1
|
Hu H, Tai J, Zhang R, Zhang H. Study on the anticancer function and mechanism of cathelicidin-DM in liver cancer. Genomics 2025; 117:111049. [PMID: 40288464 DOI: 10.1016/j.ygeno.2025.111049] [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: 10/23/2024] [Revised: 04/17/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Previous studies have shown that bioactive molecules secreted by toad skin possess anticancer properties. Cathelicidin-DM (C-DM) is a bioactive peptide secreted by toad skin, but its effects on tumor cells and the underlying molecular mechanisms remain unknown. METHODS The impact of varying amphibian peptides on the viability of various tumor cells was determined by CCK-8. The influence of C-DM on liver cancer (LC) cell function was investigated using colony formation, flow cytometry, and Transwell. The anti-tumor activity of C-DM was evaluated by xenograft models. RNA-seq was done to confirm differentially expressed genes (DEGs), which were subject to GSEA. Cellular experiments were performed to verify the molecular regulatory mechanism of C-DM in LC. RESULTS C-DM (100 μg/mL) had a significant repressive impact on the proliferation, migration, invasion, and cell cycle progression of HepG2 and Hep3B cells, and facilitated apoptosis. In vivo experiments validated the anti-tumor impact of C-DM, while its toxicity was low. DEG DHRS3 was enriched in the JAK-STAT pathway. Overexpression of DHRS3 fostered the malignant phenotype of LC cells and activated the JAK-STAT pathway. However, the addition of C-DM weakened the oncogenic properties of DHRS3 and repressed the JAK-STAT pathway. CONCLUSION C-DM exerted an anti-LC effect by downregulating DHRS3 and mediating the JAK-STAT pathway, indicating that C-DM may be a promising candidate drug for LC treatment.
Collapse
Affiliation(s)
- Huang Hu
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China; Jinhua University of Vocational Technology, Agriculture College, Jinhua 321016, China
| | - Jingjing Tai
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China; Jinhua University of Vocational Technology, Agriculture College, Jinhua 321016, China
| | - Ruiyun Zhang
- Shang Cheng Center for Disease Control and Prevention, Hangzhou 310009, China
| | - Hong Zhang
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China.
| |
Collapse
|
2
|
WANG JIAHUI, GE HONGCHENG, YU ZHENGYUAN, WU LINGZHI. Non-coding RNAs as potential mediators of resistance to lung cancer immunotherapy and chemotherapy. Oncol Res 2025; 33:1033-1054. [PMID: 40296912 PMCID: PMC12034021 DOI: 10.32604/or.2024.058256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 11/18/2024] [Indexed: 04/30/2025] Open
Abstract
Lung cancer is a common cause of cancer-related death globally. The majority of lung cancer patients initially benefit from chemotherapy and immunotherapy. However, as the treatment cycle progresses and the disease evolves, the emergence of acquired resistance leads to treatment failure. Many researches have shown that non-coding RNAs (ncRNAs) not only influence lung cancer progression but also act as potential mediators of immunotherapy and chemotherapy resistance in lung cancer, mediating drug resistance by regulating multiple targets and pathways. In addition, the regulation of immune response by ncRNAs is dualistic, forming a microenvironment for inhibits/promotes immune escape through changes in the expression of immune checkpoints. The aim of this review is to understand the effects of ncRNAs on the occurrence and development of lung cancer, focusing on the role of ncRNAs in regulating drug resistance of lung cancer.
Collapse
Affiliation(s)
- JIAHUI WANG
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - HONGCHENG GE
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310018, China
| | - ZHENGYUAN YU
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - LINGZHI WU
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| |
Collapse
|
3
|
Zhou Y, Sun R, Zhang ZW, He XY, Li L, Zhang CJ, Liu Y, Yu HT. Proliferation Inhibited by Genipin in Human Leukemia K562 Cells: Involvement of Uncoupling Protein 2 in Mitochondrial Damage. World J Oncol 2025; 16:83-94. [PMID: 39850526 PMCID: PMC11750759 DOI: 10.14740/wjon1975] [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: 09/21/2024] [Accepted: 12/16/2024] [Indexed: 01/25/2025] Open
Abstract
Background Uncoupling protein 2 (UCP2) is essential for maintaining redox homeostasis and regulating energy metabolism. Abnormal expression of UCP2 has been associated with various tumors, including leukemia. Genipin (GEN), a specific inhibitor of UCP2, has a long history of use in traditional Chinese medicine. However, the precise role and underlying mechanisms of UCP2 in the inhibition of leukemia cells by GEN remain inadequately understood. This study focuses on the expression levels of UCP2 in myeloid leukemia (ML) and investigates the effects of GEN on the proliferation, mitochondrial function, and energy metabolism of the chronic myeloid leukemia (CML) cell line K562. Methods The expression of UCP2 in clinical samples and cell lines (HL-60, U937, and K562) was confirmed using real-time quantitative polymerase chain reaction (qPCR) and western blot. The effects of GEN on K562 cell viability, morphology, and apoptosis were assessed through a cell counting kit-8 (CCK-8), Wright-Giemsa staining, and an annexin V-fluorescein isothiocyanate/propidium iodide (FITC/PI) apoptosis detection kit. Additionally, the impact of GEN on mitochondrial function and energy metabolism, including reactive oxygen species (ROS), mitochondrial membrane permeability transition pore (MPTP), lactic acid (LA), oxygen consumption rate (OCR), and adenosine triphosphate (ATP) levels in K562 cells, was also examined. Results The results showed that UCP2 was differentially expressed in clinical samples from patients with ML. Among the three cell lines examined, K562 cells exhibited a significantly higher expression level of UCP2. Functionally, GEN markedly inhibited K562 cell viability while promoting K562 cell differentiation and apoptosis. Mechanistically, UCP2 mRNA and protein expression levels were inhibited by GEN in K562 cells in a concentration- and time-dependent manner. Additionally, GEN dramatically increased ROS generation and induced mitochondrial MPTP opening in K562 cells. Furthermore, GEN significantly reduced LA production in K562 cells and markedly increased OCR and ATP production. Conclusion The results suggest that UCP2 is differentially expressed in ML patients and cell lines; GEN, a UCP2 inhibitor, induces mitochondrial damage and metabolic remodeling, thereby inhibiting proliferation and promoting apoptosis in K562 cells, and thus could be suggested as an adjuvant of an antitumor metabolic therapy.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Biochemistry and Molecular Biology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Rui Sun
- Department of Biochemistry and Molecular Biology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Zi Wen Zhang
- Department of Biochemistry and Molecular Biology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Xin Yi He
- Department of Biochemistry and Molecular Biology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Lin Li
- Department of Biochemistry and Molecular Biology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Chun Jing Zhang
- Department of Biochemistry and Molecular Biology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Ying Liu
- Department of Biochemistry and Molecular Biology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Hai Tao Yu
- Department of Cell Biology and Genetics, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| |
Collapse
|
4
|
XIA LIANGJIANG, LI GUANGBIN, ZHOU QINGWU, FENG YU, MA HAITAO. CircRNA circ_0015278 induces ferroptosis in lung adenocarcinoma through the miR-1228/P53 axis. Oncol Res 2025; 33:465-475. [PMID: 39866239 PMCID: PMC11753987 DOI: 10.32604/or.2024.050835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/11/2024] [Indexed: 01/28/2025] Open
Abstract
Background Circular RNAs play an important role in regulating lung adenocarcinoma (LUAD). Bioinformatics analysis identified circ_0015278 as differentially expressed in LUAD. However, the biological mechanism of circ_0015278 in LUAD has not been fully clarified, especially in ferroptosis. Materials and Methods Bioinformatics analysis was employed to explore the downstream mechanisms of Circ_0015278, subsequently confirmed by luciferase reporter assays. The impact of Circ_0015278 on cell proliferation, migration, invasion, and ferroptosis was investigated through a loss-of-function experiment. A xenotransplantation mouse model elucidated the effect of Circ_0015278 on tumour growth. Results Circ_0015278 exhibited downregulation in LUAD. It inhibited cell proliferation, migration, and invasion while promoting ferroptosis by interacting with miR-1228 to regulate P53 expression through a competitive endogenous RNA mechanism. Moreover, circ_0015278 suppressed tumour growth in mice. Conclusions Circ_0015278 was identified as a novel factor promoting ferroptosis in LUAD. Furthermore, it suppressed the malignant progression of LUAD through the miR-1228/P53 axis.
Collapse
Affiliation(s)
- LIANGJIANG XIA
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - GUANGBIN LI
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - QINGWU ZHOU
- The First Clinical Medical College of Nanchang University, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - YU FENG
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - HAITAO MA
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| |
Collapse
|
5
|
Wang K, Zhang L, Deng B, Zhao K, Chen C, Wang W. Mitochondrial uncoupling protein 2: a central player in pancreatic disease pathophysiology. Mol Med 2024; 30:259. [PMID: 39707176 DOI: 10.1186/s10020-024-01027-y] [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: 10/07/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024] Open
Abstract
Pancreatic diseases pose considerable health challenges due to their complex etiology and limited therapeutic options. Mitochondrial uncoupling protein 2 (UCP2), highly expressed in pancreatic tissue, participates in numerous physiological processes and signaling pathways, indicating its potential relevance in these diseases. Despite this, UCP2's role in acute pancreatitis (AP) remains underexplored, and its functions in chronic pancreatitis (CP) and pancreatic steatosis are largely unknown. Additionally, the mechanisms connecting various pancreatic diseases are intricate and not yet fully elucidated. Given UCP2's diverse functionality, broad expression in pancreatic tissue, and the distinct pathophysiological features of pancreatic diseases, this review offers a comprehensive analysis of current findings on UCP2's involvement in these conditions. We discuss recent insights into UCP2's complex regulatory mechanisms, propose that UCP2 may serve as a central regulatory factor in pancreatic disease progression, and hypothesize that UCP2 dysfunction could significantly contribute to disease pathogenesis. Understanding UCP2's role and mechanisms in pancreatic diseases may pave the way for innovative therapeutic and diagnostic approaches.
Collapse
Affiliation(s)
- Kunpeng Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- General Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lilong Zhang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- General Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Beiying Deng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Kailiang Zhao
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- General Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chen Chen
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China.
- General Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Weixing Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China.
- General Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
6
|
Lv Y, Yuan Z, Chen D, Chen Z, Zhu X, Ying X, Huang Y, Ji W, Qi D. Circular RNA LMBR1 inhibits bladder cancer progression by enhancing expression of the protein ALDH1A3. Noncoding RNA Res 2024; 9:1235-1248. [PMID: 39036604 PMCID: PMC11259990 DOI: 10.1016/j.ncrna.2024.05.004] [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: 10/19/2023] [Revised: 05/11/2024] [Accepted: 05/12/2024] [Indexed: 07/23/2024] Open
Abstract
Background Circular RNAs (circRNAs) have been identified as playing an integral role in the development of bladder cancer (BC). However, the mechanism by which circRNAs operate in the chemical carcinogenesis of BC remains unclear. Methods To explore this mechanism, we used RNA high-throughput sequencing to identify differentially expressed circRNA in bladder epithelial cells and chemically induced malignant transformed BC cells. Subsequently, in vitro experiments were conducted to investigate the biological function and molecular mechanism of circLMBR1 in BC. Finally, animal experiments were conducted to examine the clinical relevance of circLMBR1 in vivo. Results Our profiling of circular RNA expression during cellular malignant transformation induced by chemical carcinogens identified a subset of circRNAs associated with cell transformation. We verified that the expression of circLMBR1 in bladder epithelial malignant transformed cells was decreased compared with control cells, as well as in BC tissues and bladder cell lines. Furthermore, circLMBR1 was seen to inhibit the proliferation, invasion, and migration of BC cells both in vitro and in vivo. Mechanistically, circLMBR1 was found to exert its antitumor effect by binding to the protein ALDH1A3. Conclusions Our findings have revealed that circLMBR1 inhibits the progression of BC cells by binding to ALDH1A3 and upregulating its expression. As such, circLMBR1 serves as a promising predictor of BC and may provide a novel therapeutic target for the treatment of BC.
Collapse
Affiliation(s)
- Yifan Lv
- Department of Urology, Minimally Invasive Surgery Center, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510230, Guangdong, PR China
| | - Zusen Yuan
- Department of Pediatric Surgery, Maternal and Child Health Hospital of Hubei, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430071, Hubei, PR China
| | - Dongmao Chen
- Department of Urology, The First People's Hospital of Zhaoqing, Zhaoqing, 526060, Guangdong, PR China
| | - Zhibin Chen
- Department of Urology, Minimally Invasive Surgery Center, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510230, Guangdong, PR China
| | - Xiaowei Zhu
- Department of Urology, The People's Hospital of Enping, Jiangmen, 529499, Guangdong, PR China
| | - Xiaoling Ying
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, Guangdong, PR China
| | - Yapeng Huang
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510062, Guangdong, PR China
| | - Weidong Ji
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510062, Guangdong, PR China
| | - Defeng Qi
- Department of Urology, Minimally Invasive Surgery Center, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510230, Guangdong, PR China
| |
Collapse
|
7
|
Dong H, Sun K, Wang X, Cui M, Ma Y, Li K, Duan W, Zhang H, Zhang L, Sheng Z, He M, Zhang B. Repurposed genipin targeting UCP2 exhibits antitumor activity through inducing ferroptosis in glioblastoma. Acta Biochim Biophys Sin (Shanghai) 2024; 57:403-414. [PMID: 39523775 PMCID: PMC11986454 DOI: 10.3724/abbs.2024168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/20/2024] [Indexed: 11/16/2024] Open
Abstract
Uncoupling protein-2 (UCP2) controls the antioxidant response and redox homeostasis in cancer and is considered a potent molecular target for cancer treatment. However, the specific mechanism of UCP2 inhibition and its role in glioblastoma (GBM) have not yet been elucidated. Here, we attempt to identify a UCP2 inhibitor and study the underlying molecular mechanism in GBM. Bioinformatics analysis and immunohistochemistry are used to validate the high expression of UCP2 in GBM and its prognostic significance. Drug intervention and tumor xenograft experiments are conducted to determine the inhibitory effect of genipin, a UCP2 inhibitor, on UCP2. The mitochondrial membrane potential and key ferroptosis genes are examined to determine the occurrence of ferroptosis. High expression of UCP2 in GBM is associated with poor prognosis, and inhibiting UCP2 can alleviate the malignant behavior of GBM tumors. Genipin can downregulate the expression of GPX4 and upregulate the expression of ACSL4 by inhibiting UCP2, leading to ferroptosis and alleviating the malignant behavior of tumors. In summary, UCP2 is a potential therapeutic target for GBM. Genipin, which targets UCP2, effectively inhibits GBM development by inducing ferroptosis in vivo and in vitro. These findings indicate that genipin treatment based on UCP2 targeting has potential therapeutic applications with a clinical perspective for the treatment of GBM patients.
Collapse
Affiliation(s)
- Hao Dong
- Department of Diagnostic PathologySchool of Basic Medical SciencesShandong Second Medical UniversityWeifang261042China
| | - Kaixuan Sun
- Department of PathologyAffiliated Hospital of Shandong Second Medical UniversityWeifang261041China
| | - Xuejie Wang
- Department of Diagnostic PathologySchool of Basic Medical SciencesShandong Second Medical UniversityWeifang261042China
| | - Meimei Cui
- Department of Diagnostic PathologySchool of Basic Medical SciencesShandong Second Medical UniversityWeifang261042China
| | - Yaping Ma
- Department of Diagnostic PathologySchool of Basic Medical SciencesShandong Second Medical UniversityWeifang261042China
| | - Kexin Li
- Department of Diagnostic PathologySchool of Basic Medical SciencesShandong Second Medical UniversityWeifang261042China
| | - Wanli Duan
- Department of Diagnostic PathologySchool of Basic Medical SciencesShandong Second Medical UniversityWeifang261042China
| | - Hongxing Zhang
- Department of Diagnostic PathologySchool of Basic Medical SciencesShandong Second Medical UniversityWeifang261042China
| | - Liying Zhang
- Department of Diagnostic PathologySchool of Basic Medical SciencesShandong Second Medical UniversityWeifang261042China
| | - Zhimei Sheng
- Department of Diagnostic PathologySchool of Basic Medical SciencesShandong Second Medical UniversityWeifang261042China
| | - Maotao He
- Department of Diagnostic PathologySchool of Basic Medical SciencesShandong Second Medical UniversityWeifang261042China
- Department of PathologyAffiliated Hospital of Shandong Second Medical UniversityWeifang261041China
| | - Baogang Zhang
- Department of Diagnostic PathologySchool of Basic Medical SciencesShandong Second Medical UniversityWeifang261042China
| |
Collapse
|
8
|
Li H, Lei Y, Chen N, Guo G, Xiang X, Huang Y. circRNA-CPA4 Regulates Cell Proliferation and Apoptosis of Non-small Cell Lung Cancer via the miR-1183/PDPK1 Axis. Biochem Genet 2024; 62:4087-4102. [PMID: 38273153 DOI: 10.1007/s10528-023-10641-0] [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: 10/19/2023] [Accepted: 12/12/2023] [Indexed: 01/27/2024]
Abstract
Non-small-cell lung cancer (NSCLC) stands as a prevalent subtype of lung cancer, with circular RNAs emerging as key players in cancer development. This study elucidates the role of circRNA-CPA4 in NSCLC. Elevated circRNA-CPA4 expression in NSCLC lines was confirmed through qRT-PCR. Silencing circRNA-CPA4 with shRNA revealed, through CCK-8, colony formation, and flow cytometry assays, a notable constraint on proliferation and promotion of apoptosis in NSCLC cells. Subcellular localization analysis, RNA immunoprecipitation, and expression level assessments were employed to decipher the intricate interplay among miR-1183, circRNA-CPA4, and PDPK1. Results demonstrated heightened circRNA-CPA4 levels in NSCLC, and its knockdown curtailed NSCLC growth in vivo. Acting as a molecular sponge for miR-1183, circRNA-CPA4 regulated PDPK1 expression. Conversely, inhibiting miR-1183 counteracted the impact of circRNA-CPA4 silencing, reinstating NSCLC cell proliferation, and impeding apoptosis. Overall, this study unveils a novel mechanism: circRNA-CPA4 promotes PDPK1 expression by sequestering miR-1183, fostering NSCLC cell proliferation, and hindering apoptosis.
Collapse
Affiliation(s)
- Heng Li
- Department of Thoracic Surgery II, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650118, China
| | - Yujie Lei
- Department of Thoracic Surgery I, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunzhou Road 519, Kunming, 650118, China
| | - Nan Chen
- Department of Thoracic Surgery II, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650118, China
| | - Gang Guo
- Department of Thoracic Surgery II, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650118, China
| | - Xudong Xiang
- Department of Thoracic Surgery II, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650118, China
| | - Yunchao Huang
- Department of Thoracic Surgery II, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650118, China.
- Department of Thoracic Surgery I, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunzhou Road 519, Kunming, 650118, China.
| |
Collapse
|
9
|
Siedlecki E, Remiszewski P, Stec R. The Role of circHIPK3 in Tumorigenesis and Its Potential as a Biomarker in Lung Cancer. Cells 2024; 13:1483. [PMID: 39273053 PMCID: PMC11393915 DOI: 10.3390/cells13171483] [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: 07/29/2024] [Revised: 08/27/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
Lung cancer treatment and detection can be improved by the identification of new biomarkers. Novel approaches in investigating circular RNAs (circRNAs) as biomarkers have yielded promising results. A circRNA molecule circHIPK3 was found to be widely expressed in non-small-cell lung cancer (NSCLC) cells, where it plays a crucial role in lung cancer tumorigenesis. CircHIPK3 promotes lung cancer progression by sponging oncosuppressive miRNAs such as miR-124, miR-381-3p, miR-149, and miR-107, which results in increased cell proliferation, migration, and resistance to therapies. Inhibiting circHIPK3 has been demonstrated to suppress tumour growth and induce apoptosis, which suggests its potential use in the development of new lung cancer treatment strategies targeting circHIPK3-related pathways. As a biomarker, circHIPK3 shows promise for early detection and monitoring of lung cancer. CircHIPK3 increased expression levels in lung cancer cells, and its potential link to metastasis risk highlights its clinical relevance. Given the promising preliminary findings, more clinical trials are needed to validate circHIPK3 efficacy as a biomarker. Moreover, future research should determine if the mechanisms discovered in NSCLC apply to small cell lung cancer (SCLC) to investigate circHIPK3-targeted therapies for SCLC.
Collapse
Affiliation(s)
- Eryk Siedlecki
- Department of Oncology, Medical University of Warsaw, 02-097 Warsaw, Poland; (P.R.); (R.S.)
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Piotr Remiszewski
- Department of Oncology, Medical University of Warsaw, 02-097 Warsaw, Poland; (P.R.); (R.S.)
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Rafał Stec
- Department of Oncology, Medical University of Warsaw, 02-097 Warsaw, Poland; (P.R.); (R.S.)
| |
Collapse
|
10
|
Cao L, Liu H, Han Z, Huang C, Guo C, Zhao L, Gao C, Xu Y, Wang G, Feng Z, Li S. MCM8 promotes lung cancer progression through upregulating DNAJC10. J Cell Mol Med 2024; 28:e18488. [PMID: 39031896 PMCID: PMC11190951 DOI: 10.1111/jcmm.18488] [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: 11/13/2023] [Revised: 04/09/2024] [Accepted: 05/28/2024] [Indexed: 07/22/2024] Open
Abstract
MCM8 is a helicase, which participates in DNA replication and tumorigenesis and is upregulated in many human cancers, including lung cancer (LC); however, the function of MCM8 in LC tumour progression is unclear. In this study, we found that MCM8 was expressed at high levels in LC cells and tissues. Further, MCM8 upregulation was associated with advanced tumour grade and lymph node metastasis, and indicated poor prognosis. Silencing of MCM8 suppressed cell growth and migration in vitro and in vivo, while ectopic MCM8 expression promoted cell cycle progression, as well as cell migration, proliferation, and apoptosis. Mechanistically, DNAJC10 was identified as a downstream target of MCM8, using gene array and CO-IP assays. DNAJC10 overexpression combatted the inhibitory activity of MCM8 knockdown on LC progression, while silencing DNAJC10 alleviated the oncogenic function of MCM8 overexpression. MCM8 expression was positively correlated with that of DNAJC10 in LC samples from The Cancer Genome Atlas database, and DNAJC10 upregulation was also associated with poor overall survival of patients with LC. This study indicated that MCM8/DNAJC10 axis plays an important role in in LC development, and maybe as a new potential therapeutic target or a diagnostic biomarker for treating patients with LC.
Collapse
Affiliation(s)
- Lei Cao
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Hongsheng Liu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhijun Han
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Cheng Huang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Chao Guo
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Luo Zhao
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Chao Gao
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yuan Xu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Guige Wang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhe Feng
- Department of Thoracic Surgery, Beijing Sixth Hospital, Beijing, China
| | - Shanqing Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
11
|
Tang J, Yin C, Chen M, Dong M, Xu Y. Yifei Sanjie formula alleviates lung cancer progression via regulating PRMT6-YBX1-CDC25A axis. ENVIRONMENTAL TOXICOLOGY 2024; 39:3225-3237. [PMID: 38357781 DOI: 10.1002/tox.24160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/23/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024]
Abstract
Lung cancer (LC) is the most prevalent cancer type, with a high mortality rate worldwide. The current treatment options for LC have not been particularly successful in improving patient outcomes. Yifei Sanjie (YFSJ), a well-applicated traditional Chinese medicine formula, is widely used to treat pulmonary diseases, especially LC, yet little is known about its molecular mechanisms. This study was conducted to explore the molecular mechanism by which YFSJ ameliorated LC progression. The A549, NCI-H1975, and Calu-3 cells were treated with the YFSJ formula and observed for colony number, apoptosis, migration, and invasion properties recorded via corresponding assays. The PRMT6-YBX1-CDC25A axis was tested and verified through luciferase reporter, RNA immunoprecipitation, and chromatin immunoprecipitation assays and rescue experiments. Our results demonstrated that YFSJ ameliorated LC cell malignant behaviors by increasing apoptosis and suppressing proliferation, migration, and invasion processes. We also noticed that the xenograft mouse model treated with YFSJ significantly reduced tumor growth compared with the control untreated group in vivo. Mechanistically, it was found that YFSJ suppressed the expression of PRMT6, YBX1, and CDC25A, while the knockdown of these proteins significantly inhibited colony growth, migration, and invasion, and boosted apoptosis in LC cells. In summary, our results suggest that YFSJ alleviates LC progression via the PRMT6-YBX1-CDC25A axis, confirming its efficacy in clinical use. The findings of our study provide a new regulatory network for LC growth and metastasis, which could shed new insights into pulmonary medical research.
Collapse
Affiliation(s)
- Jie Tang
- Department of Oncology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Chenyan Yin
- Department of Pharmacy, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Meiyun Chen
- Department of Oncology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Mengjia Dong
- Department of Oncology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Youqi Xu
- Department of Oncology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
12
|
Tang L, Chen Z, Yang J, Li Q, Wang S, Mo T, Zeng W, Ding H, Pan S. Single-cell and Bulk RNA-Seq reveal angiogenic heterogeneity and microenvironmental features to evaluate prognosis and therapeutic response in lung adenocarcinoma. Front Immunol 2024; 15:1352893. [PMID: 38390340 PMCID: PMC10882092 DOI: 10.3389/fimmu.2024.1352893] [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: 12/09/2023] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
Background Angiogenesis stands as a pivotal hallmark in lung adenocarcinoma (LUAD), intricately shaping the tumor microenvironment (TME) and influencing LUAD progression. It emerges as a promising therapeutic target for LUAD, affecting patients' prognosis. However, its role in TME, LUAD prognosis, and its clinical applicability remain shrouded in mystery. Methods We employed integrated single-cell and bulk transcriptome sequencing to unravel the heterogeneity of angiogenesis within LUAD cells. Through "consensus clustering", we delineated distinct angiogenic clusters and deciphered their TME features. "Monocle2" was used to unravel divergent trajectories within malignant cell subpopulations of LUAD. Additionally, regulon submodules and specific cellular communication patterns of cells in different angiogenic states were analyzed by "pyscenic" and "Cellchat" algorithms. The "univariate Cox" and "LASSO" algorithms were applied to build angiogenic prognostic models. Immunohistochemistry (IHC) on clinical samples validated the role of model factors in LUAD angiogenesis. We utilized CTRP 2.0 and PRISM databases for pinpointing sensitive drugs against lung adenocarcinoma. Results Two clusters for the activation of angiogenesis were identified, with Cluster 1 showing a poor prognosis and a pro-cancerous TME. Three differentiated states of malignant epithelial LUAD cells were identified, which had different degrees of angiogenic activation, were regulated by three different regulon submodules, and had completely different crosstalk from other cells in TME. The experiments validate that SLC2A1 promotes angiogenesis in LUAD. ARS (Angiogenesis related score) had a high prognostic value; low ARSs showed immunotherapy benefits, whereas high ARSs were sensitive to 15 chemotherapeutic agents. Conclusion The assessment of angiogenic clusters helps to determine the prognostic and TME characteristics of LUAD. Angiogenic prognostic models can be used to assess the prognosis, immunotherapeutic response, and chemotherapeutic drug sensitivity of LUAD.
Collapse
Affiliation(s)
- Lijuan Tang
- Dalian Medical University, Dalian, China
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
| | - Zhike Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qifan Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Sichu Wang
- Dalian Medical University, Dalian, China
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
| | - Taoming Mo
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Weibiao Zeng
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hao Ding
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shu Pan
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Suzhou Gene Pharma Co., Ltd, Suzhou, China
| |
Collapse
|
13
|
Wang Q, Chen M, Tang X. Luteolin Inhibits Lung Cancer Cell Migration by Negatively Regulating TWIST1 and MMP2 Through Upregulation of miR-106a-5p. Integr Cancer Ther 2024; 23:15347354241247223. [PMID: 38646808 PMCID: PMC11034356 DOI: 10.1177/15347354241247223] [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: 01/04/2024] [Revised: 03/10/2024] [Accepted: 03/29/2024] [Indexed: 04/23/2024] Open
Abstract
BACKGROUND Luteolin, a common dietary flavonoid found in plants, has been shown to have anti-cancer properties. However, its exact mechanisms of action in non-small cell lung cancer (NSCLC) are still not fully understood, particularly its role in regulating broader genomic networks and specific gene targets. In this study, we aimed to elucidate the role of microRNAs (miRNAs) in NSCLC treated with luteolin, using A549 cells as a model system. MATERIALS AND METHODS miRNA profiling was conducted on luteolin-treated A549 cells using Exiqon microarrays, with validation of selected miRNAs by qRT-PCR. Bioinformatic analysis identified the regulatory roles of miRNAs in biological processes and pathways following luteolin treatment. Computational algorithms were employed to identify potential target genes. A549 cells were transfected with miR-106a-5p mimic and inhibitor or their corresponding controls. The expression levels of 2 genes, twist basic helix-loop-helix transcription factor 1 (TWIST1) and matrix metallopeptidase 2 (MMP2), and cell migration were assessed. RESULTS miRNA profiling identified 341 miRNAs, with 18 exhibiting significantly altered expression (P < 0.05). Subsequent qRT-PCR analysis confirmed altered expression of 6 selected miRNAs. KEGG and GO analyses revealed significant alterations in pathways and biological processes crucial for tumor biology. TWIST1 and MMP2, which both contain conserved miR-106a-5p binding sites, exhibited an inverse correlation with the expression levels of miR-106a-5p. Dual-luciferase reporter assays confirmed TWIST1 and MMP2 as direct targets of miR-106a-5p. Luteolin treatment led to a reduction in A549 cell migration, and this reduction was further amplified by the overexpression of miR-106a-5p. CONCLUSION Luteolin inhibits A549 cell migration by modulating the miRNA landscape, shedding light on its mechanisms and laying the foundation for miRNA-based therapeutic approaches for NSCLC.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310005, People’s Republic of China
| | - Mengyuan Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Xiaofang Tang
- Department of Cadre Health Care, Zhejiang Hospital, Hangzhou, People’s Republic of China
| |
Collapse
|