1
|
Guo Q, Yang W, Robinson G, Chaludiya K, Abdulkadir AN, Roy FG, Shivakumar D, Ahmad AN, Abdulkadir SA, Kirschner AN. Unlocking the Radiosensitizing Potential of MYC inhibition in Neuroendocrine Malignancies. Int J Radiat Oncol Biol Phys 2025:S0360-3016(25)00431-6. [PMID: 40354951 DOI: 10.1016/j.ijrobp.2025.04.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 04/04/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025]
Abstract
The MYC family of transcription factors-comprising c-MYC, N-MYC, and L-MYC-plays a pivotal role in oncogenesis, driving cancer progression and resistance to therapy. While MYC proteins have long been considered challenging drug targets due to their intricate structures, recent advances have led to the development of promising inhibitors. This review explores the role of MYC overexpression in promoting radiotherapy resistance in aggressive neuroendocrine malignancies through multiple mechanisms, including increased tumor cell invasion, enhanced DNA damage repair and oxidative stress management, pro-survival autophagy, survival of circulating tumor cells, angiogenesis, awakening from dormancy, and modulation of chronic inflammation and host immunity. Paradoxically, MYC overexpression can also enhance radiosensitivity in certain cancer cells by driving pro-apoptotic pathways, such as reactive oxygen species (ROS)-induced DNA damage that overwhelms cellular repair mechanisms, ultimately leading to cell death. Additionally, we provide a comprehensive summary of direct MYC inhibitors, detailing their current stage of preclinical and clinical development as novel anti-cancer therapeutics. This review highlights MYC's role in cancer metastasis and radiotherapy resistance while examining the potential of MYC inhibitors as radiosensitizers in adult and pediatric neuroendocrine malignancies, including small cell lung cancer, large cell neuroendocrine lung cancer, Merkel cell carcinoma, neuroendocrine-differentiated prostate cancer, neuroblastoma, central nervous system embryonal tumors, and medulloblastoma.
Collapse
Affiliation(s)
- Qianyu Guo
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, USA 32224; Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, USA 32224; Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida, USA; Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611; The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611.
| | - William Yang
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611; The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611.
| | - Guy Robinson
- Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, USA 32224; Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida, USA.
| | - Keyur Chaludiya
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905
| | | | - Falguni Ghosh Roy
- MGM Institute of Health Sciences, Navi Mumbai, Maharashtra, India 410209
| | - Divya Shivakumar
- Kamineni Academy of Medical Science and Research Centre, Hyderabad, Telangana, India 500068
| | - Ayesha N Ahmad
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611; The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611; Boonshoft School of Medicine, Wright State University, Fairborn, OH, USA 45324
| | - Sarki A Abdulkadir
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611; The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611.
| | - Austin N Kirschner
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN, USA 37232.
| |
Collapse
|
2
|
Guerini AE, Mataj E, Borghetti P, Triggiani L, Pegurri L, Nici S, Riga S, Tucci A, Belotti A, Bonù ML, Facheris G, Magrini SM, Spiazzi L, Buglione M. Impact of the Number of Administered Systemic Treatment Lines on Local Response to Radiation Therapy for Multiple Myeloma. Adv Radiat Oncol 2025; 10:101696. [PMID: 39911722 PMCID: PMC11795776 DOI: 10.1016/j.adro.2024.101696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 11/21/2024] [Indexed: 02/07/2025] Open
Abstract
Purpose Multiple myeloma (MM) tends to develop resistance to systemic therapy through multiple mechanisms that might as well induce radioresistance, as suggested by preclinical studies. The aim of the present analysis was to elucidate whether the number of systemic treatment lines received prior to radiation therapy (RT) might confer radioresistance and influence local response. Methods and Materials This single-center retrospective study enrolled patients who received RT for MM at our institution between January 1, 2005, and January 31, 2023. Information regarding RT, systemic therapy, and characteristics of the patients and disease were retrieved from medical records. The primary outcome for this analysis was radiologic local response at 6 months after RT, according to RECIST 1.1 (Response Evaluation Criteria in Solid Tumors) or PERCIST 1.0 (Positron Emission Tomography Response Criteria in Solid Tumors) criteria. The secondary outcome was toxicity reported during the RT course. Results Data from 665 MM lesions from 366 patients were analyzed. Data regarding local response at 6 months were available for 217 lesions, reporting 29 complete responses (13.4%), 141 partial responses (65%), 42 stable diseases (19.4%), and only 5 disease progressions (2.3%). The number of previous systemic treatment lines had no impact on radiologic response at 6 months (p = .721). RT BED10 (Biologically Effective Dose) had a significant impact on response at 6 months (p = .007). The toxicity profile was optimal, as grade > 2 events during RT were reported only in 0.9% of cases. Conclusions In this large retrospective cohort of MM patients, the number of systemic treatment lines administered before RT had no impact on the local response, confuting concerns of cross-resistance raised by multiple preclinical studies. Disease control after RT was optimal, and instances of severe toxicities during treatment were rare.
Collapse
Affiliation(s)
- Andrea Emanuele Guerini
- Department of Radiation Oncology, University of Brescia and Spedali Civili Hospital, Brescia, Italy
| | - Eneida Mataj
- Department of Radiation Oncology, University of Brescia and Spedali Civili Hospital, Brescia, Italy
| | - Paolo Borghetti
- Department of Radiation Oncology, University of Brescia and Spedali Civili Hospital, Brescia, Italy
| | - Luca Triggiani
- Department of Radiation Oncology, University of Brescia and Spedali Civili Hospital, Brescia, Italy
| | - Ludovica Pegurri
- Department of Radiation Oncology, University of Brescia and Spedali Civili Hospital, Brescia, Italy
| | - Stefania Nici
- Medical Physics Department, ASST Spedali Civili Hospital, Brescia, Italy
| | - Stefano Riga
- Medical Physics Department, ASST Spedali Civili Hospital, Brescia, Italy
| | - Alessandra Tucci
- Department of Haematology, ASST Spedali Civili Hospital, Brescia, Italy
| | - Angelo Belotti
- Department of Haematology, ASST Spedali Civili Hospital, Brescia, Italy
| | - Marco Lorenzo Bonù
- Department of Radiation Oncology, University of Brescia and Spedali Civili Hospital, Brescia, Italy
| | - Giorgio Facheris
- Department of Radiation Oncology, University of Brescia and Spedali Civili Hospital, Brescia, Italy
| | - Stefano Maria Magrini
- Dipartimento di Specialità Medico-Chirurgiche, Centro per lo Studio della Radioterapia guidata dalle Immagini e dai Biomarkers (BIO-RT), Scienze Radiologiche e Sanità Pubblica, University of Brescia, Brescia, Italy
| | - Luigi Spiazzi
- Department of Haematology, ASST Spedali Civili Hospital, Brescia, Italy
| | - Michela Buglione
- Department of Radiation Oncology, University of Brescia and Spedali Civili Hospital, Brescia, Italy
| |
Collapse
|
3
|
Fang XL, Cao XP, Xiao J, Hu Y, Chen M, Raza HK, Wang HY, He X, Gu JF, Zhang KJ. Overview of role of survivin in cancer: expression, regulation, functions, and its potential as a therapeutic target. J Drug Target 2024; 32:223-240. [PMID: 38252514 DOI: 10.1080/1061186x.2024.2309563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/11/2023] [Indexed: 01/24/2024]
Abstract
Survivin holds significant importance as a member of the inhibitor of apoptosis protein (IAP) family due to its predominant expression in tumours rather than normal terminally differentiated adult tissues. The high expression level of survivin in tumours is closely linked to chemotherapy resistance, heightened tumour recurrence, and increased tumour aggressiveness and serves as a negative prognostic factor for cancer patients. Consequently, survivin has emerged as a promising therapeutic target for cancer treatment. In this review, we delve into the various biological characteristics of survivin in cancers and its pivotal role in maintaining immune system homeostasis. Additionally, we explore different therapeutic strategies aimed at targeting survivin.
Collapse
Affiliation(s)
- Xian-Long Fang
- Academician Expert Workstation of Fengxian District, Shanghai Yuansong Biotechnology Limited Company, Shanghai, China
| | - Xue-Ping Cao
- Academician Expert Workstation of Fengxian District, Shanghai Yuansong Biotechnology Limited Company, Shanghai, China
| | - Jun Xiao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Yun Hu
- Academician Expert Workstation of Fengxian District, Shanghai Yuansong Biotechnology Limited Company, Shanghai, China
| | - Mian Chen
- Academician Expert Workstation of Fengxian District, Shanghai Yuansong Biotechnology Limited Company, Shanghai, China
| | - Hafiz Khuram Raza
- Academician Expert Workstation of Fengxian District, Shanghai Yuansong Biotechnology Limited Company, Shanghai, China
| | - Huai-Yuan Wang
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xu He
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jin-Fa Gu
- Academician Expert Workstation of Fengxian District, Shanghai Yuansong Biotechnology Limited Company, Shanghai, China
| | - Kang-Jian Zhang
- Academician Expert Workstation of Fengxian District, Shanghai Yuansong Biotechnology Limited Company, Shanghai, China
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
4
|
Kanagasabai T, Hawaz M, Ellis K, Fah O, Mikhaeil H, Nguyen P, Tombo N, Shanker A, Sampath C, Khoury ZH, Cade J, Ferguson A, Gangula P. Effects of Topoisomerase II alpha Inhibition on Oral Cancer Cell Metabolism and Cancer Stem Cell Function. DENTAL RESEARCH AND ORAL HEALTH 2024; 7:58-65. [PMID: 38957610 PMCID: PMC11218736 DOI: 10.26502/droh.0076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Background Topoisomerase IIα (TOP2A), is an enzyme involved in DNA replication, transcription, recombination, and chromatin remodeling and is found in a variety of cancers. However, the role of TOP2A regulation in oral cancer progression is not fully explained. We investigated the effect of TOP2A inhibition on cell survival, metabolism, and cancer stem cell self-renewal function in oral cancer cells. Methods Oral carcinoma cell line SCC25 was cultured in complete DMEM/F12 media and treated with 5μM of Etoposide (Topoisomerase II inhibitor) for 48h. The critical parameters of cellular metabolism, including extracellular acidification rate (ECAR) and mitochondrial oxidative phosphorylation based on the oxygen consumption rate of cancer cells were assessed using Seahorse assay. Western blotting was performed to assess the proteins that are associated with proliferation (Survivin, IL-6) and cancer stem cell function (Oct4, Sox2) in cell lysates prepared from control and etoposide treated groups. Statistical analysis was performed using One-way ANOVA with Dunnett's multiple comparisons test. Results The protein expression of TOP2A was significantly (P<0.05) inhibited by etoposide. Additionally, TOP2A inhibition decreased the mitochondrial respiratory parameters including basal respiration, maximal respiration and ATP production. However, TOP2A inhibition has no impact on glycolytic function. Moreover, the proliferative marker survivin and IL-6 showed a significant (P<0.05) decrease after TOP2A inhibition. Conversely, the protein expression of cancer stem cell markers Oct-4 and Sox 2 were not altered. Conclusion These results indicate that inhibition of TOP2A is more efficacious by decreasing the mitochondrial metabolic reprogramming and thereby downregulating the key anti-apoptotic and pro-survival mediators. Thus, TOP2A represents an ideal therapeutic target and offers a potential treatment strategy for OSCC.
Collapse
Affiliation(s)
- Thanigaivelan Kanagasabai
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA
| | - Mariam Hawaz
- Department of Oral Diagnostic Sciences and Research, School of Dentistry, Meharry Medical College, Nashville, TN, 37208, USA
| | - Kayla Ellis
- Department of Oral Diagnostic Sciences and Research, School of Dentistry, Meharry Medical College, Nashville, TN, 37208, USA
| | - Orlyne Fah
- Department of Oral Diagnostic Sciences and Research, School of Dentistry, Meharry Medical College, Nashville, TN, 37208, USA
| | - Helana Mikhaeil
- Department of Oral Diagnostic Sciences and Research, School of Dentistry, Meharry Medical College, Nashville, TN, 37208, USA
| | - Philip Nguyen
- Department of Oral Diagnostic Sciences and Research, School of Dentistry, Meharry Medical College, Nashville, TN, 37208, USA
| | - Nathalie Tombo
- Department of Oral Diagnostic Sciences and Research, School of Dentistry, Meharry Medical College, Nashville, TN, 37208, USA
| | - Anil Shanker
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Chethan Sampath
- Department of Oral Diagnostic Sciences and Research, School of Dentistry, Meharry Medical College, Nashville, TN, 37208, USA
| | - Zaid H Khoury
- Department of Oral Diagnostic Sciences and Research, School of Dentistry, Meharry Medical College, Nashville, TN, 37208, USA
| | - James Cade
- Department of Oral Diagnostic Sciences and Research, School of Dentistry, Meharry Medical College, Nashville, TN, 37208, USA
| | - Alexys Ferguson
- Department of Oral Diagnostic Sciences and Research, School of Dentistry, Meharry Medical College, Nashville, TN, 37208, USA
| | - Pandu Gangula
- Department of Oral Diagnostic Sciences and Research, School of Dentistry, Meharry Medical College, Nashville, TN, 37208, USA
| |
Collapse
|
5
|
Liu W, Wang Y, Xia L, Li J. Research Progress of Plant-Derived Natural Products against Drug-Resistant Cancer. Nutrients 2024; 16:797. [PMID: 38542707 PMCID: PMC10975298 DOI: 10.3390/nu16060797] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 01/04/2025] Open
Abstract
As one of the malignant diseases globally, cancer seriously endangers human physical and mental health because of its high morbidity and mortality. Conventional cancer treatment strategies, such as surgical resection and chemoradiotherapy, are effective at the early stage of cancer but have limited efficacy for advanced cancer. Along with cancer progress and treatment, resistance develops gradually within the population of tumor cells. As a consequence, drug resistance become the major cause that leads to disease progression and poor clinical prognosis in some patients. The mechanisms of cancer drug resistance are quite complex and involve various molecular and cellular mechanisms. Therefore, exploring the mechanisms and finding specific targets are becoming imperative to overcome drug resistance. In recent years, plant-derived natural products have been evaluated as potential therapeutic candidates against cancer with drug resistance due to low side effects and high anticancer efficacy. A growing number of studies have shown that natural products can achieve superior antitumor effects through multiple signaling pathways. The mechanisms include regulation of multiple drug resistance (MDR)-related genes, inhibition of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway, induction of autophagy, and blockade of the cell cycle. This paper reviews the molecular and cellular mechanisms of cancer drug resistance, as well as the therapeutic effects and mechanisms of plant-derived natural products against cancer drug resistance. It provides references for developing therapeutic medication for drug-resistant cancer treatment with high efficacy and low side effects.
Collapse
Affiliation(s)
| | | | - Lijie Xia
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China; (W.L.); (Y.W.)
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China; (W.L.); (Y.W.)
| |
Collapse
|
6
|
Rahimian S, Najafi H, Afzali B, Doroudian M. Extracellular Vesicles and Exosomes: Novel Insights and Perspectives on Lung Cancer from Early Detection to Targeted Treatment. Biomedicines 2024; 12:123. [PMID: 38255228 PMCID: PMC10813125 DOI: 10.3390/biomedicines12010123] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/24/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Lung cancer demands innovative approaches for early detection and targeted treatment. In addressing this urgent need, exosomes play a pivotal role in revolutionizing both the early detection and targeted treatment of lung cancer. Their remarkable capacity to encapsulate a diverse range of biomolecules, traverse biological barriers, and be engineered with specific targeting molecules makes them highly promising for both diagnostic markers and precise drug delivery to cancer cells. Furthermore, an in-depth analysis of exosomal content and biogenesis offers crucial insights into the molecular profile of lung tumors. This knowledge holds significant potential for the development of targeted therapies and innovative diagnostic strategies for cancer. Despite notable progress in this field, challenges in standardization and cargo loading persist. Collaborative research efforts are imperative to maximize the potential of exosomes and advance the field of precision medicine for the benefit of lung cancer patients.
Collapse
Affiliation(s)
| | | | | | - Mohammad Doroudian
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran 14911-15719, Iran; (S.R.); (H.N.); (B.A.)
| |
Collapse
|
7
|
Bagheri R, Rassouli FB, Gholamhosseinian H, Ebrahimi K, Mahdavi S, Goudarzi S, Iranshahi M, Rafatpanah H, Keramati MR. Radiation Response of Human Leukemia/Lymphoma Cells was Improved by 7-Geranyloxycoumarin. Dose Response 2022; 20:15593258221124479. [PMID: 36158737 PMCID: PMC9500271 DOI: 10.1177/15593258221124479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Objectives Adult T-cell leukemia/lymphoma (ATLL) is a blood neoplasm with specific geographic distribution. Although radiotherapy is a palliative treatment that provides long-term local control, single use of radiation leads to complications for patients. To introduce a novel multimodal approach against ATLL, we investigated combinatorial effects of 7-geranyloxycoumarin and radiation in vitro. Methods Viability of MT-2 cells was determined by resazurin assay upon administration of 7-geranyloxycoumarin alone and followed by radiation. Then, apoptosis was detected by annexin V and propidium iodide, and the expression of candidate genes was analyzed by qPCR. Results Findings revealed significant (P<.0001) improvement in radiation effects upon 7-geranyloxycoumarin pretreatment, most notably when cells were pretreated with 5 µg/ml 7-geranyloxycoumarin for 96 h, exposed to 6 Gy radiation and recovered for 48 h. These results were confirmed by flow cytometry, as the percentage of early and late apoptotic cells was increased after combinatorial treatment. In addition, significant (P< .0001) changes in CD44, c-MYC, cFLIPL, BMI-1, NF-κB (Rel A), and P53 expression was induced by 7-geranyloxycoumarin and radiation. Conclusions Current research indicated, for the first time, that combinatorial use of 7-geranyloxycoumarin and ionizing radiation could be considered as an effective therapeutic modality for ATLL.
Collapse
Affiliation(s)
- Ramin Bagheri
- Cancer Molecular Pathology Research Center, Department of Hematology and Blood Bank, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh B. Rassouli
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hamid Gholamhosseinian
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Keyhan Ebrahimi
- Cancer Molecular Pathology Research Center, Department of Hematology and Blood Bank, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shakiba Mahdavi
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Sajad Goudarzi
- Cancer Molecular Pathology Research Center, Department of Hematology and Blood Bank, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Houshang Rafatpanah
- Immunology Research Center, Inflammation and inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Keramati
- Cancer Molecular Pathology Research Center, Department of Hematology and Blood Bank, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
8
|
Liu L, He L, Li W, Zhao T, Li G, Xiu X, Xu Y, Bourbonne V, Käsmann L, Kowalchuk RO, Zhong Q. The miR-4306/IGF2R axis modulates the lung adenocarcinoma response to irradiation in vitro and in vivo. Transl Lung Cancer Res 2021; 10:4494-4510. [PMID: 35070756 PMCID: PMC8743517 DOI: 10.21037/tlcr-21-890] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 12/22/2021] [Indexed: 11/13/2022]
Abstract
BACKGROUND Lung adenocarcinoma accounts for more than 50% of non-small cell lung cancers. Dysregulated microRNAs (miRNAs) and coding genes play a critical role in lung adenocarcinoma irradiation resistance and might be promising therapeutic targets. In the present study, we demonstrate the effect of the miR-4306/IGF2R axis on malignant behaviors of lung adenocarcinoma cells and the response to irradiation. METHODS Quantitative realtime-PCR and Western blot assays were applied for miR-4306 and IGF2R expression in tumors and cells. A CCK-8 assay kit was used to detect cell viability. Colony formation assay was implied to detect cell proliferation. Transwell assay was used to detect cell invasion. A subcutaneous tumor model was performed in nude mice to detect tumor formation in vivo. Hematoxylin & eosin (H&E) staining were used to observe pathological status of tumor in nude mice. To validate the miR-4306 binding IGF2R 3'-UTR, a dual-luciferase reporter assay was performed. RESULTS The expression level of miR-4306 was dramatically upregulated in lung adenocarcinoma samples and cells, and could be induced by irradiation in a dose-dependent manner. In lung adenocarcinoma cells, miR-4306 overexpression significantly promoted cell viability and invasive abilities and attenuated the inhibitory effect of irradiation on malignant cancer cell behaviors. In a subcutaneous tumor model in nude mice, miR-4306 overexpression promoted tumor growth and attenuated the suppressive effect of irradiation on tumor growth. miR-4306 directly inhibited the expression of IGF2R. In lung adenocarcinoma cells without irradiation, IGF2R overexpression was inhibited, while IGF2R knockdown promoted cell viability and invasive abilities. The effects of miR-4306 overexpression were partially attenuated by IGF2R overexpression. In lung adenocarcinoma cells, suppressive role of irradiation on cancer cell viability and invasive abilities were enhanced by IGF2R overexpression, but attenuated by IGF2R knockdown. The effects of miR-4306 overexpression on cancer cell viability and invasive abilities were also partially attenuated by IGF2R overexpression in lung adenocarcinoma cells with irradiation. In tissue samples, expression of miR-4306 and IGF2R were negatively correlated. CONCLUSIONS The miR-4306/IGF2R axis could significantly affect lung adenocarcinoma progression and response to radiotherapy, and further investigation of the clinical implications of this axis is strongly recommended.
Collapse
Affiliation(s)
- Lipin Liu
- Department of Radiation Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Lei He
- Department of Pathology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Wenhan Li
- Department of Pharmacy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Ting Zhao
- Department of Radiation Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Gaofeng Li
- Department of Radiation Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xia Xiu
- Department of Radiation Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Yonggang Xu
- Department of Radiation Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | | | - Lukas Käsmann
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | | | - Qiuzi Zhong
- Department of Radiation Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
9
|
de Mey S, Dufait I, De Ridder M. Radioresistance of Human Cancers: Clinical Implications of Genetic Expression Signatures. Front Oncol 2021; 11:761901. [PMID: 34778082 PMCID: PMC8579106 DOI: 10.3389/fonc.2021.761901] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Although radiotherapy is given to more than 50% of cancer patients, little progress has been made in identifying optimal radiotherapy - drug combinations to improve treatment efficacy. Using molecular data from The Cancer Genome Atlas (TCGA), we extracted a total of 1016 cancer patients that received radiotherapy. The patients were diagnosed with head-and-neck (HNSC - 294 patients), cervical (CESC - 166 patients) and breast (BRCA - 549 patients) cancer. We analyzed mRNA expression patterns of 50 hallmark gene sets of the MSigDB collection, which we divided in eight categories based on a shared biological or functional process. Tumor samples were split into upregulated, neutral or downregulated mRNA expression for all gene sets using a gene set analysis (GSEA) pre-ranked analysis and assessed for their clinical relevance. We found a prognostic association between three of the eight gene set categories (Radiobiological, Metabolism and Proliferation) and overall survival in all three cancer types. Furthermore, multiple single associations were revealed in the other categories considered. To the best of our knowledge, our study is the first report suggesting clinical relevance of molecular characterization based on hallmark gene sets to refine radiation strategies.
Collapse
Affiliation(s)
- Sven de Mey
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Inès Dufait
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mark De Ridder
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
10
|
Soofiyani SR, Hosseini K, Soleimanian A, Abkhooei L, Hoseini AM, Tarhriz V, Ghasemnejad T. An Overview on the Role of miR-451 in Lung Cancer: Diagnosis, Therapy, and Prognosis. Microrna 2021; 10:181-190. [PMID: 34514995 DOI: 10.2174/2211536610666210910130828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/15/2021] [Accepted: 08/03/2021] [Indexed: 11/22/2022]
Abstract
MicroRNAs (miRNAs) are highly conserved non-coding RNAs involved in many physiological processes such as cell proliferation, inhibition, development of apoptosis, differentiation, suppresses tumorigenicity, and regulating cell growth. The description of the alterations of miRNA expression patterns in cancers will be helpful to recognize biomarkers for early detection and possible therapeutic intervention in the treatment of cancers. Recent studies have shown that miR-451 is broadly dysregulated in lung cancer and is a crucial agent in lung tumor progression. This review summarizes recent advances of the potential role of miR-451 in lung cancer diagnosis, prognosis, and treatment and provides an insight into the potential use of miR-451 for the development of advanced therapeutic methods in lung cancer.
Collapse
Affiliation(s)
- Saiedeh Razi Soofiyani
- Clinical Research Development Unit of Sina Educational, Research and Treatment Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Kamram Hosseini
- Student research committee, Shiraz University of Medical Sciences, Shiraz. Iran
| | - Alireza Soleimanian
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz. Iran
| | - Liela Abkhooei
- Department of Medical Biotechnology, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad. Iran
| | - Akbar Mohammad Hoseini
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine and Tabriz Blood Transfusion Center, Tabriz. Iran
| | - Vahideh Tarhriz
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Tohid Ghasemnejad
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz. Iran
| |
Collapse
|
11
|
Nguyen L, Schilling D, Dobiasch S, Raulefs S, Santiago Franco M, Buschmann D, Pfaffl MW, Schmid TE, Combs SE. The Emerging Role of miRNAs for the Radiation Treatment of Pancreatic Cancer. Cancers (Basel) 2020; 12:cancers12123703. [PMID: 33317198 PMCID: PMC7763922 DOI: 10.3390/cancers12123703] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/17/2020] [Accepted: 12/04/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Pancreatic cancer is an aggressive disease with a high mortality rate. Radiotherapy is one treatment option within a multimodal therapy approach for patients with locally advanced, non-resectable pancreatic tumors. However, radiotherapy is only effective in about one-third of the patients. Therefore, biomarkers that can predict the response to radiotherapy are of utmost importance. Recently, microRNAs, small non-coding RNAs regulating gene expression, have come into focus as there is growing evidence that microRNAs could serve as diagnostic, predictive and prognostic biomarkers in various cancer entities, including pancreatic cancer. Moreover, their high stability in body fluids such as serum and plasma render them attractive candidates for non-invasive biomarkers. This article describes the role of microRNAs as suitable blood biomarkers and outlines an overview of radiation-induced microRNAs changes and the association with radioresistance in pancreatic cancer. Abstract Today, pancreatic cancer is the seventh leading cause of cancer-related deaths worldwide with a five-year overall survival rate of less than 7%. Only 15–20% of patients are eligible for curative intent surgery at the time of diagnosis. Therefore, neoadjuvant treatment regimens have been introduced in order to downsize the tumor by chemotherapy and radiotherapy. To further increase the efficacy of radiotherapy, novel molecular biomarkers are urgently needed to define the subgroup of pancreatic cancer patients who would benefit most from radiotherapy. MicroRNAs (miRNAs) could have the potential to serve as novel predictive and prognostic biomarkers in patients with pancreatic cancer. In the present article, the role of miRNAs as blood biomarkers, which are associated with either radioresistance or radiation-induced changes of miRNAs in pancreatic cancer, is discussed. Furthermore, the manuscript provides own data of miRNAs identified in a pancreatic cancer mouse model as well as radiation-induced miRNA changes in the plasma of tumor-bearing mice.
Collapse
Affiliation(s)
- Lily Nguyen
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany; (L.N.); (D.S.); (S.D.); (S.R.); (M.S.F.); (T.E.S.)
- Department of Radiation Oncology, School of Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany
| | - Daniela Schilling
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany; (L.N.); (D.S.); (S.D.); (S.R.); (M.S.F.); (T.E.S.)
- Department of Radiation Oncology, School of Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany
| | - Sophie Dobiasch
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany; (L.N.); (D.S.); (S.D.); (S.R.); (M.S.F.); (T.E.S.)
- Department of Radiation Oncology, School of Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, 81675 Munich, Germany
| | - Susanne Raulefs
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany; (L.N.); (D.S.); (S.D.); (S.R.); (M.S.F.); (T.E.S.)
- Department of Radiation Oncology, School of Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany
| | - Marina Santiago Franco
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany; (L.N.); (D.S.); (S.D.); (S.R.); (M.S.F.); (T.E.S.)
| | - Dominik Buschmann
- Division of Animal Physiology and Immunology, TUM School of Life Sciences Weihenstephan, Technical University of Munich (TUM), 85354 Freising, Germany; (D.B.); (M.W.P.)
| | - Michael W. Pfaffl
- Division of Animal Physiology and Immunology, TUM School of Life Sciences Weihenstephan, Technical University of Munich (TUM), 85354 Freising, Germany; (D.B.); (M.W.P.)
| | - Thomas E. Schmid
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany; (L.N.); (D.S.); (S.D.); (S.R.); (M.S.F.); (T.E.S.)
- Department of Radiation Oncology, School of Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany
| | - Stephanie E. Combs
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany; (L.N.); (D.S.); (S.D.); (S.R.); (M.S.F.); (T.E.S.)
- Department of Radiation Oncology, School of Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, 81675 Munich, Germany
- Correspondence: ; Tel.: +49-89-4140-4501
| |
Collapse
|
12
|
LncRNA SNHG15 regulates EGFR-TKI acquired resistance in lung adenocarcinoma through sponging miR-451 to upregulate MDR-1. Cell Death Dis 2020; 11:525. [PMID: 32655137 PMCID: PMC7354989 DOI: 10.1038/s41419-020-2683-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/01/2020] [Accepted: 02/03/2020] [Indexed: 12/14/2022]
Abstract
Lung adenocarcinoma (LUAD) is the main component of non-small-cell lung cancer (NSCLC) and causes a great health concern globally. The top priority of LUAD treatment is to deal with gefitinib resistance. Long non-coding RNAs are certified to modify gefitinib resistance in the course of tumor aggravation. The study focuses on addressing the function of small nucleolar RNA host gene 15 (SNHG15) on modifying gefitinib resistance in LUAD. Previously, NOTCH pathway is implicated in LUAD chemo-resistance. SNHG15 level was boosted following the depletion of NOTCH-1 in A549/GR and H1975/GR cells. Functional studies indicated that SNHG15 and multidrug resistance protein 1 (MDR-1) were overexpressed and possess tumor-promoting functions in gefitinib-resistant LUAD cells while miR-451 was downregulated and possess tumor-suppressive behaviors in gefitinib-resistant LUAD cells. Mechanically, the SNHG15 was cytoplasmically distributed in GR LUAD cells. In addition, SNHG15 released MDR-1 from the suppression of miR-451, leading to MDR-1 promotion. In addition, the elevation of SNHG15 could be attributed to ZEB1. Rescue assays highlighted that downstream molecules MDR-1 and miR-451 could reverse the effects of SNHG15 downregulation on gefitinib-resistant LUAD cells. SNHG15 could alter chemo-resistance of LUAD cells to Gefitinib via regulating miR-451/MDR-1, which could be inspiring findings for the advancement of chemo-therapies for LUAD.
Collapse
|
13
|
Moertl S, Buschmann D, Azimzadeh O, Schneider M, Kell R, Winkler K, Tapio S, Hornhardt S, Merl-Pham J, Pfaffl MW, Atkinson MJ. Radiation Exposure of Peripheral Mononuclear Blood Cells Alters the Composition and Function of Secreted Extracellular Vesicles. Int J Mol Sci 2020; 21:ijms21072336. [PMID: 32230970 PMCID: PMC7178185 DOI: 10.3390/ijms21072336] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/20/2020] [Accepted: 03/24/2020] [Indexed: 12/24/2022] Open
Abstract
Normal tissue toxicity is a dose-limiting factor in radiation therapy. Therefore, a detailed understanding of the normal tissue response to radiation is necessary to predict the risk of normal tissue toxicity and to development strategies for tissue protection. One component of normal tissue that is continuously exposed during therapeutic irradiation is the circulating population of peripheral blood mononuclear cells (PBMC). PBMCs are highly sensitive to ionizing radiation (IR); however, little is known about how IR affects the PBMC response on a systemic level. It was the aim of this study to investigate whether IR was capable to induce changes in the composition and function of extracellular vesicles (EVs) secreted from PBMCs after radiation exposure to different doses. Therefore, whole blood samples from healthy donors were exposed to X-ray radiation in the clinically relevant doses of 0, 0.1, 2 or 6 Gy and PBMC-secreted EVs were isolated 72 h later. Proteome and miRNome analysis of EVs as well as functional studies were performed. Secreted EVs showed a dose-dependent increase in the number of significantly deregulated proteins and microRNAs. For both, proteome and microRNA data, principal component analysis showed a dose-dependent separation of control and exposed groups. Integrated pathway analysis of the radiation-regulated EV proteins and microRNAs consistently predicted an association of deregulated molecules with apoptosis, cell death and survival. Functional studies identified endothelial cells as an efficient EV recipient system, in which irradiation of recipient cells further increased the uptake. Furthermore an apoptosis suppressive effect of EVs from irradiated PBMCs in endothelial recipient cells was detected. In summary, this study demonstrates that IR modifies the communication between PBMCs and endothelial cells. EVs from irradiated PBMC donors were identified as transmitters of protective signals to irradiated endothelial cells. Thus, these data may lead to the discovery of biomarker candidates for radiation dosimetry and even more importantly, they suggest EVs as a novel systemic communication pathway between irradiated normal, non-cancer tissues.
Collapse
Affiliation(s)
- Simone Moertl
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Radiation Biology, 85764 Neuherberg, Germany; (O.A.); (M.S.); (R.K.); (K.W.); (S.T.); (M.J.A.)
- Federal Office for Radiation Protection, 85764 Oberschleißheim, Germany; (S.H.); (M.W.P.)
- Correspondence:
| | - Dominik Buschmann
- Division of Animal Physiology and Immunology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany;
| | - Omid Azimzadeh
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Radiation Biology, 85764 Neuherberg, Germany; (O.A.); (M.S.); (R.K.); (K.W.); (S.T.); (M.J.A.)
| | - Michael Schneider
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Radiation Biology, 85764 Neuherberg, Germany; (O.A.); (M.S.); (R.K.); (K.W.); (S.T.); (M.J.A.)
| | - Rosemarie Kell
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Radiation Biology, 85764 Neuherberg, Germany; (O.A.); (M.S.); (R.K.); (K.W.); (S.T.); (M.J.A.)
| | - Klaudia Winkler
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Radiation Biology, 85764 Neuherberg, Germany; (O.A.); (M.S.); (R.K.); (K.W.); (S.T.); (M.J.A.)
| | - Soile Tapio
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Radiation Biology, 85764 Neuherberg, Germany; (O.A.); (M.S.); (R.K.); (K.W.); (S.T.); (M.J.A.)
| | - Sabine Hornhardt
- Federal Office for Radiation Protection, 85764 Oberschleißheim, Germany; (S.H.); (M.W.P.)
| | - Juliane Merl-Pham
- Helmholtz Zentrum München, German Research Center for Environmental Health, Research Unit Protein Science, 80939 München, Germany;
| | - Michael W. Pfaffl
- Federal Office for Radiation Protection, 85764 Oberschleißheim, Germany; (S.H.); (M.W.P.)
| | - Michael J. Atkinson
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Radiation Biology, 85764 Neuherberg, Germany; (O.A.); (M.S.); (R.K.); (K.W.); (S.T.); (M.J.A.)
- Chair of Radiation Biology, Technical University of Munich, 80333 Munich, Germany
| |
Collapse
|
14
|
Bai H, Wu S. miR-451: A Novel Biomarker and Potential Therapeutic Target for Cancer. Onco Targets Ther 2019; 12:11069-11082. [PMID: 31908476 PMCID: PMC6924581 DOI: 10.2147/ott.s230963] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 11/28/2019] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are endogenous, non-coding, single-stranded small RNAs involved in a variety of cellular processes, including ontogeny, cell proliferation, differentiation, and apoptosis. They can also function as oncogenes or tumor suppressor genes. Recent studies have revealed that miRNA-451 (miR-451) is involved in the regulation of various human physiological and pathological processes. Furthermore, it has been shown that miR-451 not only directly affects the biological functions of tumor cells but also indirectly affects tumor cell invasion and metastasis upon secretion into the tumor microenvironment via exosomes. Thus, miR-451 also influences the progression of tumorigenesis and drug resistance. This review summarizes the expression of miR-451 in various cancer types and the relationship between miR-451 and the diagnosis, treatment, and drug resistance of solid tumors. In addition, we address possible mechanisms of action of miR-451 and its potential application as a biomarker in the diagnosis and treatment of human cancers.
Collapse
Affiliation(s)
- Hua Bai
- Department of Gynecology and Obstetrics, Shanxi Dayi Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Suhui Wu
- Department of Gynecology and Obstetrics, Shanxi Dayi Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| |
Collapse
|
15
|
|
16
|
Post AEM, Bussink J, Sweep FCGJ, Span PN. Changes in DNA Damage Repair Gene Expression and Cell Cycle Gene Expression Do Not Explain Radioresistance in Tamoxifen-Resistant Breast Cancer. Oncol Res 2019; 28:33-40. [PMID: 31046897 PMCID: PMC7851527 DOI: 10.3727/096504019x15555794826018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Tamoxifen-induced radioresistance, reported in vitro, might pose a problem for patients who receive neoadjuvant tamoxifen treatment and subsequently receive radiotherapy after surgery. Previous studies suggested that DNA damage repair or cell cycle genes are involved, and could therefore be targeted to preclude the occurrence of cross-resistance. We aimed to characterize the observed cross-resistance by investigating gene expression of DNA damage repair genes and cell cycle genes in estrogen receptor-positive MCF-7 breast cancer cells that were cultured to tamoxifen resistance. RNA sequencing was performed, and expression of genes characteristic for several DNA damage repair pathways was investigated, as well as expression of genes involved in different phases of the cell cycle. The association of differentially expressed genes with outcome after radiotherapy was assessed in silico in a large breast cancer cohort. None of the DNA damage repair pathways showed differential gene expression in tamoxifen-resistant cells compared to wild-type cells. Two DNA damage repair genes were more than two times upregulated (NEIL1 and EME2), and three DNA damage repair genes were more than two times downregulated (PCNA, BRIP1, and BARD1). However, these were not associated with outcome after radiotherapy in the TCGA breast cancer cohort. Genes involved in G1, G1/S, G2, and G2/M phases were lower expressed in tamoxifen-resistant cells compared to wild-type cells. Individual genes that were more than two times upregulated (MAPK13) or downregulated (E2F2, CKS2, GINS2, PCNA, MCM5, and EIF5A2) were not associated with response to radiotherapy in the patient cohort investigated. We assessed the expression of DNA damage repair genes and cell cycle genes in tamoxifen-resistant breast cancer cells. Though several genes in both pathways were differentially expressed, these could not explain the cross-resistance for irradiation in these cells, since no association to response to radiotherapy in the TCGA breast cancer cohort was found.
Collapse
Affiliation(s)
- Annemarie E M Post
- Radboud University Medical Center, Department of Radiation Oncology, Radiotherapy and OncoImmunology LaboratoryNijmegenThe Netherlands
| | - Johan Bussink
- Radboud University Medical Center, Department of Radiation Oncology, Radiotherapy and OncoImmunology LaboratoryNijmegenThe Netherlands
| | - Fred C G J Sweep
- Radboud University Medical Center, Department of Laboratory MedicineNijmegenThe Netherlands
| | - Paul N Span
- Radboud University Medical Center, Department of Radiation Oncology, Radiotherapy and OncoImmunology LaboratoryNijmegenThe Netherlands
| |
Collapse
|
17
|
The Roles of MicroRNA in Lung Cancer. Int J Mol Sci 2019; 20:ijms20071611. [PMID: 30935143 PMCID: PMC6480472 DOI: 10.3390/ijms20071611] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/11/2019] [Accepted: 03/26/2019] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is the most devastating malignancy in the world. Beyond genetic research, epigenomic studies—especially investigations of microRNAs—have grown rapidly in quantity and quality in the past decade. This has enriched our understanding about basic cancer biology and lit up the opportunities for potential therapeutic development. In this review, we summarize the involvement of microRNAs in lung cancer carcinogenesis and behavior, by illustrating the relationship to each cancer hallmark capability, and in addition, we briefly describe the clinical applications of microRNAs in lung cancer diagnosis and prognosis. Finally, we discuss the potential therapeutic use of microRNAs in lung cancer.
Collapse
|
18
|
MicroRNA-451 Inhibits Migration of Glioblastoma while Making It More Susceptible to Conventional Therapy. Noncoding RNA 2019; 5:ncrna5010025. [PMID: 30875963 PMCID: PMC6468936 DOI: 10.3390/ncrna5010025] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/08/2019] [Accepted: 03/11/2019] [Indexed: 12/18/2022] Open
Abstract
Malignant glioblastoma (GBM, glioma) is the most common and aggressive primary adult brain tumor. The prognosis of GBM patients remains poor, despite surgery, radiation and chemotherapy. The major obstacles for successful remedy are invasiveness and therapy resistance of GBM cells. Invasive glioma cells leave primary tumor core and infiltrate surrounding normal brain leading to inevitable recurrence, even after surgical resection, radiation and chemotherapy. Therapy resistance allowing for selection of more aggressive and resistant sub-populations including GBM stem-like cells (GSCs) upon treatment is another serious impediment to successful treatment. Through their regulation of multiple genes, microRNAs can orchestrate complex programs of gene expression and act as master regulators of cellular processes. MicroRNA-based therapeutics could thus impact broad cellular programs, leading to inhibition of invasion and sensitization to radio/chemotherapy. Our data show that miR-451 attenuates glioma cell migration in vitro and invasion in vivo. In addition, we have found that miR-451 sensitizes glioma cells to conventional chemo- and radio-therapy. Our data also show that miR-451 is regulated in vivo by AMPK pathway and that AMPK/miR-451 loop has the ability to switch between proliferative and migratory pattern of glioma cells behavior. We therefore postulate that AMPK/miR-451 negative reciprocal feedback loop allows GBM cells/GSCs to adapt to tumor “ecosystem” by metabolic and behavioral flexibility, and that disruption of such a loop reduces invasiveness and diminishes therapy resistance.
Collapse
|
19
|
Du M, Wang J, Chen H, Wang S, Chen L, Xu Y, Su F, Lu X. MicroRNA‑200a suppresses migration and invasion and enhances the radiosensitivity of NSCLC cells by inhibiting the HGF/c‑Met signaling pathway. Oncol Rep 2018; 41:1497-1508. [PMID: 30569179 PMCID: PMC6365696 DOI: 10.3892/or.2018.6925] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 11/28/2018] [Indexed: 12/16/2022] Open
Abstract
Hepatocyte growth factor (HGF), an activator of the c‑Met signaling pathway, is involved in tumor invasiveness, metastasis and radiotherapy resistance. In the present study, a novel HGF regulatory pathway in lung cancer involving micro-RNAs (miRNAs/miR) is described. Immunohistochemical staining and western blot analyses demonstrated that HGF was upregulated and associated with miR‑200a downregulation in non‑small cell lung cancer (NSCLC) samples compared with normal lung tissues. The association between HGF and miR‑200a was associated with the degree of tumor malignancy and cell migration and invasion. miR‑200a negatively regulated HGF expression by targeting the 3'‑untranslated region of the HGF mRNA. miR‑200a overexpression induced HGF downregulation, decreased NSCLC cell migration and invasion, promoted apoptosis, and decreased cell survival in A549 and H1299 cells in response to ionizing radiation. The present results revealed a previously uncharacterized role of miRNA‑200a in regulating tumor malignancy and radiosensitivity by suppressing HGF expression, a key factor in the HGF/c‑Met pathway.
Collapse
Affiliation(s)
- Menghua Du
- Department of Oncology and Radiotherapy, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Jin Wang
- Department of Oncology and Radiotherapy, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Huan Chen
- Department of Oncology and Radiotherapy, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Shouli Wang
- Department of Pathology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Liesong Chen
- Department of Oncology and Radiotherapy, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Yichang Xu
- Department of Pathology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Fengtao Su
- Cancer Institute, Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Xueguan Lu
- Department of Oncology and Radiotherapy, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| |
Collapse
|
20
|
Lu J, Zhan Y, Feng J, Luo J, Fan S. MicroRNAs associated with therapy of non-small cell lung cancer. Int J Biol Sci 2018; 14:390-397. [PMID: 29725260 PMCID: PMC5930471 DOI: 10.7150/ijbs.22243] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 02/25/2018] [Indexed: 12/30/2022] Open
Abstract
Background & Objective: The incidence of non-small cell lung cancer (NSCLC) has been rising over the past several decades. Despite various therapeutic regimens and modern diagnostic techniques are developed, NSCLC still have an extremely poor prognosis due to drug resistance. Therefore, it is critical to find a novel precise diagnosis and effective treatment approach for NSCLC patients. MicroRNAs (MiRNAs) are a class of 18-25nt non-coding small RNAs, which have been shown to be involved profoundly in the pathogenesis such as cellular proliferation, differentiation, development, apoptosis and tumorigenesis in many human tumors including of NSCLC. We reviewed existing research literature regarding correlations between miRNAs and their target's response to anticancer treatment, and summarized the recent findings between miRNAs and therapy availability in NSCLC.
Collapse
Affiliation(s)
- Junmi Lu
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuting Zhan
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juan Feng
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiadi Luo
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Songqing Fan
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
21
|
Ding L, Zhang Y, Han L, Fu L, Mei X, Wang J, Itkow J, Elabid AEI, Pang L, Yu D. Activating and sustaining c-Myc by depletion of miR-144/451 gene locus contributes to B-lymphomagenesis. Oncogene 2017; 37:1293-1307. [PMID: 29284789 PMCID: PMC6168470 DOI: 10.1038/s41388-017-0055-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 09/15/2017] [Accepted: 11/03/2017] [Indexed: 02/06/2023]
Abstract
Hyper activity of protooncogene c-Myc is one of the hallmarks of highly aggressive lymphomas. However, the mechanism of how c-Myc is subjected to activation and amplification is still not well defined. In this study, we use gene knockout strategy to show that targeted depletion of a well-conserved microRNA gene locus miR-144/451 initiates tumorigenesis including B-lymphoma development in aged mice. This is due, at least in part, to the direct activation of the c-Myc gene by loss of miR-144/451 expression in hematopoietic cells. Moreover, oncoprotein c-Myc inversely regulates miR-144/451 expression by directly binding to the miR-144/451 promoter region, forming a miRNA-Myc positive feedback loop to safeguard the high level of c-Myc in B-lymphocytes. We also demonstrate that this miRNA-Myc crosstalk is disrupted in human diffuse large B-cell lymphomas with aberrant c-Myc expression. Therefore, our findings provide strong evidence, for the first time, that deficiency of miR-144/451 expression may play a bona fide role in derepression of silenced c-Myc, which contributes to tumor development including B-lymphomagenesis.
Collapse
Affiliation(s)
- Lan Ding
- Department of Pathology, Jiangdu People's Hospital, Yangzhou University, Yangzhou, China
| | - Yanqing Zhang
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University School of Medicine, Yangzhou, Jiangsu Province, China
| | - Lingling Han
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University School of Medicine, Yangzhou, Jiangsu Province, China
| | - Lei Fu
- Department of Pathology, Jiangdu People's Hospital, Yangzhou University, Yangzhou, China
| | - Xia Mei
- Department of Pathology, Jiangdu People's Hospital, Yangzhou University, Yangzhou, China
| | - Jijun Wang
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University School of Medicine, Yangzhou, Jiangsu Province, China
| | - Jacobi Itkow
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University School of Medicine, Yangzhou, Jiangsu Province, China
| | - Afaf Elabid Ibrahim Elabid
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University School of Medicine, Yangzhou, Jiangsu Province, China
| | - Lei Pang
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University School of Medicine, Yangzhou, Jiangsu Province, China
| | - Duonan Yu
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University School of Medicine, Yangzhou, Jiangsu Province, China. .,Institute of Translational Medicine, Yangzhou University School of Medicine, Yangzhou, China. .,Institute of Comparative Medicine, Yangzhou University, Yangzhou, China. .,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou, China.
| |
Collapse
|
22
|
Wang J, Wang Y, Mei H, Yin Z, Geng Y, Zhang T, Wu G, Lin Z. The BET bromodomain inhibitor JQ1 radiosensitizes non-small cell lung cancer cells by upregulating p21. Cancer Lett 2017; 391:141-151. [DOI: 10.1016/j.canlet.2017.01.031] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 01/19/2017] [Accepted: 01/20/2017] [Indexed: 01/07/2023]
|
23
|
Cui F, Hou J, Huang C, Sun X, Zeng Y, Cheng H, Wang H, Li C. C-Myc regulates radiation-induced G2/M cell cycle arrest and cell death in human cervical cancer cells. J Obstet Gynaecol Res 2017; 43:729-735. [DOI: 10.1111/jog.13261] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 10/10/2016] [Accepted: 11/08/2016] [Indexed: 12/26/2022]
Affiliation(s)
- Fengmei 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
| | - Jun Hou
- Department of Pathology; Zhongshan Hospital, Fudan University; Shanghai China
| | - Chengcheng Huang
- 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
| | - Xiujin 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
| | - Yanan Zeng
- 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
| | - Huiying Cheng
- 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
| | - Hao Wang
- Department of Oncology; the First Affiliated Hospital, Medical University of Anhui; Hefei China
| | - Chao Li
- Department of Radiotherapy and Oncology; Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine; Suzhou China
| |
Collapse
|
24
|
Lv Q, Hu JX, Li YJ, Xie N, Song DD, Zhao W, Yan YF, Li BS, Wang PY, Xie SY. MiR-320a effectively suppresses lung adenocarcinoma cell proliferation and metastasis by regulating STAT3 signals. Cancer Biol Ther 2017; 18:142-151. [PMID: 28106481 DOI: 10.1080/15384047.2017.1281497] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
MicroRNAs play important roles in tumorigenesis of various types of cancers. MiR-320a can inhibits cell proliferation of some cancers, but the biologic roles of miR-320a in lung cancer need to be further studied. Here, we investigated the roles of miR-320a in suppressing the proliferation of lung adenocarcinoma cells. MiR-320a treatment was found to effectively suppress LTEP-a-2 and A549 cell proliferation, and induce more apoptotic cells with irradiation treatment compared with control treatment. Our results also showed that miR-320a, as a novel miRNA, directly regulated signal transducer and activator of transcription 3 (STAT3) and its signals, such as Bcl-2, Bax, and Caspase 3. The siRNA-inhibited STAT3 levels further proved its roles in regulating STAT3 signals. Moreover, miR-320a treatment effectively suppressed cancer cell growth in mice xenografts compared with controls, and significantly inhibited cell migration in vitro and in vivo. Our findings collectively demonstrated that miR-320a, by directly regulating STAT3 signals, not only suppressed cell proliferation and metastasis, but also enhanced irradiation-induced apoptosis of adenocarcinomia cells.
Collapse
Affiliation(s)
- Qing Lv
- a Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology , Binzhou Medical University , YanTai , Shandong , P.R. China
| | - Jin-Xia Hu
- a Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology , Binzhou Medical University , YanTai , Shandong , P.R. China
| | - You-Jie Li
- a Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology , Binzhou Medical University , YanTai , Shandong , P.R. China
| | - Ning Xie
- b YanTaiShan Hospital , YanTai , Shandong , P.R. China
| | - Dan Dan Song
- a Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology , Binzhou Medical University , YanTai , Shandong , P.R. China
| | - Wei Zhao
- a Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology , Binzhou Medical University , YanTai , Shandong , P.R. China
| | - Yun-Fei Yan
- a Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology , Binzhou Medical University , YanTai , Shandong , P.R. China
| | - Bao-Sheng Li
- c Department of Radiation Oncology , Shandong Cancer Hospital, Shandong Academy of Medical Sciences , Jinan , P.R. China
| | - Ping-Yu Wang
- a Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology , Binzhou Medical University , YanTai , Shandong , P.R. China
| | - Shu-Yang Xie
- a Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology , Binzhou Medical University , YanTai , Shandong , P.R. China
| |
Collapse
|
25
|
Su R, Gong JN, Chen MT, Song L, Shen C, Zhang XH, Yin XL, Ning HM, Liu B, Wang F, Ma YN, Zhao HL, Yu J, Zhang JW. c-Myc suppresses miR-451⊣YWTAZ/AKT axis via recruiting HDAC3 in acute myeloid leukemia. Oncotarget 2016; 7:77430-77443. [PMID: 27764807 PMCID: PMC5363596 DOI: 10.18632/oncotarget.12679] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 09/20/2016] [Indexed: 12/16/2022] Open
Abstract
Aberrant activation of c-Myc plays an important oncogenic role via regulating a series of coding and non-coding genes in acute myeloid leukemia (AML). Histone deacetylases (HDACs) can remove acetyl group from histone and regulate gene expression via changing chromatin structure. Here, we found miR-451 is abnormally down-regulated in AML patient samples; c-Myc recruits HDAC3 to form a transcriptional suppressor complex, co-localizes on the miR-451 promoter, epigenetically inhibits its transcription and finally induces its downregulation in AML. Furthermore, our in vitro and in vivo results suggest that miR-451 functions as a tumor suppressor via promoting apoptosis and suppressing malignant cell proliferation. The mechanistic study demonstrated that miR-451 directly targets YWHAZ mRNA and suppresses YWHAZ/AKT signaling in AML. Knockdown of c-Myc results in restoration of miR-451 and inhibition of YWHAZ/AKT signaling. In AML patients, low level of miR-451 is negatively correlated with high levels of c-Myc and YWHAZ, while c-Myc level is positively related to YWHAZ expression. These results suggested that c-Myc⊣miR-451⊣YWHAZ/AKT cascade might play a crucial role during leukemogenesis, and reintroduction of miR-451 could be as a potential strategy for AML therapy.
Collapse
Affiliation(s)
- Rui Su
- The State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jia-Nan Gong
- The State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming-Tai Chen
- The State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Song
- The State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Shen
- The State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin-Hua Zhang
- Department of Hematology, The 303 Hospital, Nanning, Guangxi, China
| | - Xiao-Lin Yin
- Department of Hematology, The 303 Hospital, Nanning, Guangxi, China
| | - Hong-Mei Ning
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital to Academy of Military Medical Sciences, The 307 Hospital, Beijing, China
| | - Bing Liu
- State Key Laboratory of Proteomics, Translational Medicine Center of Stem Cells, 307-lvy Translational Medicine Center, Laboratory of Oncology, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Fang Wang
- The State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan-Ni Ma
- The State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hua-Lu Zhao
- The State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jia Yu
- The State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun-Wu Zhang
- The State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
26
|
Huang J, Chen Y, Li J, Zhang K, Chen J, Chen D, Feng B, Song H, Feng J, Wang R, Chen L. Notch-1 Confers Chemoresistance in Lung Adenocarcinoma to Taxanes through AP-1/microRNA-451 Mediated Regulation of MDR-1. MOLECULAR THERAPY. NUCLEIC ACIDS 2016; 5:e375. [PMID: 27727250 PMCID: PMC5095685 DOI: 10.1038/mtna.2016.82] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 08/22/2016] [Indexed: 12/28/2022]
Abstract
We previously demonstrated that expression of Notch-1 is associated with poor prognosis in lung adenocarcinoma (LAD) patients. The aim of this study is to reveal whether Notch-1 was associated with Taxanes-resistant LAD and, the underlying mechanisms. We collected 39 patients of advanced LAD treated with Taxanes and found that positive Notch-1 expression is closely related to LAD lymph node metastasis, recurrence and poorer prognosis, and Notch-1 acts as an independent poor prognostic factor in LAD by multivariate analysis with Cox regression model. Then, by using the Docetaxel (DTX)-resistant LAD cell lines that we established previously, we found that Notch-1 contributes to resistance of LAD cells to DTX in vitro, and inhibition of Notch-1 sensitizes LAD to DTX in vivo. We further demonstrated that Notch-1 mediates chemoresistance response and strengthens proliferation capacity in LAD cells partially through negative regulation of miR-451 by transcription factor AP-1. Moreover, we found that MDR-1 is a direct target of miR-451 and influences chemoresistance of LAD cells. Taken together, our data revealed a novel Notch-1/AP-1/miR-451/MDR-1 signaling axis, and suggested a new therapeutic strategy of combining DTX with Notch inhibitors to treat DTX-resistant LAD.
Collapse
Affiliation(s)
- Jiayuan Huang
- Department of Medical Oncology, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Jiangsu Institute of Cancer Research, Jiangsu, China.,Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Yitian Chen
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Junyang Li
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Kai Zhang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Jing Chen
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Dongqin Chen
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Bing Feng
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Haizhu Song
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Jifeng Feng
- Department of Medical Oncology, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Jiangsu Institute of Cancer Research, Jiangsu, China
| | - Rui Wang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Longbang Chen
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
27
|
Wang J, Lei ZJ, Guo Y, Wang T, Qin ZY, Xiao HL, Fan LL, Chen DF, Bian XW, Liu J, Wang B. miRNA-regulated delivery of lincRNA-p21 suppresses β-catenin signaling and tumorigenicity of colorectal cancer stem cells. Oncotarget 2016; 6:37852-70. [PMID: 26497997 PMCID: PMC4741970 DOI: 10.18632/oncotarget.5635] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 10/06/2015] [Indexed: 12/11/2022] Open
Abstract
Cancer stem cells (CSCs) are key cellular targets for effective cancer therapy, due to their critical roles in cancer progression and chemo/radio-resistance. Emerging evidence demonstrates that long non-coding RNAs (lncRNAs) are important players in the biology of cancers. However, it remains unknown whether lncRNAs could be exploited to target CSCs. We report that large intergenic non-coding RNA p21 (lincRNA-p21) is a potent suppressor of stem-like traits of CSCs purified from both primary colorectal cancer (CRC) tissues and cell lines. A novel lincRNA-p21-expressing adenoviral vector, which was armed with miRNA responsive element (MRE) of miR-451 (Ad-lnc-p21-MRE), was generated to eliminate CRC CSCs. Integration of miR-451 MREs into the adenovirus efficiently delivered lincRNA-p21 into CSCs that contained low levels of miR-451. Moreover, lincRNA-p21 inhibited the activity of β-catenin signaling, thereby attenuating the viability, self-renewal, and glycolysis of CSCs in vitro. By limiting dilution and serial tumor formation assay, we demonstrated that Ad-lnc-p21-MRE significantly suppressed the self-renewal potential and tumorigenicity of CSCs in nude mice. Importantly, application of miR-451 MREs appeared to protect normal liver cells from off-target expression of lincRNA-p21 in both tumor-bearing and naïve mice. Taken together, these findings suggest that lncRNAs may be promising therapeutic molecules to eradicate CSCs and MREs of tumor-suppressor miRNAs, such as miR-451, may be exploited to ensure the specificity of CSC-targeting strategies.
Collapse
Affiliation(s)
- Jun Wang
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Zeng-jie Lei
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Yan Guo
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Tao Wang
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Zhong-yi Qin
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Hua-liang Xiao
- Department of Pathology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Li-lin Fan
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Dong-feng Chen
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Xiu-wu Bian
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Jia Liu
- Institute of Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Bin Wang
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| |
Collapse
|
28
|
Ou DL, Shyue SK, Lin LI, Feng ZR, Liou JY, Fan HH, Lee BS, Hsu C, Cheng AL. Growth arrest DNA damage-inducible gene 45 gamma expression as a prognostic and predictive biomarker in hepatocellular carcinoma. Oncotarget 2016; 6:27953-65. [PMID: 26172295 PMCID: PMC4695037 DOI: 10.18632/oncotarget.4446] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 06/19/2015] [Indexed: 12/29/2022] Open
Abstract
Growth arrest DNA damage-inducible gene 45 (GADD45) family proteins play a crucial role in regulating cellular stress responses and apoptosis. The present study explored the prognostic and predictive role of GADD45γ in hepatocellular carcinoma (HCC) treatment. GADD45γ expression in HCC cells was examined using quantitative reverse transcription-PCR (qRT-PCR) and Western blotting. The control of GADD45γ transcription was examined using a luciferase reporter assay and chromatin immunoprecipitation. The in vivo induction of GADD45γ was performed using adenoviral transfer. The expression of GADD45γ in HCC tumor tissues from patients who had undergone curative resection was measured using qRT-PCR. Sorafenib induced expression of GADD45γ mRNA and protein, independent of its RAF kinase inhibitor activity. GADD45γ induction was more prominent in sorafenib-sensitive HCC cells (Huh-7 and HepG2, IC50 6–7 μM) than in sorafenib-resistant HCC cells (Hep3B, Huh-7R, and HepG2R, IC50 12–15 μM). Overexpression of GADD45γ reversed sorafenib resistance in vitro and in vivo, whereas GADD45γ expression knockdown by using siRNA partially abrogated the proapoptotic effects of sorafenib on sorafenib-sensitive cells. Overexpression of survivin in HCC cells abolished the antitumor enhancement between GADD45γ overexpression and sorafenib treatment, suggesting that survivin is a crucial mediator of antitumor effects of GADD45γ. GADD45γ expression decreased in tumors from patients with HCC who had undergone curative surgery, and low GADD45γ expression was an independent prognostic factor for poor survival, in addition to old age and vascular invasion. The preceding data indicate that GADD45γ suppression is a poor prognostic factor in patients with HCC and may help predict sorafenib efficacy in HCC.
Collapse
Affiliation(s)
- Da-Liang Ou
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan.,National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Song-Kun Shyue
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Liang-In Lin
- Graduate Institute of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Zi-Rui Feng
- National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan.,Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Jun-Yang Liou
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Hsiang-Hsuan Fan
- National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan.,Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Bin-Shyun Lee
- National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan.,Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chiun Hsu
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ann-Lii Cheng
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
29
|
Li J, Yang X, Guan H, Mizokami A, Keller ET, Xu X, Liu X, Tan J, Hu L, Lu Y, Zhang J. Exosome-derived microRNAs contribute to prostate cancer chemoresistance. Int J Oncol 2016; 49:838-46. [PMID: 27278879 DOI: 10.3892/ijo.2016.3560] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Accepted: 05/20/2016] [Indexed: 02/06/2023] Open
Abstract
Certain microRNAs (miRNAs) play a key role in cancer cell chemoresistance. However, the pleiotropic functions of exosome-derived miRNAs on developing chemoresistance remain unknown. In the present study, we aimed to construct potential networks of miRNAs, which derived from the exosome of chemoresistant prostate cancer (PCa) cells, with their known target genes using miRNA expression profiling and bioinformatic tools. Global miRNA expression profiles were measured by microarray. Twelve miRNAs were initially selected and validated by qRT-PCR. Known targets of deregulated miRNAs were utilized using DIANA-TarBase database v6.0. The incorporation of deregulated miRNAs and target genes into KEGG pathways were utilized using DIANA-mirPath software. To construct potential miRNA regulatory networks, the overlapping parts of miRNAs and their targer genes from the selected KEGG pathway 'PCa progression (hsa05215)' were visualized by Cytoscape software. We identified 29 deregulated miRNAs, including 19 upregulated and 10 downregulated, in exosome samples derived from two kinds of paclitaxel resistance PCa cells (PC3-TXR and DU145-TXR) compared with their parental cells (PC3 and DU145). The enrichment results of deregulated miRNAs and known target genes showed that a few pathways were correlated with several critical cell signaling pathways. We found that hub hsa-miR3176, -141-3p, -5004-5p, -16-5p, -3915, -488‑3p, -23c, -3673 and -3654 were potential targets to hub gene androgen receptor (AR) and phosphatase and tensin homolog (PTEN). Hub gene T-cell factors/lymphoid enhancer-binding factors 4 (TCF4) target genes were mainly regulated by hub hsa-miR-32-5, -141-3p, -606, -381 and -429. These results may provide a linkage between PCa chemoresistance and exosome regulatory networks and thus lead us to propose that AR, PTEN and TCF4 genes may be the important genes which are regulated by exosome miRNAs in chemoresistance cancer cells.
Collapse
Affiliation(s)
- Jing Li
- Key Laboratory of Longevity and Aging-Related Diseases, Ministry of Education, Nanning, Guangxi, P.R. China
| | - Xin Yang
- Key Laboratory of Longevity and Aging-Related Diseases, Ministry of Education, Nanning, Guangxi, P.R. China
| | - Hao Guan
- Key Laboratory of Longevity and Aging-Related Diseases, Ministry of Education, Nanning, Guangxi, P.R. China
| | | | - Evan T Keller
- Department of Urology and Pathology, School of Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Xiaozhen Xu
- Key Laboratory of Longevity and Aging-Related Diseases, Ministry of Education, Nanning, Guangxi, P.R. China
| | - Xia Liu
- Key Laboratory of Longevity and Aging-Related Diseases, Ministry of Education, Nanning, Guangxi, P.R. China
| | - Jiyong Tan
- Key Laboratory of Longevity and Aging-Related Diseases, Ministry of Education, Nanning, Guangxi, P.R. China
| | - Longyuan Hu
- Key Laboratory of Longevity and Aging-Related Diseases, Ministry of Education, Nanning, Guangxi, P.R. China
| | - Yi Lu
- Key Laboratory of Longevity and Aging-Related Diseases, Ministry of Education, Nanning, Guangxi, P.R. China
| | - Jian Zhang
- Key Laboratory of Longevity and Aging-Related Diseases, Ministry of Education, Nanning, Guangxi, P.R. China
| |
Collapse
|
30
|
Huang JY, Zhang K, Chen DQ, Chen J, Feng B, Song H, Chen Y, Zhu Z, Lu L, De W, Wang R, Chen LB. MicroRNA-451: epithelial-mesenchymal transition inhibitor and prognostic biomarker of hepatocelluar carcinoma. Oncotarget 2016; 6:18613-30. [PMID: 26164082 PMCID: PMC4621914 DOI: 10.18632/oncotarget.4317] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 05/12/2015] [Indexed: 02/06/2023] Open
Abstract
Increasing evidence indicates that dysregulation of microRNAs (miRNAs) plays critical roles in malignant transformation and tumor progression. Previously, we have shown that microRNA-451 (miR-451) inhibits growth, increases chemo- or radiosensitivity and reverses epithelial to mesenchymal transition (EMT) in lung cancer. However, the roles of miR-451 in hepatocelluar carcinoma (HCC) progression and metastasis are still largely unknown. Reduced miR-451 in HCC tissues was observed to be significantly correlated with advanced clinical stage, metastasis and worse disease-free or overall survival. Through gain- and loss-of function experiments, we demonstrated that miR-451 inhibited cell growth, induced G0/G1 arrest and promoted apoptosis in HCC cells. Importantly, miR-451 could inhibit the migration and invasion in vitro, as well as in vivo metastasis of HCC cells through regulating EMT process. Moreover, the oncogene c-Myc was identified as a direct and functional target of miR-451 in HCC cells. Knockdown of c-Myc phenocopied the effects of miR-451 on EMT and metastasis of HCC cells, whereas overexpression of c-Myc partially attenuated the functions of miR-451 restoration. Furthermore, miR-451 downregulation-induced c-Myc overexpression leads to the activation of Erk1/2 signaling, which induces acquisition of EMT phenotype through regulation of GSK-3β/snail/E-cadherin and the increased expression of MMPs family members in HCC cells. Collectively, these data demonstrated that miR-451 is a novel prognostic biomarker for HCC patients and that function as a potential metastasis inhibitor in HCC cells through activation of the Erk1/2 signaling, at least partially by targeting c-Myc. Thus, targeting miR-451/c-Myc/Erk1/2 axis may be a potential strategy for the treatment of metastatic HCC.
Collapse
Affiliation(s)
- Jia-Yuan Huang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Kai Zhang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Dong-Qin Chen
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Jing Chen
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Bing Feng
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Haizhu Song
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Yitian Chen
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Ziman Zhu
- Department of Hepatobiliary Surgery, First Hospital Affiliated to The Chinese PLA General Hospital, Beijing, China
| | - Lei Lu
- Liver Disease Center of PLA, The 81th Hospital of PLA, Nanjing, Jiangsu, China
| | - Wei De
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rui Wang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Long-Bang Chen
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
31
|
Li Y, Zhu Y, Prochownik EV. MicroRNA-based screens for synthetic lethal interactions with c-Myc. RNA & DISEASE 2016; 3:e1330. [PMID: 27975083 PMCID: PMC5152767 DOI: 10.14800/rd.1330] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
microRNAs (miRs) are small, non-coding RNAs, which play crucial roles in the development and progression of human cancer. Given that miRs are stable, easy to synthetize and readily introduced into cells, they have been viewed as having potential therapeutic benefit in cancer. c-Myc (Myc) is one of the most commonly deregulated oncogenic transcription factors and has important roles in the pathogenesis of cancer, thus making it an important, albeit elusive therapeutic target. Here we review the miRs that have been identified as being both positive and negative targets for Myc and how these participate in the complex phenotypes that arise as a result of Myc-driven transformation. We also discussseveral recent reports of Myc-synthetic lethal interactions with miRs.These highlight the importance and complexity of miRs in Myc-mediated biological functions and the opportunities for Myc-driven human cancer therapies.
Collapse
Affiliation(s)
- Youjun Li
- College of Life Sciences, Wuhan University, Wuhan 430072, China
- Medical Research Institute, Wuhan University, Wuhan 430071, China
| | - Yahui Zhu
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Edward V. Prochownik
- Division of Hematology/Oncology, Children's Hospital of Pittsburgh of UPMC and The Department of Microbiology and Molecular Genetics, The University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15224, USA
| |
Collapse
|
32
|
Bronisz A, Godlewski J, Chiocca EA. Extracellular Vesicles and MicroRNAs: Their Role in Tumorigenicity and Therapy for Brain Tumors. Cell Mol Neurobiol 2016; 36:361-76. [PMID: 26983830 DOI: 10.1007/s10571-015-0293-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/22/2015] [Indexed: 12/12/2022]
Abstract
MicroRNAs are small non-coding RNAs which mediate post-transcriptional gene regulation. Recently, microRNAs have also been found to be localized to the extracellular space, often encapsulated in secreted extracellular vesicles (EVs). This tandem of EVs and tissue-specific expressed/secreted microRNAs that can be taken up by neighboring or distant recipient cells, leading to changes in gene expression-suggests a cell-specialized role in physiological and pathological conditions. The complexity of solid tumors and their distinct pathophysiology relies on interactive communications between the various cell types in the neoplasm (tumor, endothelial, or macrophages, for instance). Understanding how such EV/microRNA-mediated communication occurs may actually lead to avenues for therapeutic exploitation and/or intervention, particularly for the most formidable cancers, such as those in the brain. In this review, the role of microRNAs/EVs in brain tumors will be discussed with emphasis on how these molecules could be utilized for tumor therapy.
Collapse
Affiliation(s)
- Agnieszka Bronisz
- Department of Neurosurgery, Harvey Cushing Neuro-oncology Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jakub Godlewski
- Department of Neurosurgery, Harvey Cushing Neuro-oncology Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - E Antonio Chiocca
- Department of Neurosurgery, Harvey Cushing Neuro-oncology Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
33
|
Li J, Wu J, Zhang J, Wang Y, Fang L, Shen Q. Oral bioavailability and evaluation of docetaxel–nicotinamide complex loaded chitosan nanoparticles. RSC Adv 2016. [DOI: 10.1039/c5ra27590c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
DTX–NA/NPs, synergism of DTX–NA complex and positively charged chitosan nanoparticles, can considerably enhance oral bioavailability.
Collapse
Affiliation(s)
- Jing Li
- School of Pharmacy
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| | - Jian Wu
- Department of Pharmaceutical Sciences
- Shenyang Pharmaceutical University
- Shenyang 110016
- China
| | - Jun Zhang
- School of Pharmacy
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| | - Yiyue Wang
- School of Pharmacy
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| | - Liang Fang
- Department of Pharmaceutical Sciences
- Shenyang Pharmaceutical University
- Shenyang 110016
- China
| | - Qi Shen
- School of Pharmacy
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| |
Collapse
|
34
|
Xiao M, Li W. Recent Advances on Small-Molecule Survivin Inhibitors. Curr Med Chem 2015; 22:1136 - 1146. [PMID: 25613234 DOI: 10.2174/0929867322666150114102146] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 12/07/2014] [Accepted: 12/09/2014] [Indexed: 12/18/2022]
Abstract
Survivin, a member of the inhibitor of apoptosisproteins family, is highly expressed in most human neoplasms, but its expression is very low or undetectable in terminally differentiated normal tissues. Survivin has been shown to inhibit cancer cell apoptosis and promote cell proliferation. The overexpression of survivin closely correlates with tumor progression and drug resistance. Because of its key role in tumor formation and maintenance, survivin is considered as an ideal target for anticancer treatment. However, the development of small-molecule survivin inhibitors has been challenging due to the requirement to disrupt the protein-protein interactions. Currently only a limited number of survivin inhibitors have been developed in recent years, and most of these inhibitors reduce survivin levels by interacting with other biomolecules instead of directly interacting with survivin protein. Despite these challenges, developing potent and selective small-molecule survivin inhibitors will be important in both basic science to better understand survivin biology and in translational research to develop potentially more effective, broad-spectrum anticancer agents. In this review, the functions of survivin and its role in cancer are summarized. Recent developments, challenges, and future direction of small-molecule survivin inhibitors are also discussed in detail.
Collapse
Affiliation(s)
| | - Wei Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.
| |
Collapse
|