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.
Cancer stem cells (CSCs) play a pivotal role in tumorigenesis and have been extensively investigated as a prospective target for anticancer therapy (1,2). An association exists between the presence of CSCs and tendencies towards recurrence, treatment resistance and immunological tolerance in bladder cancer (BC); nevertheless, the mechanisms by which bladder CSCs (BCSCs) sustain their stemness are not fully elucidated (3). Pyrroline-5-carboxylate reductase 1 (PYCR1) has been identified as a carcinogenic factor of BC and has been shown to promote BC proliferation, metastasis and epithelial-mesenchymal transition (EMT) (4-6). PYCR1 can promote proliferation, metastasis and EMT of BC, and its role is similar to that of tumor stemness; therefore (7), it is plausible to hypothesize the potential role of PYCR1 in the regulation of BCSC stemness.
Histone methylation, a well-known epigenetic modification mediated by histone demethylases, methyltransferases and methylation reader proteins, is instrumental in cancer pathogenesis (8). SET and MYND domain-containing protein 2 (SMYD2) is a protein methyltransferase, including a catalytic SET domain, known to facilitate monomethylation of lysine residues on both histone and non-histone proteins (8,9). Expression of SMYD2 is significantly enhanced in breast cancer tissues, and patients with BC with elevated SMYD2 expression show a poor prognosis (10). Elevated levels of SMYD2 may promote the proliferation of papillary thyroid carcinoma cells (11), as well as regulate gene expression by modulating histone H3 lysine 4 trimethylation (H3K4me3) modification in the promoter region of its target gene, thereby affecting gastric cancer cell glycolysis and proliferation (12). It was proposed that SMYD2 may stimulate the transcriptional expression of PYCR1 by regulating the levels of H3K4me3 in the promoter region of PYCR1 to regulate BC stemness.
Mitophagy facilitates maintenance of stemness in CSCs (13,14). Mitophagy enhances the plasticity of CSCs via metabolic recombination, thereby improving their adaptation to the tumor microenvironment (15). The putative kinase 1 (PINK1)/Parkin signaling pathway is one of the key mechanisms regulating mitophagy (16,17). When the PINK1/Parkin mitophagy pathway is stimulated, SRY-box transcription factor 2 (Sox2) promotes Nanog expression, potentiates CSC self-renewal and preserves CSC stemness (18). PYCR1/2 serves as a crucial enzyme in the proline biosynthetic pathway, and proline is capable of inducing mitophagy via activation of AMPKα and the upregulation of the Parkin expression (19). Our previous study revealed that overexpression of PYCR1 has the potential to boost the protein expression of PINK1 and Parkin, leading to mitophagy in BC (4). Also, PYCR1-augmented proline synthesis serves a crucial role in sustaining the stemness of CSCs (20). Therefore, PYCR1 might regulate BCSC stemness by controlling mitophagy via the PINK1/Parkin pathway. It was hypothesized that SMYD2 may increase PYCR1 expression by upregulating the H3K4me3 levels in the PYCR1 promoter region and promote BCSC stemness through PINK1/Parkin-regulated mitophagy.
SMYD2 expression in BC and normal bladder tissue and the association between SMYD2 and PYCR1 in BC were analyzed using the GEPIA database (gepia.cancer-pku.cn/).
Human BC cell lines T24 (SCSP-536) and EJ (YS1803C) were procured from Institute of Chinese Academy of Sciences and Yaji Biotechnology Co., Ltd. (both Shanghai, China), respectively. T24 and EJ cells were cultured in DMEM/Ham's F-12 with 10% fetal bovine serum (both Gibco; Thermo Fisher Scientific, Inc.) and 1% penicillin-streptomycin solution (Shanghai Zeye Biotechnology Co., Ltd.) at 37°C with 5% CO2. Cells from the P3 generation with 80% confluence were collected for subsequent experiments.
Human BC cell lines were grouped as follows: Blank (untreated); small interfering (si)-PYCR1 (cells were transfected with PYCR1 siRNA for 6 h and cultured for 18 h].); si-SMYD2 (BC cells were transfected with SMYD2 siRNA for 24 h); si-negative control (NC) (transfected with scramble siRNA for 24 h); overexpression (oe)-PYCR1 (pcDNA3.1-PYCR1-transfected for 24 h); oe-SMYD2 (transfected with pcDNA3.1-SMYD2 for 24 h); oe-NC (transfected with pcDNA3.1-NC for 24 h); oe-SMYD2 + si-PYCR1 (transfection of pcDNA3.1-SMYD2 while being treated with PYCR1 siRNA for 24 h); oe-SMYD2 + si-NC (treated with scramble siRNA for 24 h while being transfected with pcDNA3.1-SMYD2); oe-PYCR1 + si-PINK1 (24-h simultaneous treatment with pcDNA3.1-PYCR1 and PINK1 siRNA) and oe-PYCR1 + si-NC group (co-treatment with pcDNA3.1-PYCR1 and scramble siRNA for 24 h). si-PYCR1 forward 5′-UGC UAU CAA CGC UGU GG-3′ and reverse 5′-CCA CAG CGU UGA UAG CA-3′; si-SMYD2: forward: 5′-CAC CAG UUC UAC UCC AAG UTT-3′, reverse: 5′-ACU UGG AGU AGA ACU GGU GTT-3′; si-NC: forward 5′-UUC UCC GAA CGU GUC ACG UTT-3′ and reverse 5′-ACG UGA CAC GUU CGG AGA ATT-3′.
Cell transfection was performed using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) at 37°C for 24 h with 10 nmol/l pcDNA3.1-PYCR1, pcDNA3.1-SMYD2, pcDNA3.1-NC, PYCR1 siRNA, SMYD2 siRNA, PINK1 siRNA, and scramble siRNA, with a final concentration of 3.75 µl/ml, as previously described (21).
Cells were prepared into a single-cell suspension for the screening of BCSCs. After being washed twice with PBS, cells were resuspended in PBS to adjust the cell concentration to 1×109/l. Cells were cultured in the presence of antibodies labeled with CSC markers, such as CD44 (IM7) Rat mAb (Alexa Fluor® 555 Conjugate; cat. no. #95235) and CD133 (A8N6N) Mouse mAb (Alexa Fluor® 647 Conjugate; cat. no. #53276; both 1:50; both Cell Signaling Technology, Inc.) on ice without light for 30 min, followed by PBS rinsing three times. Fluorescence-activated cell sorting Aria III (BD Biosciences) was employed to sort CD44+CD133+ cells.
Total RNA extraction from cells was performed using TRIzol (Invitrogen; Thermo Fisher Scientific, Inc.). Total RNA underwent RT to complementary DNA using a PrimeScript RT reagent kit (Takara Biotechnology Co., Ltd.) according to the manufacturer's instructions. qPCR was performed using TB Green® Premix Ex Taq II (Takara Biotechnology Co., Ltd.) and primers (Table I) on the ABI7900 fluorescence PCR instrument. Thermocycling conditions were as follows: Initial denaturation at 95°C for 10 min, followed by 40 cycles of denaturation at 95°C for 10 sec, annealing at 60°C for 20 sec and extension at 72°C for 34 sec. β-actin was used as the internal reference, and data analysis was conducted using the 2−ΔΔCq method (22).
BC cells were seeded in 6 cm culture dishes at a density of 1×103 cells/ml and cultured at 37°C for 14 days to colony formation. After the culture dish was washed with distilled water, cells were immobilized at room temperature with 4% paraformaldehyde (Sigma-Aldrich; Merck KGaA) for 15 min and then stained with crystal violet (Beyotime Institute of Biotechnology) for 30 min at room temperature. Images were captured and the quantification of cell colonies was performed manually. The colony was defined as a cell population large enough to be observed with the naked eye.
Serum-free DMEM/F-12 (Gibco) was supplemented with 20 ng/ml each epidermal growth factor (Invitrogen) and basic fibroblast growth factor (Gibco; all Thermo Fisher Scientific, Inc.). Then, 5 µg/ml insulin (Procell Life Science & Technology Co., Ltd.) and 100 µg/ml penicillin/streptomycin were added to a low adhesion 6-well plate. Cells were re-suspended with 1 ml medium added to the plate every 3 days. Afterward, cells were seeded at 1,000 cells/well into DMEM/F-12 (serum-free). Following 7 days of culture at 37°C, tumor spheres were collected once the diameter reached 50-100 µm. Spheres were detached and dispersed into single cells and re-seeded to form new tumor spheres once again. After 14 days at 37°C, the culture was terminated and the number of tumor sphere (diameter >50 µm) was calculated manually under the Leica DMi8 inverted light microscope (200X magnification).
Cells were stained at 37°C with 400 nM Mito-Tracker Green (cat. no. #C1046, Beyotime Institute of Biotechnology) for 40 min, and then blocked at 37°C for 1 h with 1% bovine serum albumin (BSA, cat. no. #735094; Amresco, LLC) in PBS. Following 1 h incubation at 37°C with the primary antibody light chain-3B (LC3B; cat. no. ab192890, 1:100), the secondary antibody 6478-Conjugated Goal Anti-Rabbit IgG (cat. no. ab150083, 1:200; both Abcam) was added to cells for 30-min at 37°C and nuclei were stained with 5 µg/ml DAPI (cat. no. #C1002; Beyotime Institute of Biotechnology) at room temperature for 3 min. Finally, the immunofluorescence was visualized using a laser confocal microscope (200×).
Analysis was conducted utilizing a simpleChIP®Plus ultrasonic ChIP kit (cat. no. #56383, Cell Signaling Technology, Inc.). Cells at 80% confluence were fixed using 4% paraformaldehyde at room temperature for 15 min and treated with glycine at room temperature for 15 sec at a final concentration of 0.125 M (both Beyotime Institute of Biotechnology) to halt the fixation process. Cells were lysed and centrifuged at 300 × g at 4°C for 5 min. Chromatin was resuspended in ChIP lysis buffer (50 mM HEPES-KOH pH 7.5, 140 mM NaCl, 1 mM EDTA pH 8, 1% Triton X-100, 0.1% deoxycholate, 0.1% SDS), 1% protease inhibitor) and sonicated on ice for 2 min (sonication: 3 sec on, 2 s off) to shear DNA-protein fragments to approximately 500 bp. 20 µl of cell lysate was incubated overnight at 4°C with 5 µg of rabbit Anti-H3K4me3 antibody (#9751S, 1:50, Cell Signaling Technology) or rabbit IgG (2729S, 1:100, Cell Signaling Technology). Protein-A beads (20 µl) were then used to capture the antibody complex. The beads were washed 7 times with wash buffer (50 mM HEPES, pH 7.5; 500 mM LiCl; 1 mM EDTA; 1% NP-40 and 0.7% deoxycholate), followed by one wash with Tris-EDTA buffer (10 mM Tris pH 8.0, 1 mM EDTA). The precipitated DNA was purified using a purification column and the expression of PYCR1 was detected by RT-qPCR as aforementioned. Primers were as follows: PYCR1-forward, 5′-CCT GTC ATC ATC TAA GAT AC-3′; PYCR1-reverse, 5′-CCC TGT CGG TGG CCA AGA TT-3′.
BALB/C nude mice (male, n=30; age, 5 weeks; weight 20-22 g) were purchased from Experimental Animal Resource Platform of the Chinese Academy of Sciences [animal license no. SYXK (Beijing) 2021-0063, Beijing, China]. Nude mice were housed in a standard animal house (25±2°C, 50±10% relative humidity, 12/12-h light-dark cycle and ad libitum access to food and water).
The BC xenograft model was established by subcutaneously injecting T24 cells (2×106 cells in 100 µl PBS) into nude mice. Mice were randomly divided (six mice/group) into groups as follows: BC; BC + lentivirus (Lv)-oe-SMYD2 (T24 cells delivered with pcDNA3.1-SMYD2 Lv were subcutaneously injected); BC + Lv-oe-NC (injection of T24 cells transfected with pcDNA3.1-NC Lv plasmid); BC + Lv-oe-SMYD2 + Lv-si-PYCR1 (T24 cells transfected with pcDNA3.1-SMYD2 + si-PYCR1 Lv plasmids were hypodermically injected); BC + Lv-oe-SMYD2 + Lv-si-NC (T24 cells transfected with pcDNA3.1-SMYD2 + si-NC Lv plasmids were subcutaneously injected). After 4 weeks, mice were euthanized via intraperitoneal injection of 100 mg/kg sodium pentobarbital and tumor tissue was obtained to measure the longest longitudinal (a) and transverse diameter (b). The tumor volume was calculated and weighed using the formula (a x b2)/2 (23), followed by protein extraction, hematoxylin-eosin (HE) staining and immunohistochemistry (IHC). The humane endpoints were body weight of the mice decreased by 40% or the tumor diameter more than 3 cm). No mice reached the humane endpoints.
The homogenates of cells or tumor tissues were lysed using RIPA lysis solution (Beyotime Institute of Biotechnology) containing protease inhibitors (Sigma-Aldrich; Merck KGaA) and supernatants were obtained upon 20 min centrifugation at 12,000 × g at 4°C. Protein concentration was determined using a BCA kit. The protein loading amount of each well was 10 µg. Following separation via 10% SDS-PAGE, protein bands were moved to a PVDF membrane and incubated overnight at 4°C with the following primary antibodies: Anti-PYCR1 (cat. no. ab226340, 1:2,000), anti-SMYD2 (cat. no. ab108217, 1:2,000), anti-Nanog (cat. no. ab21624, 1:200), anti-Sox2 (cat. no. ab92494, 1:1,000), anti-H3K4me3 (cat. no. ab272143, 1:500), anti-PINK1 (cat. no. ab216144, 1:1,000, all Abcam), anti-Parkin (cat. no. PA5-13399, 1:2,000, Invitrogen; Thermo Fisher Scientific, Inc.), anti-LC3B (cat. no. ab192890, 1:2,000), anti-p62 (cat. no. ab109012, 1:10,000) and β-actin (cat. no. ab8227, 1:1,000; all Abcam). Subsequently, the membrane was washed with TBS-tween (0.1% Tween 20; cat. no. ST673; Beyotime) and incubated with horseradish peroxidase-labeled goat anti-rabbit IgG (cat. no. ab205718, 1:2,000, Abcam) for 1 h at room temperature. Protein bands were visualized using the ultrasensitive enhanced chemiluminescence kit (cat. no. P0018S, Beyotime) and the gray values were quantified using Image J 1.8 software (National Institutes of Health), with β-actin serving as the internal reference. Cell experiments were repeated three times.
Rat tumor tissues were subjected to paraffin embedding after fixation with 4% paraformaldehyde at room temperature for 24 hand cut into 5-µm sections. The sections were stained at room temperature for 20 min with HE Stain kit (cat. no. G1120; Beijing Solarbio Science & Technology Co., Ltd.) and photographed under a light microscope (200×) (Olympus Corporation).
The tumor tissue subjected to 24-h fixation with 4% paraformaldehyde at room temperature was embedded in paraffin, serially sectioned (4 µm), dewaxed with xylene, and hydrated with gradient alcohol. Sections were treated with 3% H2O2 for 10 min to neutralize endogenous peroxidase activity. The samples were subjected to antigen retrieval in 0.01 mol/l sodium citrate buffer (pH=6.0, 15 min) using microwave heating at 95°C. The sections were blocked at room temperature for 20 min using 5% BSA (cat. no. A602440-0050, Sangon Biotechnology Co., Ltd.) and incubated overnight at 4°C with primary antibodies against H3K4me3 (cat. no. ab8580, 1:100), SMYD2 (cat. no. ab234862, 1:200), CD44 (cat no. ab316123, 1:5,000) and CD133 (cat. no. ab19898, 1:500; all Abcam). HRP labeled goat anti rabbit IgG (cat. no. ab6721, 1:1,000; Abcam) was incubated at room temperature for 20 min. After sections were rinsed with PBS, the color was developed using 3,3′-diaminobenzidine (cat. no. B-IMWRS51, AmyJet Scientific, Inc.). The staining was observed under a light microscope (200X magnification; Olympus Corporation). The percentages of positive cells were counted and averaged using Image J 1.8 (National Institutes of Health).
All data analysis was performed using SPSS 21.0 (IBM Corp.) and GraphPad Prism 9.5 (GraphPad Software, Inc.; Dotmatics). The data are presented as the mean ± SD of three independent experiments. Comparisons between two groups were performed using unpaired t-test, while multi-group comparisons were analyzed using one-way ANOVA followed by Tukey's multiple comparison test. P<0.05 was considered to indicate a statistically significant difference.
To investigate whether PYCR1 is involved in regulating BCSC stemness, BC cell lines (EJ and T24) were cultured in vitro. Subpopulations exhibiting SC characteristics (CD44+CD133+) were sorted from BC cells by flow cytometry and expression of PYCR1 mRNA and proteins in CD44+CD133+ and CD44−CD133− cells was assessed via RT-qPCR and western blot. The results showed that PYCR1 mRNA and its protein expression were higher in the CD44+CD133+ BC cell subpopulation than in CD44−CD133− cells (Fig. 1A and B). Following PYCR1 siRNA treatment, PYCR1 mRNA and protein expression in BC cells (Fig. 1C and D), CD44+CD133+ cell levels (Fig. 1E) and colony tumor sphere formation (Fig. 1F and G) and Nanog and Sox2 expression was reduced (Fig. 1H and I). Following transfection of pcDNA3.1-PYCR1, BC cells had increased expression of PYCR1 mRNA and protein (Fig. 1C and D) levels of CD44+CD133+ cells (Fig. 1E), increased formation of cancer cell colonies and tumor spheres (Fig. 1F and G) and elevated expression of Nanog and Sox2 (Fig. 1H and I). These results indicated that PYCR1 enhanced BCSC stemness maintenance.
Multiple post-translational modifications of proteins serve crucial roles in tumor progression, with methylation modification being common (24). SMYD2 is a protein lysine methyltransferase involved in the advancement of BC (10). SMYD2 was found to be significantly highly expressed in BC through the analysis of the GEPIA database (Fig. 2A). When BC cells were treated with SMYD2 siRNA, SMYD2 mRNA and protein expression (Fig. 2B and C), CD44+CD133+ cell levels (Fig. 2D), colony and tumor sphere formation (Fig. 2E and F) and Sox2 and Nanog expression decreased (Fig. 2G and H). However, following transfection of pcDNA3.1-SMYD2 into BC cells, SMYD2 mRNA and protein expression (Fig. 2B and C), levels of CD44+CD133+ cells (Fig. 2D), colony and tumor sphere formation (Fig. 2E and F) and the protein expression of stemness markers Nanog and Sox2 was increased (Fig. 2G and H). Altogether, these findings illustrated that the stemness retention of BCSCs was promoted by SMYD2.
SMYD2 was positively associated with PYCR1 expression in BC according to the GEPIA database analysis (Fig. 3A). Following transfection of SMYD2 siRNA in BC cells, PYCR1 mRNA and its protein expression and H3K4me3 levels decreased, while transfection of pcDNA3.1-SMYD2 into BC cells increased PYCR1 mRNA and protein expression and H3K4me3 levels (Fig. 3B and C). ChIP assay demonstrated enrichment of H3K4me3 in the PYCR1 promoter region was decreased following SMYD2 siRNA but increased following pcDNA3.1-SMYD2 transfection (Fig. 3D). Overall, SMYD2 modulated the expression of PYRC1 in BC cells by controlling the H3K4me3 levels in the PYRC1 promoter region.
To explore whether SMYD2 can promote stemness maintenance of BCSCs by modulating PYCR1, BC cells were transfected with pcDNA3.1-SMYD2 and PYCR1 siRNA. Compared with oe-SMYD2 + si-NC group, cells in the oe-SMYD2 + si-PYCR1 group exhibited decreased expression of PYCR1 mRNA and protein (Fig. 4A and B), CD44+CD133+ cell levels (Fig. 4C), colony and tumor sphere formation (Fig. 4D and E) and Nanog/Sox2 expression Fig. 4F and G). These results suggested that SMYD2 contributed to BCSC stemness maintenance by modulating expression of PYCR1.
Following PYCR1 siRNA treatment, mitochondrial LC3B expression (Fig. 5A) and LC3B II/I ratio decreased and p62 protein expression was augmented (Fig. 5B); these alterations were reversed following pcDNA3.1-PYCR1 transfection. Additionally, following treatment with PYCR1 siRNA, expression of PINK1 and Parkin proteins was decreased; after transfection of pcDNA3.1-PYCR1, expression of PINK1 and Parkin proteins increased (Fig. 5B). BC cells were transfected with PINK1 siRNA and pcDNA3.1-PYCR1. In comparison with the oe-PYCR1 + si-NC group, oe-PYCR1 + si-PINK1 group exhibited reduced PINK1/Parkin protein (Fig. 5B) and LC3B expression (Fig. 5A) and LC3B II/I ratio, raised p62 protein expression (Fig. 5B) and inhibited CD44+CD133+ cell levels (Fig. 5C), formation of tumor spheres and colonies (Fig. 5D and E) and expression of Sox2 and Nanog (Fig. 5F and G). Collectively, these findings demonstrated that PYCR1 enhanced BCSC stemness maintenance via regulation of the PINK1/Parkin mitophagy pathway.
A BC xenograft model was established for in vivo validation experiments by subcutaneously injecting T24 cells transfected with pcDNA3.1-SMYD2 or si-PYCR1 Lv plasmid into nude mice. Tumor size and weight in the BC + Lv-oe-SMYD2 group were higher than those in the BC + Lv-oe-NC group; tumor weight and size in the BC + Lv-oe-SMYD2 + Lv-si-PYCR1 group were lower than those in the BC + Lv-oe-SMYD2 + si-NC group (Fig. 6A-C). HE staining showed that compared with the BC + Lv-oe-NC group, the tumor cell density of the BC + Lv-oe-SMYD2 group was elevated and the proportion of necrotic cells (unstructured eosinophilic substances) were decreased; compared with the BC + Lv-oe-SMYD2 + si-NC group, the tumor cell density of nude mice in the BC + Lv-oe-SMYD2 + Lv-si-PYCR1 group was decreased and the proportion of necrotic cells increased (Fig. 6D). By contrast with the BC + Lv-oe-NC group, protein levels of SMYD2, PYCR1, PINK1 and Parkin in tissue homogenate of nude mice in the BC + Lv-oe-SMYD2 group were elevated (Fig. 6E). SMYD2, Parkin, PYCR1 and PINK1 expression in the BC + Lv-oe-SMYD2 + Lv-si-PYCR1 group was lower than in the BC + Lv-oe-SMYD2 + si-NC group (Fig. 6E). As demonstrated by IHC results, SMYD2-, H3K4me3-, CD44- and CD133-positive cell numbers in the BC + Lv-oe-SMYD2 group were significantly augmented relative to the BC + Lv-oe-NC group; nude mice in the BC + Lv-oe-SMYD2 + Lv-si-PYCR1 group displayed lower CD133- and CD44-positive cell numbers than those in the BC + Lv-oe-SMYD2 + si-NC group (Fig. 6F). Western blot results revealed that nude mice in the BC + Lv-oe-SMYD2 group exhibited a higher LC3B II/I ratio and lower p62 protein levels than the BC + Lv-oe-NC group; BC + Lv-oe-SMYD2 + Lv-si-PYCR1 group exhibited a higher LC3B II/I ratio and lower p62 protein expression compared with BC + Lv-oe-SMYD2 + si-NC group (Fig. 6G). Moreover, Nanog and Sox2 protein levels were raised in the BC + Lv-oe-SMYD2 group vs. BC + Lv-oe-NC group but these protein levels dropped in the BC + Lv-oe-SMYD2 + Lv-si-PYCR1 compared with BC + Lv-oe-SMYD2 + si-NC group (Fig. 6H and I). In summary, these results indicated that SMYD2 sustained BCSC stemness by increasing PYCR1 expression to stimulate the PINK1/Parkin mitophagy pathway.
Lysine methyltransferases, which affect numerous oncoproteins and tumor suppressor proteins, play a key role in the development of various types of malignancies, such as colorectal cancer and lung cancer (25-27). SMYD2, an extensively studied lysine methyltransferase, is associated with regulation of transcription, epigenetics and tumorigenesis (28,29). However, the potential impact of SMYD2 on the stemness maintenance of BCSCs remains unclear. The present study demonstrated that histone methyltransferase SMYD2 is involved in the stemness of BCSCs by upregulating PYCR1 expression and activating the PINK1/Parkin pathway (Fig. 7).
PYCR1 strengthens BC proliferation and EMT, which is associated with expression of stemness markers Nanog and Sox2 (4,30), suggesting that PYCR1 plays a role in BCSC stemness. GEPIA database analysis demonstrated that SMYD2 was significantly highly expressed in BC. Correspondingly, SMYD2 expression is significantly increased in human BC compared with normal bladder tissue, underscoring that inhibiting SMYD2 may have therapeutic promise for BC (31). The present study assessed the mechanism of SMYD2/PYCR1 in maintaining BCSC stemness. CD44, a hallmark of BCSCs, indicates stemness and activates many signaling pathways to maintain self-renewal capacity (32,33). The potential role of CD44 and CD133 as markers for BCSCs has been proposed (34). PYCR1 and SMYD2 knockdown in CD44+CD133+ BCSC subpopulation with stem cell-like characteristics decreased CD44+CD133+ cell levels, Nanog and Sox2 expression and colony and sphere formation, whereas overexpressing PYCR1 or SMYD2 resulted the opposite effects. Similarly, suppression of PYCR1 in vitro decreases colony formation and proliferation and induces cell cycle arrest (35). Cui et al (20) confirmed that ablation of PYCR1 can lead to reduced expression of Sox2 and Nanog, CSC-associated aldehyde dehydrogenase+ population and ability of breast CSCs to form spheres, suggesting that PYCR1 is involved in preserving the stemness of breast cancer cells. In addition, Shang and Wei (36) demonstrated that inhibition of SMYD2 impedes tumor sphere formation and cell migration in non-small-cell lung cancer cells. To the best of our knowledge, the impact of SMYD2 on the stemness of BCSCs has not been reported. It was hypothesized that both SMYD2 and PYCR1 amplified maintenance of stemness in BCSCs.
H3K4me3 is abnormally expressed in lung, liver and colon cancer (37-39). SMYD2 serves as a histone methyltransferase of H3K4me3, facilitating transcriptional activation of its downstream target genes by catalyzing trimethylation of histone H3K4, which is associated with modulation of target gene transcription (12,40). A notable positive association between SMYD2 and PYRC1 was observed in the present study. To the best of our knowledge, the present study is the first to demonstrate that SMYD2 may promote PYCR1 expression via H3K4me3; in vitro demonstrated increased PYCR1 and H3K4me3 levels, along with heightened enrichment levels of H3K4me3 in the PYCR1 promoter region in SMYD2-overexpressing BC cells. Furthermore, previous research has confirmed that proline produced by PYCR1 can activate the cGMP/cGMP-dependent protein kinase signaling pathway, thereby enhancing breast cancer stemness (20). Here, SMYD2 heightened stemness in BCSCs by regulating PYCR1.
The PINK1/Parkin signaling pathway is one of the primary regulatory mechanisms of mitophagy (17). The accumulation of PINK1 augments PINK1/Parkin-mediated mitophagy, which efficiently removes impaired mitochondria (41). The feedforward signaling pathway, including PINK1 and Parkin, is responsible for facilitating mitophagy (42). Here, PYCR1 silencing resulted in elevated p62 and Parkin/PINK1 expression, as well as lessened LC3B expression and LC3B II/I ratios, which were negated by PYCR1 overexpression. This indicated that PYCR1 could mediate the Parkin/PINK1 mitophagy axis. Moreover, it is well-established that mitophagy encourages the obtainment of tumor cell stemness (14,43). The present experiments verified the promoting effect of the PINK1/Parkin mitophagy pathway in BCSC stemness. Other investigations have established a connection between mitophagy and SC stemness (44,45). Augmentation of mitophagy can contribute to the stemness of mesenchymal SCs (46). Conversely, the reduction of mitophagy-related PINK1/Parkin leads to diminished stemness of bone marrow-derived mesenchymal SCs, as indicated by decreased expression of stemness markers Sox2 and Octamer-binding transcription factor 4 (44). Here, PYCR1 stimulated stemness maintenance in BCSCs via the PINK1/Parkin mitophagy pathway. Furthermore, in vivo verification demonstrated that SMYD2 preserved BCSC stemness by activating the PINK1/Parkin mitophagy pathway via enhancement of PYCR1 expression.
In conclusion, the present study demonstrated the regulatory effect and mechanism by which SMYD2 affects PYCR1. Additionally, in vitro and in vivo experiments demonstrated the role of PYCR1 in modulating the PINK1/Parkin signaling pathway, which affects mitophagy and regulates stemness of BCSCs. However, the present study only verified that PYCR1 regulates mitophagy via regulation of PINK1/Parkin to regulate BCSC stemness; other target genes and signaling pathways regulated by PYCR1 require further investigation. Upstream influencing factors of PYCR1 also need to be further explored. Future investigations investigate the role of mitophagy in preserving the stemness of CSCs, as well as the mechanisms by which PYCR1 modulates other target genes and signaling pathways.
The data generated in the present study may be requested from the corresponding author.
All authors have read and approved the final manuscript. JC contributed to the study concepts, study design; SX and XY performed the literature review. JC and YW performed experiments. JC and WS contributed to the data analysis. JC and SX wrote and revised the manuscript. JC and SX confirm the authenticity of all the raw data.
The present study was reviewed and approved by the Animal Ethics Committee of Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University (approval no. 2024-84).
Not applicable.
The authors declare that they have no competing interests.
Not applicable.
The present study was supported by Natural Science Foundation of Hunan Province (grant no. 2025JJ50640), Hunan Provincial Health High-Level Talent Scientific Research Project (grant no. R2023154) and Hunan Provincial People's Hospital Doctoral Fund Project (grant no. BSJJ202106).
|
Chen S, Zhu J, Wang F, Guan Z, Ge Y, Yang X and Cai J: LncRNAs and their role in cancer stem cells. Oncotarget. 8:110685–110692. 2017. View Article : Google Scholar | |
|
Eid RA, Alaa Edeen M, Shedid EM, Kamal AS, Warda MM, Mamdouh F, Khedr SA, Soltan MA, Jeon HW, Zaki MS and Kim B: Targeting Cancer Stem Cells as the Key Driver of Carcinogenesis and Therapeutic Resistance. Int J Mol Sci. 24:2023. View Article : Google Scholar | |
|
Aghaalikhani N, Rashtchizadeh N, Shadpour P, Allameh A and Mahmoodi M: Cancer stem cells as a therapeutic target in bladder cancer. J Cell Physiol. 234:3197–3206. 2019. View Article : Google Scholar | |
|
Li Z, Liu J, Fu H, Li Y, Liu Q, Song W and Zeng M: SENP3 affects the expression of PYCR1 to promote bladder cancer proliferation and EMT transformation by deSUMOylation of STAT3. Aging (Albany NY). 14:8032–8045. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Song W, Li Z, Yang K, Gao Z, Zhou Q and Li P: Antisense lncRNA-RP11-498C9.13 promotes bladder cancer progression by enhancing reactive oxygen species-induced mitophagy. J Gene Med. 25:e35272023. View Article : Google Scholar : PubMed/NCBI | |
|
Song W, Yang K, Luo J, Gao Z and Gao Y: Dysregulation of USP18/FTO/PYCR1 signaling network promotes bladder cancer development and progression. Aging (Albany NY). 13:3909–3925. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Zhang X, Huang X, Tang X, Zhang M, Li Z, Hu X, Zhang M, Wang X and Yan Y: Tumor stemness score to estimate epithelial-to-mesenchymal transition (EMT) and cancer stem cells (CSCs) characterization and to predict the prognosis and immunotherapy response in bladder urothelial carcinoma. Stem Cell Res Ther. 14:152023. View Article : Google Scholar : PubMed/NCBI | |
|
Ye Y, Li L, Dai Q, Liu Y and Shen L: Comprehensive analysis of histone methylation modification regulators for predicting prognosis and drug sensitivity in lung adenocarcinoma. Front Cell Dev Biol. 10:9919802022. View Article : Google Scholar : PubMed/NCBI | |
|
McCabe MT, Mohammad HP, Barbash O and Kruger RG: Targeting Histone Methylation in Cancer. Cancer J. 23:292–301. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Tan Z, Fu S, Feng R, Huang Y, Li N, Wang H and Wang J: Identification of potential biomarkers for progression and prognosis of bladder cancer by comprehensive bioinformatics analysis. J Oncol. 2022:18027062022. View Article : Google Scholar : PubMed/NCBI | |
|
Xu W, Chen F, Fei X, Yang X and Lu X: Overexpression of SET and MYND domain-containing protein 2 (SMYD2) is associated with tumor progression and poor prognosis in patients with papillary thyroid carcinoma. Med Sci Monit. 24:7357–7365. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Xu H, Ba Z, Liu C and Yu X: Long noncoding RNA DLEU1 promotes proliferation and glycolysis of gastric cancer cells via APOC1 upregulation by recruiting SMYD2 to induce trimethylation of H3K4 modification. Transl Oncol. 36:1017312023. View Article : Google Scholar : PubMed/NCBI | |
|
Whelan KA, Chandramouleeswaran PM, Tanaka K, Natsuizaka M, Guha M, Srinivasan S, Darling DS, Kita Y, Natsugoe S, Winkler JD, et al: Autophagy supports generation of cells with high CD44 expression via modulation of oxidative stress and Parkin-mediated mitochondrial clearance. Oncogene. 36:4843–4858. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Liu K, Lee J, Kim JY, Wang L, Tian Y, Chan ST, Cho C, Machida K, Chen D and Ou JJ: Mitophagy controls the activities of tumor suppressor p53 to regulate hepatic cancer stem cells. Mol Cell. 68:281–292. 2017. 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 | |
|
Lou Y, Ma C, Liu Z, Shi J, Zheng G, Zhang C and Zhang Z: Antimony exposure promotes bladder tumor cell growth by inhibiting PINK1-Parkin-mediated mitophagy. Ecotoxicol Environ Saf. 221:1124202021. View Article : Google Scholar : PubMed/NCBI | |
|
Nguyen TN, Padman BS and Lazarou M: Deciphering the molecular signals of PINK1/Parkin mitophagy. Trends Cell Biol. 26:733–744. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Lee J, Liu K, Stiles B and Ou JJ: Mitophagy and hepatic cancer stem cells. Autophagy. 14:715–716. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Choudhury D, Rong N, Senthil Kumar HV, Swedick S, Samuel RZ, Mehrotra P, Toftegaard J, Rajabian N, Thiyagarajan R, Podder AK, et al: Proline restores mitochondrial function and reverses aging hallmarks in senescent cells. Cell Rep. 43:1137382024. View Article : Google Scholar : PubMed/NCBI | |
|
Cui B, He B, Huang Y, Wang C, Luo H, Lu J, Su K, Zhang X, Luo Y, Zhao Z, et al: Pyrroline-5-carboxylate reductase 1 reprograms proline metabolism to drive breast cancer stemness under psychological stress. Cell Death Dis. 14:6822023. View Article : Google Scholar : PubMed/NCBI | |
|
Zietzer A, Hosen MR, Wang H, Goody PR, Sylvester M, Latz E, Nickenig G, Werner N and Jansen F: The RNA-binding protein hnRNPU regulates the sorting of microRNA-30c-5p into large extracellular vesicles. J Extracell Vesicles. 9:17869672020. 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 | |
|
Han Y, Liu C, Zhang D, Men H, Huo L, Geng Q, Wang S, Gao Y, Zhang W, Zhang Y and Jia Z: Mechanosensitive ion channel Piezo1 promotes prostate cancer development through the activation of the Akt/mTOR pathway and acceleration of cell cycle. Int J Oncol. 55:629–644. 2019.PubMed/NCBI | |
|
Dai X, Ren T, Zhang Y and Nan N: Methylation multiplicity and its clinical values in cancer. Expert Rev Mol Med. 23:e22021. View Article : Google Scholar : PubMed/NCBI | |
|
Komatsu S, Ichikawa D, Hirajima S, Nagata H, Nishimura Y, Kawaguchi T, Miyamae M, Okajima W, Ohashi T, Konishi H, et al: Overexpression of SMYD2 contributes to malignant outcome in gastric cancer. Br J Cancer. 112:357–364. 2015. View Article : Google Scholar : | |
|
Zhang Y, Zhou L, Xu Y, Zhou J, Jiang T, Wang J, Li C, Sun X, Song H and Song J: Targeting SMYD2 inhibits angiogenesis and increases the efficiency of apatinib by suppressing EGFL7 in colorectal cancer. Angiogenesis. 26:1–18. 2023. View Article : Google Scholar | |
|
Kim K, Ryu TY, Jung E, Han TS, Lee J, Kim SK, Roh YN, Lee MS, Jung CR, Lim JH, et al: Epigenetic regulation of SMAD3 by histone methyltransferase SMYD2 promotes lung cancer metastasis. Exp Mol Med. 55:952–964. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Sakamoto LH, Andrade RV, Felipe MS, Motoyama AB and Pittella Silva F: SMYD2 is highly expressed in pediatric acute lymphoblastic leukemia and constitutes a bad prognostic factor. Leuk Res. 38:496–502. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Yi X, Jiang XJ and Fang ZM: Histone methyltransferase SMYD2: Ubiquitous regulator of disease. Clin Epigenetics. 11:1122019. View Article : Google Scholar : PubMed/NCBI | |
|
Migita T, Ueda A, Ohishi T, Hatano M, Seimiya H, Horiguchi SI, Koga F and Shibasaki F: Epithelial-mesenchymal transition promotes SOX2 and NANOG expression in bladder cancer. Lab Invest. 97:567–576. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Cho HS, Hayami S, Toyokawa G, Maejima K, Yamane Y, Suzuki T, Dohmae N, Kogure M and Kang D: RB1 methylation by SMYD2 enhances cell cycle progression through an increase of RB1 phosphorylation. Neoplasia. 14:476–486. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Gao RL, Chen XR, Li YN, Yan XY, Sun JG, He QL and Cai FZ: Upregulation of miR-543-3p promotes growth and stem cell-like phenotype in bladder cancer by activating the Wnt/β-catenin signaling pathway. Int J Clin Exp Pathol. 10:9418–9426. 2017. | |
|
Hu Y, Zhang Y, Gao J, Lian X and Wang Y: The clinicopathological and prognostic value of CD44 expression in bladder cancer: A study based on meta-analysis and TCGA data. Bioengineered. 11:572–581. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Xia P, Liu DH, Xu ZJ and Ren F: Cancer stem cell markers for urinary carcinoma. Stem Cells Int. 2022:36116772022. View Article : Google Scholar : PubMed/NCBI | |
|
Cai F, Miao Y, Liu C, Wu T, Shen S, Su X and Shi Y: Pyrroline-5-carboxylate reductase 1 promotes proliferation and inhibits apoptosis in non-small cell lung cancer. Oncol Lett. 15:731–740. 2018.PubMed/NCBI | |
|
Shang L and Wei M: Inhibition of SMYD2 sensitized cisplatin to resistant cells in NSCLC through activating p53 pathway. Front Oncol. 9:3062019. View Article : Google Scholar : PubMed/NCBI | |
|
Lin X, Chen JD, Wang CY, Cai Z, Zhan R, Yang C, Zhang LY, Li LY, Xiao Y, Chen MK and Wu M: Cooperation of MLL1 and Jun in controlling H3K4me3 on enhancers in colorectal cancer. Genome Biol. 24:2682023. View Article : Google Scholar : PubMed/NCBI | |
|
Phoyen S, Sanpavat A, Ma-On C, Stein U, Hirankarn N, Tangkijvanich P, Jindatip D, Whongsiri P and Boonla C: H4K20me3 upregulated by reactive oxygen species is associated with tumor progression and poor prognosis in patients with hepatocellular carcinoma. Heliyon. 9:e225892023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou Z, Zhang B, Deng Y, Deng S, Li J, Wei W, Wang Y, Wang J, Feng Z, Che M, et al: FBW7/GSK3 β mediated degradation of IGF2BP2 inhibits IGF2BP2-SLC7A5 positive feedback loop and radioresistance in lung cancer. J Exp Clin Cancer Res. 43:342024. View Article : Google Scholar | |
|
Abu-Farha M, Lambert JP, Al-Madhoun AS, Elisma F, Skerjanc IS and Figeys D: The tale of two domains: proteomics and genomics analysis of SMYD2, a new histone methyltransferase. Mol Cell Proteomics. 7:560–572. 2008. View Article : Google Scholar | |
|
Wang S, Long H, Hou L, Feng B, Ma Z, Wu Y, Zeng Y, Cai J, Zhang DW and Zhao G: The mitophagy pathway and its implications in human diseases. Signal Transduct Target Ther. 8:3042023. View Article : Google Scholar : PubMed/NCBI | |
|
Silvian LF: PINK1/parkin pathway activation for mitochondrial quality control-which is the best molecular target for therapy? Front Aging Neurosci. 14:8908232022. View Article : Google Scholar | |
|
Yuan X, Chen K, Zheng F, Xu S, Li Y, Wang Y, Ni H, Wang F, Cui Z, Qin Y, et al: Low-dose BPA and its substitute BPS promote ovarian cancer cell stemness via a non-canonical PINK1/p53 mitophagic signaling. J Hazard Mater. 452:1312882023. View Article : Google Scholar : PubMed/NCBI | |
|
Feng X, Yin W, Wang J, Feng L and Kang YJ: Mitophagy promotes the stemness of bone marrow-derived mesenchymal stem cells. Exp Biol Med (Maywood). 246:97–105. 2021. View Article : Google Scholar | |
|
Liu D, Sun Z, Ye T, Li J, Zeng B, Zhao Q, Wang J and Xing HR: The mitochondrial fission factor FIS1 promotes stemness of human lung cancer stem cells via mitophagy. FEBS Open Bio. 11:1997–2007. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Vazquez-Martin A, Van den Haute C, Cufi S, Corominas-Faja B, Cuyàs E, Lopez-Bonet E, Rodr iguez-Gallego E, Fernández-Arroyo S, Joven J, Baekelandt V and Menendez JA: Mitophagy-driven mitochondrial rejuvenation regulates stem cell fate. Aging (Albany NY). 8:1330–1352. 2016. View Article : Google Scholar : PubMed/NCBI |