1
|
Ellson I, Martorell-Marugán J, Carmona-Sáez P, Ramos-Mejia V. MiRNA expression as outcome predictor in pediatric AML: systematic evaluation of a new model. NPJ Genom Med 2024; 9:40. [PMID: 39107334 PMCID: PMC11303725 DOI: 10.1038/s41525-024-00424-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 06/24/2024] [Indexed: 08/10/2024] Open
Abstract
Accurately predicting patient outcomes is essential for optimizing treatment and improving outcomes in pediatric acute myeloid leukemia (AML). In recent years, microRNAs have emerged as a promising prognostic marker, with a growing body of evidence supporting their potential predictive value. We systematically reviewed all previous studies that have analyzed the expression of microRNAs as predictors of survival in pediatric AML and found 16 microRNAs and 4 microRNA signatures previously proposed as predictors of survival. We then used a public access cohort of 1414 pediatric AML patients from the TARGET project to develop a new predictive model using penalized lasso Cox regression based on microRNA expression. Here we propose a new score based on a 37-microRNA signature that is associated with AML and is able to predict survival more accurately than previous microRNA-based methods.
Collapse
Affiliation(s)
- Ivan Ellson
- GENYO, Centre for Genomics and Oncological Research Pfizer, University of Granada, Andalusian Regional Government, PTS, 18016, Granada, Spain
| | - Jordi Martorell-Marugán
- GENYO, Centre for Genomics and Oncological Research Pfizer, University of Granada, Andalusian Regional Government, PTS, 18016, Granada, Spain
- Fundación para la Investigación Biosanitaria de Andalucía Oriental-Alejandro Otero (FIBAO), 18012, Granada, Spain
| | - Pedro Carmona-Sáez
- GENYO, Centre for Genomics and Oncological Research Pfizer, University of Granada, Andalusian Regional Government, PTS, 18016, Granada, Spain.
- Department of Statistics, University of Granada, 18071, Granada, Spain.
| | - Verónica Ramos-Mejia
- GENYO, Centre for Genomics and Oncological Research Pfizer, University of Granada, Andalusian Regional Government, PTS, 18016, Granada, Spain.
- Department of Cell Biology, Faculty of Sciences, University of Granada, 18071, Granada, Spain.
| |
Collapse
|
2
|
Bhadra M, Sachan M. An overview of challenges associated with exosomal miRNA isolation toward liquid biopsy-based ovarian cancer detection. Heliyon 2024; 10:e30328. [PMID: 38707279 PMCID: PMC11068823 DOI: 10.1016/j.heliyon.2024.e30328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/07/2024] Open
Abstract
As one of the deadliest gynaecological cancers, ovarian cancer has been on the list. With lesser-known symptoms and lack of an accurate detection method, it is still difficult to catch it early. In terms of both the diagnosis and outlook for cancer, liquid biopsy has come a long way with significant advancements. Exosomes, extracellular components commonly shed by cancerous cells, are nucleic acid-rich particles floating in almost all body fluids and hold enormous promise, leading to minimallyinvasive molecular diagnostics. They have been shown as potential biomarkers in liquid biopsy, being implicated in tumour growth and metastasis. In order to address the drawbacks of ovarian cancer tumor heterogeneity, a liquid biopsy-based approach is being investigated by detecting cell-free nucleic acids, particularly non-coding RNAs, having the advantage of being less invasive and more prominent in nature. microRNAs are known to actively contribute to cancer development and their existence inside exosomes has also been made quite apparent which can be leveraged to diagnose and treat the disease. Extraction of miRNAs and exosomes is an arduous execution, and while other approaches have been investigated, none have produced results that are as encouraging due to limits in time commitment, yield, and, most significantly, damage to the exosomal structure resulting discrepancies in miRNA-based expression profiling for disease diagnosis. We have briefly outlined and reviewed the difficulties with exosome isolation techniques and the need for their standardization. The several widely used procedures and their drawbacks in terms of the exosomal purity they may produce have also been outlined.
Collapse
Affiliation(s)
- Mridula Bhadra
- Department of Biotechnology, Motilal Nehru National Institute of Technology-Allahabad, Prayagraj, 211004, Uttar Pradesh, India
| | - Manisha Sachan
- Department of Biotechnology, Motilal Nehru National Institute of Technology-Allahabad, Prayagraj, 211004, Uttar Pradesh, India
| |
Collapse
|
3
|
Yin PY, Wang RW, Jing R, Li X, Ma JH, Li KM, Wang H. Research progress on molecular biomarkers of acute myeloid leukemia. Front Oncol 2023; 13:1078556. [PMID: 36824144 PMCID: PMC9941555 DOI: 10.3389/fonc.2023.1078556] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/26/2023] [Indexed: 02/10/2023] Open
Abstract
Acute myeloid leukemia (AML) is the most common type of adult acute leukemia. The pathophysiology of the disease has been studied intensively at the cellular and molecular levels. At present, cytogenetic markers are an important basis for the early diagnosis, prognostic stratification and treatment of AML. However, with the emergence of new technologies, the detection of other molecular markers, such as gene mutations and epigenetic changes, began to play important roles in evaluating the occurrence and development of diseases. Recent evidence shows that identifying new AML biomarkers contributes to a better understanding of the molecular mechanism of the disease and is essential for AML screening, diagnosis, prognosis monitoring, and individualized treatment response. In this review, we summarized the promising AML biomarkers from four aspects, which contributing to a better understanding of the disease. Of course, it must be soberly aware that we have not listed all biomarkers of AML. Anyway, the biomarkers we mentioned are representative. For example, mutations in TP53, FLT3, and ASXL1 suggest poor prognosis, low remission rate, short survival period, and often require allogeneic hematopoietic stem cell transplantation. The CEBPA double mutation, NPM1 and CBF mutation suggest that the prognosis is good, the remission rate is high, the survival period is long, and the effect of chemotherapy or autotherapy is good. As for other mutations mentioned in the article, they usually predict a moderate prognosis. All in all, we hope it could provide a reference for the precise diagnosis and treatment of AML.
Collapse
Affiliation(s)
- Pei-Yuan Yin
- Hematology Department, Yantai Affiliated Hospital, Binzhou Medical University, Yantai, Shandong, China,Department of Blood Supply, Yantai Center Blood Station, Yantai, Shandong, China
| | - Rui-Wen Wang
- Department of Anesthesiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Rui Jing
- Hematology Department, Yantai Affiliated Hospital, Binzhou Medical University, Yantai, Shandong, China
| | - Xing Li
- Department of Blood Supply, Yantai Center Blood Station, Yantai, Shandong, China
| | - Jing-Hua Ma
- Department of Science and Education, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong, China,*Correspondence: Hua Wang, ; Kai-Min Li, ; Jing-Hua Ma,
| | - Kai-Min Li
- Hematology Department, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China,*Correspondence: Hua Wang, ; Kai-Min Li, ; Jing-Hua Ma,
| | - Hua Wang
- Hematology Department, Yantai Affiliated Hospital, Binzhou Medical University, Yantai, Shandong, China,*Correspondence: Hua Wang, ; Kai-Min Li, ; Jing-Hua Ma,
| |
Collapse
|
4
|
Lim HC, Soneji S, Pálmason R, Lenhoff S, Laurell T, Scheding S. Development of acoustically isolated extracellular plasma vesicles for biomarker discovery in allogeneic hematopoietic stem cell transplantation. Biomark Res 2021; 9:6. [PMID: 33468257 PMCID: PMC7814576 DOI: 10.1186/s40364-020-00259-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 12/29/2020] [Indexed: 12/29/2022] Open
Abstract
Background Infection and graft-versus-host disease (GvHD) are the major causes for mortality and morbidity of allogeneic hematopoietic stem cell transplantation (allo-HSCT). Plasma-derived extracellular vesicles (EVs) contain disease-related proteins, DNAs and RNAs, and have recently been suggested as potential biomarker candidates for transplantation complications. However, EV isolation from small plasma volumes in clinical biomarker studies using conventional methods is challenging. We therefore investigated if EVs isolated by novel automated acoustic trapping could be developed as potential biomarkers for allo-HSCT complications by performing a clinical proof-of-principle study. Results Plasma samples were collected from twenty consecutive patients with high-risk/relapsed hematologic malignancies undergoing allo-HSCT before transplantation and post-transplant up to 12 weeks. EVs were isolated from small plasma sample volumes (150 μl) by an automated, acoustofluidic-based particle trapping device, which utilizes a local λ/2 ultrasonic standing wave in a borosilicate glass capillary to capture plasma EVs among pre-seeded polystyrene microbeads through sound scatter interactions. We found that EVs could be reliably isolated from all plasma samples (n = 173) and that EV numbers increased more than 2-fold in the majority of patients after transplantation. Also, sufficient quantities of RNA for downstream microRNA (miRNA) analysis were obtained from all samples and EV miRNA profiles were found to differ from whole plasma profiles. As a proof of principle, expression of platelet-specific miR-142-3p in EVs was shown to correlate with platelet count kinetics after transplantation as expected. Importantly, we identified plasma EV miRNAs that were consistently positively correlated with infection and GvHD, respectively, as well as miRNAs that were consistently negatively correlated with these complications. Conclusions This study demonstrates that acoustic enrichment of EVs in a clinical biomarker study setting is feasible and that downstream analysis of acoustically-enriched EVs presents a promising tool for biomarker development in allo-HSCT. Certainly, these findings warrant further exploration in larger studies, which will have significant implications not only for biomarker studies in transplantation but also for the broad field of EV-based biomarker discovery. Supplementary Information The online version contains supplementary material available at 10.1186/s40364-020-00259-4.
Collapse
Affiliation(s)
- Hooi Ching Lim
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Lund University, BMC B12, Klinikgatan 26, 22184, Lund, Sweden
| | - Shamit Soneji
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Lund University, BMC B12, Klinikgatan 26, 22184, Lund, Sweden
| | - Róbert Pálmason
- Department of Hematology, Skåne University Hospital, Lund, Sweden
| | - Stig Lenhoff
- Department of Hematology, Skåne University Hospital, Lund, Sweden
| | - Thomas Laurell
- Division of Nanobiotechnology and Lab-on-a-chip, Department of Biomedical Engineering, Lund University, Lund, Sweden
| | - Stefan Scheding
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Lund University, BMC B12, Klinikgatan 26, 22184, Lund, Sweden. .,Department of Hematology, Skåne University Hospital, Lund, Sweden.
| |
Collapse
|
5
|
Szczepanek J. Role of microRNA dysregulation in childhood acute leukemias: Diagnostics, monitoring and therapeutics: A comprehensive review. World J Clin Oncol 2020; 11:348-369. [PMID: 32855905 PMCID: PMC7426929 DOI: 10.5306/wjco.v11.i6.348] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/18/2020] [Accepted: 05/22/2020] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are short noncoding RNAs that regulate the expression of genes by sequence-specific binding to mRNA to either promote or block its translation; they can also act as tumor suppressors (e.g., let-7b, miR-29a, miR-99, mir-100, miR-155, and miR-181) and/or oncogenes (e.g., miR-29a, miR-125b, miR-143-p3, mir-155, miR-181, miR-183, miR-196b, and miR-223) in childhood acute leukemia (AL). Differentially expressed miRNAs are important factors associated with the initiation and progression of AL. As shown in many studies, they can be used as noninvasive diagnostic and prognostic biomarkers, which are useful in monitoring early stages of AL development or during therapy (e.g., miR-125b, miR-146b, miR-181c, and miR-4786), accurate classification of different cellular or molecular AL subgroups (e.g., let-7b, miR-98, miR-100, miR-128b, and miR-223), and identification and development of new therapeutic agents (e.g., mir-10, miR-125b, miR-203, miR-210, miR-335). Specific miRNA patterns have also been described for commonly used AL therapy drugs (e.g., miR-125b and miR-223 for doxorubicin, miR-335 and miR-1208 for prednisolone, and miR-203 for imatinib), uncovering miRNAs that are associated with treatment response. In the current review, the role of miRNAs in the development, progression, and therapy monitoring of pediatric ALs will be presented and discussed.
Collapse
Affiliation(s)
- Joanna Szczepanek
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Toruń 87100, Poland
| |
Collapse
|
6
|
Zhou JD, Li XX, Zhang TJ, Xu ZJ, Zhang ZH, Gu Y, Wen XM, Zhang W, Ji RB, Deng ZQ, Lin J, Qian J. MicroRNA-335/ ID4 dysregulation predicts clinical outcome and facilitates leukemogenesis by activating PI3K/Akt signaling pathway in acute myeloid leukemia. Aging (Albany NY) 2020; 11:3376-3391. [PMID: 31147526 PMCID: PMC6555456 DOI: 10.18632/aging.101991] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/20/2019] [Indexed: 12/22/2022]
Abstract
MircoRNA-335 (miR-335) has been reported as a significant cancer-associated microRNA, which was often epigenetically silenced and acted as a tumor suppressor gene in diverse human solid tumors. Conversely, recent studies show that miR-335 overexpression was identified in both adult and pediatric acute myeloid leukemia (AML), suggesting that it might play an oncogenic role of miR-335 in AML. However, the role of miR-335 during leukemogenesis remains to be elucidated. MiR-335/ID4 expression was detected by real-time quantitative PCR and/or western blot. Survival analysis was performed to explore the association between miR-335/ID4 expression and the prognosis, and further validated by public databases. Gain-of-function experiments determined by cell proliferation, apoptosis, and differentiation were conducted to investigate the biological functions of miR-335/ID4. Herein, we found that miR-335 expression, independent of its methylation, was significantly increased and negatively correlated with reduced ID4 expression in AML. Moreover, aberrant miR-335/ID4 expression independently affected chemotherapy response and leukemia-free/overall survival in patients with AML. Gain-of-function experiments in vitro showed the oncogenic role of miR-335 by affecting cell apoptosis and proliferation in AML, and could be rescued by ID4 restoration. Mechanistically, we identified and verified that miR-335/ID4 contributed to leukemogenesis through activating PI3K/Akt signaling pathway. Collectively, aberrant miR-335/ID4 expression was an independent prognostic biomarker in AML. MiR-335/ID4 dysregulation facilitated leukemogenesis through the activation of PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Jing-Dong Zhou
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, People's Republic of China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, People's Republic of China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, People's Republic of China
| | - Xi-Xi Li
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Ting-Juan Zhang
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, People's Republic of China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, People's Republic of China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, People's Republic of China
| | - Zi-Jun Xu
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, People's Republic of China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, People's Republic of China.,, Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, People's Republic of China
| | - Zhi-Hui Zhang
- Department of Geriatrics, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Yu Gu
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, People's Republic of China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, People's Republic of China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, People's Republic of China
| | - Xiang-Mei Wen
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, People's Republic of China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, People's Republic of China.,, Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, People's Republic of China
| | - Wei Zhang
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, People's Republic of China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, People's Republic of China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, People's Republic of China
| | - Run-Bi Ji
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, People's Republic of China.,, Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, People's Republic of China
| | - Zhao-Qun Deng
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, People's Republic of China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, People's Republic of China.,, Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, People's Republic of China
| | - Jiang Lin
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, People's Republic of China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, People's Republic of China.,, Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, People's Republic of China
| | - Jun Qian
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, People's Republic of China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, People's Republic of China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, People's Republic of China
| |
Collapse
|
7
|
Sun Y, Wang H, Luo C. MiR-100 regulates cell viability and apoptosis by targeting ATM in pediatric acute myeloid leukemia. Biochem Biophys Res Commun 2020; 522:855-861. [PMID: 31801665 DOI: 10.1016/j.bbrc.2019.11.156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 11/22/2019] [Indexed: 12/20/2022]
Abstract
Acute myeloid leukemia (AML) is the most common pediatric malignancy and a major cause of morbidity and mortality in children. miR-100 is associated with progression of various diseases including AML. The aim of this study was to explore the underlying molecule mechanisms of miR-100 involved in AML. The expressions of miR-100 and ataxia telangiectasia mutated (ATM) in pediatric AML patients and cell lines were monitored using qRT-PCR and western blot assays. MTT assay was carried to evaluate cell viability. Cell apoptosis was measured by flow cytometry. The binding sites between miR-100 and ATM were predicted by mirtarbase database. Luciferase reporter assay was used to confirm the relationship between miR-100 and ATM. miR-100 expression was highly expressed in bone marrow of AML patients and cell lines. Moreover, Knockdown of miR-100 led to the inhibition of viability and promotion of apoptosis in Kasumi-1 and MV-4-11 cells. miR-100 harbored the 3'UTR of ATM. Meanwhile, the expression of ATM was downregulated in bone marrow of AML patients and AML cell lines. Subsequently, a negative correlation between miR-100 and ATM in bone marrow of AML patients was also observed. Furthermore, ectopic expression of ATM repressed cell viability while enhanced apoptosis. Notably, loss of ATM attenuated the effect of miR-100 depletion on cell viability and apoptosis in AML cells. miR-100 participates in cell viability and apoptosis by targeting ATM in pediatric AML.
Collapse
Affiliation(s)
- Yin Sun
- Department of Pediatrics, Tengzhou Central People's Hospital, Tengzhou, 277500, Shandong, China
| | - Hongxiang Wang
- Department of Pediatrics, Tengzhou Central People's Hospital, Tengzhou, 277500, Shandong, China
| | - Chibao Luo
- Department of Pediatrics, Tengzhou Central People's Hospital, Tengzhou, 277500, Shandong, China.
| |
Collapse
|
8
|
Dou XQ, Chen XJ, Zhou Q, Wen MX, Zhang SZ, Zhang SQ. miR-335 modulates Numb alternative splicing via targeting RBM10 in endometrial cancer. Kaohsiung J Med Sci 2020; 36:171-177. [PMID: 31894898 DOI: 10.1002/kjm2.12149] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 10/13/2019] [Indexed: 12/19/2022] Open
Abstract
Numb is a conserved protein plays important roles in the development of cancer. Two Numb isoforms have been found produced by alternative splicing and play contrast roles in regulating cellular functions. It is reported that the expression of Numb long isoform (Numb-L) was increased in various kinds of cancers, but in endometrial cancer, the condition is still unknown. The level of two Numb transcripts and protein isoforms were detected by semiquantitative polymerase chain reaction and immunoblotting in 47 paired endometrial tumor and adjacent non-tumor control tissues. The level of three alternative splicing related proteins: RBM5, RBM6, and RBM10 was determined by immunoblotting. MiRNAs targeting RBM10 were predicted by bioinformatics tools and their interaction with RBM10 was confirmed by luciferase assay and immunoblotting. The function of miR-335 in endometrial cancer was examined in xenograft mouse model. Numb-L level was increased in tumors and negatively correlated with RBM10 protein level. RBM10 mRNA level was not significantly altered in endometrial tumors suggesting its expression may regulated by post transcriptional regulators such as miRNAs. We identified miR-133a, miR-133b, and miR-335 directly target RBM10, but only miR-335 level increased in tumors and negatively correlated with RBM10 protein level. miR-335 overexpression promoted tumor growth by downregulating RBM10 and upregulating Numb-L level in xenograft mouse model. miR-335 overexpression promoted Numb-L expression via targeting RBM10 in endometrial cancer, which may provide new biomarkers for EC diagnosis.
Collapse
Affiliation(s)
- Xiao-Qing Dou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Shandong University, Jinan, China
| | - Xiu-Juan Chen
- Department of Obstetrics and Gynecology, People's Hospital of Rizhao, Rizhao, China
| | - Qun Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Ming-Xiao Wen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Shu-Zhen Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Shi-Qian Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Shandong University, Jinan, China
| |
Collapse
|
9
|
Ye J, Luo D, Yu J, Zhu S. Transcriptome analysis identifies key regulators and networks in Acute myeloid leukemia. ACTA ACUST UNITED AC 2019; 24:487-491. [PMID: 31210592 DOI: 10.1080/16078454.2019.1631506] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Acute myeloid leukemia (AML) is a heterogeneous and highly recurrent hematological malignancy. Studies have shown an association between microRNAs and drive genes in AMLs. However, the regulatory roles of miRNAs in AML and how they act on downstream targets and the signaling pathway has been little studied. METHODS As to understand the mechanism of mRNA-miRNA interaction in the blood malignancy from a large scale of transcriptomic sequencing studies, we applied a comprehensive miRNA-mRNA association, co-expression gene network and ingenuity pathway analysis using TCGA AML datasets. RESULTS Our results showed that his-mir-335 was a critical regulatory of homeobox A gene family. PBX3, KAT6A, MEIS1, and COMMD3-BMI1 were predicted as top transcription regulators in the regulatory network of the HOXA family. The most significantly enriched functions were cell growth, proliferation, and survival in the mRNA-miRNA network. CONCLUSION Our work revealed that regulation of the HOXA gene family and its regulation played an important role in the development of AML.
Collapse
Affiliation(s)
- Jiaxin Ye
- a Department of Hematology , Shaoxing Shangyu People's Hospital , Shaoxing , People's Republic of China
| | - Daliang Luo
- a Department of Hematology , Shaoxing Shangyu People's Hospital , Shaoxing , People's Republic of China
| | - Jianhong Yu
- b Department of Geriatric , Affiliated Hospital of Shaoxing University (Shaoxing Municipal Hospital) , Zhejiang , People's Republic of China
| | - Sibo Zhu
- c School of Life Sciences, Fudan University , Shanghai , People's Republic of China
| |
Collapse
|
10
|
Qi J, Shi LY, Wu Y, Shen XJ, Yuan J, Jin CJ, Cong H, Ju SQ. Epigenetic silencing of miR-335 induces migration by targeting insulin-like growth factor-1 receptor in multiple myeloma. Leuk Lymphoma 2019; 60:3188-3198. [PMID: 31190579 DOI: 10.1080/10428194.2019.1627534] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Multiple myeloma (MM) is a common hematological malignancy and remains incurable. MiRNA-335 is a classic tumor suppressor, yet its expression pattern and biological role in MM is unclear. The aim of the present study was to determine the expression pattern, biological role, and mechanism of miR-335 in MM. In this study, we found that miR-335 expression was decreased in MM. The promoter of miR-335 was also hypermethylated in MM. It was found that over-expression of miR-335 or 5-azacytidine treatment suppressed migration of MM cells and down-regulated the expression of IGF-1R. MiR-335 thus acts as a metastatic suppressor by targeting IGF-1R in MM. Moreover, aberrant promoter hyper-methylation is critical for miR-335 silencing in MM. We also found that miR-335 assisted in predicting both the prognosis and progression of disease in MM patients. Observations might offer a new complementary diagnostic and therapeutic target in MM.
Collapse
Affiliation(s)
- Jing Qi
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Lin-Ying Shi
- Medical School of Nantong University, Nantong, Jiangsu Province, China
| | - Yin Wu
- Medical School of Nantong University, Nantong, Jiangsu Province, China
| | - Xian-Juan Shen
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Jie Yuan
- Center of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Chun-Jing Jin
- Center of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Hui Cong
- Center of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Shao-Qing Ju
- Center of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
11
|
Mardani R, Jafari Najaf Abadi MH, Motieian M, Taghizadeh-Boroujeni S, Bayat A, Farsinezhad A, Gheibi Hayat SM, Motieian M, Pourghadamyari H. MicroRNA in leukemia: Tumor suppressors and oncogenes with prognostic potential. J Cell Physiol 2018; 234:8465-8486. [PMID: 30515779 DOI: 10.1002/jcp.27776] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 10/30/2018] [Indexed: 02/06/2023]
Abstract
Leukemia is known as a progressive malignant disease, which destroys the blood-forming organs and results in adverse effects on the proliferation and development of leukocytes and their precursors in the blood and bone marrow. There are four main classes of leukemia including acute leukemia, chronic leukemia, myelogenous leukemia, and lymphocytic leukemia. Given that a variety of internal and external factors could be associated with the initiation and progression of different types of leukemia. One of the important factors is epigenetic regulators such as microRNAs (miRNAs) and long noncoding RNAs (ncRNA). MiRNAs are short ncRNAs which act as tumor suppressor (i.e., miR-15, miR-16, let-7, and miR-127) or oncogene (i.e., miR-155, miR-17-92, miR-21, miR-125b, miR-93, miR-143-p3, miR-196b, and miR-223) in leukemia. It has been shown that deregulation of these molecules are associated with the initiation and progression of leukemia. Hence, miRNAs could be used as potential therapeutic candidates in the treatment of patients with leukemia. Moreover, increasing evidence revealed that miRNAs could be used as diagnostic and prognostic biomarkers in monitoring patients in early stages of disease or after received chemotherapy regimen. It seems that identification and development of new miRNAs could pave to the way to the development new therapeutic platforms for patients with leukemia. Here, we summarized various miRNAs as tumor suppressor and oncogene which could be introduced as therapeutic targets in treatment of leukemia.
Collapse
Affiliation(s)
- Rajab Mardani
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | | | - Mahsa Motieian
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sima Taghizadeh-Boroujeni
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Borujen, Iran
| | - Amir Bayat
- Hematology, Oncology, and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Cell and Molecular Biology, College of Science, Kish International Campus, University of Tehran, Kish, Iran
| | - Alireza Farsinezhad
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Mahtab Motieian
- Department of Internal Medicine, Montefiore New Rochelle Hospital, Albert Einstein College of Medicine, New York, New York
| | - Hossein Pourghadamyari
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran.,Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
12
|
Carvalho de Oliveira J, Molinari Roberto G, Baroni M, Bezerra Salomão K, Alejandra Pezuk J, Sol Brassesco M. MiRNA Dysregulation in Childhood Hematological Cancer. Int J Mol Sci 2018; 19:ijms19092688. [PMID: 30201877 PMCID: PMC6165337 DOI: 10.3390/ijms19092688] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/03/2018] [Accepted: 09/08/2018] [Indexed: 12/14/2022] Open
Abstract
For decades, cancer biology focused largely on the protein-encoding genes that have clear roles in tumor development or progression: cell-cycle control, apoptotic evasion, genome instability, drug resistance, or signaling pathways that stimulate growth, angiogenesis, or metastasis. MicroRNAs (miRNAs), however, represent one of the more abundant classes of cell modulators in multicellular organisms and largely contribute to regulating gene expression. Many of the ~2500 miRNAs discovered to date in humans regulate vital biological processes, and their aberrant expression results in pathological and malignant outcomes. In this review, we highlight what has been learned about the roles of miRNAs in some of the most common human pediatric leukemias and lymphomas, along with their value as diagnostic/prognostic factors.
Collapse
Affiliation(s)
| | - Gabriela Molinari Roberto
- Department of Pediatrics, Ribeirão Preto School of Medicine, University of São Paulo, 14049-900 Ribeirão Preto, Brazil.
| | - Mirella Baroni
- Department of Pediatrics, Ribeirão Preto School of Medicine, University of São Paulo, 14049-900 Ribeirão Preto, Brazil.
| | - Karina Bezerra Salomão
- Department of Pediatrics, Ribeirão Preto School of Medicine, University of São Paulo, 14049-900 Ribeirão Preto, Brazil.
| | - Julia Alejandra Pezuk
- Programa de Pós-graduação em Farmácia, Anhanguera University of São Paulo, UNIAN/SP, 05145-200 São Paulo, Brazil.
| | - María Sol Brassesco
- Departamento de Biologia, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, 14040-901 Ribeirão Preto, Brazil.
| |
Collapse
|
13
|
Li Santi A, Gorrasi A, Alfieri M, Montuori N, Ragno P. A novel oncogenic role for urokinase receptor in leukemia cells: molecular sponge for oncosuppressor microRNAs. Oncotarget 2018; 9:27823-27834. [PMID: 29963240 PMCID: PMC6021242 DOI: 10.18632/oncotarget.25597] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 05/19/2018] [Indexed: 01/05/2023] Open
Abstract
Urokinase receptor (uPAR) expression is up-regulated and represents a negative prognostic marker in most cancers. We previously reported that uPAR and CXCR4 can be regulated by common microRNAs in leukemia cells. Transcripts containing response elements for shared microRNAs in their 3’UTR may regulate their availability. We investigated uPAR 3’UTR capability to recruit microRNAs, thus regulating the expression of their targets. uPAR 3’UTR transfection in KG1 leukemia cells up-regulates the expression of endogenous uPAR. Transfection of uPAR 3’UTR, inserted downstream a reporter gene, increases uPAR expression and simultaneously down-regulates the reporter gene expression. Transfection of uPAR 3’UTR also increases CXCR4 expression; accordingly, uPAR silencing induces down-regulation of CXCR4 expression, through a mechanism involving Dicer, the endoribonuclease required for microRNA maturation. Transfection of uPAR 3’UTR also increases the expression of pro-tumoral factors and modulates cell adhesion and migration, consistently with the capability of uPAR3’UTR-recruited microRNAs to target several and different transcripts and, thus, functions. Finally, we found 3’UTR-containing variants of uPAR transcript in U937 leukemia cells, which show higher levels of uPAR expression as compared to KG1 cells, in which these variants are not detected. These results suggest that uPAR mRNA may recruit oncosuppressor microRNAs, allowing the expression of their targets.
Collapse
Affiliation(s)
- Anna Li Santi
- Department of Chemistry and Biology, University of Salerno, Salerno, Italy
| | - Anna Gorrasi
- Department of Chemistry and Biology, University of Salerno, Salerno, Italy
| | | | - Nunzia Montuori
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Pia Ragno
- Department of Chemistry and Biology, University of Salerno, Salerno, Italy
| |
Collapse
|
14
|
Differential expression profiles of miRNAs and correlation with clinical outcomes in acute myeloid leukemia. Meta Gene 2018. [DOI: 10.1016/j.mgene.2018.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
15
|
Tian C, Zhang L, Li X, Zhang Y, Li J, Chen L. Low miR-192 expression predicts poor prognosis in pediatric acute myeloid leukemia. Cancer Biomark 2018; 22:209-215. [PMID: 29689705 DOI: 10.3233/cbm-170657] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Chunmei Tian
- Department of Pediatrics, Affiliated Hospital of Binzhou Medical University, Binzhou 256603, Shandong, China
| | - Lin Zhang
- Department of Radiology, Affiliated Hospital of Binzhou Medical University, Binzhou 256603, Shandong, China
| | - Xiaohua Li
- Department of Pediatrics, Affiliated Hospital of Binzhou Medical University, Binzhou 256603, Shandong, China
| | - Yanjun Zhang
- Department of Pediatrics, Affiliated Hospital of Binzhou Medical University, Binzhou 256603, Shandong, China
| | - Jianchang Li
- Department of Pediatrics, Affiliated Hospital of Binzhou Medical University, Binzhou 256603, Shandong, China
| | - Liang Chen
- Department of Radiology, Affiliated Hospital of Binzhou Medical University, Binzhou 256603, Shandong, China
| |
Collapse
|
16
|
Liao Q, Wang B, Li X, Jiang G. miRNAs in acute myeloid leukemia. Oncotarget 2018; 8:3666-3682. [PMID: 27705921 PMCID: PMC5356910 DOI: 10.18632/oncotarget.12343] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 09/24/2016] [Indexed: 12/30/2022] Open
Abstract
MicroRNAs (miRNAs) are small, non-coding RNAs found throughout the eukaryotes that control the expression of a number of genes involved in commitment and differentiation of hematopoietic stem cells and tumorigenesis. Widespread dysregulation of miRNAs have been found in hematological malignancies, including human acute myeloid leukemia (AML). A comprehensive understanding of the role of miRNAs within the complex regulatory networks that are disrupted in malignant AML cells is a prerequisite for the development of therapeutic strategies employing miRNA modulators. Herein, we review the roles of emerging miRNAs and the miRNAs regulatory networks in AML pathogenesis, prognosis, and miRNA-directed therapies.
Collapse
Affiliation(s)
- Qiong Liao
- Key Laboratory for Rare & Uncommon Dseases of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China.,School of Medicine and Life Sciences, Jinan University, Jinan, Shandong, P.R. China
| | - Bingping Wang
- Department of Hematology, Shengli Oilfield Central Hospital, Dongying, Shandong, P.R. China
| | - Xia Li
- Key Laboratory for Rare & Uncommon Dseases of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China.,Shandong University School of Medicine, Jinan, Shandong, P.R. China
| | - Guosheng Jiang
- Key Laboratory for Rare & Uncommon Dseases of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| |
Collapse
|
17
|
Trino S, Lamorte D, Caivano A, Laurenzana I, Tagliaferri D, Falco G, Del Vecchio L, Musto P, De Luca L. MicroRNAs as New Biomarkers for Diagnosis and Prognosis, and as Potential Therapeutic Targets in Acute Myeloid Leukemia. Int J Mol Sci 2018; 19:ijms19020460. [PMID: 29401684 PMCID: PMC5855682 DOI: 10.3390/ijms19020460] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/12/2018] [Accepted: 01/12/2018] [Indexed: 02/07/2023] Open
Abstract
Acute myeloid leukemias (AML) are clonal disorders of hematopoietic progenitor cells which are characterized by relevant heterogeneity in terms of phenotypic, genotypic, and clinical features. Among the genetic aberrations that control disease development there are microRNAs (miRNAs). miRNAs are small non-coding RNAs that regulate, at post-transcriptional level, translation and stability of mRNAs. It is now established that deregulated miRNA expression is a prominent feature in AML. Functional studies have shown that miRNAs play an important role in AML pathogenesis and miRNA expression signatures are associated with chemotherapy response and clinical outcome. In this review we summarized miRNA signature in AML with different cytogenetic, molecular and clinical characteristics. Moreover, we reviewed the miRNA regulatory network in AML pathogenesis and we discussed the potential use of cellular and circulating miRNAs as biomarkers for diagnosis and prognosis and as therapeutic targets.
Collapse
MESH Headings
- Animals
- Antagomirs/genetics
- Antagomirs/metabolism
- Antagomirs/therapeutic use
- Biomarkers, Tumor/agonists
- Biomarkers, Tumor/antagonists & inhibitors
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Chromosome Aberrations
- Extracellular Vesicles/metabolism
- Extracellular Vesicles/pathology
- Gene Expression Regulation, Leukemic
- Humans
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/therapy
- Mice
- MicroRNAs/agonists
- MicroRNAs/antagonists & inhibitors
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Molecular Targeted Therapy
- Oligoribonucleotides/genetics
- Oligoribonucleotides/metabolism
- Oligoribonucleotides/therapeutic use
- Oncogene Proteins, Fusion/antagonists & inhibitors
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Prognosis
- Signal Transduction
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Stefania Trino
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Daniela Lamorte
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Antonella Caivano
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Ilaria Laurenzana
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Daniela Tagliaferri
- Biogem Scarl, Istituto di Ricerche Genetiche 'Gaetano Salvatore', 83031 Ariano Irpino, Italy.
| | - Geppino Falco
- Biogem Scarl, Istituto di Ricerche Genetiche 'Gaetano Salvatore', 83031 Ariano Irpino, Italy.
- Department of Biology, University of Naples Federico II, 80147 Naples, Italy.
| | - Luigi Del Vecchio
- CEINGE Biotecnologie Avanzate s.c.a r.l., 80147 Naples, Italy.
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80138 Naples, Italy.
| | - Pellegrino Musto
- Scientific Direction, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Potenza, Italy.
| | - Luciana De Luca
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| |
Collapse
|
18
|
Kumar S, Bakhshi S. Diagnostic & prognostic role of microRNAs in paediatric acute myeloid leukaemia. Indian J Med Res 2018; 144:807-814. [PMID: 28474617 PMCID: PMC5433273 DOI: 10.4103/ijmr.ijmr_220_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Dysregulation in microRNAs (miRNAs) expression has been observed in distinct acute myeloid leukaemia (AML) subtypes, and their potential as an effective diagnostic and prognostic biomarker is slowly being realized. Certain miRNAs have been found to be associated with various cytogenetic and molecular abnormalities of prognostic significance in AML. Experimental evidences have indicated the potential of modulating miRNA expression as an effective antileukaemic strategy. This has opened a new window for miRNAs-based targeted therapies. In this review, we present results of some studies analyzing the dysregulation in miRNAs expression pattern in paediatric AML and also discuss their use as diagnostic and prognostic markers.
Collapse
Affiliation(s)
- Sachin Kumar
- Department of Medical Oncology, Dr B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
19
|
Feng R, Beeharry MK, Lu S, Sah BK, Yuan F, Yan M, Liu B, Li C, Zhu Z. Down-regulated serum miR-126 is associated with aggressive progression and poor prognosis of gastric cancer. Cancer Biomark 2018; 22:119-126. [PMID: 29562500 DOI: 10.3233/cbm-171099] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND miR-126 functions as a tumor suppressor in gastric cancer (GC), however, the clinical significance of serum miR-126 in GC remains unclear. OBJECTIVE To investigate the associations of serum miR-126 level with the clinicopathological characteristics and prognosis of GC patients. METHODS Quantitative real-time polymerase chain reaction was performed to examine the expression levels of miR-126 in 338 GC patients' tissues and sera, and 50 healthy controls' sera. The associations of serum miR-126 with clinicopathological characteristics and clinical outcome were evaluated. RESULTS Compared with the matched adjacent non-tumor tissues and normal sera, miR-126 expression was significantly down-regulated in both tumor tissues and sera of GC patients. Importantly, there was a positive correlation between tissue and serum levels of miR-126 in GC patients. A reduced serum miR-126 level statistically correlated with aggressive clinicopathological characteristics, such as larger tumor size, deeper local invasion, more lymph node metastasis, advanced TNM stage, and poorer prognosis. Notably, multivariate analysis identified reduced serum miR-126 level as an independent predictor for the unfavorable prognosis of GC. CONCLUSIONS These results indicate for the first time that serum miR-126 may serve as a novel prognostic biomarker in GC.
Collapse
Affiliation(s)
- Runhua Feng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Key Laboratory of Gastric Neoplasms, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Maneesh K Beeharry
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Sheng Lu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Birendra K Sah
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Fei Yuan
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Min Yan
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Bingya Liu
- Shanghai Key Laboratory of Gastric Neoplasms, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chen Li
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Key Laboratory of Gastric Neoplasms, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhenggang Zhu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Key Laboratory of Gastric Neoplasms, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
20
|
Lim EL, Trinh DL, Ries RE, Wang J, Gerbing RB, Ma Y, Topham J, Hughes M, Pleasance E, Mungall AJ, Moore R, Zhao Y, Aplenc R, Sung L, Kolb EA, Gamis A, Smith M, Gerhard DS, Alonzo TA, Meshinchi S, Marra MA. MicroRNA Expression-Based Model Indicates Event-Free Survival in Pediatric Acute Myeloid Leukemia. J Clin Oncol 2017; 35:3964-3977. [PMID: 29068783 PMCID: PMC5721230 DOI: 10.1200/jco.2017.74.7451] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Purpose Children with acute myeloid leukemia (AML) whose disease is refractory to standard induction chemotherapy therapy or who experience relapse after initial response have dismal outcomes. We sought to comprehensively profile pediatric AML microRNA (miRNA) samples to identify dysregulated genes and assess the utility of miRNAs for improved outcome prediction. Patients and Methods To identify miRNA biomarkers that are associated with treatment failure, we performed a comprehensive sequence-based characterization of the pediatric AML miRNA landscape. miRNA sequencing was performed on 1,362 samples-1,303 primary, 22 refractory, and 37 relapse samples. One hundred sixty-four matched samples-127 primary and 37 relapse samples-were analyzed by using RNA sequencing. Results By using penalized lasso Cox proportional hazards regression, we identified 36 miRNAs the expression levels at diagnosis of which were highly associated with event-free survival. Combined expression of the 36 miRNAs was used to create a novel miRNA-based risk classification scheme (AMLmiR36). This new miRNA-based risk classifier identifies those patients who are at high risk (hazard ratio, 2.830; P ≤ .001) or low risk (hazard ratio, 0.323; P ≤ .001) of experiencing treatment failure, independent of conventional karyotype or mutation status. The performance of AMLmiR36 was independently assessed by using 878 patients from two different clinical trials (AAML0531 and AAML1031). Our analysis also revealed that miR-106a-363 was abundantly expressed in relapse and refractory samples, and several candidate targets of miR-106a-5p were involved in oxidative phosphorylation, a process that is suppressed in treatment-resistant leukemic cells. Conclusion To assess the utility of miRNAs for outcome prediction in patients with pediatric AML, we designed and validated a miRNA-based risk classification scheme. We also hypothesized that the abundant expression of miR-106a could increase treatment resistance via modulation of genes that are involved in oxidative phosphorylation.
Collapse
Affiliation(s)
- Emilia L. Lim
- Emilia L. Lim, Diane L. Trinh, Yussanne Ma, James Topham, Erin Pleasance, Andrew J. Mungall, Richard Moore, Yongjun Zhao, and Marco A. Marra, Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency; Marco A. Marra, University of British Columbia, Vancouver, British Columbia; Lillian Sung, The Hospital for Sick Children, Toronto, Ontario, Canada; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, University of Washington, Seattle, WA; Jim Wang, Robert B. Gerbing, E. Anders Kolb, Alan Gamis, and Todd A. Alonzo, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, The Children’s Hospital of Philadelphia, Philadelphia, PA; Malcolm Smith and Daniela S. Gerhard, Office of Cancer Genomics, National Cancer Institute, Bethesda, MD; and Robert J. Arceci, Phoenix Children’s Hospital, Phoenix. AZ
| | - Diane L. Trinh
- Emilia L. Lim, Diane L. Trinh, Yussanne Ma, James Topham, Erin Pleasance, Andrew J. Mungall, Richard Moore, Yongjun Zhao, and Marco A. Marra, Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency; Marco A. Marra, University of British Columbia, Vancouver, British Columbia; Lillian Sung, The Hospital for Sick Children, Toronto, Ontario, Canada; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, University of Washington, Seattle, WA; Jim Wang, Robert B. Gerbing, E. Anders Kolb, Alan Gamis, and Todd A. Alonzo, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, The Children’s Hospital of Philadelphia, Philadelphia, PA; Malcolm Smith and Daniela S. Gerhard, Office of Cancer Genomics, National Cancer Institute, Bethesda, MD; and Robert J. Arceci, Phoenix Children’s Hospital, Phoenix. AZ
| | - Rhonda E. Ries
- Emilia L. Lim, Diane L. Trinh, Yussanne Ma, James Topham, Erin Pleasance, Andrew J. Mungall, Richard Moore, Yongjun Zhao, and Marco A. Marra, Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency; Marco A. Marra, University of British Columbia, Vancouver, British Columbia; Lillian Sung, The Hospital for Sick Children, Toronto, Ontario, Canada; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, University of Washington, Seattle, WA; Jim Wang, Robert B. Gerbing, E. Anders Kolb, Alan Gamis, and Todd A. Alonzo, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, The Children’s Hospital of Philadelphia, Philadelphia, PA; Malcolm Smith and Daniela S. Gerhard, Office of Cancer Genomics, National Cancer Institute, Bethesda, MD; and Robert J. Arceci, Phoenix Children’s Hospital, Phoenix. AZ
| | - Jim Wang
- Emilia L. Lim, Diane L. Trinh, Yussanne Ma, James Topham, Erin Pleasance, Andrew J. Mungall, Richard Moore, Yongjun Zhao, and Marco A. Marra, Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency; Marco A. Marra, University of British Columbia, Vancouver, British Columbia; Lillian Sung, The Hospital for Sick Children, Toronto, Ontario, Canada; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, University of Washington, Seattle, WA; Jim Wang, Robert B. Gerbing, E. Anders Kolb, Alan Gamis, and Todd A. Alonzo, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, The Children’s Hospital of Philadelphia, Philadelphia, PA; Malcolm Smith and Daniela S. Gerhard, Office of Cancer Genomics, National Cancer Institute, Bethesda, MD; and Robert J. Arceci, Phoenix Children’s Hospital, Phoenix. AZ
| | - Robert B. Gerbing
- Emilia L. Lim, Diane L. Trinh, Yussanne Ma, James Topham, Erin Pleasance, Andrew J. Mungall, Richard Moore, Yongjun Zhao, and Marco A. Marra, Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency; Marco A. Marra, University of British Columbia, Vancouver, British Columbia; Lillian Sung, The Hospital for Sick Children, Toronto, Ontario, Canada; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, University of Washington, Seattle, WA; Jim Wang, Robert B. Gerbing, E. Anders Kolb, Alan Gamis, and Todd A. Alonzo, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, The Children’s Hospital of Philadelphia, Philadelphia, PA; Malcolm Smith and Daniela S. Gerhard, Office of Cancer Genomics, National Cancer Institute, Bethesda, MD; and Robert J. Arceci, Phoenix Children’s Hospital, Phoenix. AZ
| | - Yussanne Ma
- Emilia L. Lim, Diane L. Trinh, Yussanne Ma, James Topham, Erin Pleasance, Andrew J. Mungall, Richard Moore, Yongjun Zhao, and Marco A. Marra, Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency; Marco A. Marra, University of British Columbia, Vancouver, British Columbia; Lillian Sung, The Hospital for Sick Children, Toronto, Ontario, Canada; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, University of Washington, Seattle, WA; Jim Wang, Robert B. Gerbing, E. Anders Kolb, Alan Gamis, and Todd A. Alonzo, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, The Children’s Hospital of Philadelphia, Philadelphia, PA; Malcolm Smith and Daniela S. Gerhard, Office of Cancer Genomics, National Cancer Institute, Bethesda, MD; and Robert J. Arceci, Phoenix Children’s Hospital, Phoenix. AZ
| | - James Topham
- Emilia L. Lim, Diane L. Trinh, Yussanne Ma, James Topham, Erin Pleasance, Andrew J. Mungall, Richard Moore, Yongjun Zhao, and Marco A. Marra, Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency; Marco A. Marra, University of British Columbia, Vancouver, British Columbia; Lillian Sung, The Hospital for Sick Children, Toronto, Ontario, Canada; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, University of Washington, Seattle, WA; Jim Wang, Robert B. Gerbing, E. Anders Kolb, Alan Gamis, and Todd A. Alonzo, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, The Children’s Hospital of Philadelphia, Philadelphia, PA; Malcolm Smith and Daniela S. Gerhard, Office of Cancer Genomics, National Cancer Institute, Bethesda, MD; and Robert J. Arceci, Phoenix Children’s Hospital, Phoenix. AZ
| | - Maya Hughes
- Emilia L. Lim, Diane L. Trinh, Yussanne Ma, James Topham, Erin Pleasance, Andrew J. Mungall, Richard Moore, Yongjun Zhao, and Marco A. Marra, Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency; Marco A. Marra, University of British Columbia, Vancouver, British Columbia; Lillian Sung, The Hospital for Sick Children, Toronto, Ontario, Canada; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, University of Washington, Seattle, WA; Jim Wang, Robert B. Gerbing, E. Anders Kolb, Alan Gamis, and Todd A. Alonzo, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, The Children’s Hospital of Philadelphia, Philadelphia, PA; Malcolm Smith and Daniela S. Gerhard, Office of Cancer Genomics, National Cancer Institute, Bethesda, MD; and Robert J. Arceci, Phoenix Children’s Hospital, Phoenix. AZ
| | - Erin Pleasance
- Emilia L. Lim, Diane L. Trinh, Yussanne Ma, James Topham, Erin Pleasance, Andrew J. Mungall, Richard Moore, Yongjun Zhao, and Marco A. Marra, Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency; Marco A. Marra, University of British Columbia, Vancouver, British Columbia; Lillian Sung, The Hospital for Sick Children, Toronto, Ontario, Canada; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, University of Washington, Seattle, WA; Jim Wang, Robert B. Gerbing, E. Anders Kolb, Alan Gamis, and Todd A. Alonzo, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, The Children’s Hospital of Philadelphia, Philadelphia, PA; Malcolm Smith and Daniela S. Gerhard, Office of Cancer Genomics, National Cancer Institute, Bethesda, MD; and Robert J. Arceci, Phoenix Children’s Hospital, Phoenix. AZ
| | - Andrew J. Mungall
- Emilia L. Lim, Diane L. Trinh, Yussanne Ma, James Topham, Erin Pleasance, Andrew J. Mungall, Richard Moore, Yongjun Zhao, and Marco A. Marra, Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency; Marco A. Marra, University of British Columbia, Vancouver, British Columbia; Lillian Sung, The Hospital for Sick Children, Toronto, Ontario, Canada; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, University of Washington, Seattle, WA; Jim Wang, Robert B. Gerbing, E. Anders Kolb, Alan Gamis, and Todd A. Alonzo, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, The Children’s Hospital of Philadelphia, Philadelphia, PA; Malcolm Smith and Daniela S. Gerhard, Office of Cancer Genomics, National Cancer Institute, Bethesda, MD; and Robert J. Arceci, Phoenix Children’s Hospital, Phoenix. AZ
| | - Richard Moore
- Emilia L. Lim, Diane L. Trinh, Yussanne Ma, James Topham, Erin Pleasance, Andrew J. Mungall, Richard Moore, Yongjun Zhao, and Marco A. Marra, Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency; Marco A. Marra, University of British Columbia, Vancouver, British Columbia; Lillian Sung, The Hospital for Sick Children, Toronto, Ontario, Canada; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, University of Washington, Seattle, WA; Jim Wang, Robert B. Gerbing, E. Anders Kolb, Alan Gamis, and Todd A. Alonzo, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, The Children’s Hospital of Philadelphia, Philadelphia, PA; Malcolm Smith and Daniela S. Gerhard, Office of Cancer Genomics, National Cancer Institute, Bethesda, MD; and Robert J. Arceci, Phoenix Children’s Hospital, Phoenix. AZ
| | - Yongjun Zhao
- Emilia L. Lim, Diane L. Trinh, Yussanne Ma, James Topham, Erin Pleasance, Andrew J. Mungall, Richard Moore, Yongjun Zhao, and Marco A. Marra, Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency; Marco A. Marra, University of British Columbia, Vancouver, British Columbia; Lillian Sung, The Hospital for Sick Children, Toronto, Ontario, Canada; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, University of Washington, Seattle, WA; Jim Wang, Robert B. Gerbing, E. Anders Kolb, Alan Gamis, and Todd A. Alonzo, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, The Children’s Hospital of Philadelphia, Philadelphia, PA; Malcolm Smith and Daniela S. Gerhard, Office of Cancer Genomics, National Cancer Institute, Bethesda, MD; and Robert J. Arceci, Phoenix Children’s Hospital, Phoenix. AZ
| | - Richard Aplenc
- Emilia L. Lim, Diane L. Trinh, Yussanne Ma, James Topham, Erin Pleasance, Andrew J. Mungall, Richard Moore, Yongjun Zhao, and Marco A. Marra, Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency; Marco A. Marra, University of British Columbia, Vancouver, British Columbia; Lillian Sung, The Hospital for Sick Children, Toronto, Ontario, Canada; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, University of Washington, Seattle, WA; Jim Wang, Robert B. Gerbing, E. Anders Kolb, Alan Gamis, and Todd A. Alonzo, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, The Children’s Hospital of Philadelphia, Philadelphia, PA; Malcolm Smith and Daniela S. Gerhard, Office of Cancer Genomics, National Cancer Institute, Bethesda, MD; and Robert J. Arceci, Phoenix Children’s Hospital, Phoenix. AZ
| | - Lillian Sung
- Emilia L. Lim, Diane L. Trinh, Yussanne Ma, James Topham, Erin Pleasance, Andrew J. Mungall, Richard Moore, Yongjun Zhao, and Marco A. Marra, Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency; Marco A. Marra, University of British Columbia, Vancouver, British Columbia; Lillian Sung, The Hospital for Sick Children, Toronto, Ontario, Canada; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, University of Washington, Seattle, WA; Jim Wang, Robert B. Gerbing, E. Anders Kolb, Alan Gamis, and Todd A. Alonzo, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, The Children’s Hospital of Philadelphia, Philadelphia, PA; Malcolm Smith and Daniela S. Gerhard, Office of Cancer Genomics, National Cancer Institute, Bethesda, MD; and Robert J. Arceci, Phoenix Children’s Hospital, Phoenix. AZ
| | - E. Anders Kolb
- Emilia L. Lim, Diane L. Trinh, Yussanne Ma, James Topham, Erin Pleasance, Andrew J. Mungall, Richard Moore, Yongjun Zhao, and Marco A. Marra, Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency; Marco A. Marra, University of British Columbia, Vancouver, British Columbia; Lillian Sung, The Hospital for Sick Children, Toronto, Ontario, Canada; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, University of Washington, Seattle, WA; Jim Wang, Robert B. Gerbing, E. Anders Kolb, Alan Gamis, and Todd A. Alonzo, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, The Children’s Hospital of Philadelphia, Philadelphia, PA; Malcolm Smith and Daniela S. Gerhard, Office of Cancer Genomics, National Cancer Institute, Bethesda, MD; and Robert J. Arceci, Phoenix Children’s Hospital, Phoenix. AZ
| | - Alan Gamis
- Emilia L. Lim, Diane L. Trinh, Yussanne Ma, James Topham, Erin Pleasance, Andrew J. Mungall, Richard Moore, Yongjun Zhao, and Marco A. Marra, Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency; Marco A. Marra, University of British Columbia, Vancouver, British Columbia; Lillian Sung, The Hospital for Sick Children, Toronto, Ontario, Canada; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, University of Washington, Seattle, WA; Jim Wang, Robert B. Gerbing, E. Anders Kolb, Alan Gamis, and Todd A. Alonzo, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, The Children’s Hospital of Philadelphia, Philadelphia, PA; Malcolm Smith and Daniela S. Gerhard, Office of Cancer Genomics, National Cancer Institute, Bethesda, MD; and Robert J. Arceci, Phoenix Children’s Hospital, Phoenix. AZ
| | - Malcolm Smith
- Emilia L. Lim, Diane L. Trinh, Yussanne Ma, James Topham, Erin Pleasance, Andrew J. Mungall, Richard Moore, Yongjun Zhao, and Marco A. Marra, Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency; Marco A. Marra, University of British Columbia, Vancouver, British Columbia; Lillian Sung, The Hospital for Sick Children, Toronto, Ontario, Canada; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, University of Washington, Seattle, WA; Jim Wang, Robert B. Gerbing, E. Anders Kolb, Alan Gamis, and Todd A. Alonzo, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, The Children’s Hospital of Philadelphia, Philadelphia, PA; Malcolm Smith and Daniela S. Gerhard, Office of Cancer Genomics, National Cancer Institute, Bethesda, MD; and Robert J. Arceci, Phoenix Children’s Hospital, Phoenix. AZ
| | - Daniela S. Gerhard
- Emilia L. Lim, Diane L. Trinh, Yussanne Ma, James Topham, Erin Pleasance, Andrew J. Mungall, Richard Moore, Yongjun Zhao, and Marco A. Marra, Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency; Marco A. Marra, University of British Columbia, Vancouver, British Columbia; Lillian Sung, The Hospital for Sick Children, Toronto, Ontario, Canada; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, University of Washington, Seattle, WA; Jim Wang, Robert B. Gerbing, E. Anders Kolb, Alan Gamis, and Todd A. Alonzo, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, The Children’s Hospital of Philadelphia, Philadelphia, PA; Malcolm Smith and Daniela S. Gerhard, Office of Cancer Genomics, National Cancer Institute, Bethesda, MD; and Robert J. Arceci, Phoenix Children’s Hospital, Phoenix. AZ
| | - Todd A. Alonzo
- Emilia L. Lim, Diane L. Trinh, Yussanne Ma, James Topham, Erin Pleasance, Andrew J. Mungall, Richard Moore, Yongjun Zhao, and Marco A. Marra, Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency; Marco A. Marra, University of British Columbia, Vancouver, British Columbia; Lillian Sung, The Hospital for Sick Children, Toronto, Ontario, Canada; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, University of Washington, Seattle, WA; Jim Wang, Robert B. Gerbing, E. Anders Kolb, Alan Gamis, and Todd A. Alonzo, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, The Children’s Hospital of Philadelphia, Philadelphia, PA; Malcolm Smith and Daniela S. Gerhard, Office of Cancer Genomics, National Cancer Institute, Bethesda, MD; and Robert J. Arceci, Phoenix Children’s Hospital, Phoenix. AZ
| | - Soheil Meshinchi
- Emilia L. Lim, Diane L. Trinh, Yussanne Ma, James Topham, Erin Pleasance, Andrew J. Mungall, Richard Moore, Yongjun Zhao, and Marco A. Marra, Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency; Marco A. Marra, University of British Columbia, Vancouver, British Columbia; Lillian Sung, The Hospital for Sick Children, Toronto, Ontario, Canada; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, University of Washington, Seattle, WA; Jim Wang, Robert B. Gerbing, E. Anders Kolb, Alan Gamis, and Todd A. Alonzo, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, The Children’s Hospital of Philadelphia, Philadelphia, PA; Malcolm Smith and Daniela S. Gerhard, Office of Cancer Genomics, National Cancer Institute, Bethesda, MD; and Robert J. Arceci, Phoenix Children’s Hospital, Phoenix. AZ
| | - Marco A. Marra
- Emilia L. Lim, Diane L. Trinh, Yussanne Ma, James Topham, Erin Pleasance, Andrew J. Mungall, Richard Moore, Yongjun Zhao, and Marco A. Marra, Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency; Marco A. Marra, University of British Columbia, Vancouver, British Columbia; Lillian Sung, The Hospital for Sick Children, Toronto, Ontario, Canada; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Rhonda E. Ries, Maya Hughes, and Soheil Meshinchi, University of Washington, Seattle, WA; Jim Wang, Robert B. Gerbing, E. Anders Kolb, Alan Gamis, and Todd A. Alonzo, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, The Children’s Hospital of Philadelphia, Philadelphia, PA; Malcolm Smith and Daniela S. Gerhard, Office of Cancer Genomics, National Cancer Institute, Bethesda, MD; and Robert J. Arceci, Phoenix Children’s Hospital, Phoenix. AZ
| |
Collapse
|
21
|
MicroRNAs as prognostic biomarker and relapse indicator in leukemia. Clin Transl Oncol 2017; 19:951-960. [PMID: 28271337 DOI: 10.1007/s12094-017-1638-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 02/23/2017] [Indexed: 12/15/2022]
Abstract
Despite significant progress in the treatment of different types of leukemia, relapse remains a challenging clinical problem that is observed in a number of patients who are often resistant to chemotherapy and exhibit multi-drug resistance. Identification of new functional biomarkers, including microRNAs, is essential to determine prognosis and relapse at the time of diagnosis. The aim of this study was to detect the specific microRNAs involved in predicting relapse or progression in acute and chronic leukemias, as well as their relationship with overall survival (OS) and relapse-free survival (RFS). The relevant literature was identified through a PubMed and Scholar search (2008-2016) of English-language papers using the terms Leukemia, microRNAs, survival and relapse. Different leukemia types and subtypes show specific microRNA expression profile and different changes, which can be useful in the differentiation between leukemias and evaluation of relapse at the time of diagnosis. Altered microRNA expression profiles can turn these molecules into oncogenes or tumor suppressors, which affect the expression of relapse-related genes. Therefore, monitoring of specific microRNA expression profiles from diagnosis and during follow-up of patients can contribute to the assessment of outcome and determination of relapse and prognosis of leukemic patients.
Collapse
|
22
|
Luo LJ, Wang DD, Wang J, Yang F, Tang JH. Diverse roles of miR-335 in development and progression of cancers. Tumour Biol 2016; 37:15399–15410. [PMID: 27718128 DOI: 10.1007/s13277-016-5385-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 09/09/2016] [Indexed: 12/16/2022] Open
Abstract
MicroRNAs (miRNAs), a series of small noncoding RNAs that regulate gene expression at the post-transcriptional/translational level, are pivotal in cell differentiation, biological development, occurrence, and development of diseases, especially in cancers. Early studies have shown that miRNA-335 (miR-335) is widely dysregulated in human cancers and play critical roles in tumorigenesis and tumor progression. In this review, we aim to summarize the regulation of miR-335 expression mechanisms in cancers. We focus on the target genes regulated by miR-335 and its downstream signaling pathways involved in the biological effects of tumor growth, invasion, and metastasis both in vitro and in vivo, and analyze the relationships between miR-335 expression and the clinical characteristics of tumors as well as its effects on prognosis. The collected evidences support the potential use of miR-335 in prognosis and diagnosis as well as the therapeutic prospects of miR-335 in cancers.
Collapse
Affiliation(s)
- Long-Ji Luo
- Department of General Surgery, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of General Surgery, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Baiziting 42, Nanjing, 210009, China
| | - Dan-Dan Wang
- Department of General Surgery, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Baiziting 42, Nanjing, 210009, China
- Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jing Wang
- Department of General Surgery, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Baiziting 42, Nanjing, 210009, China
- Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fan Yang
- Department of General Surgery, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of General Surgery, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Baiziting 42, Nanjing, 210009, China
| | - Jin-Hai Tang
- Department of General Surgery, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Baiziting 42, Nanjing, 210009, China.
| |
Collapse
|
23
|
Yeh CH, Moles R, Nicot C. Clinical significance of microRNAs in chronic and acute human leukemia. Mol Cancer 2016; 15:37. [PMID: 27179712 PMCID: PMC4867976 DOI: 10.1186/s12943-016-0518-2] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/01/2016] [Indexed: 01/01/2023] Open
Abstract
Small non-coding microRNAs (miRNAs) are epigenetic regulators that target specific cellular mRNA to modulate gene expression patterns and cellular signaling pathways. miRNAs are involved in a wide range of biological processes and are frequently deregulated in human cancers. Numerous miRNAs promote tumorigenesis and cancer progression by enhancing tumor growth, angiogenesis, invasion and immune evasion, while others have tumor suppressive effects (Hayes, et al., Trends Mol Med 20(8): 460-9, 2014; Stahlhut and Slack, Genome Med 5 (12): 111, 2013). The expression profile of cancer miRNAs can be used to predict patient prognosis and clinical response to treatment (Bouchie, Nat Biotechnol 31(7): 577, 2013). The majority of miRNAs are intracellular localized, however circulating miRNAs have been detected in various body fluids and represent new biomarkers of solid and hematologic cancers (Fabris and Calin, Mol Oncol 10(3):503-8, 2016; Allegra, et al., Int J Oncol 41(6): 1897-912, 2012). This review describes the clinical relevance of miRNAs, lncRNAs and snoRNAs in the diagnosis, prognosis and treatment response in patients with chronic lymphocytic leukemia (CLL), chronic myeloid leukemia (CML), acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML) and acute adult T-cell leukemia (ATL).
Collapse
Affiliation(s)
- Chien-Hung Yeh
- Department of Pathology, Center for Viral Oncology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Ramona Moles
- Department of Pathology, Center for Viral Oncology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Christophe Nicot
- Department of Pathology, Center for Viral Oncology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA.
| |
Collapse
|
24
|
Li X, Zhong H. The diagnosis, prognosis, and therapeutic application of MicroRNAs in haematological malignancies. ACTA ACUST UNITED AC 2016; 21:263-71. [PMID: 26907667 DOI: 10.1080/10245332.2015.1114766] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE MicroRNAs (miRNAs) are small noncoding RNA molecules that participate in vital cell processes such as proliferation, apoptosis, and differentiation. In recent years, they have been proven to play vital roles in haematological malignancies. In this review we briefly introduce some basic knowledge of microRNAs and summarize their ectopic expression in haematological malignancies, especially in leukaemia. We will also discuss the potential of microRNAs in the diagnosis of leukaemia, in the determination of the clinical prognosis of diverse subtypes, and in targeted therapy. DISCUSSION Despite current adoption of novel biological agents combining traditional chemotherapy regimens, leukaemia remains to have undesirable clinical outcomes due to inaccurate diagnosis, invasiveness of the disease, and patients' intolerance to chemotherapy, thus brand new therapeutic directions are urgently needed. MiRNAs regulate gene expression by means of binding to the 3'-untranslated regions of corresponding mRNAs, leading to the degradation of targeted mRNA or the inhibition of translation. It has been confirmed that they can either function as tumour inhibitors, or may trigger tumourigenesis in certain situations, this specific dual characteristic undoubtedly attract scientists to explore their roles in haematological malignancies. It is of great necessity to summarize the roles of miRNAs in haematological malignancies diagnosis, prognosis evaluation, and clinical treatment. CONCLUSIONS Future studies may take full advantage of miRNAs detection in diagnosing, in choosing targeted biological therapy, and in avoiding predictable side effect, thus the overall survival rate and cure efficiency of leukaemia should improve.
Collapse
Affiliation(s)
- Xin Li
- a Department of Hematology, Ren Ji Hospital, School of Medicine , Shanghai Jiao Tong University , P.R. China
| | - Hua Zhong
- a Department of Hematology, Ren Ji Hospital, School of Medicine , Shanghai Jiao Tong University , P.R. China
| |
Collapse
|
25
|
Abstract
In the context of breast cancer, the importance of the skeleton in the regulation of primary tumour development and as a site for subsequent metastasis is well characterized. Our understanding of the contributions made by the host bone and bone marrow cells increasingly demonstrates the extent of the interaction between tumour cells and normal host cells. As a result, the need to develop and utilize therapies that can impede the growth and/or function of tumour cells while sparing normal host bone and bone marrow cells is immense and expanding. The need for these new treatments is, however, superimposed on the orthopaedic management of patients' quality of life, where pain control and continued locomotion are paramount. Indeed, the majority of the anticancer therapies used to date often result in direct or indirect damage to bone. Thus, although the bone microenvironment regulates tumour cell growth in bone, cells within the bone marrow niche also mediate many of the orthopaedic consequences of tumour progression as well as resistance to the antitumour effects of existing therapies. In this Review, we highlight the effects of existing cancer treatments on bone and the bone marrow microenvironment as well as the mechanisms mediating these effects and the current utility of modern orthopaedic interventions.
Collapse
Affiliation(s)
- Issam Makhoul
- Department of Medicine, Division of Haematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA
| | - Corey O Montgomery
- Department of Orthopaedic Surgery, Centre for Orthopaedic Research, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA
| | - Dana Gaddy
- Department of Physiology and Biophysics, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA
| | - Larry J Suva
- Department of Orthopaedic Surgery, Centre for Orthopaedic Research, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA
| |
Collapse
|
26
|
Alečković M, Kang Y. Bone marrow stroma-derived miRNAs as regulators, biomarkers and therapeutic targets of bone metastasis. BONEKEY REPORTS 2015; 4:671. [PMID: 25908970 PMCID: PMC4398005 DOI: 10.1038/bonekey.2015.38] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 02/19/2015] [Indexed: 12/22/2022]
Abstract
MicroRNAs (miRNAs) are short, endogenous RNA molecules that have essential roles in regulating gene expression. They control numerous physiological and cellular processes, including normal bone organogenesis and homeostasis, by enhancing or inhibiting bone marrow cell growth, differentiation, functional activity and crosstalk of the multiple cell types within the bone. Hence, elucidating miRNA targets in bone marrow stromal cells has revealed novel regulations during bone development and maintenance. Moreover, recent studies have detailed the capacity for bone stromal miRNAs to influence bone metastasis from a number of primary carcinomas by interfering with bone homeostasis or by directly influencing metastatic tumor cells. Owing to the current lack of good diagnostic biomarkers of bone metastases, such changes in bone stromal miRNA expression in the presence of metastatic lesions may become useful biomarkers, and may even serve as therapeutic targets. In particular, cell-free and exosomal miRNAs shed from bone stromal cells into circulation may be developed into novel biomarkers that can be routinely measured in easily accessible samples. Taken together, these findings reveal the significant role of bone marrow stroma-derived miRNAs in the regulation of bone homeostasis and bone metastasis.
Collapse
Affiliation(s)
- Maša Alečković
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| |
Collapse
|
27
|
Abstract
DNA methylation and histone modification are epigenetic mechanisms that result in altered gene expression and cellular phenotype. The exact role of methylation in myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) remains unclear. However, aberrations (e.g. loss-/gain-of-function or up-/down-regulation) in components of epigenetic transcriptional regulation in general, and of the methylation machinery in particular, have been implicated in the pathogenesis of these diseases. In addition, many of these components have been identified as therapeutic targets for patients with MDS/AML, and are also being assessed as potential biomarkers of response or resistance to hypomethylating agents (HMAs). The HMAs 5-azacitidine (AZA) and 2'-deoxy-5-azacitidine (decitabine, DAC) inhibit DNA methylation and have shown significant clinical benefits in patients with myeloid malignancies. Despite being viewed as mechanistically similar drugs, AZA and DAC have differing mechanisms of action. DAC is incorporated 100% into DNA, whereas AZA is incorporated into RNA (80-90%) as well as DNA (10-20%). As such, both drugs inhibit DNA methyltransferases (DNMTs; dependently or independently of DNA replication) resulting in the re-expression of tumor-suppressor genes; however, AZA also has an impact on mRNA and protein metabolism via its inhibition of ribonucleotide reductase, resulting in apoptosis. Herein, we first give an overview of transcriptional regulation, including DNA methylation, post-translational histone-tail modifications, the role of micro-RNA and long-range epigenetic gene silencing. We place special emphasis on epigenetic transcriptional regulation and discuss the implication of various components in the pathogenesis of MDS/AML, their potential as therapeutic targets, and their therapeutic modulation by HMAs and other substances (if known). The main focus of this review is laid on dissecting the rapidly evolving knowledge of AZA and DAC with a special focus on their differing mechanisms of action, and the effect of HMAs on transcriptional regulation.
Collapse
Affiliation(s)
- Lisa Pleyer
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Hospital Salzburg, Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute , Salzburg , Austria
| | | |
Collapse
|