1
|
Luo XR, Huang LZ, Yin J, Xiong ZM, Li WX, Liao C, Lin ML, Huang W, Zhang S. FSTL3 promotes colorectal cancer by activating the HIF1 pathway. Gene 2025; 954:149435. [PMID: 40154584 DOI: 10.1016/j.gene.2025.149435] [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: 01/06/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Follistatin-like 3 (FSTL3) is a glycoprotein known to promote tumor growth, invasion, and angiogenesis in various cancers. However, its role in Colorectal Cancer (CRC), particularly concerning the hypoxia-inducible factor 1α (HIF1α) signaling pathway, remains unclear. The HIF1α pathway is critical in CRC progression, enabling tumor cells to adapt to hypoxia through angiogenesis, Epithelial-Mesenchymal Transition (EMT), and metabolic reprogramming. Analysis of The Cancer Genome Atlas (TCGA) and GSE39582 datasets revealed that FSTL3 is significantly upregulated in CRC tissues and correlates with poor Overall Survival (OS), Progression-Free Survival (PFS), and aggressive features such as venous, lymphatic, and perineural invasion. In vitro experiments demonstrated that FSTL3 overexpression in HCT15 and HCT116 cells promoted proliferation, migration, and cell cycle progression, whereas knockdown in LOVO and Caco2 cells suppressed these processes and induced apoptosis. Transcriptome sequencing and western blot analysis indicated that FSTL3 activated the HIF1α pathway by upregulating HIF1α, ANGPT2, and HK3, which are key regulators of angiogenesis and glycolysis. Importantly, treatment with the HIF1α inhibitor KC7F2 reversed the oncogenic effects of FSTL3 overexpression both in vitro and in vivo. In xenograft and tail vein metastasis models, KC7F2 suppressed tumor growth, reduced pulmonary metastasis, and restored lung tissue integrity, further downregulating FSTL3 and HIF1α expression. These findings suggest that FSTL3 promotes CRC progression via the HIF1α pathway and highlight its potential as a prognostic biomarker and therapeutic target for CRC treatment.
Collapse
Affiliation(s)
- Xiang-Rong Luo
- Department of Colorectal & Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, PR China; The Central Hospital of Shaoyang, No. 36, Hongqi Road, Shaoyang City, Hunan Province 422000, PR China
| | - Li-Zhe Huang
- Department of Colorectal & Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, PR China
| | - Jie Yin
- The Central Hospital of Shaoyang, No. 36, Hongqi Road, Shaoyang City, Hunan Province 422000, PR China
| | - Zu-Ming Xiong
- Department of Colorectal & Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, PR China
| | - Wen-Xin Li
- Department of Colorectal & Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, PR China
| | - Cun Liao
- Department of Colorectal & Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, PR China
| | - Ming-Lin Lin
- Department of Colorectal & Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, PR China
| | - Wei Huang
- Department of Colorectal & Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, PR China
| | - Sen Zhang
- Department of Colorectal & Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, PR China.
| |
Collapse
|
2
|
Tang C, Xu N, Gao T. Molecular mechanisms of recombinant proteins PTTG1IP, ADAM12, PAPSS1, and MYO1B and their effects on wound repair induced by tendon exposure: Analysis of key genes. Int J Biol Macromol 2025; 305:141263. [PMID: 39978508 DOI: 10.1016/j.ijbiomac.2025.141263] [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: 12/30/2024] [Revised: 02/06/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
Tendon injury repair involves a variety of cellular and molecular pathways. The aim of this study was to explore the molecular mechanisms and interactions of recombinant proteins PTTG1IP, ADAM12, PAPSS1 and MYO1B in wound repair induced by tendon exposure. The study collected a dataset of gene expression related to tendon exposure from the GEO database. Differential gene expression analysis and Venn diagram analysis were used to identify the differentially expressed genes before and after tendon exposure injury, and potential diagnostic markers were screened. Appropriate statistical methods were used to analyze all data. The results show that the expression levels of PTTG1IP, ADAM12, PAPSS1 and MYO1B change dynamically after tendon exposure injury, and PTTG1IP, ADAM12, PAPSS1 and MYO1B play an important role in the wound repair process caused by tendon exposure. The change of its expression level and function is closely related to the wound healing process.
Collapse
Affiliation(s)
- Chao Tang
- Department of Orthopedics, Shanghai Eighth People's Hospital, Shanghai 200235, China
| | - Ning Xu
- Department of Orthopedics, Shanghai Eighth People's Hospital, Shanghai 200235, China
| | - Tiantian Gao
- Department of Orthopedics, Shanghai Eighth People's Hospital, Shanghai 200235, China.
| |
Collapse
|
3
|
Li N, Gong X, Cao Z, Li Z, Wang L, Huo R, Fu C. Myosin 1b Inhibits the Phenotype of HemECs to Affect the Progression of Infantile Hemangiomas. Clin Cosmet Investig Dermatol 2025; 18:989-998. [PMID: 40296866 PMCID: PMC12036681 DOI: 10.2147/ccid.s517750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 04/15/2025] [Indexed: 04/30/2025]
Abstract
Purpose To investigate the role of myosin 1b (Myo1b) in infantile hemangiomas (IHs). Patients and Methods The expression of Myo1b in IHs and normal skin tissues was evaluated using immunohistochemical techniques. Expression of Myo1b in hemangioma endothelial cells (HemECs) was detected using quantitative real-time polymerase chain reaction and Western blotting techniques. The effects of Myo1b on cell proliferation, invasion, tube formation and other biological characteristics were analyzed. A mouse model was established to observe the inhibitory effects of Myo1b in vivo. Protein expression of peroxisome proliferator-activated receptor-gamma (PPARγ) and glucose transporter 1 protein (Glut-1) in a Myo1b knockdown HemECs was detected using Western blot. Results Myo1b was highly expressed in the proliferative stage of IHs, and promoted proliferation, invasion, and angiogenesis in vitro. Mouse models further demonstrated that Myo1b inhibited proliferation and angiogenesis of IHs. Knockdown of Myo1b inhibited Glut-1 activity, promoted expression of PPARγ transcription factor. Conclusion Myo1b has a pro-tumor effect on IHs, knockdown of Myo1b facilitated progression of the proliferating to involution phase of IHs. Therefore, Myo1b is expected to serve as a new target for the treatment of IHs.
Collapse
Affiliation(s)
- Nanxi Li
- Department of Plastic and Aesthetic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
| | - Xiangan Gong
- Department of Plastic and Aesthetic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
| | - Zhongying Cao
- Department of Plastic and Aesthetic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
| | - Zhiyu Li
- Department of Plastic and Aesthetic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
| | - Luying Wang
- Department of Plastic and Aesthetic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
| | - Ran Huo
- Department of Plastic and Aesthetic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
| | - Cong Fu
- Department of Plastic and Aesthetic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
| |
Collapse
|
4
|
Du Y, Zhang X, Xu Y, Zhou Y, Xu Y. D-mannose suppresses the angiogenesis and progression of colorectal cancer. Acta Biochim Biophys Sin (Shanghai) 2025. [PMID: 40259886 DOI: 10.3724/abbs.2025043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025] Open
Abstract
Angiogenesis is an important factor influencing the development of solid tumors, and vascular endothelial growth factor receptor-2 (VEGFR2) is a central regulator of angiogenesis. Antibodies and inhibitors against VEGFR2 have been widely used in various malignancies. However, the regulatory mechanism of VEGFR2 has not been fully clarified. Here, we show that D-mannose can significantly inhibit angiogenesis and tumor growth by degrading VEGFR2. Specifically, D-mannose inactivates GSK3β by promoting the phosphorylation of GSK3β at Ser9, enhances the nuclear translocation of TFE3, and promotes lysosomal biogenesis, thereby increasing the lysosome-mediated degradation of VEGFR2. Thus, D-mannose significantly inhibits the proliferation, migration, and capillary formation of human umbilical vein endothelial cells (HUVECs) in vitro. Oral administration of D-mannose dramatically inhibits angiogenesis and tumor growth in mice. Our findings reveal a previously unrecognized anti-tumor mechanism of D-mannose by destabilizing VEGFR2 and provide a new strategy for the clinical treatment of colorectal cancer (CRC).
Collapse
Affiliation(s)
- Yu Du
- Nourse Centre for Pet Nutrition, Wuhu 241200, China
- Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| | - Xinchao Zhang
- Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| | - Yixin Xu
- Nourse Centre for Pet Nutrition, Wuhu 241200, China
- Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| | - Yuefan Zhou
- Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| | - Yanping Xu
- Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| |
Collapse
|
5
|
Kakanj P, Bonse M, Kshirsagar A, Gökmen A, Gaedke F, Sen A, Mollá B, Vogelsang E, Schauss A, Wodarz A, Pla-Martín D. Retromer promotes the lysosomal turnover of mtDNA. SCIENCE ADVANCES 2025; 11:eadr6415. [PMID: 40184468 PMCID: PMC11970507 DOI: 10.1126/sciadv.adr6415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 02/28/2025] [Indexed: 04/06/2025]
Abstract
Mitochondrial DNA (mtDNA) is exposed to multiple insults produced by normal cellular function. Upon mtDNA replication stress, the mitochondrial genome transfers to endosomes for degradation. Using proximity biotinylation, we found that mtDNA stress leads to the rewiring of the mitochondrial proximity proteome, increasing mitochondria's association with lysosomal and vesicle-related proteins. Among these, the retromer complex, particularly VPS35, plays a pivotal role by extracting mitochondrial components. The retromer promotes the formation of mitochondrial-derived vesicles shuttled to lysosomes. The mtDNA, however, directly shuttles to a recycling organelle in a BAX-dependent manner. Moreover, using a Drosophila model carrying a long deletion on the mtDNA (ΔmtDNA), we found that ΔmtDNA activates a specific transcriptome profile to counteract mitochondrial damage. Here, Vps35 expression restores mtDNA homoplasmy and alleviates associated defects. Hence, we demonstrate the existence of a previously unknown quality control mechanism for the mitochondrial matrix and the essential role of lysosomes in mtDNA turnover to relieve mtDNA damage.
Collapse
Affiliation(s)
- Parisa Kakanj
- Institute of Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Mari Bonse
- Institute of Physiology, University Clinics and Faculty of Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Arya Kshirsagar
- Institute of Biochemistry and Molecular Biology, University Clinics and Faculty of Medicine, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Aylin Gökmen
- Institute of Physiology, University Clinics and Faculty of Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Felix Gaedke
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Ayesha Sen
- Institute of Biochemistry and Molecular Biology, University Clinics and Faculty of Medicine, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | | | - Elisabeth Vogelsang
- Department of Molecular Cell Biology, Institute I for Anatomy. University Clinics and Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Astrid Schauss
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Andreas Wodarz
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Department of Molecular Cell Biology, Institute I for Anatomy. University Clinics and Faculty of Medicine, University of Cologne, Cologne, Germany
| | - David Pla-Martín
- Institute of Physiology, University Clinics and Faculty of Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Institute of Biochemistry and Molecular Biology, University Clinics and Faculty of Medicine, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
6
|
黄 晴, 张 文, 张 小, 王 炼, 宋 雪, 耿 志, 左 芦, 王 月, 李 静, 胡 建. [High MYO1B expression promotes proliferation, migration and invasion of gastric cancer cells and is associated with poor patient prognosis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2025; 45:622-631. [PMID: 40159977 PMCID: PMC11955900 DOI: 10.12122/j.issn.1673-4254.2025.03.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Indexed: 04/02/2025]
Abstract
OBJECTIVES To analyze MYO1B expression in gastric cancer, its association with long-term prognosis and its role in regulating biological behaviors of gastric cancer cells. METHODS We analyzed MYO1B expression in gastric cancer and its correlation with tumor grade, tumor stage, and patient survival using the Cancer Public Database. We also examined MYO1B expression with immunohistochemistry in gastric cancer and paired adjacent tissues from 105 patients receiving radical surgery and analyzed its correlation with cancer progression and postoperative 5-year survival of the patients. GO and KEGG enrichment analyses were used to explore the biological functions of MYO1B and the key pathways. In cultured gastric cancer cells, we examined the changes in cell proliferation, migration and invasion following MYO1B overexpression and knockdown. RESULTS Data from the Cancer Public Database showed that MYO1B expression was significantly higher in gastric cancer tissues than in normal tissues with strong correlations with tumor grade, stage and patient prognosis (P<0.05). In the clinical tissue samples, MYO1B was significantly overexpressed in gastric cancer tissues in positive correlation with Ki67 expression (r=0.689, P<0.05) and the parameters indicative of gastric cancer progression (CEA ≥5 μg/L, CA19-9 ≥37 kU/L, G3-4, T3-4, and N2-3) (P<0.05). Kaplan-Meier analysis and multivariate Cox regression analysis suggested that high MYO1B expression was associated with decreased postoperative 5-year survival and was an independent risk factor (HR: 3.522, 95%CI: 1.783-6.985, P<0.05). MYO1B expression level was a strong predictor of postoperative survival (cut-off value: 3.11, AUC: 0.753, P<0.05). GO and KEGG analyses suggested that MYO1B may regulate cell migration and the mTOR signaling pathway. In cultured gastric cancer cells, MYO1B overexpression significantly enhanced cell proliferation, migration, and invasion and promoted the phosphorylation of Akt and mTOR. CONCLUSIONS High MYO1B expression promotes proliferation, migration and invasion of gastric cancer cells and is correlated with poor patient prognosis.
Collapse
|
7
|
Zhu J, Jin Z, Wang J, Wu Z, Xu T, Tong G, Shen E, Fan J, Jiang C, Wang J, Li X, Cong W, Lin L. FGF21 ameliorates septic liver injury by restraining proinflammatory macrophages activation through the autophagy/HIF-1α axis. J Adv Res 2025; 69:477-494. [PMID: 38599281 PMCID: PMC11954821 DOI: 10.1016/j.jare.2024.04.004] [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: 02/02/2024] [Revised: 03/26/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024] Open
Abstract
INTRODUCTION Sepsis, a systemic immune syndrome caused by severe trauma or infection, poses a substantial threat to the health of patients worldwide. The progression of sepsis is heavily influenced by septic liver injury, which is triggered by infection and cytokine storms, and has a significant impact on the tolerance and prognosis of septic patients. The objective of our study is to elucidate the biological role and molecular mechanism of fibroblast growth factor 21 (FGF21) in the process of sepsis. OBJECTIVES This study was undertaken in an attempt to elucidate the function and molecular mechanism of FGF21 in therapy of sepsis. METHODS Serum concentrations of FGF21 were measured in sepsis patients and septic mice. Liver injury was compared between mice FGF21 knockout (KO) mice and wildtype (WT) mice. To assess the therapeutic potential, recombinant human FGF21 was administered to septic mice. Furthermore, the molecular mechanism of FGF21 was investigated in mice with myeloid-cell specific HIF-1α overexpression mice (LyzM-CreDIO-HIF-1α) and myeloid-cell specific Atg7 knockout mice (Atg7△mye). RESULTS Serum level of FGF21 was significantly increased in sepsis patients and septic mice. Through the use of recombinant human FGF21 (rhFGF21) and FGF21 KO mice, we found that FGF21 mitigated septic liver injury by inhibiting the initiation and propagation of inflammation. Treatment with rhFGF21 effectively suppressed the activation of proinflammatory macrophages by promoting macroautophagy/autophagy degradation of hypoxia-inducible factor-1α (HIF-1α). Importantly, the therapeutic effect of rhFGF21 against septic liver injury was nullified in LyzM-CreDIO-HIF-1α mice and Atg7△mye mice. CONCLUSIONS Our findings demonstrate that FGF21 considerably suppresses inflammation upon septic liver injury through the autophagy/ HIF-1α axis.
Collapse
Affiliation(s)
- Junjie Zhu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Zhouxiang Jin
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Jie Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Zhaohang Wu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Tianpeng Xu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Gaozan Tong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China
| | - Enzhao Shen
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China
| | - Junfu Fan
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Chunhui Jiang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Jiaqi Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Xiaokun Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China; Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Weitao Cong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China; Haihe Laboratory of Cell Ecosystem, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Li Lin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China.
| |
Collapse
|
8
|
Nie XH, Li TZ, Peng CM. ATP ion channel-type P2X purinergic receptors as a key regulatory molecule in breast cancer progression. Pathol Res Pract 2025; 267:155844. [PMID: 39965402 DOI: 10.1016/j.prp.2025.155844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/31/2025] [Accepted: 02/13/2025] [Indexed: 02/20/2025]
Abstract
Studies have confirmed that ATP ion channel P2X purinergic receptors play a key role in tumor growth and metastasis. Similarly, P2X purinergic receptors can be used as a favorable regulatory molecule of breast cancer cells to participate in the progression of breast cancer. There are abundant ATP and its cleavage products in breast cancer microenvironment, which can be used as natural activators of P2X purinergic receptors. P2X purinergic receptors play a role in regulating the growth and metastasis of breast cancer cells by mediating signal transduction, growth regulation and immune cell activity in microenvironment. However, the application of P2X purinergic receptors antagonist has the pharmacological characteristics of inhibiting the progression of breast cancer cells. Among P2X purinergic receptors, there is a close relationship between P2X7 receptor and breast cancer patients. P2X purinergic receptors can be used as a biological marker for breast cancer patients. In this paper, we discuss the functional role and regulatory molecular mechanism of P2X purinergic receptors in the progression of breast cancer. The pharmacological effects of P2X purinergic receptors selective antagonist on the growth, metastasis and invasion of breast cancer cells were further discussed. Therefore, P2X purinergic receptors can be used as a key regulatory molecule of breast cancer and a pharmacological target for potential therapy.
Collapse
Affiliation(s)
- Xin-Hua Nie
- Department of Gastroenterology, The second affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Teng-Zheng Li
- Department of Gastroenterology, The second affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Cheng-Ming Peng
- Department of Gastroenterology, The second affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, China.
| |
Collapse
|
9
|
Zhang Y, Wang Y, Yang Y, Sun C. Long noncoding RNA SNHG4 promotes glioma progression via regulating miR-367-3p/MYO1B axis in zebrafish xenografts. Hum Cell 2025; 38:53. [PMID: 39951205 PMCID: PMC11828807 DOI: 10.1007/s13577-025-01183-1] [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: 07/26/2024] [Accepted: 01/27/2025] [Indexed: 02/17/2025]
Abstract
Glioma is one of the most malignancy and prevalent tumor in the human central nervous system, which is associated with severe morbidity and high mortality. Numerous studies have explained the clear correlation between abnormal expression of lncRNA and progression of Glioma. LncRNA small nucleolar RNA host gene 4 (SNHG4) have been proved to play oncogenesis roles in various tumors, however, the underlying mechanism remains to be explored deeply. In this study, by analysis of the public database, we found that SNHG4 was upregulated in multiple cancer tissues, including glioma. Subsequently, the functional roles of SNHG4 were investigated, and we found that knockdown of SNHG4 remarkedly inhibited cell proliferation, migration. While, overexpression of SNHG4 enhanced these functions of glioma cells in vitro. Meanwhile, as the in vivo tool, zebrafish xenograft model was used to verify the functions of SNHG4 in glioma cells. Mechanically, we identified that SNHG4 or MYO1B could bind with miR-367-3p by the luciferase reporter assays. Furthermore, the rescue experiments showed that the inhibition of miR-367-3p or the expression of MYO1B partially rescue the inhibition effects of SNHG4 in glioma cells. Our study reveals that SNHG4 promotes the proliferation, migration of glioma via regulating miR-367-3p/MYO1B axis.
Collapse
Affiliation(s)
- Yueqing Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People's Republic of China
- Department of Neurosurgery, Huai'an Hospital of Huai'an City, Huai'an, 223200, People's Republic of China
| | - Yongjin Wang
- Department of Neurosurgery, Huai'an Hospital of Huai'an City, Huai'an, 223200, People's Republic of China
| | - Yang Yang
- Department of Neurosurgery, Huai'an Hospital of Huai'an City, Huai'an, 223200, People's Republic of China
| | - Chunming Sun
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People's Republic of China.
| |
Collapse
|
10
|
Tufail M, Jiang CH, Li N. Tumor dormancy and relapse: understanding the molecular mechanisms of cancer recurrence. Mil Med Res 2025; 12:7. [PMID: 39934876 PMCID: PMC11812268 DOI: 10.1186/s40779-025-00595-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 01/26/2025] [Indexed: 02/13/2025] Open
Abstract
Cancer recurrence, driven by the phenomenon of tumor dormancy, presents a formidable challenge in oncology. Dormant cancer cells have the ability to evade detection and treatment, leading to relapse. This review emphasizes the urgent need to comprehend tumor dormancy and its implications for cancer recurrence. Despite notable advancements, significant gaps remain in our understanding of the mechanisms underlying dormancy and the lack of reliable biomarkers for predicting relapse. This review provides a comprehensive analysis of the cellular, angiogenic, and immunological aspects of dormancy. It highlights the current therapeutic strategies targeting dormant cells, particularly combination therapies and immunotherapies, which hold promise in preventing relapse. By elucidating these mechanisms and proposing innovative research methodologies, this review aims to deepen our understanding of tumor dormancy, ultimately facilitating the development of more effective strategies for preventing cancer recurrence and improving patient outcomes.
Collapse
Affiliation(s)
- Muhammad Tufail
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Can-Hua Jiang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, 410008, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ning Li
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Institute of Oral Precancerous Lesions, Central South University, Changsha, 410008, China.
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
11
|
Panthong W, Pientong C, Nukpook T, Roytrakul S, Yingchutrakul Y, Teeramatwanich W, Aromseree S, Ekalaksananan T. Integrated Proteomics and Machine Learning Approach Reveals PYCR1 as a Novel Biomarker to Predict Prognosis of Sinonasal Squamous Cell Carcinoma. Int J Mol Sci 2024; 25:13234. [PMID: 39768999 PMCID: PMC11675701 DOI: 10.3390/ijms252413234] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
Sinonasal squamous cell carcinoma (SNSCC) is a rare tumor with a high 5-year mortality rate. However, proteomic technologies have not yet been utilized to identify SNSCC-associated proteins, which could be used as biomarkers. In this study, we aimed to discover a biomarker to predict SNSCC patients using proteomic analysis integrated with machine learning models. Support vector machine (SVM), logistic regression (LR), random forest (RF), and gradient boost (GB) classifiers were developed to predict SNSCC based on proteomic profiles of SNSCC compared with nasal polyps (NP) as control. Seventeen feature proteins were found in all models, indicating possible biomarkers for SNSCC. Analysis of gene expression across multiple cancer types and their associations with cancer stage and patient survival in the TCGA-HNSC dataset identified a PYCR1 and MYO1B gene that could be a potential tumor-associated marker. The expression of PYCR1 was confirmed by RT-qPCR in SNSCC tissues, and its high expression was associated with poor overall survival, indicating PYCR1 as a potential tumor-associated biomarker to predict the prognosis of SNSCC.
Collapse
Affiliation(s)
- Watcharapong Panthong
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand; (W.P.); (T.N.); (S.A.)
- HPV&EBV and Carcinogenesis (HEC) Research Group, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand
| | - Chamsai Pientong
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand; (W.P.); (T.N.); (S.A.)
- HPV&EBV and Carcinogenesis (HEC) Research Group, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand
| | - Thawaree Nukpook
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand; (W.P.); (T.N.); (S.A.)
- HPV&EBV and Carcinogenesis (HEC) Research Group, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand
| | - Sittiruk Roytrakul
- Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Khlong Luang, Pathum Thani 12120, Thailand; (S.R.); (Y.Y.)
| | - Yodying Yingchutrakul
- Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Khlong Luang, Pathum Thani 12120, Thailand; (S.R.); (Y.Y.)
| | - Watchareporn Teeramatwanich
- Department of Otorhinolaryngology, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand;
| | - Sirinart Aromseree
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand; (W.P.); (T.N.); (S.A.)
- HPV&EBV and Carcinogenesis (HEC) Research Group, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand
| | - Tipaya Ekalaksananan
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand; (W.P.); (T.N.); (S.A.)
- HPV&EBV and Carcinogenesis (HEC) Research Group, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand
| |
Collapse
|
12
|
Bao X, Zhuang T, Xu Y, Chen L, Feng L, Yao H. Essential role of the interaction between classical swine fever virus core protein and cellular MYO1B in viral components transport to exosomes and titer maintenance. Vet Microbiol 2024; 299:110315. [PMID: 39603013 DOI: 10.1016/j.vetmic.2024.110315] [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: 08/25/2024] [Revised: 11/15/2024] [Accepted: 11/23/2024] [Indexed: 11/29/2024]
Abstract
Classical swine fever (CSF) is a severe disease caused by the highly contagious CSFV. Our previous study demonstrated that exosomes from CSFV-infected cells contained significant amounts of viral genome and Core (C) protein and were infectious. To further elucidate the mechanisms underlying the formation of these infectious exosomes, we investigated the intracellular transport of the C protein in this study. We first identified the synchronized transport of the C protein and viral genome to exosomes, distinguishing it from other structural proteins. This suggests that the C protein likely binds to the viral genome and is transported to exosomes as a nucleocapsid. Subsequently, Co-IP and co-localization experiments confirmed the interaction between the host Myosin 1B (MYO1B) protein and the C protein. Key interaction sites were identified by generating and analyzing various C protein point mutations and truncation variants. The results indicate that specific sites at the N-terminus of the C protein significantly impact its interaction with MYO1B. Ultimately, by modulating MYO1B expression, we found that MYO1B knockdown significantly reduced the C protein and viral genome content in exosomes, leading to a decrease in CSFV titers. These findings underscore the critical role of MYO1B in facilitating the transport of the C protein and viral genome into exosomes during CSFV infection. Overall, this study explores the mechanism of infectious exosome formation during CSFV infection, revealing the critical role of the host MYO1B in this process. This is the first study to identify the involvement of MYO1B in viral infection, not only offering important insights into host-virus interactions but also identifying a new target for antiviral drug development.
Collapse
Affiliation(s)
- Xi Bao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China; Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing, Jiangsu 210014, China
| | - Tenhan Zhuang
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing, Jiangsu 210014, China
| | - Yue Xu
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing, Jiangsu 210014, China
| | - Li Chen
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing, Jiangsu 210014, China
| | - Lei Feng
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing, Jiangsu 210014, China; GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China; Laboratory for Food Quality and Safety State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing, Jiangsu, China; School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu, China.
| | - Huochun Yao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China.
| |
Collapse
|
13
|
Lv W, Yang F, Ge Z, Xin L, Zhang L, Zhai Y, Liu X, Guo Q, Mao X, Luo P, Zhang L, Jiang X, Zhang Y. Aberrant overexpression of myosin 1b in glioblastoma promotes angiogenesis via VEGF-myc-myosin 1b-Piezo1 axis. J Biol Chem 2024; 300:107807. [PMID: 39307302 PMCID: PMC11532902 DOI: 10.1016/j.jbc.2024.107807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 09/06/2024] [Accepted: 09/12/2024] [Indexed: 10/25/2024] Open
Abstract
Glioblastoma (GBM) is the most aggressive intracranial malignancy with poor prognosis. Enhanced angiogenesis is an essential hallmark of GBM, which demonstrates extensive microvascular proliferation and abnormal vasculature. Here, we uncovered the key role of myosin 1b in angiogenesis and vascular abnormality in GBM. Myosin 1b is upregulated in GBM endothelial cells (ECs) compared to the paired nonmalignant brain tissue. In our study, we found that myosin 1b promotes migration, proliferation, and angiogenesis of human/mouse brain ECs. We also found that myosin 1b expression in ECs can be regulated by vascular endothelial growth factor (VEGF) signaling through myc. Moreover, myosin 1b promotes angiogenesis via Piezo1 by enhancing Ca2+ influx, in which process VEGF can be the trigger. In conclusion, our results identified myosin 1b as a key mediator in promoting angiogenesis via mechanosensitive ion channel component 1 (Piezo1) and suggested that VEGF/myc signaling pathway could be responsible for driving the changes of myosin 1b overexpression in GBM ECs.
Collapse
Affiliation(s)
- Weifeng Lv
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Fan Yang
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden; Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neuro-injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Zhengmao Ge
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lele Xin
- China-Sweden International Joint Research Center for Brain Diseases, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Lingxue Zhang
- China-Sweden International Joint Research Center for Brain Diseases, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yaohong Zhai
- China-Sweden International Joint Research Center for Brain Diseases, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xian Liu
- China-Sweden International Joint Research Center for Brain Diseases, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Qingdong Guo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xinggang Mao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Peng Luo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lei Zhang
- China-Sweden International Joint Research Center for Brain Diseases, College of Life Sciences, Shaanxi Normal University, Xi'an, China; Jinfeng Laboratory, Chongqing, China
| | - Xiaofan Jiang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Yanyu Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
14
|
Gjorgoska M, Šturm L, Lanišnik Rižner T. Pre-receptor regulation of 11-oxyandrogens differs between normal and cancerous endometrium and across endometrial cancer grades and molecular subtypes. Front Endocrinol (Lausanne) 2024; 15:1404804. [PMID: 39205690 PMCID: PMC11349532 DOI: 10.3389/fendo.2024.1404804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Background Endometrial cancer (EC) is a prevalent gynecological malignancy globally, with a rising incidence trend. While classic androgens have been implicated with EC risk, the role of their 11-oxygenated metabolites is poorly understood. Here, we studied 11-oxyandrogen formation from steroid precursors in EC for the first time. Methods We performed in vitro studies on a panel of four EC cell lines of varying differentiation degree and molecular subtype and a control cell line of normal endometrium to assess 11-oxyandrogen formation from steroid precursors. We also characterized the transcriptomic effects of dihydrotestosterone (DHT) and 11-keto-DHT on Ishikawa and RL95-2. Key molecular players in 11-oxyandrogen metabolism and action were explored in endometrial tumors using public transcriptomic datasets. Results We discovered that within endometrial tumors, the formation of 11-oxyandrogens does not occur from classic androgen precursors. However, we observed distinct regulatory mechanisms at a pre-receptor level in normal endometrium compared to cancerous tissue, and between low- and high-grade tumors. Specifically, in vitro models of low-grade EC formed higher levels of bioactive 11-keto-testosterone from 11-oxyandrogen precursors compared to models of noncancerous endometrium and high-grade, TP53-mutated EC. Moreover, the potent androgen, DHT and its 11-keto homologue induced mild transcriptomic effects on androgen receptor (AR)-expressing EC model, Ishikawa. Finally, using public transcriptomic datasets, we found HSD11B2 and SRD5A2, coding for key enzymes in steroid metabolism, to be associated with better disease-specific survival, whereas higher intra-tumoral AR expression correlated with lower recurrence in TP53-wt tumors. Conclusions The intra-tumoral metabolism of 11-oxyandrogen precursors is characteristic for low-grade EC of non-TP53-alt molecular subtypes. Our findings support further exploration of circulating 11-oxyandrogens as prognostic biomarkers in EC.
Collapse
Affiliation(s)
| | | | - Tea Lanišnik Rižner
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
15
|
Fu J, Lin J, Dai Z, Lin B, Zhang J. Hypoxia-associated autophagy flux dysregulation in human cancers. Cancer Lett 2024; 590:216823. [PMID: 38521197 DOI: 10.1016/j.canlet.2024.216823] [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: 02/16/2024] [Revised: 03/09/2024] [Accepted: 03/18/2024] [Indexed: 03/25/2024]
Abstract
A general feature of cancer is hypoxia, determined as low oxygen levels. Low oxygen levels may cause cells to alter in ways that contribute to tumor growth and resistance to treatment. Hypoxia leads to variations in cancer cell metabolism, angiogenesis and metastasis. Furthermore, a hypoxic tumor microenvironment might induce immunosuppression. Moreover, hypoxia has the potential to impact cellular processes, such as autophagy. Autophagy refers to the catabolic process by which damaged organelles and toxic macromolecules are broken down. The abnormal activation of autophagy has been extensively recorded in human tumors and it serves as a regulator of cell growth, spread to other parts of the body, and resistance to treatment. There is a correlation between hypoxia and autophagy in human malignancies. Hypoxia can regulate the activity of AMPK, mTOR, Beclin-1, and ATGs to govern autophagy in human malignancies. Furthermore, HIF-1α, serving as an indicator of low oxygen levels, controls the process of autophagy. Hypoxia-induced autophagy has a crucial role in regulating the growth, spread, and resistance to treatment in human malignancies. Hypoxia-induced regulation of autophagy can impact other mechanisms of cell death, such as apoptosis. Chemoresistance and radioresistance have become significant challenges in recent years. Hypoxia-mediated autophagy plays a crucial role in determining the response to these therapeutic treatments.
Collapse
Affiliation(s)
- Jiding Fu
- Department of Intensive Care Unit, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China
| | - Jie Lin
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China
| | - Zili Dai
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China
| | - Baisheng Lin
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China
| | - Jian Zhang
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China.
| |
Collapse
|
16
|
Magar AG, Morya VK, Kwak MK, Oh JU, Noh KC. A Molecular Perspective on HIF-1α and Angiogenic Stimulator Networks and Their Role in Solid Tumors: An Update. Int J Mol Sci 2024; 25:3313. [PMID: 38542288 PMCID: PMC10970012 DOI: 10.3390/ijms25063313] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/10/2024] [Accepted: 03/11/2024] [Indexed: 01/02/2025] Open
Abstract
Hypoxia-inducible factor-1α (HIF-1α) is a major transcriptional factor, which plays an important role in cellular reprogramming processes under hypoxic conditions, which facilitate solid tumors' progression. HIF-1α is directly involved in the regulation of the angiogenesis, metabolic reprogramming, and extracellular matrix remodeling of the tumor microenvironment. Therefore, an in-depth study on the role of HIF-1α in solid tumor malignancies is required to develop novel anti-cancer therapeutics. HIF-1α also plays a critical role in regulating growth factors, such as the vascular endothelial growth factor, fibroblast growth factor, and platelet-derived growth factor, in a network manner. Additionally, it plays a significant role in tumor progression and chemotherapy resistance by regulating a variety of angiogenic factors, including angiopoietin 1 and angiopoietin 2, matrix metalloproteinase, and erythropoietin, along with energy pathways. Therefore, this review attempts to provide comprehensive insight into the role of HIF-1α in the energy and angiogenesis pathways of solid tumors.
Collapse
Affiliation(s)
- Anuja Gajanan Magar
- Hallym University Dongtan Sacred Heart Hospital, Dongtan 18450, Republic of Korea
- School of Medicine, Hallym University, Chuncheon-si 24252, Republic of Korea
| | - Vivek Kumar Morya
- Hallym University Dongtan Sacred Heart Hospital, Dongtan 18450, Republic of Korea
| | - Mi Kyung Kwak
- Hallym University Dongtan Sacred Heart Hospital, Dongtan 18450, Republic of Korea
| | - Ji Ung Oh
- Hallym University Dongtan Sacred Heart Hospital, Dongtan 18450, Republic of Korea
| | - Kyu Cheol Noh
- Hallym University Dongtan Sacred Heart Hospital, Dongtan 18450, Republic of Korea
| |
Collapse
|
17
|
Hashemi M, Esbati N, Rashidi M, Gholami S, Raesi R, Bidoki SS, Goharrizi MASB, Motlagh YSM, Khorrami R, Tavakolpournegari A, Nabavi N, Zou R, Mohammadnahal L, Entezari M, Taheriazam A, Hushmandi K. Biological landscape and nanostructural view in development and reversal of oxaliplatin resistance in colorectal cancer. Transl Oncol 2024; 40:101846. [PMID: 38042134 PMCID: PMC10716031 DOI: 10.1016/j.tranon.2023.101846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/09/2023] [Accepted: 11/20/2023] [Indexed: 12/04/2023] Open
Abstract
The treatment of cancer patients has been mainly followed using chemotherapy and it is a gold standard in improving prognosis and survival rate of patients. Oxaliplatin (OXA) is a third-platinum anti-cancer agent that reduces DNA synthesis in cancer cells to interfere with their growth and cell cycle progression. In spite of promising results of using OXA in cancer chemotherapy, the process of drug resistance has made some challenges. OXA is commonly applied in treatment of colorectal cancer (CRC) as a malignancy of gastrointestinal tract and when CRC cells increase their proliferation and metastasis, they can obtain resistance to OXA chemotherapy. A number of molecular factors such as CHK2, SIRT1, c-Myc, LATS2 and FOXC1 have been considered as regulators of OXA response in CRC cells. The non-coding RNAs are able to function as master regulator of other molecular pathways in modulating OXA resistance. There is a close association between molecular mechanisms such as apoptosis, autophagy, glycolysis and EMT with OXA resistance, so that apoptosis inhibition, pro-survival autophagy induction and stimulation of EMT and glycolysis can induce OXA resistance in CRC cells. A number of anti-tumor compounds including astragaloside IV, resveratrol and nobiletin are able to enhance OXA sensitivity in CRC cells. Nanoparticles for increasing potential of OXA in CRC suppression and reversing OXA resistance have been employed in cancer chemotherapy. These subjects are covered in this review article to shed light on molecular factors resulting in OXA resistance.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Nastaran Esbati
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sadaf Gholami
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Rasoul Raesi
- Department of Health Services Management, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Shahabadin Bidoki
- Faculty of medicine, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | | | - Ramin Khorrami
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Alireza Tavakolpournegari
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Rongjun Zou
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China
| | - Leila Mohammadnahal
- Department of Health Services Management, School of Health, Tehran University of Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
18
|
Webster NJG, Kumar D, Wu P. Dysregulation of RNA splicing in early non-alcoholic fatty liver disease through hepatocellular carcinoma. Sci Rep 2024; 14:2500. [PMID: 38291075 PMCID: PMC10828381 DOI: 10.1038/s41598-024-52237-7] [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: 09/06/2023] [Accepted: 01/16/2024] [Indexed: 02/01/2024] Open
Abstract
While changes in RNA splicing have been extensively studied in hepatocellular carcinoma (HCC), no studies have systematically investigated changes in RNA splicing during earlier liver disease. Mouse studies have shown that disruption of RNA splicing can trigger liver disease and we have shown that the splicing factor SRSF3 is decreased in the diseased human liver, so we profiled RNA splicing in liver samples from twenty-nine individuals with no-history of liver disease or varying degrees of non-alcoholic fatty liver disease (NAFLD). We compared our results with three publicly available transcriptome datasets that we re-analyzed for splicing events (SEs). We found many changes in SEs occurred during early liver disease, with fewer events occurring with the onset of inflammation and fibrosis. Many of these early SEs were enriched for SRSF3-dependent events and were associated with SRSF3 binding sites. Mapping the early and late changes to gene ontologies and pathways showed that the genes harboring these early SEs were involved in normal liver metabolism, whereas those harboring late SEs were involved in inflammation, fibrosis and proliferation. We compared the SEs with HCC data from the TCGA and observed that many of these early disease SEs are found in HCC samples and, furthermore, are correlated with disease survival. Changes in splicing factor expression are also observed, which may be associated with distinct subsets of the SEs. The maintenance of these SEs through the multi-year oncogenic process suggests that they may be causative. Understanding the role of these splice variants in metabolic liver disease progression may shed light on the triggers of liver disease progression and the pathogenesis of HCC.
Collapse
Affiliation(s)
- Nicholas J G Webster
- Jennifer Moreno VA Medical Center, San Diego, CA, 92161, USA.
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, CA, 92093, USA.
- Moores Cancer Center, University of California, San Diego, CA, 92093, USA.
| | - Deepak Kumar
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, CA, 92093, USA
| | - Panyisha Wu
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, CA, 92093, USA
| |
Collapse
|
19
|
Zhou S, Huang H, Zheng Z, Zheng K, Xie L. MOGS promotes stemness acquisition and invasion via enhancing NOTCH1-glycosylation dependent NOTCH pathway in colorectal cancer. Am J Cancer Res 2023; 13:5996-6010. [PMID: 38187061 PMCID: PMC10767340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/22/2023] [Indexed: 01/09/2024] Open
Abstract
Colorectal cancer (CRC) ranks as the third most prevalent cancer globally, and about half of CRC patients eventually succumb to tumor metastasis. Despite this, treatment options for metastatic colon cancer remain severely limited, reflected by a 12% 5-year overall survival rate. Increasing evidence suggests that cancer stem cells (CSCs) are pivotal in driving CRC metastasis. Our study found a significant upregulation of MOGS in metastatic colorectal cancer, with high MOGS expression inversely correlating with patient prognosis. Additionally, MOGS enhances the NOTCH pathway, thus promoting stemness in CRC cells, both in vitro and in vivo. Mechanistically, MOGS may facilitate the maturation of NOTCH1 protein by promoting NOTCH1 glycosylation. Correspondingly, silencing MOGS markedly reduced invasion and stemness of CRC cells in vivo. In summary, our findings highlight the critical role of MOGS in fostering stemness and activating the NOTCH pathway in colorectal cancer cells. Disrupting the function of the MOGS/NOTCH could represent a feasible therapeutic strategy for CRC management.
Collapse
Affiliation(s)
- Shihai Zhou
- Department of Tumor Surgery, Zhongshan City People’s HospitalZhongshan 528403, Guangdong, China
| | - Hongyun Huang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijing 100730, China
| | - Zheng Zheng
- Department of Tumor Surgery, Zhongshan City People’s HospitalZhongshan 528403, Guangdong, China
| | - Kehong Zheng
- Department of General Surgery, Zhujiang Hospital, Southern Medical UniversityGuangzhou 510280, Guangdong, China
| | - Lang Xie
- Department of General Surgery, Zhujiang Hospital, Southern Medical UniversityGuangzhou 510280, Guangdong, China
| |
Collapse
|
20
|
Bajracharya R, Baral KC, Lee SH, Song JG, Han HK. Organometallic Phyllosilicate-Gold Nanocomplex: An Effective Oral Delivery System of Methotrexate for Enhanced in vivo Efficacy Against Colorectal Cancer. Int J Nanomedicine 2023; 18:7257-7266. [PMID: 38076733 PMCID: PMC10710251 DOI: 10.2147/ijn.s437860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Purpose Oral administration, although convenient and preferred for treating colorectal cancer (CRC), faces challenges due to limited CRC-related intestinal positioning and a dense mucus barrier. In the present study, a gold-nanoparticle decorated-organometallic phyllosilicate nanocomposite (AC-Au), with a pH-dependent surface coating, was employed for more effective oral delivery of anticancer drugs to treat CRC. Methods The organometallic AC-Au was synthesized using the in-situ sol-gel method. Subsequently, methotrexate (MTX) was loaded into AC-Au, and the complex (AC-Au/MTX) was surface-coated with poly (methacrylic acid-co-methyl methacrylate) (1:2), a pH-dependent polymer (E/AC-Au /MTX). The in vitro characteristics of nanoparticles were examined using various analytical methods. In vivo efficacy studies were also conducted using an HCT-116 orthotopic colorectal cancer model. Results AC-Au emerged as a spherical nanoparticle with a mean size of 26.5 ± 0.43 nm, displaying a positive charge over the pH range of 2-10. Both the uncoated and coated drug-loaded nanocomplexes (AC-Au/MTX and E/AC-Au/MTX) were fabricated with high entrapment efficiency (> 80%). Various analyses, including ultraviolet-visible spectroscopy, X-ray powder diffraction, transmission electron microscopy, and energy dispersive X-ray spectroscopy, confirmed the formation of the nanocomplexes. While AC-Au/MTX achieved rapid and extensive drug release at the pH range of 1.2-7.4, E/AC-Au/MTX exhibited pH-dependent drug release, with approximately 23% at pH 1.2 and 74% at pH 7.4. Relative to free MTX, the AC-Au-based nanocomplex significantly enhanced the cytotoxicity of MTX in HCT-116 cells. Furthermore, orally administered E/AC-Au/MTX significantly improved the anti-tumor activity of MTX in an HCT-116 orthotopic colorectal cancer model, resulting in approximately 60% suppression of tumor mass compared with the positive control. Conclusion The organometallic AC-Au nanocomplex coated with a pH-dependent polymer has the potential to be an effective colonic drug delivery system of MTX, enhancing in vivo efficacy against colorectal cancer.
Collapse
Affiliation(s)
| | | | - Sang Hoon Lee
- College of Pharmacy, Dongguk University-Seoul, Goyang, Korea
| | - Jae Geun Song
- College of Pharmacy, Dongguk University-Seoul, Goyang, Korea
| | - Hyo-Kyung Han
- College of Pharmacy, Dongguk University-Seoul, Goyang, Korea
| |
Collapse
|