1
|
Shi X, Zhang R, Liu Z, Mao X, Fan B, Guo J. Specific expression profiles of lncRNAs in cis-regulatory responses to gill in Litopenaeus vannamei under high alkalinity. Int J Biol Macromol 2025; 305:141272. [PMID: 39978500 DOI: 10.1016/j.ijbiomac.2025.141272] [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: 09/18/2024] [Revised: 01/15/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
As a pivotal regulatory molecule, long non-coding RNA (lncRNA) can coordinate the network regulation of transcription and post-transcriptional processes. However, little is known about their cis-regulatory contributions in gene expression of crustaceans under alkalinity stress. In this study, the purpose was to explore the potential role of lncRNA in gill of Pacific white shrimp (Litopenaeus vannamei) under different alkalinity stress (C: 50 mg/L and T: 350 mg/L). We found 5034 high confidence lncRNAs in gills. Among them, 82 differentially expressed lncRNA (DELs) in C and T groups may affect gill stress by regulating gene expression in cis- or trans- mode at transcriptional level. The cis-functions DELs were enriched in several pathways, including Glutathione metabolism, Peroxisome and Lysosome. The trans-functions DELs were enriched in Toll-like receptor signaling pathway, Peroxisome and Lysosome. We also obtained 12 DELs-mRNA pairs consisting of 6 DELs and 11 mRNAs, and DELs have potential for positively affecting their neighboring genes, predominantly in a cis-regulatory manner. In addition, we found that two hub genes, ARSB and gba-3, were significantly enriched in Lysosome. In the silencing experiment, XR_003476973.1 and MSTRG.20723.1 were found to regulate the Lysosomal pathway under alkalinity stress through cis-acting. Under alkalinity stress, physiological parameters related to homeostasis regulation, immune response and lysosomal hydrolysis changed significantly. These results provide new insights into the mechanism of lncRNA in shrimp gills under alkaline stress, and also offer valuable perspectives into the implications of gill regulation for crustaceans in saline-alkaline water.
Collapse
Affiliation(s)
- Xiang Shi
- College of Animal Science & Technology, Gansu Agricultural University, No. 1 Yingmen Village, Anning District, Lanzhou 730070, Gansu Province, China
| | - Ruiqi Zhang
- College of Animal Science & Technology, Gansu Agricultural University, No. 1 Yingmen Village, Anning District, Lanzhou 730070, Gansu Province, China.
| | - Zhe Liu
- College of Animal Science & Technology, Gansu Agricultural University, No. 1 Yingmen Village, Anning District, Lanzhou 730070, Gansu Province, China
| | - Xue Mao
- College of Animal Science & Technology, Gansu Agricultural University, No. 1 Yingmen Village, Anning District, Lanzhou 730070, Gansu Province, China
| | - Baoyi Fan
- College of Animal Science & Technology, Gansu Agricultural University, No. 1 Yingmen Village, Anning District, Lanzhou 730070, Gansu Province, China
| | - Jintao Guo
- College of Animal Science & Technology, Gansu Agricultural University, No. 1 Yingmen Village, Anning District, Lanzhou 730070, Gansu Province, China
| |
Collapse
|
2
|
Sun LI, Zhuo S, Li X, Kong H, DU W, Zhou C, Huang J. Astragalus polysaccharide enhances the therapeutic efficacy of cisplatin in triple-negative breast cancer through multiple mechanisms. Oncol Res 2025; 33:641-651. [PMID: 40109863 PMCID: PMC11915043 DOI: 10.32604/or.2024.050057] [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: 01/26/2024] [Accepted: 06/03/2024] [Indexed: 03/22/2025] Open
Abstract
Background Cisplatin (DDP) has been used in the treatment of various human cancers. However, DDP alone lacks efficacy in treating triple-negative breast cancer (TNBC), and its clinical application is often hampered by side effects. Astragalus polysaccharide (APS) is one of the active components extracted from Astragalus membranaceus and has gained attention for its various biological properties. This research is aimed to evaluate the effectiveness of a combination of APS and DDP on TNBC and explore the potential mechanisms. Methods The efficacy and mechanisms of single or combined treatment were evaluated using Cell Counting Kit-8 (CCK8) assay, Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) staining, wound healing assay, trans-well invasion/migration assay, hematoxylin-eosin (HE) staining, immunohistochemical (IHC) staining, Western Blot (WB) analysis, and fluorescence-activated cell sorting (FACS). An orthotopic model of TNBC was used to assess the in vivo treatment efficacy of single or combination treatment. Results APS significantly enhanced the anti-proliferative, anti-migratory, and anti-invasive effects of DDP on TNBC cells. The combination of APS and DDP downregulated anti-apoptotic genes (Bcl2 and Bcl-xL) while upregulating pro-apoptotic genes (Puma, Cle-Caspase3, Cle-PARP), leading to enhanced apoptosis. This combination treatment increased E-cadherin levels, decreased Vimentin, Snail, Slug, and Twist levels, and effectively suppressed epithelial-mesenchymal transition (EMT)-associated cell invasion. In the orthotopic model of TNBC, a synergistic reduction in tumor growth was observed in mice treated with APS and DDP. Additionally, the combination of APS and DDP induced the infiltration of CD8+ T lymphocytes into the tumor immune microenvironment. Conclusion The combination of APS and DDP exhibits more potent tumor inhibition and anti-tumor immunity than either agent alone, representing a novel approach to enhance therapeutic efficacy without increasing the side effects of DDP.
Collapse
Affiliation(s)
- L I Sun
- Department of Oncology, Taizhou People's Hospital Affiliated to Nanjing University of Chinese Medicine, Taizhou, 225300, China
| | - Shichao Zhuo
- Department of Pathology, Xuzhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Xuzhou, 221009, China
| | - Xiaoxin Li
- Department of Pathology, Xuzhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Xuzhou, 221009, China
| | - Husheng Kong
- Department of Oncology, Xuzhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Xuzhou, 221009, China
| | - Weiwei DU
- Department of Oncology, Xuzhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Xuzhou, 221009, China
| | - Chong Zhou
- Department of Radiotherapy, Xuzhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Xuzhou, 221009, China
| | - Junxing Huang
- Department of Oncology, Taizhou People's Hospital Affiliated to Nanjing University of Chinese Medicine, Taizhou, 225300, China
| |
Collapse
|
3
|
Fu Y, Zeng S, Wang Z, Huang H, Zhao X, Li M. Mechanisms of Copper-Induced Autophagy and Links with Human Diseases. Pharmaceuticals (Basel) 2025; 18:99. [PMID: 39861161 PMCID: PMC11768742 DOI: 10.3390/ph18010099] [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: 12/18/2024] [Revised: 01/04/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
As a structural and catalytic cofactor, copper is involved in many biological pathways and is required for the biochemistry of all living organisms. However, excess intracellular copper can induce cell death due to its potential to catalyze the generation of reactive oxygen species, thus copper homeostasis is strictly regulated. And the deficiency or accumulation of intracellular copper is connected with various pathological conditions. Since the success of platinum-based compounds in the clinical treatment of various types of neoplasias, metal-based drugs have shown encouraging perspectives for drug development. Compared to platinum, copper is an essential intracellular trace element that may have better prospects for drug development than platinum. Recently, the potential therapeutic role of copper-induced autophagy in chronic diseases such as Parkinson's, Wilson's, and cardiovascular disease has already been demonstrated. In brief, copper ions, numerous copper complexes, and copper-based nano-preparations could induce autophagy, a lysosome-dependent process that plays an important role in various human diseases. In this review, we not only focus on the current advances in elucidating the mechanisms of copper or copper-based compounds/preparations on the regulation of autophagy but also outline the association between copper-induced autophagy and human diseases.
Collapse
Affiliation(s)
- Yuanyuan Fu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Shuyan Zeng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Zhenlin Wang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Huiting Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Xin Zhao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Min Li
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| |
Collapse
|
4
|
Liu Y, Zou Y, Ye Y, Chen Y. Advances in the Understanding of the Pathogenesis of Triple-Negative Breast Cancer. Cancer Med 2024; 13:e70410. [PMID: 39558881 PMCID: PMC11574469 DOI: 10.1002/cam4.70410] [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: 09/02/2024] [Revised: 10/24/2024] [Accepted: 10/30/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a heterogeneous disease characterized by high aggressiveness, high malignancy, and poor prognosis compared to other breast cancer subtypes. OBJECTIVE This review aims to explore recent advances in understanding TNBC and to provide new insights and potential references for clinical treatment. METHODS We examined current literature on TNBC to analyze molecular subtypes, genetic mutations, signaling pathways, mechanisms of drug resistance, and emerging therapies. RESULTS Findings highlight key aspects of TNBC's molecular subtypes, relevant mutations, and pathways, alongside emerging treatments that target drug resistance mechanisms. CONCLUSION These insights into TNBC pathogenesis may help guide future therapeutic strategies and improve clinical outcomes for patients with TNBC.
Collapse
Affiliation(s)
- Yuhan Liu
- School of Clinical MedicineShandong Second Medical UniversityWeifangChina
| | - Yuhan Zou
- School of Clinical MedicineShandong Second Medical UniversityWeifangChina
| | - Yangli Ye
- College of Life Sciences and TechnologyShandong Second Medical UniversityWeifangChina
| | - Yong Chen
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical SciencesShandong Second Medical UniversityWeifangChina
| |
Collapse
|
5
|
Chen L, Zhu S, Zhao L, Ye W. Identification of exosome-related gene features in psoriasis and construction of a diagnostic model via integrated bioinformatics analysis. Comput Methods Biomech Biomed Engin 2024:1-12. [PMID: 39358990 DOI: 10.1080/10255842.2024.2410224] [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: 05/23/2024] [Revised: 09/11/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Psoriasis, a chronic inflammatory dermatosis, profoundly affects patients' well-being. Although exosomes are key in disease etiology, diagnostic potentials of associated genes are unclear. Our research targeted bioinformatics-based characterization of exosome-related genes and the development of a diagnostic model for psoriasis. METHODS Within GSE30999 dataset, an exosome-centric diagnostic model was formulated. Its diagnostic capability was appraised in GSE30999 and GSE14905 cohorts. Human keratinocytes (HaCaT) were used to construct psoriasis cell model. qRT-PCR was used to detect expression of diagnostic genes in the model. Construction of a protein-protein interaction network was undertaken, complemented by enrichment analyses. Comparative evaluation of immunological microenvironments between healthy controls and disease cohort was executed. Prospective miRNAs and transcription factors (TFs) were prognosticated using online prediction tools. RESULTS A distinctive diagnostic model with superior diagnostic performance, evidenced by an AUC value greater than 0.88, was unveiled. The model featured seven exosome-related biomarker genes (CCNA2, NDC80, CCNB1, CDCA8, KIF11, CENPF, and ASPM) interwoven in a complex network and chiefly linked in the regulation of Cell Cycle and Cellular Senescence. These genes were significantly overexpressed in psoriasis cell models. Immune infiltration analysis distinguished profound discrepancies (p < 0.05) in immunological microenvironment between disease and control groups with enrichment of T cells CD4 memory activated, Macrophages M1, and Neutrophils in the disease group. 11 miRNAs and 27 TFs were identified. CONCLUSION The study introduces a new and potent diagnostic model for psoriasis, with selection of credible exosome-associated biomarker genes. These discoveries aid in clinical diagnostics and research on exosome involvement in psoriasis.
Collapse
Affiliation(s)
- Lifen Chen
- Department of Clinical Laboratory, Lishui People's Hospital, Lishui City, Zhejiang Province, China
| | - Shuangmei Zhu
- Department of Radiation Oncology, Lishui People's Hospital, Lishui City, Zhejiang Province, China
| | - Lu Zhao
- Department of Ultrasound, Lishui People's Hospital, Lishui City, Zhejiang Province, China
| | - Wenxia Ye
- Department of Dermatology, Lishui People's Hospital, Lishui City, Zhejiang Province, China
| |
Collapse
|
6
|
Anyin Y, Jianping L, Mengru L, Hong Z, Xulei Z, Lianping W. Integrating bioinformatics and machine learning methods to analyze diagnostic biomarkers for HBV-induced hepatocellular carcinoma. Diagn Pathol 2024; 19:105. [PMID: 39095799 PMCID: PMC11295615 DOI: 10.1186/s13000-024-01528-8] [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: 06/21/2024] [Accepted: 07/21/2024] [Indexed: 08/04/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor. It is estimated that approximately 50-80% of HCC cases worldwide are caused by hepatitis b virus (HBV) infection, and other pathogenic factors have been shown to promote the development of HCC when coexisting with HBV. Understanding the molecular mechanisms of HBV-induced hepatocellular carcinoma (HBV-HCC) is crucial for the prevention, diagnosis, and treatment of the disease. In this study, we analyzed the molecular mechanisms of HBV-induced HCC by combining bioinformatics and deep learning methods. Firstly, we collected a gene set related to HBV-HCC from the GEO database, performed differential analysis and WGCNA analysis to identify genes with abnormal expression in tumors and high relevance to tumors. We used three deep learning methods, Lasso, random forest, and SVM, to identify key genes RACGAP1, ECT2, and NDC80. By establishing a diagnostic model, we determined the accuracy of key genes in diagnosing HBV-HCC. In the training set, RACGAP1(AUC:0.976), ECT2(AUC:0.969), and NDC80 (AUC: 0.976) showed high accuracy. They also exhibited good accuracy in the validation set: RACGAP1(AUC:0.878), ECT2(AUC:0.731), and NDC80(AUC:0.915). The key genes were found to be highly expressed in liver cancer tissues compared to normal liver tissues, and survival analysis indicated that high expression of key genes was associated with poor prognosis in liver cancer patients. This suggests a close relationship between key genes RACGAP1, ECT2, and NDC80 and the occurrence and progression of HBV-HCC. Molecular docking results showed that the key genes could spontaneously bind to the anti-hepatocellular carcinoma drugs Lenvatinib, Regorafenib, and Sorafenib with strong binding activity. Therefore, ECT2, NDC80, and RACGAP1 may serve as potential biomarkers for the diagnosis of HBV-HCC and as targets for the development of targeted therapeutic drugs.
Collapse
Affiliation(s)
- Yang Anyin
- Department of Pharmacy, Gaochun People's Hospital, Gaochun Hospital Affiliated to Jiangsu University, Nanjing, 211300, China
| | - Liu Jianping
- Department of Pharmacy, Gaochun People's Hospital, Gaochun Hospital Affiliated to Jiangsu University, Nanjing, 211300, China
| | - Li Mengru
- Department of Hospital Infection Management Section, Gaochun People's Hospital, Gaochun Hospital Affiliated to Jiangsu University, Nanjing, 211300, China
| | - Zhang Hong
- Department of Pharmacy, Gaochun People's Hospital, Gaochun Hospital Affiliated to Jiangsu University, Nanjing, 211300, China
| | - Zhang Xulei
- Department of Liver Disease, Gaochun People's Hospital, Gaochun Hospital Affiliated to Jiangsu University, Nanjing, 211300, China.
| | - Wu Lianping
- Department of Pharmacy, Gaochun People's Hospital, Gaochun Hospital Affiliated to Jiangsu University, Nanjing, 211300, China.
| |
Collapse
|
7
|
Wang Q, Li H, Wu T, Yu B, Cong H, Shen Y. Nanodrugs based on co-delivery strategies to combat cisplatin resistance. J Control Release 2024; 370:14-42. [PMID: 38615892 DOI: 10.1016/j.jconrel.2024.04.020] [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: 12/30/2023] [Revised: 03/24/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
Cisplatin (CDDP), as a broad-spectrum anticancer drug, is able to bind to DNA and inhibit cell division. Despite the widespread use of cisplatin since its discovery, cisplatin resistance developed during prolonged chemotherapy, similar to other small molecule chemotherapeutic agents, severely limits its clinical application. Cisplatin resistance in cancer cells is mainly caused by three reasons: DNA repair, decreased cisplatin uptake/increased efflux, and cisplatin inactivation. In earlier combination therapies, the emergence of multidrug resistance (MDR) in cancer cells prevented the achievement of the desired therapeutic effect even with the accurate combination of two chemotherapeutic drugs. Therefore, combination therapy using nanocarriers for co-delivery of drugs is considered to be ideal for alleviating cisplatin resistance and reducing cisplatin-related toxicity in cancer cells. This article provides an overview of the design of cisplatin nano-drugs used to combat cancer cell resistance, elucidates the mechanisms of action of cisplatin and the pathways through which cancer cells develop resistance, and finally discusses the design of drugs and related carriers that can synergistically reduce cancer resistance when combined with cisplatin.
Collapse
Affiliation(s)
- Qiubo Wang
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Hui Li
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Taixia Wu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Bing Yu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China.
| | - Hailin Cong
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China; School of Materials Science and Engineering, Shandong University of Technology, Zibo 255000, China.
| | - Youqing Shen
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bio-nanoengineering, and Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| |
Collapse
|
8
|
Zhao PW, Cui JX, Wang XM. Upregulation of p300 in paclitaxel-resistant TNBC: implications for cell proliferation via the PCK1/AMPK axis. THE PHARMACOGENOMICS JOURNAL 2024; 24:5. [PMID: 38378770 DOI: 10.1038/s41397-024-00324-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/18/2024] [Accepted: 01/24/2024] [Indexed: 02/22/2024]
Abstract
OBJECTIVE To explore the role of p300 in the context of paclitaxel (PTX) resistance in triple-negative breast cancer (TNBC) cells, focusing on its interaction with the phosphoenolpyruvate carboxykinase 1 (PCK1)/adenosine monophosphate-activated protein kinase (AMPK) pathway. METHODS The expression of p300 and PCK1 at the messenger ribonucleic acid (mRNA) level was detected using a quantitative polymerase chain reaction. The GeneCards and GEPIA databases were used to investigate the relationship between p300 and PCK1. The MDA-MB-231/PTX cell line, known for its PTX resistance, was chosen to understand the specific role of p300 in such cells. The Lipofectamine™ 3000 reagent was used to transfer the p300 small interfering RNA and the overexpression of PCK1 plasmid into MDA-MB-231/PTX. The expression levels of p300, PCK1, 5'AMPK and phosphorylated AMPK (p-AMPK) were determined using the western blot test. RESULTS In TNBC cancer tissue, the expression of p300 was increased compared with TNBC paracancerous tissue (P < 0.05). In the MDA-MB-231 cell line of TNBC, the expression of p300 was lower than in the PTX-resistant TNBC cells (MDA-MB-231/PTX) (P < 0.05). The PCK1 expression was decreased in the TNBC cancer tissue compared with TNBC paracancerous tissue, and the PCK1 expression was reduced in MDA-MB-231/PTX than in MDA-MB-231 (P < 0.05) indicating that PCK1 was involved in the resistance function. Additionally, p-AMPK was decreased in MDA-MB-231/PTX compared with MDA-MB-231 (P < 0.05). The adenosine triphosphate (ATP) level was also detected and was significantly lower in MDA-MB-231/PTX than in MDA-MB-231 (P < 0.05). Additionally, cell proliferation increased significantly in MDA-MB-231/PTX at 48 and 72 h (P < 0.05) suggesting that MDA-MB-231/PTX cells obtained the resistance function which was associated with AMPK and ATP level. When p300 was inhibited, p-AMPK and ATP levels elevated in MDA-MB-231/PTX (P < 0.05). When PCK1 was suppressed, the ATP consumption rate decreased, and cell proliferation increased (P < 0.05). However, there were no changes in p300. CONCLUSIONS In MDA-MB-231/PTX, p300 can inhibit p-AMPK and ATP levels by inhibiting PCK1 expression. Our findings suggest that targeting p300 could modulate the PCK1/AMPK axis, offering a potential therapeutic avenue for overcoming PTX resistance in TNBC.
Collapse
Affiliation(s)
- Peng-Wei Zhao
- Laboratory of Microbiology and Immunology, School of Basic Medical Science, Inner Mongolia Medical University, No.5 Xinhua Street, Huimin District, Hohhot, 010059, China
| | - Jia-Xian Cui
- Laboratory of Microbiology and Immunology, School of Basic Medical Science, Inner Mongolia Medical University, No.5 Xinhua Street, Huimin District, Hohhot, 010059, China
| | - Xiu-Mei Wang
- Medical Oncology, Affiliated Cancer Hospital of Inner Mongolia Medical University, No. 42 Zhaowuda Road, Saihan District, Hohhot, 010020, Inner Mongolia, China.
| |
Collapse
|
9
|
Nie X, Fu L, Cheng Y, Wu X, Lv K, Li R, Wu Y, Leung GPH, Fu C, Lee SMY, Seto SW, Zhang J, Li J. Garcinone E suppresses breast cancer growth and metastasis by modulating tumor-associated macrophages polarization via STAT6 signaling. Phytother Res 2023; 37:4442-4456. [PMID: 37259475 DOI: 10.1002/ptr.7909] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 05/16/2023] [Accepted: 05/24/2023] [Indexed: 06/02/2023]
Abstract
Cancer metastasis remains the most common cause of death in breast cancer patients. Tumor-associated macrophages (TAMs) are a novel therapeutic target for the treatment of metastatic breast cancer. Despite the good anti-cancer activity of garcinone E (GE), there are no reports on its therapeutic effects on breast cancer metastasis. The objective of this study was to examine the anti-cancer effects of GE on metastatic breast cancer. RAW 264.7 and THP-1 cells were polarized to M2 macrophages by IL-4/IL-13 in vitro. A 4T1 mouse breast cancer model and the tail vein breast cancer metastasis model were used to explore the effect of GE on breast cancer growth and metastasis in vivo. In vitro studies showed that GE dose-dependently suppressed IL-4 + IL-13-induced expression of CD206 in both RAW 264.7 cells and differentiated THP-1 macrophages. However, GE did not affect the LPS + IFN-γ-induced polarization to the M1-like macrophages in vitro. GE inhibited the expression of the M2 macrophage specific genes in RAW 264.7 cells, and simultaneously impaired M2 macrophage-induced breast cancer cell proliferation and migration, and angiogenesis. In animal studies, GE significantly suppressed tumor growth, angiogenesis, and lung metastasis in 4T1 tumor-bearing mice, without causing toxicity. In both tumor and lung tissues, the proportion of M2-like TAMs was significantly decreased while the proportion of M1-like TAMs was markedly increased by GE treatment. Mechanistically, GE inhibited phosphorylation of STAT6 in vitro and in vivo. Our results demonstrate for the first time that GE suppresses breast cancer growth and pulmonary metastasis by modulating M2-like macrophage polarization through the STAT6 signaling pathway.
Collapse
Affiliation(s)
- Xin Nie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, SAR, China
| | - Li Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanfen Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoping Wu
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Kongpeng Lv
- Department of Interventional Radiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Renkai Li
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Yihan Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - George Pak-Heng Leung
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, SAR, China
| | - Sai-Wang Seto
- Department of Food Science and Nutrition, Faculty of Science, Hong Kong Polytechnic University, Hong Kong, China
- The Research Center for Chinese Medicine Innovation, Hong Kong Polytechnic University, Hong Kong, SAR, China
| | - Jinming Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingjing Li
- The Research Center for Chinese Medicine Innovation, Hong Kong Polytechnic University, Hong Kong, SAR, China
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong, SAR, China
| |
Collapse
|
10
|
Liang C, Zhang HY, Wang YQ, Yang LA, Du YS, Luo Y, Zhang TC, Xu Y. TMED2 Induces Cisplatin Resistance in Breast Cancer via Targeting the KEAP1-Nrf2 Pathway. Curr Med Sci 2023; 43:1023-1032. [PMID: 37615927 DOI: 10.1007/s11596-023-2777-7] [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: 03/13/2023] [Accepted: 06/13/2023] [Indexed: 08/25/2023]
Abstract
OBJECTIVE Cisplatin is the first-line treatment for breast cancer, but it faces challenges of drug resistance. This study investigated new molecular mechanisms underlying cisplatin resistance in breast cancer. METHODS We analyzed sequencing data from the TCGA database to identify potential associations between transmembrane emp24 protein transport domain containing 2 (TMED2) and breast cancer. Western blotting, real-time PCR, CCK-8, and TUNEL assays were used to measure the effects and molecular mechanism of TMED2 on cisplatin resistance in MCF-7 and MDA-MB-231 cell lines. RESULTS TMED2 was overexpressed in breast cancer and associated with poor prognosis. TMED2 increased cisplatin resistance in breast cancer cells in vitro via promoting ubiquitination of Kelch-like ECH-associated protein 1 (KEAP1), relieving inhibition of KEAP1 on nuclear factor erythroid 2-related factor 2 (Nrf2), and increasing expression of downstream drug resistance related genes, such as heme oxygenase 1 (HO-1) and NAD (P) H quinone oxidoreductase 1 (NQO1). CONCLUSION We identified a new molecular mechanism by which TMED2 affects cisplatin resistance in breast cancer. Our results provide theoretical guidance for future clinical applications.
Collapse
Affiliation(s)
- Chen Liang
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430070, China
| | - Han-Yong Zhang
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yi-Qian Wang
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430070, China
| | - Ling-Ang Yang
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430070, China
| | - Yu-Sen Du
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430070, China
| | - Ying Luo
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430070, China
| | - Tong-Cun Zhang
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430070, China
| | - Yao Xu
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430070, China.
| |
Collapse
|
11
|
Jin Y, Qiu L, Bao W, Lu M, Cao F, Ni H, Zhao B. High expression of IGHG1 promotes breast cancer malignant development by activating the AKT pathway. Cell Cycle 2023; 22:718-731. [PMID: 36404682 PMCID: PMC9980652 DOI: 10.1080/15384101.2022.2147141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 10/17/2022] [Accepted: 10/27/2022] [Indexed: 11/22/2022] Open
Abstract
This study researched the exact function of IgG1 heavy chain (IGHG1) on breast cancer (BC) progression. IGHG1 level within BC and paired normal tissues was acquired in Gene Expression Profiling Interactive Analysis dataset. Meanwhile, this work harvested tumor and paired healthy tissues in 42 BC cases. siRNA targeting IGHG1 was transfected into BC cells. SC79 was used to treat the transfected BC cells. CCK-8 assay, clone formation experiment, BrdU assay, Transwell experiment and flow cytometry were carried out to measure the viability, colony formation, proliferation, invasion, and apoptosis of BC cells. Paclitaxel and cisplatin sensitivity of BC cells was evaluated by MTT assay. Real-time quantitative reverse transcription-polymerase chain reaction and Western-blot were performed for measuring mRNA and protein expression. The overexpressed IGHG1 indicated dismal BC survival. IGHG1 silencing attenuated the viability, invasion, proliferation, epithelial-mesenchymal transition, but enhanced the apoptosis of BC cells. IGHG1 silencing enhanced the paclitaxel and cisplatin sensitivity of BC cells. IGHG1 silencing suppressed the activity of the MEK, AKT, and ERK pathways. AKT agonist partially reversed the inhibition of IGHG1 silencing on BC cell malignant phenotype and resistance to paclitaxel and cisplatin. IGHG1 promotes the malignant development of BC by activating the AKT pathway. It may be an effective target for BC treatment.
Collapse
Affiliation(s)
- Yongmei Jin
- Department of Nursing, The Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lin Qiu
- Department of General Surgery, The Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenqing Bao
- School of Medicine, Tongji University, Shanghai, China
| | - Minhao Lu
- Department of General Surgery, The Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Feng Cao
- Department of General Surgery, The Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hanchen Ni
- Department of Nursing, The Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bin Zhao
- Department of General Surgery, The Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
12
|
Yan B, Shi R, Lu YY, Fang DD, Ye MN, Zhou QM. Shenqi Fuzheng injection reverses M2 macrophage-mediated cisplatin resistance through the PI3K pathway in breast cancer. PLoS One 2023; 18:e0279752. [PMID: 36693064 PMCID: PMC9873177 DOI: 10.1371/journal.pone.0279752] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 12/14/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Shenqi Fuzheng injection (SQFZ) combined with chemotherapy can sensitize tumour cells. However, the mechanisms underlying SQFZ's effects remain unknown. In human breast cancer cell lines and M2 macrophages, we showed that SQFZ was a significantly potent agent of sensitization. METHODS The human breast cancer cell line, MDA-MB-231/DDP, and the human acute leukaemia mononuclear cell line, THP-1, were used. MDA-MB-231/DDP breast cancer xenografts were established to monitor tumour growth. Resistance-associated proteins were examined by western blotting. Levels of cytokines and chemokines were detected by ELISA. Cell viability was measured using the MTT assay. Apoptosis was detected by flow cytometric analysis. RESULTS SQFZ significantly enhanced the capability of cisplatin to reduce tumour mass. SQFZ and cisplatin decreased the expression of CD206 by 1.89-fold and increased that of CD86 by 1.76-fold as compared to cisplatin alone. The levels of PGE2, IL-6, and CCL1 decreased significantly, and the activation of p-PI3K and the expressions of P-gp and ABCG2 were also inhibited by SQFZ in combination with cisplatin treatment in vivo. The survival following cisplatin administration of 60 μM and 120 μM reduced significantly in the presence of SQFZ in MDA-MB-231/DDP and M2 co-cultured cells. IGF-1, a PI3K activator, combined with SQFZ weakened the effects of SQFZ-induced apoptosis from 28.7% to 10.5%. The effects of IGF-1 on increasing the expressions of P-gp, ABCG2, and Bcl-2, and decreasing that of Bax were reversed by SQFZ. CONCLUSION Our findings provide evidence that SQFZ is a potential therapeutic drug for cancer therapy.
Collapse
Affiliation(s)
- Bin Yan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rong Shi
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi-yu Lu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dong-dong Fang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mei-na Ye
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qian-mei Zhou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- * E-mail:
| |
Collapse
|
13
|
Alterations in the p53 isoform ratio govern breast cancer cell fate in response to DNA damage. Cell Death Dis 2022; 13:907. [PMID: 36307393 PMCID: PMC9616954 DOI: 10.1038/s41419-022-05349-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/07/2022]
Abstract
Our previous studies have shown that p53 isoform expression is altered in breast cancer and related to prognosis. In particular, a high ∆40p53:p53α ratio is associated with worse disease-free survival. In this manuscript, the influence of altered Δ40p53 and p53α levels on the response to standard of care DNA-damaging agents used in breast cancer treatment was investigated in vitro. Our results revealed that a high Δ40p53:p53α ratio causes cells to respond differently to doxorubicin and cisplatin treatments. Δ40p53 overexpression significantly impairs the cells' sensitivity to doxorubicin through reducing apoptosis and DNA damage, whereas Δ40p53 knockdown has the opposite effect. Further, a high Δ40p53:p53α ratio inhibited the differential expression of several genes following doxorubicin and promoted DNA repair, impairing the cells' canonical response. Overall, our results suggest that the response of breast cancer cells to standard of care DNA-damaging therapies is dependent on the expression of p53 isoforms, which may contribute to outcomes in breast cancer.
Collapse
|