1
|
Krause C, Britsemmer JH, Bernecker M, Molenaar A, Taege N, Lopez-Alcantara N, Geißler C, Kaehler M, Iben K, Judycka A, Wagner J, Wolter S, Mann O, Pfluger P, Cascorbi I, Lehnert H, Stemmer K, Schriever SC, Kirchner H. Liver microRNA transcriptome reveals miR-182 as link between type 2 diabetes and fatty liver disease in obesity. eLife 2024; 12:RP92075. [PMID: 39037913 PMCID: PMC11262792 DOI: 10.7554/elife.92075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024] Open
Abstract
Background The development of obesity-associated comorbidities such as type 2 diabetes (T2D) and hepatic steatosis has been linked to selected microRNAs in individual studies; however, an unbiased genome-wide approach to map T2D induced changes in the miRNAs landscape in human liver samples, and a subsequent robust identification and validation of target genes are still missing. Methods Liver biopsies from age- and gender-matched obese individuals with (n=20) or without (n=20) T2D were used for microRNA microarray analysis. The candidate microRNA and target genes were validated in 85 human liver samples, and subsequently mechanistically characterized in hepatic cells as well as by dietary interventions and hepatic overexpression in mice. Results Here, we present the human hepatic microRNA transcriptome of type 2 diabetes in liver biopsies and use a novel seed prediction tool to robustly identify microRNA target genes, which were then validated in a unique cohort of 85 human livers. Subsequent mouse studies identified a distinct signature of T2D-associated miRNAs, partly conserved in both species. Of those, human-murine miR-182-5 p was the most associated with whole-body glucose homeostasis and hepatic lipid metabolism. Its target gene LRP6 was consistently lower expressed in livers of obese T2D humans and mice as well as under conditions of miR-182-5 p overexpression. Weight loss in obese mice decreased hepatic miR-182-5 p and restored Lrp6 expression and other miR-182-5 p target genes. Hepatic overexpression of miR-182-5 p in mice rapidly decreased LRP6 protein levels and increased liver triglycerides and fasting insulin under obesogenic conditions after only seven days. Conclusions By mapping the hepatic miRNA-transcriptome of type 2 diabetic obese subjects, validating conserved miRNAs in diet-induced mice, and establishing a novel miRNA prediction tool, we provide a robust and unique resource that will pave the way for future studies in the field. As proof of concept, we revealed that the repression of LRP6 by miR-182-5 p, which promotes lipogenesis and impairs glucose homeostasis, provides a novel mechanistic link between T2D and non-alcoholic fatty liver disease, and demonstrate in vivo that miR-182-5 p can serve as a future drug target for the treatment of obesity-driven hepatic steatosis. Funding This work was supported by research funding from the Deutsche Forschungsgemeinschaft (KI 1887/2-1, KI 1887/2-2, KI 1887/3-1 and CRC-TR296), the European Research Council (ERC, CoG Yoyo LepReSens no. 101002247; PTP), the Helmholtz Association (Initiative and Networking Fund International Helmholtz Research School for Diabetes; MB) and the German Center for Diabetes Research (DZD Next Grant 82DZD09D1G).
Collapse
Affiliation(s)
- Christin Krause
- Institute for Human Genetics, Division Epigenetics & Metabolism, University of LübeckLübeckGermany
- Center of Brain, Behaviour and Metabolism (CBBM), University of LübeckLübeckGermany
- German Center for Diabetes Research (DZD)MunichGermany
| | - Jan H Britsemmer
- Institute for Human Genetics, Division Epigenetics & Metabolism, University of LübeckLübeckGermany
- Center of Brain, Behaviour and Metabolism (CBBM), University of LübeckLübeckGermany
- German Center for Diabetes Research (DZD)MunichGermany
| | - Miriam Bernecker
- German Center for Diabetes Research (DZD)MunichGermany
- Research Unit NeuroBiology of Diabetes, Institute for Diabetes and Obesity, Helmholtz CentreMunichGermany
| | - Anna Molenaar
- German Center for Diabetes Research (DZD)MunichGermany
- Research Unit NeuroBiology of Diabetes, Institute for Diabetes and Obesity, Helmholtz CentreMunichGermany
| | - Natalie Taege
- Institute for Human Genetics, Division Epigenetics & Metabolism, University of LübeckLübeckGermany
- Center of Brain, Behaviour and Metabolism (CBBM), University of LübeckLübeckGermany
- German Center for Diabetes Research (DZD)MunichGermany
| | - Nuria Lopez-Alcantara
- Center of Brain, Behaviour and Metabolism (CBBM), University of LübeckLübeckGermany
- Institute for Experimental Endocrinology, University of LübeckLübeckGermany
| | - Cathleen Geißler
- Institute for Human Genetics, Division Epigenetics & Metabolism, University of LübeckLübeckGermany
- Center of Brain, Behaviour and Metabolism (CBBM), University of LübeckLübeckGermany
| | - Meike Kaehler
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus KielKielGermany
| | - Katharina Iben
- Institute for Human Genetics, Division Epigenetics & Metabolism, University of LübeckLübeckGermany
- Center of Brain, Behaviour and Metabolism (CBBM), University of LübeckLübeckGermany
| | - Anna Judycka
- Institute for Human Genetics, Division Epigenetics & Metabolism, University of LübeckLübeckGermany
- Center of Brain, Behaviour and Metabolism (CBBM), University of LübeckLübeckGermany
| | - Jonas Wagner
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-EppendorfHamburgGermany
| | - Stefan Wolter
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-EppendorfHamburgGermany
| | - Oliver Mann
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-EppendorfHamburgGermany
| | - Paul Pfluger
- German Center for Diabetes Research (DZD)MunichGermany
- Research Unit NeuroBiology of Diabetes, Institute for Diabetes and Obesity, Helmholtz CentreMunichGermany
- Chair of Neurobiology of Diabetes, TUM School of Medicine, Technical University of MunichMunichGermany
| | - Ingolf Cascorbi
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus KielKielGermany
| | - Hendrik Lehnert
- Center of Brain, Behaviour and Metabolism (CBBM), University of LübeckLübeckGermany
- German Center for Diabetes Research (DZD)MunichGermany
- University Hospital of Coventry and WarwickshireCoventryUnited Kingdom
| | - Kerstin Stemmer
- German Center for Diabetes Research (DZD)MunichGermany
- Molecular Cell Biology, Institute of Theoretical Medicine, Faculty of Medicine, University of AugsburgAugsburgGermany
| | - Sonja C Schriever
- German Center for Diabetes Research (DZD)MunichGermany
- Research Unit NeuroBiology of Diabetes, Institute for Diabetes and Obesity, Helmholtz CentreMunichGermany
| | - Henriette Kirchner
- Institute for Human Genetics, Division Epigenetics & Metabolism, University of LübeckLübeckGermany
- Center of Brain, Behaviour and Metabolism (CBBM), University of LübeckLübeckGermany
- German Center for Diabetes Research (DZD)MunichGermany
| |
Collapse
|
2
|
Palkina N, Aksenenko M, Zemtsov D, Lavrentev S, Zinchenko I, Belenyuk V, Kirichenko A, Savchenko A, Ruksha T. miR-204-5p in vivo inhibition cause diminished CD45RO cells rate in lungs of melanoma B16-bearing mice. Noncoding RNA Res 2022; 7:133-141. [PMID: 35756165 PMCID: PMC9188961 DOI: 10.1016/j.ncrna.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 10/25/2022] Open
|
3
|
Shen W, Zhou Q, Peng C, Li J, Yuan Q, Zhu H, Zhao M, Jiang X, Liu W, Ren C. FBXW7 and the Hallmarks of Cancer: Underlying Mechanisms and Prospective Strategies. Front Oncol 2022; 12:880077. [PMID: 35515121 PMCID: PMC9063462 DOI: 10.3389/fonc.2022.880077] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/15/2022] [Indexed: 12/13/2022] Open
Abstract
FBXW7, a member of the F-box protein family within the ubiquitin–proteasome system, performs an indispensable role in orchestrating cellular processes through ubiquitination and degradation of its substrates, such as c-MYC, mTOR, MCL-1, Notch, and cyclin E. Mainly functioning as a tumor suppressor, inactivation of FBXW7 induces the aberrations of its downstream pathway, resulting in the occurrence of diseases especially tumorigenesis. Here, we decipher the relationship between FBXW7 and the hallmarks of cancer and discuss the underlying mechanisms. Considering the interplay of cancer hallmarks, we propose several prospective strategies for circumventing the deficits of therapeutic resistance and complete cure of cancer patients.
Collapse
Affiliation(s)
- Wenyue Shen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Quanwei Zhou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chenxi Peng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jiaheng Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qizhi Yuan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hecheng Zhu
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China.,Changsha Kexin Cancer Hospital, Changsha, China
| | - Ming Zhao
- Changsha Kexin Cancer Hospital, Changsha, China
| | - Xingjun Jiang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Weidong Liu
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, School of Basic Medicine, Central South University, Changsha, China
| | - Caiping Ren
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, School of Basic Medicine, Central South University, Changsha, China
| |
Collapse
|
4
|
Moubarak RS, Koetz-Ploch L, Mullokandov G, Gaziel A, de Pablos-Aragoneses A, Argibay D, Kleffman K, Sokolova E, Berwick M, Thomas NE, Osman I, Brown BD, Hernando E. In Vivo miRNA Decoy Screen Reveals miR-124a as a Suppressor of Melanoma Metastasis. Front Oncol 2022; 12:852952. [PMID: 35480113 PMCID: PMC9036958 DOI: 10.3389/fonc.2022.852952] [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: 01/11/2022] [Accepted: 02/24/2022] [Indexed: 11/30/2022] Open
Abstract
Melanoma is a highly prevalent cancer with an increasing incidence worldwide and high metastatic potential. Brain metastasis is a major complication of the disease, as more than 50% of metastatic melanoma patients eventually develop intracranial disease. MicroRNAs (miRNAs) have been found to play an important role in the tumorigenicity of different cancers and have potential as markers of disease outcome. Identification of relevant miRNAs has generally stemmed from miRNA profiling studies of cells or tissues, but these approaches may have missed miRNAs with relevant functions that are expressed in subfractions of cancer cells. We performed an unbiased in vivo screen to identify miRNAs with potential functions as metastasis suppressors using a lentiviral library of miRNA decoys. Notably, we found that a significant fraction of melanomas that metastasized to the brain carried a decoy for miR-124a, a miRNA that is highly expressed in the brain/neurons. Additional loss- and gain-of-function in vivo validation studies confirmed miR-124a as a suppressor of melanoma metastasis and particularly of brain metastasis. miR-124a overexpression did not inhibit tumor growth in vivo, underscoring that miR-124a specifically controls processes required for melanoma metastatic growth, such as seeding and growth post-extravasation. Finally, we provide proof of principle of this miRNA as a promising therapeutic agent by showing its ability to impair metastatic growth of melanoma cells seeded in distal organs. Our efforts shed light on miR-124a as an antimetastatic agent, which could be leveraged therapeutically to impair metastatic growth and improve patient survival.
Collapse
Affiliation(s)
- Rana S. Moubarak
- Department of Pathology, New York University (NYU) School of Medicine, New York, NY, United States
- Interdisciplinary Melanoma Cooperative Group (IMCG), New York University (NYU) Cancer Institute, New York, NY, United States
- Laura and Isaac Perlmutter Cancer Center, New York University (NYU) Langone Health, New York, NY, United States
| | - Lisa Koetz-Ploch
- Department of Pathology, New York University (NYU) School of Medicine, New York, NY, United States
- Interdisciplinary Melanoma Cooperative Group (IMCG), New York University (NYU) Cancer Institute, New York, NY, United States
| | - Gavriel Mullokandov
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Avital Gaziel
- Department of Pathology, New York University (NYU) School of Medicine, New York, NY, United States
- Interdisciplinary Melanoma Cooperative Group (IMCG), New York University (NYU) Cancer Institute, New York, NY, United States
| | - Ana de Pablos-Aragoneses
- Department of Pathology, New York University (NYU) School of Medicine, New York, NY, United States
- Interdisciplinary Melanoma Cooperative Group (IMCG), New York University (NYU) Cancer Institute, New York, NY, United States
| | - Diana Argibay
- Department of Pathology, New York University (NYU) School of Medicine, New York, NY, United States
- Interdisciplinary Melanoma Cooperative Group (IMCG), New York University (NYU) Cancer Institute, New York, NY, United States
| | - Kevin Kleffman
- Department of Pathology, New York University (NYU) School of Medicine, New York, NY, United States
- Interdisciplinary Melanoma Cooperative Group (IMCG), New York University (NYU) Cancer Institute, New York, NY, United States
| | - Elena Sokolova
- Department of Pathology, New York University (NYU) School of Medicine, New York, NY, United States
- Interdisciplinary Melanoma Cooperative Group (IMCG), New York University (NYU) Cancer Institute, New York, NY, United States
| | - Marianne Berwick
- Division of Epidemiology, Biostatistics and Preventive Medicine, Department of Internal Medicine, University of New Mexico, Albuquerque, NM, United States
| | - Nancy E. Thomas
- Department of Dermatology, University of North Carolina, Chapel Hill, NC, United States
| | - Iman Osman
- Interdisciplinary Melanoma Cooperative Group (IMCG), New York University (NYU) Cancer Institute, New York, NY, United States
- Laura and Isaac Perlmutter Cancer Center, New York University (NYU) Langone Health, New York, NY, United States
- Ronald O. Perelman Department of Dermatology, New York University (NYU) School of Medicine, New York, NY, United States
| | - Brian D. Brown
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Eva Hernando
- Department of Pathology, New York University (NYU) School of Medicine, New York, NY, United States
- Interdisciplinary Melanoma Cooperative Group (IMCG), New York University (NYU) Cancer Institute, New York, NY, United States
- Laura and Isaac Perlmutter Cancer Center, New York University (NYU) Langone Health, New York, NY, United States
| |
Collapse
|
5
|
Peng Q, Wang J. Non-coding RNAs in melanoma: Biological functions and potential clinical applications. MOLECULAR THERAPY-ONCOLYTICS 2021; 22:219-231. [PMID: 34514101 PMCID: PMC8424110 DOI: 10.1016/j.omto.2021.05.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Malignant melanoma (MM) is a malignant tumor that originates from melanocytes and has a high mortality rate. Therefore, early diagnosis and treatment are very important for survival. So far, the exact molecular mechanism leading to the occurrence of melanoma, especially the molecular metastatic mechanism, remains largely unknown. Non-coding RNAs (ncRNAs), such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNA (circRNAs), have been investigated and found to play vital roles in regulating tumor occurrence and development, including melanoma. In this review, we summarize the progress of recent research on the effects of ncRNAs on melanoma and attempt to elucidate the role of ncRNAs as molecular markers or potential targets that will provide promising application perspectives on melanoma.
Collapse
Affiliation(s)
- Qiu Peng
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, China
| | - Jia Wang
- Department of Immunology, Changzhi Medical College, Changzhi, Shanxi 046000 China
| |
Collapse
|
6
|
Chen P, Zhang G, Zhou Q, Li Z. Plasma microRNA-720 may predict prognosis and diagnosis in glioma patients. Biosci Rep 2020; 40:BSR20201449. [PMID: 32639004 PMCID: PMC7364510 DOI: 10.1042/bsr20201449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/16/2020] [Accepted: 07/08/2020] [Indexed: 12/18/2022] Open
Abstract
We enrolled 122 patients with glioma who received surgery treatment in our hospital from June 2010 to May 2012, and 60 healthy individuals. We found that the plasma miR-720 in the glioma group was significantly higher than that in the healthy control group (3.19 ± 1.26 vs 0.98 ± 0.65, P<0.001). The sensitivity and specificity were 71.3% (95%CI: 62.4-79.1%) and 83.3% (71.5-91.7%), respectively. The results indicated that the plasma miR-720 level was associated with tumor grade (t = 104.418, P<0.001). The advanced tumor tended to have higher miR-720 expression level. No significant association was found between miR-720 and age, sex, tumor size, KPS and tumor position (P=0.438, 0.514, 0.518, 0.058, 0.226). The multivariate cox analysis indicated that the high expression of miR-720 (HR = 1.48, 95%CI: 1.12-2.97, P=0.023) was independently predictors of adverse prognosis in patients with glioma. The high expression of miR-720 was also associated with recurrence or development in patients with glioma (HR = 1.47, 95%CI: 1.18-3.14, P=0.012). Plasma miR-720 has a moderate diagnostic ability in early diagnostic of glioma and may be a potential tumor biomarker. The high plasma miR-720 was related to adverse prognosis in patients with glioma and could be a prognosis predictor of glioma patients.
Collapse
Affiliation(s)
- Peng Chen
- Department of orthopedic, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Guangying Zhang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Qin Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Zhanzhan Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| |
Collapse
|
7
|
miR-1285-3p Controls Colorectal Cancer Proliferation and Escape from Apoptosis through DAPK2. Int J Mol Sci 2020; 21:ijms21072423. [PMID: 32244500 PMCID: PMC7177834 DOI: 10.3390/ijms21072423] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/23/2020] [Accepted: 03/29/2020] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs are tiny but powerful regulators of gene expression at the post-transcriptional level. Aberrant expression of oncogenic and tumor-suppressor microRNAs has been recognized as a common feature of human cancers. Colorectal cancer represents a major clinical challenge in the developed world and the design of innovative therapeutic approaches relies on the identification of novel biological targets. Here, we perform a functional screening in colorectal cancer cells using a library of locked nucleic acid (LNA)-modified anti-miRs in order to unveil putative oncogenic microRNAs whose inhibition yields a cytotoxic effect. We identify miR-1285-3p and further explore the effect of its targeting in both commercial cell lines and primary colorectal cancer stem cells, finding induction of cell cycle arrest and apoptosis. We show that DAPK2, a known tumor-suppressor, is a novel miR-1285 target and mediates both the anti-proliferative and the pro-apoptotic effects of miR-1285 depletion. Altogether, our findings uncover a novel oncogenic microRNA in colorectal cancer and lay the foundation for further studies aiming at the development of possible therapeutic strategies based on miR-1285 targeting.
Collapse
|
8
|
Mansouri M, Peymani M, Mohamadynejad P. A genetic variant in the flanking region of miR-182 could decrease the susceptibility to the breast cancer risk in the iranian population. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2020; 39:806-817. [PMID: 31994428 DOI: 10.1080/15257770.2019.1704778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Breast cancer is one of the most malignant tumors in the world. It is, in fact, the second leading cause of cancer death in women. Recent research has identified the role of miR-182 in this disease as an oncogene agent. In this study, the association of rs4541843 in the flanking region of the miR-182 sequence with the susceptibility to breast cancer risk has been studied in the Iranian population. By using the PCR_RFLP, the genotype rs4541843 was determined in 161 patients and 164 control subjects. The genotypes of the individuals were analyzed statistically to find the association between rs4541843 and the breast cancer incidence and its pathological characteristics. The results revealed that due to the dominance of the G allele, the frequency of GG + AG genotypes, as compared with AA, had a significant correlation with the incidence of this disease in controls and cases (P = 0.022; OR = 0.523). Moreover, the genotypes AG and AA could significantly decrease the susceptibility to the breast cancer risk; also in the presence of the A allele (OR, 0.565; P = 0.015), the incidence of the disease could be decreased. Our results indicated that this SNP was associated with the breast cancer risk of the Iranian population. We suppose that rs4541843 may influence the processing of the mature miRNA by affecting the cleavage of Drosha. Therefore, this SNP can be considered as a candidate genetic marker for the susceptibility to breast cancer in the Iranian women.
Collapse
Affiliation(s)
- Maryam Mansouri
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Parisa Mohamadynejad
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| |
Collapse
|
9
|
He L, Zhao M, Yu X, Zhao Y, Fu L, Qiao X, Lin H, Zhang Y, Li G, Li S, Lu D. MicroRNA-182-3p negatively regulates cytokines expression by targeting TLR5M in orange-spotted grouper, Epinephelus coioides. FISH & SHELLFISH IMMUNOLOGY 2019; 93:589-596. [PMID: 31351112 DOI: 10.1016/j.fsi.2019.07.063] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/20/2019] [Accepted: 07/24/2019] [Indexed: 06/10/2023]
Abstract
Toll-like receptors (TLRs) as essential pattern recognition receptors in innate immunity, can recognize pathogens and trigger immune response to eliminate invading pathogens. MicroRNAs regulates multiple biological processes by suppressing mRNA translation or resulting in mRNA degradation. MiR-182 has previously been implicated in DNA repair, disease and cancer aspects. The potential role of miR-182-3p in TLR signaling pathway against pathogens is unclear. In this study, we found that the expression of miR-182-3p was up-regulated after Vibrio parahaemolyticus flagellin stimulation in grouper spleen (GS) cells, and negatively correlated with the expression of orange-spotted grouper (Epinephelus coioides) TLR5M (EcTLR5M). Then we found that miR-182-3p could directly target EcTLR5M by using bioinformatic analysis and dual-luciferase reporter assay. Dual-luciferase reporter assay also showed that miR-182-3p down-regulated the wild-type EcTLR5M 3'UTR in luciferase activity rather than the mutant group in HEK 293T cells. We further verified the effect of miR-182-3p on the activation of Nuclear factor-κB (NF-κB) signaling pathway, and found that miR-182-3p inhibitors significantly augmented flagellin-induced NF-κB phosphorylation. Additionally, we also demonstrated that the increased expression of miR-182-3p significantly suppressed the flagellin-induced EcTLR5M, pro-inflammatory cytokines interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) mRNA expression. And the endogenous miR-182-3p knockdown experiments reversely verified the regulatory effect of miR-182-3p. These results suggested that miR-182-3p post-transcriptionally controls EcTLR5M expression and thereby suppresses the expression of pro-inflammatory cytokines. This study is the first to demonstrate that miR-182-3p suppresses pro-inflammatory cytokines expression by regulating the TLR signaling pathway.
Collapse
Affiliation(s)
- Liangge He
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals, Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, P.R. China
| | - Mi Zhao
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals, Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, P.R. China
| | - Xue Yu
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals, Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, P.R. China
| | - Yulin Zhao
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals, Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, P.R. China
| | - Lijun Fu
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals, Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, P.R. China
| | - Xifeng Qiao
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals, Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, P.R. China
| | - Haoran Lin
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals, Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, P.R. China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, P.R. China; College of Ocean, Hainan University, Haikou, 570228, P.R. China
| | - Yong Zhang
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals, Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, P.R. China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, P.R. China; Southern Laboratory of Ocean Science and Engineering (Guangdong, Zhuhai), Zhuhai, 51900, P.R. China
| | - Guangli Li
- Guangdong South China Sea Key Laboratory of Aquaculture for Aquatic Economic Animals, Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, P.R. China
| | - Shuisheng Li
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals, Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, P.R. China; Southern Laboratory of Ocean Science and Engineering (Guangdong, Zhuhai), Zhuhai, 51900, P.R. China.
| | - Danqi Lu
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals, Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, P.R. China; Southern Laboratory of Ocean Science and Engineering (Guangdong, Zhuhai), Zhuhai, 51900, P.R. China.
| |
Collapse
|
10
|
Bertucci A, Kim KH, Kang J, Zuidema JM, Lee SH, Kwon EJ, Kim D, Howell SB, Ricci F, Ruoslahti E, Jang HJ, Sailor MJ. Tumor-Targeting, MicroRNA-Silencing Porous Silicon Nanoparticles for Ovarian Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2019; 11:23926-23937. [PMID: 31251556 DOI: 10.1021/acsami.9b07980] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Silencing of aberrantly expressed microRNAs (miRNAs or miRs) has emerged as one of the strategies for molecular targeted cancer therapeutics. In particular, miR-21 is an oncogenic miRNA overexpressed in many tumors, including ovarian cancer. To achieve efficient administration of anti-miR therapeutics, delivery systems are needed that can ensure local accumulation in the tumor environment, low systemic toxicity, and reduced adverse side effects. In order to develop an improved anti-miR therapeutic agent for the treatment of ovarian cancer, a nanoformulation is engineered that leverages biodegradable porous silicon nanoparticles (pSiNPs) encapsulating an anti-miR-21 locked nucleic acid payload and displaying a tumor-homing peptide for targeted distribution. Targeting efficacy, miR-21 silencing, and anticancer activity are optimized in vitro on a panel of ovarian cancer cell lines, and a formulation of anti-miR-21 in a pSiNP displaying the targeting peptide CGKRK is identified for in vivo evaluation. When this nanoparticulate agent is delivered to mice bearing tumor xenografts, a substantial inhibition of tumor growth is achieved through silencing of miR-21. This study presents the first successful application of tumor-targeted anti-miR porous silicon nanoparticles for the treatment of ovarian cancer in a mouse xenograft model.
Collapse
Affiliation(s)
- Alessandro Bertucci
- Department of Chemical Sciences and Technologies , University of Rome Tor Vergata , Rome , 00133 , Italy
| | | | | | | | | | | | | | | | - Francesco Ricci
- Department of Chemical Sciences and Technologies , University of Rome Tor Vergata , Rome , 00133 , Italy
| | - Erkki Ruoslahti
- Cancer Center , Sanford Burnham Prebys Medical Discovery Institute , La Jolla , California 92037 , United States
| | | | | |
Collapse
|
11
|
Miroshnichenko S, Patutina O. Enhanced Inhibition of Tumorigenesis Using Combinations of miRNA-Targeted Therapeutics. Front Pharmacol 2019; 10:488. [PMID: 31156429 PMCID: PMC6531850 DOI: 10.3389/fphar.2019.00488] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 04/17/2019] [Indexed: 12/18/2022] Open
Abstract
The search for effective strategies to inhibit tumorigenesis remains one of the most relevant scientific challenges. Among the most promising approaches is the direct modulation of the function of short non-coding RNAs, particularly miRNAs. These molecules are propitious targets for anticancer therapy, since they perform key regulatory roles in a variety of signaling cascades related to cell proliferation, apoptosis, migration, and invasion. The development of pathological states is often associated with deregulation of miRNA expression. The present review describes in detail the strategies aimed at modulating miRNA activity that invoke antisense oligonucleotide construction, such as small RNA zippers, miRNases (miRNA-targeted artificial ribonucleases), miRNA sponges, miRNA masks, anti-miRNA oligonucleotides, and synthetic miRNA mimics. The broad impact of developed miRNA-based therapeutics on the various events of tumorigenesis is also discussed. Above all, the focus of this review is to evaluate the results of the combined application of different miRNA-based agents and chemotherapeutic drugs for the inhibition of tumor development. Many studies indicate a considerable increase in the efficacy of anticancer therapy as a result of additive or synergistic effects of simultaneously applied therapies. Different drug combinations, such as a cocktail of antisense oligonucleotides or multipotent miRNA sponges directed at several oncogenic microRNAs belonging to the same/different miRNA families, a mixture of anti-miRNA oligonucleotides and cytostatic drugs, and a combination of synthetic miRNA mimics, have a more complex and profound effect on the various events of tumorigenesis as compared with treatment with a single miRNA-based agent or chemotherapeutic drug. These data provide strong evidence that the simultaneous application of several distinct strategies aimed at suppressing different cellular processes linked to tumorigenesis is a promising approach for cancer therapy.
Collapse
Affiliation(s)
- Svetlana Miroshnichenko
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | - Olga Patutina
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| |
Collapse
|
12
|
Abstract
Stem cells are a rare subpopulation defined by the potential to self-renew and differentiate into specific cell types. A population of stem-like cells has been reported to possess the ability of self-renewal, invasion, metastasis, and engraftment of distant tissues. This unique cell subpopulation has been designated as cancer stem cells (CSC). CSC were first identified in leukemia, and the contributions of CSC to cancer progression have been reported in many different types of cancers. The cancer stem cell hypothesis attempts to explain tumor cell heterogeneity based on the existence of stem cell-like cells within solid tumors. The elimination of CSC is challenging for most human cancer types due to their heightened genetic instability and increased drug resistance. To combat these inherent abilities of CSC, multi-pronged strategies aimed at multiple aspects of CSC biology are increasingly being recognized as essential for a cure. One of the most challenging aspects of cancer biology is overcoming the chemotherapeutic resistance in CSC. Here, we provide an overview of autotaxin (ATX), lysophosphatidic acid (LPA), and their signaling pathways in CSC. Increasing evidence supports the role of ATX and LPA in cancer progression, metastasis, and therapeutic resistance. Several studies have demonstrated the ATX-LPA axis signaling in different cancers. This lipid mediator regulatory system is a novel potential therapeutic target in CSC. In this review, we summarize the evidence linking ATX-LPA signaling to CSC and its impact on cancer progression and metastasis. We also provide evidence for the efficacy of cancer therapy involving the pharmacological inhibition of this signaling pathway.
Collapse
|
13
|
Krichevsky AM, Uhlmann EJ. Oligonucleotide Therapeutics as a New Class of Drugs for Malignant Brain Tumors: Targeting mRNAs, Regulatory RNAs, Mutations, Combinations, and Beyond. Neurotherapeutics 2019; 16:319-347. [PMID: 30644073 PMCID: PMC6554258 DOI: 10.1007/s13311-018-00702-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Malignant brain tumors are rapidly progressive and often fatal owing to resistance to therapies and based on their complex biology, heterogeneity, and isolation from systemic circulation. Glioblastoma is the most common and most aggressive primary brain tumor, has high mortality, and affects both children and adults. Despite significant advances in understanding the pathology, multiple clinical trials employing various treatment strategies have failed. With much expanded knowledge of the GBM genome, epigenome, and transcriptome, the field of neuro-oncology is getting closer to achieve breakthrough-targeted molecular therapies. Current developments of oligonucleotide chemistries for CNS applications make this new class of drugs very attractive for targeting molecular pathways dysregulated in brain tumors and are anticipated to vastly expand the spectrum of currently targetable molecules. In this chapter, we will overview the molecular landscape of malignant gliomas and explore the most prominent molecular targets (mRNAs, miRNAs, lncRNAs, and genomic mutations) that provide opportunities for the development of oligonucleotide therapeutics for this class of neurologic diseases. Because malignant brain tumors focally disrupt the blood-brain barrier, this class of diseases might be also more susceptible to systemic treatments with oligonucleotides than other neurologic disorders and, thus, present an entry point for the oligonucleotide therapeutics to the CNS. Nevertheless, delivery of oligonucleotides remains a crucial part of the treatment strategy. Finally, synthetic gRNAs guiding CRISPR-Cas9 editing technologies have a tremendous potential to further expand the applications of oligonucleotide therapeutics and take them beyond RNA targeting.
Collapse
Affiliation(s)
- Anna M Krichevsky
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Initiative for RNA Medicine, Boston, Massachusetts, 02115, USA.
| | - Erik J Uhlmann
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Initiative for RNA Medicine, Boston, Massachusetts, 02115, USA
| |
Collapse
|
14
|
Fattore L, Mancini R, Ascierto PA, Ciliberto G. The potential of BRAF-associated non-coding RNA as a therapeutic target in melanoma. Expert Opin Ther Targets 2018; 23:53-68. [PMID: 30507327 DOI: 10.1080/14728222.2019.1554057] [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: 02/07/2023]
Abstract
INTRODUCTION The advent of targeted therapies and immune checkpoints inhibitors has enhanced the treatment of metastatic melanomas. Despite striking improvements of patients' survival, drug resistance continues to limit the efficacy of such treatments. Genetic and nongenetic/adaptive mechanisms of resistance could be involved; in the latter mechanism, noncoding RNAs (ncRNAs) are emerging as key players. Areas covered: This article outlines the current knowledge of ncRNA involvement in BRAF-mutant melanomas and the development of resistance to targeted/immunotherapies. We also discuss how ncRNAs can be exploited for the development of therapeutic and diagnostic approaches. Expert opinion: ncRNAs can be envisaged as powerful diagnostics and therapeutics. Despite progress in our knowledge about their deregulation in cancer, it is still difficult to derive universal and robust ncRNAs unique signatures of malignancy for diagnostic purposes, which need validation in large cohort of patients. Also, ncRNA specific targeting to melanoma cells in vivo requires the development of improved systemic delivery tools. In this regard, the development of stable nanodelivery particles seems to offer renewed hope for success in the clinic.
Collapse
Affiliation(s)
- Luigi Fattore
- a IRCCS , Regina Elena National Cancer Institute , Rome , Italy
| | - Rita Mancini
- b Department of Molecular and Clinical Medicine , University of Roma "Sapienza" , Rome , Italy
| | | | | |
Collapse
|
15
|
Ushio N, Rahman MM, Maemura T, Lai YC, Iwanaga T, Kawaguchi H, Miyoshi N, Momoi Y, Miura N. Identification of dysregulated microRNAs in canine malignant melanoma. Oncol Lett 2018; 17:1080-1088. [PMID: 30655868 PMCID: PMC6313064 DOI: 10.3892/ol.2018.9692] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 09/27/2018] [Indexed: 12/29/2022] Open
Abstract
Inhibiting aberrantly upregulated microRNAs (miR/miRNAs) has emerged as a novel focus for therapeutic intervention in human melanoma. Thus, identifying upregulated miRNAs is essential for identifying additional melanoma-associated therapeutic targets. In the present study, microarray-based miRNA profiling of canine malignant melanoma (CMM) tissue obtained from the oral cavity was performed and differential expression was confirmed by a reverse transcription-quantitative polymerase chain reaction (RT-qPCR). An analysis of the microarray data revealed 17 dysregulated miRNAs; 5 were upregulated and 12 were downregulated. RT-qPCR analysis was performed for 2 upregulated (miR-204 and miR-383), 3 downregulated (miR-122, miR-143 and miR-205) and 6 additional oncogenic miRNAs (oncomiRs; miR-16, miR-21, miR-29b, miR-92a, miR-125b and miR-222). The expression levels of seven of the miRNAs, miR-16, miR-21, miR-29b, miR-122, miR-125b, miR-204 and miR-383 were significantly upregulated; however, the expression of miR-205 was downregulated in CMM tissues compared with normal oral tissues. The microarray and RT-qPCR analyses validated the upregulation of two potential oncomiRs miR-204 and miR-383. The present study additionally constructed a protein interaction network and a miRNA-target regulatory interaction network using STRING and Cytoscape. In the proposed network, cyclin dependent kinase 2 was a target for miR-383, sirtuin 1 and tumor protein p53 were targets for miR-204 and ATR serine/threonine kinase was a target for both. It was concluded that miR-383 and miR-204 were potential oncomiRs that may be involved in regulating melanoma development by evading DNA repair and apoptosis.
Collapse
Affiliation(s)
- Norio Ushio
- Department of Clinical Veterinary Science, United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi 753-8511, Japan
| | - Md Mahfuzur Rahman
- Department of Clinical Veterinary Science, United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi 753-8511, Japan
| | - Tadashi Maemura
- Kagoshima University Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima 890-0065, Japan
| | - Yu-Chang Lai
- Department of Clinical Veterinary Science, United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi 753-8511, Japan.,Kagoshima University Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima 890-0065, Japan
| | - Tomoko Iwanaga
- Kagoshima University Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima 890-0065, Japan
| | - Hiroaki Kawaguchi
- Department of Hygiene and Health Promotion Medicine, Graduate School of Medicine and Dental Sciences, Kagoshima University, Korimoto, Kagoshima 890-0065, Japan
| | - Noriaki Miyoshi
- Department of Veterinary Histopathology, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima 890-0065, Japan
| | - Yasuyuki Momoi
- Department of Veterinary Diagnostic Imaging, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima 890-0065, Japan
| | - Naoki Miura
- Department of Clinical Veterinary Science, United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi 753-8511, Japan.,Kagoshima University Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima 890-0065, Japan
| |
Collapse
|
16
|
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: 6] [Impact Index Per Article: 1.0] [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
|
17
|
Chai L, Kang XJ, Sun ZZ, Zeng MF, Yu SR, Ding Y, Liang JQ, Li TT, Zhao J. MiR-497-5p, miR-195-5p and miR-455-3p function as tumor suppressors by targeting hTERT in melanoma A375 cells. Cancer Manag Res 2018; 10:989-1003. [PMID: 29760567 PMCID: PMC5937487 DOI: 10.2147/cmar.s163335] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background hTERT gene plays an important role in melanoma, although the specific mechanism involved is unclear. The aim of this study was to screen and identify the relative miRNAs with the regulation of hTERT in melanoma. Materials and methods Quantitative real-time polymerase chain reaction (q-PCR) and immunohistochemistry were performed to detect hTERT mRNA and protein expression in 36 formalin-fixed paraffin-embedded melanoma tissues and 36 age- and sex-matched pigmented nevi cases, respectively. Bioinformatics analysis and custom miRNA polymerase chain reaction array were determined for predicting, screening and verifying miRNAs with the regulation of the hTERT gene. To investigate the biological functions, miRNAs mimics or inhibitors were transfected into melanoma A375 cells. The relative expression of miR-497-5p, miR-195-5p, miR-455-3p and hTERT mRNA was determined by q-PCR. The protein expression of hTERT was detected by Western blot. 3-(4,5-Dimethylthiazolyl-2-yl)-2,5-biphenyl tetrazolium bromide and flow cytometry were employed to detect cell proliferation ability, cell apoptosis and cell cycle. Transwell and wound healing assays were used to observe cell invasion and migration abilities. A direct target gene of miRNAs was analyzed by a dual luciferase reporter activity assay. Results MiR-497-5p, miR-195-5p, miR-455-3p were significantly downregulated, while hTERT was upregulated in melanoma tissues. hTERT expression level was inversely correlated with miR-497-5p, miR-195-5p and miR-455-3p. Overexpression of miR-497-5p, miR-195-5p and miR-455-3p inhibited A375 cell proliferation, migration and invasion, arrested the cell cycle, induced cell apoptosis and decreased hTERT expression at both mRNA and protein levels. Suppression of miR-497-5p, miR-195-5p and miR-455-3p partially reversed the inhibitory effects. Finally, hTERT was identified as a direct target of miR-497-5p, miR-195-5p and miR-455-3p. Conclusions MiR-497-5p, miR-195-5p and miR-455-3p act as tumor suppressors by targeting hTERT in melanoma A375 cells. Therefore, miR-497-5p, miR-195-5p and miR-455-3p could be potential targeted therapeutic choice for melanoma.
Collapse
Affiliation(s)
- Li Chai
- Xinjiang Medical University, Urumqi, China
| | - Xiao-Jing Kang
- Department of Dermatology and Venereology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Zhen-Zhu Sun
- Department of Pathology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Ming-Feng Zeng
- Department of Dermatology and Venereology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Shi-Rong Yu
- Department of Dermatology and Venereology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Yuan Ding
- Department of Dermatology and Venereology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Jun-Qin Liang
- Department of Dermatology and Venereology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Ting-Ting Li
- Department of Dermatology and Venereology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Juan Zhao
- Department of Dermatology and Venereology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| |
Collapse
|
18
|
Ma Y, Liang AJ, Fan YP, Huang YR, Zhao XM, Sun Y, Chen XF. Dysregulation and functional roles of miR-183-96-182 cluster in cancer cell proliferation, invasion and metastasis. Oncotarget 2018; 7:42805-42825. [PMID: 27081087 PMCID: PMC5173173 DOI: 10.18632/oncotarget.8715] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 03/31/2016] [Indexed: 02/07/2023] Open
Abstract
Previous studies have reported aberrant expression of the miR-183-96-182 cluster in a variety of tumors, which indicates its' diagnostic or prognostic value. However, a key characteristic of the miR-183-96-182 cluster is its varied expression levels, and pleomorphic functional roles in different tumors or under different conditions. In most tumor types, the cluster is highly expressed and promotes tumorigenesis, cancer progression and metastasis; yet tumor suppressive effects have also been reported in some tumors. In the present study, we discuss the upstream regulators and the downstream target genes of miR-183-96-182 cluster, and highlight the dysregulation and functional roles of this cluster in various tumor cells. Newer insights summarized in this review will help readers understand the different facets of the miR-183-96-182 cluster in cancer development and progression.
Collapse
Affiliation(s)
- Yi Ma
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - A-Juan Liang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yu-Ping Fan
- Reproductive Medicine Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi-Ran Huang
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Ming Zhao
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yun Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Xiang-Feng Chen
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China.,Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
19
|
Rodríguez-Cerdeira C, Molares-Vila A, Carnero-Gregorio M, Corbalán-Rivas A. Recent advances in melanoma research via "omics" platforms. J Proteomics 2017; 188:152-166. [PMID: 29138111 DOI: 10.1016/j.jprot.2017.11.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/25/2017] [Accepted: 11/08/2017] [Indexed: 02/09/2023]
Abstract
Melanoma has a high mortality rate and metastatic melanoma is highly resistant to conventional therapies. "Omics" fields such as proteomics and microRNA and exosome studies have provided new knowledge to complement the information generated by genomic studies. This work aimed to review the current status of biomarker discovery for melanoma through multi-"omics" platforms. A few sets of novel microRNAs and proteins are described, some of them with important implications in suppressing melanoma at different stages. Upregulation of genes involved in angiogenesis, immunosuppressive factors, modification of stroma, capture of melanoma cells in lymph nodes and factors responsible for tumour cell recruitment have been identified in exosomes, among molecules with other functions. A remarkable series of proteins involved in epithelial-mesenchymal/mesenchymal-epithelial transitions, inflammation, motility, proliferation and progression processes, centrosome amplification, aneuploidy, inhibition of CD8+ effector T-cells, and metastasis in general were identified. Genomic and protein-protein interactions or metabolome levels were not analysed. Proteomics tools such as Orbitrap shotgun mass spectrometry or deep mining proteomic analysis utilizing high-resolution reversed phase nanoseparation in combination with mass spectrometry are also discussed. The application of these tools together with bioinformatics approaches applied to the clinical setting will enable the implementation of personalized medicine in the near future.
Collapse
Affiliation(s)
- Carmen Rodríguez-Cerdeira
- Efficiency, Quality and Costs in Health Services Research Group (EFISALUD), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Spain; Dermatology Department, Complexo Hospitalario Universitario de Vigo (CHUVI), SERGAS, Vigo, Spain.
| | - Alberto Molares-Vila
- Efficiency, Quality and Costs in Health Services Research Group (EFISALUD), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Spain; Department of Analytical & Food Chemistry, Universidade de Vigo (UVIGO), Spain
| | - Miguel Carnero-Gregorio
- Efficiency, Quality and Costs in Health Services Research Group (EFISALUD), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Spain; Department of Biochemistry, Genetics & Immunology, Universidade de Vigo (UVIGO), Spain
| | - Alberte Corbalán-Rivas
- Nursery Department, Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS, A Coruña, Spain
| |
Collapse
|
20
|
Starkey MP, Compston-Garnett L, Malho P, Dunn K, Dubielzig R. Metastasis-associated microRNA expression in canine uveal melanoma. Vet Comp Oncol 2017; 16:81-89. [PMID: 28512868 DOI: 10.1111/vco.12315] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 03/01/2017] [Accepted: 03/22/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Uveal melanoma (UM) is the most common primary intraocular tumour in dogs. There is no effective means of predicting whether a tumour will metastasize. microRNA (miRNA) metastasis signatures have been identified for several human cancers, including UM. AIMS In this study we investigated whether metastasizing and non-metastasizing canine UMs can be distinguished by miRNA expression levels. MATERIALS AND METHODS miRNA microarray profiling was used to compare miRNA expression in 8 metastasizing and 12 non-metastasizing formalin-fixed, paraffin-embedded (FFPE) primary UM biopsies. RESULTS Fourteen miRNAs exhibited statistically significant differences in expression between the metastasizing and non-metastasizing tumours. Class prediction analysis pinpointed 9 miRNAs which categorized tumours as metastasizing or non-metastasizing with an accuracy of 89%. Of the discriminating miRNAs, 8 were up-regulated in metastasizing UM, and included 3 miRNAs implicated as potential "metastasis activators" in human cutaneous melanoma. The expression of 4 of the miRNAs was subsequently measured using the quantitative reverse transcription polymerase chain reaction (RT-qPCR), and their up-regulation in metastasizing tumours validated. CONCLUSION miRNA expression profiles may potentially be used to identify UMs that will metastasize, and miRNAs that are up-regulated in metastasizing tumours may be targets for therapeutic intervention.
Collapse
Affiliation(s)
- M P Starkey
- Molecular Oncology Group, Animal Health Trust, Newmarket, UK
| | | | - P Malho
- Comparative Ophthalmology Unit, Animal Health Trust, Newmarket, UK
| | - K Dunn
- FOCUS-EyePathLab, Murarrie, Australia
| | - R Dubielzig
- Comparative Ocular Pathology Laboratory, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
21
|
Manne U, Jadhav T, Putcha BDK, Samuel T, Soni S, Shanmugam C, Suswam EA. Molecular Biomarkers of Colorectal Cancer and Cancer Disparities: Current Status and Perspective. CURRENT COLORECTAL CANCER REPORTS 2016. [PMID: 28626361 DOI: 10.1007/s11888-016-0338-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
This review provides updates on the efforts for the development of prognostic and predictive markers in colorectal cancer based on the race/ethnicity of patients. Since the clinical consequences of genetic and molecular alterations differ with patient race and ethnicity, the usefulness of these molecular alterations as biomarkers needs to be evaluated in different racial/ethnic groups. To accomplish personalized patient care, a combined analysis of multiple molecular alterations in DNA, RNA, microRNAs (miRNAs), metabolites, and proteins in a single test is required to assess disease status in a precise way. Therefore, a special emphasis is placed on issues related to utility of recently identified genetic and molecular alterations in genes, miRNAs, and various "-omes" (e.g., proteomes, kinomes, metabolomes, exomes, methylomes) as candidate molecular markers to determine cancer progression (disease recurrence/relapse and metastasis) and to assess the efficacy of therapy in colorectal cancer in relation to patient race and ethnicity. This review will be useful for oncologists, pathologists, and basic and translational researchers.
Collapse
Affiliation(s)
- Upender Manne
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.,Comprehensive Cancer Center, Wallace Tumor Institute, University of Alabama at Birmingham, Room # 420A, 1530 3rd Avenue South, Birmingham, AL 35294, USA
| | - Trafina Jadhav
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.,Wallace Tumor Institute, University of Alabama at Birmingham, Room # 430A, 1530 3rd Avenue South, Birmingham, AL 35294, USA.,Present address: Division of Cardiovascular Medicine, Vanderbilt University, 1215 21st Avenue South, Medical Center East, Suite 5050, Nashville, TN 37232-8802, USA
| | - Balananda-Dhurjati Kumar Putcha
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.,Wallace Tumor Institute, University of Alabama at Birmingham, Room # 430A, 1530 3rd Avenue South, Birmingham, AL 35294, USA.,Present address: 2502 East Woodlands, Saint Joseph, MO 64506, USA
| | - Temesgen Samuel
- Department of Pathobiology, College of Veterinary Medicine, Nursing and Allied Health, Tuskegee University, Tuskegee, AL 36088, USA
| | - Shivani Soni
- Department of Biological Sciences, Alabama State University, Room # 325, Life Science Building, 1627, Hall Street, Montgomery, AL 36104, USA
| | - Chandrakumar Shanmugam
- Wallace Tumor Institute, University of Alabama at Birmingham, Room # 430A, 1530 3rd Avenue South, Birmingham, AL 35294, USA.,Present address: Department of Pathology, ESIC Medical College and Hospital, Sanathnagar, Hyderabad, Telangana 500 038, India
| | - Esther A Suswam
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Pathology, Wallace Tumor Institute, University of Alabama at Birmingham, 1720 2nd Avenue South, # 410C, Birmingham, AL 35294-3300, USA
| |
Collapse
|
22
|
Blaya D, Coll M, Rodrigo-Torres D, Vila-Casadesús M, Altamirano J, Llopis M, Graupera I, Perea L, Aguilar-Bravo B, Díaz A, Banales JM, Clària J, Lozano JJ, Bataller R, Caballería J, Ginès P, Sancho-Bru P. Integrative microRNA profiling in alcoholic hepatitis reveals a role for microRNA-182 in liver injury and inflammation. Gut 2016; 65:1535-45. [PMID: 27196584 DOI: 10.1136/gutjnl-2015-311314] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 04/17/2016] [Indexed: 12/12/2022]
Abstract
OBJECTIVE MicroRNAs (miRNAs) are well-known regulators of disease pathogenesis and have great potential as biomarkers and therapeutic targets. We aimed at profiling miRNAs in alcoholic hepatitis (AH) and identifying miRNAs potentially involved in liver injury. DESIGN MiRNA profiling was performed in liver samples from patients with AH, alcohol liver disease, non-alcoholic steatohepatitis, HCV disease and normal liver tissue. Expression of miRNAs was assessed in liver and serum from patients with AH and animal models. Mimic and decoy miR-182 were used in vitro and in vivo to evaluate miR-182's biological functions. RESULTS MiRNA expression profile in liver was highly altered in AH and distinctive from alcohol-induced cirrhotic livers. Moreover, we identified a set of 18 miRNAs predominantly expressed in AH as compared with other chronic liver conditions. Integrative miRNA-mRNA functional analysis revealed the association of AH-altered miRNAs with nuclear receptors, IGF-1 signalling and cholestasis. Interestingly, miR-182 was the most highly expressed miRNA in AH, which correlated with degree of ductular reaction, disease severity and short-term mortality. MiR-182 mimic induced an upregulation of inflammatory mediators in biliary cells. At experimental level, miR-182 was increased in biliary cells in mice fed with 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) diet but not upregulated by alcohol intake or fibrosis. Inhibition of miR-182 in DDC-fed mice reduced liver damage, bile acid accumulation and inflammatory response. CONCLUSIONS AH is characterised by a deregulated miRNA profile, including miR-182, which is associated with disease severity and liver injury. These results highlight the potential of miRNAs as therapeutic targets and biomarkers in AH.
Collapse
Affiliation(s)
- Delia Blaya
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Mar Coll
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Daniel Rodrigo-Torres
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Maria Vila-Casadesús
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - José Altamirano
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Marta Llopis
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Isabel Graupera
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Luis Perea
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Beatriz Aguilar-Bravo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Alba Díaz
- Department of Pathology, Hospital Clínic, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Jesus M Banales
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute-Donostia University Hospital, University of the Basque Country (UPV/EHU), Ikerbasque, San Sebastian, Spain
| | - Joan Clària
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain Department of Biochemistry and Molecular Genetics, Hospital Clínic and Department of Physiological Sciences I, University of Barcelona, Barcelona, Spain
| | - Juan José Lozano
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Ramon Bataller
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain Division of Gastroenterology and Hepatology, Departments of Medicine and Nutrition, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Juan Caballería
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain Liver Unit, Hospital Clínic, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Pere Ginès
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain Liver Unit, Hospital Clínic, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Pau Sancho-Bru
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| |
Collapse
|
23
|
Abstract
Noncoding RNAs are important regulatory molecules of cellular processes. MicroRNAs (miRNAs) are small noncoding RNAs that bind to complementary sequences in the 3' untranslated region of target mRNAs, leading to degradation of the target mRNAs and/or inhibition of their translation. Some miRNAs are essential for normal animal development; however, many other miRNAs are dispensable for development but play a critical role in pathological conditions, including tumorigenesis and metastasis. miRNA genes often reside at fragile chromosome sites and are deregulated in cancer. Some miRNAs function as oncogenes or tumor suppressors, collectively termed "oncomirs." Specific metastasis-regulating miRNAs, collectively termed "metastamirs," govern molecular processes and pathways in malignant progression in either a tumor cell-autonomous or a cell-nonautonomous manner. Recently, exosome-transferred miRNAs have emerged as mediators of the tumor-stroma cross talk. In this chapter, we focus on the functions, mechanisms of action, and therapeutic potential of miRNAs, particularly oncomirs and metastamirs.
Collapse
Affiliation(s)
- L Ma
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States.
| |
Collapse
|
24
|
Aghagolzadeh P, Radpour R. New trends in molecular and cellular biomarker discovery for colorectal cancer. World J Gastroenterol 2016; 22:5678-5693. [PMID: 27433083 PMCID: PMC4932205 DOI: 10.3748/wjg.v22.i25.5678] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 05/16/2016] [Accepted: 06/15/2016] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer death worldwide, which is consequence of multistep tumorigenesis of several genetic and epigenetic events. Since CRC is mostly asymptomatic until it progresses to advanced stages, the early detection using effective screening approaches, selection of appropriate therapeutic strategies and efficient follow-up programs are essential to reduce CRC mortalities. Biomarker discovery for CRC based on the personalized genotype and clinical information could facilitate the classification of patients with certain types and stages of cancer to tailor preventive and therapeutic approaches. These cancer-related biomarkers should be highly sensitive and specific in a wide range of specimen(s) (including tumor tissues, patients’ fluids or stool). Reliable biomarkers which enable the early detection of CRC, can improve early diagnosis, prognosis, treatment response prediction, and recurrence risk. Advances in our understanding of the natural history of CRC have led to the development of different CRC associated molecular and cellular biomarkers. This review highlights the new trends and approaches in CRC biomarker discovery, which could be potentially used for early diagnosis, development of new therapeutic approaches and follow-up of patients.
Collapse
|
25
|
Sun L, Bai Y, Zhao R, Sun T, Cao R, Wang F, He G, Zhang W, Chen Y, Ye P, Du G. Oncological miR-182-3p, a Novel Smooth Muscle Cell Phenotype Modulator, Evidences From Model Rats and Patients. Arterioscler Thromb Vasc Biol 2016; 36:1386-97. [PMID: 27199451 DOI: 10.1161/atvbaha.115.307412] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 04/21/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Vascular smooth muscle cell (VSMC) phenotype change is a hallmark of vascular remodeling, which contributes to atherosclerotic diseases and can be regulated via microRNA-dependent mechanisms. We recently identified that asymmetrical dimethylarginine positively correlates to vascular remodeling-based diseases. We hypothesized that asymmetrical dimethylarginine induces smooth muscle cell (SMC) phenotypic change via a microRNA-dependent mechanism. APPROACH AND RESULTS Microarray analysis enabled the identification of downregulation of miR-182-3p in asymmetrical dimethylarginine-treated human aortic artery SMCs. The myeloid-associated differentiation marker (MYADM) was identified as the downstream target of miR-182-3p and implicated to contribute to miR-182-3p knockdown-mediated SMC phenotype change, which was evidenced by the increased proliferation and migration and reduced expression levels of phenotype-related genes in human aortic artery SMCs through the ERK/MAP (extracellular signal-regulated kinase/mitogen-activated protein) kinase-dependent mechanism. When inhibiting MYADM in the presence of miR-182-3p inhibitor or overexpressing MYADM in the presence of pre-miR-182-3p, human aortic artery SMCs were reversed to the differentiation phenotype. In vivo, adeno-miR-182-3p markedly suppressed carotid neointimal formation by using balloon-injured rat carotid artery model, specifically via decreased MYADM expression, whereas adeno-miR-182-3p inhibitor significantly promoted neointimal formation. Atherosclerotic lesions from patients with high asymmetrical dimethylarginine plasma levels exhibited decreased miR-182-3p expression levels and elevated MYADM expression levels. CONCLUSIONS miR-182-3p is a novel SMC phenotypic modulator by targeting MYADM.
Collapse
Affiliation(s)
- Lan Sun
- From the State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing, China (Y.B., R.C., P.Y.); Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China (T.S.); and Department of Neurosurgery, PLA General Hospital, Haidian District, Beijing, China (F.W.).
| | - Yongyi Bai
- From the State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing, China (Y.B., R.C., P.Y.); Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China (T.S.); and Department of Neurosurgery, PLA General Hospital, Haidian District, Beijing, China (F.W.)
| | - Rui Zhao
- From the State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing, China (Y.B., R.C., P.Y.); Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China (T.S.); and Department of Neurosurgery, PLA General Hospital, Haidian District, Beijing, China (F.W.)
| | - Tao Sun
- From the State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing, China (Y.B., R.C., P.Y.); Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China (T.S.); and Department of Neurosurgery, PLA General Hospital, Haidian District, Beijing, China (F.W.)
| | - Ruihua Cao
- From the State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing, China (Y.B., R.C., P.Y.); Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China (T.S.); and Department of Neurosurgery, PLA General Hospital, Haidian District, Beijing, China (F.W.)
| | - Fuyu Wang
- From the State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing, China (Y.B., R.C., P.Y.); Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China (T.S.); and Department of Neurosurgery, PLA General Hospital, Haidian District, Beijing, China (F.W.)
| | - Guorong He
- From the State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing, China (Y.B., R.C., P.Y.); Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China (T.S.); and Department of Neurosurgery, PLA General Hospital, Haidian District, Beijing, China (F.W.)
| | - Wen Zhang
- From the State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing, China (Y.B., R.C., P.Y.); Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China (T.S.); and Department of Neurosurgery, PLA General Hospital, Haidian District, Beijing, China (F.W.)
| | - Ying Chen
- From the State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing, China (Y.B., R.C., P.Y.); Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China (T.S.); and Department of Neurosurgery, PLA General Hospital, Haidian District, Beijing, China (F.W.)
| | - Ping Ye
- From the State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing, China (Y.B., R.C., P.Y.); Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China (T.S.); and Department of Neurosurgery, PLA General Hospital, Haidian District, Beijing, China (F.W.).
| | - Guanhua Du
- From the State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China (L.S., R.Z., G.H., W.Z., Y.C., G.D.); Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing, China (Y.B., R.C., P.Y.); Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China (T.S.); and Department of Neurosurgery, PLA General Hospital, Haidian District, Beijing, China (F.W.).
| |
Collapse
|
26
|
Xiao Y, Zhang L, Song Z, Guo C, Zhu J, Li Z, Zhu S. Potential Diagnostic and Prognostic Value of Plasma Circulating MicroRNA-182 in Human Glioma. Med Sci Monit 2016; 22:855-62. [PMID: 26978735 PMCID: PMC4795091 DOI: 10.12659/msm.897164] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Background Previous studies showed the aberrant expression of microRNA-182 (miR-182) in glioma tissue. However, the exact role of circulating miR-182 in glioma remains unclear. Here, we confirmed the expression of plasma circulating miR-182 in glioma patients, and further explored its potential diagnostic and prognostic value. Material/Methods Real-time quantitative PCR (RT-PCR) was used to measure circulating cell-free miR-182 from 112 glioma patients and 54 healthy controls. Results Our findings showed that the level of circulating miR-182 in glioma patients was higher than that in healthy controls (P<0.001), which was significantly associated with KPS score (P=0.025) and WHO grade (P<0.001). The area under the receiver operating characteristic (ROC) curve (AUC) was 0.778. The optimal cut-off value was 1.56, and the sensitivity and specificity were 58.5% and 85.2%, respectively. Interestingly, a high predictive value of circulating miR-182 was observed in high-grade glioma (AUC=0.815). However, the AUC was lower in low-grade glioma (AUC=0.621). Kaplan-Meier analysis demonstrated that the cumulative 5-year overall survival rate in the high miR-182 group was significantly lower than that in the low miR-182 group in both overall survival (OS) (P=0.003) and disease-free survival (DFS) (P=0.006). Moreover, multivariate Cox analysis revealed that circulating miR-182 was an independent prognostic indicator for OS (P=0.034) and DFS (P=0.013). Conclusions These results suggest that circulating miR-182 may be a potential noninvasive biomarker for the diagnosis and prognosis of human glioma.
Collapse
Affiliation(s)
- Yilei Xiao
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Lina Zhang
- Department of Intensive Care Medicine, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| | - Zikun Song
- Department of Intensive Care Medicine, The People's Second Hospital of Liaocheng, Linqing, Shandong, China (mainland)
| | - Chuanjun Guo
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| | - Jianxin Zhu
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| | - Zhongmin Li
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| | - Shugan Zhu
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
27
|
Zhou J, Xu D, Xie H, Tang J, Liu R, Li J, Wang S, Chen X, Su J, Zhou X, Xia K, He Q, Chen J, Xiong W, Cao P, Cao K. miR-33a functions as a tumor suppressor in melanoma by targeting HIF-1α. Cancer Biol Ther 2016; 16:846-55. [PMID: 25891797 DOI: 10.1080/15384047.2015.1030545] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Our previous findings showed that miR-33 expressed abnormally in clinical specimens of melanoma, but the exact molecular mechanism has not been elucidated. OBJECT To determine miR-33's roles in melanoma and confirm whether HIF-1α is a direct target gene of miR-33a. METHODS First miR-33a/b expression levels were detected in HM, WM35, WM451, A375 and SK-MEL-1. Then lentiviral vectors were constructed to intervene miR-33a expression in melanoma cells. Cell proliferation, invasion and metastasis were detected. A375 cells mice model was performed to test the tumorigenesis of melanoma in vivo. Finally the dual reporter gene assay was carried out to confirm whether HIF-1α is a direct target gene of miR-33a. RESULTS MiR-33a/b exhibited a lower expression in WM35, WM451, A375 and SK-MEL-1 of the metastatic skin melanoma cell lines than that in HM. Then inhibition of miR-33a expression in WM35 and WM451 cell lines could promote cell proliferation, invasion and metastasis. Conversely, increased expression of miR-33a in A375 cells could inhibit cellproliferation, invasion and metastasis. In vivo tests also confirmed that overexpression of miR-33a in A375 cells significantly inhibited melanoma tumorigenesis. Finally, we confirmed that HIF-1α is a direct target gene of miR-33a. CONCLUSION The newly identified miR-33a/HIF-1α axis might provide a new strategy for the treatment of melanoma.
Collapse
Affiliation(s)
- Jianda Zhou
- a Department of Plastic Surgery ; Third Xiangya Hospital; Central South University ; Changsha City , China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Van Keuren‐Jensen KR, Malenica I, Courtright AL, Ghaffari LT, Starr AP, Metpally RP, Beecroft TA, Carlson EW, Kiefer JA, Pockros PJ, Rakela J. microRNA changes in liver tissue associated with fibrosis progression in patients with hepatitis C. Liver Int 2016; 36:334-43. [PMID: 26189820 PMCID: PMC5049661 DOI: 10.1111/liv.12919] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 07/12/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Accumulating evidence indicates that microRNAs play a role in a number of disease processes including the pathogenesis of liver fibrosis in hepatitis C infection. Our goal is to add to the accruing information regarding microRNA deregulation in liver fibrosis to increase our understanding of the underlying mechanisms of pathology and progression. METHODS We used next generation sequencing to profile all detectable microRNAs in liver tissue and serum from patients with hepatitis C, stages F1-F4 of fibrosis. RESULTS We found altered expression of several microRNAs, in particular, miR-182, miR199a-5p, miR-200a-5p and miR-183 were found to be significantly upregulated in tissue from liver biopsies of hepatitis C patients with advanced fibrosis, stage F3 and F4, when compared with liver biopsies from patients with early fibrosis, stages F1 and F2. We also found miR-148-5p, miR-1260b, miR-122-3p and miR-378i among the microRNAs most significantly down-regulated from early to advanced fibrosis of the liver. We also sequenced the serum microRNAs; however, we were not able to detect significant changes in circulating microRNAs associated with fibrosis stage after adjusting for multiple tests. CONCLUSIONS Adding measurements of tissue microRNAs acquired during routine biopsies will continue to increase our knowledge of underlying mechanisms of fibrosis. Our goal is that these data, in combination with studies from other researchers and future long-term studies, could be used to enhance the staging accuracy of liver biopsies and expand the surveillance of patients at increased risk for cancer and progression to advanced fibrosis.
Collapse
Affiliation(s)
| | - Ivana Malenica
- NeurogenomicsTranslational Genomics Research InstitutePhoenixAZUSA
| | | | | | - Alex P. Starr
- NeurogenomicsTranslational Genomics Research InstitutePhoenixAZUSA
| | | | | | | | | | | | - Jorge Rakela
- Gastroenterology and HepatologyMayo ClinicScottsdaleAZUSA
| |
Collapse
|
29
|
Cekaite L, Eide PW, Lind GE, Skotheim RI, Lothe RA. MicroRNAs as growth regulators, their function and biomarker status in colorectal cancer. Oncotarget 2016; 7:6476-505. [PMID: 26623728 PMCID: PMC4872728 DOI: 10.18632/oncotarget.6390] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/16/2015] [Indexed: 02/07/2023] Open
Abstract
Gene expression is in part regulated by microRNAs (miRNAs). This review summarizes the current knowledge of miRNAs in colorectal cancer (CRC); their role as growth regulators, the mechanisms that regulate the miRNAs themselves and the potential of miRNAs as biomarkers. Although thousands of tissue samples and bodily fluids from CRC patients have been investigated for biomarker potential of miRNAs (>160 papers presented in a comprehensive tables), none single miRNA nor miRNA expression signatures are in clinical use for this disease. More than 500 miRNA-target pairs have been identified in CRC and we discuss how these regulatory nodes interconnect and affect signaling pathways in CRC progression.
Collapse
Affiliation(s)
- Lina Cekaite
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K.G.Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Peter W. Eide
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K.G.Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Guro E. Lind
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K.G.Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Rolf I. Skotheim
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K.G.Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Ragnhild A. Lothe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K.G.Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
30
|
Mirzaei H, Gholamin S, Shahidsales S, Sahebkar A, Jaafari MR, Mirzaei HR, Hassanian SM, Avan A. MicroRNAs as potential diagnostic and prognostic biomarkers in melanoma. Eur J Cancer 2015; 53:25-32. [PMID: 26693896 DOI: 10.1016/j.ejca.2015.10.009] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 10/08/2015] [Accepted: 10/12/2015] [Indexed: 12/12/2022]
Abstract
Melanoma is a life-threatening malignancy with poor prognosis and a relatively high burden of mortality in advanced stages. The efficacy of current available therapeutic strategies is limited, with a survival rate of less than 10%. Despite rapid advances in biomarker-guided drug development in different tumour types, including melanoma, only a very small number of biomarkers have been identified. Recently, microRNAs (miRNAs) have emerged as a molecular regulator in the development and progression of melanoma. Aberrant activation of some known miRNAs, e.g. let-7a and b, miR-148, miR-155, miR-182, miR-200c, miR-211, miR-214, miR-221 and 222, has been recognised to be linked with melanoma-associated genes such as NRAS, microphthalmia-associated transcription factor, receptor tyrosine kinase c-KIT, AP-2 transcription factor, etc. There is accumulating evidence suggesting the potential impact of circulating miRNAs as diagnostic and therapeutic markers in diseases. In addition, miRNAs have turned out to play important roles in drug-resistance mechanisms; suggesting their modulation as a potential approach to overcome chemoresistance. This review highlights recent preclinical and clinical studies on circulating miRNAs and their potential role as diagnosis, and therapeutic targets in melanoma.
Collapse
Affiliation(s)
- Hamed Mirzaei
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sharareh Gholamin
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Soodabeh Shahidsales
- Cancer Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Biotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Mirzaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mahdi Hassanian
- Biochemistry of Nutrition Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Microanatomy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Molecular Medicine Group, Department of Modern Sciences and Technologies, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Biochemistry of Nutrition Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
31
|
Mannavola F, Tucci M, Felici C, Stucci S, Silvestris F. miRNAs in melanoma: a defined role in tumor progression and metastasis. Expert Rev Clin Immunol 2015; 12:79-89. [PMID: 26505837 DOI: 10.1586/1744666x.2016.1100965] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The crosstalk of melanoma cells with components of the microenvironment promotes malignant cell proliferation and spread to distant tissues. Although the major pathogenetic events have already been elucidated, the mechanisms that drive the metastatic behavior of tumor cells are still undefined. MicroRNAs (miRNAs) are small non-coding RNAs that control post-transcriptional gene expression through interconnected kinases upstream of functional genes involved in tumor progression. Here, we review the biological relevance of melanoma-related miRNAs and focus on their potential role in propagating signals that may cause tumor microenvironment rearrangements, as well as disablement of the immune system and melanoma cell proliferation.
Collapse
Affiliation(s)
- Francesco Mannavola
- a DIMO, Department of Internal Medicine and Clinical Oncology , University of Bari 'Aldo Moro' , Bari , Italy
| | - Marco Tucci
- a DIMO, Department of Internal Medicine and Clinical Oncology , University of Bari 'Aldo Moro' , Bari , Italy
| | - Claudia Felici
- a DIMO, Department of Internal Medicine and Clinical Oncology , University of Bari 'Aldo Moro' , Bari , Italy
| | - Stefania Stucci
- a DIMO, Department of Internal Medicine and Clinical Oncology , University of Bari 'Aldo Moro' , Bari , Italy
| | - Franco Silvestris
- a DIMO, Department of Internal Medicine and Clinical Oncology , University of Bari 'Aldo Moro' , Bari , Italy
| |
Collapse
|
32
|
El Sobky SA, El-Ekiaby NM, Mekky RY, Elemam NM, Mohey Eldin MA, El-Sayed M, Esmat G, Abdelaziz AI. Contradicting roles of miR-182 in both NK cells and their host target hepatocytes in HCV. Immunol Lett 2015; 169:52-60. [PMID: 26518141 DOI: 10.1016/j.imlet.2015.10.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 10/16/2015] [Accepted: 10/21/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIM Natural killer cells are part of the innate immunity involved in viral eradication and were shown to be greatly affected by HCV infection. Epigenetic regulation of NK cell function by microRNAs was not efficiently studied before and was never studied in HCV infection; therefore the aim of this study was to assess for the first time the role of microRNAs in regulating the function of NK cells of HCV-infected patients and hence viral replication in the target HCV-infected Huh7 cells. METHODOLOGY NK cells were isolated from PBMCs of HCV-infected patients as well as controls, and HCV-infected liver biopsies as well as Huh7 cells infected with the virus were used. For the infection of Huh7 cells, first viral vector was in-vitro transcribed into viral RNA that was then used to infect naїve Huh7 cells. Supernatant from the infected cells was then collected and used for further infection. For manipulation of NK cells or Huh7 cells, miR-182 mimics and inhibitors were transfected via lipofection method. RNA was extracted from each cell population, reverse transcribed. Gene expression as well as viral load was quantified using qPCR. RESULTS Screening of NKG2A and NKG2D between patients and controls showed no difference in expression of NKG2A, while NKG2D was found to be downregulated. In view of that, bioinformatics analysis was performed and showed that miR-182 has potential binding sites on both the inhibitory receptor NKG2A and the activating receptor NKG2D, and on its ligand ULBP2, as well as on the viral genome itself. In NK cells of HCV-infected patients, miR-182 was found to be over-expressed compared to controls; its ectopic expression was found to decrease NKG2D mRNA level, while miR-182 inhibitors were able to decrease NKG2A mRNA compared to untransfected cells. In addition, co-culturing genotype 4 or 2 HCV-infected Huh7 cells with miR-182 mimicked NK cells of HCV-infected patients showed decreased viral replication, suggesting an enhanced NK cell function. On the other hand, miR-182 and ULBP2 were both found to be downregulated in HCV liver tissues and HCV-infected Huh7 cells compared to their controls. miR-182 mimics were found to decrease ULBP2 mRNA and increase viral replication in genotypes 4 and 2 HCV-infected target (Huh7) cells compared to controls, while miR-182 inhibitor decreased viral replication in the cell models. CONCLUSION miR-182 was never investigated before, neither in HCV infection nor in NK cells, and we found it to have dysregulated expression in both liver tissues and NK cells of HCV-infected patients compared to control. In addition to that, miR-182 was found to have a contradicting effect in both effector cell and its HCV-infected target cell regarding HCV replication.
Collapse
Affiliation(s)
- Shereen A El Sobky
- Department of Pharmaceutical Biology, German University in Cairo, New Cairo City, Main Entrance Al Tagamoa Al Khames, 11835 Cairo, Egypt
| | - Nada M El-Ekiaby
- Department of Pharmacology & Toxicology, German University in Cairo, New Cairo City, Main Entrance Al Tagamoa Al Khames, 11835 Cairo, Egypt
| | - Radwa Y Mekky
- Department of Pharmacology & Toxicology, German University in Cairo, New Cairo City, Main Entrance Al Tagamoa Al Khames, 11835 Cairo, Egypt
| | - Noha M Elemam
- Department of Pharmacology & Toxicology, German University in Cairo, New Cairo City, Main Entrance Al Tagamoa Al Khames, 11835 Cairo, Egypt
| | - Mohammad A Mohey Eldin
- Department of Tropical Medicine and Hepatology, Cairo University Kasr El-Aini St, Cairo, Egypt
| | - Mohammed El-Sayed
- Department of Tropical Medicine and Hepatology, Cairo University Kasr El-Aini St, Cairo, Egypt
| | - Gamal Esmat
- Department of Tropical Medicine and Hepatology, Cairo University Kasr El-Aini St, Cairo, Egypt
| | - Ahmed I Abdelaziz
- Department of Biology, American University in Cairo New Cairo, Egypt.
| |
Collapse
|
33
|
Wang YH, Scadden DT. Harnessing the apoptotic programs in cancer stem-like cells. EMBO Rep 2015; 16:1084-98. [PMID: 26253117 DOI: 10.15252/embr.201439675] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 06/19/2015] [Indexed: 12/12/2022] Open
Abstract
Elimination of malignant cells is an unmet challenge for most human cancer types even with therapies targeting specific driver mutations. Therefore, a multi-pronged strategy to alter cancer cell biology on multiple levels is increasingly recognized as essential for cancer cure. One such aspect of cancer cell biology is the relative apoptosis resistance of tumor-initiating cells. Here, we provide an overview of the mechanisms affecting the apoptotic process in tumor cells emphasizing the differences in the tumor-initiating or stem-like cells of cancer. Further, we summarize efforts to exploit these differences to design therapies targeting that important cancer cell population.
Collapse
Affiliation(s)
- Ying-Hua Wang
- Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - David T Scadden
- Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| |
Collapse
|
34
|
Zhang W, Qian P, Zhang X, Zhang M, Wang H, Wu M, Kong X, Tan S, Ding K, Perry JK, Wu Z, Cao Y, Lobie PE, Zhu T. Autocrine/Paracrine Human Growth Hormone-stimulated MicroRNA 96-182-183 Cluster Promotes Epithelial-Mesenchymal Transition and Invasion in Breast Cancer. J Biol Chem 2015; 290:13812-29. [PMID: 25873390 PMCID: PMC4447958 DOI: 10.1074/jbc.m115.653261] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 04/11/2015] [Indexed: 12/28/2022] Open
Abstract
Human growth hormone (hGH) plays critical roles in pubertal mammary gland growth, development, and sexual maturation. Accumulated studies have reported that autocrine/paracrine hGH is an orthotopically expressed oncoprotein that promotes normal mammary epithelial cell oncogenic transformation. Autocrine/paracrine hGH has also been reported to promote mammary epithelial cell epithelial-mesenchymal transition (EMT) and invasion. However, the underlying mechanism remains largely obscure. MicroRNAs (miRNAs) are reported to be involved in regulation of multiple cellular functions of cancer. To determine whether autocrine/paracrine hGH promotes EMT and invasion through modulation of miRNA expression, we performed microarray profiling using MCF-7 cells stably expressing wild type or a translation-deficient hGH gene and identified miR-96-182-183 as an autocrine/paracrine hGH-regulated miRNA cluster. Forced expression of miR-96-182-183 conferred on epithelioid MCF-7 cells a mesenchymal phenotype and promoted invasive behavior in vitro and dissemination in vivo. Moreover, we observed that miR-96-182-183 promoted EMT and invasion by directly and simultaneously suppressing BRMS1L (breast cancer metastasis suppressor 1-like) gene expression. miR-96 and miR-182 also targeted GHR, providing a potential negative feedback loop in the hGH-GHR signaling pathway. We further demonstrated that autocrine/paracrine hGH stimulated miR-96-182-183 expression and facilitated EMT and invasion via STAT3 and STAT5 signaling. Consistent with elevated expression of autocrine/paracrine hGH in metastatic breast cancer tissue, miR-96-182-183 expression was also remarkably enhanced. Hence, we delineate the roles of the miRNA-96-182-183 cluster and elucidate a novel hGH-GHR-STAT3/STAT5-miR-96-182-183-BRMS1L-ZEB1/E47-EMT/invasion axis, which provides further understanding of the mechanism of autocrine/paracrine hGH-stimulated EMT and invasion in breast cancer.
Collapse
Affiliation(s)
- Weijie Zhang
- From the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China, the Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
| | - Pengxu Qian
- From the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Xiao Zhang
- From the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Min Zhang
- From the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China, the Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
| | - Hong Wang
- From the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Mingming Wu
- From the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Xiangjun Kong
- From the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Sheng Tan
- From the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Keshuo Ding
- From the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Jo K Perry
- the Liggins Institute and Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland 1023, New Zealand
| | - Zhengsheng Wu
- From the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China, the Department of Pathology, Anhui Medical University, Hefei, Anhui 230027, China, and
| | - Yuan Cao
- the Department of Pathology, Anhui Medical University, Hefei, Anhui 230027, China, and
| | - Peter E Lobie
- the Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore 117599, Singapore
| | - Tao Zhu
- From the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China, the Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China,
| |
Collapse
|
35
|
Li Y, Sun Y, Ren F, Li Y, Liu M, Liu H, Chen J. [Methylation Status of miR-182 Promoter in Lung Cancer Cell Lines]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2015; 18:260-5. [PMID: 25975295 PMCID: PMC6015212 DOI: 10.3779/j.issn.1009-3419.2015.05.02] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
背景与目的 已有的研究证明MiR-182的异常调控与恶性肿瘤的发生发展密切相关,本研究旨在探讨肺癌细胞中miR-182启动子的甲基化状态对miR-182表达的影响。 方法 荧光定量PCR检测肺癌细胞中miR-182表达水平,甲基化特异性PCR检测各细胞株中miR-182启动子区的甲基化状态,并通过测序进行验证。DNA甲基转移酶抑制剂5’-Aza-dC处理后检测各肺癌细胞株中miR-182表达变化。 结果 MiR-182在不同肺癌细胞株的表达水平不同,其中,在高转移性肺癌细胞株如A549和L9981中相对呈低表达;而在低转移性细胞株95C则相对呈高表达。MSP及测序分析显示多株肺癌细胞株中miR-182启动子区域存在DNA甲基化,其中A549细胞甲基化程度最高。在5'-氮杂-脱氧胞苷酸(5’-Aza-dC)作用下,A549细胞及其他肺癌细胞中miR-182表达水平均明显升高。 结论 在肺癌细胞中miR-182启动子区域存在异常甲基化,miR-182的表达受DNA甲基化的调控。MiR-182的甲基化在肺癌中的作用尚需进一步研究。
Collapse
Affiliation(s)
- Yongwen Li
- Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital,
Tianjin 300052, China
| | - Yonglin Sun
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital,
Tianjin 300052, China
| | - Fan Ren
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital,
Tianjin 300052, China
| | - Ying Li
- Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital,
Tianjin 300052, China
| | - Minghui Liu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital,
Tianjin 300052, China
| | - Hongyu Liu
- Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital,
Tianjin 300052, China
| | - Jun Chen
- Tianjin Lung Cancer Institute;Department of Lung Cancer Surgery, Tianjin Medical University General Hospital,
Tianjin 300052, China
| |
Collapse
|
36
|
The miR-200 family and the miR-183~96~182 cluster target Foxf2 to inhibit invasion and metastasis in lung cancers. Oncogene 2015; 35:173-86. [PMID: 25798833 PMCID: PMC4580489 DOI: 10.1038/onc.2015.71] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 01/25/2015] [Accepted: 02/02/2015] [Indexed: 12/12/2022]
Abstract
Metastatic lung cancer is one of the most lethal forms of cancer and molecular pathways driving metastasis are still not clearly elucidated. Metastatic cancer cells undergo an epithelial-mesenchymal transition (EMT) where they lose their epithelial properties and acquire a migratory and invasive phenotype. Here we identify that expression of microRNAs from the miR-200 family and the miR-183~96~182 cluster are significantly co-repressed in non-small cell lung cancer (NSCLC) cell lines and primary tumors from multiple TCGA data sets with high EMT scores. Ectopic expression of the miR-183~96~182 cluster inhibited cancer cell migration and invasion, while its expression was tightly modulated by miR-200. We identified Foxf2 as a common, novel and direct target of both these microRNA families. Foxf2 expression tightly correlates with the transcription factor Zeb1 and is elevated in mesenchymal-like metastatic lung cancer cells. Foxf2 expression induced robust EMT, migration, invasion and metastasis in lung cancer cells, whereas Foxf2 inhibition significantly repressed these phenotypes. We also demonstrated that Foxf2 transcriptionally represses E-Cadherin and miR-200, independent of Zeb1, to form a double negative feedback loop. We therefore identified a novel mechanism whereby the miR-200 family and the miR-183~96~182 cluster inhibit lung cancer invasion and metastasis by targeting Foxf2.
Collapse
|
37
|
Wei Q, Lei R, Hu G. Roles of miR-182 in sensory organ development and cancer. Thorac Cancer 2015; 6:2-9. [PMID: 26273328 PMCID: PMC4448460 DOI: 10.1111/1759-7714.12164] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 08/03/2014] [Indexed: 01/21/2023] Open
Abstract
Micro ribonucleic acids (miRNAs) are a cluster of small non-coding RNA molecules predicted to regulate more than 30% of coding messenger (m)RNAs in the human genome and proven to be essential in developmental and pathological processes. The miR-182 gene was first found to be abundantly expressed in sensory organs and regulates the development of the retina and inner ear. Further studies revealed its roles in osteogenesis and T cell differentiation. In addition, the involvement of miR-182 in cancer initiation and progression has recently been uncovered by a growing body of evidence, the majority of which supports its promoting effects in cell proliferation, angiogenesis, and invasion, as well as distant metastasis of various cancer types. Clinical analyses demonstrated the link of miR-182 expression to poor prognosis in cancer patients. Mechanistically, multiple downstream genes including missing-in-metastasis, microphthalm-associated transcription factor, FoxO1, cylindromatiosis, and others, can be targeted by miR-182 and mediate its roles in cancer. miR-182 is also interconnected with prominent cancer-related signaling pathways, such as transforming growth factor beta and nuclear factor kappa beta. Interestingly, it was shown that in vivo targeting of miR-182 prevented liver metastasis of melanoma. miR-182 is emerging as an important regulator of malignancies, which warrants further study to establish the application potential of miR-182 in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Qing Wei
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine Shanghai, China
| | - Rong Lei
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine Shanghai, China
| | - Guohong Hu
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine Shanghai, China
| |
Collapse
|
38
|
Clinical implications of miRNAs in the pathogenesis, diagnosis and therapy of pancreatic cancer. Adv Drug Deliv Rev 2015; 81:16-33. [PMID: 25453266 DOI: 10.1016/j.addr.2014.10.020] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 10/12/2014] [Accepted: 10/15/2014] [Indexed: 02/06/2023]
Abstract
Despite considerable progress being made in understanding pancreatic cancer (PC) pathogenesis, it still remains the 10th most often diagnosed malignancy in the world and 4th leading cause of cancer related deaths in the United States with a five year survival rate of only 6%. The aggressive nature, lack of early diagnostic and prognostic markers, late clinical presentation, and limited efficacy of existing treatment regimens make PC a lethal cancer with high mortality and poor prognosis. Therefore, novel reliable biomarkers and molecular targets are urgently needed to combat this deadly disease. MicroRNAs (miRNAs) are short (19-24 nucleotides) non-coding RNA molecules implicated in the regulation of gene expression at post-transcriptional level and play significant roles in various physiological and pathological conditions. Aberrant expression of miRNAs has been reported in several cancers including PC and is implicated in PC pathogenesis and progression, suggesting their utility in diagnosis, prognosis and therapy. In this review, we summarize the role of several miRNAs that regulate various oncogenes (KRAS) and tumor suppressor genes (p53, p16, SMAD4, etc.) involved in PC development, their prospective roles as diagnostic and prognostic markers and as a therapeutic targets.
Collapse
|
39
|
Aftab MN, Dinger ME, Perera RJ. The role of microRNAs and long non-coding RNAs in the pathology, diagnosis, and management of melanoma. Arch Biochem Biophys 2014; 563:60-70. [PMID: 25065585 PMCID: PMC4221535 DOI: 10.1016/j.abb.2014.07.022] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 07/14/2014] [Accepted: 07/17/2014] [Indexed: 12/21/2022]
Abstract
Melanoma is frequently lethal and its global incidence is steadily increasing. Despite the rapid development of different modes of targeted treatment, durable clinical responses remain elusive. A complete understanding of the molecular mechanisms that drive melanomagenesis is required, both genetic and epigenetic, in order to improve prevention, diagnosis, and treatment. There is increased appreciation of the role of microRNAs (miRNAs) in melanoma biology, including in proliferation, cell cycle, migration, invasion, and immune evasion. Data are also emerging on the role of long non-coding RNAs (lncRNAs), such as SPRY4-IT1, BANCR, and HOTAIR, in melanomagenesis. Here we review the data on the miRNAs and lncRNAs implicated in melanoma biology. An overview of these studies will be useful for providing insights into mechanisms of melanoma development and the miRNAs and lncRNAs that might be useful biomarkers or future therapeutic targets.
Collapse
Affiliation(s)
- Muhammad Nauman Aftab
- Sanford-Burnham Medical Research Institute, Orlando, FL 32827, USA; Institute of Industrial Biotechnology, Government College University, Katchery Road, Lahore 54000, Pakistan
| | - Marcel E Dinger
- Garvan Institute of Medical Research and St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW 2010, Australia
| | - Ranjan J Perera
- Sanford-Burnham Medical Research Institute, Orlando, FL 32827, USA.
| |
Collapse
|
40
|
Circulating microRNA-182 in plasma and its potential diagnostic and prognostic value for pancreatic cancer. Med Oncol 2014; 31:225. [PMID: 25326859 DOI: 10.1007/s12032-014-0225-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 09/02/2014] [Indexed: 12/30/2022]
Abstract
MicroRNA-182 (miR-182) is overexpressed in several tumors and is found to be associated with adverse clinical characteristics. However, less information on the circulating miR-182 in pancreatic cancer (PCa) is available. The aim of this study was to detect the circulating miR-182 in plasma and to explore its potential diagnostic and prognostic value in PCa. Real-time quantitative PCR was employed to detect circulating miR-182 from 109 PCa and 38 chronic pancreatitis (CP) as well as 50 healthy controls. Our findings revealed that the level of circulating miR-182 in PCa patients was higher than that in CP patients and healthy controls (both at P < 0.05), which was significantly associated with clinical stages (P < 0.001) and lymph node metastasis (P = 0.018). The area under the receiver operating characteristic curve was 0.775, and the optimal cutoff value was 1.63, thus providing a sensitivity of 64.1 % and a specificity of 82.6 %. The diagnosis capability of circulating miR-182 was significantly higher than that of CA19-9, and the combination of two molecules had higher diagnosis capacity (sensitivity of 84.68 % and specificity of 86.77 %). Kaplan-Meier analysis demonstrated that the elevated circulating miR-182 was closely correlated with both shorten overall survival (OS) (P < 0.001) and disease-free survival (DFS) (P < 0.001). Cox analysis indicated that it was an independent prognostic factor for OS and DFS. Our data suggest that circulating miR-182 may be a potential and useful noninvasive tumor marker for diagnosis and prognosis of pancreatic cancer.
Collapse
|
41
|
Li E, Ji P, Ouyang N, Zhang Y, Wang XY, Rubin DC, Davidson NO, Bergamaschi R, Shroyer KR, Burke S, Zhu W, Williams JL. Differential expression of miRNAs in colon cancer between African and Caucasian Americans: implications for cancer racial health disparities. Int J Oncol 2014; 45:587-94. [PMID: 24865442 PMCID: PMC4091964 DOI: 10.3892/ijo.2014.2469] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 03/26/2014] [Indexed: 01/04/2023] Open
Abstract
Colorectal cancer (CRC) incidence and mortality are higher in African Americans (AAs) than in Caucasian Americans (CAs) and microRNAs (miRNAs) have been found to be dysregulated in colonic and other neoplasias. The aim of this exploratory study was to identify candidate miRNAs that could contribute to potential biological differences between AA and CA colon cancers. Total RNA was isolated from tumor and paired adjacent normal colon tissue from 30 AA and 31 CA colon cancer patients archived at Stony Brook University (SBU) and Washington University (WU)‑St. Louis Medical Center. miRNA profiles were determined by probing human genome-wide miRNA arrays with RNA isolated from each sample. Using repeated measures analysis of variance (RANOVA), miRNAs were selected that exhibited significant (p<0.05) interactions between race and tumor or significant (fold change >1.5, p<0.05) main effects of race and/or tumor. Quantitative polymerase chain reaction (q-PCR) was used to confirm miRNAs identified by microarray analysis. Candidate miRNA targets were analyzed using immunohistochemistry. RANOVA results indicated that miR-182, miR152, miR-204, miR-222 and miR-202 exhibited significant race and tumor main effects. Of these miRNAs, q-PCR analysis confirmed that miR-182 was upregulated in AA vs. CA tumors and exhibited significant race:tumor interaction. Immunohistochemical analysis revealed that the levels of FOXO1 and FOXO3A, two potential miR-182 targets, are reduced in AA tumors. miRNAs may play a role in the differences between AA and CA colon cancer. Specifically, differences in miRNA expression levels of miR-182 may contribute to decreased survival in AA colon cancer patients.
Collapse
Affiliation(s)
- Ellen Li
- Division of Gastroenterology, Stony Brook University, Stony Brook, NY 11794-8160, USA
| | - Ping Ji
- Division of Cancer Prevention, Stony Brook University, Stony Brook, NY 11794-8160, USA
| | - Nengtai Ouyang
- Division of Cancer Prevention, Stony Brook University, Stony Brook, NY 11794-8160, USA
| | - Yuanhao Zhang
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794-8160, USA
| | - Xin Yu Wang
- Division of Cancer Prevention, Stony Brook University, Stony Brook, NY 11794-8160, USA
| | - Deborah C Rubin
- Division of Gastroenterology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nicholas O Davidson
- Division of Gastroenterology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Roberto Bergamaschi
- Division of Colon and Rectal Surgery, Stony Brook University, Stony Brook, NY 11794-8160, USA
| | - Kenneth R Shroyer
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794-8160, USA
| | - Stephanie Burke
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794-8160, USA
| | - Wei Zhu
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794-8160, USA
| | - Jennie L Williams
- Division of Cancer Prevention, Stony Brook University, Stony Brook, NY 11794-8160, USA
| |
Collapse
|
42
|
Ferreira R, Santos T, Amar A, Gong A, Chen TC, Tahara SM, Giannotta SL, Hofman FM. Argonaute-2 promotes miR-18a entry in human brain endothelial cells. J Am Heart Assoc 2014; 3:e000968. [PMID: 24837588 PMCID: PMC4309089 DOI: 10.1161/jaha.114.000968] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Cerebral arteriovenous malformation (AVM) is a vascular disease exhibiting abnormal blood vessel morphology and function. miR-18a ameliorates the abnormal characteristics of AVM-derived brain endothelial cells (AVM-BEC) without the use of transfection reagents. Hence, our aim was to identify the mechanisms by which miR-18a is internalized by AVM-BEC. Since AVM-BEC overexpress RNA-binding protein Argonaute-2 (Ago-2) we explored the clinical potential of Ago-2 as a systemic miRNA carrier. METHODS AND RESULTS Primary cultures of AVM-BEC were isolated from surgical specimens and tested for endogenous miR-18a levels using qPCR. Conditioned media (CM) was derived from AVM-BEC cultures (AVM-BEC-CM). AVM-BEC-CM significantly enhanced miR-18a internalization. Ago-2 was detected using western blotting and immunostaining techniques. Ago-2 was highly expressed in AVM-BEC; and siAgo-2 decreased miR-18a entry into brain-derived endothelial cells. Only brain-derived endothelial cells were responsive to the Ago-2/miR-18a complex and not other cell types tested. Secreted products (eg, thrombospondin-1 [TSP-1]) were tested using ELISA. Brain endothelial cells treated with the Ago-2/miR-18a complex in vitro increased TSP-1 secretion. In the in vivo angiogenesis glioma model, animals were treated with miR-18a in combination with Ago-2. Plasma was obtained and tested for TSP-1 and vascular endothelial growth factor (VEGF)-A. In this angiogenesis model, the Ago-2/miR-18a complex caused a significant increase in TSP-1 and decrease in VEGF-A secretion in the plasma. CONCLUSIONS Ago-2 facilitates miR-18a entry into brain endothelial cells in vitro and in vivo. This study highlights the clinical potential of Ago-2 as a miRNA delivery platform for the treatment of brain vascular diseases.
Collapse
Affiliation(s)
- Raquel Ferreira
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA (R.F., A.A., T.C.C., S.L.G., F.M.H.)
| | - Tiago Santos
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA (T.S., A.G., T.C.C., F.M.H.)
| | - Arun Amar
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA (R.F., A.A., T.C.C., S.L.G., F.M.H.)
| | - Alex Gong
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA (T.S., A.G., T.C.C., F.M.H.)
| | - Thomas C Chen
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA (R.F., A.A., T.C.C., S.L.G., F.M.H.) Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA (T.S., A.G., T.C.C., F.M.H.)
| | - Stanley M Tahara
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA (S.M.T.)
| | - Steven L Giannotta
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA (R.F., A.A., T.C.C., S.L.G., F.M.H.)
| | - Florence M Hofman
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA (R.F., A.A., T.C.C., S.L.G., F.M.H.) Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA (T.S., A.G., T.C.C., F.M.H.)
| |
Collapse
|
43
|
Xu X, Ayub B, Liu Z, Serna VA, Qiang W, Liu Y, Hernando E, Zabludoff S, Kurita T, Kong B, Wei JJ. Anti-miR182 reduces ovarian cancer burden, invasion, and metastasis: an in vivo study in orthotopic xenografts of nude mice. Mol Cancer Ther 2014; 13:1729-39. [PMID: 24825857 DOI: 10.1158/1535-7163.mct-13-0982] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
High-grade serous ovarian carcinoma (HGSOC) is a fatal disease, and its grave outcome is largely because of widespread metastasis at the time of diagnosis. Current chemotherapies reduce tumor burden, but they do not provide long-term benefits for patients with cancer. The aggressive tumor growth and metastatic behavior characteristic of these tumors demand novel treatment options such as anti-microRNA treatment, which is emerging as a potential modality for cancer therapy. MicroRNA-182 (miR182) overexpression contributes to aggressive ovarian cancer, largely by its negative regulation of multiple tumor suppressor genes involved in tumor growth, invasion, metastasis, and DNA instability. In this study, we examined the therapeutic potential of anti-miR182 utilizing the animal orthotopic model to mimic human ovarian cancer using ovarian cancer cells SKOV3 (intrabursal xenografts) and OVCAR3 (intraperitoneal injection). These models provide a valuable model system for the investigation of ovarian cancer therapy in vivo. Through a combination of imaging, histological, and molecular analyses, we found that anti-miR182 treatment can significantly reduce tumor burden (size), local invasion, and distant metastasis compared with its control in both models. The bases of anti-miR182 treatment are mainly through the restoration of miR182 target expression, including but not limited to BRCA1, FOXO3a, HMGA2, and MTSS1. Overall, our results strongly suggest that anti-miR182 can potentially be used as a therapeutic modality in treating HGSOC.
Collapse
Affiliation(s)
- Xiaofei Xu
- Authors' Affiliations: Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University; Pathology and
| | | | - Zhaojian Liu
- Institute of Genetics, Shandong University School of Medicine, Jinan, Shandong, China; Departments of Pathology and
| | | | - Wenan Qiang
- Pathology and Obstetrics and Gynecology; Robert H. Lurie Comprehensive Cancer Center, Northwestern University School of Medicine, Chicago, Illinois
| | | | - Eva Hernando
- Department of Pathology, New York University, New York, New York; and
| | | | - Takeshi Kurita
- Obstetrics and Gynecology; Robert H. Lurie Comprehensive Cancer Center, Northwestern University School of Medicine, Chicago, Illinois
| | - Beihua Kong
- Authors' Affiliations: Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University;
| | - Jian-Jun Wei
- Pathology and Obstetrics and Gynecology; Robert H. Lurie Comprehensive Cancer Center, Northwestern University School of Medicine, Chicago, Illinois;
| |
Collapse
|
44
|
Yang MH, Yu J, Jiang DM, Li WL, Wang S, Ding YQ. microRNA-182 targets special AT-rich sequence-binding protein 2 to promote colorectal cancer proliferation and metastasis. J Transl Med 2014; 12:109. [PMID: 24884732 PMCID: PMC4020308 DOI: 10.1186/1479-5876-12-109] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Accepted: 04/25/2014] [Indexed: 02/07/2023] Open
Abstract
Background Increasing evidence has revealed that microRNAs (miRNA) played a pivotal role in regulating cancer cell proliferation and metastasis. The deregulation of miR-182 has been identified in colorectal cancer (CRC). However, the role and mechanism of miR-182 in CRC have not been completely understood yet. Methods The expression levels of miR-182 in CRC tissues and CRC cell lines were examined by performing stem-loop quantitative RT-PCR. The stable over-expression miR-182 cell lines and control cell lines were constructed by lentivirus infection. Subsequently, CCK-8 assay, plate colony formation assay, cell migration, invasion assay and experimental animal models were performed to detect the biological functions of miR-182 in vitro and in vivo. A luciferase reporter assay was conducted to confirm target associations. Western blot and immunohistochemical analysis were performed to examine the expression changes of molecular markers that are regulated by miR-182. Results We found that miR-182 expression is increased in CRC cells that originated from metastatic foci and human primary CRC tissues with lymph node metastases. The ectopic expression of miR-182 enhanced cell proliferation, invasion, and migration in vitro. Stable overexpression of miR-182 also facilitated tumor growth and metastasis in vivo too. Further research showed that miR-182 could directly target the 3’untranslated region (3’UTR) of SATB2 mRNA and subsequently repress both the mRNA and protein expressions of SATB2, which we identified in previous studies as a CRC metastasis-associated protein. Restoring SATB2 expression could reverse the effects of miR-182 on CRC cell proliferation and migration. Investigations of possible mechanisms underlying these behaviors induced by miR-182 revealed that miR-182 induced epithelial-mesenchymal transition (EMT) by modulating the expression of key cellular molecules in EMT. Conclusions Our results illustrated that the up-regulation of miR-182 played a pivotal role in CRC tumorigenesis and metastasis, which suggesting a potential implication of miR-182 in the molecular therapy for CRC.
Collapse
Affiliation(s)
| | | | | | | | - Shuang Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | | |
Collapse
|
45
|
Díaz-López A, Moreno-Bueno G, Cano A. Role of microRNA in epithelial to mesenchymal transition and metastasis and clinical perspectives. Cancer Manag Res 2014; 6:205-16. [PMID: 24812525 PMCID: PMC4008290 DOI: 10.2147/cmar.s38156] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The microRNAs (miRNAs) are a class of small, 20–22 nucleotides in length, endogenously expressed noncoding RNAs that regulate multiple targets posttranscriptionally. Interestingly, miRNAs have emerged as regulators of most physiological and pathological processes, including metastatic tumor progression, in part by controlling a reversible process called epithelial-to-mesenchymal transition (EMT). The activation of EMT increases the migratory and invasive properties fundamental for tumor cell spread while activation of the reverse mesenchymal-to-epithelial transition is required for metastasis outgrowth. The EMT triggering leads to the activation of a core of transcription factors (EMT-TFs) – SNAIL1/SNAIL2, bHLH (E47, E2-2, and TWIST1/TWIST2), and ZEB1/ZEB2 – that act as E-cadherin repressors and, ultimately, coordinate EMT. Recent evidence indicates that several miRNAs regulate the expression of EMT-TFs or EMT-activating signaling pathways. Interestingly, some miRNAs and EMT-TFs form tightly interconnected negative feedback loops that control epithelial cell plasticity, providing self-reinforcing signals and robustness to maintain the epithelial or mesenchymal cell status. Among the most significant feedback loops, we focus on the ZEB/miR-200 and the SNAIL1/miR-34 networks that hold a clear impact in the regulation of the epithelial-mesenchymal state. Recent insights into the p53 modulation of the EMT-TF/miRNA loops and epigenetic regulatory mechanisms in the context of metastasis dissemination will also be discussed. Understanding the regulation of EMT by miRNAs opens new avenues for the diagnosis and prognosis of tumors and identifies potential therapeutic targets that might help to negatively impact on metastasis dissemination and increasing patient survival.
Collapse
Affiliation(s)
- Antonio Díaz-López
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid, Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain
| | - Gema Moreno-Bueno
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid, Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain ; Fundación MDAnderson Internacional, Madrid, Spain
| | - Amparo Cano
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid, Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain
| |
Collapse
|
46
|
Stenvold H, Donnem T, Andersen S, Al-Saad S, Busund LT, Bremnes RM. Stage and tissue-specific prognostic impact of miR-182 in NSCLC. BMC Cancer 2014; 14:138. [PMID: 24575749 PMCID: PMC3996062 DOI: 10.1186/1471-2407-14-138] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 02/12/2014] [Indexed: 11/10/2022] Open
Abstract
Background MicroRNA (miR)-182 is frequently upregulated in cancers, has generally been viewed as an oncogene and is possibly connected to angiogenesis. We aimed to explore what impact miR-182 has in non-small cell lung cancer (NSCLC), and more explicitly its correlation with angiogenic markers. Methods From 335 unselected stage I to IIIA NSCLC carcinomas, duplicate tumor and tumor-associated stromal cores were collected in tissue microarray blocks (TMAs). In situ hybridization (ISH) was used to detect the expression of miR-182 in tumor cells, and immunohistochemistry (IHC) was used to detect the expression of angiogenesis related protein markers. Results In univariate analyses, high tumor cell expression of miR-182 was a positive prognostic factor for patients with squamous cell carcinoma (SCC, P = 0.042) and stage II patients (P = 0.003). Also in the multivariate analysis, high tumor cell miR-182 expression was associated with a good prognosis in the same groups (SCC: HR 0.57, CI 95% 0.33-0.99, P = 0.048; stage II: HR 0.50, CI 95% 0.28-0.90, P = 0.020). We found significant correlations between miR-182 and the angiogenesis related markers FGF2, HIF2α and MMP-7. Conclusion In patients with SCC and in stage II patients, high tumor cell miR-182 expression is an independent positive prognostic factor.
Collapse
Affiliation(s)
- Helge Stenvold
- Institute of Clinical Medicine, University of Tromso, Tromso, Norway.
| | | | | | | | | | | |
Collapse
|
47
|
Vilming Elgaaen B, Olstad OK, Haug KBF, Brusletto B, Sandvik L, Staff AC, Gautvik KM, Davidson B. Global miRNA expression analysis of serous and clear cell ovarian carcinomas identifies differentially expressed miRNAs including miR-200c-3p as a prognostic marker. BMC Cancer 2014; 14:80. [PMID: 24512620 PMCID: PMC3928323 DOI: 10.1186/1471-2407-14-80] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 02/07/2014] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Improved insight into the molecular characteristics of the different ovarian cancer subgroups is needed for developing a more individualized and optimized treatment regimen. The aim of this study was to a) identify differentially expressed miRNAs in high-grade serous ovarian carcinoma (HGSC), clear cell ovarian carcinoma (CCC) and ovarian surface epithelium (OSE), b) evaluate selected miRNAs for association with clinical parameters including survival and c) map miRNA-mRNA interactions. METHODS Differences in miRNA expression between HGSC, CCC and OSE were analyzed by global miRNA expression profiling (Affymetrix GeneChip miRNA 2.0 Arrays, n = 12, 9 and 9, respectively), validated by RT-qPCR (n = 35, 19 and 9, respectively), and evaluated for associations with clinical parameters. For HGSC, differentially expressed miRNAs were linked to differentially expressed mRNAs identified previously. RESULTS Differentially expressed miRNAs (n = 78) between HGSC, CCC and OSE were identified (FDR < 0.01%), of which 18 were validated (p < 0.01) using RT-qPCR in an extended cohort. Compared with OSE, miR-205-5p was the most overexpressed miRNA in HGSC. miR-200 family members and miR-182-5p were the most overexpressed in HGSC and CCC compared with OSE, whereas miR-383 was the most underexpressed. miR-205-5p and miR-200 members target epithelial-mesenchymal transition (EMT) regulators, apparently being important in tumor progression. miR-509-3-5p, miR-509-5p, miR-509-3p and miR-510 were among the strongest differentiators between HGSC and CCC, all being significantly overexpressed in CCC compared with HGSC. High miR-200c-3p expression was associated with poor progression-free (p = 0.031) and overall (p = 0.026) survival in HGSC patients. Interacting miRNA and mRNA targets, including those of a TP53-related pathway presented previously, were identified in HGSC. CONCLUSIONS Several miRNAs differentially expressed between HGSC, CCC and OSE have been identified, suggesting a carcinogenetic role for these miRNAs. miR-200 family members, targeting EMT drivers, were mostly overexpressed in both subgroups, among which miR-200c-3p was associated with survival in HGSC patients. A set of miRNAs differentiates CCC from HGSC, of which miR-509-3-5p and miR-509-5p are the strongest classifiers. Several interactions between miRNAs and mRNAs in HGSC were mapped.
Collapse
Affiliation(s)
- Bente Vilming Elgaaen
- Department of Gynecological Oncology, Oslo University Hospital (OUH), The Norwegian Radium Hospital, Postbox 4953 Nydalen 0424, Oslo, Norway.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Luo C, Weber CEM, Osen W, Bosserhoff AK, Eichmüller SB. The role of microRNAs in melanoma. Eur J Cell Biol 2014; 93:11-22. [PMID: 24602414 DOI: 10.1016/j.ejcb.2014.02.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 01/22/2014] [Accepted: 02/03/2014] [Indexed: 12/21/2022] Open
Abstract
Melanoma is the most dangerous form of skin cancer, being largely resistant to conventional therapies at advanced stages. Understanding the molecular mechanisms behind this disease might be the key for the development of novel therapeutic strategies. MicroRNAs (miRNAs) are small non-coding RNAs that post-transcriptionally control gene expression, thereby regulating various cellular signaling pathways involved in the initiation and progression of different cancer types, including melanoma. In this review, we summarize approaches for the identification of candidate miRNAs and their target genes and review the functions of miRNAs in melanoma. Finally, we highlight the recent progress in pre-clinical use of miRNAs as prognostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Chonglin Luo
- Translational Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Claudia E M Weber
- Translational Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Wolfram Osen
- Translational Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | | | - Stefan B Eichmüller
- Translational Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
49
|
Zhou J, Liu R, Wang Y, Tang J, Tang S, Chen X, Xia K, Xiong W, Xu D, Wang S, He Q, Cao K. miR-199a-5p regulates the expression of metastasis-associated genes in B16F10 melanoma cells. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:7182-90. [PMID: 25400815 PMCID: PMC4230055 DOI: pmid/25400815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 09/10/2014] [Indexed: 02/05/2023]
Abstract
MicroRNAs are regulatory factors that play important roles in tumor development, invasion and metastasis. Previously, we showed that miR-199a is abnormally expressed in clinical melanoma specimens and expression was closely associated with clinical features of metastasis. However, the exact molecular mechanisms by which miR-199a-5p influences melanoma invasion and metastasis remains unclear. In this study, we investigated gene expression changes of metastasis-associated genes in B16F10 melanoma cells following targeted silencing or overexpression of miR-199a-5p, using mouse tumor metastasis PCR arrays. Comparison of gene expression changes in miR-199a-5p-silenced versus overexpressing cells identified a set of upregulated genes (> 2-fold) including Cd44, Cdh1, Cxcr4, Etv4, Fxyd5, Rpsa, Mmp3, Myc, Rb1, Tcf20, Hprt1, Actb1 and downregulated genes (> 2-fold) including Ctsk, Itga7 and Tnfsf10. Regulation of a subset of these genes (Myc, Tnfsf10 and Cd44) following miR-199a-5p silencing or overexpression was validated by reverse transcription-polymerase chain reaction (RT-PCR) and western blot. In conclusion, our study demonstrates that miR-199a-5p regulates melanoma metastasis-related genes, and may provide a basis for the development of novel, molecularly targeted drugs.
Collapse
Affiliation(s)
- Jianda Zhou
- Department of Plastic and Reconstructive Surgery, Third Xiangya Hospital, Central South UniversityChangsha, China
| | - Rui Liu
- Department of Plastic and Reconstructive Surgery, Third Xiangya Hospital, Central South UniversityChangsha, China
| | - Yang Wang
- Department of Plastic and Reconstructive Surgery, Third Xiangya Hospital, Central South UniversityChangsha, China
| | - Jingtian Tang
- Institute of Medical Physics and Engineering, Department of Engineering Physics, Tsinghua UniversityBeijing, China
| | - Shijie Tang
- Cleft Lip and Palate Treatment Center, Second Affiliated Hospital, Shantou University Medical CollegeShantou, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South UniversityChangsha, China
| | - Kun Xia
- State Key Laboratory of Medical Genetics, Central South UniversityChangsha, China
| | - Wei Xiong
- Cancer Research Institute, Key Laboratory of Carcinogenesis of Ministry of Health, Central South UniversityChangsha, China
| | - Dan Xu
- Department of Plastic and Reconstructive Surgery, Third Xiangya Hospital, Central South UniversityChangsha, China
| | - Shaohua Wang
- Department of Plastic and Reconstructive Surgery, Third Xiangya Hospital, Central South UniversityChangsha, China
| | - Quanyong He
- Department of Plastic and Reconstructive Surgery, Third Xiangya Hospital, Central South UniversityChangsha, China
| | - Ke Cao
- Department of Oncology, Third Xiangya Hospital, Central South UniversityChangsha, China
| |
Collapse
|
50
|
Kunz M. Oncogenes in melanoma: an update. Eur J Cell Biol 2013; 93:1-10. [PMID: 24468268 DOI: 10.1016/j.ejcb.2013.12.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 12/16/2013] [Accepted: 12/17/2013] [Indexed: 12/13/2022] Open
Abstract
Melanoma is a highly aggressive tumour with poor prognosis in the metastatic stage. BRAF, NRAS, and KIT are three well-known oncogenes involved in melanoma pathogenesis. Targeting of mutated BRAF kinase has recently been shown to significantly improve overall survival of metastatic melanoma patients, underscoring the particular role of this oncogene in melanoma biology. However, recurrences regularly occur within several months, which supposedly involve further oncogenes. Moreover, oncogenic driver mutations have not been described for up to 30% of all melanomas. In order to obtain a more complete picture of the mutational landscape of melanoma, more recent studies used high-throughput DNA sequencing technologies. A number of new oncogene candidates such as MAPK1/2, ERBB4, GRIN2A, GRM3, RAC1, and PREX2 were identified. Their particular role in melanoma biology is currently under investigation. Evidence for the functional relevance of some of these new oncogene candidates has been provided in in vitro and in vivo experiments. However, these findings await further validation in clinical studies. This review provides an overview on well-known melanoma oncogenes and new oncogene candidates, based on recent high-throughput sequencing studies. The list of genes discussed herein is of course not complete but highlights some of the most significant of recent findings in this area. The new candidates may support more individualized treatment approaches for metastatic melanoma patients in the future.
Collapse
Affiliation(s)
- Manfred Kunz
- Department of Dermatology, Venereology and Allergology, University of Leipzig, 04103 Leipzig, Germany.
| |
Collapse
|