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.
Infantile hemangioma (IH) is the most common vascular tumor in children, with an incidence of 3–10% (1). Due to its rapid growth, some IHs lead to long-term residual permanent skin damage and might affect the physical and mental health of children (2). Although a number of studies have focused on the mechanisms underlying IH progression, the detailed regulatory mechanisms are still not fully understood.
Long non-coding RNAs (lncRNAs), with a length of >200 nucleotides, have crucial roles in hemangioma cell proliferation and invasion (3,4). A recent study reported that CTBP1-AS2 sponges microRNA (miR)-335-5p, upregulates C-C motif chemokine ligand 2 and subsequently enhances hemangioma angiogenesis and progression (5). MIR4435-2HG, which is secreted by M2 macrophages, targets Heterogeneous nuclear ribonucleoprotein A1 and promotes IH progression (4). Most notably, our previous study revealed that knocking down lncRNA NEAT1 repressed IH progression by sponging miR-33a-5p and regulating the downstream hypoxia inducible factor 1 subunit α (HIF1α)/NF-κB pathway (6). Additional studies have further reported that in hemangioma, lncRNA NEAT1 is positively regulated by alkB homolog 5, RNA demethylase in an m6A modification-dependent manner and promotes hemangioma development by regulating the miR-361-5p/VEGFA pathway (7,8). However, the exact regulatory roles of lncRNA NEAT1 in hemangioma progression are still largely unknown.
Histone lactylation (addition of a lactyl group to a lysine residue) by P300 and KAT8 is a protein modification newly discovered in 2019 that affects transcription (9,10), and has been found to widely participate in disease progression including cancer, atrial fibrillation and keloids (11,12). Recent studies have shown that H3 lysine 18 lactylation (H3K18la) can activate gene transcription (13,14). However, the roles of protein lactylation in hemangioma development are still unclear.
In the present study, the effects of the lncRNA NEAT1/H3K18la/β-catenin signaling pathway on the colony formation ability and viability of hemangioma cells were investigated. Additionally, the detailed mechanism by which NEAT1 regulates β-catenin in hemangioma cells was further investigated.
IH tissues (proliferating stage; median age, 7 months; n=10, Table I) and normal adjacent subcutaneous tissues (n=10) were collected from the Kunming Children's Hospital (Kunming, China) between February 2022 and September 2024. Samples were kept at −80°C until analysis. Informed consent was obtained from the parents/legal guardians of each patient. The patients received no treatment before surgery. The Ethics Committee of Kunming Children's Hospital approved the present study (approval no. 2021-03-181-K01). The inclusion criteria were as follows: i) The patients were diagnosed with IH (proliferating stage); ii) age of <1 years old; and iii) The hemangioma lesion was located in concealed areas such as the trunk and limbs, and the parents voluntarily opted for surgical removal. The exclusion criteria were as follows: i) Multiple hemangiomas; ii) vascular malformation; and iii) the patients had received any other treatment.
Hemangioma endothelial cells (HemECs) were prepared following the protocol described in a previous study (15) using the hemangioma sample from patient no. 4 in Table I. The HemECs were cultured in human endothelial serum-free medium (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% FBS (Gibco; Thermo Fisher Scientific, Inc.) at 5% CO2 and 37°C.
Lipofectamine 3000 (Thermo Fisher Scientific, Inc.) was used to transfect small interfering RNAs (siRNA; 50 nM; Shanghai GenePharma Co., Ltd.) into the cells at 37°C for 48 h. Then, the cells were collected for reverse transcription-quantitative PCR (RT-qPCR), western blotting, chromatin immunoprecipitation (ChIP)-PCR and cell viability assays. Table SI lists the sequences of the siRNAs.
2-Deoxy-D-glucose (2-DG), oxamate, lactate, MG132, bafilomycin A1 (BafA1), cycloheximide (CHX) and A-485 were purchased from Selleck Chemicals. In the present study, 10 mM 2-DG, 20 mM oxamate and 10 mM lactate were used to treat the HemECs (16–19) (RT-qPCR, western blotting, ChIP-PCR and cell viability assays: 37°C for 48 h; colony formation assay: 37°C for 7–9 days), and the protein lactylation was inactivated (2-DG and oxamate) and activated (lactate). At 46.5 and 46 h after transfection of NEAT1 siRNA, 20 µM MG132 (treatment: 1.5 h at 37°C) and 100 nM BafA1 (treatment: 2 h at 37°C) were used to suppress the proteosome and lysosome, respectively (20), and the cells were harvested for western blotting after a total time of 48 h of transfection with NEAT1 siRNA. At 33, 36, 39, 42 and 45 h after transfection of NEAT1 siRNA (corresponding to the 15, 12, 9, 6 and 3 h treatment group), 100 µg/ml CHX was used to treat the cells (20), and the cells were harvested for western blotting after a total time of 48 h of transfection with NEAT1 siRNA. Additionally, 5 µM A-485 (RT-qPCR, western blotting, cell viability assays: 37°C for 48 h; colony formation assay: 37°C for 7–9 days) were applied to block p300 (21).
Total RNA from tissues and cells was extracted using RNAiso Esay Plus (Wuhan Servicebio Technology Co., Ltd.) according to the manufacturer's instructions. First-strand cDNA synthesis and qPCR were performed using the HifiScript cDNA Synthesis Kit (Cwbiotech) and PowerUp™ SYBR™ Green Master Mix (Thermo Fisher Scientific, Inc.), respectively, according to the manufacturer's protocol. The qPCR program was as follows: 95°C for 2 min, followed by 40 cycles of 95°C for 15 sec and 60°C for 1 min. The 2−ΔΔcq method was used to calculate the relative expression of the genes (22). GAPDH was used for normalization. Table SII lists the primer sequences.
A total of 500 cells per well were plated in 12-well plate and then incubated for 7–9 days [lactate dehydrogenase B (LDHB)-knock down cells: cells were seeded after transfection for 24 h; 2-DG/oxamate/lactate/A-485 treated cells: drugs were used to treat after seeding for 24 h]. At the end of the experiment, the cells were fixed with 4% paraformaldehyde for 15 min at room temperature and then stained with 0.1% crystal violet (Beyotime Biotechnology) for 30 min at room temperature. Colonies (>50 cells) were visualized and quantified by light microscopy. The cell viability was detected using Cell Counting Kit-8 (Dojindo Laboratories, Inc.) assays as previously described (6).
According to the protocol previously described (23), the hemangioma cells were harvested and washed twice with ice-cold PBS. The hemangioma cells were suspended in RIP buffer (1 ml) containing 50 mmol/l Tris pH 7.4, 0.5% NP-40, 1X RNasin plus (Promega Corporation), 150 mmol/l NaCl, 2 mmol/l ribonucleoside vanadyl complex (New England BioLabs, Inc.), 1X protease inhibitor cocktail (MilliporeSigma) and 1 mmol/l PMSF. After brief sonication, cell lysates were centrifuged at 10,000 × g for 10 min at 4°C and the supernatants were precleared using 10 µl Dynabeads Protein G (Thermo Fisher Scientific, Inc.). The precleared supernatants were divided into two equal parts, and one part was incubated with 1 µg LDHB antibody (cat. no. 14824-1-AP; Proteintech Group, Inc.) and 20 µl Protein A/G Magnetic Beads (cat. No. HY-K0202; MedChemExpress) overnight at 4°C. The other part was incubated with 1 µg IgG control antibody (cat. no. 30000-0-AP; Proteintech Group, Inc.) and 20 µl Protein A/G Magnetic Beads overnight at 4°C. After washing the beads three times with RIP buffer, the samples were decrosslinked by proteinase K buffer (50 mmol/l Tris pH 7.4, 150 mmol/l NaCl, 1X RNasin plus, 0.5% SDS and 200 µg/ml proteinase K) at room temperature for 30 min. Then, the RNA was extracted and RT-qPCR was performed.
The SimpleChIP Enzymatic Chromatin IP Kit (Magnetic Beads) (cat. no. CST-9003; Cell Signaling Technology, Inc.) were used to analyze the H3K18 lactylation level of the promoter of the β-catenin gene (CTNNB1) following the manufacturer's protocol. The 2-DG/oxamate-treated cells, lactate-treated cells, NEAT1-knockdown cells and LDHB-knockdown cells were crosslinked with 1% formaldehyde for 10 min at room temperature, then collected after washing twice using PBS (containing 0.5 mM EDTA). The cell pellet was lysed with 0.3 ml of cell lysis buffer (50 mM Tris-HCl pH 8.1, 10 mM EDTA, 1% SDS and protease inhibitor) and incubated for 10 min on ice. The cell lysates were sonicated (sonication for 15 sec, with a 10-sec interval, totaling 3 min of sonication) to obtain DNA fragments (150–900 base pair in length) and ~50 µg of cross-linked sheared chromatin solution was then used for immunoprecipitation. The L-lactyl-histone H3 (Lys18) antibody (cat. no. PTM-1427RM; PTM BIO LLC; 6 µg/5×106 cells) was incubated with the sample overnight at 4°C on a rotating shaker. Magnetic beads were added to the solution, incubated at 4°C for 1 h and then washed with washing buffer. The cross-linking was reversed by adding NaCl at a final concentration of 200 mM and heating at 65°C for 30 min. The DNA fragments were purified using a spin column and qPCR was performed. The sequences of the primers used were as follows: CTNNB1 promoter forward primer, 5′-CCTAGTGACAAGTGGAACCAGA-3′; and reverse primer, 5′-GAACTCTCCGTAGAACGGGC-3′.
The western blot procedure was described in our previous study (6). The primary antibodies used were LDHB (cat. no. 14824-1-AP; 1:5,000), β-catenin (cat. no. 51067-2-AP; 1:5,000), alanyl-tRNA synthetase 1 (AARS1; cat. no. 17394-1-AP; 1:2,000), P300 (cat. no. 20695-1-AP; 1:1,000) and GAPDH (cat. no. 60004-1-Ig; 1:50,000) from Proteintech Group, Inc., and L-lactyl-Histone H3 (Lys18) (cat. no. PTM-1406RM; 1:1,000), L-lactyl lysine (pan-lac; cat. no. PTM-1401RM; 1:1,000) and Histone H3 (cat. no. PTM-1002RM; 1:1,000) from PTM BIO LLC. All primary antibodies were incubated overnight at 4°C. The secondary antibodies used were HRP-conjugated Goat Anti-Mouse IgG (H+L) (cat. no. SA00001-1; 1:10,000; Proteintech Group, Inc.) and HRP-conjugated Goat Anti-Rabbit IgG (H+L) (cat, no. SA00001-2; 1:10,000; Proteintech Group, Inc.), which were incubated for 1 h at room temperature.
The cellular lactate concentrations were detected using a Lactic Acid assay kit (cat. no. BC2230; Beijing Solarbio Science & Technology Co., Ltd.) according to the manufacturer's protocol.
The NEAT1-interacting proteins were predicted using catRAPID software (http://s.tartaglialab.com/page/catrapid_group; accessed on September 5, 2024). Based on the NEAT1-interacting proteins (predicted by catRAPID; Interaction Propensity >50), Gene Oncology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed using the DAVID platform (24,25).
GraphPad Prism 10 software (Dotmatics) was used for data analysis. The quantitative data are shown as the mean ± SD and were analyzed via one-way with Tukey's multiple comparisons test (multiple groups) or unpaired Student's t-test (two groups). P<0.05 was considered to indicate a statistically significant difference.
Our previous study reported that NEAT1 was highly expressed in IH tissues (6). In the present study, to investigate the regulatory mechanisms underlying NEAT1-affected hemangioma progression, the proteins that interact with NEAT1 were analyzed using catRAPID software. In total, 2,064 proteins (such as HSP90B1 and CDK11B) were predicted to interact with NEAT1. GO and pathway enrichment analyses of these genes were subsequently performed using DAVID. The biological process enrichment analysis revealed that NEAT1-interacting proteins were associated with processes such as ‘mRNA processing’, ‘mRNA splicing, via spliceosome’, ‘RNA splicing’, ‘Translation’ and ‘rRNA processing’ (Fig. 1A). Cellular component enrichment analysis revealed that NEAT1-interacting proteins were associated with components such as ‘Nucleoplasm’, ‘Nucleolus’, ‘Ribonucleoprotein complex’, ‘Nucleus’ and ‘Nuclear speck’ (Fig. 1B). Molecular function enrichment analysis revealed that NEAT1-interacting proteins were associated with functions such as ‘RNA binding’, ‘mRNA binding’, ‘Double-stranded RNA binding’, ‘RNA helicase activity’ and ‘Structural constituent of ribosome’ (Fig. 1C). KEGG enrichment analysis revealed that NEAT1-interacting proteins were associated with ‘Nucleocytoplasmic transport’, ‘Spliceosome,’ ‘Ribosome’, ‘RNA degradation’ and ‘mRNA surveillance pathway’ (Fig. 1D).
Notably, it was found that LDHB was predicted to bind with NEAT1 using catRAPID software (Fig. 2A and B). The interaction between NEAT1 and LDHB in hemangioma cells was validated using a RIP assay (Fig. 2C). Additionally, after knocking down NEAT1 expression (Fig. 2D), the protein level (Fig. 2E) but not the mRNA level of LDHB (Fig. 2F) was reduced, indicating that NEAT1 may regulate LDHB at the protein level. After protein translation was inhibited via CHX, LDHB degradation occurred faster in NEAT1-knock down cells than in the negative control cells (Fig. 2G). The proteasome inhibitor MG132, but not the lysosome inhibitor BafA1, reversed the effect of NEAT1 knockdown on the protein level of LDHB, indicating that NEAT1 knockdown reduced the LDHB protein levels via activation of the proteasome activity (Fig. 2H). These results suggested that the lncRNA NEAT1 interacted with and positively regulated LDHB in hemangioma cells.
LDHB and LDHA have critical roles in converting pyruvate to lactate during glycolysis and are involved in regulating protein lactylation (26,27). In the present study, the knockdown of NEAT1 reduced the cellular lactate concentration and attenuated the levels of both pan lactylation (pan-lac) and H3K18 lactylation in HemECs (Fig. 3A-D). After LDHB expression was knocked down (Fig. 3E), the viability and colony formation ability of HemECs was decreased (Fig. 3F and G).
Activation of the Wnt/β-catenin pathway potentiates the invasion, migration, proliferation and epithelial-mesenchymal transition (EMT) of HemECs (28,29). Therefore, it was next evaluated whether lncRNA NEAT1 promotes the colony formation and viability of HemECs by regulating the Wnt/β-catenin signaling pathway. The results revealed that knocking down NEAT1 downregulated β-catenin at both the mRNA and protein levels, and that knocking down LDHB also reduced the mRNA and protein levels of β-catenin in HemECs (Fig. 3H-L). These data revealed that knocking down of lncRNA NEAT1 inhibited cellular protein lactylation and downregulated β-catenin in hemangioma cells.
Oxamate suppressed cellular protein and H3K18 lactylation in hemangioma cells and 2-DG slightly reduced the H3K18 lactylation level and inhibited cellular protein lactylation in hemangioma cells (Fig. 4A and B). Conversely, lactate addition markedly increased cellular protein lactylation and H3K18 lactylation in hemangioma cells (Fig. 4C). Both 2-DG and oxamate significantly impeded the viability and colony formation of hemangioma cells (Fig. 4D, E, G and H), and lactate significantly enhanced the viability and colony formation of hemangioma cells (Fig. 4F and I). These data indicated that inhibition of lactylation suppressed the viability and colony formation of hemangioma cells and that promotion of lactylation exhibited promotive effects.
Lactylation of promoters or enhancers is an indication of transcription activation (13); hence, whether NEAT1 regulates β-catenin by affecting lactylation of the CTNNB1 promoter was investigated. The results showed that both 2-DG and oxamate treatment reduced the H3K18 lactylation level of the CTNNB1 promoter; conversely, lactate addition increased the H3K18 lactylation level (Fig. 5A and B). Notably, knocking down NEAT1 or LDHB inhibited the H3K18 lactylation of the CTNNB1 promoter (Fig. 5C and D).
Both 2-DG and oxamate treatment reduced the CTNNB1 mRNA level; by contrast, lactate increased CTNNB1 mRNA expression (Fig. 5E and F). Additionally, western blotting revealed that 2-DG and oxamate treatment downregulated β-catenin at the protein level (Fig. 5G and H) and that lactate upregulated β-catenin at the protein level (Fig. 5I and J). These findings suggested that NEAT1 promoted H3K18 lactylation of the CTNNB1 promoter and that blocking lactylation downregulated β-catenin expression in hemangioma cells.
AARS1, AARS2, CREB binding lysine acetyltransferase (CBP), lysine acetyltransferase (KAT) 5, KAT8 and P300 are reported to positively regulate protein lactylation as lactyltransferases, and Sirtuin (SIRT) 1, SIRT2, SIRT3, Histone deacetylase (HDAC) 1, HDAC2 and HDAC3 have been found to negatively affect protein lactylation as delactyltransferases (30). Hence, which molecules regulate β-catenin in hemangioma cells were screened next. Notably, after separately knocking down every lactyltransferase (Fig. 6A) and delactyltransferase (Fig. 6C), it was found that the knockdown of AARS1 and P300 reduced the mRNA level of CTNNB1 (Fig. 6B). Additionally, knocking down HDAC1 also downregulated CTNNB1 at the mRNA level in hemangioma cells (Fig. 6D). The knockdown efficiency of AARS1 and P300 was further confirmed using western blotting (Fig. 6E and F). It was subsequently found that the knocking down of AARS1 and P300 in hemangioma cells also downregulated β-catenin at the protein level (Fig. 6G and H). Furthermore, knockdown of NEAT1 significantly decreased the mRNA and protein levels of AARS1 and P300 (Fig. 6I-K). Notably, blocking P300 using A-485 suppressed the viability and colony formation of HemECs (Fig. 7A-C), and A-485 treatment also decreased the mRNA and protein levels of β-catenin (Fig. 7D-F). These results indicated that NEAT1 regulates β-catenin in hemangioma cells possibly by positively affecting the lactyltransferases AARS1 and P300.
The status of β-catenin expression and H3K18 lactylation levels in IH tissues were further evaluated. The RT-qPCR and western blotting results revealed that both the mRNA and protein levels of β-catenin were greater in the IH tissues than in normal tissues (Fig. 7G and H). Additionally, western blotting indicated that the H3K18 lactylation level was greater in IH tissues than in normal tissues (Fig. 7I). In summary, the results revealed that lncRNA NEAT1, which is upregulated in hemangioma, binds with and upregulates LDHB, subsequently elevates the levels of cellular lactate and H3K18 lactylation, potentiates β-catenin transcription and ultimately enhances the proliferation of hemangioma cells (Fig. 7G).
Our previous study revealed that lncRNA NEAT1 is highly expressed in IH tissues and promotes the proliferation, migration and invasion of hemangioma cells by sponging miR-33a-5p and regulating the downstream HIF1α/NF-κB pathway (6). Nevertheless, the functional roles and detailed mechanisms of NEAT1 in hemangioma are still not fully understood.
The Wnt/β-catenin pathway plays critical promotional roles in hemangioma progression, and inactivation of the Wnt/β-catenin pathway has therapeutic effects on hemangioma (28,29). IL13RA2 was overexpressed in IH tissues, and exogenous expression of IL13RA2 potentiated hemangioma progression by interacting with and activating the β-catenin pathway (28). Renin has been shown to promote the proliferation of IH cells by activating the Wnt pathway (31). Additionally, blockade of the Wnt/β-catenin pathway by fucoidan has been shown to inhibit the proliferation and EMT of hemangioma cells (29). Dai et al (32) reported that luteolin suppresses IH progression by blocking the Wnt signaling pathway. Notably, the results of the present study indicated that lncRNA NEAT1, which is upregulated in IH tissues, upregulated β-catenin expression in an H3K18 lactylation-dependent manner.
Lactic acid was first discovered in 1780 and was formerly considered a byproduct of metabolism (33). However, at present, lactate is considered to act as both a metabolic substance and a signaling molecule (34,35). In 2019, Zhang et al (9) discovered protein lysine lactylation, a new posttranslational modification, and provided a new insight into the novel function of lactate. H3K18 lactylation is a critical type of histone lactylation that typically activates gene transcription (36) and is recognized as a biomarker for poor prognosis in epithelial ovarian cancer (37). In non-small cell lung carcinoma, H3K18 lactylation directly transcriptionally activates POM121 transmembrane nucleoporin and ultimately induces programmed cell death protein 1 expression and enhances immune escape (27). In ovarian cancer, lactate-induced H3K18 lactylation increases CCL18 expression (38). In pancreatic ductal adenocarcinoma (PDAC), H3K18 lactylation, which is elevated in PDAC, promotes the transcription of BUB1 mitotic checkpoint serine/threonine kinase B and TTK protein kinase and eventually causes tumorigenesis (39). H3K18 lactylation transcriptionally activates SOX9 and facilitates liver fibrosis development (40). Notably, the present study revealed that H3K18 lactylation transcriptionally activates β-catenin in hemangioma.
H3K18 lactylation is typically regulated by glycolysis, lactyltransferases and delactyltransferases. In breast cancer, potassium two pore domain channel subfamily K member 1 binds with LDHA and affects histone lysine lactylation (41). In PDAC, HDAC2 and P300 regulate histone lactylation as the eraser and writer of protein lactylation (39). The present study revealed that NEAT1 binds to LDHB and positively regulates LDHB expression in hemangioma cells. A previous study reported that LDHA was highly expressed in HemECs compared with human umbilical vein endothelial cells (42). Another study revealed that the protein expression of phosphofructokinase-1 (FPK-1) is greater in proliferating IHs than in involuting IHs, and that suppression of FPK-1 impedes the proliferation and migration of HemECs and reduces lactate production (43). However, the role of LDHB in hemangioma progression is still unclear.
The proliferative (first 6–12 months after birth), involuting (starting at ~13 months) and involuted (4–7 years of age) phases are the three stages of the self-limiting disease course in IH (44). Unraveling the mechanisms underlying IH development from proliferation to involution is crucial for developing a new therapeutic strategy for IH. The findings of the present study suggest that the levels of H3K18la and β-catenin are increased in proliferating IH tissues. Molecular analysis further revealed that H3K18la-driven transcriptional activation of CTNNB1 enhances the proliferation of hemangioma cells. Therefore, blocking this mechanism might be a benefit for IH therapy.
Taken together, the results of the present study revealed that lncRNA NEAT1, which is upregulated in hemangioma, binds with and stabilizes LDHB, subsequently elevates the levels of cellular lactate and H3K18 lactylation, potentiates CTNNB1 transcription and finally enhances the proliferation of hemangioma cells. Nevertheless, several limitations of the present study remain. First, when the candidate lactyltransferases or delactyltransferases that may regulate β-catenin expression were screened, the changes in expression were only examined at the mRNA level and not the protein level. Second, in addition to H3K18 lactylation, NEAT1 also regulates the lactylation of non-histone proteins; therefore, whether the lactylation of non-histone proteins is also involved in the NEAT1-affected hemangioma cell proliferation still needs to be investigated. Third, how NEAT1 regulates AARS1 and P300 requires further exploration.
Not applicable.
This study was funded by the Joint Special Funds for the Department of Science and Technology of Yunnan Province-Kunming Medical University (grant no. 202201AY070001-202).
The data generated in the present study may be requested from the corresponding author.
HS conceived and designed the research. LY performed the experiments and wrote the manuscript. NZ, XLZ, XJP, LX, YJP, LZ and JNW analyzed the data. All authors read and approved the final version of the manuscript. HS and LY confirm the authenticity of all the raw data.
The Ethics Committee of Kunming Children's Hospital (Kunming, China) approved this study (approval no. 2021-03-181-K01). Informed consent was obtained from the parents/legal guardians of each patient.
Not applicable.
The authors declare that they have no competing interests.
|
Liu C, Zhao ZL, Ji ZD, Jiang Y and Zheng J: MiR-187-3p enhances propranolol sensitivity of hemangioma stem cells. Cell Struct Funct. 44:41–50. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Acevedo LM and Cheresh DA: Suppressing NFAT increases VEGF signaling in hemangiomas. Cancer Cell. 14:429–430. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Q, Zhao C, Du Q, Cao Z and Pan J: Non-coding RNA in infantile hemangioma. Pediatr Res. 96:1594–1602. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Li Z, Cao Z, Li N, Wang L, Fu C, Huo R, Xu G, Tian C and Bi J: M2 macrophage-derived exosomal lncRNA MIR4435-2HG promotes progression of infantile hemangiomas by targeting HNRNPA1. Int J Nanomedicine. 18:5943–5960. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Li R, Liu Y, Liu J, Chen B, Ji Z, Xu A and Zhang T: CCL2 regulated by the CTBP1-AS2/miR-335-5p axis promotes hemangioma progression and angiogenesis. Immunopharmacol Immunotoxicol. 46:385–394. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Yu L, Shu H, Xing L, Lv MX, Li L, Xie YC, Zhang Z, Zhang L and Xie YY: Silencing long non-coding RNA NEAT1 suppresses the tumorigenesis of infantile hemangioma by competitively binding miR-33a-5p to stimulate HIF1α/NF-κB pathway. Mol Med Rep. 22:3358–3366. 2020.PubMed/NCBI | |
|
Yu X, Liu X, Wang R and Wang L: Long non-coding RNA NEAT1 promotes the progression of hemangioma via the miR-361-5p/VEGFA pathway. Biochem Biophys Res Commun. 512:825–831. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Peng K, Xia RP, Zhao F, Xiao Y, Ma TD, Li M, Feng Y and Zhou CG: ALKBH5 promotes the progression of infantile hemangioma through regulating the NEAT1/miR-378b/FOSL1 axis. Mol Cell Biochem. 477:1527–1540. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang D, Tang Z, Huang H, Zhou G, Cui C, Weng Y, Liu W, Kim S, Lee S, Perez-Neut M, et al: Metabolic regulation of gene expression by histone lactylation. Nature. 574:575–580. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Xiao SY, Zhang SH, Sun K, Huang Q and Hu C: Lactate and lactylation: Molecular insights into histone and non-histone lactylation in tumor progression, tumor immune microenvironment, and therapeutic strategies. Biomark Res. 13:1342025. View Article : Google Scholar : PubMed/NCBI | |
|
Fang N, Zhang N, Jiang X, Yan S, Wang Z, Gao Q, Xu M, Mu L, Li X, Chen J, et al: PFKM-Driven lactate overproduction promotes atrial fibrillation via triggering cardiac fibroblasts histone lactylation. Adv Sci (Weinh). 12:e009632025. View Article : Google Scholar : PubMed/NCBI | |
|
Gu JJ, Deng CC, An Q, Zhu DH, Xu XY, Fu ZZ, Zhou ZY, Rong Z and Yang B: Lactate promotes collagen expression, proliferation and migration through H3K18 lactylation-dependent stimulation of LTBP3/TGF-beta1 axis in keloid fibroblasts. J Invest Dermatol. July 7–2025.(Epub ahead of print). View Article : Google Scholar | |
|
Galle E, Wong CW, Ghosh A, Desgeorges T, Melrose K, Hinte LC, Castellano-Castillo D, Engl M, de Sousa JA, Ruiz-Ojeda FJ, et al: H3K18 lactylation marks tissue-specific active enhancers. Genome Biol. 23:2072022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang H, Xu M, Zhang T, Pan J, Li C, Pan B, Zhou L, Huang Y, Gao C, He M, et al: PYCR1 promotes liver cancer cell growth and metastasis by regulating IRS1 expression through lactylation modification. Clin Transl Med. 14:e700452024. View Article : Google Scholar : PubMed/NCBI | |
|
Khan ZA, Melero-Martin JM, Wu X, Paruchuri S, Boscolo E, Mulliken JB and Bischoff J: Endothelial progenitor cells from infantile hemangioma and umbilical cord blood display unique cellular responses to endostatin. Blood. 108:915–921. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Kong C, Li R, Wang X, Li L, Kang N, Zhen X, Dong Y and Yan G: Environmental aminomethylphosphonic acid (AMPA) exposure increased the risk of spontaneous abortion through lactate-induced JunB lactylation in trophoblast. Ecotoxicol Environ Saf. 302:1187432025. View Article : Google Scholar : PubMed/NCBI | |
|
Lu Y, Zhu J, Zhang Y, Li W, Xiong Y, Fan Y, Wu Y, Zhao J, Shang C, Liang H and Zhang W: Lactylation-Driven IGF2BP3-mediated serine metabolism reprogramming and RNA m6A-modification promotes lenvatinib resistance in HCC. Adv Sci (Weinh). 11:e24013992024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao SS, Liu J, Wu QC and Zhou XL: Lactate regulates pathological cardiac hypertrophy via histone lactylation modification. J Cell Mol Med. 28:e700222024. View Article : Google Scholar : PubMed/NCBI | |
|
Wei S, Zhang J, Zhao R, Shi R, An L, Yu Z, Zhang Q, Zhang J, Yao Y, Li H and Wang H: Histone lactylation promotes malignant progression by facilitating USP39 expression to target PI3K/AKT/HIF-1α signal pathway in endometrial carcinoma. Cell Death Discov. 10:1212024. View Article : Google Scholar : PubMed/NCBI | |
|
Lu G, Yi J, Gubas A, Wang YT, Wu Y, Ren Y, Wu M, Shi Y, Ouyang C, Tan HWS, et al: Suppression of autophagy during mitosis via CUL4-RING ubiquitin ligases-mediated WIPI2 polyubiquitination and proteasomal degradation. Autophagy. 15:1917–1934. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Whitworth CP, Aw WY, Doherty EL, Handler C, Ambekar Y, Sawhney A, Scarcelli G and Polacheck WJ: P300 modulates endothelial mechanotransduction of fluid shear stress. Cell Mol Bioeng. 17:507–523. 2024. 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 : PubMed/NCBI | |
|
Chen B, Deng S, Ge T, Ye M, Yu J, Lin S, Ma W and Songyang Z: Live cell imaging and proteomic profiling of endogenous NEAT1 lncRNA by CRISPR/Cas9-mediated knock-in. Protein Cell. 11:641–660. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Huang da W, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 4:44–57. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Huang da W, Sherman BT and Lempicki RA: Bioinformatics enrichment tools: Paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37:1–13. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Cai S, Xia Q, Duan D, Fu J, Wu Z, Yang Z and Yu C: Creatine kinase mitochondrial 2 promotes the growth and progression of colorectal cancer via enhancing Warburg effect through lactate dehydrogenase B. PeerJ. 12:e176722024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang C, Zhou L, Zhang M, Du Y, Li C, Ren H and Zheng L: H3K18 Lactylation potentiates immune escape of Non-small cell lung cancer. Cancer Res. 84:3589–3601. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Z, Ma T, Li J, Ren W and Zhang Z: IL13RA2 promotes progression of infantile haemangioma by activating glycolysis and the Wnt/β-catenin signaling pathway. Oncol Res. 32:1453–1465. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu Z, Luo J, Li L, Wang D, Xu Q, Teng J, Zhou J, Sun L, Yu N and Zuo D: Fucoidan suppresses proliferation and epithelial-mesenchymal transition process via Wnt/β-catenin signalling in hemangioma. Exp Dermatol. 33:e150272024. View Article : Google Scholar : PubMed/NCBI | |
|
Chen J, Zhao D, Wang Y, Liu M, Zhang Y, Feng T, Xiao C, Song H, Miao R, Xu L, et al: Lactylated apolipoprotein C-II induces immunotherapy resistance by promoting extracellular lipolysis. Adv Sci (Weinh). 11:e24063332024. View Article : Google Scholar : PubMed/NCBI | |
|
van Schaijik B, Tan ST, Marsh RW and Itinteang T: Expression of (pro)renin receptor and its effect on endothelial cell proliferation in infantile hemangioma. Pediatr Res. 86:202–207. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Dai Y, Zheng H, Liu Z, Wang Y and Hu W: The flavonoid luteolin suppresses infantile hemangioma by targeting FZD6 in the Wnt pathway. Invest New Drugs. 39:775–784. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Li X, Yang Y, Zhang B, Lin X, Fu X, An Y, Zou Y, Wang JX, Wang Z and Yu T: Lactate metabolism in human health and disease. Signal Transduct Target Ther. 7:3052022. View Article : Google Scholar : PubMed/NCBI | |
|
Yu H, Zhu T, Ma D, Cheng X, Wang S and Yao Y: The role of nonhistone lactylation in disease. Heliyon. 10:e362962024. View Article : Google Scholar : PubMed/NCBI | |
|
Brooks GA: Lactate as a fulcrum of metabolism. Redox Biol. 35:1014542020. View Article : Google Scholar : PubMed/NCBI | |
|
Raychaudhuri D, Singh P, Chakraborty B, Hennessey M, Tannir AJ, Byregowda S, Natarajan SM, Trujillo-Ocampo A, Im JS and Goswami S: Histone lactylation drives CD8+ T cell metabolism and function. Nat Immunol. 25:2140–2151. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Chao J, Chen GD, Huang ST, Gu H, Liu YY, Luo Y, Lin Z, Chen ZZ, Li X, Zhang B, et al: High histone H3K18 lactylation level is correlated with poor prognosis in epithelial ovarian cancer. Neoplasma. 71:319–332. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Sun J, Feng Q, He Y, Wang M and Wu Y: Lactate activates CCL18 expression via H3K18 lactylation in macrophages to promote tumorigenesis of ovarian cancer. Acta Biochim Biophys Sin (Shanghai). 56:1373–1386. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Li F, Si W, Xia L, Yin D, Wei T, Tao M, Cui X, Yang J, Hong T and Wei R: Positive feedback regulation between glycolysis and histone lactylation drives oncogenesis in pancreatic ductal adenocarcinoma. Mol Cancer. 23:902024. View Article : Google Scholar : PubMed/NCBI | |
|
Wu S, Li J and Zhan Y: H3K18 lactylation accelerates liver fibrosis progression through facilitating SOX9 transcription. Exp Cell Res. 440:1141352024. View Article : Google Scholar : PubMed/NCBI | |
|
Hou X, Ouyang J, Tang L, Wu P, Deng X, Yan Q, Shi L, Fan S, Fan C, Guo C, et al: KCNK1 promotes proliferation and metastasis of breast cancer cells by activating lactate dehydrogenase A (LDHA) and up-regulating H3K18 lactylation. PLoS Biol. 22:e30026662024. View Article : Google Scholar : PubMed/NCBI | |
|
Chen J, Wu D, Dong Z, Chen A and Liu S: The expression and role of glycolysis-associated molecules in infantile hemangioma. Life Sci. 259:1182152020. View Article : Google Scholar : PubMed/NCBI | |
|
Yang K, Zhang X, Chen L, Chen S and Ji Y: Microarray expression profile of mRNAs and long noncoding RNAs and the potential role of PFK-1 in infantile hemangioma. Cell Div. 16:12021. View Article : Google Scholar : PubMed/NCBI | |
|
Rodriguez Bandera AI, Sebaratnam DF, Wargon O and Wong LF: Infantile hemangioma. Part 1: Epidemiology, pathogenesis, clinical presentation and assessment. J Am Acad Dermatol. 85:1379–1392. 2021. View Article : Google Scholar : PubMed/NCBI |