101
|
Caracciolo D, Di Martino MT, Amodio N, Morelli E, Montesano M, Botta C, Scionti F, Talarico D, Altomare E, Gallo Cantafio ME, Zuccalà V, Maltese L, Todoerti K, Rossi M, Arbitrio M, Neri A, Tagliaferri P, Tassone P. miR-22 suppresses DNA ligase III addiction in multiple myeloma. Leukemia 2019; 33:487-498. [PMID: 30120376 PMCID: PMC6365379 DOI: 10.1038/s41375-018-0238-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 07/04/2018] [Accepted: 07/13/2018] [Indexed: 12/15/2022]
Abstract
Multiple myeloma (MM) is a hematologic malignancy characterized by high genomic instability. Here we provide evidence that hyper-activation of DNA ligase III (LIG3) is crucial for genomic instability and survival of MM cells. LIG3 mRNA expression in MM patients correlates with shorter survival and even increases with more advanced stage of disease. Knockdown of LIG3 impairs MM cells viability in vitro and in vivo, suggesting that neoplastic plasmacells are dependent on LIG3-driven repair. To investigate the mechanisms involved in LIG3 expression, we investigated the post-transcriptional regulation. We identified miR-22-3p as effective negative regulator of LIG3 in MM. Enforced expression of miR-22 in MM cells downregulated LIG3 protein, which in turn increased DNA damage inhibiting in vitro and in vivo cell growth. Taken together, our findings demonstrate that myeloma cells are addicted to LIG3, which can be effectively inhibited by miR-22, promoting a novel axis of genome stability regulation.
Collapse
Affiliation(s)
- Daniele Caracciolo
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | - Eugenio Morelli
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | - Martina Montesano
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | - Cirino Botta
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | - Francesca Scionti
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | | | - Emanuela Altomare
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | - Maria Eugenia Gallo Cantafio
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | | | | | - Katia Todoerti
- Department of Oncology and Hemato-oncology, University of Milan, and Hematology, Fondazione Cà Granda IRCCS Policlinico, Milan, Italy
| | - Marco Rossi
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | - Mariamena Arbitrio
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | - Antonino Neri
- Department of Oncology and Hemato-oncology, University of Milan, and Hematology, Fondazione Cà Granda IRCCS Policlinico, Milan, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy.
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
102
|
Pucella JN, Cols M, Yen WF, Xu S, Chaudhuri J. The B Cell Activation-Induced miR-183 Cluster Plays a Minimal Role in Canonical Primary Humoral Responses. THE JOURNAL OF IMMUNOLOGY 2019; 202:1383-1396. [PMID: 30683701 DOI: 10.4049/jimmunol.1800071] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 12/05/2018] [Indexed: 12/18/2022]
Abstract
Although primary humoral responses are vital to durable immunity, fine-tuning is critical to preventing catastrophes such as autoimmunity, chronic inflammation, and lymphomagenesis. MicroRNA (miRNA)-mediated regulation is particularly well suited for fine-tuning roles in physiology. Expression of clustered paralogous miR-182, miR-96, and miR-183 (collectively, 183c) is robustly induced upon B cell activation, entry into the germinal center, and plasmablast differentiation. 183cGT/GT mice lacking 183c miRNA expression exhibit largely normal primary humoral responses, encompassing class switch recombination, affinity maturation, and germinal center reaction, as well as plasmablast differentiation. Our rigorous analysis included ex vivo class switch recombination and plasmablast differentiation models as well as in vivo immunization with thymus-dependent and thymus-independent Ags. Our work sways the debate concerning the role of miR-182 in plasmablast differentiation, strongly suggesting that 183c miRNAs are dispensable. In the process, we present a valuable framework for systematic evaluation of primary humoral responses. Finally, our work bolsters the notion of robustness in miRNA:target interaction networks and advocates a paradigm shift in miRNA studies.
Collapse
Affiliation(s)
- Joseph N Pucella
- Immunology Program, Memorial Sloan Kettering Cancer Center, Gerstner Sloan Kettering Graduate School, New York, NY 10065; and
| | - Montserrat Cols
- Immunology Program, Memorial Sloan Kettering Cancer Center, Gerstner Sloan Kettering Graduate School, New York, NY 10065; and
| | - Wei-Feng Yen
- Immunology Program, Memorial Sloan Kettering Cancer Center, Gerstner Sloan Kettering Graduate School, New York, NY 10065; and
| | - Shunbin Xu
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48201
| | - Jayanta Chaudhuri
- Immunology Program, Memorial Sloan Kettering Cancer Center, Gerstner Sloan Kettering Graduate School, New York, NY 10065; and
| |
Collapse
|
103
|
Jimeno S, Mejías-Navarro F, Prados-Carvajal R, Huertas P. Controlling the balance between chromosome break repair pathways. DNA Repair (Amst) 2019; 115:95-134. [DOI: 10.1016/bs.apcsb.2018.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
104
|
Sun K, Mikule K, Wang Z, Poon G, Vaidyanathan A, Smith G, Zhang ZY, Hanke J, Ramaswamy S, Wang J. A comparative pharmacokinetic study of PARP inhibitors demonstrates favorable properties for niraparib efficacy in preclinical tumor models. Oncotarget 2018; 9:37080-37096. [PMID: 30647846 PMCID: PMC6324689 DOI: 10.18632/oncotarget.26354] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/28/2018] [Indexed: 01/19/2023] Open
Abstract
Niraparib is an orally bioavailable and selective poly (ADP-ribose) polymerase (PARP)-1/-2 inhibitor approved for maintenance treatment of both BRCA mutant (mut) and BRCA wildtype (wt) adult patients with recurrent epithelial ovarian, fallopian tube, or primary peritoneal cancers who have demonstrated a complete or partial response to platinum-based chemotherapy. In patients without germline BRCA mutations (non-gBRCAmut), niraparib improved progression-free survival (PFS) by 5.4 months, whereas another PARP inhibitor (PARPi) olaparib supplied only 1.9 months of improvement in a similar patient population. Previous studies revealed higher cell membrane permeability and volume of distribution (VD) as unique features of niraparib in comparison to other PARPi including olaparib. Here, we explore the potential correlation of these pharmacokinetic properties to preclinical antitumor effects in BRCAwt tumors. Our results show that at steady state, tumor exposure to niraparib is 3.3 times greater than plasma exposure in tumor xenograft mouse models. In comparison, the tumor exposure to olaparib is less than observed in plasma. In addition, niraparib crosses the blood-brain barrier and shows good sustainability in the brain, whereas sustained brain exposure to olaparib is not observed in the same models. Consistent with its favorable tumor and brain distribution, niraparib achieves more potent tumor growth inhibition than olaparib in BRCAwt models and an intracranial tumor model at maximum tolerated doses (MTD). These findings demonstrate favorable pharmacokinetic profiles and potent antitumor effects of niraparib in BRCAwt tumors, consistent with its broader clinical effect in patients with both BRCAmut and BRCAwt tumors.
Collapse
Affiliation(s)
| | | | | | | | - Aparajitha Vaidyanathan
- Division of Cellular Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital & Medical School, Dundee, UK
| | - Gillian Smith
- Division of Cellular Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital & Medical School, Dundee, UK
| | | | | | | | | |
Collapse
|
105
|
Dhawan A, Scott JG, Harris AL, Buffa FM. Pan-cancer characterisation of microRNA across cancer hallmarks reveals microRNA-mediated downregulation of tumour suppressors. Nat Commun 2018; 9:5228. [PMID: 30531873 PMCID: PMC6286392 DOI: 10.1038/s41467-018-07657-1] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 11/16/2018] [Indexed: 12/12/2022] Open
Abstract
microRNAs are key regulators of the human transcriptome across a number of diverse biological processes, such as development, aging and cancer, where particular miRNAs have been identified as tumour suppressive and oncogenic. In this work, we elucidate, in a comprehensive manner, across 15 epithelial cancer types comprising 7316 clinical samples from the Cancer Genome Atlas, the association of miRNA expression and target regulation with the phenotypic hallmarks of cancer. Utilising penalised regression techniques to integrate transcriptomic, methylation and mutation data, we find evidence for a complex map of interactions underlying the relationship of miRNA regulation and the hallmarks of cancer. This highlighted high redundancy for the oncomiR-1 cluster of oncogenic miRNAs, in particular hsa-miR-17-5p. In addition, we reveal extensive miRNA regulation of tumour suppressor genes such as PTEN, FAT4 and CDK12, uncovering an alternative mechanism of repression in the absence of mutation, methylation or copy number changes.
Collapse
Affiliation(s)
- Andrew Dhawan
- Department of Oncology, Medical Sciences Division, University of Oxford, Oxford, OX3 7DQ, UK
| | - Jacob G Scott
- Translational Hematology and Radiology, Cleveland Clinic, Cleveland, 44195, USA
| | - Adrian L Harris
- Department of Oncology, Medical Sciences Division, University of Oxford, Oxford, OX3 7DQ, UK
| | - Francesca M Buffa
- Department of Oncology, Medical Sciences Division, University of Oxford, Oxford, OX3 7DQ, UK.
| |
Collapse
|
106
|
Faraoni I, Graziani G. Role of BRCA Mutations in Cancer Treatment with Poly(ADP-ribose) Polymerase (PARP) Inhibitors. Cancers (Basel) 2018; 10:E487. [PMID: 30518089 PMCID: PMC6316750 DOI: 10.3390/cancers10120487] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 11/29/2018] [Accepted: 11/30/2018] [Indexed: 12/29/2022] Open
Abstract
Inhibition of poly(ADP-ribose) polymerase (PARP) activity induces synthetic lethality in mutated BRCA1/2 cancers by selectively targeting tumor cells that fail to repair DNA double strand breaks (DSBs). Clinical studies have confirmed the validity of the synthetic lethality approach and four different PARP inhibitors (PARPi; olaparib, rucaparib, niraparib and talazoparib) have been approved as monotherapies for BRCA-mutated or platinum-sensitive recurrent ovarian cancer and/or for BRCA-mutated HER2-negative metastatic breast cancer. PARPi therapeutic efficacy is higher against tumors harboring deleterious germline or somatic BRCA mutations than in BRCA wild-type tumors. BRCA mutations or intrinsic tumor sensitivity to platinum compounds are both regarded as indicators of deficiency in DSB repair by homologous recombination as well as of favorable response to PARPi. However, not all BRCA-mutated or platinum-responsive patients obtain clinical benefit from these agents. Conversely, a certain percentage of patients with wild-type BRCA or platinum-resistant tumors can still get benefit from PARPi. Thus, additional reliable markers need to be validated in clinical trials to select patients potentially eligible for PARPi-based therapies, in the absence of deleterious BRCA mutations or platinum sensitivity. In this review, we summarize the mechanisms of action of PARPi and the clinical evidence supporting their use as anticancer drugs as well as the additional synthetic lethal partners that might confer sensitivity to PARPi in patients with wild-type BRCA tumors.
Collapse
Affiliation(s)
- Isabella Faraoni
- Department of Systems Medicine, University of Rome Tor Vergata, 00173 Rom, Italy.
| | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, 00173 Rom, Italy.
| |
Collapse
|
107
|
Konstantinopoulos PA, Matulonis UA. Targeting DNA Damage Response and Repair as a Therapeutic Strategy for Ovarian Cancer. Hematol Oncol Clin North Am 2018; 32:997-1010. [DOI: 10.1016/j.hoc.2018.07.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
108
|
miR-509-3p enhances platinum drug sensitivity in ovarian cancer. Gene 2018; 686:63-67. [PMID: 30408550 DOI: 10.1016/j.gene.2018.11.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/23/2018] [Accepted: 11/03/2018] [Indexed: 01/08/2023]
Abstract
Drug-resistance of platinum remains a big challenge for effective treatment of patients with ovarian cancer. MicroRNAs (miRNAs) act as post-transcriptional regulators of gene expression and are associated with multi-drug resistance. Our study aims on identifying role of miRNAs in drug-resistance of platinum in ovarian cancer. In present study, we compared the expression profiles of miRNAs between three pairs of platinum-resistant and platinum-sensitive ovarian tissues and found that miR-509-3p was significantly down-regulated in cisplatin-resistant ovarian cancer tissues. The different expression of miR-509-3p was further determined by RT-qPCR analyses of tissue samples from groups of 20 patients with cisplatin-sensitive ovarian cancer and 7 patients with cisplatin-resistant ovarian cancer. Functional studies demonstrated that miR-509-3p inhibitor decreased cell response to cisplatin (CDDP) and promoted cell survival in SKOV3 ovarian cancer cells. Furthermore, we found gene expression level of Golgi phosphoprotein-3 (GOLPH3) and wntless Wnt ligand secretion mediator (WLS) were regulated by miR-509-3p. The direct bindings of miR-509-3p to GOLPH3 and WLS genes were confirmed by dual-luciferase reporter assay. And the negative correlation between their expression levels in SKOV3 cells was further verified with RT-qPCR. Altogether, our data provide preliminary evidence, supporting that targeting miR-509-3p might be a potential therapeutic strategy for patients with platinum-resistant ovarian cancer.
Collapse
|
109
|
Tomasik B, Chałubińska-Fendler J, Chowdhury D, Fendler W. Potential of serum microRNAs as biomarkers of radiation injury and tools for individualization of radiotherapy. Transl Res 2018; 201:71-83. [PMID: 30021695 DOI: 10.1016/j.trsl.2018.06.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 05/31/2018] [Accepted: 06/04/2018] [Indexed: 12/30/2022]
Abstract
Due to tremendous technological advances, radiation oncologists are now capable of personalized treatment plans and deliver the dose in a highly precise manner. However, a crucial challenge is how to escalate radiation doses to cancer cells while reducing damage to surrounding healthy tissues. This determines the probability of achieving therapeutic success whilst safeguarding patients from complications. The current dose constraints rely on observational data. Therefore, incidental toxicity observed in a minority of patients limits the admissible dose thresholds for the whole population, theoretically narrowing down the curative potential of radiotherapy. Future tools for measurements of individual's radiosensitivity before and during treatment would allow proper treatment personalization. Variation in tissue tolerance is at least partially genetically-determined and recent progress in the field of molecular biology raises the possibility that novel assays will allow to predict the response to ionizing radiation. Recently, microRNAs have garnered interest as stable biomarkers of tumor radiation response and normal-tissue toxicity. Preclinical studies in mice and nonhuman primates have shown that serum circulating microRNAs can be used to accurately distinguish pre- and postirradiation states and predict the biological impact of high-dose irradiation. First reports from human studies are also encouraging, however biology-driven precision radiation oncology, which tailors treatment to individual patient's needs, still remains to be translated into clinical studies. In this review, we summarize current knowledge about the potential of serum microRNAs as biodosimeters and biomarkers for radiation injury to lung and hematopoietic cells.
Collapse
Affiliation(s)
- Bartłomiej Tomasik
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland; Postgraduate School of Molecular Medicine, Warsaw Medical University, Warsaw, Poland
| | | | - Dipanjan Chowdhury
- Department of Radiation Oncology, Harvard Medical School, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.
| | - Wojciech Fendler
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland; Department of Radiation Oncology, Harvard Medical School, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.
| |
Collapse
|
110
|
Thapar R. Regulation of DNA Double-Strand Break Repair by Non-Coding RNAs. Molecules 2018; 23:molecules23112789. [PMID: 30373256 PMCID: PMC6278438 DOI: 10.3390/molecules23112789] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/25/2018] [Accepted: 10/26/2018] [Indexed: 01/12/2023] Open
Abstract
DNA double-strand breaks (DSBs) are deleterious lesions that are generated in response to ionizing radiation or replication fork collapse that can lead to genomic instability and cancer. Eukaryotes have evolved two major pathways, namely homologous recombination (HR) and non-homologous end joining (NHEJ) to repair DSBs. Whereas the roles of protein-DNA interactions in HR and NHEJ have been fairly well defined, the functions of small and long non-coding RNAs and RNA-DNA hybrids in the DNA damage response is just beginning to be elucidated. This review summarizes recent discoveries on the identification of non-coding RNAs and RNA-mediated regulation of DSB repair.
Collapse
Affiliation(s)
- Roopa Thapar
- Department of Molecular and Cellular Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
111
|
Walker SE, Spencer GE, Necakov A, Carlone RL. Identification and Characterization of microRNAs during Retinoic Acid-Induced Regeneration of a Molluscan Central Nervous System. Int J Mol Sci 2018; 19:E2741. [PMID: 30217012 PMCID: PMC6163488 DOI: 10.3390/ijms19092741] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/06/2018] [Accepted: 09/08/2018] [Indexed: 12/11/2022] Open
Abstract
Retinoic acid (RA) is the biologically active metabolite of vitamin A and has become a well-established factor that induces neurite outgrowth and regeneration in both vertebrates and invertebrates. However, the underlying regulatory mechanisms that may mediate RA-induced neurite sprouting remain unclear. In the past decade, microRNAs have emerged as important regulators of nervous system development and regeneration, and have been shown to contribute to processes such as neurite sprouting. However, few studies have demonstrated the role of miRNAs in RA-induced neurite sprouting. By miRNA sequencing analysis, we identify 482 miRNAs in the regenerating central nervous system (CNS) of the mollusc Lymnaeastagnalis, 219 of which represent potentially novel miRNAs. Of the remaining conserved miRNAs, 38 show a statistically significant up- or downregulation in regenerating CNS as a result of RA treatment. We further characterized the expression of one neuronally-enriched miRNA upregulated by RA, miR-124. We demonstrate, for the first time, that miR-124 is expressed within the cell bodies and neurites of regenerating motorneurons. Moreover, we identify miR-124 expression within the growth cones of cultured ciliary motorneurons (pedal A), whereas expression in the growth cones of another class of respiratory motorneurons (right parietal A) was absent in vitro. These findings support our hypothesis that miRNAs are important regulators of retinoic acid-induced neuronal outgrowth and regeneration in regeneration-competent species.
Collapse
Affiliation(s)
- Sarah E Walker
- Department of Biological Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada.
| | - Gaynor E Spencer
- Department of Biological Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada.
| | - Aleksandar Necakov
- Department of Biological Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada.
| | - Robert L Carlone
- Department of Biological Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada.
| |
Collapse
|
112
|
Molecular pathways involved in microRNA-mediated regulation of multidrug resistance. Mol Biol Rep 2018; 45:2913-2923. [DOI: 10.1007/s11033-018-4358-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 09/03/2018] [Indexed: 12/23/2022]
|
113
|
Plantamura I, Cosentino G, Cataldo A. MicroRNAs and DNA-Damaging Drugs in Breast Cancer: Strength in Numbers. Front Oncol 2018; 8:352. [PMID: 30234015 PMCID: PMC6129576 DOI: 10.3389/fonc.2018.00352] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 08/10/2018] [Indexed: 11/25/2022] Open
Abstract
MicroRNAs are a class of small non-coding regulatory RNAs playing key roles in cancer. Breast cancer is the most common female malignancy worldwide and is categorized into four molecular subtypes: luminal A and B, HER2+ and triple-negative breast cancer (TNBC). Despite the development of multiple targeted therapies for luminal and HER2+ breast tumors, TNBC lacks specific therapeutic approaches, thus they are treated mainly with radio- and chemotherapy. The effectiveness of these therapeutic regimens is based on their ability to induce DNA damage, which is differentially resolved and repaired by normal vs. cancer cells. Recently, drugs directly targeting DNA repair mechanisms, such as PARP inhibitors, have emerged as attractive candidates for the future molecular targeted-therapy in breast cancer. These compounds prevent cancer cells to appropriate repair DNA double strand breaks and induce a phenomenon called synthetic lethality, that results from the concurrent inhibition of PARP and the absence of functional BRCA genes which prompt cell death. MicroRNAs are relevant players in most of the biological processes including DNA damage repair mechanisms. Consistently, the downregulation of DNA repair genes by miRNAs have been probe to improve the therapeutic effect of genotoxic drugs. In this review, we discuss how microRNAs can sensitize cancer cells to DNA-damaging drugs, through the regulation of DNA repair genes, and examine the most recent findings on their possible use as a therapeutic tools of treatment response in breast cancer.
Collapse
Affiliation(s)
- Ilaria Plantamura
- Molecular Targeting Unit, Research Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giulia Cosentino
- Molecular Targeting Unit, Research Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessandra Cataldo
- Molecular Targeting Unit, Research Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
114
|
Genetic Variants in pre-miR-146a, pre-miR-499, pre-miR-125a, pre-miR-605, and pri-miR-182 Are Associated with Breast Cancer Susceptibility in a South American Population. Genes (Basel) 2018; 9:genes9090427. [PMID: 30135399 PMCID: PMC6162394 DOI: 10.3390/genes9090427] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 08/13/2018] [Accepted: 08/17/2018] [Indexed: 12/17/2022] Open
Abstract
Breast cancer (BC) is one of the most frequent tumors affecting women worldwide. microRNAs (miRNAs) single-nucleotide polymorphisms (SNPs) likely contribute to BC susceptibility. We evaluated the association of five SNPs with BC risk in non-carriers of the BRCA1/2-mutation from a South American population. The SNPs were genotyped in 440 Chilean BRCA1/2-negative BC cases and 1048 controls. Our data do not support an association between rs2910164:G>C or rs3746444:A>G and BC risk. The rs12975333:G>T is monomorphic in the Chilean population. The pre-miR-605 rs2043556-C allele was associated with a decreased risk of BC, both in patients with a strong family history of BC and in early-onset non-familial BC (Odds ratio (OR) = 0.5 [95% confidence interval (CI) 0.4⁻0.9] p = 0.006 and OR = 0.6 [95% CI 0.5⁻0.9] p = 0.02, respectively). The rs4541843-T allele is associated with increased risk of familial BC. This is the first association study on rs4541843 and BC risk. Previously, we showed that the TOX3-rs3803662:C>T was significantly associated with increased risk of familial BC. Given that TOX3 mRNA is a target of miR-182, and that both the TOX3 rs3803662-T and pri-miR-182 rs4541843-T alleles are associated with increased BC risk, we evaluated their combined effect. Risk of familial BC increased in a dose-dependent manner with the number of risk alleles (p-trend = 0.0005), indicating an additive effect.
Collapse
|
115
|
Oxidative stress-induced miRNAs modulate AKT signaling and promote cellular senescence in uterine leiomyoma. J Mol Med (Berl) 2018; 96:1095-1106. [PMID: 30097674 DOI: 10.1007/s00109-018-1682-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 07/12/2018] [Accepted: 08/06/2018] [Indexed: 02/08/2023]
Abstract
Uterine leiomyomas (ULM) grow under high oxidative stress due to a hypoxic microenvironment and defects in redox metabolism. AKT is one major pathway activated by reactive oxygen species (ROS) that maintains ULM growth and survival. We previously reported that AKT inactivated by AKT inhibitors can significantly induce cellular senescence in ULM cells. Since some miRNAs are induced by AKT inhibitors in an ROS-dependent manner, we proposed that these miRNAs may modulate AKT function and cellular senescence in ULM. We therefore established ex vivo models of a three-dimensional ULM spheroid culture system to study the role of miRNAs in cellular senescence. Four miRNAs, miR-29b, miR-181a, miR-182, and miR-200c, were found to induce cellular senescence in primary ULM and myometrium spheroid cultures when stably overexpressed. miR-181a and miR-182 were found to repress AKT3 and CCND2, respectively. Correspondingly, RNAi of AKT3 or CCND2 also induced cellular senescence and G0/G1 arrest. Thus, miR-181a and miR-182 may drive cellular senescence in ULM by repressing AKT3 and CCND2 activity, respectively. We further demonstrated that senescent ULM cells can be effectively removed by BH3 mimetic ABT263, which provides a new therapeutic venue for the treatment of ULM. Our findings suggest that miRNAs are potent modulators in regulating the ROS-AKT-cell cycle axis in uterine leiomyoma. KEY MESSAGES A subset of oxidative stress-induced miRNAs is involved in AKT signaling in uterine leiomyoma. Overexpression of miR-181a and miR-182 resulted in cellular senescence in leiomyoma through repression of AKT3 and CCND2, respectively. Silencing of AKT3 and CCND2 drives leiomyoma cell into senescence and cycle arrest. Application of our newly developed 3D leiomyoma spheroids can provide a quick and reliable ex vivo model for cytopathologic and functional analysis. BH3 mimetics can effectively reduce the viability of miRNA-mediated senescent cells in leiomyoma.
Collapse
|
116
|
Liu Q, Ma L, Jones T, Palomero L, Pujana MA, Martinez-Ruiz H, Ha PK, Murnane J, Cuartas I, Seoane J, Baumann M, Linge A, Barcellos-Hoff MH. Subjugation of TGFβ Signaling by Human Papilloma Virus in Head and Neck Squamous Cell Carcinoma Shifts DNA Repair from Homologous Recombination to Alternative End Joining. Clin Cancer Res 2018; 24:6001-6014. [PMID: 30087144 DOI: 10.1158/1078-0432.ccr-18-1346] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/15/2018] [Accepted: 08/02/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE Following cytotoxic therapy, 70% of patients with human papillomavirus (HPV)-positive oropharyngeal head and neck squamous cell carcinoma (HNSCC) are alive at 5 years compared with 30% of those with similar HPV-negative cancer. Loss of TGFβ signaling is a poorly studied consequence of HPV that could contribute to patient outcome by compromising DNA repair. EXPERIMENTAL DESIGN Human HNSCC cell lines (n = 9), patient-derived xenografts (n = 9), tissue microarray (n = 194), TCGA expression data (n = 279), and primary tumor specimens (n = 10) were used to define the relationship between TGFβ competency, response to DNA damage, and type of DNA repair. RESULTS Analysis of HNSCC specimens in situ and in vitro showed that HPV associated with loss of TGFβ signaling that increased response to radiation or cisplatin. TGFβ suppressed miR-182, which inhibited both BRCA1, necessary for homologous recombination repair (HRR), and FOXO3, required for ATM kinase activity. TGFβ signaling blockade by either HPV or inhibitors released miR182 control, compromised HRR and increased response to PARP inhibition. Antagonizing miR-182 rescued the HRR deficit in HPV-positive cells. Loss of TGFβ signaling unexpectedly increased repair by error prone, alternative end-joining (alt-EJ). CONCLUSIONS HPV-positive HNSCC cells are unresponsive to TGFβ. Abrogated TGFβ signaling compromises repair by HRR and increases reliance on alt-EJ, which provides a mechanistic basis for sensitivity to PARP inhibitors. The effect of HPV in HNSCC provides critical validation of TGFβ's role in DNA repair proficiency and further raises the translational potential of TGFβ inhibitors in cancer therapy.
Collapse
Affiliation(s)
- Qi Liu
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Lin Ma
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Trevor Jones
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Luis Palomero
- ProCURE, Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - Miquel Angel Pujana
- ProCURE, Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | | | - Patrick K Ha
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - John Murnane
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Isabel Cuartas
- Vall d Hebron Institute of Oncology (VHIO), Institució Catalana de Recerca i Estudis Avançats (ICREA), Universitat Autònoma de Barcelona, CIBERONC, Barcelona, Spain
| | - Joan Seoane
- Vall d Hebron Institute of Oncology (VHIO), Institució Catalana de Recerca i Estudis Avançats (ICREA), Universitat Autònoma de Barcelona, CIBERONC, Barcelona, Spain
| | - Michael Baumann
- German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, Germany; and Department of Radiotherapy and Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Annett Linge
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mary Helen Barcellos-Hoff
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California.
| |
Collapse
|
117
|
Kaina B, Izzotti A, Xu J, Christmann M, Pulliero A, Zhao X, Dobreanu M, Au WW. Inherent and toxicant-provoked reduction in DNA repair capacity: A key mechanism for personalized risk assessment, cancer prevention and intervention, and response to therapy. Int J Hyg Environ Health 2018; 221:993-1006. [PMID: 30041861 DOI: 10.1016/j.ijheh.2018.07.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 07/03/2018] [Accepted: 07/04/2018] [Indexed: 02/05/2023]
Abstract
Genomic investigations reveal novel evidence which indicates that genetic predisposition and inherent drug response are key factors for development of cancer and for poor response to therapy. However, mechanisms for these outcomes and interactions with environmental factors have not been well-characterized. Therefore, cancer risk, prevention, intervention and prognosis determinations have still mainly been based on population, rather than on individualized, evaluations. The objective of this review was to demonstrate that a key mechanism which contributes to the determination is inherent and/or toxicant-provoked reduction in DNA repair capacity. In addition, functional and quantitative determination of DNA repair capacity on an individual basis would dramatically change the evaluation and management of health problems from a population to a personalized basis. In this review, justifications for the scenario were delineated. Topics to be presented include assays for detection of functional DNA repair deficiency, mechanisms for DNA repair defects, toxicant-perturbed DNA repair capacity, epigenetic mechanisms (methylation and miRNA expression) for alteration of DNA repair function, and bioinformatics approach to analyze large amount of genomic data. Information from these topics has recently been and will be used for better understanding of cancer causation and of response to therapeutic interventions. Consequently, innovative genomic- and mechanism-based evidence can be increasingly used to develop more precise cancer risk assessment, and target-specific and personalized medicine.
Collapse
Affiliation(s)
| | - Alberto Izzotti
- University of Genoa, Genoa, Italy; IRCCS Policlinico San Martino Genoa, Italy
| | - Jianzhen Xu
- Shantou University Medical College, Shantou, China
| | | | | | - Xing Zhao
- Shantou University Medical College, Shantou, China
| | | | - William W Au
- Shantou University Medical College, Shantou, China; University of Medicine and Pharmacy, Tirgu Mures, Romania; University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
118
|
Nie J, Li CP, Li JH, Chen X, Zhong X. Analysis of non‑alcoholic fatty liver disease microRNA expression spectra in rat liver tissues. Mol Med Rep 2018; 18:2669-2680. [PMID: 30015905 PMCID: PMC6102666 DOI: 10.3892/mmr.2018.9268] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 06/14/2018] [Indexed: 12/14/2022] Open
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) has been increasing in recent years. Previous studies have suggested that micro (mi)RNAs may be involved in the pathogenesis of NAFLD. To investigate the role of miRNAs in rat NAFLD, a total of 16 male Sprague Dawley rats were randomly divided into a control group and a model group. Rats in the control group were fed a normal diet for 12 weeks, whereas the rats in the model group were fed a high‑fat and high‑sugar diet for 12 weeks. Following this, the animals were sacrificed and liver tissues were rapidly removed to investigate the severity of NAFLD. Blood samples were collected to investigate liver function, in addition to total cholesterol, total triglyceride and fasting plasma glucose levels. Total RNA from three fresh liver samples per experimental group was extracted for subsequent miRNA gene chip analysis using GeneChip miRNA 4.0 to investigate differentially expressed miRNAs, and miRNA expression was further verified via reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). Compared with the control group, the results revealed that there were 10 differentially expressed miRNAs in the model group, five of which were overexpressed and five of which were underexpressed compared with the control group. The results of the RT‑qPCR analysis revealed that miR‑182, miR‑29b‑3p and miR‑741‑3p were significantly overexpressed in the model group compared with the control group, which was largely consistent with the results of the microarray analysis. The results suggested that the differentially expressed microRNAs demonstrated in the present study may be involved in the pathogenesis of NAFLD; however, the mechanism underlying the differential expression of miRNAs in NAFLD requires further investigation.
Collapse
Affiliation(s)
- Jiao Nie
- Department of Gastroenterology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Chang-Ping Li
- Department of Gastroenterology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jue-Hong Li
- Graduate School, College of Medicine, Shanghai Jiao Tong University, Shanghai 200025, P.R. China
| | - Xia Chen
- Department of Gastroenterology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Xiaoling Zhong
- Department of Gastroenterology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
119
|
Novel miRNA biomarkers for genotoxicity screening in mouse. Toxicology 2018; 404-405:68-75. [PMID: 29777724 DOI: 10.1016/j.tox.2018.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 05/08/2018] [Accepted: 05/13/2018] [Indexed: 12/31/2022]
Abstract
The genotoxic potential of drugs is a serious problem, and its evaluation is one of the most critical processes of drug development. Although the comet assay of compound-exposed tissue is a frequently used genotoxicity test, its high false-positive rate is a major complication, and we consistently obtained false-positive results using the comet assay of mouse liver for nine hepatotoxic non-genotoxins (NGTXs). To identify novel genotoxin (GTX)-specific biomarkers, we screened the expression of 750 microRNAs (miRNAs) in the livers of mice treated with GTXs or NGTXs. Three miRNAs, miR-22-3p, miR-409-3p, and miR-543-3p, were significantly down-regulated in GTX-treated mouse liver. In contrast, these three miRNAs were significantly up-regulated in plasma. A discrimination model based on the expression levels of these biomarkers successfully identified GTXs and NGTXs. This novel biomarker expression-based discrimination model analysis using both liver and plasma is effective for detecting genotoxicity with high sensitivity and reliability to support drug development.
Collapse
|
120
|
Mandujano-Tinoco EA, García-Venzor A, Melendez-Zajgla J, Maldonado V. New emerging roles of microRNAs in breast cancer. Breast Cancer Res Treat 2018; 171:247-259. [PMID: 29948402 DOI: 10.1007/s10549-018-4850-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/03/2018] [Indexed: 01/12/2023]
Abstract
BACKGROUND MicroRNAs constitute a large family of non-coding RNAs, which actively participate in tumorigenesis by regulating a set of mRNAs of distinct signaling pathways. An altered expression of these molecules has been found in different tumorigenic processes of breast cancer, the most common type of cancer in the female population worldwide. PURPOSE The objective of this review is to discuss how miRNAs become master regulators in breast tumorigenesis. METHODS An integrative review of miRNAs and breast cancer literature from the last 5 years was done on PubMed. We summarize recent works showing that the defects on the biogenesis of miRNAs are associated with different breast cancer characteristics. Then, we show several examples that demonstrate the link between cellular processes regulated by miRNAs and the hallmarks of breast cancer. Finally, we examine the complexity in the regulation of these molecules as they are modulated by other non-coding RNAs and the clinical applications of miRNAs as they could serve as good diagnostic and classification tools. CONCLUSION The information presented in this review is important to encourage new directed studies that consider microRNAs as a good tool to improve the diagnostic and treatment alternatives in breast cancer.
Collapse
Affiliation(s)
- Edna Ayerim Mandujano-Tinoco
- Epigenetics Laboratory, Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, 14610, Mexico, CDMX, Mexico.,Laboratory of Connective Tissue, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra". Calz., México-Xochimilco 289, Arenal de Guadalupe, 14389, Mexico, CDMX, Mexico
| | - Alfredo García-Venzor
- Epigenetics Laboratory, Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, 14610, Mexico, CDMX, Mexico
| | - Jorge Melendez-Zajgla
- Functional Genomics Laboratory, Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, 14610, Mexico, CDMX, Mexico
| | - Vilma Maldonado
- Epigenetics Laboratory, Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, 14610, Mexico, CDMX, Mexico.
| |
Collapse
|
121
|
Duma N, Gast KC, Choong GM, Leon-Ferre RA, O'Sullivan CC. Where Do We Stand on the Integration of PARP Inhibitors for the Treatment of Breast Cancer? Curr Oncol Rep 2018; 20:63. [PMID: 29884921 DOI: 10.1007/s11912-018-0709-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW To provide an overview of the clinical development of poly(ADP-ribose) polymerase inhibitors (PARPi) in breast cancer to date and to review existing challenges and future research directions. RECENT FINDINGS We summarize the clinical development of PARPi in breast cancer from bench to bedside, and discuss the results of recent phase 3 trials in patients with metastatic breast cancer (MBC) and germline mutations in BRCA1/2 (gBRCAm). We will also provide an update regarding mechanisms of action and resistance to PARPi, and review clinical trials of PARPi as monotherapy or in combination regimens. PARPi are a novel treatment approach in persons with gBRCA1/2m-associated MBC. Going forward, the clinical applicability of these compounds outside the gBRCAm setting will be studied in greater detail. The identification of accurate predictive biomarkers of response is a research priority.
Collapse
Affiliation(s)
- Narjust Duma
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Kelly C Gast
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Grace M Choong
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | | | | |
Collapse
|
122
|
Javadi H, Lotfi AS, Hosseinkhani S, Mehrani H, Amani J, Soheili ZS, Hojati Z, Kamali M. The combinational effect of E6/E7 siRNA and anti-miR-182 on apoptosis induction in HPV16-positive cervical cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:727-736. [PMID: 29873516 DOI: 10.1080/21691401.2018.1468770] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In the present research, we assumed that reducing the amounts of E6 and E7 oncoproteins by a specific siRNA sequence and recovering p53 and RB proteins, along with the recovery of the FOXO1 protein by applying anti-miR-182, would increase apoptosis and reduce proliferation rate in cancer cells. The HPV16-positive CaSki cervical cancer cell line was used. 48 hours after transfection of siRNA for targeting E6 and E7 oncoproteins and anti-miR-182, expression of its cellular targets p53, p21 and FOXO1 was assessed by real-time PCR, western blot analysis and immunocytofluorescence staining. In all treatments, apoptosis rate and viability were evaluated using Annexin-V-FITC apoptosis detection kits and MTT assays, respectively. Among the designed siRNAs, E6-1 and E7-2 proved the most effective in reducing E6 and E7 expressions by increasing the apoptotic rates to 12.4% and 16%, respectively, after 48 hours. Also, using anti-miR-182 increased apoptotic rate to 12.7% 48 hours after transfection of cervical cancer cells. The combinational use of either E6-1 or E7-2 siRNAs with anti-miR-182 resulted in a rise in apoptosis to 19.3% and 26%, respectively, higher than those obtained from the individual application of either without anti-miR-182. The simultaneous use of siRNA E6-1 and siRNA E7-2 with cisplatin increased sensitivity to cisplatin and reduced the viability of the cancer cells as compared to the use of cisplatin alone. The simultaneous use of cisplatin and anti-miR-182 had no considerable effect on viability or apoptosis rate compared to cisplatin alone.
Collapse
Affiliation(s)
- Hamidreza Javadi
- a Nanobiotechnology Research Center , Baqiyatallah University of Medical Sciences , Tehran , Iran.,b Department of Molecular Medicine , Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology , Tehran , Iran
| | - Abbas Sahebghadam Lotfi
- c Department of Clinical Biochemistry, Faculty of Medicine , Tarbiat Modares University , Tehran , Iran
| | - Saman Hosseinkhani
- d Department of Biochemistry, Faculty of Basic Sciences , Tarbiat Modares University , Tehran , Iran
| | - Hossein Mehrani
- e Department of Biochemistry, Faculty of Science , Islamic Azad University Branch of Neyshabur , Neyshabur , Iran
| | - Jafar Amani
- f Applied Microbiology Research Center, System Biology and Poisonings Institute , Baqiyatallah University of Medical Sciences , Tehran , Iran
| | - Zahra Soheila Soheili
- b Department of Molecular Medicine , Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology , Tehran , Iran
| | - Zahra Hojati
- b Department of Molecular Medicine , Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology , Tehran , Iran
| | - Mehdi Kamali
- a Nanobiotechnology Research Center , Baqiyatallah University of Medical Sciences , Tehran , Iran
| |
Collapse
|
123
|
Abstract
Terminally differentiated cells have a reduced capacity to repair double-stranded breaks (DSB) in DNA, however, the underlying molecular mechanism remains unclear. Here, we show that miR-22 is upregulated during postmitotic differentiation of human breast MCF-7 cells, hematopoietic HL60 and K562 cells. Increased expression of miR-22 in differentiated cells was associated with decreased expression of MDC1, a protein that plays a key role in the response to DSBs. This downregulation of MDC1 was accompanied by reduced DSB repair, impaired recruitment of the protein to the site of DNA damage following IR. Conversely, inhibiting miR-22 enhanced MDC1 protein levels, recovered MDC1 foci, fully rescued DSB repair in terminally differentiated cells. Moreover, MDC1 levels, IR-induced MDC1 foci, and the efficiency of DSB repair were fully rescued by siRNA-mediated knockdown of c-Fos in differentiated cells. These findings indicate that the c-Fos/miR-22/MDC1 axis plays a relevant role in DNA repair in terminally differentiated cells, which may facilitate our understanding of molecular mechanism underlying the downregulating DNA repair in differentiated cells.
Collapse
|
124
|
Liu X, Li H, Wu G, Cui S. miR-182 promotes cell proliferation and invasion by inhibiting APC in melanoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:1900-1908. [PMID: 31938296 PMCID: PMC6958208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 02/14/2018] [Indexed: 06/10/2023]
Abstract
OBJECTIVE Common treatment methods have shown a lack of therapeutic effect in melanoma, a type of malignant tumor. The pathogenesis of melanoma is not yet fully clear, therefore, search for a new treatment strategy is urgent. Recent studies have demonstrated that miR-182 is remarkably over-expressed in human melanoma. Our study aimed to explore the underlying mechanism of miR-182 on melanoma. METHODS In this study, the expression level of miR-182 was detected using RT-PCR in melanoma and adjacent tissues as well as in HEM-m, A375, A2058, and WM35 cell lines. Functions of miR-182 were investigated by using CCK-8 on cell proliferation, apoptosis, invasion, and cell cycle on A375 cells. Moreover, expression levels of Frz, Dsh, β-catenin, APC, Axin, GSK-3β, and CK1 were detected by Western blotting after knockdown and overexpression of miR-182. Overexpression of miR-182 and knockdown of APC was used to demonstrate regulated functions in melanoma. RESULTS Expression levels of miR-182 were significantly upregulated in melanoma tissues and cell lines. Overexpression of miR-182 promoted cell proliferation, migration, and invasion while inhibiting cell apoptosis and cell cycle in S phase. Overexpression of miR-182 upregulated expression levels of β-catenin and APC. Overexpression of miR-182 and knockdown of APC inhibited proliferation of melanoma cells and tumors. CONCLUSION Overexpression of miR-182 promotes cell proliferation and invasion by targeting to APC in melanoma. The pathogenesis of miR-182 and APC might provide therapeutic targets for treatment of melanoma in the molecular level.
Collapse
Affiliation(s)
- Xilin Liu
- Department of Hand Surgery, China-Japan Union Hospital of Jilin UniversityChangchun, Jilin Province, China
| | - Hong Li
- Department of Rehabilitation, China-Japan Union Hospital of Jilin UniversityChangchun, Jilin Province, China
| | - Guangzhi Wu
- Department of Hand Surgery, China-Japan Union Hospital of Jilin UniversityChangchun, Jilin Province, China
| | - Shusen Cui
- Department of Hand Surgery, China-Japan Union Hospital of Jilin UniversityChangchun, Jilin Province, China
| |
Collapse
|
125
|
Ji C, Xu Q, Guo L, Wang X, Ren Y, Zhang H, Zhu W, Ming Z, Yuan Y, Ren X, Song J, Cheng Y, Yang J, Zhang Y. eEF-2 Kinase-targeted miR-449b confers radiation sensitivity to cancer cells. Cancer Lett 2018; 418:64-74. [DOI: 10.1016/j.canlet.2018.01.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 12/27/2017] [Accepted: 01/08/2018] [Indexed: 10/18/2022]
|
126
|
Zoni E, Karkampouna S, Thalmann GN, Kruithof-de Julio M, Spahn M. Emerging aspects of microRNA interaction with TMPRSS2-ERG and endocrine therapy. Mol Cell Endocrinol 2018; 462:9-16. [PMID: 28189568 DOI: 10.1016/j.mce.2017.02.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 12/22/2016] [Accepted: 02/07/2017] [Indexed: 11/22/2022]
Abstract
Prostate cancer (PCa) is the most common malignancy detected in males and the second most common cause of cancer death in western countries. The development of the prostate gland, is finely regulated by androgens which modulate also its growth and function. Importantly, androgens exert a major role in PCa formation and progression and one of the hypothesized mechanism proposed has been linked to the chromosomal rearrangement of the androgen regulated gene TMPRSS2 with ERG. Androgens have been therefore used as main target for therapies in the past. However, despite the development of endocrine therapies (e.g. androgen ablation), when PCa progress, tumors become resistant to this therapeutic castration and patients develop incurable metastases. A strategy to better understand how patients respond to therapy, in order to achieve a better patient stratification, consists in monitoring the levels of small noncoding RNAs (microRNAs). microRNAs are a class of small molecules that regulate protein abundance and their application as biomarkers to monitor disease progression has been intensely studied in the last years. In this review, we highlight the interactions between microRNAs and endocrine-related aspects of PCa in tissues. We focus on the modulation of TMPRSS2-ERG and Glucocorticoid Receptor (GR) by microRNAs and detail the influence of steroidal hormonal therapies on microRNAs expression.
Collapse
Affiliation(s)
- Eugenio Zoni
- Urology Research Laboratory, Department of Urology, University of Bern, Bern, Switzerland; Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Sofia Karkampouna
- Urology Research Laboratory, Department of Urology, University of Bern, Bern, Switzerland; Department of Clinical Research, University of Bern, Bern, Switzerland
| | - George N Thalmann
- Urology Research Laboratory, Department of Urology, University of Bern, Bern, Switzerland; Department of Clinical Research, University of Bern, Bern, Switzerland; Department of Urology, Bern University Hospital, Bern, Switzerland
| | - Marianna Kruithof-de Julio
- Urology Research Laboratory, Department of Urology, University of Bern, Bern, Switzerland; Department of Clinical Research, University of Bern, Bern, Switzerland; Urology Research Laboratory, Department of Urology, Leiden University Medical Center, Leiden, The Netherlands
| | - Martin Spahn
- Urology Research Laboratory, Department of Urology, University of Bern, Bern, Switzerland; Department of Urology, Bern University Hospital, Bern, Switzerland.
| |
Collapse
|
127
|
Sengodan SK, K H S, Nadhan R, Srinivas P. Regulation of epithelial to mesenchymal transition by BRCA1 in breast cancer. Crit Rev Oncol Hematol 2018; 123:74-82. [PMID: 29482782 DOI: 10.1016/j.critrevonc.2018.01.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/06/2017] [Accepted: 01/17/2018] [Indexed: 12/13/2022] Open
Abstract
Reports till its discovery has proven multiple facets of Breast Cancer type 1 susceptibility gene (BRCA1) from nucleus to cytoplasm; from DNA repair to drug resistance; from Homologous Recombination (HR) to Ubiquitination; from breast to brain; from cancer to HIV and many of the roles are still unexplored. One of the recent attractions of BRCA1 is its role in regulating breast cancer metastasis though the exact mechanism is poorly understood. In this review, we will discuss the molecular interactions between BRCA1 and the key molecules of Epithelial to Mesenchymal Transition (EMT) associated with metastasis, its associated drug resistance and the possible treatment strategy for BRCA1 mutated breast cancer.
Collapse
Affiliation(s)
- Satheesh Kumar Sengodan
- Cancer Research Program 5, Rajiv Gandhi Centre for Biotechnology, Thycaud P O, Poojappura, Thiruvananthapuram 695 014, Kerala, India
| | - Sreelatha K H
- Cancer Research Program 5, Rajiv Gandhi Centre for Biotechnology, Thycaud P O, Poojappura, Thiruvananthapuram 695 014, Kerala, India
| | - Revathy Nadhan
- Cancer Research Program 5, Rajiv Gandhi Centre for Biotechnology, Thycaud P O, Poojappura, Thiruvananthapuram 695 014, Kerala, India
| | - Priya Srinivas
- Cancer Research Program 5, Rajiv Gandhi Centre for Biotechnology, Thycaud P O, Poojappura, Thiruvananthapuram 695 014, Kerala, India.
| |
Collapse
|
128
|
Matamala N, Vargas MT, González-Cámpora R, Arias JI, Menéndez P, Andrés-León E, Yanowsky K, Llaneza-Folgueras A, Miñambres R, Martínez-Delgado B, Benítez J. MicroRNA deregulation in triple negative breast cancer reveals a role of miR-498 in regulating BRCA1 expression. Oncotarget 2018; 7:20068-79. [PMID: 26933805 PMCID: PMC4991439 DOI: 10.18632/oncotarget.7705] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 02/16/2016] [Indexed: 12/21/2022] Open
Abstract
Emerging evidence suggests that BRCA1 pathway contributes to the behavior of sporadic triple negative breast cancer (TNBC), but little is known about the mechanisms underlying this association. Considering the central role that microRNAs (miRNAs) play in gene expression regulation, the aim of this study was to identify miRNAs specifically deregulated in TNBC and investigate their involvement in BRCA1 regulation. Using locked nucleic acid (LNA)-based microarrays, expression levels of 1919 miRNAs were measured in paraffin-embedded tissues from 122 breast tumors and 11 healthy breast tissue samples. Differential miRNA expression was explored among the main subtypes of breast cancer, and 105 miRNAs were identified as specific for triple negative tumors. In silico prediction revealed that miR-498 and miR-187-5p target BRCA1, and these results were confirmed by luciferase reporter assay. While miR-187-5p was found overexpressed in a luminal B cell line, miR-498 was highly expressed in a triple negative cell line, Hs578T, and its expression was negatively correlated with the levels of BRCA1. We functionally demonstrated that miR-498 inhibits BRCA1 in breast cancer cell lines, and showed that inhibition of miR-498 led to reduced proliferation in the triple negative cell line Hs578T. Our results indicate that miR-498 regulates BRCA1 expression in breast cancer and its overexpression could contribute to the pathogenesis of sporadic TNBC via BRCA1 downregulation.
Collapse
Affiliation(s)
- Nerea Matamala
- Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,Current address: Molecular Genetics Unit, Research Institute of Rare Diseases (IIER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Maria Teresa Vargas
- Pathology Service, Hospital Virgen de la Macarena, Sevilla, Spain.,Current address: Hematology Service, Hospital Virgen de la Macarena, Sevilla, Spain
| | | | | | | | - Eduardo Andrés-León
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,Current address: Bioinformatics Unit, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas (IPBLN-CSIC), Granada, Spain
| | - Kira Yanowsky
- Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | | | - Beatriz Martínez-Delgado
- Molecular Genetics Unit, Research Institute of Rare Diseases (IIER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Javier Benítez
- Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,Network on Rare Diseases (CIBERER), Madrid, Spain
| |
Collapse
|
129
|
Sundaravinayagam D, Kim HR, Wu T, Kim HH, Lee HS, Jun S, Cha JH, Kee Y, You HJ, Lee JH. miR146a-mediated targeting of FANCM during inflammation compromises genome integrity. Oncotarget 2018; 7:45976-45994. [PMID: 27351285 PMCID: PMC5216775 DOI: 10.18632/oncotarget.10275] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 06/03/2016] [Indexed: 02/07/2023] Open
Abstract
Inflammation is a potent inducer of tumorigenesis. Increased DNA damage or loss of genome integrity is thought to be one of the mechanisms linking inflammation and cancer development. It has been suggested that NF-κB-induced microRNA-146 (miR146a) may be a mediator of the inflammatory response. Based on our initial observation that miR146a overexpression strongly increases DNA damage, we investigated its potential role as a modulator of DNA repair. Here, we demonstrate that FANCM, a component in the Fanconi Anemia pathway, is a novel target of miR146a. miR146a suppressed FANCM expression by directly binding to the 3′ untranslated region of the gene. miR146a-induced downregulation of FANCM was associated with inhibition of FANCD2 monoubiquitination, reduced DNA homologous recombination repair and checkpoint response, failed recovery from replication stress, and increased cellular sensitivity to cisplatin. These phenotypes were recapitulated when miR146a expression was induced by overexpressing the NF-κB subunit p65/RelA or Helicobacter pylori infection in a human gastric cell line; the phenotypes were effectively reversed with an anti-miR146a antagomir. These results suggest that undesired inflammation events caused by a pathogen or over-induction of miR146a can impair genome integrity via suppression of FANCM.
Collapse
Affiliation(s)
- Devakumar Sundaravinayagam
- Laboratory of Genomic Instability and Cancer Therapeutics, Cancer Mutation Research Center, Chosun University School of Medicine, Gwangju, Republic of Korea.,Department of Pharmacology, Chosun University School of Medicine, Gwangju, Republic of Korea
| | - Hye Rim Kim
- Laboratory of Genomic Instability and Cancer Therapeutics, Cancer Mutation Research Center, Chosun University School of Medicine, Gwangju, Republic of Korea.,Department of Cellular and Molecular Medicine, Chosun University School of Medicine, Gwangju, Republic of Korea
| | - TingTing Wu
- Laboratory of Genomic Instability and Cancer Therapeutics, Cancer Mutation Research Center, Chosun University School of Medicine, Gwangju, Republic of Korea.,Department of Cellular and Molecular Medicine, Chosun University School of Medicine, Gwangju, Republic of Korea
| | - Hyun Hee Kim
- Laboratory of Genomic Instability and Cancer Therapeutics, Cancer Mutation Research Center, Chosun University School of Medicine, Gwangju, Republic of Korea.,Department of Cellular and Molecular Medicine, Chosun University School of Medicine, Gwangju, Republic of Korea
| | - Hyun-Seo Lee
- Laboratory of Genomic Instability and Cancer Therapeutics, Cancer Mutation Research Center, Chosun University School of Medicine, Gwangju, Republic of Korea.,Department of Pharmacology, Chosun University School of Medicine, Gwangju, Republic of Korea
| | - Semo Jun
- Laboratory of Genomic Instability and Cancer Therapeutics, Cancer Mutation Research Center, Chosun University School of Medicine, Gwangju, Republic of Korea.,Department of Pharmacology, Chosun University School of Medicine, Gwangju, Republic of Korea
| | - Jeong-Heon Cha
- Department of Oral Biology, Department of Applied Life Science, The Graduate School, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Younghoon Kee
- Department of Cell Biology, Microbiology, and Molecular Biology, College of Arts and Sciences, University of South Florida, Tampa, Florida, United States of America
| | - Ho Jin You
- Laboratory of Genomic Instability and Cancer Therapeutics, Cancer Mutation Research Center, Chosun University School of Medicine, Gwangju, Republic of Korea.,Department of Pharmacology, Chosun University School of Medicine, Gwangju, Republic of Korea
| | - Jung-Hee Lee
- Laboratory of Genomic Instability and Cancer Therapeutics, Cancer Mutation Research Center, Chosun University School of Medicine, Gwangju, Republic of Korea.,Department of Cellular and Molecular Medicine, Chosun University School of Medicine, Gwangju, Republic of Korea
| |
Collapse
|
130
|
Sharma B, Preet Kaur R, Raut S, Munshi A. BRCA1 mutation spectrum, functions, and therapeutic strategies: The story so far. Curr Probl Cancer 2018; 42:189-207. [PMID: 29452958 DOI: 10.1016/j.currproblcancer.2018.01.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 12/19/2017] [Accepted: 01/04/2018] [Indexed: 02/07/2023]
Abstract
BRCA1 gene mutations account for about 25-28% of hereditary Breast Cancer as BRCA1 is included in the category of high penetrance genes. Except for few commonmutations, there is a heterogenous spectrum of BRCA1 mutations in various ethnic groups. 185AGdel and 5382ins Care the most common BRCA1 alterations (founder mutations) which have been identified in most of the population. This review has been compiled with an aim to consolidate the information on genetic variants reported in BRCA1 found in various ethnic groups, their functional implications if known; involvement of BRCA1 in various cellular pathways/processes and potential BRCA1 targeted therapies. The pathological variations of BRCA1 vary among different ethical groups. A systematic search in PubMed and Google scholar for the literature on BRCA1 gene was carried out to figure out structure and function of BRCA1 gene. BRCA1 is a large protein having 1863 amino acids with multiple functional domains and interacts with multiple proteins to carry out various crucial cellular processes. BRCA1 plays a major role in maintaining genome integrity, transcription regulation, chromatin remodeling, cell cycle checkpoint control, DNA damage repair, chromosomal segregation, and apoptosis. Studies investigating the phenotypic response of mutant BRCA1 protein and comparing it to wildtype BRCA1 protein are clinically important as they are involved in homologous recombination and other repair mechanisms. These studies may help in developing more targetted therapies, detecting novel interacting partners, identification of new signaling pathways that BRCA1 is a part of or downstream target genes that BRCA1 affects.
Collapse
Affiliation(s)
- Babita Sharma
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, Punjab, India
| | - Raman Preet Kaur
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, Punjab, India
| | - Sonali Raut
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, Punjab, India
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, Punjab, India.
| |
Collapse
|
131
|
Cyriac SL, Karakasis K, Oza AM. Rucaparib for the treatment of ovarian cancer. Expert Opin Orphan Drugs 2017. [DOI: 10.1080/21678707.2018.1419129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Sunu Lazar Cyriac
- Gynecology and Drug Development Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Katherine Karakasis
- Research Program Development, The Princess Margaret, Drug Development Program, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Amit M. Oza
- Division of Medical Oncology and Hematology, Clinical Cancer Research Unit and Bras Drug Development Program, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
132
|
Yu J, Tian X, Chang J, Liu P, Zhang R. RUNX3 inhibits the proliferation and metastasis of gastric cancer through regulating miR-182/HOXA9. Biomed Pharmacother 2017; 96:782-791. [DOI: 10.1016/j.biopha.2017.08.144] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 08/25/2017] [Accepted: 08/28/2017] [Indexed: 01/11/2023] Open
|
133
|
He M, Lin Y, Tang Y, Liu Y, Zhou W, Li C, Sun G, Guo M. miR-638 suppresses DNA damage repair by targeting SMC1A expression in terminally differentiated cells. Aging (Albany NY) 2017; 8:1442-56. [PMID: 27405111 PMCID: PMC4993341 DOI: 10.18632/aging.100998] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 06/28/2016] [Indexed: 12/27/2022]
Abstract
The reduction of DNA damage repair capacity in terminally differentiated cells may be involved in sensitivity to cancer chemotherapy drugs; however, the underlying molecular mechanism is still not fully understood. Herein, we evaluated the role of miR-638 in the regulation of DNA damage repair in terminally differentiated cells. Our results show that miR-638 expression was up-regulated during cellular terminal differentiation and involved in mediating DNA damage repair processes. Results from a luciferase reporting experiment show that structural maintenance of chromosomes (SMC)1A was a potential target of miR-638; this was verified by western blot assays during cell differentiation and DNA damage induction. Overexpression of miR-638 enhanced the sensitivity of cancer cells to cisplatin, thus reducing cell viability in response to chemotherapy drug treatment. Furthermore, miR-638 overexpression affected DNA damage repair processes by interfering with the recruitment of the DNA damage repair-related protein, γH2AX, to DNA break sites. These findings indicate that miR-638 might act as a sensitizer in cancer chemotherapy and accompany chemotherapy drugs to enhance chemotherapeutic efficacy and to improve the chance of recovery from cancer.
Collapse
Affiliation(s)
- Mingyang He
- College of Life Sciences, Wuhan University, 430072 Wuhan, P. R. China
| | - Yi Lin
- College of Life Sciences, Wuhan University, 430072 Wuhan, P. R. China
| | - Yunlan Tang
- College of Life Sciences, Wuhan University, 430072 Wuhan, P. R. China
| | - Yi Liu
- College of Life Sciences, Wuhan University, 430072 Wuhan, P. R. China
| | - Weiwei Zhou
- College of Life Sciences, Wuhan University, 430072 Wuhan, P. R. China
| | - Chuang Li
- College of Life Sciences, Wuhan University, 430072 Wuhan, P. R. China
| | - Guihong Sun
- School of Basic Medical Sciences, Wuhan University, 430071 Wuhan, P.R. China
| | - Mingxiong Guo
- College of Life Sciences, Wuhan University, 430072 Wuhan, P. R. China
| |
Collapse
|
134
|
The expression of miRNAs is associated with tumour genome instability and predicts the outcome of ovarian cancer patients treated with platinum agents. Sci Rep 2017; 7:14736. [PMID: 29116111 PMCID: PMC5677022 DOI: 10.1038/s41598-017-12259-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 09/04/2017] [Indexed: 12/19/2022] Open
Abstract
miRNAs, a class of short but stable noncoding RNA molecules, have been revealed to play important roles in the DNA damage response (DDR). However, their functions in cancer genome instability and the consequent clinical effect as the response to chemotherapy have not been fully elucidated. In this study, we utilized multidimensional TCGA data and the known miRNAs involved in DDR to identify a miRNA-regulatory network that responds to DNA damage. Additionally, based on the expression of ten miRNAs in this network, we developed a 10-miRNA-score that predicts defects in the homologous recombination (HR) pathway and genome instability in ovarian cancer. Importantly, consistent with the association between HR defects and improved response to chemotherapeutic agents, the 10-miRNA-score predicts the outcome of ovarian cancer patients treated with platinum agents, with a surprisingly better performance than the indexes of DNA damage. Therefore, our study demonstrates the implication of miRNA expression on cancer genome instability and provides an alternative method to identify DDR defects in patients who show the best effect with platinum drug treatment.
Collapse
|
135
|
Bitler BG, Watson ZL, Wheeler LJ, Behbakht K. PARP inhibitors: Clinical utility and possibilities of overcoming resistance. Gynecol Oncol 2017; 147:695-704. [PMID: 29037806 DOI: 10.1016/j.ygyno.2017.10.003] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/01/2017] [Accepted: 10/03/2017] [Indexed: 12/21/2022]
Abstract
PARP inhibitors represent a major breakthrough in ovarian cancer care. Almost half of all ovarian cancers have deficiencies in the homologous recombination (HR) DNA repair pathway, namely BRCA1/2 mutations. Given the limited therapeutic options for recurrent ovarian cancer patients there has been a significant effort to develop novel therapies to exploit DNA repair deficiencies. In 2005 and 2006, inhibiting PARP enzymes was first observed to be highly effective against cancers with HR deficiencies. PARP inhibitors are being utilized in the clinic to manage recurrent ovarian cancers that display defects in the HR repair pathway. However, PARP inhibitors also show significant clinical benefit in patients without HR deficiencies. There are currently three FDA-approved PARP inhibitors for recurrent ovarian cancer and an additional two PARP inhibitors being evaluated in late stage clinical trials. Given the expanding clinical use of PARP inhibitors and the high likelihood of acquired resistance, there is a significant need for clinical strategies to manage PARP inhibitor resistant disease. This review will examine PARP inhibitors in the context of: indications and toxicities, novel biomarkers to predict response, targeted-therapy resistance, and potential approaches to manage resistant disease.
Collapse
Affiliation(s)
- Benjamin G Bitler
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, The University of Colorado, Aurora, CO, United States.
| | - Zachary L Watson
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, The University of Colorado, Aurora, CO, United States
| | - Lindsay J Wheeler
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, The University of Colorado, Aurora, CO, United States; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, The University of Colorado, Aurora, CO, United States
| | - Kian Behbakht
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, The University of Colorado, Aurora, CO, United States; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, The University of Colorado, Aurora, CO, United States.
| |
Collapse
|
136
|
Strumidło A, Skiba S, Scott RJ, Lubiński J. The potential role of miRNAs in therapy of breast and ovarian cancers associated with BRCA1 mutation. Hered Cancer Clin Pract 2017; 15:15. [PMID: 29021870 PMCID: PMC5622493 DOI: 10.1186/s13053-017-0076-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 09/17/2017] [Indexed: 11/29/2022] Open
Abstract
Germline variants within BRCA1 or BRCA2 genes account for approximately 25% of familial aggregations of breast-ovarian cancers. Low or no expression of BRCA1 in breast and ovarian cancers is associated with a good clinical response to treatment with platinum therapies and PARP1 inhibitors. Recent studies demonstrated that microRNAs - small non-coding RNAs, involved in the control of gene expression, can decrease BRCA1 expression by targeting the 3’UTR region of the gene. This article reviews reported relationships between various miRNAs, such as miRNA-9, miRNA-146a, miRNA-182 miRNA-218, miRNA-638 and the response to cytostatic drugs, mainly to platins and PARP1 inhbitors, for the treatment of breast and ovarian cancer associated with BRCA1 mutations.
Collapse
Affiliation(s)
| | - Sylwia Skiba
- Pomeranian University of Medicine, Szczecin, Poland
| | - Rodney J Scott
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia.,Division of Genetics, Hunter Area Pathology Service, Newcastle, Australia
| | - Jan Lubiński
- Department of Genetics and Pathology, Pomeranian University of Medicine, Szczecin, Poland
| |
Collapse
|
137
|
Zhang X, Ma G, Liu J, Zhang Y. MicroRNA-182 promotes proliferation and metastasis by targeting FOXF2 in triple-negative breast cancer. Oncol Lett 2017; 14:4805-4811. [PMID: 29085483 PMCID: PMC5649577 DOI: 10.3892/ol.2017.6778] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 08/11/2017] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancer (TNBC), the most aggressive subtype of breast cancer (BC), is characterized as high proliferation, young age and poor prognosis. MicroRNA-182 (miR-182) was reported to have oncogenic potential in many cancers. We aimed to elucidate pathobiological effects of miR-182 expression by targeting forkhead-box F2 (FOXF2) in TNBC. In this study, we explored the functional role of miR-182 expression in TNBC. Quantitative real-time PCR (qRT-PCR) was applied to evaluate the expression of miR-182 in cell lines and tissues. A series of in vitro and in vivo assays were performed in the MCF-7 and MDA-MB-231 cell lines with miR-182 overexpression. Luciferase reporter assays and western blot analysis were used to identify FOXF2 as the direct and functional target of miR-182. In TNBC tissues and cell lines, we found that miR-182 was significantly upregulated. Transwell assay showed that re-expression of miR-182 increased cell migration and invasion abilities and MTT assay showed that it promoted cell growth in vitro. In vivo assay, re-expression of miR-182 significantly increase tumor volume and enhanced instant metastasis in the lungs of mice. Besides, FOXF2 was identified as a direct and functional target of miR-182. These results indicated that miR-182 plays an important role in the initiation and progression of TNBC by targeting FOXF2 and the miR-182/FOXF2 axis may present a new therapeutic strategy for TNBC in the future.
Collapse
Affiliation(s)
- Xingzeng Zhang
- Department of General Surgery, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Genshun Ma
- Department of General Surgery, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Jianchao Liu
- Department of General Surgery, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Yajun Zhang
- Department of General Surgery, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| |
Collapse
|
138
|
Dambal S, Baumann B, McCray T, Williams L, Richards Z, Deaton R, Prins GS, Nonn L. The miR-183 family cluster alters zinc homeostasis in benign prostate cells, organoids and prostate cancer xenografts. Sci Rep 2017; 7:7704. [PMID: 28794468 PMCID: PMC5550464 DOI: 10.1038/s41598-017-07979-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/03/2017] [Indexed: 12/18/2022] Open
Abstract
The miR-183 cluster, which is comprised of paralogous miRs-183, -96 and -182, is overexpressed in many cancers, including prostate adenocarcinoma (PCa). Prior studies showed that overexpression of individual pre-miRs-182, -96 and -183 in prostate cells decreased zinc import, which is a characteristic feature of PCa tumours. Zinc is concentrated in healthy prostate 10-fold higher than any other tissue, and an >80% decrease in zinc is observed in PCa specimens. Here, we studied the effect of overexpression of the entire 4.8 kb miR-183 family cluster, including the intergenic region which contains highly conserved genomic regions, in prostate cells. This resulted in overexpression of mature miR-183 family miRs at levels that mimic cancer-related changes. Overexpression of the miR-183 cluster reduced zinc transporter and intracellular zinc levels in benign prostate cells, PCa xenografts and fresh prostate epithelial organoids. Microarray analysis of miR-183 family cluster overexpression in prostate cells showed an enrichment for cancer-related pathways including adhesion, migration and wound healing. An active secondary transcription start site was identified within the intergenic region of the miR-183 cluster, which may regulate expression of miR-182. Taken together, this study shows that physiologically relevant expression of the miR-183 family regulates zinc levels and carcinogenic pathways in prostate cells.
Collapse
Affiliation(s)
- Shweta Dambal
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Bethany Baumann
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Tara McCray
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - LaTanya Williams
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Zachary Richards
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Ryan Deaton
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Gail S Prins
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, 60612, USA.,Department of Urology, University of Illinois at Chicago, Chicago, IL, 60612, USA.,University of Illinois Cancer Center, Chicago, IL, 60612, USA
| | - Larisa Nonn
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, 60612, USA. .,University of Illinois Cancer Center, Chicago, IL, 60612, USA.
| |
Collapse
|
139
|
Markopoulos GS, Roupakia E, Tokamani M, Chavdoula E, Hatziapostolou M, Polytarchou C, Marcu KB, Papavassiliou AG, Sandaltzopoulos R, Kolettas E. A step-by-step microRNA guide to cancer development and metastasis. Cell Oncol (Dordr) 2017; 40:303-339. [PMID: 28748501 DOI: 10.1007/s13402-017-0341-9] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2017] [Indexed: 01/17/2023] Open
|
140
|
Delia D, Mizutani S. The DNA damage response pathway in normal hematopoiesis and malignancies. Int J Hematol 2017; 106:328-334. [PMID: 28707218 DOI: 10.1007/s12185-017-2300-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 07/05/2017] [Indexed: 11/29/2022]
Abstract
In mammalian cells, the DNA damage response (DDR) prevents the replication and propagation of DNA errors to the next generation, thus maintaining genomic stability. At the heart of the DDR are the related signaling kinases ATM, ATR, and DNA-PK, which regulate DNA repair and associated events such as cell cycle checkpoints, chromatin remodeling, transcription, and ultimately apoptosis. Several findings highlight the occurrence of DDR in hemopoietic stem cells (HSCs), and persistence of DNA lesions in these cells promotes their functional decline and accumulation of leukemogenic mutations. Besides favoring tumor formation and progression, molecular defects that directly or indirectly inactivate certain DDR pathways can provide a therapeutic opportunity, since a reduced ability to repair DNA lesions renders hemopoietic malignancies vulnerable to genotoxic drugs acting also through synthetic lethal interactions. Here, we discuss the essential role of DDR in HSC maintenance and protection against leukemogenesis, and how acquired DDR dysfunctions or pharmacological agents that block this pathway can be effectively exploited for the treatment of various hematopoietic malignancies.
Collapse
Affiliation(s)
- Domenico Delia
- Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy.
| | - Shuki Mizutani
- Kawasaki North Center for Childhood Developmental Disorder/Tokyo Medical and Dental University, 5-26-1 Katahira, Aso-ku, Kawasaki, 215-0003, Japan
| |
Collapse
|
141
|
Zhu L, Sturgis EM, Zhang H, Lu Z, Tao Y, Wei Q, Li G. Genetic variants in microRNA-binding sites of DNA repair genes as predictors of recurrence in patients with squamous cell carcinoma of the oropharynx. Int J Cancer 2017. [PMID: 28646528 DOI: 10.1002/ijc.30849] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The incidence of squamous cell carcinoma of the oropharynx (SCCOP) continues to rise because of increasing rates of human papillomavirus (HPV) infection. Inherited polymorphisms in DNA repair pathways may influence the risk of SCCOP development and the prognosis of SCCOP. We sought to determine whether polymorphisms in microRNA (miRNA)-binding sites within 3'-untranslated regions (3'UTRs) of genes in DNA repair pathways modulate the risk of SCCOP recurrence. We evaluated the associations between nine such polymorphisms and SCCOP recurrence in 1,008 patients with incident SCCOP using the log-rank test and multivariable Cox models. In an analysis of all the patients, patients with variant genotypes of BRCA1 rs12516 and RAD51 rs7180135 had better disease-free survival (log-rank, p = 0.0002 and p = 0.0003, respectively) and lower risk of SCCOP recurrence (hazard ratio [HR], 0.5, 95% confidence interval [CI], 0.2-0.8, and HR, 0.5, 95% CI, 0.3-0.9, respectively) than patients with common homozygous genotypes of the two polymorphisms after multivariable adjustment. Moreover, in tumor HPV16-positive patients, patients with variant genotypes of the same two polymorphisms also had better disease-free survival (log-rank, p = 0.004 and p = 0.003, respectively) and lower recurrence risk (HR, 0.2, 95% CI, 0.1-0.6, and HR, 0.2, 95% CI, 0.0-0.7, respectively) than patients with common homozygous genotypes of the two polymorphisms. No such significant associations were found for other polymorphisms. These findings support significant roles of BRCA1 rs12516 and RAD51 rs7180135 in modifying the risk of recurrence of SCCOP, particularly HPV16-positive SCCOP. However, these results must be validated in larger studies.
Collapse
Affiliation(s)
- Lijun Zhu
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Oral and Maxillofacial Surgery, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Erich M Sturgis
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hua Zhang
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Otorhinolaryngology-Head and Neck Surgery, Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Zhongming Lu
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Otolaryngology-Head and Neck Surgery, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China
| | - Ye Tao
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Otolaryngology & head and neck Surgery, 2nd affiliated hospital of Anhui Medical University, Hefei, China
| | - Qingyi Wei
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX.,Duke Cancer Institute, Duke University Medical Center, Durham, NC
| | - Guojun Li
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
142
|
Stefansson OA, Hermanowicz S, van der Horst J, Hilmarsdottir H, Staszczak Z, Jonasson JG, Tryggvadottir L, Gudjonsson T, Sigurdsson S. CpG promoter methylation of the ALKBH3 alkylation repair gene in breast cancer. BMC Cancer 2017; 17:469. [PMID: 28679371 PMCID: PMC5498885 DOI: 10.1186/s12885-017-3453-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 06/26/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND DNA repair of alkylation damage is defective in various cancers. This occurs through somatically acquired inactivation of the MGMT gene in various cancer types, including breast cancers. In addition to MGMT, the two E. coli AlkB homologs ALKBH2 and ALKBH3 have also been linked to direct reversal of alkylation damage. However, it is currently unknown whether ALKBH2 or ALKBH3 are found inactivated in cancer. METHODS Methylome datasets (GSE52865, GSE20713, GSE69914), available through Omnibus, were used to determine whether ALKBH2 or ALKBH3 are found inactivated by CpG promoter methylation. TCGA dataset enabled us to then assess the impact of CpG promoter methylation on mRNA expression for both ALKBH2 and ALKBH3. DNA methylation analysis for the ALKBH3 promoter region was carried out by pyrosequencing (PyroMark Q24) in 265 primary breast tumours and 30 proximal normal breast tissue samples along with 8 breast-derived cell lines. ALKBH3 mRNA and protein expression were analysed in cell lines using RT-PCR and Western blotting, respectively. DNA alkylation damage assay was carried out in cell lines based on immunofluorescence and confocal imaging. Data on clinical parameters and survival outcomes in patients were obtained and assessed in relation to ALKBH3 promoter methylation. RESULTS The ALKBH3 gene, but not ALKBH2, undergoes CpG promoter methylation and transcriptional silencing in breast cancer. We developed a quantitative alkylation DNA damage assay based on immunofluorescence and confocal imaging revealing higher levels of alkylation damage in association with epigenetic inactivation of the ALKBH3 gene (P = 0.029). In our cohort of 265 primary breast cancer, we found 72 cases showing aberrantly high CpG promoter methylation over the ALKBH3 promoter (27%; 72 out of 265). We further show that increasingly higher degree of ALKBH3 promoter methylation is associated with reduced breast-cancer specific survival times in patients. In this analysis, ALKBH3 promoter methylation at >20% CpG methylation was found to be statistically significantly associated with reduced survival (HR = 2.3; P = 0.012). By thresholding at the clinically relevant CpG methylation level (>20%), we find the incidence of ALKBH3 promoter methylation to be 5% (13 out of 265). CONCLUSIONS ALKBH3 is a novel addition to the catalogue of DNA repair genes found inactivated in breast cancer. Our results underscore a link between defective alkylation repair and breast cancer which, additionally, is found in association with poor disease outcome.
Collapse
Affiliation(s)
- Olafur Andri Stefansson
- Cancer Research Laboratory, Biomedical Center, Vatnsmyrarvegur 16 (4th floor), 101 Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16 (4th floor), 101 Reykjavik, Iceland
| | - Stefan Hermanowicz
- Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16 (4th floor), 101 Reykjavik, Iceland
- Department of Biochemistry and Molecular Biology, Biomedical Center, Vatnsmyrarvegur 16 (5th floor), 101 Reykjavik, Iceland
| | - Jasper van der Horst
- Cancer Research Laboratory, Biomedical Center, Vatnsmyrarvegur 16 (4th floor), 101 Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16 (4th floor), 101 Reykjavik, Iceland
| | - Holmfridur Hilmarsdottir
- Cancer Research Laboratory, Biomedical Center, Vatnsmyrarvegur 16 (4th floor), 101 Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16 (4th floor), 101 Reykjavik, Iceland
| | - Zuzanna Staszczak
- Cancer Research Laboratory, Biomedical Center, Vatnsmyrarvegur 16 (4th floor), 101 Reykjavik, Iceland
| | - Jon Gunnlaugur Jonasson
- Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16 (4th floor), 101 Reykjavik, Iceland
- Icelandic Cancer Registry, Skogarhlid 8, Reykjavik, Iceland
- Department of Pathology, Landspitali University Hospital, Reykjavik, Iceland
| | - Laufey Tryggvadottir
- Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16 (4th floor), 101 Reykjavik, Iceland
- Icelandic Cancer Registry, Skogarhlid 8, Reykjavik, Iceland
| | - Thorkell Gudjonsson
- Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16 (4th floor), 101 Reykjavik, Iceland
- Department of Biochemistry and Molecular Biology, Biomedical Center, Vatnsmyrarvegur 16 (5th floor), 101 Reykjavik, Iceland
| | - Stefan Sigurdsson
- Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16 (4th floor), 101 Reykjavik, Iceland
- Department of Biochemistry and Molecular Biology, Biomedical Center, Vatnsmyrarvegur 16 (5th floor), 101 Reykjavik, Iceland
| |
Collapse
|
143
|
Petrovic N, Davidovic R, Bajic V, Obradovic M, Isenovic RE. MicroRNA in breast cancer: The association with BRCA1/2. Cancer Biomark 2017; 19:119-128. [DOI: 10.3233/cbm-160319] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
144
|
Spolverini A, Fuchs G, Bublik DR, Oren M. let-7b and let-7c microRNAs promote histone H2B ubiquitylation and inhibit cell migration by targeting multiple components of the H2B deubiquitylation machinery. Oncogene 2017; 36:5819-5828. [PMID: 28604753 PMCID: PMC5600258 DOI: 10.1038/onc.2017.187] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 04/18/2017] [Accepted: 05/10/2017] [Indexed: 02/07/2023]
Abstract
Monoubiquitylation of histone H2B (H2Bub1) is catalyzed mainly by the RNF20/RNF40 complex and erased by multiple deubiquitylating enzymes (DUBs). H2Bub1 influences many aspects of chromatin function, including transcription regulation and DNA repair. Cancer cells often display reduced levels of H2Bub1, and this reduction may contribute to cancer progression. The let-7 family of microRNAs comprises multiple members with reported tumor suppressive features, whose expression is frequently downregulated in cancer. We now report that let-7b and let-7c can positively regulate cellular H2Bub1 levels. Overexpression of let-7b and let-7c in a variety of non-transformed and cancer-derived cell lines results in H2Bub1 elevation. The positive effect of let-7b and let-7c on H2Bub1 levels is achieved through targeting of multiple mRNAs, coding for distinct components of the H2B deubiquitylation machinery. Specifically, let-7b and let-7c bind directly and inhibit the mRNAs encoding the DUBs USP42 and USP44, and also the mRNA encoding the adapter protein ATXN7L3, which is part of the DUB module of the SAGA complex. RNF20 knockdown strongly reduces H2Bub1 levels and increases the migration of non-transformed mammary epithelial cells and breast cancer-derived cells. Remarkably, overexpression of let-7b, which partly counteracts the effect of RNF20 knockdown on H2Bub1 levels, also reverses the pro-migratory effect of RNF20 knockdown. Likewise, ATXN7L3 knockdown also increases H2Bub1 levels and reduces cell migration, and this anti-migratory effect is abolished by simultaneous knockdown of RNF20. Together, our findings uncover a novel function of let-7 microRNAs as regulators of H2B ubiquitylation, suggesting an additional mechanism whereby these microRNAs can exert their tumor suppressive effects.
Collapse
Affiliation(s)
- A Spolverini
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - G Fuchs
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - D R Bublik
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - M Oren
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
145
|
Gasparri ML, Casorelli A, Bardhi E, Besharat AR, Savone D, Ruscito I, Farooqi AA, Papadia A, Mueller MD, Ferretti E, Benedetti Panici P. Beyond circulating microRNA biomarkers: Urinary microRNAs in ovarian and breast cancer. Tumour Biol 2017; 39:1010428317695525. [PMID: 28459207 DOI: 10.1177/1010428317695525] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most common malignancy in women worldwide, and ovarian cancer is the most lethal gynecological malignancy. Women carrying a BRCA1/2 mutation have a very high lifetime risk of developing breast and ovarian cancer. The only effective risk-reducing strategy in BRCA-mutated women is a prophylactic surgery with bilateral mastectomy and bilateral salpingo-oophorectomy. However, many women are reluctant to undergo these prophylactic surgeries due to a consequent mutilated body perception, unfulfilled family planning, and precocious menopause. In these patients, an effective screening strategy is available only for breast cancer, but it only consists in close radiological exams with a significant burden for the health system and a significant distress to the patients. No biomarkers have been shown to effectively detect breast and ovarian cancer at an early stage. MicroRNAs (miRNAs) are key regulatory molecules operating in a post-transcriptional regulation of gene expression. Aberrant expression of miRNAs has been documented in several pathological conditions, including solid tumors, suggesting their involvement in tumorigenesis. miRNAs can be detected in blood and urine and could be used as biomarkers in solid tumors. Encouraging results are emerging in gynecological malignancy as well, and suggest a different pattern of expression of miRNAs in biological fluids of breast and ovarian cancer patients as compared to healthy control. Aim of this study is to highlight the role of the urinary miRNAs which are specifically associated with cancer and to investigate their role in early diagnosis and in determining the prognosis in breast and ovarian cancer.
Collapse
Affiliation(s)
- Maria Luisa Gasparri
- 1 Department of Gynecology, Obstetrics and Urology, Sapienza University of Rome, Rome, Italy.,2 Department of Obstetrics and Gynecology, University Hospital of Berne, University of Berne, Berne, Switzerland
| | - Assunta Casorelli
- 1 Department of Gynecology, Obstetrics and Urology, Sapienza University of Rome, Rome, Italy
| | - Erlisa Bardhi
- 1 Department of Gynecology, Obstetrics and Urology, Sapienza University of Rome, Rome, Italy
| | - Aris Raad Besharat
- 1 Department of Gynecology, Obstetrics and Urology, Sapienza University of Rome, Rome, Italy
| | - Delia Savone
- 1 Department of Gynecology, Obstetrics and Urology, Sapienza University of Rome, Rome, Italy
| | - Ilary Ruscito
- 1 Department of Gynecology, Obstetrics and Urology, Sapienza University of Rome, Rome, Italy
| | - Ammad Ahmad Farooqi
- 3 Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, Pakistan
| | - Andrea Papadia
- 2 Department of Obstetrics and Gynecology, University Hospital of Berne, University of Berne, Berne, Switzerland
| | - Michael David Mueller
- 2 Department of Obstetrics and Gynecology, University Hospital of Berne, University of Berne, Berne, Switzerland
| | - Elisabetta Ferretti
- 4 Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.,5 Neuromed Institute, Pozzilli, Italy
| | | |
Collapse
|
146
|
El Bairi K, Amrani M, Kandhro AH, Afqir S. Prediction of therapy response in ovarian cancer: Where are we now? Crit Rev Clin Lab Sci 2017; 54:233-266. [PMID: 28443762 DOI: 10.1080/10408363.2017.1313190] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 03/27/2017] [Indexed: 02/08/2023]
Abstract
Therapy resistance is a major challenge in the management of ovarian cancer (OC). Advances in detection and new technology validation have led to the emergence of biomarkers that can predict responses to available therapies. It is important to identify predictive biomarkers to select resistant and sensitive patients in order to reduce important toxicities, to reduce costs and to increase survival. The discovery of predictive and prognostic biomarkers for monitoring therapy is a developing field and provides promising perspectives in the era of personalized medicine. This review article will discuss the biology of OC with a focus on targetable pathways; current therapies; mechanisms of resistance; predictive biomarkers for chemotherapy, antiangiogenic and DNA-targeted therapies, and optimal cytoreductive surgery; and the emergence of liquid biopsy using recent studies from the Medline database and ClinicalTrials.gov.
Collapse
Affiliation(s)
- Khalid El Bairi
- a Faculty of Medicine and Pharmacy , Mohamed Ist University , Oujda , Morocco
| | - Mariam Amrani
- b Equipe de Recherche ONCOGYMA, Faculty of Medicine, Pathology Department , National Institute of Oncology, Université Mohamed V , Rabat , Morocco
| | - Abdul Hafeez Kandhro
- c Department of Biochemistry , Healthcare Molecular and Diagnostic Laboratory , Hyderabad , Pakistan
| | - Said Afqir
- d Department of Medical Oncology , Mohamed VI University Hospital , Oujda , Morocco
| |
Collapse
|
147
|
Zhang Y, Dai D, Chang Y, Li Y, Zhang M, Zhou G, Peng Z, Zeng C. Cryopreservation of boar sperm induces differential microRNAs expression. Cryobiology 2017; 76:24-33. [DOI: 10.1016/j.cryobiol.2017.04.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 04/28/2017] [Accepted: 04/28/2017] [Indexed: 01/29/2023]
|
148
|
Rajawat J, Shukla N, Mishra DP. Therapeutic Targeting of Poly(ADP-Ribose) Polymerase-1 (PARP1) in Cancer: Current Developments, Therapeutic Strategies, and Future Opportunities. Med Res Rev 2017; 37:1461-1491. [PMID: 28510338 DOI: 10.1002/med.21442] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/31/2017] [Accepted: 02/16/2017] [Indexed: 12/16/2022]
Abstract
Poly(ADP-ribose) polymerase-1 (PARP-1) plays a central role in numerous cellular processes including DNA repair, replication, and transcription. PARP interacts directly, indirectly or via PARylation with various oncogenic proteins and regulates several transcription factors thereby modulating carcinogenesis. Therapeutic inhibition of PARP is therefore perceived as a promising anticancer strategy and a number of PARP inhibitors (PARPi) are currently under development and clinical evaluation. PARPi inhibit the DNA repair pathway and thus form the concept of synthetic lethality in cancer therapeutics. Preclinical and clinical studies have shown the potential of PARPi as chemopotentiator, radiosensitizer, or as adjuvant therapeutic agents. Recent studies have shown that PARP-1 could be either oncogenic or tumor suppressive in different cancers. PARP inhibitor resistance is also a growing concern in the clinical setting. Recently, changes in the levels of PARP-1 activity or expression in cancer patients have provided the basis for consideration of PARP-1 regulatory proteins as potential biomarkers. This review focuses on the current developments related to the role of PARP in cancer progression, therapeutic strategies targeting PARP-associated oncogenic signaling, and future opportunities in use of PARPi in anticancer therapeutics.
Collapse
Affiliation(s)
- Jyotika Rajawat
- Cell Death Research Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India
| | - Nidhi Shukla
- Cell Death Research Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India
| | - Durga Prasad Mishra
- Cell Death Research Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India
| |
Collapse
|
149
|
Nieborowska-Skorska M, Sullivan K, Dasgupta Y, Podszywalow-Bartnicka P, Hoser G, Maifrede S, Martinez E, Di Marcantonio D, Bolton-Gillespie E, Cramer-Morales K, Lee J, Li M, Slupianek A, Gritsyuk D, Cerny-Reiterer S, Seferynska I, Stoklosa T, Bullinger L, Zhao H, Gorbunova V, Piwocka K, Valent P, Civin CI, Muschen M, Dick JE, Wang JC, Bhatia S, Bhatia R, Eppert K, Minden MD, Sykes SM, Skorski T. Gene expression and mutation-guided synthetic lethality eradicates proliferating and quiescent leukemia cells. J Clin Invest 2017; 127:2392-2406. [PMID: 28481221 DOI: 10.1172/jci90825] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 03/07/2017] [Indexed: 02/02/2023] Open
Abstract
Quiescent and proliferating leukemia cells accumulate highly lethal DNA double-strand breaks that are repaired by 2 major mechanisms: BRCA-dependent homologous recombination and DNA-dependent protein kinase-mediated (DNA-PK-mediated) nonhomologous end-joining, whereas DNA repair pathways mediated by poly(ADP)ribose polymerase 1 (PARP1) serve as backups. Here we have designed a personalized medicine approach called gene expression and mutation analysis (GEMA) to identify BRCA- and DNA-PK-deficient leukemias either directly, using reverse transcription-quantitative PCR, microarrays, and flow cytometry, or indirectly, by the presence of oncogenes such as BCR-ABL1. DNA-PK-deficient quiescent leukemia cells and BRCA/DNA-PK-deficient proliferating leukemia cells were sensitive to PARP1 inhibitors that were administered alone or in combination with current antileukemic drugs. In conclusion, GEMA-guided targeting of PARP1 resulted in dual cellular synthetic lethality in quiescent and proliferating immature leukemia cells, and is thus a potential approach to eradicate leukemia stem and progenitor cells that are responsible for initiation and manifestation of the disease. Further, an analysis of The Cancer Genome Atlas database indicated that this personalized medicine approach could also be applied to treat numerous solid tumors from individual patients.
Collapse
Affiliation(s)
- Margaret Nieborowska-Skorska
- Temple University Lewis Katz School of Medicine, Department of Microbiology and Immunology and Fels Institute for Cancer Research & Molecular Biology, Philadelphia, Pennsylvania, USA
| | - Katherine Sullivan
- Temple University Lewis Katz School of Medicine, Department of Microbiology and Immunology and Fels Institute for Cancer Research & Molecular Biology, Philadelphia, Pennsylvania, USA
| | - Yashodhara Dasgupta
- Temple University Lewis Katz School of Medicine, Department of Microbiology and Immunology and Fels Institute for Cancer Research & Molecular Biology, Philadelphia, Pennsylvania, USA
| | | | - Grazyna Hoser
- The Center of Postgraduate Medical Education, Laboratory of Flow Cytometry, Warsaw, Poland
| | - Silvia Maifrede
- Temple University Lewis Katz School of Medicine, Department of Microbiology and Immunology and Fels Institute for Cancer Research & Molecular Biology, Philadelphia, Pennsylvania, USA
| | - Esteban Martinez
- Research Institute of Fox Chase Cancer Center, Immune Cell Development and Host Defense, Philadelphia, Pennsylvania, USA
| | - Daniela Di Marcantonio
- Research Institute of Fox Chase Cancer Center, Immune Cell Development and Host Defense, Philadelphia, Pennsylvania, USA
| | - Elisabeth Bolton-Gillespie
- Temple University Lewis Katz School of Medicine, Department of Microbiology and Immunology and Fels Institute for Cancer Research & Molecular Biology, Philadelphia, Pennsylvania, USA
| | - Kimberly Cramer-Morales
- Temple University Lewis Katz School of Medicine, Department of Microbiology and Immunology and Fels Institute for Cancer Research & Molecular Biology, Philadelphia, Pennsylvania, USA
| | - Jaewong Lee
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Min Li
- Department of Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California, USA
| | - Artur Slupianek
- Temple University Lewis Katz School of Medicine, Department of Microbiology and Immunology and Fels Institute for Cancer Research & Molecular Biology, Philadelphia, Pennsylvania, USA
| | - Daniel Gritsyuk
- Temple University Lewis Katz School of Medicine, Department of Microbiology and Immunology and Fels Institute for Cancer Research & Molecular Biology, Philadelphia, Pennsylvania, USA
| | - Sabine Cerny-Reiterer
- Medical University of Vienna and Ludwig Boltzmann-Cluster Oncology, and Department of Internal Medicine I, Division of Hematology and Hemostaseology, Vienna, Austria
| | - Ilona Seferynska
- Department of Hematology, Institute of Hematology and Blood Transfusion, Warsaw, Poland
| | - Tomasz Stoklosa
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Lars Bullinger
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Huaqing Zhao
- Temple University Lewis Katz School of Medicine, Department of Clinical Sciences, Philadelphia, Pennsylvania, USA
| | - Vera Gorbunova
- Department of Biology, University of Rochester, Rochester, New York, USA
| | | | - Peter Valent
- Medical University of Vienna and Ludwig Boltzmann-Cluster Oncology, and Department of Internal Medicine I, Division of Hematology and Hemostaseology, Vienna, Austria
| | - Curt I Civin
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Markus Muschen
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Jean Cy Wang
- Princess Margaret Cancer Centre, UHN, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Division of Medical Oncology and Hematology, UHN, Toronto, Ontario, Canada
| | | | - Ravi Bhatia
- Division of Hematology-Oncology, Department of Medicine, University of Alabama Birmingham, Birmingham, Alabama, USA
| | - Kolja Eppert
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Mark D Minden
- Princess Margaret Cancer Center, Ontario Cancer Institute, Toronto, Ontario, Canada
| | - Stephen M Sykes
- Research Institute of Fox Chase Cancer Center, Immune Cell Development and Host Defense, Philadelphia, Pennsylvania, USA
| | - Tomasz Skorski
- Temple University Lewis Katz School of Medicine, Department of Microbiology and Immunology and Fels Institute for Cancer Research & Molecular Biology, Philadelphia, Pennsylvania, USA
| |
Collapse
|
150
|
MicroRNA-182-5p Regulates Nerve Injury–induced Nociceptive Hypersensitivity by Targeting Ephrin Type-b Receptor 1. Anesthesiology 2017; 126:967-977. [PMID: 28248712 DOI: 10.1097/aln.0000000000001588] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Abstract
Background
The authors and others have previously shown that the up-regulation of spinal ephrin type-b receptor 1 plays an essential role in the pathologic process of nerve injury–induced nociceptive hypersensitivity, but the regulatory mechanism remains unclear.
Methods
Radiant heat and von Frey filaments were applied to assess nociceptive behaviors. Real-time quantitative polymerase chain reaction, Western blotting, fluorescence in situ hybridization, immunofluorescence, immunohistochemistry, dual-luciferase reporter gene assays, recombinant lentivirus, and small interfering RNA were used to characterize the likely mechanisms.
Results
Periphery nerve injury induced by chronic constriction injury of the sciatic nerve significantly reduced spinal microRNA-182-5p (miR-182-5p) expression levels, which were inversely correlated with spinal ephrin type-b receptor 1 expression (R2 = 0.90; P < 0.05; n = 8). The overexpression of miR-182-5p in the spinal cord prevented and reversed the nociceptive behaviors induced by sciatic nerve injury, accompanied by a decreased expression of spinal ephrin type-b receptor 1 (recombinant lentiviruses containing pre-microRNA-182: 1.91 ± 0.34 vs. 1.24 ± 0.31, n = 4; miR-182-5p mimic: 2.90 ± 0.48 vs. 1.51 ± 0.25, n = 4). In contrast, the down-regulation of spinal miR-182-5p facilitated the nociceptive behaviors induced by sciatic nerve injury and increased the expression of spinal ephrin type-b receptor 1 (1.0 ± 0.26 vs. 1.74 ± 0.31, n = 4). Moreover, the down-regulation of miR-182-5p and up-regulation of ephrin type-b receptor 1 caused by sciatic nerve injury were mediated by the N-methyl-d-aspartate receptor.
Conclusions
Collectively, our findings reveal that the spinal ephrin type-b receptor 1 is regulated by miR-182-5p in nerve injury–induced nociceptive hypersensitivity.
Collapse
|