h. For the invasion assay, a layer of Matrigel matrix glue (Corning, Inc.; ratio of serum-free medium:matrix glue 8:1) was coated within the lower chamber, and the cells were incubated for 24 h at 37°C. After incubation, a 4% paraformaldehyde solution was used to fix the cells in the Transwell. The Transwell was subsequently stained with 0.1% crystal violet for 20 min. Cell counts for migration and invasion were recorded by bright-field microscopy. Three biological replicates were performed. The migration and invasion abilities were expressed as the number of stained cells counted under the microscope.
Flow cytometry was performed to evaluate the cell cycle and apoptosis of the cells. HGC-27 cells were stained with propidium iodide (PI; cat. Y267501; Beyotime Institute of Biotechnology) for the cell cycle assay and Annexin Ⅴ and PI (Annexin Ⅴ; cat. C1062M; Beyotime Institute of Biotechnology) were used for the apoptosis assay. Cells were stained with PI for cell cycle and Annexin Ⅴ and PI for apoptosis at 37°C for 30 min. FACSCalibur and FACSCelesta (BD Biosciences) were used for cell cycle and apoptosis detection, respectively. The flow cytometer was operated with a 488 nm excitation laser at standard power. For cell cycle analysis, gating was based on PI fluorescence intensity to distinguish populations with different DNA content, corresponding to G0/G1, S and G2/M phases. For apoptosis analysis, gating was performed on Annexin V/PI two-parameter dot plots to classify cells as viable (Annexin V−/PI−), early apoptotic (Annexin V+/PI-), or late apoptotic/necrotic (Annexin V+/PI+). Three biological replicates were performed. Cell cycle distribution was expressed as the percentage of cells in G0/G1, S and G2/M phases and apoptosis results were expressed as the percentage of apoptotic cells.
ROS were detected using the Reactive Oxygen Specific Test Kit (Beyotime Institute of Biotechnology). ROS levels were measured using 2,7-dichlorodihydrofluorescein diacetate (DCFH-DA). HGC-27 cells (1×106) were divided equally. In the positive control group, 1 μl of Rosup stimulating drug was added and incubated at 37°C for 30 min. The treatment group was directly treated with 10 μM DCFH-DA dissolved in serum-free culture medium (1 ml) and incubated at 37°C for 30 min. Fluorescence was detected by flow cytometry at an excitation wavelength of 488 nm and an emission wavelength of 530 nm and changes in MMP were detected using the Mitochondrial Membrane Potential Detection Kit (Beyotime Institute of Biotechnology). The HGC-27 cells were harvested by centrifugation at 1,000 × g for 5 min at 37°C and resuspended in 50 mM phosphate-buffered saline (pH 7.0). A total of 5×104 cells collected by centrifugation were resuspended in 188 μl Annexin V-FITC conjugate. Then 2 μl Mito-Tracker Red CMXRos was added and incubated at 25°C for 30 min and the cells placed in an ice bath. Fluorescence was detected by flow cytometry using an excitation wavelength of 579 nm and an emission wavelength of 599 nm. ROS levels were expressed as mean fluorescence intensity and MMP was expressed as fluorescence intensity. The experiment was performed three times.
The String website (https://cn.string-db.org/) was used to retrieve target genes for analysis of protein-protein interactions (PPI). The species was limited to Homo sapiens and the parameters were set as follows: Choice of significance term for network edges; evidence: Choice of active interaction source; experimental: Choice of minimum required interaction score; confidence: 0.2.
After downloading the enriched gene set, de-duplication of the gene set was carried out to obtain the set of interacting genes. The final PPI network map was calculated by selecting the top 10 pivotal genes in the Hubba node using the Cytoscape (https://cytoscape.org/; version 3.8.0) plugin cyto-Hubba. R software (https://www.r-project.org/; version 3.6.3) was used to analyze the RHOT1 mRNA expression levels of STAD RNA-seq based on The Cancer Genome Atlas (TCGA) database and a single-gene co-expression heatmap was constructed. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses of selected data were performed using the clusterProfiler package (https://www.bioconductor.org/packages/release/bioc/html/clusterProfiler.html; version 3.14.3) and visualized using the ggplot2 package (https://cran.r-project.org/web/packages/ggplot2/index.html; version 3.3.3). The intersection of mitophagy genes obtained from the Reactome database (https://curator.reactome.org; version 87) with those obtained from the KEGG database was taken and plotted as a Venn diagram. The expression matrix of 20 mitophagy pathway core genes was screened based on the intersection of mitophagy pathways from KEGG and Reactome. Principal component analysis (PCA) was performed (after normalization) using Omicstudio (https://www.omicstudio.cn/tool) and the correlation heatmap of these core genes was plotted.
Total proteins from transfected HGC-27 cells were isolated by RIPA buffer (cat. no. R0010; Beijing Solarbio Science & Technology Co., Ltd.) and then separated by 8-11% SDS-PAGE. Protein concentration was determined using the BCA protein assay kit (cat. no. WLA019; Wanleibio Co., Ltd.). The 20 μg protein samples were dissolved and transferred to 0.45 μm polyvinylidene fluoride (PVDF) membranes (cat. no. IPVH00010; MilliporeSigma). The PVDF membranes were blocked with 5% skimmed milk powder for 2 h at room temperature and then incubated with primary antibodies at 4°C overnight. The antibodies were RHOT1 (cat. no. A5838; 1:500; ABclonal Biotech Co., Ltd.), PINK1 (cat. no. WL04963; 1:500; Wanleibio Co., Ltd.), Parkin (cat. no. WL02512; 1:500; Wanleibio Co., Ltd.), GAPDH (cat. no. 5174, 1:1,000; Cell Signaling Technology, Inc.). The PVDF membranes were then washed with tris-buffered saline-Tween 20 (0.1% TBST-20; cat. GC204002; Wuhan Servicebio Technology Co., Ltd.) and incubated with horseradish peroxidase-conjugated goat anti-rabbit IgG (cat. no. WLA023; 1:5,000; Wanleibio Co., Ltd.) for 1.5 h at room temperature. The membranes were scanned and analyzed using the Gel-Pro Analyzer 4.5 program for Windows (Media Cybernetics, Inc.). Three biological replicates were performed. Protein expression levels were expressed as band intensities normalized to GAPDH.
The data were presented as the mean ± standard deviation. The findings from each experiment were corroborated through independent replicates. Normality of data distribution was assessed using the Shapiro-Wilk test prior to parametric analyses. For comparisons between two groups, a two-tailed unpaired Student's t-test was applied. For comparisons among multiple groups with a single variable, one-way ANOVA followed by the Tukey-Kramer post hoc test was used. For datasets involving more than one variable, two-way ANOVA with appropriate post-hoc analyses was performed. SPSS (version 29.0; IBM Corp.) and GraphPad Prism (version 9.5.0; Dotmatics) were used for statistical analyses. P<0.05 was considered to indicate a statistically significant difference.
To investigate the function of RHOT1, the interacting proteins of RHOT1 were investigated from the STRING search sets. Analysis of RHOT1-interacting proteins identified RHOT2, LRRK2, GRID2IP, PLEKHA4 and CIT as hub genes (Fig. 1A). Single-gene co-expression analysis using TCGA-STAD RNA-seq data showed that RHOT2, LRRK2, GRID2IP and CIT were significantly co-expressed with RHOT1 (P<0.001; Fig. 1B). GO and KEGG enrichment analysis indicated involvement in mitochondrion organization, mitophagy, microtubule binding and pathways including Parkinson's disease and neurodegeneration (Fig. 1C and D).
A Venn diagram identified 20 mitophagy-related genes from Reactome and KEGG databases (Fig. 1E) and PCA showed significant differences between tumor and normal groups (Fig. 1F). Next, a correlation heatmap was constructed from the 20 mitophagy genes (Fig. 1G). The correlation heatmap displayed the genes with a statistically significant difference P<0.01 (Fig. 1H). Intersection of RHOT1-interacting genes and mitophagy genes revealed PINK1 and Parkin (Fig. 1I). RHOT1 expression was upregulated in GC cell lines HGC-27, MKN-45, AGS and SNU-1 compared with GES-1, with HGC-27 showing the highest expression (Fig. 2A). Among four siRHOT1 constructs, siRNA RHOT1-homo-393 achieved the highest silencing efficiency (Fig. 2B). Subsequently, the mRNA expression levels of the genes were examined. The result showed that the mRNA expression of PINK1 and Parkin were both downregulated after silencing RHOT1, with PINK1 decreased by 59.75% (P=0.025) and Parkin decreased by 65.12% compared with the si-NC group (P=0.0189) (Fig. 2C and D). Additionally, the mRNA expression of mitochondrial membrane-related genes TOMM20 (translocase of the outer mitochondrial membrane 20) and TIMM23 (translocase of the inner mitochondrial membrane 23) were increased after silencing RHOT1 (Fig. 2E and F). The protein expression of PINK1 and Parkin were downregulated after silencing RHOT1 (Fig. 2G).
These results indicated that silencing RHOT1 leads to decreased PINK1 and Parkin expression and inhibition of mitophagy.
Based on the aforementioned results for PINK1, Parkin and mitochondrial membrane-related genes, mitochondrial function was further examined to assess mitophagy. Flow cytometry analysis showed that the average ROS fluorescence intensity in HGC-27 cells was increased by 84.73% (P<0.001) after silencing RHOT1 compared with the negative control, while it remained lower than the positive control (Fig. 3A). MMP assessment indicated that silencing RHOT1 caused a 36.94% decrease in MMP compared with the negative control (P=0.0061), reflecting mitochondrial depolarization (Fig. 3B).
These results demonstrate that silencing RHOT1 elevates ROS levels and depolarizes the mitochondrial membrane, indicating mitochondrial damage and dysfunction in HGC-27 cells.
Mitochondria were damaged and dysfunctional, which would cause abnormal cellular energy metabolism and affect cell growth. The effect of silencing RHOT1 on the growth state and apoptosis of GC cells was assessed in HGC-27 cells. CCK-8 assay showed that silencing RHOT1 markedly slowed HGC-27 cell proliferation from 48 h to 96 h compared with the si-NC group (Fig. 4A). Flow cytometry analysis revealed that silencing RHOT1 increased the proportion of cells in G0/G1 phase, causing cell cycle arrest. Specifically, the G0/G1 phase proportion was markedly higher than the si-NC group (P<0.05) (Fig. 4B). Apoptosis assessment demonstrated that silencing RHOT1 elevated the percentage of apoptotic cells compared with the si-NC group (P<0.05) (Fig. 4C).
These results indicated that silencing RHOT1 affected mitophagy to reduce the proliferation of GC cells, arrest the cell cycle in the G0/G1 phase and promote cell apoptosis.
The present results showed that the expression of PINK1 and Parkin was downregulated after silencing RHOT1, so RHOT1 may affect mitophagy, leading to the accumulation of damaged mitochondria, which further increased ROS and depleted energy in the cells and consequently affected the invasion and migration phenotypes of the cells. The effects of RHOT1 on the invasion and migration of GC cells were subsequently investigated. Wound healing and Transwell migration assays were employed to evaluate the migration ability of HGC-27 cells. Wound healing assay showed that silencing RHOT1 markedly reduced the migration of HGC-27 cells compared with the si-NC group (Fig. 5A); Transwell migration assay confirmed that the number of migrating cells in the si-RHOT1 group was markedly lower than in the si-NC group (P<0.05) (Fig. 5B). Transwell invasion assay further demonstrated that silencing RHOT1 markedly reduced the number of invasive HGC-27 cells through Matrigel compared with the control group (P<0.05; Fig. 5C).
These results indicated that silencing RHOT1 affected mitophagy to reduce the migration and invasion of GC cells.
GC is the fifth most common cause of cancer-related death worldwide, contributing to 8.8% of all such deaths (27). Screening programs have reduced mortality in some cancers by detecting early-stage and precancerous lesions in asymptomatic patients, but organized GC screening has only been implemented in a few high-prevalence countries (1). The etiology of GC is complex and multifactorial, with numerous molecular alterations influencing tumorigenesis and progression through aberrant gene expression or protein dysfunction (28). At present, there remains a necessity to explore treatment targets and effective biomarkers.
Mitochondrial health is critical for cellular function and mitophagy, the selective degradation of damaged mitochondria, is central to maintaining mitochondrial quality (29,30). Mitophagy exerts dual roles in cancer, either prompting or suppressing carcinogenesis (31). RHOT1, an outer mitochondrial membrane protein, serves as a molecular switch in the mitochondrial dynamics and mitophagy (11,20,32). Mutations in RHOT1 disrupt mitochondria transport, impair quality control and have been linked to neurodegenerative diseases such as Parkinson's (11,33-35). RHOT1 is upregulated in several cancers: Jiang et al (36) predicted its prognostic value in pancreatic cancer and Li et al (37) found that silencing RHOT1 inhibited pancreatic cancer cell proliferation and migration. In mice, RHOT1 knockdown reduced mitochondrial motility and impaired quality control (38). It was also found that RHOT1 interacted with LRRK2, consistent with findings that LRRK2 knockdown or mutation impaired basal mitophagy (39). Collectively, these data position RHOT1 as a critical regulator of mitochondrial dynamics and mitophagy. Database analyses revealed strong associations between RHOT1 and PINK1/Parkin, the canonical ubiquitin-dependent mitophagy pathway (40). In the present study, silencing RHOT1 altered PINK1 and Parkin expression, suggesting that RHOT1 modulates mitophagy via this axis. GO and KEGG enrichment of RHOT1-interacting genes further supported predominant effects on mitochondrial function and the involvement of the PINK1/Parkin pathway.
Silencing RHOT1 also induced mitochondrial dysfunction, characterized by increased ROS production and MMP depolarization. ROS are central regulators of proliferation, differentiation and metabolism, but excessive ROS promotes oxidative stress, mtDNA mutations and mitochondrial damage (41,42). RHOT1 has been implicated in peroxisomal transport, which contributes to ROS clearance (43,44). Thus, the present findings of ROS accumulation and MMP depolarization aligned with reports that RHOT1 deficiency aggravates oxidative stress and compromises mitochondrial homeostasis. Consequently, dysfunctional mitochondria accumulate, reinforcing oxidative stress and energy imbalance.
In addition, silencing RHOT1 upregulated TOMM20 and TIMM23, proteins essential for mitochondrial protein import. This upregulation probably reflects a compensatory response to mitochondrial stress (45-47), consistent with previous studies showing increased import machinery and UPRmt components to preserve organelle integrity (48-50). Thus, RHOT1 deficiency appears to couple impaired mitophagy with a UPRmt-like compensatory mechanism, which will be a subgoal for future research.
Mitochondrial dysfunction also influenced cell cycle progression and apoptosis. Oxidative stress stabilizes p53, upregulates p21, inhibits CDK2/Cyclin E and blocks G1-S transition (48,51-53). Disruption of MMP triggers cytochrome c release, caspase activation and PARP cleavage, promoting apoptosis (54-56). In agreement, silencing RHOT1 in GC cells induced G0/G1 arrest and apoptosis, linking mitochondrial quality control to cell-cycle regulation and survival. Furthermore, RHOT1 loss impaired migration and invasion. Reduced ATP production and ROS accumulation inhibit cytoskeletal remodeling and pseudopodia formation (57,58), while ROS modulates EMT gene expression via NF-κB and HIF-1α signaling (59). Myc has been shown to transcriptionally regulate RHOT1, coordinating mitochondrial trafficking with cytoskeletal dynamics (60). Dysregulated mitophagy and accumulation of damaged mitochondria further promote tumor progression (61).
In summary, RHOT1 deficiency in GC cells perturbs mitophagy, exacerbates oxidative stress, disrupts mitochondrial homeostasis and impairs key cellular processes, including cell cycle progression, apoptosis and invasion. These findings highlighted RHOT1 as a potential therapeutic target in GC. A potential limitation of the present study is that functional assays were mainly conducted in the HGC-27 cell line, which showed the highest RHOT1 expression among the GC cell lines examined. While this strengthens the rationale for its selection as the primary model, further validation in additional GC subtypes would help to generalize the findings. Addressing this limitation will be an important focus of future research. In addition, the results remained correlative, as no additional experiments were designed to directly validate the causal relationship. This limitation was acknowledged and the findings were therefore interpreted with caution. Future studies will be required to provide more definitive evidence of causality.
Silencing RHOT1 induced mitochondrial dynamics disorder, resulting in the inhibition of the PINK1/Parkin mitophagy pathway, which impaired mitophagy and caused the accumulation of intracellular ROS and depolarization of MMP. The aforementioned multiple ways triggered cell cycle arrest and promoted apoptosis, resulting in decreased proliferation of GC cells and weakened invasion and migration ability. Therefore, suppression of RHOT1 may become a new strategy for the treatment of GC and provide a new direction for the study of targeted treatment and drug resistance mechanisms in GC.
The data generated in the present study may be requested from the corresponding author.
YQP and XC conceived and designed the research; YQP, XC, FK, JZ, SY were responsible for data analysis. RF, JW and YYP were provided supervision and project administration. XC provided supervision. YQP wrote the first draft and YYP reviewed and edited the manuscript. FK, JZ and YYP confirm the authenticity of all the raw data. All authors read and approved the final manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
Not applicable.
The present study was supported by the Doctoral Start-up Foundation of Liaoning Province, China (grant no. 2022-BS-340); the Science and Technology Research Project of Department of Education of Liaoning Province (grant no. JYTZD2023145), the Science and Technology Key Research Project of Liaoning Province (grant no. 2024JH2/102500059) and the Research Project on Integrated Traditional Chinese and Western Medicine for Chronic Disease Management (grant no. CXZH2024134).
|
Conti CB, Agnesi S, Scaravaglio M, Masseria P, Dinelli ME, Oldani M and Uggeri F: Early gastric cancer: Update on prevention, diagnosis and treatment. Int J Environ Res Public Health. 20:21492023. View Article : Google Scholar : PubMed/NCBI | |
|
Rocken C: Predictive biomarkers in gastric cancer. J Cancer Res Clin Oncol. 149:467–481. 2023. View Article : Google Scholar : | |
|
Smyth EC, Nilsson M, Grabsch HI, Van Grieken NC and Lordick F: Gastric cancer. Lancet. 396:635–648. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Drizlionoks E, Tercioti V Junior, Coelho Neto JS, Andreollo NA and Lopes LR: Surgical treatment of gastric stump cancer: A cohort study of 51 patients. Arq Bras Cir Dig. 37:e18502025. View Article : Google Scholar : PubMed/NCBI | |
|
Yang WJ, Zhao HP, Yu Y, Wang JH, Guo L, Liu JY, Pu J and Lv J: Updates on global epidemiology, risk and prognostic factors of gastric cancer. World J Gastroenterol. 29:2452–2468. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Qiu H, Cao S and Xu R: Cancer incidence, mortality, and burden in China: A time-trend analysis and comparison with the United States and United Kingdom based on the global epidemiological data released in 2020. Cancer Commun (Lond). 41:1037–1048. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Veitch AM, Uedo N, Yao K and East JE: Optimizing early upper gastrointestinal cancer detection at endoscopy. Nat Rev Gastroenterol Hepatol. 12:660–667. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Haggstrom L, Chan WY, Nagrial A, Chantrill LA, Sim HW, Yip D and Chin V: Chemotherapy and radiotherapy for advanced pancreatic cancer. Cochrane Database Syst Rev. 12:CD0110442024.PubMed/NCBI | |
|
Gao S, Wang Y, Shan Y, Wang W, Li J and Tan H: Rhizoma Coptidis polysaccharides: Extraction, separation, purification, structural characteristics and bioactivities. Int J Biol Macromol. 320(Pt 1): 1456772025. View Article : Google Scholar : PubMed/NCBI | |
|
Gao S, Xu T, Wang W, Li J, Shan Y, Wang Y and Tan H: Polysaccharides from Anemarrhena asphodeloides Bge, the extraction, purification, structure characterization, biological activities and application of a traditional herbal medicine. Int J Biol Macromol. 311(Pt 1): 1434972025. View Article : Google Scholar : PubMed/NCBI | |
|
Kavyashree S, Harithpriya K and Ramkumar KM: Miro1-a key player in β-cell function and mitochondrial dynamics under diabetes mellitus. Mitochondrion. 84:1020392025. View Article : Google Scholar | |
|
Li JM, Li X, Chan LWC, Hu R, Zheng T, Li H and Yang S: Lipotoxicity-polarised macrophage-derived exosomes regulate mitochondrial fitness through Miro1-mediated mitophagy inhibition and contribute to type 2 diabetes development in mice. Diabetologia. 66:2368–2386. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Jeong YY, Jia N, Ganesan D and Cai Q: Broad activation of the PRKN pathway triggers synaptic failure by disrupting synaptic mitochondrial supply in early tauopathy. Autophagy. 18:1472–1474. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Maremanda KP, Sundar IK and Rahman I: Protective role of mesenchymal stem cells and mesenchymal stem cell-derived exosomes in cigarette smoke-induced mitochondrial dysfunction in mice. Toxicol Appl Pharmacol. 385:1147882019. View Article : Google Scholar : PubMed/NCBI | |
|
Li D, Su H, Deng X, Huang Y, Wang Z, Zhang J, Chen C, Zheng Z, Wang Q, Zhao S, et al: DARS2 promotes bladder cancer progression by enhancing PINK1-mediated mitophagy. Int J Biol Sci. 21:1530–1544. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Deng X, Huang Y, Zhang J, Chen Y, Jiang F, Zhang Z, Li T, Hou L, Tan W and Li F: Histone lactylation regulates PRKN-Mediated mitophagy to promote M2 Macrophage polarization in bladder cancer. Int Immunopharmacol. 148:1141192025. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Z, Yu C, Xie G, Tao K, Yin Z and Lv Q: USP14 inhibits mitophagy and promotes tumorigenesis and chemosensitivity through deubiquitinating BAG4 in microsatellite instability-high colorectal cancer. Mol Med. 31:1632025. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng B, Wang Y, Zhou B, Qian F, Liu D, Ye D, Zhou X and Fang L: Urolithin A inhibits breast cancer progression via activating TFEB-mediated mitophagy in tumor macrophages. J Adv Res. 69:125–138. 2025. View Article : Google Scholar : | |
|
Zheng S, Zhang Y, Gong X, Teng Z and Chen J: CREB1 regulates RECQL4 to inhibit mitophagy and promote esophageal cancer metastasis. J Clin Biochem Nutr. 75:102–110. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Chemla A, Arena G, Sacripanti G, Barmpa K, Zagare A, Garcia P, Gorgogietas V, Antony P, Ohnmacht J, Baron A, et al: Parkinson's disease mutant Miro1 causes mitochondrial dysfunction and dopaminergic neuron loss. Brain. Feb 6–2025.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI | |
|
Cavinato M: Mitochondrial dysfunction and cisplatin sensitivity in gastric cancer: GDF15 as a master player. FEBS J. 291:1111–1114. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Lo YL, Wang TY, Chen CJ, Chang YH and Lin AM: Two-in-one nanoparticle formulation to deliver a tyrosine kinase inhibitor and microRNA for targeting metabolic reprogramming and mitochondrial dysfunction in gastric cancer. Pharmaceutics. 14:17592022. View Article : Google Scholar : PubMed/NCBI | |
|
Chen W, Zou P, Zhao Z, Weng Q, Chen X, Ying S, Ye Q, Wang Z, Ji J and Liang G: Selective killing of gastric cancer cells by a small molecule via targeting TrxR1 and ROS-mediated ER stress activation. Oncotarget. 7:16593–16609. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Rong Y, Teng Y and Zhou X: Advances in the study of metabolic reprogramming in gastric cancer. Cancer Med. 14:e709482025. View Article : Google Scholar : PubMed/NCBI | |
|
Peng YY, Sun D and Xin Y: Hsa_circ_0005230 is up-regulated and promotes gastric cancer cell invasion and migration via regulating the miR-1299/RHOT1 axis. Bioengineered. 13:5046–5063. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar | |
|
Li M, Pi H, Yang Z, Reiter RJ, Xu S, Chen X, Chen C, Zhang L, Yang M, Li Y, et al: Melatonin antagonizes cadmium-induced neurotoxicity by activating the transcription factor EB-dependent autophagy-lysosome machinery in mouse neuroblastoma cells. J Pineal Res. 61:353–369. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Procaccio L, Schirripa M, Fassan M, Vecchione L, Bergamo F, Prete AA, Intini R, Manai C, Dadduzio V, Boscolo A, et al: Immunotherapy in gastrointestinal cancers. Biomed Res Int. 2017:43465762017. View Article : Google Scholar : PubMed/NCBI | |
|
Leites EP and Morais VA: Mitochondrial quality control pathways: PINK1 acts as a gatekeeper. Biochem Biophys Res Commun. 500:45–50. 2018. View Article : Google Scholar | |
|
Yao RQ, Ren C, Xia ZF and Yao YM: Organelle-specific autophagy in inflammatory diseases: A potential therapeutic target underlying the quality control of multiple organelles. Autophagy. 17:385–401. 2021. View Article : Google Scholar : | |
|
Wang Z, Chen C, Ai J, Shu J, Ding Y, Wang W, Gao Y, Jia Y and Qin Y: Identifying mitophagy-related genes as prognostic biomarkers and therapeutic targets of gastric carcinoma by integrated analysis of single-cell and bulk-RNA sequencing data. Comput Biol Med. 163:1072272023. View Article : Google Scholar : PubMed/NCBI | |
|
Eberhardt EL, Ludlam AV, Tan Z and Cianfrocco MA: Miro: A molecular switch at the center of mitochondrial regulation. Protein Sci. 29:1269–1284. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Lopez-Domenech G, Howden JH, Covill-Cooke C, Morfill C, Patel JV, Burli R, Crowther D, Birsa N, Brandon NJ and Kittler JT: Loss of neuronal Miro1 disrupts mitophagy and induces hyperactivation of the integrated stress response. EMBO J. 40:e1007152021. View Article : Google Scholar : PubMed/NCBI | |
|
Diaz-Moreno I and De La Rosa MA: IUBMB focused meeting/FEBS workshop: Crosstalk between nucleus and mitochondria in human disease (CrossMitoNus). IUBMB Life. 73:489–491. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Grossmann D, Berenguer-Escuder C, Chemla A, Arena G and Kruger R: The Emerging role of RHOT1/Miro1 in the pathogenesis of parkinson's disease. Front Neurol. 11:5872020. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang H, He C, Geng S, Sheng H, Shen X, Zhang X, Li H, Zhu S, Chen X, Yang C and Gao H: RhoT1 and Smad4 are correlated with lymph node metastasis and overall survival in pancreatic cancer. PLoS One. 7:e422342012. View Article : Google Scholar : PubMed/NCBI | |
|
Li Q, Yao L, Wei Y, Geng S, He C and Jiang H: Role of RHOT1 on migration and proliferation of pancreatic cancer. Am J Cancer Res. 5:1460–1470. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Lopez-Domenech G, Higgs NF, Vaccaro V, Ros H, Arancibia-Carcamo IL, Macaskill AF and Kittler JT: Loss of dendritic complexity precedes neurodegeneration in a mouse model with disrupted mitochondrial distribution in mature dendrites. Cell Rep. 17:317–327. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Singh F, Prescott AR, Rosewell P, Ball G, Reith AD and Ganley IG: Pharmacological rescue of impaired mitophagy in Parkinson's disease-related LRRK2 G2019S knock-in mice. Elife. 10:e676042021. View Article : Google Scholar : PubMed/NCBI | |
|
Cao Y, Zheng J, Wan H, Sun Y, Fu S, Liu S, He B, Cai G, Cao Y, Huang H, et al: A mitochondrial SCF-FBXL4 ubiquitin E3 ligase complex degrades BNIP3 and NIX to restrain mitophagy and prevent mitochondrial disease. EMBO J. 42:e1130332023. View Article : Google Scholar : PubMed/NCBI | |
|
Lu Y, Li Z, Zhang S, Zhang T, Liu Y and Zhang L: Cellular mitophagy: Mechanism, roles in diseases and small molecule pharmacological regulation. Theranostics. 13:736–766. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Lemasters JJ: Selective mitochondrial autophagy, or mitophagy, as a targeted defense against oxidative stress, mitochondrial dysfunction, and aging. Rejuvenation Res. 8:3–5. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Covill-Cooke C, Toncheva VS, Drew J, Birsa N, Lopez-Domenech G and Kittler JT: Peroxisomal fission is modulated by the mitochondrial Rho-GTPases, Miro1 and Miro2. EMBO Rep. 21:e498652020. View Article : Google Scholar : PubMed/NCBI | |
|
Costello JL, Castro IG, Camoes F, Schrader TA, Mcneall D, Yang J, Giannopoulou EA, Gomes S, Pogenberg V, Bonekamp NA, et al: Predicting the targeting of tail-anchored proteins to subcellular compartments in mammalian cells. J Cell Sci. 130:1675–1687. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Raiff A, Zhao S, Bekturova A, Zenge C, Mazor S, Chen X, Ru W, Makaros Y, Ast T, Ordureau A, et al: TOM20-driven E3 ligase recruitment regulates mitochondrial dynamics through PLD6. Nat Chem Biol. Apr 22–2025.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI | |
|
Saleem A, Iqbal S, Zhang Y and Hood DA: Effect of p53 on mitochondrial morphology, import, and assembly in skeletal muscle. Am J Physiol Cell Physiol. 308:C319–C329. 2015. View Article : Google Scholar : | |
|
Zha J, Li J, Yin H, Shen M and Xia Y: TIMM23 overexpression drives NSCLC cell growth and survival by enhancing mitochondrial function. Cell Death Dis. 16:1742025. View Article : Google Scholar : PubMed/NCBI | |
|
Kumar N, Shukla A, Kumar S, Ulasov I, Singh RK, Kumar S, Patel A, Yadav L, Tiwari R, Paswan R, et al: FNC (4'-azido-2'-deoxy-2'-fluoro(arbino)cytidine) as an effective therapeutic agent for NHL: ROS generation, cell cycle arrest, and mitochondrial-mediated apoptosis. Cell Biochem Biophys. 82:623–639. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Tian X, Yuan M, Li L, Chen D, Liu B, Zou X, He M and Wu Z: Enterovirus 71 induces mitophagy via PINK1/Parkin signaling pathway to promote viral replication. FASEB J. 39:e706592025. View Article : Google Scholar : PubMed/NCBI | |
|
Lu J, Zhang Y, Song H, Wang F, Wang L, Xiong L and Shen X: A novel polysaccharide from tremella fuciformis alleviated high-fat diet-induced obesity by promoting AMPK/PINK1/PRKN-Mediated mitophagy in mice. Mol Nutr Food Res. 69:e2024006992025. View Article : Google Scholar : PubMed/NCBI | |
|
Bai LY, Su JH, Chiu CF, Lin WY, Hu JL, Feng CH, Shu CW and Weng JR: Antitumor effects of a sesquiterpene derivative from marine sponge in human breast cancer cells. Mar Drugs. 19:2442021. View Article : Google Scholar : PubMed/NCBI | |
|
Bhavya K, Mantipally M, Roy S, Arora L, Badavath VN, Gangireddy M, Dasgupta S, Gundla R and Pal D: Novel imidazo[1,2-a]pyridine derivatives induce apoptosis and cell cycle arrest in non-small cell lung cancer by activating NADPH oxidase mediated oxidative stress. Life Sci. 294:1203342022. View Article : Google Scholar : PubMed/NCBI | |
|
Minella AC, Swanger J, Bryant E, Welcker M, Hwang H and Clurman BE: p53 and p21 form an inducible barrier that protects cells against cyclin E-cdk2 deregulation. Curr Biol. 12:1817–1827. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Yu C, Sun J, Lai X, Tan Z, Wang Y, Du H, Pan Z, Chen T, Yang Z, Ye S, et al: Gefitinib induces apoptosis in Caco-2 cells via ER stress-mediated mitochondrial pathways and the IRE1alpha/JNK/p38 MAPK signaling axis. Med Oncol. 42:712025. View Article : Google Scholar | |
|
Guefack MF, Talukdar D, Mukherjee R, Guha S, Mitra D, Saha D, Das G, Damen F, Kuete V and Murmu N: Hypericum roeperianum bark extract suppresses breast cancer proliferation via induction of apoptosis, downregulation of PI3K/Akt/mTOR signaling cascade and reversal of EMT. J Ethnopharmacol. 319(Pt 1): 1170932024. View Article : Google Scholar | |
|
Huang J, Zhang Y, Cheng A, Wang M, Liu M, Zhu D, Chen S, Zhao X, Yang Q, Wu Y, et al: Duck Circovirus genotype 2 ORF3 protein induces apoptosis through the mitochondrial pathway. Poult Sci. 102:1025332023. View Article : Google Scholar : PubMed/NCBI | |
|
Peng J, Yang Z, Li H, Hao B, Cui D, Shang R, Lv Y, Liu Y, Pu W, Zhang H, et al: Quercetin reprograms immunometabolism of macrophages via the SIRT1/PGC-1alpha signaling pathway to ameliorate lipopolysaccharide-induced oxidative damage. Int J Mol Sci. 24:55422023. View Article : Google Scholar | |
|
Chen JT, Wei L, Chen TL, Huang CJ and Chen RM: Regulation of cytochrome P450 gene expression by ketamine: A review. Expert Opin Drug Metab Toxicol. 14:709–720. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Qin W, Li C, Zheng W, Guo Q, Zhang Y, Kang M, Zhang B, Yang B, Li B, Yang H and Wu Y: Inhibition of autophagy promotes metastasis and glycolysis by inducing ROS in gastric cancer cells. Oncotarget. 6:39839–39854. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Agarwal E, Altman BJ, Ho Seo J, Bertolini I, Ghosh JC, Kaur A, Kossenkov AV, Languino LR, Gabrilovich DI, Speicher DW, et al: Myc regulation of a mitochondrial trafficking network mediates tumor cell invasion and metastasis. Mol Cell Biol. 39:e00109–19. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Panigrahi DP, Praharaj PP, Bhol CS, Mahapatra KK, Patra S, Behera BP, Mishra SR and Bhutia SK: The emerging, multifaceted role of mitophagy in cancer and cancer therapeutics. Semin Cancer Biol. 66:45–58. 2020. View Article : Google Scholar |