1
|
Kong M, Zhai Y, Liu H, Zhang S, Chen S, Li W, Ma X, Ji Y. Insights into the mechanisms of angiogenesis in hepatoblastoma. Front Cell Dev Biol 2025; 13:1535339. [PMID: 40438141 PMCID: PMC12116456 DOI: 10.3389/fcell.2025.1535339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 05/02/2025] [Indexed: 06/01/2025] Open
Abstract
Hepatoblastoma (HB), the most common pediatric liver malignancy, is characterized by aggressive growth and metastasis driven by complex angiogenic mechanisms. This review elucidates the pivotal role of angiogenesis in HB progression, emphasizing metabolic reprogramming, tumor microenvironment (TME) dynamics, and oncogenic signalling pathways. The Warburg effect in HB cells fosters a hypoxic microenvironment, stabilizing hypoxia-inducible factor-1α (HIF-1α) and upregulating vascular endothelial growth factor (VEGF), which synergistically enhances angiogenesis. Key pathways such as the Wnt/β-catenin, VEGF, PI3K/AKT, and JAK2/STAT3 pathways are central to endothelial cell proliferation, migration, and vascular maturation, whereas interactions with tumor-associated macrophages (TAMs) and pericytes further remodel the TME to support neovascularization. Long noncoding RNAs and glycolytic enzymes have emerged as critical regulators of angiogenesis, linking metabolic activity with vascular expansion. Anti-angiogenic therapies, including VEGF inhibitors and metabolic pathway-targeting agents, show preclinical promise but face challenges such as resistance and off-target effects. Future directions advocate for dual-target strategies, spatial multiomics technologies to map metabolic-angiogenic crosstalk, and personalized approaches leveraging biomarkers for risk stratification. This synthesis underscores the need for interdisciplinary collaboration to translate mechanistic insights into durable therapies, ultimately improving outcomes for HB patients.
Collapse
Affiliation(s)
- Meng Kong
- Department of Pediatric Surgery, Children’s Hospital Affiliated to Shandong University, Jinan, China
- Department of Pediatric Surgery, Jinan Children’s Hospital, Jinan, China
| | - Yunpeng Zhai
- Department of Pediatric Surgery, Children’s Hospital Affiliated to Shandong University, Jinan, China
- Department of Pediatric Surgery, Jinan Children’s Hospital, Jinan, China
| | - Hongzhen Liu
- Department of Pediatric Surgery, Children’s Hospital Affiliated to Shandong University, Jinan, China
- Department of Pediatric Surgery, Jinan Children’s Hospital, Jinan, China
| | - Shisong Zhang
- Department of Pediatric Surgery, Children’s Hospital Affiliated to Shandong University, Jinan, China
- Department of Pediatric Surgery, Jinan Children’s Hospital, Jinan, China
| | - Shuai Chen
- Department of Pediatric Surgery, Children’s Hospital Affiliated to Shandong University, Jinan, China
- Department of Pediatric Surgery, Jinan Children’s Hospital, Jinan, China
| | - Wenfei Li
- School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiang Ma
- Department of Respiratory Disease, Children’s Hospital Affiliated to Shandong University, Jinan, China
- Jinan Key Laboratory of Pediatric Respiratory Diseases, Jinan Children’s Hospital, Jinan, China
| | - Yi Ji
- Division of Oncology, Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Zhou M, Liu Y, Li C, Yang X, Ji C, Li W, Song M, Yang Z, Liu G, Liang X, Liang J, Zhang B, Wang L. INSL3 promotes macrophage polarization to an immunosuppressive phenotype via the cAMP downstream signaling pathway and Akt/mTOR pathway. Int Immunopharmacol 2025; 154:114540. [PMID: 40168802 DOI: 10.1016/j.intimp.2025.114540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 04/03/2025]
Abstract
Insulin-like peptide 3 (INSL3) is a small peptide hormone produced almost exclusively by testicular Leydig cells in males and thus serves as an essential biomarker of the maturation and functionality of these cells. Accumulated evidence suggests that INSL3 is a crucial factor affecting testicular descent during fetal development by regulating the growth of the gubernaculum. However, the physiological roles of INSL3 in adults remain unclear. Here, we reported that relaxin family peptide 2 (RXFP2), the receptor of INSL3, is expressed on macrophages, and treatment with INSL3 can promote M2 macrophage polarization via the Akt/mTOR/S6K and PKA/CREB pathways. In addition, INSL3 can inhibit macrophage phagocytosis and promote their migration via the Epac and PKA signaling pathways, respectively. These findings reveal a new role for INSL3 in regulating macrophage function and shed new light on our understanding of the role of INSL3 in adulthood.
Collapse
Affiliation(s)
- Mengting Zhou
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yi Liu
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Cuiping Li
- Laboratory medicine department, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xizhong Yang
- Department of Spine Surgery, Qingdao Haici Medical Group, Qingdao, China
| | - Cuijie Ji
- Department of Spine Surgery, Qingdao Haici Medical Group, Qingdao, China
| | - Wei Li
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Meiying Song
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Zijie Yang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Guixian Liu
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xinping Liang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jie Liang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Bei Zhang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, China.
| | - Luoyang Wang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, China; Department of Spine Surgery, Qingdao Haici Medical Group, Qingdao, China.
| |
Collapse
|
3
|
Zhuo W, Zhang M, Tan J, Gao Y, Wang Y, Wang N, Ma J, Zhang J, Liu Z, Lv H, Liu Y. Lysine lactylation analysis of proteins in the heart of the Kawasaki disease mouse model. Front Cell Dev Biol 2025; 13:1550220. [PMID: 40114965 PMCID: PMC11922914 DOI: 10.3389/fcell.2025.1550220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/14/2025] [Indexed: 03/22/2025] Open
Abstract
Introduction Kawasaki disease (KD) is a medium-vessel vasculitis predominantly affecting children under 5 years of age and may involve the coronary arteries. Methods A mouse KD model was induced by Candida albicans cell wall extracts (CAWS), cardiac tissues were analyzed through integrated lactylomic and proteomic profiling. The lysine lactylation (Kla) results were normalized to the proteomic data. Results Elevated serum lactate and lactate dehydrogenase (LDH) levels were observed in KD patients. Given lactate's role as a substrate for Kla, this study investigated Kla modifications in KD. Proteomic analysis identified 150 upregulated proteins and 18 downregulated proteins, with 38.1% located in the cytoplasm and significant enrichment in immune-related pathways. After normalization, 41 sites in 37 proteins were found to be upregulated in the Kla data, with no downregulated sites. Approximately 67.57% of the altered proteins were localized in the mitochondria. Bioinformatics analysis indicated alterations in aerobic respiration, energy production and conversion, and key immune- and metabolism-related pathways. Discussion This study enhances the understanding of Kla modifications in the development of KD and may inform targeted therapies for its prevention and improved prognosis.
Collapse
Affiliation(s)
- Wenyu Zhuo
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Mingyang Zhang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jiajia Tan
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yang Gao
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Pediatrics, The First People's Hospital of Lianyungang, Xuzhou Medical University Affiliated Hospital of Lianyungang (Lianyungang Clinical College of Nanjing Medical University), Lianyungang, China
| | - Yan Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Cardiology, The Affiliated Xuzhou Children's Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Nana Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jin Ma
- Department of Pharmacy, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jiaying Zhang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhiheng Liu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Haitao Lv
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ying Liu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
4
|
He L, Chen Q, Wu X. Tumour-derived exosomal miR-205 promotes ovarian cancer cell progression through M2 macrophage polarization via the PI3K/Akt/mTOR pathway. J Ovarian Res 2025; 18:28. [PMID: 39955607 PMCID: PMC11829414 DOI: 10.1186/s13048-025-01616-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/01/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Tumour-associated macrophages (TAMs) are the most abundant immune cells in the tumour environment and are considered similar to M2 macrophages, which facilitate cancer progression. Exosomes, as important mediators of the cross-talk between tumour cells and tumour-associated macrophages, can facilitate the development and metastasis of ovarian cancer by mediating M2 macrophage polarization. However, the exact mechanisms underlying the communication between ovarian cancer (OC) cells and tumour-associated macrophages during OC progression remain unclear. RESULTS Here, we demonstrated that high expression of miR-205 was associated with M2 macrophage infiltration, which affected the prognosis of OC patients. Importantly, tumour-derived miR-205 could be transported from OC cells to macrophages via exosomes and promote cancer cell invasion and metastasis by inducing M2-like macrophage polarization. Animal experiments further confirmed that exosomal miR-205-induced M2 macrophages accelerated OC progression in vivo. Mechanistically, miR-205 downregulated PTEN, activating the PI3K/AKT/mTOR signalling pathway, which is critical for M2 polarization. CONCLUSIONS These results reveal that exosomal miR-205 plays a pivotal role in macrophage polarization within the OC microenvironment, highlighting its potential as a therapeutic target for OC treatment. This study not only enhances our understanding of the interactions between tumour and immune cells but also opens new avenues for targeted therapies against exosomal miR-205 in ovarian cancer.
Collapse
Affiliation(s)
- Liuqing He
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Quan Chen
- Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Xiaoying Wu
- Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China.
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China.
| |
Collapse
|