1
|
Cortellesi E, Savini I, Veneziano M, Gambacurta A, Catani MV, Gasperi V. Decoding the Epigenome of Breast Cancer. Int J Mol Sci 2025; 26:2605. [PMID: 40141248 PMCID: PMC11942310 DOI: 10.3390/ijms26062605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/06/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Breast cancer (BC) is the most prevalent malignancy among women, characterized by extensive heterogeneity stemming from molecular and genetic alterations. This review explores the intricate epigenetic landscape of BC, highlighting the significant role of epigenetic modifications-particularly DNA methylation, histone modifications, and the influence of non-coding RNAs-in the initiation, progression, and prognosis of the disease. Epigenetic alterations drive crucial processes, including gene expression regulation, cell differentiation, and tumor microenvironment interactions, contributing to tumorigenesis and metastatic potential. Notably, aberrations in DNA methylation patterns, including global hypomethylation and hypermethylation of CpG islands, have been associated with distinct BC subtypes, with implications for early detection and risk assessment. Furthermore, histone modifications, such as acetylation and methylation, affect cancer cell plasticity and aggressiveness by profoundly influencing chromatin dynamics and gene transcription. Finally, non-coding RNAs contribute by modulating epigenetic machinery and gene expression. Despite advances in our knowledge, clinical application of epigenetic therapies in BC is still challenging, often yielding limited efficacy when used alone. However, combining epi-drugs with established treatments shows promise for enhancing therapeutic outcomes. This review underscores the importance of integrating epigenetic insights into personalized BC treatment strategies, emphasizing the potential of epigenetic biomarkers for improving diagnosis, prognosis, and therapeutic response in affected patients.
Collapse
Affiliation(s)
- Elisa Cortellesi
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (E.C.); (I.S.); (M.V.); (A.G.); (M.V.C.)
| | - Isabella Savini
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (E.C.); (I.S.); (M.V.); (A.G.); (M.V.C.)
| | - Matteo Veneziano
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (E.C.); (I.S.); (M.V.); (A.G.); (M.V.C.)
| | - Alessandra Gambacurta
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (E.C.); (I.S.); (M.V.); (A.G.); (M.V.C.)
- NAST Centre (Nanoscience & Nanotechnology & Innovative Instrumentation), Tor Vergata University of Rome, 00133 Rome, Italy
| | - Maria Valeria Catani
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (E.C.); (I.S.); (M.V.); (A.G.); (M.V.C.)
| | - Valeria Gasperi
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (E.C.); (I.S.); (M.V.); (A.G.); (M.V.C.)
| |
Collapse
|
2
|
Almutairi MH, Alrubie TM, Alshareeda AT, Albarakati N, Almotiri A, Alamri AM, Almutairi BO, Alanazi M. Differential expression and regulation of ADAD1, DMRTC2, PRSS54, SYCE1, SYCP1, TEX101, TEX48, and TMPRSS12 gene profiles in colon cancer tissues and their in vitro response to epigenetic drugs. PLoS One 2024; 19:e0307724. [PMID: 39208330 PMCID: PMC11361649 DOI: 10.1371/journal.pone.0307724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/10/2024] [Indexed: 09/04/2024] Open
Abstract
Colon cancer (CC) is a significant cause of death worldwide, particularly in Saudi Arabia. To increase the accuracy of diagnosis and treatment, it is important to discover new specific biomarkers for CC. The main objectives of this research are to identify potential specific biomarkers for the early diagnosis of CC by analyzing the expressions of eight cancer testis (CT) genes, as well as to analyze how epigenetic mechanisms control the expression of these genes in CC cell lines. Tissue samples were collected from 15 male patients with CC tissues and matched NC tissues for gene expression analysis. The expression levels of specific CT genes, including ADAD1, DMRTC2, PRSS54, SYCE1, SYCP1, TEX101, TEX48, and TMPRSS12, were assessed using quantitative techniques. To validate the gene expression patterns, we used publicly available CC statistics. To investigate the effect of inhibition of DNA methylation and histone deacetylation on CT gene expression, in vitro experiments were performed using HCT116 and Caco-2 cell lines. There was no detected expression of the genes neither in the patient samples nor in NC tissues, except for TEX48, which exhibited upregulation in CC samples compared to NC tissues in online datasets. Notably, CT genes showed expression in testis samples. In vitro, experiments demonstrated significant enhancement in mRNA expression levels of ADAD1, DMRTC2, PRSS54, SYCE1, SYCP1, TEX101, TEX48, and TMPRSS12 following treatment with 5-aza-2'-deoxycytidine and trichostatin A in HCT116 and Caco-2 cell lines. Epigenetic treatments modify the expression of CT genes, indicating that these genes can potentially be used as biomarkers for CC. The importance of conducting further research to understand and target epigenetic mechanisms to improve CC treatment cannot be overemphasized.
Collapse
Affiliation(s)
- Mikhlid H. Almutairi
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Turki M. Alrubie
- Laboratories Directorate, General Directorate of Animal Health, Ministry Deputyship for Agriculture, Ministry of Environment, Water and Agriculture, Riyadh, Saudi Arabia
| | - Alaa T. Alshareeda
- Blood and Cancer Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Nada Albarakati
- Blood and Cancer Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia
| | - Alhomidi Almotiri
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Ad Dawadmi, Saudi Arabia
| | - Abdullah M. Alamri
- Department of Biochemistry, Genome Research Chair, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Bader O. Almutairi
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad Alanazi
- Department of Biochemistry, Genome Research Chair, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
3
|
Ye J, Deng R, Wang X, Song S, Xu X, Zhang JY, Xu BB, Wang X, Yu JK. Intra-articular Histone Deacetylase Inhibitor Microcarrier Delivery to Reduce Osteoarthritis. NANO LETTERS 2023; 23:10832-10840. [PMID: 38009465 PMCID: PMC10722529 DOI: 10.1021/acs.nanolett.3c03037] [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: 08/12/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 11/29/2023]
Abstract
The histone deacetylase inhibitor (HDACi) was a milestone in the treatment of refractory T-cell lymphoma. However, the beneficial effects of HDACi have not been appreciated in osteoarthritis (OA). Herein, we implemented a microcarrier system because of the outstanding advantages of controlled and sustained release, biodegradability, and biocompatibility. The poly(d,l-lactide-co-glycolide) (PLGA) microcapsules have a regulated and sustained release profile with a reduced initial burst release, which can improve the encapsulation efficiency of the Chidamide. The emulsion solvent evaporation strategy was used to encapsulate Chidamide in PLGA microcapsules. The encapsulation of Chidamide was established by UV-vis spectra and scanning electron microscopy. Additionally, the inhibition of Tnnt3 and immune stimulation by Chidamide helped to inhibit cartilage destruction and prevent articular cartilage degeneration. Based on the results, the Chidamide in PLGA microcapsules provides a transformative therapeutic strategy for the treatment of osteoarthritis patients to relieve symptoms and protect against cartilage degeneration.
Collapse
Affiliation(s)
- Jing Ye
- Sports
Medicine Department, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, No. 49 North Garden Road, Haidian
District, Beijing 100191, China
- Institute
of Sports Medicine, Peking University, No. 49 North Garden Road, Haidian
District, Beijing 100191, China
| | - Ronghui Deng
- Sports
Medicine Department, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, No. 49 North Garden Road, Haidian
District, Beijing 100191, China
- Institute
of Sports Medicine, Peking University, No. 49 North Garden Road, Haidian
District, Beijing 100191, China
| | - Xinjie Wang
- Sports
Medicine Department, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, No. 49 North Garden Road, Haidian
District, Beijing 100191, China
- Institute
of Sports Medicine, Peking University, No. 49 North Garden Road, Haidian
District, Beijing 100191, China
| | - Shitang Song
- Sports
Medicine Department, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, No. 49 North Garden Road, Haidian
District, Beijing 100191, China
- Institute
of Sports Medicine, Peking University, No. 49 North Garden Road, Haidian
District, Beijing 100191, China
| | - Xiong Xu
- Beijing
National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Ji-Ying Zhang
- Sports
Medicine Department, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, No. 49 North Garden Road, Haidian
District, Beijing 100191, China
- Institute
of Sports Medicine, Peking University, No. 49 North Garden Road, Haidian
District, Beijing 100191, China
| | - Bing-bing Xu
- Sports
Medicine Department, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, No. 49 North Garden Road, Haidian
District, Beijing 100191, China
- Institute
of Sports Medicine, Peking University, No. 49 North Garden Road, Haidian
District, Beijing 100191, China
| | - Xing Wang
- Beijing
National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Jia-Kuo Yu
- Sports
Medicine Department, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, No. 49 North Garden Road, Haidian
District, Beijing 100191, China
- Institute
of Sports Medicine, Peking University, No. 49 North Garden Road, Haidian
District, Beijing 100191, China
| |
Collapse
|
4
|
Ediriweera MK. The Histone Deacetylase Inhibitory Potential of Chicken Egg Yolk Fat and Their Fatty Acid Composition. SCIENTIFICA 2023; 2023:6360487. [PMID: 37885471 PMCID: PMC10599844 DOI: 10.1155/2023/6360487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 10/28/2023]
Abstract
Histone deacetylation is a key biochemical event associated with transcriptional regulation. Histone deacetylases (HDACs) mediate the deacetylation of histones. Fatty acids have been reported to function as histone deacetylase inhibitors (HDACi). The present instigation reports the HDAC inhibitory activity of egg yolks and egg yolk-derived fat of country and farm chicken for the first time. Egg yolks and fatty acids derived from both country (CCEF) and farm chicken (FCEF) demonstrated significant HDAC enzyme activity inhibition. Furthermore, egg yolks, CCEF, and FCEF exhibited DPPH free radical scavenging effects. The analysis of fatty acid profiles revealed varying degrees of saturated, mono-, and polyunsaturated fatty acids in the egg yolks. Palmitic acid (C16 : 0) was found to be the most abundant saturated fatty acid in both CCEF and FCEF. Among the monounsaturated fatty acids, oleic acid (C18 : 1) was the most abundant in both CCEF and FCEF. In terms of polyunsaturated fatty acids, a significant difference was observed in the content of linoleic acid (C18 : 2), an omega-6 fatty acid, and docosahexaenoic acid (C22 : 6), an omega-3 fatty acid, between CCEF and FCEF. These findings present exciting prospects for the development of histone deacetylase inhibitors based on egg yolk fat.
Collapse
Affiliation(s)
- Meran Keshawa Ediriweera
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Colombo, Colombo 08, Sri Lanka
| |
Collapse
|
5
|
Tian Y, Chen Z, Wu P, Zhang D, Ma Y, Liu X, Wang X, Ding D, Cao X, Yu Y. MIR497HG-Derived miR-195 and miR-497 Mediate Tamoxifen Resistance via PI3K/AKT Signaling in Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204819. [PMID: 36815359 PMCID: PMC10131819 DOI: 10.1002/advs.202204819] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/14/2022] [Indexed: 05/28/2023]
Abstract
Tamoxifen is commonly used for the treatment of patients with estrogen receptor-positive (ER+) breast cancer, but the acquired resistance to tamoxifen presents a critical challenge of breast cancer therapeutics. Recently, long noncoding RNA MIR497HG and its embedded miR-497 and miR-195 are proved to play significant roles in many types of human cancers, but their roles in tamoxifen-resistant breast cancer remain unknown. The results indicate that MIR497HG deficiency induces breast cancer progression and tamoxifen resistance by inducing downregulation of miR-497/195. miR-497/195 coordinately represses five positive PI3K-AKT regulators (MAP2K1, AKT3, BCL2, RAF1, and CCND1), resulting in inhibition of PI3K-AKT signaling, and PI3K-AKT inhibition in tamoxifen-resistant cells restored tamoxifen responsiveness. Furthermore, ER α binds the MIR497HG promoter to activate its transcription in an estrogen-dependent manner. ZEB1 interacts with HDAC1/2 and DNMT3B at the MIR497HG promoter, resulting in promoter hypermethylation and histone deacetylation. The findings reveal that ZEB1-induced MIR497HG depletion contributes to breast cancer progression and tamoxifen resistance through PI3K-AKT signaling. MIR497HG can be used as a biomarker for predicting tamoxifen sensitivity in patients with ER+ breast cancer.
Collapse
Affiliation(s)
- Yao Tian
- The First Department of Breast CancerTianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical UniversityMinistry of EducationTianjin300060China
- Department of General SurgeryTianjin Medical University General HospitalTianjin300052China
| | - Zhao‐Hui Chen
- The First Department of Breast CancerTianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical UniversityMinistry of EducationTianjin300060China
| | - Peng Wu
- The First Department of Breast CancerTianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical UniversityMinistry of EducationTianjin300060China
| | - Di Zhang
- The First Department of Breast CancerTianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical UniversityMinistry of EducationTianjin300060China
| | - Yue Ma
- The First Department of Breast CancerTianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical UniversityMinistry of EducationTianjin300060China
| | - Xiao‐Feng Liu
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical UniversityMinistry of EducationTianjin300060China
| | - Xin Wang
- The First Department of Breast CancerTianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical UniversityMinistry of EducationTianjin300060China
| | - Dan Ding
- State Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive MaterialsMinistry of Educationand College of Life SciencesNankai UniversityTianjin300071China
| | - Xu‐Chen Cao
- The First Department of Breast CancerTianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical UniversityMinistry of EducationTianjin300060China
| | - Yue Yu
- The First Department of Breast CancerTianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical UniversityMinistry of EducationTianjin300060China
| |
Collapse
|
6
|
Keshawa Ediriweera M. Fatty acids as histone deacetylase inhibitors: old biochemistry tales in a new life sciences town. Drug Discov Today 2023; 28:103569. [PMID: 36990144 DOI: 10.1016/j.drudis.2023.103569] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023]
Abstract
Histone acetylation is a key epigenetic event. Although the keywords fatty acids, histones, and histone acetylation have a long history in biochemistry, these topics continue to attract much attention among researchers. The acetylation of histones is controlled by the activities of histone acetyltransferases (HATs) and histone deacetylases (HDACs). An imbalance in the activities of HATs and HDACs is common in a range of human cancers. Histone deacetylase inhibitors (HDACi) can restore dysregulated histone acetylation profiles in cancer cells and have been identified as promising anti-cancer therapeutics. Short-chain fatty acids mediate anti-cancer effects by inhibiting the activity of HDACs. Recent studies have identified odd-chain fatty acids as novel HDACi. This review summarizes recent findings regarding fatty acids as HDACi in cancer therapy. Teaser: Inhibition of histone deacetylase (HDAC) activity by fatty acids.
Collapse
|
7
|
The Role of Reprogrammed Glucose Metabolism in Cancer. Metabolites 2023; 13:metabo13030345. [PMID: 36984785 PMCID: PMC10051753 DOI: 10.3390/metabo13030345] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/19/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Cancer cells reprogram their metabolism to meet biosynthetic needs and to adapt to various microenvironments. Accelerated glycolysis offers proliferative benefits for malignant cells by generating glycolytic products that move into branched pathways to synthesize proteins, fatty acids, nucleotides, and lipids. Notably, reprogrammed glucose metabolism and its associated events support the hallmark features of cancer such as sustained cell proliferation, hijacked apoptosis, invasion, metastasis, and angiogenesis. Overproduced enzymes involved in the committed steps of glycolysis (hexokinase, phosphofructokinase-1, and pyruvate kinase) are promising pharmacological targets for cancer therapeutics. In this review, we summarize the role of reprogrammed glucose metabolism in cancer cells and how it can be manipulated for anti-cancer strategies.
Collapse
|
8
|
Feng X, Han H, Guo Y, Feng X, Guo S, Zhou W. LncRNA ENST869 Targeting Nestin Transcriptional Region to Affect the Pharmacological Effects of Chidamide in Breast Cancer Cells. Front Oncol 2022; 12:874343. [PMID: 35444938 PMCID: PMC9014306 DOI: 10.3389/fonc.2022.874343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 03/07/2022] [Indexed: 01/04/2023] Open
Abstract
Breast cancer is one of the leading threats to the health of women. It has the highest incidence and mortality in women worldwide. Although progress has been made in the development and application of anti-breast cancer drugs such as Chidamide and others, the occurrence of drug resistance limits the effective application of chemotherapies. The purpose of this study is to explore the role of LncRNA in the pharmacological effect of Chidamide in breast cancer therapy. The human breast cancer MCF-7 or MDA-MB-231 cells were used as the research cell models. The RNA library screening and high-throughput sequencing comparative analysis was conducted. The binding of LncRNA and its downstream target genes in RNA and protein levels was tested. The results showed that the expression of LncRNA ENST869 in cells treated with Chidamide increased significantly, as demonstrated by real-time PCR and cell viability assay. RNAplex analysis showed that LncRNA ENST869 and Nestin mRNA may interact. RNA interference and Western blot analysis indicated that LncRNA ENST869 could target and regulate the expression of Nestin. Luciferase assay and RNA-protein pulldown showed that LncRNA ENST869 affected Nestin transcription. There might be a highly active binding region of LncRNA ENST869 in regulating Nestin transcriptional activity within the site of 250 bp upstream of the transcription starting point of Nestin. In addition, LncRNA ENST869 did not directly interact with Nestin protein to affect its activity. In conclusion, our results demonstrated that LncRNA ENST869 could affect the function of Nestin in breast cancer cells treated with Chidamide. Nestin is a key player in influencing the pharmacological activity of Chidamide and an essential factor in drug resistance of breast cancer cells.
Collapse
Affiliation(s)
- Xiuyan Feng
- Medical Administration Division, The Second Affiliated Hospital of Shenyang Medical College, Shenyang City, China
| | - Han Han
- Department of Biochemistry and Molecular Biology, Shenyang Medical College, Shenyang City, China
| | - Yarui Guo
- Department of Pathogen Biology, Shenyang Medical College, Shenyang City, China
| | - Xue Feng
- Department of Pathogen Biology, Shenyang Medical College, Shenyang City, China
| | - Shanchun Guo
- RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA, United States
| | - Weiqiang Zhou
- Department of Pathogen Biology, Shenyang Medical College, Shenyang City, China
| |
Collapse
|
9
|
Hu R, Yu Y, Wang H. The LMCD1-AS1/miR-526b-3p/OSBPL5 axis promotes cell proliferation, migration and invasion in non-small cell lung cancer. BMC Pulm Med 2022; 22:30. [PMID: 35000595 PMCID: PMC8744214 DOI: 10.1186/s12890-022-01820-7] [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: 10/25/2021] [Accepted: 12/31/2021] [Indexed: 11/10/2022] Open
Abstract
PURPOSE To explore the specific role and regulatory mechanism of oxysterol binding protein like 5 (OSBPL5) in non-small cell lung cancer (NSCLC). METHODS AND RESULTS Quantitative real-time polymerase chain reaction (qRT-PCR) analysis demonstrated that OSBPL5 expression was notably elevated in NSCLC tissues and cell lines, and Kaplan-Meier analysis manifested that high OSBPL5 expression was closely related to the poor prognosis of NSCLC patients. Besides, according to the results from western blot analysis, cell counting kit-8, EdU and Transwell assays, knockdown of OSBPL5 suppressed NSCLC cell proliferation, migration, invasion and epithelial-mesenchymal transition (EMT) process. Additionally, by performing qRT-PCR analysis, luciferase reporter and RNA pull-down assays, we verified that OSBPL5 was a downstream target of miR-526b-3p and long noncoding RNA (lncRNA) LMCD1-AS1 served as a sponge for miR-526b-3p. Moreover, from rescue assays, we observed that OSBPL5 overexpression offset LMCD1-AS1 knockdown-mediated inhibition in cell proliferation, migration, invasion and EMT in NSCLC. CONCLUSIONS This paper was the first to probe the molecular regulatory mechanism of OSBPL5 involving the LMCD1-AS1/miR-526b-3p axis in NSCLC and our results revealed that the LMCD1-AS1/miR-526b-3p/OSBPL5 axis facilitates NSCLC cell proliferation, migration, invasion and EMT, which may offer a novel therapeutic direction for NSCLC.
Collapse
Affiliation(s)
- Rui Hu
- Department of Thoracic Surgery, Shengli Oilfield Central Hospital, 31 Jinan Road, Dongying, 257034, Shandong, China
| | - Yankai Yu
- Department of Thoracic Surgery, Shengli Oilfield Central Hospital, 31 Jinan Road, Dongying, 257034, Shandong, China
| | - Haining Wang
- Department of Thoracic Surgery, Shengli Oilfield Central Hospital, 31 Jinan Road, Dongying, 257034, Shandong, China.
| |
Collapse
|
10
|
Ding R, Duan Z, Yang M, Wang X, Li D, Kan Q. High miR-3609 expression is associated with better prognosis in TNBC based on mining using systematic integrated public sequencing data. Exp Ther Med 2021; 23:54. [PMID: 34934431 PMCID: PMC8652383 DOI: 10.3892/etm.2021.10976] [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: 05/25/2021] [Accepted: 09/09/2021] [Indexed: 12/09/2022] Open
Abstract
MicroRNAs (miRNAs/miRs) are small endogenous RNAs that regulate gene expression post-transcriptionally. Abnormal miR-3609 expression is associated with the occurrence of pancreatic cancer, glioma and other diseases, such as polycystic ovary syndrome. However, the prognostic potential of miR-3609 has been reported in breast cancer. Thus, the present study aimed to investigate the differential expression and prognostic value of miR-3609 in patients with breast cancer from the UALCAN, cBioportal and Kaplan-Meier Plotter databases, respectively. Furthermore, the co-expression genes of miR-3609 in breast cancer were investigated using data from the LinkedOmics database, and functional enrichment analysis was performed using the LinkInterpreter module in LinkedOmics. The co-expression gene network was constructed using the Search Tool for the Retrieval of Interacting Genes/Proteins database, and the cytoHubba plug-in was used to identify the hub genes, which were visualized using Cytoscape software. The prognoses of the hub genes were performed using the Kaplan-Meier Plotter database. The Cell Counting Kit-8 and cell cycle assays were performed to confirm the functions of miR-3609 mimics transfection in MDA-MB-231 cells. Survival analysis using the Kaplan-Meier Plotter database demonstrated that high miR-3609 expression in triple-negative breast cancer (TNBC) was associated with a better prognosis. Furthermore, the experimental results indicated that high miR-3609 expression inhibited the proliferation of TNBC cells and induced cell cycle arrest of TNBC cells in the G0/G1 phase. Taken together, the results of the present study suggest that miR-3609 plays a vital role in mediating cell cycle arrest and inhibiting the proliferation of TNBC cells.
Collapse
Affiliation(s)
- Rumeng Ding
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zhenfeng Duan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China.,Sarcoma Biology Laboratory, Department of Orthopedic Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Meng Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xinru Wang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Duolu Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Quancheng Kan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
11
|
Zhou M, Yuan M, Zhang M, Lei C, Aras O, Zhang X, An F. Combining histone deacetylase inhibitors (HDACis) with other therapies for cancer therapy. Eur J Med Chem 2021; 226:113825. [PMID: 34562854 DOI: 10.1016/j.ejmech.2021.113825] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/16/2021] [Accepted: 08/20/2021] [Indexed: 12/30/2022]
Abstract
Histone deacetylases (HDACs) play an important role in regulating the expression of genes involved in tumorigenesis and tumor maintenance, and hence they have been considered as key targets in cancer therapy. As a novel category of antitumor agents, histone deacetylase inhibitors (HDACis) can induce cell cycle arrest, apoptosis, and differentiation in cancer cells, ultimately combating cancer. Although in the United States, the use of HDACis for the treatment of certain cancers has been approved, the therapeutic efficacy of HDACis as a single therapeutic agent in solid tumorshas been unsatisfactory and drug resistance may yet occur. To enhance therapeutic efficacy and limit drug resistance, numerous combination therapies involving HDACis in synergy with other antitumor therapies have been studied. In this review, we describe the classification of HDACs. Moreover, we summarize the antitumor mechanism of the HDACis for targeting key cellular processes of cancers (cell cycle, apoptosis, angiogenesis, DNA repair, and immune response). In addition, we outline the major developments of other antitumor therapies in combination with HDACis, including chemotherapy, radiotherapy, phototherapy, targeted therapy, and immunotherapy. Finally, we discuss the current state and challenges of HDACis-drugs combinations in future clinical studies, with the aim of optimizing the antitumor effect of such combinations.
Collapse
Affiliation(s)
- Mengjiao Zhou
- Department of Pharmacology, School of Pharmacy, Nantong University, 226000, Nantong, Jiangsu, PR China
| | - Minjian Yuan
- Department of Pharmacology, School of Pharmacy, Nantong University, 226000, Nantong, Jiangsu, PR China
| | - Meng Zhang
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Chenyi Lei
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Omer Aras
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, United States
| | - Xiaohong Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, PR China.
| | - Feifei An
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, 710061, Shaanxi, People's Republic of China; Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, PR China.
| |
Collapse
|
12
|
Akbarzadeh M, Mihanfar A, Akbarzadeh S, Yousefi B, Majidinia M. Crosstalk between miRNA and PI3K/AKT/mTOR signaling pathway in cancer. Life Sci 2021; 285:119984. [PMID: 34592229 DOI: 10.1016/j.lfs.2021.119984] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/09/2021] [Accepted: 09/19/2021] [Indexed: 01/07/2023]
Abstract
Phosphoinositide-3 kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) signaling pathway is one of the most important proliferative signaling pathways with critical undeniable function in various aspects of cancer initiation/progression, including proliferation, apoptosis, metastasis, angiogenesis, and drug resistance. On the other hand, numerous genetic alterations in the key genes involved in the PI3K/AKT/mTOR signaling pathway have been identified in multiple solid and hematological tumors. In addition, accumulating recent evidences have demonstrated a reciprocal interaction between this signaling pathway and microRNAs, a large group of small non-coding RNAs. Therefore, in this review, it was attempted to discuss about the interaction between key components of PI3K/AKT/mTOR signaling pathway with various miRNAs and their importance in cancer biology.
Collapse
Affiliation(s)
- Maryam Akbarzadeh
- Department of biochemistry, Urmia University of Medical Sciences, Urmia, Iran
| | - Ainaz Mihanfar
- Department of biochemistry, Urmia University of Medical Sciences, Urmia, Iran
| | - Shabnam Akbarzadeh
- Department of Physical Education and Sport Medicine, University of Tabriz, Tabriz, Iran
| | - Bahman Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
13
|
Liu A, Zhou K, Martínez MA, Lopez-Torres B, Martínez M, Martínez-Larrañaga MR, Wang X, Anadón A, Ares I. A "Janus" face of the RASSF4 signal in cell fate. J Cell Physiol 2021; 237:466-479. [PMID: 34553373 DOI: 10.1002/jcp.30592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 09/06/2021] [Accepted: 09/11/2021] [Indexed: 12/19/2022]
Abstract
RASSF4 (Ras-association domain family 4) is a protein-coding gene, regarded as a tumor suppressor regulated by DNA methylation. However, RASSF4 acts as a "Janus" in cell fate: death and survival. This review article focuses on the regulatory mechanisms of RASSF4 on cell death and cell survival and puts forward a comprehensive analysis of the relevant signaling pathways. The participation of RASSF4 in the regulation of intracellular store-operated Ca2+ entry also affects cell survival. Moreover, the mechanism of inducing abnormal expression of RASSF4 was summarized. We highlight recent advances in our knowledge of RASSF4 function in the development of cancer and other clinical diseases, which may provide insight into the controversial functions of RASSF4 and its potential application in disease therapy.
Collapse
Affiliation(s)
- Aimei Liu
- Department of National Reference, Laboratory of Veterinary Drug Residues (HZAU) and MOA Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, China.,Department of MOA, Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Wuhan, Hubei, China
| | - Kaixiang Zhou
- Department of National Reference, Laboratory of Veterinary Drug Residues (HZAU) and MOA Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, China.,Department of MOA, Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Wuhan, Hubei, China
| | - María Aránzazu Martínez
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid(UCM), and Research Institute Hospital 12 de October (i+12), Madrid, Spain
| | - Bernardo Lopez-Torres
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid(UCM), and Research Institute Hospital 12 de October (i+12), Madrid, Spain
| | - Marta Martínez
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid(UCM), and Research Institute Hospital 12 de October (i+12), Madrid, Spain
| | - María-Rosa Martínez-Larrañaga
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid(UCM), and Research Institute Hospital 12 de October (i+12), Madrid, Spain
| | - Xu Wang
- Department of National Reference, Laboratory of Veterinary Drug Residues (HZAU) and MOA Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, China.,Department of MOA, Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Wuhan, Hubei, China
| | - Arturo Anadón
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid(UCM), and Research Institute Hospital 12 de October (i+12), Madrid, Spain
| | - Irma Ares
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid(UCM), and Research Institute Hospital 12 de October (i+12), Madrid, Spain
| |
Collapse
|
14
|
Ediriweera MK, To NB, Lim Y, Cho SK. Odd-chain fatty acids as novel histone deacetylase 6 (HDAC6) inhibitors. Biochimie 2021; 186:147-156. [PMID: 33965456 DOI: 10.1016/j.biochi.2021.04.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/30/2021] [Accepted: 04/30/2021] [Indexed: 01/26/2023]
Abstract
The dysregulation of histone deacetylases (HDACs) is closely associated with tumorigenesis and has emerged as a promising target for anti-cancer drugs. Some odd-chain fatty acids are present in trace levels in human tissue. Despite limited health benefits, there is increasing experimental evidence of nutritional benefits of odd-chain fatty acids. This study examines the effects of five odd-chain fatty acids (valeric, heptanoic, nonanoic, undecanoic, and pentadecanoic acid) as novel HDAC6 inhibitors. Examination of these fatty acids on the proliferation and clonogenic ability in various cancer cell lines revealed that pentadecanoic and undecanoic acid can strongly inhibit cancer cell proliferation. Heptanoic and nonanoic acid showed moderate anti-proliferative effects, while valeric acid demonstrated weak anti-proliferative effects. HDAC6 inhibitory activities were in the order of pentadecanoic acid (C15:0) > undecanoic acid (C11:0) > nonanoic acid (C9:0) > heptanoic acid (C7:0) > valeric acid (C5:0), consistent with the anti-proliferative assay results. All of these fatty acids promoted the acetylation of α-tubulin in MCF-7 breast and A549 lung cancer cells dose-dependently. In-silico molecular docking analysis showed that increasing the aliphatic carbon chain length facilitates binding to HDAC6 residues, which might be important for the inhibitory potential of HDAC6. This study shows the potential utility of odd-chain fatty acids for epigenetic-based cancer therapy.
Collapse
Affiliation(s)
| | - Ngoc Bao To
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, 63243, South Korea.
| | - Yoongho Lim
- Department of Biological Sciences, Konkuk University, Seoul, 05029, Republic of Korea.
| | - Somi Kim Cho
- Subtropical/Tropical Organism Gene Bank, Jeju National University, Jeju, 63243, South Korea; Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, 63243, South Korea; Faculty of Biotechnology, College of Applied Life Sciences, SARI, Jeju National University, Jeju, 63243, Republic of Korea.
| |
Collapse
|
15
|
Dong SS, Dong DD, Yang ZF, Zhu GQ, Gao DM, Chen J, Zhao Y, Liu BB. Exosomal miR-3682-3p Suppresses Angiogenesis by Targeting ANGPT1 via the RAS-MEK1/2-ERK1/2 Pathway in Hepatocellular Carcinoma. Front Cell Dev Biol 2021; 9:633358. [PMID: 33869178 PMCID: PMC8044774 DOI: 10.3389/fcell.2021.633358] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/04/2021] [Indexed: 12/24/2022] Open
Abstract
Background Angiogenesis is a crucial process in tumorigenesis and development. The role of exosomes derived from hepatocellular carcinoma (HCC) cells in angiogenesis has not been clearly elucidated. Methods and Results Exosomes were isolated from HCC cell lines (HCCLM3, MHCC97L, and PLC/RFP/5) by ultracentrifugation and identified by nano transmission electron microscopy (TEM), NanoSight analysis and western blotting, respectively. In vitro and in vivo analyses showed that exosomes isolated from highly metastatic HCC cells enhanced the migration, invasion and tube formation of human umbilical vein endothelial cells (HUVECs) compared to exosomes derived from poorly metastatic HCC cells. In addition, microarray analysis of HCC-Exos was conducted to identify potential functional molecules, and miR-3682-3p expression was found to be significantly downregulated in exosomes isolated from highly metastatic HCC cells. By in vitro gain-of-function experiments, we found that HCC cells secreted exosomal miR-3682-3p, which negatively regulates angiopoietin-1 (ANGPT1), and this led to inhibition of RAS-MEK1/2-ERK1/2 signaling in endothelial cells and eventually impaired angiogenesis. Conclusion Our study elucidates that exosomal miR-3682-3p attenuates angiogenesis by targeting ANGPT1 through RAS-MEK1/2-ERK1/2 signaling and provides novel potential targets for liver cancer therapy.
Collapse
Affiliation(s)
- Shuang-Shuang Dong
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Dan-Dan Dong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhang-Fu Yang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Gui-Qi Zhu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Dong-Mei Gao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Jie Chen
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Yan Zhao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Bin-Bin Liu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| |
Collapse
|
16
|
Markouli M, Strepkos D, Basdra EK, Papavassiliou AG, Piperi C. Prominent Role of Histone Modifications in the Regulation of Tumor Metastasis. Int J Mol Sci 2021; 22:2778. [PMID: 33803458 PMCID: PMC7967218 DOI: 10.3390/ijms22052778] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/08/2021] [Indexed: 12/22/2022] Open
Abstract
Tumor aggressiveness and progression is highly dependent on the process of metastasis, regulated by the coordinated interplay of genetic and epigenetic mechanisms. Metastasis involves several steps of epithelial to mesenchymal transition (EMT), anoikis resistance, intra- and extravasation, and new tissue colonization. EMT is considered as the most critical process allowing cancer cells to switch their epithelial characteristics and acquire mesenchymal properties. Emerging evidence demonstrates that epigenetics mechanisms, DNA methylation, histone modifications, and non-coding RNAs participate in the widespread changes of gene expression that characterize the metastatic phenotype. At the chromatin level, active and repressive histone post-translational modifications (PTM) in association with pleiotropic transcription factors regulate pivotal genes involved in the initiation of the EMT process as well as in intravasation and anoikis resistance, playing a central role in the progression of tumors. Herein, we discuss the main epigenetic mechanisms associated with the different steps of metastatic process, focusing in particular on the prominent role of histone modifications and the modifying enzymes that mediate transcriptional regulation of genes associated with tumor progression. We further discuss the development of novel treatment strategies targeting the reversibility of histone modifications and highlight their importance in the future of cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | - Christina Piperi
- Correspondence: (A.G.P.); (C.P.); Tel.: +30-210-7462610 (C.P.); Fax: +30-210-7462703 (C.P.)
| |
Collapse
|
17
|
Turner N, Abeysinghe P, Sadowski P, Mitchell MD. Exosomal Cargo May Hold the Key to Improving Reproductive Outcomes in Dairy Cows. Int J Mol Sci 2021; 22:ijms22042024. [PMID: 33670752 PMCID: PMC7922264 DOI: 10.3390/ijms22042024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/15/2021] [Accepted: 02/15/2021] [Indexed: 12/25/2022] Open
Abstract
The reproductive status of dairy cows remains a challenge for dairy farmers worldwide, with impaired fertility linked to a significant reduction in herd profitability, due in part to impaired immunity, increased metabolic pressure, and longer postpartum anestrous interval (PPAI). Exosomes are nanovesicles released from a variety of cell types and end up in circulation, and carry proteins, bioactive peptides, lipids, and nucleic acids specific to the place of origin. As such, their role in health and disease has been investigated in humans and animals. This review discusses research into exosomes in the context of reproduction in dairy herds and introduces recent advances in mass-spectrometry (MS) based proteomics that have a potential to advance quantitative profiling of exosomal protein cargo in a search for early biomarkers of cattle fertility.
Collapse
Affiliation(s)
- Natalie Turner
- Institute of Health and Biomedical Innovation—Centre for Children’s Health Research, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4029, Australia; (N.T.); (P.A.)
| | - Pevindu Abeysinghe
- Institute of Health and Biomedical Innovation—Centre for Children’s Health Research, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4029, Australia; (N.T.); (P.A.)
| | - Pawel Sadowski
- Central Analytical Research Facility—Queensland University of Technology, Gardens Point, Brisbane, QLD 4000, Australia;
| | - Murray D. Mitchell
- Institute of Health and Biomedical Innovation—Centre for Children’s Health Research, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4029, Australia; (N.T.); (P.A.)
- Correspondence: ; Tel.: +61-7-3069-7438
| |
Collapse
|
18
|
Xu C, Yu H, Yin X, Zhang J, Liu C, Qi H, Liu P. Circular RNA circNINL promotes breast cancer progression through activating β-catenin signaling via miR-921/ADAM9 axis. J Biochem 2021; 169:693-700. [PMID: 33479730 DOI: 10.1093/jb/mvab005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 01/08/2021] [Indexed: 01/22/2023] Open
Abstract
We investigated the expression and functions of circular RNA (circRNA) circNINL and miR-921 in breast cancer (BC) in this study. We found that the expression of circNINL increased while the expression of miR-921 decreased in BC tissues and cell lines, and their anomalous expressions were associated with malignant features and poor prognostic of BC. Then, we demonstrated that circNINL could interact with miR-921 and facilitate BC cells malignant process including proliferation acceleration, migration enhancement and apoptosis evasion via sponging miR-921 in vitro. Further investigations revealed that circNINL/miR-921 axis could mediate the expression of ADAM9 which was a direct target of miR-921. In addition, we exhibited that ADAM9 may activate β-catenin signaling by interacting with E-cadherin. We presented the vital roles of circNINL/miR-921/ADAM9/β-catenin signaling in the progression of BC.
Collapse
Affiliation(s)
- Chuanbo Xu
- Department of General Surgery, The Huangdao District People's Hospital, Qingdao, Shandong, China
| | - Haitao Yu
- Department of General Surgery, The Huangdao District People's Hospital, Qingdao, Shandong, China
| | - Xianghua Yin
- Department of General Surgery, The Huangdao District People's Hospital, Qingdao, Shandong, China
| | - Jishi Zhang
- Department of General Surgery, The Huangdao District People's Hospital, Qingdao, Shandong, China
| | - Chunlin Liu
- Department of General Surgery, The Huangdao District People's Hospital, Qingdao, Shandong, China
| | - Hong Qi
- Department of General Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Peng Liu
- Department of General Surgery, The Huangdao District People's Hospital, Qingdao, Shandong, China
| |
Collapse
|
19
|
Qattan A. Novel miRNA Targets and Therapies in the Triple-Negative Breast Cancer Microenvironment: An Emerging Hope for a Challenging Disease. Int J Mol Sci 2020; 21:ijms21238905. [PMID: 33255471 PMCID: PMC7727826 DOI: 10.3390/ijms21238905] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/15/2020] [Accepted: 11/20/2020] [Indexed: 12/11/2022] Open
Abstract
Treatment of triple-negative breast cancer (TNBC) remains challenging because of the heterogeneity of the disease and lack of single targetable driving mutations. TNBC does not rely on estrogen, progesterone or epidermal growth factor receptors and is associated with aggressive disease progression and poor prognosis. TNBC is also characterized by resistance to chemotherapeutics, and response to immunotherapies is limited despite promising results in a subset of TNBC patients. MicroRNAs (miRNAs) have emerged as significant drivers of tumorigenesis and tumor progression in triple-negative breast cancer (TNBC) and present unique opportunities to target various components of the TNBC microenvironment for improved efficacy against this difficult to treat cancer. Effects of miRNAs on multiple targets may improve response rates in the context of this genetically and biologically heterogeneous disease. In this review, we offer a comprehensive view of miRNA regulation in TNBC, treatment challenges presented by TNBC in the context of the tumor microenvironment and stem cell subpopulations, and current and emerging miRNA-based therapeutic strategies targeting various components of the TNBC microenvironment. In addition, we offer insight into novel targets that have potential for treating TNBC through multiple mechanisms in the tumor microenvironment simultaneously and those that may be synergistic with standard chemotherapies.
Collapse
Affiliation(s)
- Amal Qattan
- Breast Cancer Research Unit, Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; or
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences (SMHS), George Washington University, Washington, DC 20073, USA
| |
Collapse
|
20
|
Zhuang J, Huo Q, Yang F, Xie N. Perspectives on the Role of Histone Modification in Breast Cancer Progression and the Advanced Technological Tools to Study Epigenetic Determinants of Metastasis. Front Genet 2020; 11:603552. [PMID: 33193750 PMCID: PMC7658393 DOI: 10.3389/fgene.2020.603552] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022] Open
Abstract
Metastasis is a complex process that involved in various genetic and epigenetic alterations during the progression of breast cancer. Recent evidences have indicated that the mutation in the genome sequence may not be the key factor for increasing metastatic potential. Epigenetic changes were revealed to be important for metastatic phenotypes transition with the development in understanding the epigenetic basis of breast cancer. Herein, we aim to present the potential epigenetic drivers that induce dysregulation of genes related to breast tumor growth and metastasis, with a particular focus on histone modification including histone acetylation and methylation. The pervasive role of major histone modification enzymes in cancer metastasis such as histone acetyltransferases (HAT), histone deacetylases (HDACs), DNA methyltransferases (DNMTs), and so on are demonstrated and further discussed. In addition, we summarize the recent advances of next-generation sequencing technologies and microfluidic-based devices for enhancing the study of epigenomic landscapes of breast cancer. This feature also introduces several important biotechnologists for identifying robust epigenetic biomarkers and enabling the translation of epigenetic analyses to the clinic. In summary, a comprehensive understanding of epigenetic determinants in metastasis will offer new insights of breast cancer progression and can be achieved in the near future with the development of innovative epigenomic mapping tools.
Collapse
Affiliation(s)
- Jialang Zhuang
- Biobank, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Qin Huo
- Biobank, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Fan Yang
- Biobank, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Ni Xie
- Biobank, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
21
|
Ediriweera MK, Moon JY, Nguyen YTK, Cho SK. 10-Gingerol Targets Lipid Rafts Associated PI3K/Akt Signaling in Radio-Resistant Triple Negative Breast Cancer Cells. Molecules 2020; 25:E3164. [PMID: 32664351 PMCID: PMC7397170 DOI: 10.3390/molecules25143164] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 12/14/2022] Open
Abstract
10-gingerol is a major phenolic lipid found in the rhizomes of ginger (Zingiber officinale). Being amphiphilic in nature, phenolic lipids have the ability to incorporate into cell membranes and modulate membrane properties. The purpose of the present study was to evaluate the effects of 10-gingerol on lipid raft/membrane raft modulation in radio-resistant triple negative breast cancer (MDA-MB-231/IR) cells. The effects of 10-gingerol on MDA-MB-231/IR cells' proliferation, clonogenic growth, migration, and invasion were assayed using MTT, colony formation, cell migration, and invasion assays, respectively. Sucrose density gradient centrifugation was used to extract lipid rafts. Western blotting and immunofluorescence were employed to assess the effects of 10-gingerol on lipid raft/membrane raft modulation and lipid rafts-associated PI3K/Akt signaling. Cholesterol measurements were carried out using a commercially available kit. 10-gingerol suppressed the proliferation, migration, invasion, and induced apoptosis through targeting the PI3K/Akt signaling pathway in MDA-MB-231/IR cells. Moreover, 10-gingerol was found to modulate the lipid rafts of MDA-MB-231/IR cells and attenuate the key PI3K/Akt signaling components in lipid rafts. The cholesterol content of the lipid rafts and rafts-resident Akt signaling were also affected by exposure to 10-gingerol. The results of the present study highlight rafts-associated PI3K/Akt signaling as a new target of 10-gingerol in MDA-MB-231/IR cells, thus rationalizing a new rafts-mediated treatment approach for radio-resistant triple negative breast cancer cells.
Collapse
Affiliation(s)
- Meran Keshawa Ediriweera
- Subtropical/tropical organism gene bank, Jeju National University, Jeju 63243, Korea or (M.K.E.); (J.Y.M.)
| | - Jeong Yong Moon
- Subtropical/tropical organism gene bank, Jeju National University, Jeju 63243, Korea or (M.K.E.); (J.Y.M.)
| | - Yen Thi-Kim Nguyen
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju 63243, Korea;
| | - Somi Kim Cho
- Subtropical/tropical organism gene bank, Jeju National University, Jeju 63243, Korea or (M.K.E.); (J.Y.M.)
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju 63243, Korea;
- Faculty of Biotechnology, College of Applied Life Sciences, SARI, Jeju National University, Jeju 63243, Korea
| |
Collapse
|
22
|
Aziz MH, Ahmad A. Epigenetic basis of cancer drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:113-116. [PMID: 35582041 PMCID: PMC9094056 DOI: 10.20517/cdr.2020.06] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 01/31/2020] [Indexed: 06/15/2023]
Affiliation(s)
- Moammir H. Aziz
- James H. Quillen VA Medical Center, Johnson City, TN 37604, USA
| | - Aamir Ahmad
- School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35205, USA
| |
Collapse
|