1
|
Bilski M, Ciesielka M, Orzechowska M, Jarosz B, Całka P, Bilska S, Banach A, Czaja G, Fijuth J, Kuncman Ł. miR-200 family as new potential prognostic factor of overall survival of patients with WHO G2 and WHO G3 brain gliomas. Sci Rep 2024; 14:29345. [PMID: 39592656 PMCID: PMC11599569 DOI: 10.1038/s41598-024-80656-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/21/2024] [Indexed: 11/28/2024] Open
Abstract
Gliomas are the predominant cause of cancer-related deaths among the young population. Even after incorporation of IDH1/2 mutations and 1p19q codeletion there are doubts regarding adjuvant treatment in WHO G2/G3 gliomas. miRNA molecules control about 30% of all genes, also many oncogenes, tumor suppressor genes and genes responsible for the response to ionizing radiation and systemic treatment. Patients with brain gliomas exhibit miRNA disorders. We aimed to evaluate the expression of miR-200 family members in relation to selected clinico-pathological factors and their prognostic value. We enrolled 53 patients diagnosed with WHO G2/G3 brain gliomas treated between 2012-2016. RT-qPCR based expression of miR-200 family was assessed in tumor and surrounding non-cancerous tissue. An analysis of selected clinico-pathological features was carried out. A logistic regression model was prepared for the miRNA signature. The predictive potential of the signature was assessed using the ROC curve. A stepwise backward regression model was used to select variables with a significant predictive potential related to OS. It was shown that miR-200a-3p, miR-200a-5p, miR-200c-5p, miR-141-3p and miR-429 can be independent predictors of survival. Better 2- and 5-year OS was associated with higher expression of miR-200a-3p, miR141-3p and lower expression of miR-200a-5p, miR-200c-5p, miR-429. The strongest predictors of survival were miR-200a-5p, miR-200b-3p, miR-200c-5p, miR-141-3p, miR-429, tumor volume and CTV. Members of the miR-200 family exhibit prognostic value for 2- and 5-year OS. Presented predictive models of survival may be clinically useful for treatment optimization.
Collapse
Affiliation(s)
- Mateusz Bilski
- Department of Radiotherapy, Medical University of Lublin, Lublin, Poland
- Department of Brachytherapy, St. John's Cancer Center, Lublin, Poland
- Department of Radiotherapy, St. John's Cancer Center, Lublin, Poland
| | - Marzanna Ciesielka
- Chair and Department of Forensic Medicine, Medical University of Lublin, Lublin, Poland
| | | | - Bożena Jarosz
- Chair and Department of Neurosurgery and Paediatric Neurosurgery, Medical University of Lublin, Lublin, Poland
| | - Paulina Całka
- Chair and Department of Forensic Medicine, Medical University of Lublin, Lublin, Poland
| | - Sylwia Bilska
- Clinical Genetics Center, St. John's Cancer Center, Lublin, Poland
| | - Agata Banach
- Chair and Department of Neurosurgery and Paediatric Neurosurgery, Medical University of Lublin, Lublin, Poland
| | - Gabriela Czaja
- Chair and Department of Neurosurgery and Paediatric Neurosurgery, Medical University of Lublin, Lublin, Poland
| | - Jacek Fijuth
- Department of Radiotherapy, Medical University of Łódź, Łódź, Poland
- Department of External Beam Radiotherapy, Copernicus Memorial Hospital in Lodz Comprehensive Cancer Center and Traumatology, Pabianicka 62, 93-513, Łódź, PL, Poland
| | - Łukasz Kuncman
- Department of Radiotherapy, Medical University of Łódź, Łódź, Poland.
- Department of External Beam Radiotherapy, Copernicus Memorial Hospital in Lodz Comprehensive Cancer Center and Traumatology, Pabianicka 62, 93-513, Łódź, PL, Poland.
| |
Collapse
|
2
|
Tofolo MV, Berti FCB, Nunes-Souza E, Ruthes MO, Berti LF, Fonseca AS, Rosolen D, Cavalli LR. Non-coding RNAs as modulators of radioresponse in triple-negative breast cancer: a systematic review. J Biomed Sci 2024; 31:93. [PMID: 39354523 PMCID: PMC11445946 DOI: 10.1186/s12929-024-01081-y] [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: 06/13/2024] [Accepted: 08/30/2024] [Indexed: 10/03/2024] Open
Abstract
Triple-negative breast cancer (TNBC), characterized by high invasiveness, is associated with poor prognosis and elevated mortality rates. Despite the development of effective therapeutic targets for TNBC, systemic chemotherapy and radiotherapy (RdT) remain prevalent treatment modalities. One notable challenge of RdT is the acquisition of radioresistance, which poses a significant obstacle in achieving optimal treatment response. Compelling evidence implicates non-coding RNAs (ncRNAs), gene expression regulators, in the development of radioresistance. This systematic review focuses on describing the role, association, and/or involvement of ncRNAs in modulating radioresponse in TNBC. In adhrence to the PRISMA guidelines, an extensive and comprehensive search was conducted across four databases using carefully selected entry terms. Following the evaluation of the studies based on predefined inclusion and exclusion criteria, a refined selection of 37 original research articles published up to October 2023 was obtained. In total, 33 different ncRNAs, including lncRNAs, miRNAs, and circRNAs, were identified to be associated with radiation response impacting diverse molecular mechanisms, primarily the regulation of cell death and DNA damage repair. The findings highlighted in this review demonstrate the critical roles and the intricate network of ncRNAs that significantly modulates TNBC's responsiveness to radiation. The understanding of these underlying mechanisms offers potential for the early identification of non-responders and patients prone to radioresistance during RdT, ultimately improving TNBC survival outcomes.
Collapse
Affiliation(s)
- Maria Vitoria Tofolo
- Research Institute Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim, 1632, Curitiba, 80250-060, Brazil
| | - Fernanda Costa Brandão Berti
- Research Institute Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim, 1632, Curitiba, 80250-060, Brazil
| | - Emanuelle Nunes-Souza
- Research Institute Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim, 1632, Curitiba, 80250-060, Brazil
| | - Mayara Oliveira Ruthes
- Research Institute Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim, 1632, Curitiba, 80250-060, Brazil
| | - Lucas Freitas Berti
- Department of Mechanical Engineering, Postgraduate Program in Mechanical and Materials Engineering, Universidade Tecnológica Federal do Paraná, Curitiba, 81280-340, Brazil
| | - Aline Simoneti Fonseca
- Research Institute Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim, 1632, Curitiba, 80250-060, Brazil
| | - Daiane Rosolen
- Research Institute Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim, 1632, Curitiba, 80250-060, Brazil
| | - Luciane Regina Cavalli
- Research Institute Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim, 1632, Curitiba, 80250-060, Brazil.
- Department of Oncology, Lombardi Comprenhensive Cancer Center, Washington, DC, 20007, USA.
| |
Collapse
|
3
|
Giordano G, Tucciarello C, Merlini A, Cutrupi S, Pignochino Y. Targeting the EphA2 pathway: could it be the way for bone sarcomas? Cell Commun Signal 2024; 22:433. [PMID: 39252029 PMCID: PMC11382444 DOI: 10.1186/s12964-024-01811-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024] Open
Abstract
Bone sarcomas are malignant tumors of mesenchymal origin. Complete surgical resection is the cornerstone of multidisciplinary treatment. However, advanced, unresectable forms remain incurable. A crucial step towards addressing this challenge involves comprehending the molecular mechanisms underpinning tumor progression and metastasis, laying the groundwork for innovative precision medicine-based interventions. We previously showed that tyrosine kinase receptor Ephrin Type-A Receptor 2 (EphA2) is overexpressed in bone sarcomas. EphA2 is a key oncofetal protein implicated in metastasis, self-renewal, and chemoresistance. Molecular, genetic, biochemical, and pharmacological approaches have been developed to target EphA2 and its signaling pathway aiming to interfere with its tumor-promoting effects or as a carrier for drug delivery. This review synthesizes the main functions of EphA2 and their relevance in bone sarcomas, providing strategies devised to leverage this receptor for diagnostic and therapeutic purposes, with a focus on its applicability in the three most common bone sarcoma histotypes: osteosarcoma, chondrosarcoma, and Ewing sarcoma.
Collapse
Affiliation(s)
- Giorgia Giordano
- Sarcoma Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, TO, Italy
- Department of Oncology, University of Turin, 10043, Orbassano, TO, Italy
| | - Cristina Tucciarello
- Sarcoma Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, TO, Italy
- Department of Clinical and Biological Sciences, University of Turin, 10043, Orbassano, TO, Italy
| | - Alessandra Merlini
- Department of Oncology, University of Turin, 10043, Orbassano, TO, Italy
| | - Santina Cutrupi
- Department of Clinical and Biological Sciences, University of Turin, 10043, Orbassano, TO, Italy
| | - Ymera Pignochino
- Sarcoma Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, TO, Italy.
- Department of Clinical and Biological Sciences, University of Turin, 10043, Orbassano, TO, Italy.
| |
Collapse
|
4
|
Alavanda C, Dirimtekin E, Mortoglou M, Arslan Ates E, Guney AI, Uysal-Onganer P. BRCA Mutations and MicroRNA Expression Patterns in the Peripheral Blood of Breast Cancer Patients. ACS OMEGA 2024; 9:17217-17228. [PMID: 38645356 PMCID: PMC11025100 DOI: 10.1021/acsomega.3c10086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 03/08/2024] [Accepted: 03/27/2024] [Indexed: 04/23/2024]
Abstract
Breast cancer (BC) persists as the predominant malignancy globally, standing as the foremost cause of cancer-related mortality among women. Despite notable advancements in prevention and treatment, encompassing the incorporation of targeted immunotherapies, a continued imperative exists for the development of innovative methodologies. These methodologies would facilitate the identification of women at heightened risk, enhance the optimization of therapeutic approaches, and enable the vigilant monitoring of emergent treatment resistance. Circulating microRNAs (miRNAs), found either freely circulating in the bloodstream or encapsulated within extracellular vesicles, have exhibited substantial promise for diverse clinical applications. These applications range from diagnostic and prognostic assessments to predictive purposes. This study aimed to explore the potential associations between BRCA mutations and specific miRNAs (miR-21, miR-155, miR-126, and miR-200c) expression that are known to be dysregulated in BC patient samples. Our findings indicate a robust correlation between miRNA expression status and disease subtypes. We found a correlation between the expression status of miRNAs and distinct disease subtypes. Intriguingly, however, no significant associations were discerned between disease status, subtypes, or miRNA expression levels and the presence of BRCA mutations. To advance the validation of miRNAs as clinically relevant biomarkers, additional investigations within larger and meticulously selected patient cohorts are deemed imperative. These microRNA entities hold the potential to emerge as groundbreaking and readily accessible tools, poised for seamless integration into the landscape of clinical practice.
Collapse
Affiliation(s)
- Ceren Alavanda
- Department
of Medical Genetics, School of Medicine, Marmara University, 34854 Istanbul, Turkey
- Department
of Medical Genetics, Van Research and Training
Hospital, 10300 Van, Turkey
| | - Esra Dirimtekin
- Department
of Medical Genetics, School of Medicine, Marmara University, 34854 Istanbul, Turkey
| | - Maria Mortoglou
- Cancer
Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, W1W 6UW London, U.K.
| | - Esra Arslan Ates
- Department
of Medical Genetics, Istanbul University-Cerrahpasa,
Cerrahpasa Faculty of Medicine, 34098 Istanbul, Turkey
| | - Ahmet Ilter Guney
- Department
of Medical Genetics, School of Medicine, Marmara University, 34854 Istanbul, Turkey
| | - Pinar Uysal-Onganer
- Cancer
Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, W1W 6UW London, U.K.
| |
Collapse
|
5
|
Jawad SF, Altalbawy FMA, Hussein RM, Fadhil AA, Jawad MA, Zabibah RS, Taraki TY, Mohan CD, Rangappa KS. The strict regulation of HIF-1α by non-coding RNAs: new insight towards proliferation, metastasis, and therapeutic resistance strategies. Cancer Metastasis Rev 2024; 43:5-27. [PMID: 37552389 DOI: 10.1007/s10555-023-10129-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 07/21/2023] [Indexed: 08/09/2023]
Abstract
The hypoxic environment is prominently witnessed in most solid tumors and is associated with the promotion of cell proliferation, epithelial-mesenchymal transition (EMT), angiogenesis, metabolic reprogramming, therapeutic resistance, and metastasis of tumor cells. All the effects are mediated by the expression of a transcription factor hypoxia-inducible factor-1α (HIF-1α). HIF-1α transcriptionally modulates the expression of genes responsible for all the aforementioned functions. The stability of HIF-1α is regulated by many proteins and non-coding RNAs (ncRNAs). In this article, we have critically discussed the crucial role of ncRNAs [such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), Piwi-interacting RNAs (piRNAs), and transfer RNA (tRNA)-derived small RNAs (tsRNAs)] in the regulation of stability and expression of HIF-1α. We have comprehensively discussed the molecular mechanisms and relationship of HIF-1α with each type of ncRNA in either promotion or repression of human cancers and therapeutic resistance. We have also elaborated on ncRNAs that are in clinical examination for the treatment of cancers. Overall, the majority of aspects concerning the relationship between HIF-1α and ncRNAs have been discussed in this article.
Collapse
Affiliation(s)
- Sabrean Farhan Jawad
- Department of Pharmacy, Al-Mustaqbal University College, Hilla, Babylon, 51001, Iraq
| | - Farag M A Altalbawy
- National Institute of Laser Enhanced Sciences, University of Cairo, Giza, 12613, Egypt
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
| | | | - Ali Abdulhussain Fadhil
- College of Medical Technology, Medical Lab Techniques, Al-Farahidi University, Baghdad, Iraq
| | - Mohammed Abed Jawad
- Department of Medical Laboratories Technology, Al-Nisour University College, Baghdad, Iraq
| | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | | | - Chakrabhavi Dhananjaya Mohan
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore, 570006, India.
- FEST Division, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226 001, India.
| | | |
Collapse
|
6
|
Jaiswal A, Kaushik N, Choi EH, Kaushik NK. Functional impact of non-coding RNAs in high-grade breast carcinoma: Moving from resistance to clinical applications: A comprehensive review. Biochim Biophys Acta Rev Cancer 2023; 1878:188915. [PMID: 37196783 DOI: 10.1016/j.bbcan.2023.188915] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 04/08/2023] [Accepted: 05/12/2023] [Indexed: 05/19/2023]
Abstract
Despite the recent advances in cancer therapy, triple-negative breast cancers (TNBCs) are the most relapsing cancer sub-type. It is partly due to their propensity to develop resistance against the available therapies. An intricate network of regulatory molecules in cellular mechanisms leads to the development of resistance in tumors. Non-coding RNAs (ncRNAs) have gained widespread attention as critical regulators of cancer hallmarks. Existing research suggests that aberrant expression of ncRNAs modulates the oncogenic or tumor suppressive signaling. This can mitigate the responsiveness of efficacious anti-tumor interventions. This review presents a systematic overview of biogenesis and down streaming molecular mechanism of the subgroups of ncRNAs. Furthermore, it explains ncRNA-based strategies and challenges to target the chemo-, radio-, and immunoresistance in TNBCs from a clinical standpoint.
Collapse
Affiliation(s)
- Apurva Jaiswal
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Republic of Korea
| | - Neha Kaushik
- Department of Biotechnology, College of Engineering, The University of Suwon, Suwon 18323, Republic of Korea.
| | - Eun Ha Choi
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Republic of Korea.
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Republic of Korea.
| |
Collapse
|
7
|
Varzandeh M, Sabouri L, Mansouri V, Gharibshahian M, Beheshtizadeh N, Hamblin MR, Rezaei N. Application of nano-radiosensitizers in combination cancer therapy. Bioeng Transl Med 2023; 8:e10498. [PMID: 37206240 PMCID: PMC10189501 DOI: 10.1002/btm2.10498] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 11/08/2022] [Accepted: 01/27/2023] [Indexed: 02/12/2023] Open
Abstract
Radiosensitizers are compounds or nanostructures, which can improve the efficiency of ionizing radiation to kill cells. Radiosensitization increases the susceptibility of cancer cells to radiation-induced killing, while simultaneously reducing the potentially damaging effect on the cellular structure and function of the surrounding healthy tissues. Therefore, radiosensitizers are therapeutic agents used to boost the effectiveness of radiation treatment. The complexity and heterogeneity of cancer, and the multifactorial nature of its pathophysiology has led to many approaches to treatment. The effectiveness of each approach has been proven to some extent, but no definitive treatment to eradicate cancer has been discovered. The current review discusses a broad range of nano-radiosensitizers, summarizing possible combinations of radiosensitizing NPs with several other types of cancer therapy options, focusing on the benefits and drawbacks, challenges, and future prospects.
Collapse
Affiliation(s)
- Mohammad Varzandeh
- Department of Materials EngineeringIsfahan University of TechnologyIsfahanIran
| | - Leila Sabouri
- AmitisGen TECH Dev GroupTehranIran
- Regenerative Medicine Group (REMED)Universal Scientific Education and Research Network (USERN)TehranIran
| | - Vahid Mansouri
- Regenerative Medicine Group (REMED)Universal Scientific Education and Research Network (USERN)TehranIran
- Gene Therapy Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical SciencesTehranIran
| | - Maliheh Gharibshahian
- Regenerative Medicine Group (REMED)Universal Scientific Education and Research Network (USERN)TehranIran
- Student Research CommitteeSchool of Medicine, Shahroud University of Medical SciencesShahroudIran
| | - Nima Beheshtizadeh
- Regenerative Medicine Group (REMED)Universal Scientific Education and Research Network (USERN)TehranIran
- Department of Tissue EngineeringSchool of Advanced Technologies in Medicine, Tehran University of Medical SciencesTehranIran
| | - Michael R. Hamblin
- Laser Research Center, Faculty of Health ScienceUniversity of JohannesburgDoornfonteinSouth Africa
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA)Universal Scientific Education and Research Network (USERN)TehranIran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA)Universal Scientific Education and Research Network (USERN)TehranIran
- Research Center for ImmunodeficienciesChildren's Medical Center, Tehran University of Medical SciencesTehranIran
- Department of ImmunologySchool of Medicine, Tehran University of Medical SciencesTehranIran
| |
Collapse
|
8
|
Darvish L, Bahreyni Toossi MT, Azimian H, Shakeri M, Dolat E, Ahmadizad Firouzjaei A, Rezaie S, Amraee A, Aghaee-Bakhtiari SH. The role of microRNA-induced apoptosis in diverse radioresistant cancers. Cell Signal 2023; 104:110580. [PMID: 36581218 DOI: 10.1016/j.cellsig.2022.110580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/07/2022] [Accepted: 12/21/2022] [Indexed: 12/27/2022]
Abstract
Resistance to cancer radiotherapy is one of the biggest concerns for success in treating and preventing recurrent disease. Malignant tumors may develop when they block genetic mutations associated with apoptosis or abnormal expression of apoptosis; Tumor treatment may induce the expression of apoptosis-related genes to promote tumor cell apoptosis. MicroRNAs have been shown to contribute to forecasting prognosis, distinguishing between cancer subtypes, and affecting treatment outcomes in cancer. Constraining these miRNAs may be an attractive treatment strategy to help overcome radiation resistance. The delivery of these future treatments is still challenging due to the excess downstream targets that each miRNA can control. Understanding the role of miRNAs brings us one step closer to attaining patient treatment and improving patient outcomes. This review summarized the current information on the role of microRNA-induced apoptosis in determining the radiosensitivity of various cancers.
Collapse
Affiliation(s)
- Leili Darvish
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hosein Azimian
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Physics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahsa Shakeri
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Dolat
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Ahmadizad Firouzjaei
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samaneh Rezaie
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Azadeh Amraee
- Department of Medical Physics, Faculty of Medicine, School of Medicine, Lorestan University of Medical Sciences, khorramabad, Iran
| | - Seyed Hamid Aghaee-Bakhtiari
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Bioinformatics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Anlotinib Inhibits Tumor Angiogenesis and Promotes the Anticancer Effect of Radiotherapy on Esophageal Cancer through Inhibiting EphA2. JOURNAL OF ONCOLOGY 2022; 2022:5632744. [PMID: 36090890 PMCID: PMC9452983 DOI: 10.1155/2022/5632744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/10/2022] [Accepted: 08/17/2022] [Indexed: 11/18/2022]
Abstract
Background Anlotinib is a novel multitarget tyrosine kinase inhibitor for tumor angiogenesis and has antitumor activity in a variety of solid tumors. Given that, our study was designed to unearth the mechanism of anlotinib in radioresistant esophageal cancer (EC) cells. Methods Radioresistant EC cell lines TE-1R and KYSE-150R were established by multiple fractionated irradiation. Detection of cell proliferation was governed by the MTT assay, angiogenesis by the tube formation assay, and cell migration and invasion by the transwell assay. Lastly, RT-qPCR Western blotting was employed to detect the expression of related genes. Cancerous cells showing tumor growth were then detected by tumor xenografts in mice. Results Radioresistant EC cell lines TE-1R and KYSE-150R were successfully established. Anlotinib downregulated EphA2 inhibited proliferation, angiogenesis, migration, and invasion of radioresistant EC cells in vitro. The up-regulated expression of EphA2 in both EC cell lines and radioresistant EC cells, along with anlotinib, in turn, inhibited the expression of EphA2 in radioresistant EC cells. Inhibiting EphA2 also enhanced anlotinib-mediated effects on radioresistant EC cells, so as to restrain cell proliferation, angiogenesis, migration, and invasion. Correspondingly, overexpression of EphA2 is capable of reversing the therapeutic effect of anlotinib on radioresistant EC cells. Also, anlotinib enhances the inhibitory effect of irradiation on mice. Conclusion It is concluded that anlotinib inhibits EphA2 expression, thereby suppressing angiogenesis and resensitizing EC cells to radiotherapy, providing another perspective to overcome radioresistance in EC.
Collapse
|
10
|
Radiation therapy for triple-negative breast cancer: emerging role of microRNAs as biomarkers and radiosensitivity modifiers. A systematic review. Breast Cancer Res Treat 2022; 193:265-279. [PMID: 35397079 DOI: 10.1007/s10549-022-06533-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 01/19/2022] [Indexed: 11/02/2022]
Abstract
PURPOSE Radiation therapy (RT) for triple-negative breast cancer (TNBC) treatment is currently delivered in the adjuvant setting and is under investigation as a booster of neoadjuvant treatments. However, TNBC radioresistance remains an obstacle, so new biomarkers are needed to select patients for any integration of RT in the TNBC therapy sequence. MicroRNAs (miRs) are important regulators of gene expression, involved in cancer response to ionizing radiation (IR) and assessable by tumor tissue or liquid biopsy. This systematic review aimed to evaluate the relationships between miRs and response to radiation in TNBC, as well as their potential predictive and prognostic values. METHODS A thorough review of studies related to miRs and RT in TNBC was performed on PubMed, EMBASE, and Web of Science. We searched for original English articles that involved dysregulation of miRs in response to IR on TNBC-related preclinical and clinical studies. After a rigorous selection, 44 studies were chosen for further analysis. RESULTS Thirty-five miRs were identified to be TNBC related, out of which 21 were downregulated, 13 upregulated, and 2 had a double-side expression in this cancer. Expression modulation of many of these miRs is radiosensitizing, among which miR-7, -27a, -34a, -122, and let-7 are most studied, still only in experimental models. The miRs reported as most influencing/reflecting TNBC response to IR are miR-7, -27a, -155, -205, -211, and -221, whereas miR-21, -33a, -139-5p, and -210 are associated with TNBC patient outcome after RT. CONCLUSION miRs are emerging biomarkers and radiosensitizers in TNBC, worth further investigation. Dynamic assessment of circulating miRs could improve monitoring and TNBC RT efficacy, which are of particular interest in the neoadjuvant and the high-risk patients' settings.
Collapse
|
11
|
Masoudi-Khoram N, Abdolmaleki P. Role of non-coding RNAs in response of breast cancer to radiation therapy. Mol Biol Rep 2022; 49:5199-5208. [PMID: 35217966 DOI: 10.1007/s11033-022-07234-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 02/04/2022] [Indexed: 12/13/2022]
Abstract
Breast cancer ranks as the first common cancer with a high incidence rate and mortality among women. Radiation therapy is the main therapeutic method for breast cancer patients. However, radiation resistance of tumor cells can reduce the efficacy of treatment and lead to recurrence and mortality in patients. Non-coding RNA (ncRNAs) refers to a group of small RNA molecules that are not translated into protein, while they have the ability to modulate the translation of target mRNA. Several studies have reported the altered expression of ncRNAs in response to radiation in breast cancer. NcRNAs have been found to influence on radiation response of breast cancer by regulating various mechanisms, including DNA damage response, cell cycle regulation, cell death, inflammatory response, cancer stem cell and EGFR related pathways. This paper aimed to provide a summary of current findings on ncRNAs dysregulation after irradiation. We also present the function and mechanism of ncRNAs in modulating radiosensitivity or radioresistance of breast cancer cells.
Collapse
Affiliation(s)
- Nastaran Masoudi-Khoram
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box 1415-154, Tehran, Iran
| | - Parviz Abdolmaleki
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box 1415-154, Tehran, Iran.
| |
Collapse
|
12
|
Aranza-Martínez A, Sánchez-Pérez J, Brito-Elias L, López-Camarillo C, Cantú de León D, Pérez-Plasencia C, López-Urrutia E. Non-Coding RNAs Associated With Radioresistance in Triple-Negative Breast Cancer. Front Oncol 2021; 11:752270. [PMID: 34804940 PMCID: PMC8599982 DOI: 10.3389/fonc.2021.752270] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/06/2021] [Indexed: 12/12/2022] Open
Abstract
The resistance that Triple-Negative Breast Cancer (TNBC), the most aggressive breast cancer subtype, develops against radiotherapy is a complex phenomenon involving several regulators of cell metabolism and gene expression; understanding it is the only way to overcome it. We focused this review on the contribution of the two leading classes of regulatory non-coding RNAs, microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), against ionizing radiation-based therapies. We found that these regulatory RNAs are mainly associated with DNA damage response, cell death, and cell cycle regulation, although they regulate other processes like cell signaling and metabolism. Several regulatory RNAs regulate multiple pathways simultaneously, such as miR-139-5p, the miR-15 family, and the lncRNA HOTAIR. On the other hand, proteins such as CHK1 and WEE1 are targeted by several regulatory RNAs simultaneously. Interestingly, the study of miRNA/lncRNA/mRNA regulation axes increases, opening new avenues for understanding radioresistance. Many of the miRNAs and lncRNAs that we reviewed here can be used as molecular markers or targeted by upcoming therapeutic options, undoubtedly contributing to a better prognosis for TNBC patients.
Collapse
Affiliation(s)
- Alberto Aranza-Martínez
- Laboratorio de Genómica Funcional, Facultad de Estudios Superiores Iztacala Universidad Nacional Autónoma de México (UNAM), Tlalnepantla, Mexico
| | - Julio Sánchez-Pérez
- Laboratorio de Genómica Funcional, Facultad de Estudios Superiores Iztacala Universidad Nacional Autónoma de México (UNAM), Tlalnepantla, Mexico
| | - Luis Brito-Elias
- Laboratorio de Genómica Funcional, Facultad de Estudios Superiores Iztacala Universidad Nacional Autónoma de México (UNAM), Tlalnepantla, Mexico
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Mexico City, Mexico
| | - David Cantú de León
- Dirección de Investigación, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Carlos Pérez-Plasencia
- Laboratorio de Genómica Funcional, Facultad de Estudios Superiores Iztacala Universidad Nacional Autónoma de México (UNAM), Tlalnepantla, Mexico.,Laboratorio de Genómica, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Eduardo López-Urrutia
- Laboratorio de Genómica Funcional, Facultad de Estudios Superiores Iztacala Universidad Nacional Autónoma de México (UNAM), Tlalnepantla, Mexico
| |
Collapse
|
13
|
Simion V, Loussouarn C, Laurent Y, Roncali L, Gosset D, Reverchon F, Rousseau A, Martin F, Midoux P, Pichon C, Garcion E, Baril P. LentiRILES, a miRNA-ON sensor system for monitoring the functionality of miRNA in cancer biology and therapy. RNA Biol 2021; 18:198-214. [PMID: 34570661 DOI: 10.1080/15476286.2021.1978202] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
A major unresolved challenge in miRNA biology is the capacity to monitor the spatiotemporal activity of miRNAs expressed in animal disease models. We recently reported that the miRNA-ON monitoring system called RILES (RNAi-inducible expression Luciferase system) implanted in lentivirus expression system (LentiRILES) offers unique opportunity to decipher the kinetics of miRNA activity in vitro, in relation with their intracellular trafficking in glioblastoma cells. In this study, we describe in detail the method for the production of LentiRILES stable cell lines and employed it in several applications in the field of miRNA biology and therapy. We show that LentiRILES is a robust, highly specific and sensitive miRNA sensor system that can be used in vitro as a single-cell miRNA monitoring method, cell-based screening platform for miRNA therapeutics and as a tool to analyse the structure-function relationship of the miRNA duplex. Furthermore, we report the kinetics of miRNA activity upon the intracranial delivery of miRNA mimics in an orthotopic animal model of glioblastoma. This information is exploited to evaluate the tumour suppressive function of miRNA-200c as locoregional therapeutic modality to treat glioblastoma. Our data provide evidence that LentiRILES is a robust system, well suited to resolve the activity of endogenous and exogenously expressed miRNAs from basic research to gene and cell therapy.
Collapse
Affiliation(s)
- Viorel Simion
- Centre De Biophysique Moléculaire, CNRS UPR4301, Université d'Orléans, Orléans, France
| | - Claire Loussouarn
- Université d'Angers, Université de Nantes, Inserm, CRCINA, Angers, France
| | - Yoan Laurent
- Centre De Biophysique Moléculaire, CNRS UPR4301, Université d'Orléans, Orléans, France
| | - Loris Roncali
- Université d'Angers, Université de Nantes, Inserm, CRCINA, Angers, France
| | - David Gosset
- Centre De Biophysique Moléculaire, CNRS UPR4301, Université d'Orléans, Orléans, France
| | - Flora Reverchon
- Centre De Biophysique Moléculaire, CNRS UPR4301, Université d'Orléans, Orléans, France
| | - Audrey Rousseau
- Université d'Angers, Université de Nantes, Inserm, CRCINA, Angers, France
| | - Francisco Martin
- GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, Spain
| | - Patrick Midoux
- Centre De Biophysique Moléculaire, CNRS UPR4301, Université d'Orléans, Orléans, France
| | - Chantal Pichon
- Centre De Biophysique Moléculaire, CNRS UPR4301, Université d'Orléans, Orléans, France
| | - Emmanuel Garcion
- Université d'Angers, Université de Nantes, Inserm, CRCINA, Angers, France
| | - Patrick Baril
- Centre De Biophysique Moléculaire, CNRS UPR4301, Université d'Orléans, Orléans, France
| |
Collapse
|
14
|
Sai S, Kim EH, Koom WS, Vares G, Suzuki M, Yamada S, Hayashi M. Carbon-Ion Beam Irradiation and the miR-200c Mimic Effectively Eradicate Pancreatic Cancer Stem Cells Under in vitro and in vivo Conditions. Onco Targets Ther 2021; 14:4749-4760. [PMID: 34556996 PMCID: PMC8453446 DOI: 10.2147/ott.s311567] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose The study investigated the molecular mechanisms that killed pancreatic cancer cells, including cancer stem cells (CSCs), by carbon ion beam irradiation alone or in combination with miRNA-200c under in vitro and in vivo conditions. Methods Human pancreatic cancer (PC) cells, PANC1 and PK45, were treated with carbon-ion beam irradiation alone or in combination with microRNA-200c (miR-200c) mimic. Cell viability assay, colony and spheroid formation assay, quantitative real-time PCR analysis of apoptosis-, autophagy-, and angiogenesis-related gene expression, xenograft tumor control and histopathological analyses were performed. Results The cell viability assay showed that transfection of the miRNA-200c (10 nM) mimic into pancreatic CSC (CD44+/ESA+) and non-CSC (CD44-/ESA-) significantly suppressed proliferation of both types of cell populations described above. Combining carbon-ion beam irradiation with the miRNA-200c mimic significantly reduced the colony as well as spheroid formation abilities compared to that observed with the treatment of carbon-ion beam alone or X-ray irradiation combined with the miRNA-200c mimic. Moreover, the combination of carbon ion beam irradiation and miRNA-200c mimic increased the expression of apoptosis-related gene BAX, autophagy-related genes Beclin-1 and p62, addition of gemcitabine (GEM) further enhanced the expression of these genes. In vivo data showed that carbon-ion beam irradiation in combination with the miRNA-200c mimic effectively suppressed xenograft tumor growth and significantly induced tumor necrosis and cavitation. Conclusion The combination of miRNA-200c mimic and carbon ion beam irradiation may be powerful radiotherapy that significantly kills pancreatic cancer cells containing CSCs and enhances the effect of carbon-ion beam irradiation compared to carbon-ion beam irradiation alone.
Collapse
Affiliation(s)
- Sei Sai
- Department of Charged Particle Therapy Research, Institute for Quantum Medical Science, National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Eun Ho Kim
- Department of Biochemistry, School of Medicine, Daegu Catholic University, Nam-gu, Daegu, 42472, South Korea
| | - Woong Sub Koom
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Guillaume Vares
- Institute of Radioprotection and Nuclear Safety (IRSN), Fontenay-aux-Roses Cedex, France
| | - Masao Suzuki
- Department of Charged Particle Therapy Research, Institute for Quantum Medical Science, National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Shigeru Yamada
- QST Hospital, National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Mitsuhiro Hayashi
- Breast Center, Dokkyo Medical University Hospital, Tochigi, 321-0293, Japan
| |
Collapse
|
15
|
The emerging role of miR-200 family in metastasis: focus on EMT, CSCs, angiogenesis, and anoikis. Mol Biol Rep 2021; 48:6935-6947. [PMID: 34510322 DOI: 10.1007/s11033-021-06666-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Cancer is the second major threat to human society and one of the main challenges facing healthcare systems. One of the main problems of cancer care is the metastases of cancer cells that cause 90% of deaths due to cancer. Multiple molecular mechanisms are involved in cancer cell metastasis. Therefore, a better understanding of these molecular mechanisms is necessary for designing restrictive strategies against cancer cell metastasis. Accumulating data suggests that MicroRNAs (miRNAs) are involved in metastasis and invasion of human tumors through regulating multiple genes expression levels that are involved in molecular mechanisms of metastasis. The goal of this review is to present the molecular pathways by which the miR 200 family manifests its effects on EMT, cancer stem cells, angiogenesis, anoikis, and the effects of tumor cell metastases. METHODS A detailed literature search was conducted to find information about the role of the miR-200 family in the processes involved in metastasis in various databases. RESULTS Numerous lines of evidence revealed an association between the mir-200 family and metastasis of human tumors by impressing processes such as cancer stem cells, EMT, angiogenesis, and anoikis. CONCLUSIONS Understanding the molecular mechanisms associated with metastasis in which the miR-200 family is involved can be effective in treating metastatic cancers.
Collapse
|
16
|
Salazar DA, Pržulj N, Valencia CF. Multi-project and Multi-profile joint Non-negative Matrix Factorization for cancer omic datasets. Bioinformatics 2021; 37:4801-4809. [PMID: 34375392 DOI: 10.1093/bioinformatics/btab579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 07/31/2021] [Accepted: 08/06/2021] [Indexed: 11/12/2022] Open
Abstract
MOTIVATION The integration of multi-omic data using machine learning methods has been focused on solving relevant tasks such as predicting sensitivity to a drug or subtyping patients. Recent integration methods, such as joint Non-negative Matrix Factorization (jNMF), have allowed researchers to exploit the information in the data to unravel the biological processes of multi-omic datasets. RESULTS We present a novel method called Multi-project and Multi-profile joint Non-negative Matrix Factorization (M&M-jNMF) capable of integrating data from different sources, such as experimental and observational multi-omic data. The method can generate co-clusters between observations, predict profiles and relate latent variables. We applied the method to integrate low-grade glioma omic profiles from The Cancer Genome Atlas (TCGA) and Cell Line Encyclopedia (CCLE) projects. The method allowed us to find gene clusters mainly enriched in cancer-associated terms. We identified groups of patients and cell lines similar to each other by comparing biological processes. We predicted the drug profile for patients, and we identified genetic signatures for resistant and sensitive tumors to a specific drug. AVAILABILITY AND IMPLEMENTATION Source code repository is publicly available at https://bitbucket.org/dsalazarb/mmjnmf/ - Zenodo DOI: 10.5281/zenodo.5150920. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- D A Salazar
- Industrial Engineering Department, University of los Andes, Bogota, 111711, Colombia.,Center for optimization and applied probability, University of los Andes, Bogota, 111711, Colombia
| | - N Pržulj
- Barcelona Supercomputing Center (BSC), Barcelona, 08034, Spain.,Department of Computer Science, University College London, London, WC1E 6BT, UK.,ICREA, Pg. Lluis Companys 23, Barcelona, 08010, Spain
| | - C F Valencia
- Industrial Engineering Department, University of los Andes, Bogota, 111711, Colombia.,Center for optimization and applied probability, University of los Andes, Bogota, 111711, Colombia
| |
Collapse
|
17
|
Qi R, Zhao Y, Guo Q, Mi X, Cheng M, Hou W, Zheng H, Hua B. Exosomes in the lung cancer microenvironment: biological functions and potential use as clinical biomarkers. Cancer Cell Int 2021; 21:333. [PMID: 34193120 PMCID: PMC8247080 DOI: 10.1186/s12935-021-01990-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 05/22/2021] [Indexed: 12/14/2022] Open
Abstract
Lung cancer is one of the most common malignant tumours worldwide. however, emerging immunotherapy and targeted therapies continue to show limited efficacy. In the search for new targets for lung cancer treatment, exosomes have become a major focus of research. Exosomes play an important role in the tumour microenvironment (TME) of lung cancer and affect invasion, metastasis, and treatment responses. This review describes our current understanding of the release of exosomes derived from different cells in the TME, the effects of exosomes on T/Tregs, myeloid-derived suppressor cells, tumour-associated macrophages, dendritic cells, and natural killer cells, and the role of exosomes in the endothelial–mesenchymal transition, angiogenesis, and cancer-associated fibroblasts. In particular, this review focuses on the potential clinical applications of exosomes in the lung cancer microenvironment and their prognostic and diagnostic value.
Collapse
Affiliation(s)
- Runzhi Qi
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Xicheng District, Beijing, China
| | - Yuwei Zhao
- Beijing University of Chinese Medicine, Chaoyang District, Beijing, China
| | - Qiujun Guo
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Xicheng District, Beijing, China
| | - Xue Mi
- Shaanxi University of Chinese Medicine, Qindu District, Xianyang, Shaanxi, China
| | - Mengqi Cheng
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Xicheng District, Beijing, China
| | - Wei Hou
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Xicheng District, Beijing, China
| | - Honggang Zheng
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Xicheng District, Beijing, China.
| | - Baojin Hua
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Xicheng District, Beijing, China.
| |
Collapse
|
18
|
Bai JC, Huang GY. miR-1825 Accelerates Cell Proliferation and Inhibits Cell Apoptosis of Prostate Cancer via Targeting Suppressor of Cancer Cell Invasion. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Prostate cancer (PC) is one major carcinoma threat to the health of males. microRNAs (miRNAs) are short non-coding transcripts with about 23 nt in length. Booming evidence has verified the various roles of miRNAs in human tumors. miR-1825 was once demonstrated to be highly expressed
in PC, but the potential role of miR-1825 in PC has never been clarified yet. This work aimed to explore the function of miR-1825 and reveal the underlying modulation mechanism in PC. First, miR-1825 was detected to be elevated in PC cells compared with normal prostate cells, as proved by
RT-qPCR. After miR-1825 expression was inhibited, cell proliferation was hindered and cell apoptosis was promoted, which was observed by CCK8, colony formation, TUNEL staining and western blot assays. Bioinformatics tools discovered the targeting of suppressor of cancer cell invasion (SCAI)
by miR-1825, which was further confirmed by luciferase reporter assay. Then the suppression of miR-1825 on SCAI protein expression was verified by western blotting. Eventually, rescue assays were implemented and affirmed the miR-1825/SCAI axis in PC cells. In conclusion, our present research
disclosed the oncogenic role of miR-1825 and the miR-1825/SCAI pathway in PC. These findings gave new clues for the therapy of PC.
Collapse
Affiliation(s)
- Jun-Chao Bai
- Department of Urology, The Fourth Affiliated Hospital Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Guang-Yi Huang
- Department of Urology, The Fourth Affiliated Hospital Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| |
Collapse
|
19
|
MiRNA-200C expression in Fanconi anemia pathway functionally deficient lung cancers. Sci Rep 2021; 11:4420. [PMID: 33627769 PMCID: PMC7904768 DOI: 10.1038/s41598-021-83884-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 02/09/2021] [Indexed: 12/16/2022] Open
Abstract
The Fanconi Anemia (FA) pathway is essential for human cells to maintain genomic integrity following DNA damage. This pathway is involved in repairing damaged DNA through homologous recombination. Cancers with a defective FA pathway are expected to be more sensitive to cross-link based therapy or PARP inhibitors. To evaluate downstream effectors of the FA pathway, we studied the expression of 734 different micro RNAs (miRNA) using NanoString nCounter miRNA array in two FA defective lung cancer cells and matched control cells, along with two lung tumors and matched non-tumor tissue samples that were deficient in the FA pathway. Selected miRNA expression was validated with real-time PCR analysis. Among 734 different miRNAs, a cluster of microRNAs were found to be up-regulated including an important cancer related micro RNA, miR-200C. MiRNA-200C has been reported as a negative regulator of epithelial-mesenchymal transition (EMT) and inhibits cell migration and invasion by promoting the upregulation of E-cadherin through targeting ZEB1 and ZEB2 transcription factors. miRNA-200C was increased in the FA defective lung cancers as compared to controls. AmpliSeq analysis showed significant reduction in ZEB1 and ZEB2 mRNA expression. Our findings indicate the miRNA-200C potentially play a very important role in FA pathway downstream regulation.
Collapse
|
20
|
Mansoori B, Silvestris N, Mohammadi A, Khaze V, Baghbani E, Mokhtarzadeh A, Shanehbandi D, Derakhshani A, Duijf PHG, Baradaran B. miR-34a and miR-200c Have an Additive Tumor-Suppressive Effect on Breast Cancer Cells and Patient Prognosis. Genes (Basel) 2021; 12:267. [PMID: 33673143 PMCID: PMC7918749 DOI: 10.3390/genes12020267] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 01/28/2021] [Accepted: 02/02/2021] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is the most common women's malignancy in the world and, for subgroups of patients, treatment outcomes remain poor. Thus, more effective therapeutic strategies are urgently needed. MicroRNAs (miRNAs) have emerged as promising therapeutic tools and targets, as they play significant roles in regulating key cellular processes by suppressing gene expression. However, additive opportunities involving miRNAs have been underexplored. For example, both miR-34a and miR-200c individually suppress the development of different types of cancer, but the cellular effects of their combined actions remain unknown. Here, we show that miR-34a and miR-200c levels are reduced in breast tumors compared to adjacent normal tissues and that this additively predicts poor patient survival. In addition, in cell lines, miR-34a and miR-200c additively induce apoptosis and cell cycle arrest, while also inhibiting proliferation, invasion, migration, stemness and epithelial-to-mesenchymal transition (EMT). Mechanistically, both miRNA-34a and miR-200c directly target HIF1-α and subsequently downregulate VEGFR, MMP9 and CXCR4, although combined miRNA-34a and miR-200c delivery suppresses mouse xenograft tumor development as effectively as individual delivery. We establish a model, supported by in vitro and clinical data, which collectively suggest that the co-delivery of miR-34a and miR-200c represents a promising novel therapeutic strategy for breast cancer patients.
Collapse
Affiliation(s)
- Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran; (B.M.); (V.K.); (E.B.); (A.M.); (D.S.); (A.D.)
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, 5000C Odense, Denmark;
| | - Nicola Silvestris
- Medical Oncology Unit-IRCCS IstitutoTumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
- Department of Biomedical Sciences and Human Oncology, DIMO-University of Bari, 70124 Bari, Italy
| | - Ali Mohammadi
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, 5000C Odense, Denmark;
| | - Vahid Khaze
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran; (B.M.); (V.K.); (E.B.); (A.M.); (D.S.); (A.D.)
| | - Elham Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran; (B.M.); (V.K.); (E.B.); (A.M.); (D.S.); (A.D.)
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran; (B.M.); (V.K.); (E.B.); (A.M.); (D.S.); (A.D.)
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran; (B.M.); (V.K.); (E.B.); (A.M.); (D.S.); (A.D.)
| | - Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran; (B.M.); (V.K.); (E.B.); (A.M.); (D.S.); (A.D.)
| | - Pascal H. G. Duijf
- Faculty of Health, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, 37 Kent Street, Brisbane, QLD 4102, Australia
- University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, 37 Kent Street, Brisbane, QLD 4102, Australia
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran; (B.M.); (V.K.); (E.B.); (A.M.); (D.S.); (A.D.)
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran
| |
Collapse
|
21
|
Pesch AM, Pierce LJ, Speers CW. Modulating the Radiation Response for Improved Outcomes in Breast Cancer. JCO Precis Oncol 2021; 5:PO.20.00297. [PMID: 34250414 DOI: 10.1200/po.20.00297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/12/2020] [Accepted: 12/22/2020] [Indexed: 12/25/2022] Open
Affiliation(s)
- Andrea M Pesch
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI.,Department of Pharmacology, University of Michigan, Ann Arbor, MI.,Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| | - Lori J Pierce
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI.,Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| | - Corey W Speers
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI.,Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| |
Collapse
|
22
|
Cardoso AM, Morais CM, Rebelo O, Tão H, Barbosa M, Pedroso de Lima MC, Jurado AS. Downregulation of long non-protein coding RNA MVIH impairs glioblastoma cell proliferation and invasion through an miR-302a-dependent mechanism. Hum Mol Genet 2021; 30:46-64. [PMID: 33438023 DOI: 10.1093/hmg/ddab009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/18/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma (GB) is the most frequent and malignant type of brain tumor, for which no effective therapy exists. The high proliferative and invasive nature of GB, as well as its acquired resistance to chemotherapy, makes this type of cancer extremely lethal shortly after diagnosis. Long non-protein coding RNAs (lncRNA) are a class of regulatory RNAs whose levels can be dysregulated in the context of diseases, unbalancing several physiological processes. The lncRNA associated with microvascular invasion in hepatocellular carcinoma (lncRNA-MVIH), overexpressed in several cancers, was described to co-precipitate with phosphoglycerate kinase 1 (PGK1), preventing secretion of this enzyme to the extracellular environment and promoting cell migration and invasion. We hypothesized that, by silencing the expression of lncRNA-MVIH, the secretion of PGK1 would increase, reducing GB cell migration and invasion capabilities. We observed that lncRNA-MVIH silencing in human GB cells significantly decreased glycolysis, cell growth, migration, and invasion and sensitized GB cells to cediranib. However, no increase in extracellular PGK1 was observed as a consequence of lncRNA-MVIH silencing, and therefore, we investigated the possibility of a mechanism of miRNA sponge of lncRNA-MVIH being in place. We found that the levels of miR-302a loaded onto RISC increased in GB cells after lncRNA-MVIH silencing, with the consequent downregulation of several miR-302a molecular targets. Our findings suggest a new mechanism of action of lncRNA-MVIH as a sponge of miR-302a. We suggest that lncRNA-MVIH knockdown may be a promising strategy to address GB invasiveness and chemoresistance, holding potential towards its future application in a clinical context.
Collapse
Affiliation(s)
- Ana M Cardoso
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, IIIUC - Institute for Interdisciplinary Research, Coimbra, Portugal
| | - Catarina M Morais
- Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
| | - Olinda Rebelo
- Neuropathology Laboratory, Neurology Service, University Hospital of Coimbra, 3004-561 Coimbra, Portugal
| | - Hermínio Tão
- Neurosurgery Service, University Hospital of Coimbra, 33004-561 Coimbra, Portugal
| | - Marcos Barbosa
- Neurosurgery Service, University Hospital of Coimbra, 33004-561 Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Maria C Pedroso de Lima
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, IIIUC - Institute for Interdisciplinary Research, Coimbra, Portugal
| | - Amália S Jurado
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, IIIUC - Institute for Interdisciplinary Research, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
| |
Collapse
|
23
|
Potential Diagnostic and Prognostic Utility of miR-141, miR-181b1, and miR-23b in Breast Cancer. Int J Mol Sci 2020; 21:ijms21228589. [PMID: 33202602 PMCID: PMC7697480 DOI: 10.3390/ijms21228589] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/05/2020] [Accepted: 11/12/2020] [Indexed: 12/14/2022] Open
Abstract
miRNAs, a group of short noncoding RNAs, are key regulators of fundamental cellular processes and signaling pathways. Dysregulation of miRNA expression with known oncogenic or tumor suppressor functions has been associated with neoplastic transformation. Numerous studies have reported dysregulation of miRNA-141, miR-181b1, and miR-23b in a wide range of malignancies, including breast cancer. To the best of our knowledge, no previous study had demonstrated the expression of miR-141-3p, miR-181b1-5p, and miR-23b-3p in different histological grades and molecular subtypes of breast cancer. Here, we identified differential expression of these three miRNAs in breast cancer tissues compared with benign breast fibroadenomas. In addition, high expression levels of miR-141-3p and miR-181b1-5p are strongly associated with aggressive breast carcinomas. We also confirmed the clinical potential of using the three miRNAs individually or combined as diagnostic and prognostic markers in breast cancer. Using bioinformatics analyses, we identified 23 hub genes of these three miRNAs which are involved in key signaling pathways in breast cancer. Furthermore, the KM plotter online database analysis demonstrates the association between elevated expression of miR-141 and miR-181b and shorter overall survival of breast cancer patients. Together, our data suggest an oncogenic role of the studied miRNAs and highlight their molecular roles and potential clinical applications in breast cancer.
Collapse
|
24
|
Rezaei S, Mahjoubin-Tehran M, Aghaee-Bakhtiari SH, Jalili A, Movahedpour A, Khan H, Moghoofei M, Shojaei Z, R Hamblin M, Mirzaei H. Autophagy-related MicroRNAs in chronic lung diseases and lung cancer. Crit Rev Oncol Hematol 2020; 153:103063. [DOI: 10.1016/j.critrevonc.2020.103063] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 06/11/2020] [Accepted: 07/12/2020] [Indexed: 12/24/2022] Open
|
25
|
Masoudi-Khoram N, Abdolmaleki P, Hosseinkhan N, Nikoofar A, Mowla SJ, Monfared H, Baldassarre G. Differential miRNAs expression pattern of irradiated breast cancer cell lines is correlated with radiation sensitivity. Sci Rep 2020; 10:9054. [PMID: 32493932 PMCID: PMC7270150 DOI: 10.1038/s41598-020-65680-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/07/2020] [Indexed: 02/08/2023] Open
Abstract
Radiotherapy is a fundamental step in the treatment of breast cancer patients. The treatment efficiency is however reduced by the possible onset of radiation resistance. In order to develop the effective treatment approach, it is important to understand molecular basis of radiosensitivity in breast cancer. The purpose of the present study was to investigate different radiation response of breast cancer cell lines, and find out if this response may be related to change in the microRNAs expression profile. MDA-MB-231 and T47D cells were subjected to different doses of radiation, then MTT and clonogenic assays were performed to assess radiation sensitivity. Cytofluorometric and western blot analysis were performed to gain insight into cell cycle distribution and protein expression. MicroRNA sequencing and bioinformatics prediction methods were used to identify the difference in microRNAs expression between two breast cancer cells and the related genes and pathways. T47D cells were more sensitive to radiation respect to MDA-MB-231 cells as demonstrated by a remarkable G2 cell cycle arrest followed by a greater reduction in cell viability and colony forming ability. Accordingly, T47D cells showed higher increase in the phosphorylation of ATM, TP53 and CDK1 (markers of radiation response) and faster and more pronounced increase in RAD51 and γH2AX expression (markers of DNA damage), when compared to MDA-MB-231 cells. The two cell lines had different microRNAs expression profiles with a confirmed significant differential expression of miR-16-5p, which targets cell cycle related genes and predicts longer overall survival of breast cancer patients, as determined by bioinformatics analysis. These results suggest a possible role for miR-16-5p as radiation sensitizing microRNA and as prognostic/predictive biomarker in breast cancer.
Collapse
Affiliation(s)
- Nastaran Masoudi-Khoram
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Parviz Abdolmaleki
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Nazanin Hosseinkhan
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Alireza Nikoofar
- Department of Radiotherapy, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Seyed Javad Mowla
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hamideh Monfared
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Gustavo Baldassarre
- Division of Experimental Oncology 2, Department of Translational Research, CRO, National Cancer Institute, Aviano, Italy
| |
Collapse
|
26
|
Zheng JJ, He Y, Liu Y, Li FS, Cui Z, Du XM, Wang CP, Wu YM. Novel role of PAF1 in attenuating radiosensitivity in cervical cancer by inhibiting IER5 transcription. Radiat Oncol 2020; 15:131. [PMID: 32471508 PMCID: PMC7257241 DOI: 10.1186/s13014-020-01580-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 05/20/2020] [Indexed: 01/16/2023] Open
Abstract
Background Radiosensitivity is limited in cervical cancer (CC) patients due to acquired radiation resistance. In our previous studies, we found that immediate-early response 5 (IER5) is upregulated in CC cells upon radiation exposure and decreases cell survival by promoting apoptosis. The details on the transcriptional regulation of radiation-induced IER5 expression are unknown. Studies in recent years have suggested that Pol II-associated factor 1 (PAF1) is a pivotal transcription factor for certain genes “induced” during tumor progression. In this study, we investigated the role of PAF1 in regulating IER5 expression during CC radiotherapy. Methods PAF1 expression in CC cells was measured by western blotting, immunohistochemistry, and qRT-PCR, and the localization of PAF1 and IER5 was determined by immunofluorescence. The effect of PAF1 and IER5 knockdown by siRNA in Siha and Hela cells was studied by western blotting, qRT-PCR, CCK-8 assay, and flow cytometry. The physical interaction of PAF1 with the IER5 promoter and enhancers was confirmed using chromatin immunoprecipitation and qPCR with or without enhancers knockout by CRISPR/Cas9. Results We confirmed that PAF1 was highly expressed in CC cells and that relatively low expression of IER5 was observed in cells with highly expressed PAF1 in the nucleus. PAF1 knockdown in Siha and Hela cells was associated with increased expression of IER5, reduced cell viability and higher apoptosis rate in response to radiation exposure, while simultaneous PAF1 and IER5 knockdown had little effect on the proportion of apoptotic cells. We also found that PAF1 hindered the transcription of IER5 by promoting Pol II pausing at the promoter-proximal region, which was primarily due to the binding of PAF1 at the enhancers. Conclusions PAF1 reduces CC radiosensitivity by inhibiting IER5 transcription, at least in part by regulating its enhancers. PAF1 might be a potential therapeutic target for overcoming radiation resistance in CC patients.
Collapse
Affiliation(s)
- Jing-Jie Zheng
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Present address: Dong-Cheng District, Qi-He-Lou Street No.17, Beijing, 100006, China
| | - Yue He
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Present address: Dong-Cheng District, Qi-He-Lou Street No.17, Beijing, 100006, China
| | - Yang Liu
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Present address: Dong-Cheng District, Qi-He-Lou Street No.17, Beijing, 100006, China
| | - Feng-Shuang Li
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Present address: Dong-Cheng District, Qi-He-Lou Street No.17, Beijing, 100006, China
| | - Zhen Cui
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Present address: Dong-Cheng District, Qi-He-Lou Street No.17, Beijing, 100006, China
| | - Xiao-Meng Du
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Present address: Dong-Cheng District, Qi-He-Lou Street No.17, Beijing, 100006, China
| | - Chun-Peng Wang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Present address: Dong-Cheng District, Qi-He-Lou Street No.17, Beijing, 100006, China
| | - Yu-Mei Wu
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Present address: Dong-Cheng District, Qi-He-Lou Street No.17, Beijing, 100006, China.
| |
Collapse
|
27
|
Gilyazova IR, Klimentova EA, Bulygin KV, Izmailov AA, Bermisheva MA, Galimova EF, Safiullin RI, Galimov SN, Pavlov VN, Khusnutdinova EK. MicroRNA-200 family expression analysis in metastatic clear cell renal cell carcinoma patients. Cancer Gene Ther 2019; 27:768-772. [PMID: 31680118 DOI: 10.1038/s41417-019-0149-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/10/2019] [Accepted: 10/16/2019] [Indexed: 12/13/2022]
Abstract
The aim of this study is to analyse the of expression levels of microRNA-200 family members in patients with metastatic clear cell renal cell carcinoma (ccRCC). Analysis of microRNA expression was performed on 23 paired DNA samples extracted from kidney tumour tissue and the surrounding normal renal parenchyma. MicroRna-200c was found to have significantly lower expression (in kidney tumour tissue compared to normal renal parenchyma. No other microRna-200 family members showed statistically significant differences in expression levels between tumour and normal kidney tissue. Recent data suggest that the role of microRNA-200c in tumour pathogenesis is rather contradictory, and the underlying mechanisms by which microRNA-200c affects the carcinogenic potential of malignant cells remains unclear and requires further investigation at the molecular level.
Collapse
Affiliation(s)
- Irina R Gilyazova
- Institute of Biochemistry and Genetics, Ufa Federal Research Center of the Russian Academy of Sciences, Ufa, Russian Federation.,Bashkir State Medical University, Ufa, Russian Federation
| | - Elizaveta A Klimentova
- Institute of Biochemistry and Genetics, Ufa Federal Research Center of the Russian Academy of Sciences, Ufa, Russian Federation
| | - Kirill V Bulygin
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation. .,M.V. Lomonosov Moscow State University, Moscow, Russian Federation.
| | | | - Marina A Bermisheva
- Institute of Biochemistry and Genetics, Ufa Federal Research Center of the Russian Academy of Sciences, Ufa, Russian Federation
| | | | | | | | | | - Elsa K Khusnutdinova
- Institute of Biochemistry and Genetics, Ufa Federal Research Center of the Russian Academy of Sciences, Ufa, Russian Federation.,Bashkir State Medical University, Ufa, Russian Federation
| |
Collapse
|
28
|
Bahreyni-Toossi MT, Dolat E, Khanbabaei H, Zafari N, Azimian H. microRNAs: Potential glioblastoma radiosensitizer by targeting radiation-related molecular pathways. Mutat Res 2019; 816-818:111679. [PMID: 31715522 DOI: 10.1016/j.mrfmmm.2019.111679] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 09/30/2019] [Accepted: 10/12/2019] [Indexed: 06/10/2023]
Abstract
Glioblastoma (GBM) is the most lethal type of primary brain tumor. Currently, even with optimal and multimodal cancer therapies, the survival rate of GBM patients remains poor. One reason for inadequate response of GBM tumors to radiotherapy is radioresistance (RR). Thus, there is a critical need for new insights about GBM treatment to increase the chance of treatment. microRNAs (miRNAs) are important regulatory molecules that can effectively control GBM radiosensitivity (RS) by affecting radiation-related signal transduction pathways such as apoptosis, proliferation, DNA repair and cell cycle regulation. miRNAs provide new clinical perspectives for developing effective GBM treatments. A growing body of literature has demonstrated that GBM RS can be modified by modulating the expression of miRNAs such as miR-7, miR-10b, miR-124, miR-128, miR-320, miR-21, miR-203, and miR-153. This paper highlights the miRNAs and the underlying molecular mechanisms that are involved in the RS of GBM. Besides highlighting the role of miRNAs in different signaling pathways, we explain the mechanisms that affect RS of GBM for modulating radiation response at the clinical level.
Collapse
Affiliation(s)
| | - Elham Dolat
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hashem Khanbabaei
- Medical Physics Department, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Navid Zafari
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Hosein Azimian
- Medical Physics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
29
|
Bahreyni-Toossi MT, Dolat E, Khanbabaei H, Zafari N, Azimian H. microRNAs: Potential glioblastoma radiosensitizer by targeting radiation-related molecular pathways. Mutat Res 2019; 816-818:111679. [DOI: https:/doi.org/10.1016/j.mrfmmm.2019.111679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
|
30
|
Wang B, Zheng J, Li R, Tian Y, Lin J, Liang Y, Sun Q, Xu A, Zheng R, Liu M, Ji A, Bu J, Yuan Y. Long noncoding RNA LINC02582 acts downstream of miR-200c to promote radioresistance through CHK1 in breast cancer cells. Cell Death Dis 2019; 10:764. [PMID: 31601781 PMCID: PMC6787210 DOI: 10.1038/s41419-019-1996-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 09/14/2019] [Accepted: 09/17/2019] [Indexed: 01/17/2023]
Abstract
Radiotherapy is essential to treat breast cancer and microRNA (miRNA) miR-200c is considered as a radiosensitizer of breast cancer. However, the molecular mechanisms by which miR-200c regulates radiosensitivity remain largely unknown. In the present study, we showed that induction of miR-200c led to widespread alteration in long noncoding RNA (lncRNA) expression in breast cancer cells. We identified lncRNA LINC02582 as a target of miR-200c. Inhibition of LINC02582 expression increased radiosensitvity, while overexpression of LINC02582 promoted radioresistance. Mechanistically, LINC02582 interacts with deubiquitinating enzyme ubiquitin specific peptidase 7 (USP7) to deubiquitinate and stabilize checkpoint kinase 1 (CHK1), a critical effector kinase in DNA damage response, thus promoting radioresistance. Furthermore, we detected an inverse correlation between the expression of miR-200c vs. LINC02582 and CHK1 in breast cancer samples. These findings identified LINC02582 as a downstream target of miR-200c linking miR-200c to CHK1, in which miR-200c increases radiosensitivity by downregulation of CHK1.
Collapse
Affiliation(s)
- Baiyao Wang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Jieling Zheng
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Rong Li
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Yunhong Tian
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Jie Lin
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Yingying Liang
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Quanquan Sun
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, People's Republic of China
| | - Anan Xu
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Ronghui Zheng
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Mengzhong Liu
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Aimin Ji
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Junguo Bu
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.
| | - Yawei Yuan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China. .,Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong Province, People's Republic of China.
| |
Collapse
|
31
|
Non-coding RNAs: Regulators of glioma cell epithelial-mesenchymal transformation. Pathol Res Pract 2019; 215:152539. [DOI: 10.1016/j.prp.2019.152539] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 06/29/2019] [Accepted: 07/12/2019] [Indexed: 12/14/2022]
|
32
|
Long L, Zhang X, Bai J, Li Y, Wang X, Zhou Y. Tissue-specific and exosomal miRNAs in lung cancer radiotherapy: from regulatory mechanisms to clinical implications. Cancer Manag Res 2019; 11:4413-4424. [PMID: 31191004 PMCID: PMC6525830 DOI: 10.2147/cmar.s198966] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 04/15/2019] [Indexed: 12/16/2022] Open
Abstract
Lung cancer is the most prevalent and deadly malignancy. Radiotherapy is a major treatment modality for lung cancer. Nevertheless, radioresistance poses a daunting challenge that largely limits the efficacy of radiotherapy. There is a pressing need for deciphering molecular mechanisms underlying radioresistance and elucidating novel therapeutic targets for individualized radiotherapy. MicroRNAs are categorized as small noncoding RNAs that modulate target-gene expression posttranscriptionally and are implicated in carcinogenesis and cancer resistance to treatment. Overwhelming evidence has unraveled that tissue-specific miRNAs are essential for regulation of the radiosensitivity in lung cancer cells through a complex interaction with multiple biological processes and radiation-induced pathways. Moreover, exosome-derived miRNAs are a novel horizon in lung cancer treatment in which exosomal miRNAs act as potential diagnostic and therapeutic biomarkers of radiotherapy. In the present review, we discuss the mediation of key biological processes and signaling pathways by tissue-specific miRNAs in lung cancer radiotherapy. Additionally, we provide new insight into the potential significance of exosomal miRNAs in radiation response. Lastly, we highlight miRNAs as promising predictors and therapeutic targets to tailor personalized lung cancer radiotherapy.
Collapse
Affiliation(s)
- Long Long
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430071, People's Republic of China
| | - Xue Zhang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430071, People's Republic of China
| | - Jian Bai
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuhan, 430071, People's Republic of China
| | - Yizhou Li
- Department of Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, People's Republic of China
| | - Xiaolong Wang
- Department of Urology, Research Lab/LIFE-Zentrum, University of Munich (LMU), München, Germany
| | - Yunfeng Zhou
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430071, People's Republic of China
| |
Collapse
|
33
|
Vigorito C, Anishchenko E, Mele L, Capolongo G, Trepiccione F, Zacchia M, Lombari P, Capasso R, Ingrosso D, Perna AF. Uremic Toxin Lanthionine Interferes with the Transsulfuration Pathway, Angiogenetic Signaling and Increases Intracellular Calcium. Int J Mol Sci 2019; 20:E2269. [PMID: 31071929 PMCID: PMC6539355 DOI: 10.3390/ijms20092269] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/03/2019] [Accepted: 05/06/2019] [Indexed: 01/08/2023] Open
Abstract
(1) The beneficial effects of hydrogen sulfide (H2S) on the cardiovascular and nervous system have recently been re-evaluated. It has been shown that lanthionine, a side product of H2S biosynthesis, previously used as a marker for H2S production, is dramatically increased in circulation in uremia, while H2S release is impaired. Thus, lanthionine could be classified as a novel uremic toxin. Our research was aimed at defining the mechanism(s) for lanthionine toxicity. (2) The effect of lanthionine on H2S release was tested by a novel lead acetate strip test (LAST) in EA.hy926 cell cultures. Effects of glutathione, as a redox agent, were assayed. Levels of sulfane sulfur were evaluated using the SSP4 probe and flow cytometry. Protein content and glutathionylation were analyzed by Western Blotting and immunoprecipitation, respectively. Gene expression and miRNA levels were assessed by qPCR. (3) We demonstrated that, in endothelial cells, lanthionine hampers H2S release; reduces protein content and glutathionylation of transsulfuration enzyme cystathionine-β-synthase; modifies the expression of miR-200c and miR-423; lowers expression of vascular endothelial growth factor VEGF; increases Ca2+ levels. (4) Lanthionine-induced alterations in cell cultures, which involve both sulfur amino acid metabolism and calcium homeostasis, are consistent with uremic dysfunctional characteristics and further support the uremic toxin role of this amino acid.
Collapse
Affiliation(s)
- Carmela Vigorito
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," 80131 Naples, Italy.
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli," 80138 Naples, Italy.
| | - Evgeniya Anishchenko
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," 80131 Naples, Italy.
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli," 80138 Naples, Italy.
| | - Luigi Mele
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli," 80138 Naples, Italy.
| | - Giovanna Capolongo
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," 80131 Naples, Italy.
| | - Francesco Trepiccione
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," 80131 Naples, Italy.
- Biogem A. C. S. R. L. Contrada Camporeale, 83031 Ariano Irpino AV, Italy.
| | - Miriam Zacchia
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," 80131 Naples, Italy.
| | - Patrizia Lombari
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," 80131 Naples, Italy.
| | - Rosanna Capasso
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli," 80138 Naples, Italy.
| | - Diego Ingrosso
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli," 80138 Naples, Italy.
| | - Alessandra F Perna
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," 80131 Naples, Italy.
| |
Collapse
|
34
|
Hawkins PG, Sun Y, Dess RT, Jackson WC, Sun G, Bi N, Tewari M, Hayman JA, Kalemkerian GP, Gadgeel SM, Lawrence TS, Haken RKT, Matuszak MM, Kong FMS, Schipper MJ, Jolly S. Circulating microRNAs as biomarkers of radiation-induced cardiac toxicity in non-small-cell lung cancer. J Cancer Res Clin Oncol 2019; 145:1635-1643. [PMID: 30923943 DOI: 10.1007/s00432-019-02903-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 03/22/2019] [Indexed: 12/13/2022]
Abstract
PURPOSE Radiation-induced cardiac toxicity (RICT) is an increasingly well-appreciated source of morbidity and mortality in patients receiving thoracic radiotherapy (RT). Currently available methods to predict RICT are suboptimal. We investigated circulating microRNAs (c-miRNAs) as potential biomarkers of RICT in patients undergoing definitive RT for non-small-cell lung cancer (NSCLC). METHODS Data from 63 patients treated on institutional trials were analyzed. Prognostic models of grade 3 or greater (G3 +) RICT based on pre-treatment c-miRNA levels ('c-miRNA'), mean heart dose (MHD) and pre-existing cardiac disease (PCD) ('clinical'), and a combination of these ('c-miRNA + clinical') were developed. Elastic net Cox regression and full cross validation were used for variable selection, model building, and model evaluation. Concordance statistic (c-index) and integrated Brier score (IBS) were used to evaluate model performance. RESULTS MHD, PCD, and serum levels of 14 c-miRNA species were identified as jointly prognostic for G3 + RICT. The 'c-miRNA and 'clinical' models yielded similar cross-validated c-indices (0.70 and 0.72, respectively) and IBSs (0.26 and 0.28, respectively). However, prognostication was not improved by combining c-miRNA and clinical factors (c-index 0.70, IBS 0.28). The 'c-miRNA' and 'clinical' models were able to significantly stratify patients into high- and low-risk groups of developing G3 + RICT. Chi-square testing demonstrated a marginally significantly higher prevalence of PCD in patients with high- compared to low-risk c-miRNA profile (p = 0.09), suggesting an association between some c-miRNAs and PCD. CONCLUSIONS We identified a pre-treatment c-miRNA signature prognostic for G3 + RICT. With further development, pre- and mid-treatment c-miRNA profiling could contribute to patient-specific dose selection and treatment adaptation.
Collapse
Affiliation(s)
- Peter G Hawkins
- Department of Radiation Oncology, University of Michigan, 1500 E Medical Center Drive, UH B2 C490 SPC 5010, Ann Arbor, MI, 48109, USA
| | - Yilun Sun
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Robert T Dess
- Department of Radiation Oncology, University of Michigan, 1500 E Medical Center Drive, UH B2 C490 SPC 5010, Ann Arbor, MI, 48109, USA
| | - William C Jackson
- Department of Radiation Oncology, University of Michigan, 1500 E Medical Center Drive, UH B2 C490 SPC 5010, Ann Arbor, MI, 48109, USA
| | - Grace Sun
- Department of Radiation Oncology, University of Michigan, 1500 E Medical Center Drive, UH B2 C490 SPC 5010, Ann Arbor, MI, 48109, USA
| | - Nan Bi
- Department of Radiation Oncology, Cancer Hospital and Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Muneesh Tewari
- Department of Biomedical Engineering, Biointerfaces Institute, and Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - James A Hayman
- Department of Radiation Oncology, University of Michigan, 1500 E Medical Center Drive, UH B2 C490 SPC 5010, Ann Arbor, MI, 48109, USA
| | - Gregory P Kalemkerian
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Shirish M Gadgeel
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan, 1500 E Medical Center Drive, UH B2 C490 SPC 5010, Ann Arbor, MI, 48109, USA
| | - Randall K Ten Haken
- Department of Radiation Oncology, University of Michigan, 1500 E Medical Center Drive, UH B2 C490 SPC 5010, Ann Arbor, MI, 48109, USA
| | - Martha M Matuszak
- Department of Radiation Oncology, University of Michigan, 1500 E Medical Center Drive, UH B2 C490 SPC 5010, Ann Arbor, MI, 48109, USA
| | - Feng-Ming Spring Kong
- Department of Radiation Oncology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Matthew J Schipper
- Department of Radiation Oncology, University of Michigan, 1500 E Medical Center Drive, UH B2 C490 SPC 5010, Ann Arbor, MI, 48109, USA
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Shruti Jolly
- Department of Radiation Oncology, University of Michigan, 1500 E Medical Center Drive, UH B2 C490 SPC 5010, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
35
|
D'Almeida O, Mothar O, Bondzie EA, Lieumo Y, Tagne L, Gupta S, Volkert T, Levine S, Tagne JB. Encapsulated miR-200c and Nkx2.1 in a nuclear/mitochondria transcriptional regulatory network of non-metastatic and metastatic lung cancer cells. BMC Cancer 2019; 19:136. [PMID: 30744585 PMCID: PMC6371494 DOI: 10.1186/s12885-019-5337-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 01/31/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND MicroRNAs are noncoding RNA molecules of ~ 22 nucleotides with diagnostic and therapeutic action [Curr Drug Targets, 2015. 16(12): p. 1381-403], affecting the expression of mRNAs involved in invasion, migration, and development [Oncotarget, 2015. 6(9): p. 6472-98, Cancer Manag Res, 2014. 6: p. 205-16]. miR-200c is part of the miR-200c/141 cluster on chromosome 12p13. Its mechanism of action when encapsulated is critical in lung cancer when patients express changes in miRNAs. miR-200c be a potential biomarkers for various lung diseases. As a potential therapy, miR-200c can impacts lives as target lung cancer is a leading cause of death with about 234,000 cases annually, high heterogeneity, complex screening, and a 5-year survival rate of 16% [CA Cancer J Clin, 2016.66(1): p. 7-30]. Encapsulated miR-200c efficiently enhances bioavailability, pharmacokinetics of therapeutics and targeting to cells, improves efficacy and provides potential cure. METHODS The functions of miR-200c were determined in non-metastatic KW-634 and metastatic 821-T4 and 821-LN mouse lung cancer cell lines after various Nano vehicle treatments. Viability and cytotoxicity were determined by cell cycle and quantitative real-time PCR analyses were used to quantify levels of miR-200c and its target genes. In situ hybridization was used to visualize patterns of expression of miR-200c and others in the lung and many organs. Next-generation sequencing accession number GSE125000, invasion and migration assays using transwell chambers, and ActivSignal were used to elucidate the activation and inhibition profiles and perform direct expression measurements and modification of cellular components. RESULTS Due to their effectiveness as intracellular vesicles transporting miR-200c into, out, and between parts of the cells, miR-200c is encapsulated with cholesterol, an integral part of the biological membranes with very important physical properties of the vehicle. Nano miR-200c showed efficient cellular uptake in KW-634, 821-T4, and 821-LN cells with important changes in gene expression and new isoforms. In KW-634, when treated with encapsulated miR-200c and compare to the non-encapsulated control; miR-29b increased by 5261-fold, and in 821-T4/LN, miR-1247 increased by 150-fold. Conversely, miR-1247 and miR-675 decreased by 348 and 1029.5-fold, respectively. miR-189 decreased by 34-fold in treated 821-T4 cells. A reduction of growth was observed only after 48 h of treatment with Nano miR-200c. Moreover, labeling the vehicle with carboxy-fluorescein showed that the encapsulated particles enter the nucleus and mitochondria. Encapsulated miR-200c by entering the cells, the nucleus and mitochondria, trigger changes in cell cycle phases with 4 up to 12 fold percentage in G2 and S phase respectively compare to miR-200c. Endogenous expression of Nkx2.1, miR-200c, and their targets Myb, Nfib, Six4 and Six1 showed an inverse correlation, as observed in development. CONCLUSIONS Little is known about miR-200c involvement in regulatory processes. Nano miR-200c affects invasion and migration mechanisms. The expression of encapsulated miR-200c contributes to the inhibition/activation of Kras, EMT, Hippo, regulatory pathways and blockers of metastasis. Delivery of miR-200c increases the expression of miR-29b, an EMY regulator, and miR-1247, an inhibitor of cancer genes, both tumor suppressors involved in lung metastasis. Encapsulated miR-200c act on different proteins that regulates cell cycle pathways. These findings represent a part of a regulatory network providing new insights towards improvement of therapy.
Collapse
Affiliation(s)
- Olga D'Almeida
- The Pulmonary Center, Boston University School of Medicine (BUSM), East Concord Street R304, Boston, MA, 02118, USA.,Faculté de Pharmacie, Université D'Auvergne, Clermont Ferrand, France
| | - Omar Mothar
- The Pulmonary Center, Boston University School of Medicine (BUSM), East Concord Street R304, Boston, MA, 02118, USA
| | - Esther Apraku Bondzie
- The Pulmonary Center, Boston University School of Medicine (BUSM), East Concord Street R304, Boston, MA, 02118, USA
| | - Yolande Lieumo
- The Pulmonary Center, Boston University School of Medicine (BUSM), East Concord Street R304, Boston, MA, 02118, USA
| | - Laure Tagne
- The Pulmonary Center, Boston University School of Medicine (BUSM), East Concord Street R304, Boston, MA, 02118, USA
| | - Sumeet Gupta
- Whitehead Institute for Biomedical Research (WIBR), Nine Cambridge Center Cambridge, Cambridge, MA, 02142, USA
| | - Thomas Volkert
- Whitehead Institute for Biomedical Research (WIBR), Nine Cambridge Center Cambridge, Cambridge, MA, 02142, USA
| | - Stuart Levine
- Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA, 02139, USA
| | - Jean-Bosco Tagne
- The Pulmonary Center, Boston University School of Medicine (BUSM), East Concord Street R304, Boston, MA, 02118, USA.
| |
Collapse
|
36
|
El Bezawy R, Tinelli S, Tortoreto M, Doldi V, Zuco V, Folini M, Stucchi C, Rancati T, Valdagni R, Gandellini P, Zaffaroni N. miR-205 enhances radiation sensitivity of prostate cancer cells by impairing DNA damage repair through PKCε and ZEB1 inhibition. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:51. [PMID: 30717752 PMCID: PMC6360656 DOI: 10.1186/s13046-019-1060-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/27/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Radiotherapy is one of the main treatment options for non-metastatic prostate cancer (PCa). Although treatment technical optimization has greatly improved local tumor control, a considerable fraction of patients still experience relapse due to the development of resistance. Radioresistance is a complex and still poorly understood phenomenon involving the deregulation of a variety of signaling pathways as a consequence of several genetic and epigenetic abnormalities. In this context, cumulative evidence supports a functional role of microRNAs in affecting radioresistance, suggesting the modulation of their expression as a novel radiosensitizing approach. Here, we investigated for the first time the ability of miR-205 to enhance the radiation response of PCa models. METHODS miR-205 reconstitution by a miRNA mimic in PCa cell lines (DU145 and PC-3) was used to elucidate miR-205 biological role. Radiation response in miRNA-reconstituted and control cells was assessed by clonogenic assay, immunofluorescence-based detection of nuclear γ-H2AX foci and comet assay. RNAi was used to silence the miRNA targets PKCε or ZEB1. In addition, target-protection experiments were carried out using a custom oligonucleotide designed to physically disrupt the pairing between the miR-205 and PKCε. For in vivo experiments, xenografts generated in SCID mice by implanting DU145 cells stably expressing miR-205 were exposed to 5-Gy single dose irradiation using an image-guided animal micro-irradiator. RESULTS miR-205 reconstitution was able to significantly enhance the radiation response of prostate cancer cell lines and xenografts through the impairment of radiation-induced DNA damage repair, as a consequence of PKCε and ZEB1 inhibition. Indeed, phenocopy experiments based on knock-down of either PKCε or ZEB1 reproduced miR-205 radiosensitizing effect, hence confirming a functional role of both targets in the process. At the molecular level, miR-205-induced suppression of PKCε counteracted radioresistance through the impairment of EGFR nuclear translocation and the consequent DNA-PK activation. Consistently, disruption of miR-205-PKCε 3'UTR pairing almost completely abrogated the radiosensitizing effect. CONCLUSIONS Our results uncovered the molecular and cellular mechanisms underlying the radiosensitizing effect of miR-205. These findings support the clinical interest in developing a novel therapeutic approach based on miR-205 reconstitution to increase PCa response to radiotherapy.
Collapse
Affiliation(s)
- Rihan El Bezawy
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, 20133, Milan, Italy
| | - Stella Tinelli
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, 20133, Milan, Italy
| | - Monica Tortoreto
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, 20133, Milan, Italy
| | - Valentina Doldi
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, 20133, Milan, Italy
| | - Valentina Zuco
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, 20133, Milan, Italy
| | - Marco Folini
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, 20133, Milan, Italy
| | - Claudio Stucchi
- Medical Physics Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133, Milan, Italy
| | - Tiziana Rancati
- Prostate Cancer Program, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133, Milan, Italy
| | - Riccardo Valdagni
- Prostate Cancer Program, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133, Milan, Italy.,Radiation Oncology 1 Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133, Milan, Italy
| | - Paolo Gandellini
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, 20133, Milan, Italy
| | - Nadia Zaffaroni
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, 20133, Milan, Italy.
| |
Collapse
|
37
|
Tang L, Hu H, He Y, Mcleod HL, Xiao D, Chen P, Shen L, Zeng S, Yin X, Ge J, Li L, Ma J, Chen Z, Huang J. The relationship between miR-302b and EphA2 and their clinical significance in gastric cancer. J Cancer 2018; 9:3109-3116. [PMID: 30210634 PMCID: PMC6134821 DOI: 10.7150/jca.25235] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 06/16/2018] [Indexed: 12/18/2022] Open
Abstract
Introduction: EphA2 is a crucial oncogene in gastric cancer (GC) development and metastasis, and miR-302b can target EphA2 in gastric cancer. This study plans to investigate their relationship and clinical significance in clinical samples. Materials and Methods: We explored the correlation of the expression of EphA2 and miR-302b, and their clinical significance in the training (n=226) cohort of GC patients, and then validated the results in the validation (n=128) cohort. Results: miR-302b was remarkably downregulated in GC tissues, while high EphA2 expression were detected, and they were inversely correlated both in mRNA and protein, (r=-0.4209, P<0.0001; r=-0.336, P <0.001, respectively). Furthermore, the pattern of high EphA2 and low miR-302b expression were found to be associated with poor overall survival in stage IV GC patients in both training and validation cohort. Conclusions: The expression of miR-302b and EphA2 was inversely correlated, and had prognostic significance on GC in clinic.
Collapse
Affiliation(s)
- Lanhua Tang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Huabin Hu
- The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510655, China
| | - Yijing He
- Department of Dermatology, XiangYa Hospital, Central South University, Changsha, 410008, China
| | - Howard L Mcleod
- Department of Dermatology, XiangYa Hospital, Central South University, Changsha, 410008, China.,Department of Clinical Pharmacology, XiangYa Hospital, Central South University, Changsha, 410008, China.,Moffitt Cancer Center, DeBartolo Family Personalized Medicine Institute, Tampa, FL 33612, USA
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Pan Chen
- Department of Hepatobiliary Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Liangfang Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xianli Yin
- Department of gastroenterology and urology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013,China
| | - Jie Ge
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Li Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jian Ma
- Cancer Research Institute, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Central South University, Changsha, 410008, China
| | - Zihua Chen
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jin Huang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| |
Collapse
|
38
|
Abstract
Gliomas are the most common type of primary brain tumors. MicroRNAs (miRNAs) are small noncoding RNAs that can epigenetically regulate target gene expression. The microRNA 200 family includes miR-200a, 200b, 200c, 141 and 429. Numerous studies have indicated that members of the miR-200 family play an important role in glioma development and metastasis. In this review, we summarize the data from various studies and highlight the effects of miR-200 on glioma metastasis, therapeutic response and prognosis.
Collapse
Affiliation(s)
- Lilei Peng
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, People's Republic of China,
| | - Jie Fu
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, People's Republic of China
| | - Yang Ming
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, People's Republic of China,
| |
Collapse
|
39
|
Abisoye-Ogunniyan A, Lin H, Ghebremedhin A, Salam AB, Karanam B, Theodore S, Jones-Trich J, Davis M, Grizzle W, Wang H, Yates C. Transcriptional repressor Kaiso promotes epithelial to mesenchymal transition and metastasis in prostate cancer through direct regulation of miR-200c. Cancer Lett 2018; 431:1-10. [PMID: 29751044 DOI: 10.1016/j.canlet.2018.04.044] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 04/26/2018] [Accepted: 04/27/2018] [Indexed: 01/06/2023]
Abstract
The loss of miR-200 family, through DNA methylation, results in cancer cells undergoing an epithelial to mesenchymal transition (EMT), and metastasis. In this study, we established that the transcriptional repressor Kaiso directly binds methylated regions of the miR-200 family, and this is reversed with 5-aza treatment. sh-Kaiso PC-3 cells display increased miR-200-a/b/c, miR-141, and miR-429 expression, with miR-200c demonstrating the most significant increase. Interestingly, overexpression of EGFR or treatment with EGF decreases miR-200c expression and this is reversed after treatment with EGFR specific kinase inhibitor PD153035. However, EGF did not have a significant effect on miR-200c in sh-Kaiso DU-145 or PC-3 cell lines, suggesting Kaiso silences miR-200c through the activation of EGFR signaling. Overexpression of Kaiso in LNCaP cells results in decreased expression of miR-200-a/b/c, miR-141, and miR-429, along with increased expression of ZEB1, p-EGFR and total EGFR levels. Overexpression of miR200c in PC-3 cells results in decreased expression of EGFR, ZEB1, ERK1/2 and Kaiso. Additionally, sh-Kaiso PC-3 demonstrates reduced in vivo tumor formation and metastasis. Thus, our data suggests that EGFR signaling regulates the silencing of miR-200 family through Kaiso binding to methylated regions in the promoter.
Collapse
Affiliation(s)
| | - Huxian Lin
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA
| | - Anghesom Ghebremedhin
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA
| | - Ahmad Bin Salam
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA
| | - Balasubramanyam Karanam
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA
| | - Shaniece Theodore
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA
| | | | - Melissa Davis
- Department of Surgery, Henry Ford Medical Center, Detroit, MI, USA
| | - William Grizzle
- Department of Pathology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Honghe Wang
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA
| | - Clayton Yates
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA.
| |
Collapse
|
40
|
Molecular Determinants of Malignant Brain Cancers: From Intracellular Alterations to Invasion Mediated by Extracellular Vesicles. Int J Mol Sci 2017; 18:ijms18122774. [PMID: 29261132 PMCID: PMC5751372 DOI: 10.3390/ijms18122774] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/29/2017] [Accepted: 12/19/2017] [Indexed: 12/15/2022] Open
Abstract
Malignant glioma cells invade the surrounding brain parenchyma, by migrating along the blood vessels, thus promoting cancer growth. The biological bases of these activities are grounded in profound alterations of the metabolism and the structural organization of the cells, which consequently acquire the ability to modify the surrounding microenvironment, by altering the extracellular matrix and affecting the properties of the other cells present in the brain, such as normal glial-, endothelial- and immune-cells. Most of the effects on the surrounding environment are probably exerted through the release of a variety of extracellular vesicles (EVs), which contain many different classes of molecules, from genetic material to defined species of lipids and enzymes. EV-associated molecules can be either released into the extracellular matrix (ECM) and/or transferred to neighboring cells: as a consequence, both deep modifications of the recipient cell phenotype and digestion of ECM components are obtained, thus causing cancer propagation, as well as a general brain dysfunction. In this review, we first analyze the main intracellular and extracellular transformations required for glioma cell invasion into the brain parenchyma; then we discuss how these events may be attributed, at least in part, to EVs that, like the pawns of a dramatic chess game with cancer, open the way to the tumor cells themselves.
Collapse
|
41
|
Wang H, Mu X, He H, Zhang XD. Cancer Radiosensitizers. Trends Pharmacol Sci 2017; 39:24-48. [PMID: 29224916 DOI: 10.1016/j.tips.2017.11.003] [Citation(s) in RCA: 351] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/08/2017] [Accepted: 11/09/2017] [Indexed: 02/07/2023]
Abstract
Radiotherapy (RT) is a mainstay treatment for many types of cancer, although it is still a large challenge to enhance radiation damage to tumor tissue and reduce side effects to healthy tissue. Radiosensitizers are promising agents that enhance injury to tumor tissue by accelerating DNA damage and producing free radicals. Several strategies have been exploited to develop highly effective and low-toxicity radiosensitizers. In this review, we highlight recent progress on radiosensitizers, including small molecules, macromolecules, and nanomaterials. First, small molecules are reviewed based on free radicals, pseudosubstrates, and other mechanisms. Second, nanomaterials, such as nanometallic materials, especially gold-based materials that have flexible surface engineering and favorable kinetic properties, have emerged as promising radiosensitizers. Finally, emerging macromolecules have shown significant advantages in RT because these molecules can be combined with biological therapy as well as drug delivery. Further research on the mechanisms of radioresistance and multidisciplinary approaches will accelerate the development of radiosensitizers.
Collapse
Affiliation(s)
- Hao Wang
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Number 238, Baidi Road, Tianjin 300192, China; These authors have contributed equally
| | - Xiaoyu Mu
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China; These authors have contributed equally
| | - Hua He
- State Key Laboratory of Heavy Oil Processing and Center for Bioengineering and Biotechnology, China University of Petroleum (East China), Qingdao 266580, China
| | - Xiao-Dong Zhang
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China; Tianjin Collaborative Innovation Center of Chemical Science and Engineering, Tianjin 300072, China.
| |
Collapse
|