1
|
Zhang Y, Liu J, Yang G, Zou J, Tan Y, Xi E, Geng Q, Wang Z. Asiaticoside Inhibits Growth and Metastasis in Non-Small Cell Lung Cancer by Disrupting EMT via Wnt/β-Catenin Pathway. ENVIRONMENTAL TOXICOLOGY 2024; 39:4859-4870. [PMID: 38888371 DOI: 10.1002/tox.24359] [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: 06/08/2023] [Revised: 04/07/2024] [Accepted: 05/11/2024] [Indexed: 06/20/2024]
Abstract
Non-small cell lung cancer (NSCLC) is the primary inducer of cancer-related death worldwide. Asiaticoside (ATS) is a triterpenoid saponin that has been indicated to possess an antitumor activity in several malignancies. Nonetheless, its detailed functions in NSCLC remain unclarified. In this study, NSCLC cells were exposed to various doses of ATS. Functional experiments were employed to estimate the ATS effect on NSCLC cell behaviors. Western blotting was implemented for protein expression evaluation. A xenograft mouse model was established to assess the ATS effect on NSCLC in vivo. The results showed that ATS restrained NSCLC cell proliferation, cell cycle progression, migration, and invasiveness. ATS reversed TGF-β-induced promotion in epithelial-mesenchymal transition (EMT). Mechanistically, ATS inhibited Wnt/β-catenin signaling in NSCLC. Upregulating β-catenin restored ATS-mediated suppression of NSCLC cell aggressiveness. Moreover, ATS administration repressed tumorigenesis in tumor-bearing mice. In conclusion, ATS represses growth and metastasis in NSCLC by blocking EMT via the inhibition of Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Yanan Zhang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiangyong Liu
- Department of Radiography, Central Theater Command General Hospital of Chinese People's Liberation Army, Wuhan, China
| | - Gang Yang
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, Wuhan, China
| | - Jiani Zou
- Department of Radiography, Central Theater Command General Hospital of Chinese People's Liberation Army, Wuhan, China
| | - Yan Tan
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, Wuhan, China
| | - Erping Xi
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, Wuhan, China
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zheng Wang
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, Wuhan, China
| |
Collapse
|
2
|
Guo M, Ying Y, Chen Y, Miao X, Yu Z. Asiaticoside inhibits breast cancer progression and tumor angiogenesis via YAP1/VEGFA signal pathway. Heliyon 2024; 10:e37169. [PMID: 39309801 PMCID: PMC11416243 DOI: 10.1016/j.heliyon.2024.e37169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 08/25/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Objective Breast cancer poses a major health risk to millions of females globally. Asiaticoside (AC) is a naturally occurring compound derived from Centella asiatica, a widely used medicinal plant in the oriental countries and has potential antitumor properties. The primary aim of this study was to investigate the anti-cancer effects of synthesized AC at the cellular level and assess its ability to inhibit tumor growth and angiogenesis in breast cancer. Methods The proliferative capacities of MCF-7 and MDA-MB-231 cells were determined using CCK-8 assay. To analyze invasion and migration, Transwell assays were conducted on the same cell lines. Additionally, apoptosis was analyzed in vitro using flow cytometry. Real-time RT-PCR was used to examine mRNA expression, and Western-blotting assay was employed to examine protein expression. Subcutaneous injection of MDA-MB-231 cells into female BALB/c nude mice was followed by treatment with AC to study its anti-tumor effects in vivo. Results AC treatment reduced cell proliferation and triggered apoptosis in MCF-7 and MDA-MB-231 cells. The invasive and pro-angiogenesis ability were also impaired upon AC treatment. AC administration also impeded the tumor growth and tumor-associated angiogenesis of MDA-MB-231 cells in nude mice, which was accompanied by the decreased levels of YAP1 and VEGFA. Conclusion Taken together, our results demonstrated the anti-cancer activity of AC in breast cancer. AC is able to suppress the malignancy of breast cancercells via YAP1/VEGFA signal pathway.
Collapse
Affiliation(s)
- Mengmeng Guo
- General Surgery Department, Nantong Hospital Affiliated to Nanjing University of Chinese Medicine, No.41, Jianshe Road, Chongchuan District, Nantong, 220000, Jiangsu, China
| | - Yu Ying
- Breast Disease Department, Jiangsu Provincial Hospital of Traditional Chinese Medicine, No. 155, Hanzhong Road, Qinhuai District, Nanjing, Jiangsu, China
| | - Yun Chen
- Department of Medical Oncology, Jiangsu Cancer Hospital, No. 42, Baizi Pavilion, Kunlun Road, Xuanwu District, Nanjing, Jiangsu, China
| | - Xian Miao
- Oncology Department, Nantong Hospital Affiliated to Nanjing University of Chinese Medicine, No.41, Jianshe Road, Chongchuan District, Nantong, 226000, Jiangsu, China
| | - Zhenghong Yu
- Rheumatology and Immunology Department, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, No. 278, Central Road, Nanjing City, China
| |
Collapse
|
3
|
Wang J, Wang J. Asiaticoside protected brain injury in hypertensive intracerebral hemorrhage via activation of the PI3K/AKT pathway. J Biochem Mol Toxicol 2024; 38:e23843. [PMID: 39253885 DOI: 10.1002/jbt.23843] [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: 01/23/2024] [Revised: 08/06/2024] [Accepted: 08/29/2024] [Indexed: 09/11/2024]
Abstract
Hypertensive intracerebral hemorrhage (HICH) is a destructive disease with high mortality, incidence, and disability. Asiaticoside (AC) is a triterpenoid derivative that has demonstrated to exert a protective effect on neuron and blood vessel. To investigate the function and potential mechanism of AC on HICH. Human brain microvascular endothelial cells (hBMECs) were treated with 20 U/mL thrombin for 24 h to establish the HICH model in vitro, and AC with the concentration of 1, 2 and 4 µM were used to incubate hBMECs. The effect and potential mechanism of AC on HICH were investigated by using cell counting kit-8, flow cytometry, tube forming assays, vascular permeability experiments and western blot assays. In vivo, rats were injected with 20 µL hemoglobin with a concentration of 150 mg/mL, and then intragastrically administrated with 1.25, 2.5 and 5 mg/kg AC. Behavioral tests, brain water content measurement, hematoxylin-eosin (HE) staining, terminal deoxynucleotidyl transferase deoxyuridine triphosphate (dUTP) nick end labeling assays, and western blot were used to assess the effect and potential mechanism of AC on HICH. AC (at 2 and 4 µM) improved the proliferation, apoptosis, angiogenesis and vascular permeability in thrombin-induced hBMECs (p < 0.05). Besides, AC (2.5 and 5 mg/kg) ameliorated behavioral scores, brain water content, pathological lesion, apoptosis and the expression of vascular permeability-related proteins in rats with HICH (p < 0.05). In addition, AC elevated the expression of PI3K/AKT pathway after HICH both in cell and animal models (p < 0.05). Application of LY294002, an inhibitor of PI3K/AKT pathway, reversed the ameliorative effect of AC on the proliferation, apoptosis, angiogenesis and vascular permeability in thrombin-induced hBMECs (p < 0.05). AC reduced brain damage by increasing the expression of the PI3K/AKT pathway after HICH.
Collapse
Affiliation(s)
- Jicun Wang
- Department of Neurology, The Hospital of Shunyi District Beijing, Beijing, China
| | - Jianxin Wang
- Department of Neurosurgery, Henan Provincial People's Hospital, Henan Provincial Cerebrovascular Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, China
- Department of Neurosurgery, The Hospital of Shunyi District Beijing, Beijing, China
| |
Collapse
|
4
|
Li Y, Yuan S, Zhou Y, Zhou J, Zhang X, Zhang P, Xiao W, Zhang Y, Deng J, Lou S. Long non-coding RNA PXN-AS1 promotes glutamine synthetase-mediated chronic myeloid leukemia BCR::ABL1-independent resistance to Imatinib via cell cycle signaling pathway. Cancer Cell Int 2024; 24:186. [PMID: 38811958 PMCID: PMC11138077 DOI: 10.1186/s12935-024-03363-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 05/08/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Chronic myeloid leukemia (CML) is a common hematological malignancy, and tyrosine kinase inhibitors (TKIs) represent the primary therapeutic approach for CML. Activation of metabolism signaling pathway has been connected with BCR::ABL1-independent TKIs resistance in CML cells. However, the specific mechanism by which metabolism signaling mediates this drug resistance remains unclear. Here, we identified one relationship between glutamine synthetase (GS) and BCR::ABL1-independent Imatinib resistance in CML cells. METHODS GS and PXN-AS1 in bone marrow samples of CML patients with Imatinib resistance (IR) were screened and detected by whole transcriptome sequencing. GS expression was upregulated using LVs and blocked using shRNAs respectively, then GS expression, Gln content, and cell cycle progression were respectively tested. The CML IR mice model were established by tail vein injection, prognosis of CML IR mice model were evaluated by Kaplan-Meier analysis, the ratio of spleen/body weight, HE staining, and IHC. PXN-AS1 level was blocked using shRNAs, and the effects of PXN-AS1 on CML IR cells in vitro and in vivo were tested the same as GS. Several RNA-RNA tools were used to predict the potential target microRNAs binding to both GS and PXN-AS1. RNA mimics and RNA inhibitors were used to explore the mechanism through which PXN-AS1 regulates miR-635 or miR-635 regulates GS. RESULTS GS was highly expressed in the bone marrow samples of CML patients with Imatinib resistance. In addition, the lncRNA PXN-AS1 was found to mediate GS expression and disorder cell cycle in CML IR cells via mTOR signaling pathway. PXN-AS1 regulated GS expression by binding to miR-635. Additionally, knockdown of PXN-AS1 attenuated BCR::ABL1-independent Imatinib resistance in CML cells via PXN-AS1/miR-635/GS/Gln/mTOR signaling pathway. CONCLUSIONS Thus, PXN-AS1 promotes GS-mediated BCR::ABL1-independent Imatinib resistance in CML cells via cell cycle signaling pathway.
Collapse
Affiliation(s)
- Yifei Li
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, 400010, China
| | - Shiyi Yuan
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, 400010, China
| | - Ying Zhou
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, 400010, China
| | - Jingwen Zhou
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, 400010, China
| | - Xuan Zhang
- Department of Oncology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400316, China
| | - Ping Zhang
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, 400010, China
| | - Wenrui Xiao
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, 400010, China
| | - Ying Zhang
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, 400010, China.
| | - Jianchuan Deng
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, 400010, China.
| | - Shifeng Lou
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
5
|
Zhang Y, Han Y, Dong J, Li F, Sun Y. Asiaticoside Down-Regulates HIF-1α to Inhibit Proliferation, Migration, and Angiogenesis in Thyroid Cancer Cells. Balkan Med J 2024; 41:23-29. [PMID: 38044598 PMCID: PMC10767772 DOI: 10.4274/balkanmedj.galenos.2023.2023-7-123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/26/2023] [Indexed: 12/05/2023] Open
Abstract
Background Thyroid cancer (TC), the most prevalent endocrine malignancy, has been subjected to various treatment methods. However, the efficacy of asiaticoside (AC) for treating TC remains uncertain. Aims To explore the impact of AC on TC and determine its potential mechanisms of action. Study Design In vitro and in vivo cell line study. Methods We evaluated the effects of AC on human TC cell lines, namely TPC-1 and FTC-133. Both in vitro and in vivo experimental validations were conducted. Results AC significantly diminished the viability and proliferation of TC cells based on the CCK-8 assay and Edu staining findings. Migration and invasion assays revealed that AC effectively curtailed the migration and invasiveness of TC cells. The tube formation assay demonstrated that AC substantially impeded TC cell-induced angiogenesis. Western blot assay revealed that AC significantly reduced the expression levels of TRAF6, HIF-1α, and VEGFA, indicating that AC could potentially exert its anticancer effect by inhibiting the TRAF6/HIF1α pathway. Our in vivo experiments, which involved administering AC to BALB/c nude mice injected with TPC-1 cells, demonstrated significant inhibition of tumor growth and reduction in the expression of Ki-67, TRAF6, HIF-1α, and VEGFA. Conclusion Our study highlights the significant inhibitory effect of AC on TC, offering fresh insights and potential drug candidates for TC treatment.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Neck Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yifan Han
- Department of Neck Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianda Dong
- Department of Neck Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Feilei Li
- Department of Neck Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yihan Sun
- Department of Neck Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
6
|
Jia J, Guo P, Zhang L, Kong W, Wang F. LINC01614 Promotes Colorectal Cancer Cell Growth and Migration by Regulating miR-217-5p/HMGA1 Axis. Anal Cell Pathol (Amst) 2023; 2023:6833987. [PMID: 39282156 PMCID: PMC11401691 DOI: 10.1155/2023/6833987] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 05/10/2023] [Accepted: 05/23/2023] [Indexed: 09/18/2024] Open
Abstract
Colorectal cancer (CRC) substantially contributes to cancer-related deaths worldwide. Recently, a long non-coding RNA (lncRNA), LINC01614, has emerged as a vital gene regulator in cancer progression. Yet, how LINC01614 affects CRC progression remains enigmatic. Here, we defined LINC01614 expression in CRC, investigated the performance of CRC cells lacking LINC01614, and elucidated the underpinning mechanism. We observed that LINC01614 was upregulated in both CRC tissues and cell lines. LINC01614 knockdown repressed the proliferation and metastasis capacity of CRC cell lines. Consistently, an in vivo LINC01614 deficiency model exhibited slow tumor growth rate. Moreover, luciferase reporter assay, RNA pull-down, and immunoprecipitation confirmed that LINC01614 targeted miR-217-5p. LINC01614 knockdown reduced the expression of HMGA1 and N-cadherin, while increasing that of E-cadherin, resulting in decreased viability, proliferation, migration, and invasion capacity of CRC cells. Our results demonstrate that LINC01614 regulates CRC progression by modulating the miR-217-5p/HMGA1 axis, thus holding great potential as a prognostic biomarker for CRC diagnosis and treatment.
Collapse
Affiliation(s)
- Jiwei Jia
- Department of Radiation Oncology, Yantai Yuhuangding Hospital, 20 Yuhuangding East Road, Yantai, Shandong 264000, China
| | - Pei Guo
- Department of Radiation Oncology, Yantai Yuhuangding Hospital, 20 Yuhuangding East Road, Yantai, Shandong 264000, China
| | - Li Zhang
- Department of Pathology, Yantai Yuhuangding Hospital, 20 Yuhuangding East Road, Yantai, Shandong 264000, China
| | - Wenqing Kong
- Central Ward Operating Room, Yantai Yuhuangding Hospital, 20 Yuhuangding East Road, Yantai, Shandong 264000, China
| | - Fangfang Wang
- Outpatient Operating Room, Yantai Yuhuangding Hospital, 20 Yuhuangding East Road, Yantai, Shandong 264000, China
| |
Collapse
|