1
|
Xu S, Wang L, Zhao Y, Mo T, Wang B, Lin J, Yang H. Metabolism-regulating non-coding RNAs in breast cancer: roles, mechanisms and clinical applications. J Biomed Sci 2024; 31:25. [PMID: 38408962 PMCID: PMC10895768 DOI: 10.1186/s12929-024-01013-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/17/2024] [Indexed: 02/28/2024] Open
Abstract
Breast cancer is one of the most common malignancies that pose a serious threat to women's health. Reprogramming of energy metabolism is a major feature of the malignant transformation of breast cancer. Compared to normal cells, tumor cells reprogram metabolic processes more efficiently, converting nutrient supplies into glucose, amino acid and lipid required for malignant proliferation and progression. Non-coding RNAs(ncRNAs) are a class of functional RNA molecules that are not translated into proteins but regulate the expression of target genes. NcRNAs have been demonstrated to be involved in various aspects of energy metabolism, including glycolysis, glutaminolysis, and fatty acid synthesis. This review focuses on the metabolic regulatory mechanisms and clinical applications of metabolism-regulating ncRNAs involved in breast cancer. We summarize the vital roles played by metabolism-regulating ncRNAs for endocrine therapy, targeted therapy, chemotherapy, immunotherapy, and radiotherapy resistance in breast cancer, as well as their potential as therapeutic targets and biomarkers. Difficulties and perspectives of current targeted metabolism and non-coding RNA therapeutic strategies are discussed.
Collapse
Affiliation(s)
- Shiliang Xu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Lingxia Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Yuexin Zhao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Tong Mo
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Bo Wang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Jun Lin
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, People's Republic of China.
| | - Huan Yang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, People's Republic of China.
| |
Collapse
|
2
|
Chen Y, Gong Y, Qin H, Wei S, Wei Y, Yu Y, Lin X, Shuai P, Wang T, Guo C, Wang Q, Li G, Meng L, Yi L. MDM2-p53 mediate a miR-181c-3p/LIF axis to regulate low dose-rate radiation-induced DNA damage in human B lymphocytes. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 270:115848. [PMID: 38134636 DOI: 10.1016/j.ecoenv.2023.115848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/18/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023]
Abstract
PURPOSE Prolonged exposure to low dose-rate radiation (LDRR) is of growing concern to public health. Recent evidences indicates that LDRR causes deleterious health effects and is closely related to miRNAs. The aim of our study is to investigate the relationship between miRNAs and DNA damage caused by LDRR. MATERIALS AND METHODS In this study, we irradiated C57BL/6J mice with 12.5μGy/h dose of γ ray emitted from uranium ore for 8 h a day for 120 days at a total dose of 12 mGy, and identified differentially expressed miRNAs from the mice long-term exposed to LDRR through isolating serum RNAs, constructing small RNA library, Illumina sequencing. To further investigate the role of differential miRNA under LDRR,we first built DNA damage model in Immortal B cells irradiated with 12.5μGy/h dose of γ ray for 28 days at a total dose of 9.4 mGy. Then, we chose the highly conserved miR-181c-3p among 12 miRNA and its mechanism in alleviating DNA damage induced by LDRR was studied by transfection, quantitative PCR, luciferase assay, and Western blot. RESULTS AND CONCLUSIONS We have found that 12 differentially expressed miRNAs including miR-181c-3p in serum isolated from irradiated mice. Analysis of GO and KEGG indicated that target genes of theses 12 miRNA enriched in pathways related to membrane, protein binding and cancer. Long-term exposure to LDRR induced upregulation of gamma-H2A histone family member X (γ-H2AX) expression, a classical biomarker for DNA damage in B cells. miR-181c-3p inhibited Leukemia inhibitory factor (LIF) expression via combining its 3'UTR. LIF, MDM2, p53, and p-p53-s6 were upregulated after exposure to LDRR. In irradiated B cells, Transfection of miR-181c-3p reduced γ-H2AX expression and suppressed LIF and MDM2 protein levels, whereas p-p53-s6 expression was increased. As expected, the effect of LIF inhibition on irradiated B cells was similar to miR-181c-3p overexpression. Our results suggest that LDRR alters miRNA expression and induces DNA damage. Furthermore, miR-181c-3p can alleviate LDRR-induced DNA damage via the LIF/MDM2/p-p53-s6 pathway in human B lymphocytes. This could provide the basis for prevention and treatment of LDRR injury.
Collapse
Affiliation(s)
- Yonglin Chen
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yaqi Gong
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Hui Qin
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Shuang Wei
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yuanyun Wei
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yueqiu Yu
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiang Lin
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Peimeng Shuai
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Tiantian Wang
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Caimao Guo
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Qingyu Wang
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Guoqing Li
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Lijuan Meng
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| | - Lan Yi
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
3
|
Habelt B, Dörr W. Relative biological effectiveness of low-energy X-rays (25 kV) in mutant p53 cancer cells. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2023; 62:161-170. [PMID: 36609923 PMCID: PMC9950242 DOI: 10.1007/s00411-022-01014-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 12/25/2022] [Indexed: 06/17/2023]
Abstract
Low-energy X-rays as used in radiation therapy and diagnostics such as mammography are associated with a certain risk of promoting tumour development, especially in patients with mutations in cancer-related genes like TP53. The present study therefore addressed the relative biological effectiveness (RBE) of low-energy X-rays for two human adenocarcinoma cell lines of the breast (MDA-MB-468) and pancreas (BxPC-3) with a mutation in the TP53 gene. Clonogenic survival and cytogenetic changes in terms of micronuclei (MN) formation were determined following irradiation with 25 kV X-rays and 200 kV reference irradiation in the dose range of 1-8 Gy. Except the frequency of MN-containing binucleated cells (BNC) (BNC + MN/BNC) in breast cancer cells yielding an RBE between 0.6 and 0.8, both cell lines displayed dose-dependent variations of RBE values between 1 and 2 for all biological end points (cell survival, (BNC + MN/BNC), MN/BNC, MN/(BNC + MN)) with increased effectiveness of 25 kV irradiation in pancreatic compared to breast cancer cells. The results confirm previous findings indicating increased effectiveness of low-energy X-rays and underline the necessity of careful risk estimation for cancer screening programmes.
Collapse
Affiliation(s)
- Bettina Habelt
- Department of Radiotherapy and Radiation Oncology, Medical Faculty Carl Gustav Carus, University of Technology Dresden, Dresden, Germany.
- Department of Psychiatry & Psychotherapy, Medical Faculty Carl Gustav Carus, University of Technology Dresden, Dresden, Germany.
| | - Wolfgang Dörr
- Department of Radiation Oncology, Medical University Vienna, Vienna, Austria
| |
Collapse
|
4
|
miR-34a and miR-21 as biomarkers in evaluating the response of chemo-radiotherapy in Egyptian breast cancer patients. JOURNAL OF RADIATION RESEARCH AND APPLIED SCIENCES 2022. [DOI: 10.1016/j.jrras.2022.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
5
|
Alonso-González C, González-Abalde C, Menéndez-Menéndez J, González-González A, Álvarez-García V, González-Cabeza A, Martínez-Campa C, Cos S. Melatonin Modulation of Radiation-Induced Molecular Changes in MCF-7 Human Breast Cancer Cells. Biomedicines 2022; 10:biomedicines10051088. [PMID: 35625825 PMCID: PMC9138876 DOI: 10.3390/biomedicines10051088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 02/07/2023] Open
Abstract
Radiation therapy is an important component of cancer treatment scheduled for cancer patients, although it can cause numerous deleterious effects. The use of adjuvant molecules aims to limit the damage in normal surrounding tissues and enhance the effects of radiation therapy, either killing tumor cells or slowing down their growth. Melatonin, an indoleamine released by the pineal gland, behaves as a radiosensitizer in breast cancer, since it enhances the therapeutic effects of ionizing radiation and mitigates side effects on normal cells. However, the molecular mechanisms through which melatonin modulates the molecular changes triggered by radiotherapy remain mostly unknown. Here, we report that melatonin potentiated the anti-proliferative effect of radiation in MCF-7 cells. Treatment with ionizing radiation induced changes in the expression of many genes. Out of a total of 25 genes altered by radiation, melatonin potentiated changes in 13 of them, whereas the effect was reverted in another 10 cases. Among them, melatonin elevated the levels of PTEN and NME1, and decreased the levels of SNAI2, ERBB2, AKT, SERPINE1, SFN, PLAU, ATM and N3RC1. We also analyzed the expression of several microRNAs and found that melatonin enhanced the effect of radiation on the levels of miR-20a, miR-19a, miR-93, miR-20b and miR-29a. Rather surprisingly, radiation induced miR-17, miR-141 and miR-15a but melatonin treatment prior to radiation counteracted this stimulatory effect. Radiation alone enhanced the expression of the cancer suppressor miR-34a, and melatonin strongly stimulated this effect. Melatonin further enhanced the radiation-mediated inhibition of Akt. Finally, in an in vivo assay, melatonin restrained new vascularization in combination with ionizing radiation. Our results confirm that melatonin blocks many of the undesirable effects of ionizing radiation in MCF-7 cells and enhances changes that lead to optimized treatment results. This article highlights the effectiveness of melatonin as both a radiosensitizer and a radioprotector in breast cancer. Melatonin is an effective adjuvant molecule to radiotherapy, promoting anti-cancer therapeutic effects in cancer treatment. Melatonin modulates molecular pathways altered by radiation, and its use in clinic might lead to improved therapeutic outcomes by enhancing the sensitivity of cancerous cells to radiation and, in general, reversing their resistance toward currently applied therapeutic modalities.
Collapse
Affiliation(s)
- Carolina Alonso-González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain; (C.A.-G.); (C.G.-A.); (J.M.-M.); (V.Á.-G.); (S.C.)
| | - Cristina González-Abalde
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain; (C.A.-G.); (C.G.-A.); (J.M.-M.); (V.Á.-G.); (S.C.)
| | - Javier Menéndez-Menéndez
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain; (C.A.-G.); (C.G.-A.); (J.M.-M.); (V.Á.-G.); (S.C.)
| | - Alicia González-González
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria and Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain;
| | - Virginia Álvarez-García
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain; (C.A.-G.); (C.G.-A.); (J.M.-M.); (V.Á.-G.); (S.C.)
| | - Alicia González-Cabeza
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain; (C.A.-G.); (C.G.-A.); (J.M.-M.); (V.Á.-G.); (S.C.)
- Correspondence: (A.G.-C.); (C.M.-C.); Tel.: +34-942-201965 (A.G.-C.); +34-942-201963 (C.M.-C.)
| | - Carlos Martínez-Campa
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain; (C.A.-G.); (C.G.-A.); (J.M.-M.); (V.Á.-G.); (S.C.)
- Correspondence: (A.G.-C.); (C.M.-C.); Tel.: +34-942-201965 (A.G.-C.); +34-942-201963 (C.M.-C.)
| | - Samuel Cos
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain; (C.A.-G.); (C.G.-A.); (J.M.-M.); (V.Á.-G.); (S.C.)
| |
Collapse
|
6
|
Oghabi Bakhshaiesh T, Esmaeili R. Effects of noncoding RNAs in radiotherapy response in breast cancer: a systematic review. Cell Cycle 2022; 21:883-893. [PMID: 35108162 PMCID: PMC9037412 DOI: 10.1080/15384101.2022.2035915] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Radiotherapy has an essential role in breast cancer treatment. However, tumor cells may be resistant to radiotherapy. Noncoding RNAs are considered regulators of different pathways which modulate radiotherapy. This systematic review classifies long noncoding RNAs, and microRNAs precipitated in the radiation response of breast cancer patients. A total of 14 microRNAs and 8 long noncoding RNAs were studied in this review. MiR-22, miR-200 c, Let7, and LINP1 as tumor suppressors increase the effect of radiotherapy in BC. However, some noncoding RNAs such as HOTAIR, NEAT1, and miR-21 are precipitated in radio-resistance breast cancers. Significant changes in the pattern of noncoding RNAs expression before and after radiotherapy make them a good candidate for the prognosis and prediction of radiotherapy response. MiR-21 and miR-182 can promote radio-resistance via cancer stem cells. At last, the molecular mechanisms initiating radio-resistance were also examined to find the candidate noncoding RNAs for the development of radiation-sensitized agents.
Collapse
Affiliation(s)
| | - Rezvan Esmaeili
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran,CONTACT Rezvan Esmaeili No 146, Gandhi Street, Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| |
Collapse
|
7
|
Masoudi-Khoram N, Abdolmaleki P. Role of non-coding RNAs in response of breast cancer to radiation therapy. Mol Biol Rep 2022; 49:5199-5208. [PMID: 35217966 DOI: 10.1007/s11033-022-07234-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 02/04/2022] [Indexed: 12/13/2022]
Abstract
Breast cancer ranks as the first common cancer with a high incidence rate and mortality among women. Radiation therapy is the main therapeutic method for breast cancer patients. However, radiation resistance of tumor cells can reduce the efficacy of treatment and lead to recurrence and mortality in patients. Non-coding RNA (ncRNAs) refers to a group of small RNA molecules that are not translated into protein, while they have the ability to modulate the translation of target mRNA. Several studies have reported the altered expression of ncRNAs in response to radiation in breast cancer. NcRNAs have been found to influence on radiation response of breast cancer by regulating various mechanisms, including DNA damage response, cell cycle regulation, cell death, inflammatory response, cancer stem cell and EGFR related pathways. This paper aimed to provide a summary of current findings on ncRNAs dysregulation after irradiation. We also present the function and mechanism of ncRNAs in modulating radiosensitivity or radioresistance of breast cancer cells.
Collapse
Affiliation(s)
- Nastaran Masoudi-Khoram
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box 1415-154, Tehran, Iran
| | - Parviz Abdolmaleki
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box 1415-154, Tehran, Iran.
| |
Collapse
|
8
|
Lee ES, Kim WT, Park GY, Lee M, Gen Son T. Calsyntenin 1 mRNA expression sensitivity to ionizing radiation in human hepatocytes and carcinoma cells and blood cells of BALB/c mice. JOURNAL OF RADIATION RESEARCH AND APPLIED SCIENCES 2020. [DOI: 10.1080/16878507.2020.1855911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Eon-Seok Lee
- Research Center, Dongnam Institute of Radiological and Medical Science, Busan, Republic of Korea
- Department of Biochemistry, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Won-Tae Kim
- Research Center, Dongnam Institute of Radiological and Medical Science, Busan, Republic of Korea
| | - Ga-Young Park
- Research Center, Dongnam Institute of Radiological and Medical Science, Busan, Republic of Korea
| | - Manwoo Lee
- Research Center, Dongnam Institute of Radiological and Medical Science, Busan, Republic of Korea
| | - Tae Gen Son
- Research Center, Dongnam Institute of Radiological and Medical Science, Busan, Republic of Korea
| |
Collapse
|
9
|
Chen T, Yan J, Li Z. Expression of miR-34a is a sensitive biomarker for exposure to genotoxic agents in human lymphoblastoid TK6 cells. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2020; 856-857:503232. [PMID: 32928372 DOI: 10.1016/j.mrgentox.2020.503232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/10/2020] [Accepted: 07/11/2020] [Indexed: 01/07/2023]
Abstract
miR-34a has been identified as a tumor suppressor microRNA (miRNA) involved in the P53 network. Its expression levels correlate to carcinogenesis, which are generally lower in tumor tissue and higher in response to DNA damage. In this study, the response of miR-34a from exposure to genotoxic agents in human lymphoblastoid TK6 cells was evaluated to assess whether the expression of this miRNA could be used as an early indicator for genotoxic damage in mammalian cells. TK6 cells were treated with seven genotoxic agents with different mode-of-actions (cisplatin, N-ethyl-N-nitrosourea, etoposide, mitomycin C, methyl methanesulphonate, taxol, and X-ray radiation) and a non-genetic toxin (usnic acid) at different concentrations for four hours (except for X-rays) and the expression levels of miR-34a were measured 24 h after the beginning of the treatments. The expression levels of miR-34a were significantly increased by these genetic toxins in a dose-dependent manner, while no significant change in miRNA expression was found in the usnic acid-treated cells. These results suggest that miR-34a can respond to genotoxic insults sensitively; thus, miR-34a expression has the potential to be used to evaluate genotoxicity of agents.
Collapse
Affiliation(s)
- Tao Chen
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR, 72079, United States.
| | - Jian Yan
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR, 72079, United States
| | - Zhiguang Li
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR, 72079, United States
| |
Collapse
|
10
|
Regulation of DNA Damage Response and Homologous Recombination Repair by microRNA in Human Cells Exposed to Ionizing Radiation. Cancers (Basel) 2020; 12:cancers12071838. [PMID: 32650508 PMCID: PMC7408912 DOI: 10.3390/cancers12071838] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022] Open
Abstract
Ionizing radiation may be of both artificial and natural origin and causes cellular damage in living organisms. Radioactive isotopes have been used significantly in cancer therapy for many years. The formation of DNA double-strand breaks (DSBs) is the most dangerous effect of ionizing radiation on the cellular level. After irradiation, cells activate a DNA damage response, the molecular path that determines the fate of the cell. As an important element of this, homologous recombination repair is a crucial pathway for the error-free repair of DNA lesions. All components of DNA damage response are regulated by specific microRNAs. MicroRNAs are single-stranded short noncoding RNAs of 20–25 nt in length. They are directly involved in the regulation of gene expression by repressing translation or by cleaving target mRNA. In the present review, we analyze the biological mechanisms by which miRNAs regulate cell response to ionizing radiation-induced double-stranded breaks with an emphasis on DNA repair by homologous recombination, and its main component, the RAD51 recombinase. On the other hand, we discuss the ability of DNA damage response proteins to launch particular miRNA expression and modulate the course of this process. A full understanding of cell response processes to radiation-induced DNA damage will allow us to develop new and more effective methods of ionizing radiation therapy for cancers, and may help to develop methods for preventing the harmful effects of ionizing radiation on healthy organisms.
Collapse
|
11
|
Helm JS, Rudel RA. Adverse outcome pathways for ionizing radiation and breast cancer involve direct and indirect DNA damage, oxidative stress, inflammation, genomic instability, and interaction with hormonal regulation of the breast. Arch Toxicol 2020. [PMID: 32399610 DOI: 10.1007/s00204-020-02752-z)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
Knowledge about established breast carcinogens can support improved and modernized toxicological testing methods by identifying key mechanistic events. Ionizing radiation (IR) increases the risk of breast cancer, especially for women and for exposure at younger ages, and evidence overall supports a linear dose-response relationship. We used the Adverse Outcome Pathway (AOP) framework to outline and evaluate the evidence linking ionizing radiation with breast cancer from molecular initiating events to the adverse outcome through intermediate key events, creating a qualitative AOP. We identified key events based on review articles, searched PubMed for recent literature on key events and IR, and identified additional papers using references. We manually curated publications and evaluated data quality. Ionizing radiation directly and indirectly causes DNA damage and increases production of reactive oxygen and nitrogen species (RONS). RONS lead to DNA damage and epigenetic changes leading to mutations and genomic instability (GI). Proliferation amplifies the effects of DNA damage and mutations leading to the AO of breast cancer. Separately, RONS and DNA damage also increase inflammation. Inflammation contributes to direct and indirect effects (effects in cells not directly reached by IR) via positive feedback to RONS and DNA damage, and separately increases proliferation and breast cancer through pro-carcinogenic effects on cells and tissue. For example, gene expression changes alter inflammatory mediators, resulting in improved survival and growth of cancer cells and a more hospitable tissue environment. All of these events overlap at multiple points with events characteristic of "background" induction of breast carcinogenesis, including hormone-responsive proliferation, oxidative activity, and DNA damage. These overlaps make the breast particularly susceptible to ionizing radiation and reinforce that these biological activities are important characteristics of carcinogens. Agents that increase these biological processes should be considered potential breast carcinogens, and predictive methods are needed to identify chemicals that increase these processes. Techniques are available to measure RONS, DNA damage and mutation, cell proliferation, and some inflammatory proteins or processes. Improved assays are needed to measure GI and chronic inflammation, as well as the interaction with hormonally driven development and proliferation. Several methods measure diverse epigenetic changes, but it is not clear which changes are relevant to breast cancer. In addition, most toxicological assays are not conducted in mammary tissue, and so it is a priority to evaluate if results from other tissues are generalizable to breast, or to conduct assays in breast tissue. Developing and applying these assays to identify exposures of concern will facilitate efforts to reduce subsequent breast cancer risk.
Collapse
Affiliation(s)
- Jessica S Helm
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA
| | - Ruthann A Rudel
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA.
| |
Collapse
|
12
|
Helm JS, Rudel RA. Adverse outcome pathways for ionizing radiation and breast cancer involve direct and indirect DNA damage, oxidative stress, inflammation, genomic instability, and interaction with hormonal regulation of the breast. Arch Toxicol 2020; 94:1511-1549. [PMID: 32399610 PMCID: PMC7261741 DOI: 10.1007/s00204-020-02752-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/16/2020] [Indexed: 12/15/2022]
Abstract
Knowledge about established breast carcinogens can support improved and modernized toxicological testing methods by identifying key mechanistic events. Ionizing radiation (IR) increases the risk of breast cancer, especially for women and for exposure at younger ages, and evidence overall supports a linear dose-response relationship. We used the Adverse Outcome Pathway (AOP) framework to outline and evaluate the evidence linking ionizing radiation with breast cancer from molecular initiating events to the adverse outcome through intermediate key events, creating a qualitative AOP. We identified key events based on review articles, searched PubMed for recent literature on key events and IR, and identified additional papers using references. We manually curated publications and evaluated data quality. Ionizing radiation directly and indirectly causes DNA damage and increases production of reactive oxygen and nitrogen species (RONS). RONS lead to DNA damage and epigenetic changes leading to mutations and genomic instability (GI). Proliferation amplifies the effects of DNA damage and mutations leading to the AO of breast cancer. Separately, RONS and DNA damage also increase inflammation. Inflammation contributes to direct and indirect effects (effects in cells not directly reached by IR) via positive feedback to RONS and DNA damage, and separately increases proliferation and breast cancer through pro-carcinogenic effects on cells and tissue. For example, gene expression changes alter inflammatory mediators, resulting in improved survival and growth of cancer cells and a more hospitable tissue environment. All of these events overlap at multiple points with events characteristic of "background" induction of breast carcinogenesis, including hormone-responsive proliferation, oxidative activity, and DNA damage. These overlaps make the breast particularly susceptible to ionizing radiation and reinforce that these biological activities are important characteristics of carcinogens. Agents that increase these biological processes should be considered potential breast carcinogens, and predictive methods are needed to identify chemicals that increase these processes. Techniques are available to measure RONS, DNA damage and mutation, cell proliferation, and some inflammatory proteins or processes. Improved assays are needed to measure GI and chronic inflammation, as well as the interaction with hormonally driven development and proliferation. Several methods measure diverse epigenetic changes, but it is not clear which changes are relevant to breast cancer. In addition, most toxicological assays are not conducted in mammary tissue, and so it is a priority to evaluate if results from other tissues are generalizable to breast, or to conduct assays in breast tissue. Developing and applying these assays to identify exposures of concern will facilitate efforts to reduce subsequent breast cancer risk.
Collapse
Affiliation(s)
- Jessica S Helm
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA
| | - Ruthann A Rudel
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA.
| |
Collapse
|
13
|
Chiba M, Uehara H, Niiyama I, Kuwata H, Monzen S. Changes in miRNA expressions in the injured small intestine of mice following high‑dose radiation exposure. Mol Med Rep 2020; 21:2452-2458. [PMID: 32323814 PMCID: PMC7185298 DOI: 10.3892/mmr.2020.11054] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 01/20/2020] [Indexed: 12/02/2022] Open
Abstract
The small intestine is one of the most highly regenerative and radiosensitive tissues in mammals, including humans. Exposure to high doses of ionizing radiation causes serious intestinal damage. Recently, several investigations have been conducted using radioprotective agents to determine ways for reducing intestinal damage caused by radiation exposure. However, a thorough understanding of functional changes occurring in the small intestine of mice exposed to high-dose radiation is necessary for developing novel and more potent radioprotective agents. In this study, we examined changes in microRNA (miRNA/miR) expressions in the small intestine of mice at 72 h after X-ray exposure (10 Gy). We identified seven upregulated miRNAs and six downregulated miRNAs in the small intestine of mice following radiation exposure using miRNA microarray analysis. Particularly, miR-34a-5p was highly expressed, which was confirmed by reverse transcription-quantitative PCR. Forkhead box P1 (Foxp1) was predicted to be a target of the mRNA of miR-34a-5p using OmicsNet. Decreased Foxp1 expression in the small intestine following radiation exposure was confirmed, suggesting that Foxp1 expression recovery may induce the suppression of radiation-induced enteritis. Therefore, miR-34a-5p is a potential target molecule for developing novel radioprotective agents.
Collapse
Affiliation(s)
- Mitsuru Chiba
- Department of Bioscience and Laboratory Medicine, Graduate School of Health Sciences, Hirosaki University, Hirosaki, Aomori 036‑8564, Japan
| | - Haruka Uehara
- Department of Medical Technology, School of Health Sciences, Hirosaki University, Hirosaki, Aomori 036‑8564, Japan
| | - Ikumi Niiyama
- Department of Medical Technology, School of Health Sciences, Hirosaki University, Hirosaki, Aomori 036‑8564, Japan
| | - Haruka Kuwata
- Department of Medical Technology, School of Health Sciences, Hirosaki University, Hirosaki, Aomori 036‑8564, Japan
| | - Satoru Monzen
- Department of Radiation Science, Graduate School of Health Sciences, Hirosaki University, Hirosaki, Aomori 036‑8564, Japan
| |
Collapse
|
14
|
Bulvik R, Biton M, Berkman N, Breuer R, Wallach-Dayan SB. Forefront: MiR-34a-Knockout Mice with Wild Type Hematopoietic Cells, Retain Persistent Fibrosis Following Lung Injury. Int J Mol Sci 2020; 21:2228. [PMID: 32210149 PMCID: PMC7139923 DOI: 10.3390/ijms21062228] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/15/2020] [Accepted: 03/19/2020] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRs) are known to limit gene expression at the post-transcriptional level and have important roles in the pathogenesis of various conditions, including acute lung injury (ALI) and fibrotic diseases such as idiopathic pulmonary fibrosis (IPF). In this study, we found increased levels of miR-34 at times of fibrosis resolution following injury, in myofibroblasts from Bleomycin-treated mouse lungs, which correlates with susceptibility to cell death induced by immune cells. On the contrary, a substantial downregulation of miR-34 was detected at stages of evolution, when fibroblasts resist cell death. Concomitantly, we found an inverse correlation between miR-34 levels with that of the survival molecule FLICE-like inhibitory protein (FLIP) in lung myofibroblasts from humans with IPF and the experimental model. Forced upregulation of miR-34 with miR-34 mimic in human IPF fibrotic-lung myofibroblasts led to decreased cell survival through downregulation of FLIP. Using chimeric miR-34 knock-out (KO)-C57BL/6 mice with miR34KO myofibroblasts but wild-type (WT) hematopoietic cells, we found, in contrast to WT mice, increased and persistent FLIP levels with a more severe fibrosis and with no signs of resolution as detected in pathology and collagen accumulation. Moreover, a mimic of miR-34a decreased FLIP expression and susceptibility to cell death was regained in miR-34KO fibroblasts. Through this study, we show for the first time an inverse correlation between miR-34a and FLIP expression in myofibroblasts, which affects survival, and accumulation in lung fibrosis. Reprogramming fibrotic-lung myofibroblasts to regain susceptibility to cell-death by specifically increasing their miR34a and downregulating FLIP, may be a useful strategy, enabling tissue regeneration following lung injury.
Collapse
Affiliation(s)
- Raanan Bulvik
- Lung Cellular and Molecular Biology Laboratory, Institute of Pulmonary Medicine, Hadassah—Hebrew University Medical Center, POB 12000, Jerusalem 9112102, Israel
| | - Moshe Biton
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hadassah Hebrew University Medical Center, Jerusalem 9112102, Israel
| | - Neville Berkman
- Lung Cellular and Molecular Biology Laboratory, Institute of Pulmonary Medicine, Hadassah—Hebrew University Medical Center, POB 12000, Jerusalem 9112102, Israel
| | - Raphael Breuer
- Lung Cellular and Molecular Biology Laboratory, Institute of Pulmonary Medicine, Hadassah—Hebrew University Medical Center, POB 12000, Jerusalem 9112102, Israel
- Department of Pathology and Laboratory Medicine, 670 Albany St, 4th Floor, Boston University School of Medicine, Boston, MA 02118, USA
| | - Shulamit B. Wallach-Dayan
- Lung Cellular and Molecular Biology Laboratory, Institute of Pulmonary Medicine, Hadassah—Hebrew University Medical Center, POB 12000, Jerusalem 9112102, Israel
| |
Collapse
|
15
|
Kang Q, Zhang X, Cao N, Chen C, Yi J, Hao L, Ji Y, Liu X, Lu J. EGCG enhances cancer cells sensitivity under 60Coγ radiation based on miR-34a/Sirt1/p53. Food Chem Toxicol 2019; 133:110807. [DOI: 10.1016/j.fct.2019.110807] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/01/2019] [Accepted: 09/05/2019] [Indexed: 01/04/2023]
|
16
|
Huang R, Zhou P. Double-edged effects of noncoding RNAs in responses to environmental genotoxic insults: Perspectives with regards to molecule-ecology network. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 247:64-71. [PMID: 30654255 DOI: 10.1016/j.envpol.2019.01.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 01/04/2019] [Accepted: 01/04/2019] [Indexed: 06/09/2023]
Abstract
Numerous recent studies have underlined the crucial players of noncoding RNAs (ncRNAs), i.e., microRNAs(miRNAs), long noncoding RNAs(lncRNAs) and circle RNAs(circRNAs) participating in genotoxic responses induced by a wide variety of environmental genotoxicants consistently. Genotoxic-derived ncRNAs provide us a new epigenetic molecular-ecological network (MEN) insights into the underlying mechanisms regarding genotoxicant exposure and genotoxic effects, which can modify ncRNAs to render them "genotoxic" and inheritable, thus potentially leading to disease risk via epigenetic changes. In fact, the spatial structures of ncRNAs, particularly of secondary and three-dimensional structures, diverse environmental genotoxicants as well as RNA splicing and editing forma dynamic pool of ncRNAs, which constructs a MEN in cells together with their enormous targets and interactions, making biological functions more complicated. We nonetheless suggest that ncRNAs have both beneficial(positive) and harmful(negative) effects, i.e., are "double-edged" in regulating genotoxicant toxic responses. Understanding the "double-edged" effects of ncRNAs is of crucial importance for our further comprehension of the pathogenesis of human diseases induced by environmental toxicants and for the construction of novel prevention and therapy targets. Furthermore, the MEN formed by ncRNAs and their interactions each other as well as downstream targets in the cells is important for considering the active relationships between external agents (environmental toxicants) and inherent genomic ncRNAs, in terms of suppression or promotion (down- or upregulation), and engineered ncRNA therapies can suppress or promote the expression of inherent genomic ncRNAs that are targets of environmental toxicants. Moreover, the MEN would be expected to be would be applied to the mechanistic explanation and risk assessment at whole scene level in environmental genotoxicant exposure. As molecular biology evolves rapidly, the proposed MEN perspective will provide a clearer or more comprehensive holistic view.
Collapse
Affiliation(s)
- Ruixue Huang
- Department of Occupational and Environmental Health, Central South University, Changsha, 410078, China.
| | - PingKun Zhou
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, AMMS, Beijing, 100850, PR China.
| |
Collapse
|
17
|
Liolios T, Kastora SL, Colombo G. MicroRNAs in Female Malignancies. Cancer Inform 2019; 18:1176935119828746. [PMID: 30792572 PMCID: PMC6376555 DOI: 10.1177/1176935119828746] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 12/27/2018] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are endogenous 22-nucleotide RNAs that can play a fundamental regulatory role in the gene expression of various organisms. Current research suggests that miRNAs can assume pivotal roles in carcinogenesis. In this article, through bioinformatics mining and computational analysis, we determine a single miRNA commonly involved in the development of breast, cervical, endometrial, ovarian, and vulvar cancer, whereas we underline the existence of 7 more miRNAs common in all examined malignancies with the exception of vulvar cancer. Furthermore, we identify their target genes and encoded biological functions. We also analyze common biological processes on which all of the identified miRNAs act and we suggest a potential mechanism of action. In addition, we analyze exclusive miRNAs among the examined malignancies and bioinformatically explore their functionality. Collectively, our data can be employed in in vitro assays as a stepping stone in the identification of a universal machinery that is derailed in female malignancies, whereas exclusive miRNAs may be employed as putative targets for future chemotherapeutic agents or cancer-specific biomarkers.
Collapse
Affiliation(s)
- Themis Liolios
- Hellenic Republic National and
Kapodistrian, University of Athens, Faculty of Biology, Athens, Greece
| | | | - Giorgia Colombo
- University of Aberdeen, School of
Medicine and Dentistry, Aberdeen, UK
| |
Collapse
|
18
|
Tharmalingam S, Sreetharan S, Brooks AL, Boreham DR. Re-evaluation of the linear no-threshold (LNT) model using new paradigms and modern molecular studies. Chem Biol Interact 2019; 301:54-67. [PMID: 30763548 DOI: 10.1016/j.cbi.2018.11.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/13/2018] [Accepted: 11/22/2018] [Indexed: 02/06/2023]
Abstract
The linear no-threshold (LNT) model is currently used to estimate low dose radiation (LDR) induced health risks. This model lacks safety thresholds and postulates that health risks caused by ionizing radiation is directly proportional to dose. Therefore even the smallest radiation dose has the potential to cause an increase in cancer risk. Advances in LDR biology and cell molecular techniques demonstrate that the LNT model does not appropriately reflect the biology or the health effects at the low dose range. The main pitfall of the LNT model is due to the extrapolation of mutation and DNA damage studies that were conducted at high radiation doses delivered at a high dose-rate. These studies formed the basis of several outdated paradigms that are either incorrect or do not hold for LDR doses. Thus, the goal of this review is to summarize the modern cellular and molecular literature in LDR biology and provide new paradigms that better represent the biological effects in the low dose range. We demonstrate that LDR activates a variety of cellular defense mechanisms including DNA repair systems, programmed cell death (apoptosis), cell cycle arrest, senescence, adaptive memory, bystander effects, epigenetics, immune stimulation, and tumor suppression. The evidence presented in this review reveals that there are minimal health risks (cancer) with LDR exposure, and that a dose higher than some threshold value is necessary to achieve the harmful effects classically observed with high doses of radiation. Knowledge gained from this review can help the radiation protection community in making informed decisions regarding radiation policy and limits.
Collapse
Affiliation(s)
- Sujeenthar Tharmalingam
- Northern Ontario School of Medicine, Laurentian University, 935 Ramsey Lake Rd, Sudbury, ON, P3E 2C6, Canada.
| | - Shayenthiran Sreetharan
- Department of Medical Physics and Applied Radiation Sciences, McMaster University, 1280 Main Street W, Hamilton ON, L8S 4K1, Canada
| | - Antone L Brooks
- Environmental Science, Washington State University, Richland, WA, USA
| | - Douglas R Boreham
- Northern Ontario School of Medicine, Laurentian University, 935 Ramsey Lake Rd, Sudbury, ON, P3E 2C6, Canada; Bruce Power, Tiverton, ON(3), UK.
| |
Collapse
|
19
|
Khordadmehr M, Shahbazi R, Ezzati H, Jigari-Asl F, Sadreddini S, Baradaran B. Key microRNAs in the biology of breast cancer; emerging evidence in the last decade. J Cell Physiol 2018; 234:8316-8326. [PMID: 30422324 DOI: 10.1002/jcp.27716] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 10/16/2018] [Indexed: 12/12/2022]
Abstract
microRNAs (miRNAs) are a family of small noncoding RNAs that play a pivotal role in the regulation of main biological and physiological processes, including cell cycle regulation, proliferation, differentiation, apoptosis, stem cell maintenance, and organ development. Dysregulation of these tiny molecules has been related to different human diseases, such as cancer. It has been estimated that more than 50% of these noncoding RNA sequences are placed on fragile sites or cancer-associated genomic regions. After the discovery of the first specific miRNA signatures in breast cancer, many studies focused on the involvement of these small RNAs in the pathophysiology of breast tumors and their possible clinical implications as reliable prognostic biomarkers or as a new therapeutic approach. Therefore, the present review will focus on the recent findings on the involvement of miRNAs in the biology of breast cancer associated with their clinical implications.
Collapse
Affiliation(s)
- Monireh Khordadmehr
- Department of Pathology, Faculty of Veterinary, Medicine, University of Tabriz, Tabriz, East Azerbaijan, Iran
| | - Roya Shahbazi
- Department of Pathology, Faculty of Veterinary, Medicine, University of Tabriz, Tabriz, East Azerbaijan, Iran
| | - Hamed Ezzati
- Department of Pathology, Faculty of Veterinary, Medicine, University of Tabriz, Tabriz, East Azerbaijan, Iran
| | - Farinaz Jigari-Asl
- Department of Pathology, Faculty of Veterinary, Medicine, University of Tabriz, Tabriz, East Azerbaijan, Iran
| | - Sanam Sadreddini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
20
|
Nicolini A, Ferrari P, Duffy MJ. Prognostic and predictive biomarkers in breast cancer: Past, present and future. Semin Cancer Biol 2018; 52:56-73. [DOI: 10.1016/j.semcancer.2017.08.010] [Citation(s) in RCA: 209] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/14/2017] [Accepted: 08/24/2017] [Indexed: 12/19/2022]
|
21
|
Plantamura I, Cosentino G, Cataldo A. MicroRNAs and DNA-Damaging Drugs in Breast Cancer: Strength in Numbers. Front Oncol 2018; 8:352. [PMID: 30234015 PMCID: PMC6129576 DOI: 10.3389/fonc.2018.00352] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 08/10/2018] [Indexed: 11/25/2022] Open
Abstract
MicroRNAs are a class of small non-coding regulatory RNAs playing key roles in cancer. Breast cancer is the most common female malignancy worldwide and is categorized into four molecular subtypes: luminal A and B, HER2+ and triple-negative breast cancer (TNBC). Despite the development of multiple targeted therapies for luminal and HER2+ breast tumors, TNBC lacks specific therapeutic approaches, thus they are treated mainly with radio- and chemotherapy. The effectiveness of these therapeutic regimens is based on their ability to induce DNA damage, which is differentially resolved and repaired by normal vs. cancer cells. Recently, drugs directly targeting DNA repair mechanisms, such as PARP inhibitors, have emerged as attractive candidates for the future molecular targeted-therapy in breast cancer. These compounds prevent cancer cells to appropriate repair DNA double strand breaks and induce a phenomenon called synthetic lethality, that results from the concurrent inhibition of PARP and the absence of functional BRCA genes which prompt cell death. MicroRNAs are relevant players in most of the biological processes including DNA damage repair mechanisms. Consistently, the downregulation of DNA repair genes by miRNAs have been probe to improve the therapeutic effect of genotoxic drugs. In this review, we discuss how microRNAs can sensitize cancer cells to DNA-damaging drugs, through the regulation of DNA repair genes, and examine the most recent findings on their possible use as a therapeutic tools of treatment response in breast cancer.
Collapse
Affiliation(s)
- Ilaria Plantamura
- Molecular Targeting Unit, Research Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giulia Cosentino
- Molecular Targeting Unit, Research Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessandra Cataldo
- Molecular Targeting Unit, Research Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
22
|
Differential miRNA expression profiling reveals miR-205-3p to be a potential radiosensitizer for low- dose ionizing radiation in DLD-1 cells. Oncotarget 2018; 9:26387-26405. [PMID: 29899866 PMCID: PMC5995186 DOI: 10.18632/oncotarget.25405] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 04/28/2018] [Indexed: 12/12/2022] Open
Abstract
Enhanced radiosensitivity at low doses of ionizing radiation (IR) (0.2 to 0.6 Gy) has been reported in several cell lines. This phenomenon, known as low doses hyper-radiosensitivity (LDHRS), appears as an opportunity to decrease toxicity of radiotherapy and to enhance the effects of chemotherapy. However, the effect of low single doses IR on cell death is subtle and the mechanism underlying LDHRS has not been clearly explained, limiting the utility of LDHRS for clinical applications. To understand the mechanisms responsible for cell death induced by low-dose IR, LDHRS was evaluated in DLD-1 human colorectal cancer cells and the expression of 80 microRNAs (miRNAs) was assessed by qPCR array. Our results show that DLD-1 cells display an early DNA damage response and apoptotic cell death when exposed to 0.6 Gy. miRNA expression profiling identified 3 over-expressed (miR-205-3p, miR-1 and miR-133b) and 2 down-regulated miRNAs (miR-122-5p, and miR-134-5p) upon exposure to 0.6 Gy. This miRNA profile differed from the one in cells exposed to high-dose IR (12 Gy), supporting a distinct low-dose radiation-induced cell death mechanism. Expression of a mimetic miR-205-3p, the most overexpressed miRNA in cells exposed to 0.6 Gy, induced apoptotic cell death and, more importantly, increased LDHRS in DLD-1 cells. Thus, we propose miR-205-3p as a potential radiosensitizer to low-dose IR.
Collapse
|
23
|
A novel miR-34a target, protein kinase D1, stimulates cancer stemness and drug resistance through GSK3/β-catenin signaling in breast cancer. Oncotarget 2018; 7:14791-802. [PMID: 26895471 PMCID: PMC4924752 DOI: 10.18632/oncotarget.7443] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 01/31/2016] [Indexed: 01/06/2023] Open
Abstract
One of the properties of human breast cancer cells is cancer stemness, which is characterized by self-renewal capability and drug resistance. Protein kinase D1 (PRKD1) functions as a key regulator of many cellular processes and is downregulated in invasive breast cancer cells. In this study, we found that PRKD1 was upregulated in MCF-7-ADR human breast cancer cells characterized by drug resistance. Additionally, we discovered that PRKD1 expression was negatively regulated by miR-34a binding to the PRKD1 3′-UTR. PRKD1 expression increased following performance of a tumorsphere formation assay in MCF-7-ADR cells. We also found that reduction of PRKD1 by ectopic miR-34a expression or PRKD1 siRNA treatment resulted in suppressed self-renewal ability in breast cancer stem cells. Furthermore, we confirmed that the PRKD1 inhibitor CRT0066101 reduced phosphorylated PKD/PKCμ, leading to suppression of breast cancer stemness through GSK3/β-catenin signaling. PRKD1 inhibition also influenced apoptosis initiation in MCF-7-ADR cells. Tumors from nude mice treated with miR-34a or CRT0066101 showed suppressed tumor growth, proliferation, and induced apoptosis. These results provide evidence that regulation of PRKD1, a novel miR-34a target, contributes to overcoming cancer stemness and drug resistance in human breast cancer.
Collapse
|
24
|
Xie X, Xu Z, Wang C, Fang C, Zhao J, Xu L, Qian X, Dai J, Sun F, Xu D, He W. Tip60 is associated with resistance to X-ray irradiation in prostate cancer. FEBS Open Bio 2017; 8:271-278. [PMID: 29435417 PMCID: PMC5794467 DOI: 10.1002/2211-5463.12371] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 11/23/2017] [Accepted: 12/04/2017] [Indexed: 01/25/2023] Open
Abstract
Tip60, an oncogene, accelerates cell growth by regulating androgen receptor translocation into the nucleus in prostate cancer. However, the mechanism of Tip60 in the response of prostate cancer to radiotherapy, and radioresistance, has not been studied. Using human prostate cancer samples and two human prostate cancer cell lines (LNCaP and DU145), Tip60 protein expression and the acetylation of ataxia telangiectasia mutant (ATM) were analysed by western blotting and immunoprecipitation. Tip60 was downregulated with small interfering RNA. Cells were irradiated using X‐rays at 0.25 Gy·min−1. Cell viability was assessed by the MTT assay. The expression of Tip60 protein was increased in radioresistant prostate cancer tissues in comparison with radiosensitive tissues, which was also confirmed in both irradiated DU145 and LNCaP cells. Furthermore, the acetylation of ATM was also upregulated in a time‐dependent manner after irradiation of both DU145 and LNCaP cells. Additionally, depletion of Tip60 decreased the survival of LNCaP and DU145 cells by inducing apoptosis, reduced the acetylation of ATM and decreased the expression of phosphorylated ATM, Chk2 and cdc25A in both DU145 and LNCaP cells after X‐ray irradiation. The results of this study demonstrated that the expression of Tip60 may be related to the radioresistance of prostate cancer and could serve as a promising predictive factor for prostate cancer patients receiving radiotherapy.
Collapse
Affiliation(s)
- Xin Xie
- Department of Urology Ruijin Hospital Shanghai Jiaotong University, School of Medicine China
| | - Zhaoping Xu
- Department of Urology Ruijin Hospital Shanghai Jiaotong University, School of Medicine China
| | - Chenghe Wang
- Department of Urology Ruijin Hospital Shanghai Jiaotong University, School of Medicine China
| | - Chen Fang
- Department of Urology Ruijin Hospital Shanghai Jiaotong University, School of Medicine China
| | - Juping Zhao
- Department of Urology Ruijin Hospital Shanghai Jiaotong University, School of Medicine China
| | - Le Xu
- Department of Urology Ruijin Hospital Shanghai Jiaotong University, School of Medicine China
| | - Xiaoqiang Qian
- Department of Urology Ruijin Hospital Shanghai Jiaotong University, School of Medicine China
| | - Jun Dai
- Department of Urology Ruijin Hospital Shanghai Jiaotong University, School of Medicine China
| | - Fukang Sun
- Department of Urology Ruijin Hospital Shanghai Jiaotong University, School of Medicine China
| | - Danfeng Xu
- Department of Urology Ruijin Hospital Shanghai Jiaotong University, School of Medicine China
| | - Wei He
- Department of Urology Ruijin Hospital Shanghai Jiaotong University, School of Medicine China
| |
Collapse
|
25
|
Lee S, Jeong H, Seong M, Kim JG. Change of tumor vascular reactivity during tumor growth and postchemotherapy observed by near-infrared spectroscopy. JOURNAL OF BIOMEDICAL OPTICS 2017; 22:121603. [PMID: 28698890 DOI: 10.1117/1.jbo.22.12.121603] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 05/31/2017] [Indexed: 05/22/2023]
Abstract
Breast cancer is one of the most common cancers in females. To monitor chemotherapeutic efficacy for breast cancer, medical imaging systems such as x-ray mammography, computed tomography, magnetic resonance imaging, and ultrasound imaging have been used. Currently, it can take up to 3 to 6 weeks to see the tumor response from chemotherapy by monitoring tumor volume changes. We used near-infrared spectroscopy (NIRS) to predict breast cancer treatment efficacy earlier than tumor volume changes by monitoring tumor vascular reactivity during inhalational gas interventions. The results show that the amplitude of oxy-hemoglobin changes (vascular reactivity) during hyperoxic gas inhalation is well correlated with tumor growth and responded one day earlier than tumor volume changes after chemotherapy. These results may imply that NIRS with respiratory challenges can be useful in early detection of tumor and in the prediction of tumor response to chemotherapy.
Collapse
Affiliation(s)
- Songhyun Lee
- Gwangju Institute of Science and Technology, Department of Biomedical Science and Engineering, Gwangju, Republic of Korea
| | - Hyeryun Jeong
- Gwangju Institute of Science and Technology, Department of Biomedical Science and Engineering, Gwangju, Republic of Korea
| | - Myeongsu Seong
- Gwangju Institute of Science and Technology, Department of Biomedical Science and Engineering, Gwangju, Republic of Korea
| | - Jae Gwan Kim
- Gwangju Institute of Science and Technology, Department of Biomedical Science and Engineering, Gwangju, Republic of KoreabGwangju Institute of Science and Technology, School of Electrical Engineering and Computer Science, Gwangju, Republic of Korea
| |
Collapse
|
26
|
Imani S, Wei C, Cheng J, Khan MA, Fu S, Yang L, Tania M, Zhang X, Xiao X, Zhang X, Fu J. MicroRNA-34a targets epithelial to mesenchymal transition-inducing transcription factors (EMT-TFs) and inhibits breast cancer cell migration and invasion. Oncotarget 2017; 8:21362-21379. [PMID: 28423483 PMCID: PMC5400590 DOI: 10.18632/oncotarget.15214] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 01/25/2017] [Indexed: 12/31/2022] Open
Abstract
MicroRNA-34a (miR-34a) plays an essential role against tumorigenesis and progression of cancer metastasis. Here, we analyzed the expression, targets and functional effects of miR-34a on epithelial to mesenchymal transition-inducing transcription factors (EMT-TFs), such as TWIST1, SLUG and ZEB1/2, and an EMT-inducing protein NOTCH1 in breast cancer (BC) cell migration and invasion and its correlation with tumorigenesis and clinical outcomes. Expression of miR-34a is downregulated in human metastatic breast cancers (MBC) compared to normal breast tissues and is negatively correlated with clinicopathological features of MBC patients. Ectopic expression of miR-34a in MBC cell-line BT-549 significantly inhibits cell migration and invasion, but exhibits no clear effect on BC cell growth. We found that miR-34a is able to inactivate EMT signaling pathway with mediatory of NOTCH1, TWIST1, and ZEB1 upon 3′-UTR activity in MBC cell lines, but has no inhibitory effects on SLUG and ZEB2. Furthermore, we investigated the synergistic effects of Thymoquinone (TQ) and miR-34a together on the expression of EMT-associated proteins. Results showed that co-delivery of miR-34a and TQ is able to inactivate EMT signaling pathway by directly targeting TWIST1 and ZEB1 in BT-549 cell line, indicating that they might be a promising therapeutic combination against breast cancer metastasis. Epigenetic inactivation of the EMT-TFs/miR-34a pathway can potentially alter the equilibrium of these regulations, facilitating EMT and metastasis in BC. Altogether, our findings suggest that miR-34a alone could serve as a potential therapeutic agent for MBC, and together with TQ, their therapeutic potential is synergistically enhanced.
Collapse
Affiliation(s)
- Saber Imani
- Key Laboratory of Epigenetics and Oncology, Research Center for Precision Medicine, Southwest Medical University, Luzhou, Sichuan, China.,Chemical Injuries Research Center, Baqiyatallah Medical Sciences University (BMSU), Tehran, Iran
| | - Chunli Wei
- Key Laboratory of Epigenetics and Oncology, Research Center for Precision Medicine, Southwest Medical University, Luzhou, Sichuan, China.,State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau (SAR), China
| | - Jingliang Cheng
- Key Laboratory of Epigenetics and Oncology, Research Center for Precision Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Md Asaduzzaman Khan
- Key Laboratory of Epigenetics and Oncology, Research Center for Precision Medicine, Southwest Medical University, Luzhou, Sichuan, China.,Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Shangyi Fu
- The Honors College, University of Houston, Houston, TX, USA
| | - Luquan Yang
- Key Laboratory of Epigenetics and Oncology, Research Center for Precision Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Mousumi Tania
- Key Laboratory of Epigenetics and Oncology, Research Center for Precision Medicine, Southwest Medical University, Luzhou, Sichuan, China.,Division of Computer Aided Drug Design, Red-Green Computing Centre, Dhaka, Bangladesh
| | - Xianqin Zhang
- Key Laboratory of Epigenetics and Oncology, Research Center for Precision Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiuli Xiao
- Pathology Department, Southwest Medical University, Luzhou, Sichuan, China
| | - Xianning Zhang
- Department of Cell Biology and Medical Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, Research Center for Precision Medicine, Southwest Medical University, Luzhou, Sichuan, China.,State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau (SAR), China
| |
Collapse
|
27
|
3,6-Dihydroxyflavone regulates microRNA-34a through DNA methylation. BMC Cancer 2017; 17:619. [PMID: 28870206 PMCID: PMC5584326 DOI: 10.1186/s12885-017-3638-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 08/29/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Breast cancer is the common cancer in China. In previous study, we determined that 3,6-dihydroxyflavone (3,6-DHF) increases miR-34a significantly in breast carcinogenesis, but the mechanism remains unclear. METHODS We used qRT-PCR to analyze miR-34a and ten-eleven translocation (TET)1, TET2, TET3 levels in breast cancer cells. With a cellular breast carcinogenesis model and an experimental model of carcinogenesis in rats, TET1 levels were evaluated by western blot analysis and immunofluorescence. TET1 and 5hmC (5-hydroxymethylcytosine) levels were evaluated by immunofluorescence in nude mouse xenografts of MDA-MB-231 cells. Chromatin immunoprecipitation(ChIP) assayed for TET1 on the TET1 promoter, and dot blot analysis of DNA 5hmC was performed in MDA-MB-231 cells. We evaluated the mechanism of 3,6-DHF on the expression of tumor suppressor miR-34a by transfecting them with DNA methyltransferase (DNMT)1 plasmid and TET1 siRNA in breast cancer cells. Methylation-specific PCR detected methylation of the miR-34a promoter. RESULTS First, we found that 3,6-DHF promotes the expression of TET1 during carcinogen-induced breast carcinogenesis in MCF10A cells and in rats. 3,6-DHF also increased TET1 and 5hmC levels in MDA-MB-231 cells. Further study indicated that TET1 siRNA and pcDNA3/Myc-DNMT1 inhibited the 3,6-DHF reactivation effect on expression of miR-34a in breast cancer cells. Methylation-specific PCR assays indicated that TET1 siRNA and pcDNA3/Myc-DNMT1 inhibit the effect of 3,6-DHF on the demethylation of the miR-34a promoter. CONCLUSIONS Our study showed that 3,6-DHF effectively increases TET1 expression by inhibiting DNMT1 and DNA hypermethylation, and consequently up-regulates miR-34a in breast carcinogenesis.
Collapse
|
28
|
Tharmalingam S, Sreetharan S, Kulesza AV, Boreham DR, Tai TC. Low-Dose Ionizing Radiation Exposure, Oxidative Stress and Epigenetic Programing of Health and Disease. Radiat Res 2017; 188:525-538. [PMID: 28753061 DOI: 10.1667/rr14587.1] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Ionizing radiation exposure from medical diagnostic imaging has greatly increased over the last few decades. Approximately 80% of patients who undergo medical imaging are exposed to low-dose ionizing radiation (LDIR). Although there is widespread consensus regarding the harmful effects of high doses of radiation, the biological effects of low-linear energy transfer (LET) LDIR is not well understood. LDIR is known to promote oxidative stress, however, these levels may not be large enough to result in genomic mutations. There is emerging evidence that oxidative stress causes heritable modifications via epigenetic mechanisms (DNA methylation, histone modification, noncoding RNA regulation). These epigenetic modifications result in permanent cellular transformations without altering the underlying DNA nucleotide sequence. This review summarizes the major concepts in the field of epigenetics with a focus on the effects of low-LET LDIR (<100 mGy) and oxidative stress on epigenetic gene modification. In this review, we show evidence that suggests that LDIR-induced oxidative stress provides a mechanistic link between LDIR and epigenetic gene regulation. We also discuss the potential implication of LDIR exposure during pregnancy where intrauterine fetal development is highly susceptible to oxidative stress-induced epigenetic programing.
Collapse
Affiliation(s)
| | | | - Adomas V Kulesza
- b Department of Biology, McMaster University, Hamilton, Canada, L8S 4K1
| | - Douglas R Boreham
- a Northern Ontario School of Medicine, Laurentian University, Sudbury, Canada, P3E 2C6.,c Department of Medical Physics and Applied Radiation Sciences, McMaster University, Hamilton, Canada, L8S 4K1
| | - T C Tai
- a Northern Ontario School of Medicine, Laurentian University, Sudbury, Canada, P3E 2C6
| |
Collapse
|
29
|
Lacombe J, Zenhausern F. Emergence of miR-34a in radiation therapy. Crit Rev Oncol Hematol 2017; 109:69-78. [PMID: 28010900 PMCID: PMC5199215 DOI: 10.1016/j.critrevonc.2016.11.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 11/14/2016] [Accepted: 11/29/2016] [Indexed: 12/20/2022] Open
Abstract
Expressions of many microRNAs (miRNAs) in response to ionizing radiation (IR) have already been investigated and some of them seem to play an important role in the tumor radioresistance, normal tissue radiotoxicity or as predictive biomarkers to radiation. miR-34a is an emerging miRNA in recent radiobiology studies. Here, we review this miR-34 family member by detailing its different roles in radiation response and we will discuss about the role that it can play in radiation treatment. Thus, we will show that IR regulates miR-34a by increasing its expression. We will also highlight different biological processes involved in cellular response to IR and regulated by miR-34a in order to demonstrate the role it can play in tumor radio-response or normal tissue radiotoxicity as a radiosensitizer or radioprotector. miR-34a is poised to assert itself as an important player in radiobiology and should become more and more important in radiation therapy management.
Collapse
Affiliation(s)
- Jerome Lacombe
- Center for Applied NanoBioscience and Medicine, University of Arizona, 145 S. 79th Street, Chandler, AZ 85226, USA.
| | - Frederic Zenhausern
- Center for Applied NanoBioscience and Medicine, University of Arizona, 145 S. 79th Street, Chandler, AZ 85226, USA; Translational Genomics Research Institute, 445 N. Fifth Street, Phoenix, AZ 85004, USA; Department of Basic Medical Sciences, College of Medicine Phoenix, 425 N. 5th Street, Phoenix, AZ 85004, USA.
| |
Collapse
|
30
|
Li SJ, Liang XY, Li HJ, Li W, Zhou L, Chen HQ, Ye SG, Yu DH, Cui JW. Low-dose irradiation promotes proliferation of the human breast cancer MDA-MB-231 cells through accumulation of mutant P53. Int J Oncol 2016; 50:290-296. [DOI: 10.3892/ijo.2016.3795] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 11/29/2016] [Indexed: 11/05/2022] Open
|
31
|
Sharma S, Patnaik PK, Aronov S, Kulshreshtha R. ApoptomiRs of Breast Cancer: Basics to Clinics. Front Genet 2016; 7:175. [PMID: 27746811 PMCID: PMC5041507 DOI: 10.3389/fgene.2016.00175] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 09/15/2016] [Indexed: 12/21/2022] Open
Abstract
Apoptosis, a form of programmed cell death, is a highly regulated process, the deregulation of which has been associated with the tumor initiation, progression, and metastasis in various cancers including breast cancer. Induction of apoptosis is a popular target of various therapies currently being tested or used for breast cancer treatment. Thus, identifying apoptotic mediators and regulators is imperative for molecular biologists and clinicians for benefit of patients. The regulation of apoptosis is complex and involves a tight equilibrium between the pro- and anti-apoptotic factors. Recent studies have highlighted the role of miRNAs in the control of apoptosis and their interplay with p53, the master guardian of apoptosis. Here, we summarize and integrate the data on the role of miRNAs in apoptosis in breast cancer and the clinical advantage it may offer for the prognosis or treatment of breast cancer patients.
Collapse
Affiliation(s)
- Shivani Sharma
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi New Delhi, India
| | | | - Stella Aronov
- Department of Molecular Biology, Ariel University Ariel, Israel
| | - Ritu Kulshreshtha
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi New Delhi, India
| |
Collapse
|
32
|
Lu J, Chen C, Hao L, Zheng Z, Zhang N, Wang Z. MiRNA expression profile of ionizing radiation-induced liver injury in mouse using deep sequencing. Cell Biol Int 2016; 40:873-86. [PMID: 27214643 DOI: 10.1002/cbin.10627] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 05/13/2016] [Indexed: 12/20/2022]
Abstract
In order to investigate the potential regulatory roles of microRNAs (miRNAs) in mouse response to ionizing radiation (IR), the small RNA libraries from liver tissues of mice with or without ionizing radiation (IR) were sequenced by high-throughput deep sequencing technology. A total of 270 miRNAs including 212 known and 58 potentially novel miRNAs were identified. Within these miRNAs, there were 48 miRNAs that were differentially expressed, including 27 known and 21 novel miRNAs. The results of quantitative RT-polymerase chain reaction (qRT-PCR) were in consistent with the sequencing analysis. Target gene prediction, function annotation, and pathway of the identified miRNAs were analyzed using RNAhybrid, miRanda software and Swiss-Prot, Gene Ontology (GO), Clusters of Orthologous Groups (COG), Kyoto Encyclopedia of Genes, and Genomes (KEGG) and non-redundant (NR) databases. These results should be useful to investigate the biological function of miRNAs under IR-induced liver injury.
Collapse
Affiliation(s)
- Jike Lu
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China.,Department of People's Liberation Army, The Quartermaster Equipment Institute of General Logistics, Beijing, 100010, China
| | - Chen Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Limin Hao
- Department of People's Liberation Army, The Quartermaster Equipment Institute of General Logistics, Beijing, 100010, China
| | - Zhiqiang Zheng
- Department of People's Liberation Army, The Quartermaster Equipment Institute of General Logistics, Beijing, 100010, China
| | - Naixun Zhang
- College of Forestry, Northeast Forestry University, Harbin, Heilongjiang, 150040, China
| | - Zhenyu Wang
- Department of Food Science and Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, 150090, China
| |
Collapse
|
33
|
Cytotoxicity and Genotoxicity Assessment of Sandalwood Essential Oil in Human Breast Cell Lines MCF-7 and MCF-10A. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:3696232. [PMID: 27293457 PMCID: PMC4879231 DOI: 10.1155/2016/3696232] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 04/10/2016] [Accepted: 04/19/2016] [Indexed: 11/17/2022]
Abstract
Sandalwood essential oil (SEO) is extracted from Santalum trees. Although α-santalol, a main constituent of SEO, has been studied as a chemopreventive agent, the genotoxic activity of the whole oil in human breast cell lines is still unknown. The main objective of this study was to assess the cytotoxic and genotoxic effects of SEO in breast adenocarcinoma (MCF-7) and nontumorigenic breast epithelial (MCF-10A) cells. Proteins associated with SEO genotoxicity were identified using a proteomics approach. Commercially available, high-purity, GC/MS characterized SEO was used to perform the experiments. The main constituents reported in the oil were (Z)-α-santalol (25.34%), (Z)-nuciferol (18.34%), (E)-β-santalol (10.97%), and (E)-nuciferol (10.46%). Upon exposure to SEO (2-8 μg/mL) for 24 hours, cell proliferation was determined by the MTT assay. Alkaline and neutral comet assays were used to assess genotoxicity. SEO exposure induced single- and double-strand breaks selectively in the DNA of MCF-7 cells. Quantitative LC/MS-based proteomics allowed identification of candidate proteins involved in this response: Ku70 (p = 1.37E - 2), Ku80 (p = 5.8E - 3), EPHX1 (p = 3.3E - 3), and 14-3-3ζ (p = 4.0E - 4). These results provide the first evidence that SEO is genotoxic and capable of inducing DNA single- and double-strand breaks in MCF-7 cells.
Collapse
|
34
|
Cui FM, Liu L, Zheng LL, Bao GL, Tu Y, Sun L, Zhu W, Cao JP, Zhou PK, Chen Q, He YM. The Role of miR-34a in Tritiated Water Toxicity in Human Umbilical Vein Endothelial Cells. Dose Response 2016; 14:1559325816638585. [PMID: 27099602 PMCID: PMC4822198 DOI: 10.1177/1559325816638585] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
In this work, we investigated the toxic effects of tritiated water (HTO) on the cardiovascular system. We examined the role of microRNA-34a (miR-34a) in DNA damage and repair in human umbilical vein endothelial cells (HUVECs) exposed to HTO. Cell proliferation capacity was evaluated by cell counting, and miR-34a expression was detected using quantitative PCR (QT-PCR). The Comet assay and γ-H2AX immunostaining were used to measure DNA double-strand breaks (DSBs). Reverse transcription polymerase chain reaction was used to measure the expression level of c-myc messenger RNA (mRNA). The cells exposed to HTO showed significantly lower proliferation than the control cells over 3 days. The DNA damage in the HTO group was more severe than that in the control group, at each time point examined. The expression of miR-34a mimics caused increased DNA DSBs whereas that of the miR-34a inhibitor caused decreased DNA DSBs. The proliferation viability was the opposite for the miR-34a mimics and inhibitor groups. The expression levels of c-myc mRNA in cells transfected with miR-34a mimics were lower than that in cells transfected with the miR-34a-5p inhibitor, at 0.5 hours and 2 hours after transfection. In summary, miR-34a mediates HTO toxicity in HUVECs by downregulating the expression of c-myc.
Collapse
Affiliation(s)
- Feng Mei Cui
- Department of Radiation Medicine, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China; Collaborative Innovation Center of Radiation Medicine, Jiangsu Higher Education Institutions, Suzhou, China
| | - Liang Liu
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lu Lin Zheng
- Department of Radiation Medicine, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China
| | - Guang Liang Bao
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China
| | - Yu Tu
- Department of Radiation Medicine, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China; Collaborative Innovation Center of Radiation Medicine, Jiangsu Higher Education Institutions, Suzhou, China
| | - Liang Sun
- Department of Radiation Medicine, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China; Collaborative Innovation Center of Radiation Medicine, Jiangsu Higher Education Institutions, Suzhou, China
| | - Wei Zhu
- Department of Radiation Medicine, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China; Collaborative Innovation Center of Radiation Medicine, Jiangsu Higher Education Institutions, Suzhou, China
| | - Jian Ping Cao
- Department of Radiation Medicine, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China; Collaborative Innovation Center of Radiation Medicine, Jiangsu Higher Education Institutions, Suzhou, China
| | - Ping Kun Zhou
- Department of Radiation Medicine, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China; Collaborative Innovation Center of Radiation Medicine, Jiangsu Higher Education Institutions, Suzhou, China
| | - Qiu Chen
- Department of Radiation Medicine, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China; Collaborative Innovation Center of Radiation Medicine, Jiangsu Higher Education Institutions, Suzhou, China
| | - Yong Ming He
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
35
|
Frères P, Josse C, Bovy N, Boukerroucha M, Struman I, Bours V, Jerusalem G. Neoadjuvant Chemotherapy in Breast Cancer Patients Induces miR-34a and miR-122 Expression. J Cell Physiol 2015; 230:473-81. [PMID: 25078559 DOI: 10.1002/jcp.24730] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 07/25/2014] [Indexed: 12/17/2022]
Abstract
Circulating microRNAs (miRNAs) have been extensively studied in cancer as biomarkers but little is known regarding the influence of anti-cancer drugs on their expression levels. In this article, we describe the modifications of circulating miRNAs profile after neoadjuvant chemotherapy (NAC) for breast cancer. The expression of 188 circulating miRNAs was assessed in the plasma of 25 patients before and after NAC by RT-qPCR. Two miRNAs, miR-34a and miR-122, that were significantly increased after NAC, were measured in tumor tissue before and after chemotherapy in 7 patients with pathological partial response (pPR) to NAC. These two chemotherapy-induced miRNAs were further studied in the plasma of 22 patients with adjuvant chemotherapy (AC) as well as in 12 patients who did not receive any chemotherapy. Twenty-five plasma miRNAs were modified by NAC. Among these miRNAs, miR-34a and miR-122 were highly upregulated, notably in pPR patients with aggressive breast cancer. Furthermore, miR-34a level was elevated in the remaining tumor tissue after NAC treatment. Studying the kinetics of circulating miR-34a and miR-122 expression during NAC revealed that their levels were especially increased after anthracycline-based chemotherapy. Comparisons of the plasma miRNA profiles after NAC and AC suggested that chemotherapy-induced miRNAs originated from both tumoral and non-tumoral compartments. This study is the first to demonstrate that NAC specifically induces miRNA expression in plasma and tumor tissue, which might be involved in the anti-tumor effects of chemotherapy in breast cancer patients.
Collapse
Affiliation(s)
- Pierre Frères
- University of Liège, Laboratory of Medical Oncology, Liège, Belgium
| | - Claire Josse
- University of Liège, Laboratory of Medical Oncology, Liège, Belgium
| | - Nicolas Bovy
- University of Liège, GIGA-Research, Molecular Angiogenesis Laboratory, Liège, Belgium
| | | | - Ingrid Struman
- University of Liège, GIGA-Research, Molecular Angiogenesis Laboratory, Liège, Belgium
| | - Vincent Bours
- University of Liège, GIGA-Research, Human Genetics, Liège, Belgium
| | - Guy Jerusalem
- University of Liège, Laboratory of Medical Oncology, Liège, Belgium
| |
Collapse
|
36
|
Takeda Y, Venkitaraman AR. Micro(mi) RNA-34a targets protein phosphatase (PP)1γ to regulate DNA damage tolerance. Cell Cycle 2015; 14:3830-41. [PMID: 26111201 PMCID: PMC4825746 DOI: 10.1080/15384101.2015.1064202] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The DNA damage response (DDR) triggers widespread changes in gene expression, mediated partly by alterations in micro(mi) RNA levels, whose nature and significance remain uncertain. Here, we report that miR-34a, which is upregulated during the DDR, modulates the expression of protein phosphatase 1γ (PP1γ) to regulate cellular tolerance to DNA damage. Multiple bio-informatic algorithms predict that miR-34a targets the PP1CCC gene encoding PP1γ protein. Ionising radiation (IR) decreases cellular expression of PP1γ in a dose-dependent manner. An miR-34a-mimic reduces cellular PP1γ protein. Conversely, an miR-34a inhibitor antagonizes IR-induced decreases in PP1γ protein expression. A wild-type (but not mutant) miR-34a seed match sequence from the 3' untranslated region (UTR) of PP1CCC when transplanted to a luciferase reporter gene makes it responsive to an miR-34a-mimic. Thus, miR-34a upregulation during the DDR targets the 3' UTR of PP1CCC to decrease PP1γ protein expression. PP1γ is known to antagonize DDR signaling via the ataxia-telangiectasia-mutated (ATM) kinase. Interestingly, we find that cells exposed to DNA damage become more sensitive - in an miR-34a-dependent manner - to a second challenge with damage. Increased sensitivity to the second challenge is marked by enhanced phosphorylation of ATM and p53, increased γH2AX formation, and increased cell death. Increased sensitivity can be partly recapitulated by a miR-34a-mimic, or antagonized by an miR-34a-inhibitor. Thus, our findings suggest a model in which damage-induced miR-34a induction reduces PP1γ expression and enhances ATM signaling to decrease tolerance to repeated genotoxic challenges. This mechanism has implications for tumor suppression and the response of cancers to therapeutic radiation.
Collapse
Affiliation(s)
- Yuko Takeda
- The Medical Research Council Cancer Unit; University of Cambridge; Cambridge, UK
| | - Ashok R Venkitaraman
- The Medical Research Council Cancer Unit; University of Cambridge; Cambridge, UK,Correspondence to: Ashok R Venkitaraman;
| |
Collapse
|
37
|
van Schooneveld E, Wildiers H, Vergote I, Vermeulen PB, Dirix LY, Van Laere SJ. Dysregulation of microRNAs in breast cancer and their potential role as prognostic and predictive biomarkers in patient management. Breast Cancer Res 2015; 17:21. [PMID: 25849621 PMCID: PMC4332424 DOI: 10.1186/s13058-015-0526-y] [Citation(s) in RCA: 215] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) are an emerging class of gene expression modulators with relevant roles in several biological processes, including cell differentiation, development, apoptosis, and regulation of the cell cycle. Deregulation of those tiny RNA molecules has been described frequently as a major determinant for the initiation and progression of diseases, including cancer. Not only miRNAs but also the enzymes responsible for miRNA processing could be deregulated in cancer. In this review, we address the role of miRNAs in the pathogenesis of breast cancer, since there are oncogenic, tumor-suppressive, and metastatic-influencing miRNAs. Additionally, the different detection platforms and normalization strategies for miRNAs will be discussed. The major part of this review, however, will focus on the capability of miRNAs to act as diagnostic, predictive, or prognostic biomarkers. We will give an overview of their potential to correlate with response to or benefit from a given treatment and we will consider their ability to give information on prognosis in breast cancer. We will focus on miRNAs validated by more than one study or verified in independent cohorts or where results rely on preclinical as well as clinical evidence. As such, we will discuss their potential use in the personalized management of breast cancer.
Collapse
|
38
|
Misso G, Di Martino MT, De Rosa G, Farooqi AA, Lombardi A, Campani V, Zarone MR, Gullà A, Tagliaferri P, Tassone P, Caraglia M. Mir-34: a new weapon against cancer? MOLECULAR THERAPY-NUCLEIC ACIDS 2014; 3:e194. [PMID: 25247240 PMCID: PMC4222652 DOI: 10.1038/mtna.2014.47] [Citation(s) in RCA: 384] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Accepted: 08/11/2014] [Indexed: 02/07/2023]
Abstract
The microRNA(miRNA)-34a is a key regulator of tumor suppression. It controls the
expression of a plethora of target proteins involved in cell cycle, differentiation
and apoptosis, and antagonizes processes that are necessary for basic cancer cell
viability as well as cancer stemness, metastasis, and chemoresistance. In this
review, we focus on the molecular mechanisms of miR-34a-mediated tumor suppression,
giving emphasis on the main miR-34a targets, as well as on the principal regulators
involved in the modulation of this miRNA. Moreover, we shed light on the miR-34a role
in modulating responsiveness to chemotherapy and on the phytonutrients-mediated
regulation of miR-34a expression and activity in cancer cells. Given the broad
anti-oncogenic activity of miR-34a, we also discuss the substantial benefits of a new
therapeutic concept based on nanotechnology delivery of miRNA mimics. In fact, the
replacement of oncosuppressor miRNAs provides an effective strategy against tumor
heterogeneity and the selective RNA-based delivery systems seems to be an excellent
platform for a safe and effective targeting of the tumor.
Collapse
Affiliation(s)
- Gabriella Misso
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Graecia University and Medical OncologyUnit, T. Campanella Cancer Center, Salvatore Venuta University Campus, Catanzaro, Italy
| | - Giuseppe De Rosa
- Department of Pharmacy, University "Federico II" of Naples, Naples, Italy
| | - Ammad Ahmad Farooqi
- Laboratory for Translational Oncology and Personalized Medicine, Rashid Latif Medical College, Lahore, Pakistan
| | - Angela Lombardi
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Virginia Campani
- Department of Pharmacy, University "Federico II" of Naples, Naples, Italy
| | - Mayra Rachele Zarone
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Annamaria Gullà
- Department of Experimental and Clinical Medicine, Magna Graecia University and Medical OncologyUnit, T. Campanella Cancer Center, Salvatore Venuta University Campus, Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University and Medical OncologyUnit, T. Campanella Cancer Center, Salvatore Venuta University Campus, Catanzaro, Italy
| | - Pierfrancesco Tassone
- 1] Department of Experimental and Clinical Medicine, Magna Graecia University and Medical OncologyUnit, T. Campanella Cancer Center, Salvatore Venuta University Campus, Catanzaro, Italy [2] Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, USA
| | - Michele Caraglia
- 1] Department of Experimental and Clinical Medicine, Magna Graecia University and Medical OncologyUnit, T. Campanella Cancer Center, Salvatore Venuta University Campus, Catanzaro, Italy [2] Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
39
|
Long Z, Wang B, Tao D, Huang Y, Tao Z. Hypofractionated radiotherapy induces miR-34a expression and enhances apoptosis in human nasopharyngeal carcinoma cells. Int J Mol Med 2014; 34:1388-94. [PMID: 25231528 DOI: 10.3892/ijmm.2014.1937] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Accepted: 09/10/2014] [Indexed: 11/05/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a relatively radiosensitive disease. However, the therapeutic effects of radiotherapy are not always satisfactory due to radioresistance. The hypofractionated schema is currently widely used in clinical practice. In the present study, we investigated the effects of hypofractionated radiotherapy on NPC cells and explored the mechanisms involved. In addition, we aimed to determine the role of miR-34a in the effects of hypofractionated radiotherapy and whether these effects occur in a p53-dependent manner. For this purpose, we used CNE1 and CNE2 NPC cells which were subjected to hyperfractionated and hypofractionated radiotherapy. The viability of the cells was measured by MTT assay and acridine orange (AO) and ethidium bromide (EB) staining was used to observe morphological changes. In addition, Annexin V-propidium iodide (PI) staining and flow cytometry were used to determine the number of apoptotic cells and mRNA and protein expression was measured by qPCR and western blot analysis, respectively. The results revealed that hypofractionated radiotherapy enhanced apoptosis and increased the expression of miR-34a and p53 in the NPC cells. In addition, it stimulated p53 promoter activity and downregulated the protein expression of c-Myc in the human NPC cells. Furthermore, the knockdown of miR-34a suppressed the growth inhibitory effects induced by hypofractionated radiotherapy. Thus, our results suggest that the enhanced apoptosis of NPC cells may be associated with the miR-34a-mediated suppression of c-Myc in a p53-dependent manner.
Collapse
Affiliation(s)
- Zhixiong Long
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Bin Wang
- Department of Otolaryngology (ENT), The Fifth Hospital of Wuhan, Wuhan, Hubei 430051, P.R. China
| | - Dan Tao
- Department of Oncology, The Fifth Hospital of Wuhan, Wuhan, Hubei 430051, P.R. China
| | - Ying Huang
- Department of Oncology, The Fifth Hospital of Wuhan, Wuhan, Hubei 430051, P.R. China
| | - Zezhang Tao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
40
|
The role of microRNAs in human breast cancer progression. Tumour Biol 2014; 35:6235-44. [DOI: 10.1007/s13277-014-2202-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Accepted: 06/06/2014] [Indexed: 12/15/2022] Open
|
41
|
Yu G, Li H, Wang J, Gumireddy K, Li A, Yao W, Tang K, Xiao W, Hu J, Xiao H, Lang B, Ye Z, Huang Q, Xu H. miRNA-34a suppresses cell proliferation and metastasis by targeting CD44 in human renal carcinoma cells. J Urol 2014; 192:1229-37. [PMID: 24866595 DOI: 10.1016/j.juro.2014.05.094] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2014] [Indexed: 01/05/2023]
Abstract
PURPOSE We investigated the potential functions of miR-34a in CD44 transcriptional complexes in renal cell carcinoma. MATERIALS AND METHODS We detected miR-34a expression by quantitative real-time polymerase chain reaction. Oligonucleotides were used to over express miR-34a. Cell proliferation and xenograft assays, colony formation and flow cytometry were done to examine effects on cancer cell proliferation in vitro and in vivo. Luciferase assay was performed to verify the precise target of miR-34a. RESULTS Promoter methylation contributed to miR-34a loss in the ACHN, 786-O and SN12PM6 renal carcinoma cell lines. Ectopic over expression of miR-34a restrained cell growth, tube formation and migration/invasion, and significantly suppressed the growth of renal carcinoma xenografts and metastasis in nude mice. Dual luciferase assay revealed that CD44 was a direct target of miR-34a in renal cancer cells and CD44 knockdown by RNAi in renal cancer cells suppressed tumor progression. In contrast, CD44 ectopic expression partially reversed the antitumor effects of miR-34a in renal cancer cells. CONCLUSIONS Our findings indicate that miR-34a targets CD44 in renal cancer cells and suppresses renal cancer cell growth, tube formation and metastasis in vitro and in vivo. Thus, miR-34a may be a potential molecular target for novel therapeutic strategies for clear cell renal carcinoma.
Collapse
Affiliation(s)
- Gan Yu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Heng Li
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Ji Wang
- Department of Cell Death and Cancer Genetics, The Hormel Institute, University of Minnesota, Austin, Minnesota
| | | | - Anping Li
- The Wistar Institute, Philadelphia, Pennsylvania
| | - Weimin Yao
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Kun Tang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Wei Xiao
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Junhui Hu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Haibing Xiao
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Bin Lang
- School of Health Sciences, Macao Polytechnic Institute, Macao, People's Republic of China
| | - Zhangqun Ye
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Qihong Huang
- The Wistar Institute, Philadelphia, Pennsylvania
| | - Hua Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|