401
|
Role of KRAS-LCS6 polymorphism in advanced NSCLC patients treated with erlotinib or docetaxel in second line treatment (TAILOR). Sci Rep 2015; 5:16331. [PMID: 26573509 PMCID: PMC4648064 DOI: 10.1038/srep16331] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 10/13/2015] [Indexed: 11/10/2022] Open
Abstract
MicroRNAs were described to target mRNA and regulate the transcription of genes involved in processes de-regulated in tumorigenesis, such as proliferation, differentiation and survival. In particular, the miRNA let-7 has been suggested to regulate the expression of the KRAS gene, a common mutated gene in non-small cell lung cancer (NSCLC), through a let-7 complementary site (LCS) in 3′UTR of KRAS mRNA. We have reported the analysis performed on the role of the polymorphism located in the KRAS-LCS (rs61764370) which is involved in the disruption of the let-7 complementary site in NSCLC patients enrolled within the TAILOR trial, a randomised trial comparing erlotinib versus docetaxel in second line treatment. In our cohort of patients, KRAS-LCS6 polymorphism did not have any impact on both overall survival (OS) and progression free survival (PFS) and was not associated with any patient’s baseline characteristics included in the study. Overall, patients had a better prognosis when treated with docetaxel instead of erlotinib for both OS and PFS. Considering KRAS-LCS6 status, the TG/GG patients had a benefit from docetaxel treatment (HR(docetaxel vs erlotinib) = 0.35, 95% CI 0.15–0.79, p = 0.011) compared with the TT patients (HR(docetaxel vs erlotinib) = 0.72, 95% CI 0.52–1.01, p = 0.056) in terms of PFS.
Collapse
|
402
|
Naidu S, Garofalo M. microRNAs: An Emerging Paradigm in Lung Cancer Chemoresistance. Front Med (Lausanne) 2015; 2:77. [PMID: 26583081 PMCID: PMC4631988 DOI: 10.3389/fmed.2015.00077] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 10/19/2015] [Indexed: 12/18/2022] Open
Abstract
Lung cancer is considered the most deadly of all cancers, with limited therapeutic options. Although advanced drugs have been tried in clinic, the therapeutic success has largely been hampered due to rapid development of drug-resistance mechanisms. Recently, microRNAs (miRNAs), a class of small non-coding RNAs, have occupied center stage in cancer biology. miRNAs negatively regulate gene expression either by promoting degradation or by interfering with translation of messenger RNA targets. Several lines of evidence have confirmed the crucial role of miRNAs in carcinogenesis, and, importantly, in the acquisition of resistance to chemotherapeutics. Modulation of miRNA expression levels has been proven to increase the efficacy of genotoxic drugs in various preclinical cancer studies. Therefore, comprehensive understanding of the role(s) of these key players in drug resistance may provide novel opportunities to design effective combinatorial therapeutic strategies for cancer treatment. In this review, we highlight recent findings on miRNAs acting as oncomiRs and tumor suppressor genes in lung cancer. Moreover, we discuss the involvement of miRNAs in different mechanisms of drug resistance in this deadly disease.
Collapse
Affiliation(s)
- Srivatsava Naidu
- Transcriptional Networks in Lung Cancer Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| | - Michela Garofalo
- Transcriptional Networks in Lung Cancer Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| |
Collapse
|
403
|
Garcia-Rendueles MER, Ricarte-Filho JC, Untch BR, Landa I, Knauf JA, Voza F, Smith VE, Ganly I, Taylor BS, Persaud Y, Oler G, Fang Y, Jhanwar SC, Viale A, Heguy A, Huberman KH, Giancotti F, Ghossein R, Fagin JA. NF2 Loss Promotes Oncogenic RAS-Induced Thyroid Cancers via YAP-Dependent Transactivation of RAS Proteins and Sensitizes Them to MEK Inhibition. Cancer Discov 2015; 5:1178-93. [PMID: 26359368 PMCID: PMC4642441 DOI: 10.1158/2159-8290.cd-15-0330] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 09/08/2015] [Indexed: 11/16/2022]
Abstract
UNLABELLED Ch22q LOH is preferentially associated with RAS mutations in papillary and in poorly differentiated thyroid cancer (PDTC). The 22q tumor suppressor NF2, encoding merlin, is implicated in this interaction because of its frequent loss of function in human thyroid cancer cell lines. Nf2 deletion or Hras mutation is insufficient for transformation, whereas their combined disruption leads to murine PDTC with increased MAPK signaling. Merlin loss induces RAS signaling in part through inactivation of Hippo, which activates a YAP-TEAD transcriptional program. We find that the three RAS genes are themselves YAP-TEAD1 transcriptional targets, providing a novel mechanism of promotion of RAS-induced tumorigenesis. Moreover, pharmacologic disruption of YAP-TEAD with verteporfin blocks RAS transcription and signaling and inhibits cell growth. The increased MAPK output generated by NF2 loss in RAS-mutant cancers may inform therapeutic strategies, as it generates greater dependency on the MAPK pathway for viability. SIGNIFICANCE Intensification of mutant RAS signaling through copy-number imbalances is commonly associated with transformation. We show that NF2/merlin inactivation augments mutant RAS signaling by promoting YAP/TEAD-driven transcription of oncogenic and wild-type RAS, resulting in greater MAPK output and increased sensitivity to MEK inhibitors.
Collapse
MESH Headings
- Animals
- Binding Sites
- Cell Cycle Proteins
- Cell Line, Tumor
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Chromosome Deletion
- Chromosomes, Human, Pair 22
- DNA Copy Number Variations
- Disease Models, Animal
- Drug Resistance, Neoplasm/genetics
- Gene Deletion
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Order
- Gene Targeting
- Genes, ras
- Humans
- Mice
- Mice, Transgenic
- Mitogen-Activated Protein Kinases/antagonists & inhibitors
- Models, Biological
- Neoplasm Staging
- Neurofibromin 2/genetics
- Nuclear Proteins/metabolism
- Nucleotide Motifs
- Position-Specific Scoring Matrices
- Promoter Regions, Genetic
- Protein Binding
- Protein Kinase Inhibitors/pharmacology
- Signal Transduction/drug effects
- Thyroid Neoplasms/drug therapy
- Thyroid Neoplasms/genetics
- Thyroid Neoplasms/metabolism
- Thyroid Neoplasms/pathology
- Transcription Factors/metabolism
- Transcriptional Activation
Collapse
Affiliation(s)
| | - Julio C Ricarte-Filho
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brian R Untch
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Iňigo Landa
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jeffrey A Knauf
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Francesca Voza
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vicki E Smith
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ian Ganly
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Barry S Taylor
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yogindra Persaud
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gisele Oler
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yuqiang Fang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Suresh C Jhanwar
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Agnes Viale
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Adriana Heguy
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kety H Huberman
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Filippo Giancotti
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Ronald Ghossein
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - James A Fagin
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York. Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Medicine, Weill Cornell Medical College, New York, New York.
| |
Collapse
|
404
|
Non-coding RNA: a new tool for the diagnosis, prognosis, and therapy of small cell lung cancer. J Thorac Oncol 2015; 10:28-37. [PMID: 25654726 DOI: 10.1097/jto.0000000000000394] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In recent years, novel classes of noncoding RNAs (ncRNAs) have been discovered, which are implicated in diverse functional and regulatory activities. Growing evidence indicates that deregulated ncRNAs play crucial roles in the onset and progression of cancer, including small-cell lung cancer. In this review, we highlight nearly all of the findings regarding the roles and the possible mechanisms of ncRNAs as oncogenes or tumor suppressors in small-cell lung cancer. Furthermore, we discuss the possible role of ncRNAs as diagnostic biomarkers, their significant contribution to the prognosis, and their functions in regulating the response to therapy.
Collapse
|
405
|
Role of Exosomal Noncoding RNAs in Lung Carcinogenesis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:125807. [PMID: 26583084 PMCID: PMC4637011 DOI: 10.1155/2015/125807] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 07/05/2015] [Accepted: 07/16/2015] [Indexed: 12/28/2022]
Abstract
Lung cancer is the major cause of cancer death worldwide. Novel, recently discovered classes of noncoding RNAs (ncRNAs) have diverse functional and regulatory activities and increasing evidence suggests crucial roles for deregulated ncRNAs in the onset and progression of cancer, including lung cancer. Exosomes are small extracellular membrane vesicles of endocytic origin that are released by many cells and are found in most body fluids. Tumor-derived exosomes mediate tumorigenesis by facilitating tumor growth and metastasis. MicroRNAs (miRNAs) are a subclass of ncRNAs that are present in exosomes. miRNAs are taken up by neighboring or distant cells and modulate various functions of recipient cells. Here, we review exosome-derived ncRNAs with a focus on miRNAs and their role in lung cancer biology.
Collapse
|
406
|
Nannini M, Ravegnini G, Angelini S, Astolfi A, Biasco G, Pantaleo MA. miRNA profiling in gastrointestinal stromal tumors: implication as diagnostic and prognostic markers. Epigenomics 2015; 7:1033-49. [PMID: 26447534 DOI: 10.2217/epi.15.52] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs are a class of short noncoding RNAs, that play a relevant role in multiple biological processes, such as differentiation, proliferation and apoptosis. Gastrointestinal stromal tumors (GIST) are considered as a paradigm of molecular biology in solid tumors worldwide, and after the discovery of specific alterations in the KIT and PDGFRA genes, they have emerged from anonymity to become a model for targeted therapy. Epigenetics have an emerging and relevant role in different steps of GIST biology such as tumorigenesis, disease progression, prognosis and drug resistance. The aim of the present review was to summarize the current evidence about the role of microRNAs in GIST, including their potential application as well as their limits.
Collapse
Affiliation(s)
- Margherita Nannini
- Department of Specialized, Experimental & Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Gloria Ravegnini
- Department of Pharmacy & Biotechnology, FaBit; University of Bologna, Italy
| | - Sabrina Angelini
- Department of Pharmacy & Biotechnology, FaBit; University of Bologna, Italy
| | - Annalisa Astolfi
- "Giorgio Prodi" Cancer Research Center, University of Bologna, Bologna, Italy
| | - Guido Biasco
- Department of Specialized, Experimental & Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy.,"Giorgio Prodi" Cancer Research Center, University of Bologna, Bologna, Italy
| | - Maria A Pantaleo
- Department of Specialized, Experimental & Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy.,"Giorgio Prodi" Cancer Research Center, University of Bologna, Bologna, Italy
| |
Collapse
|
407
|
Wu L, Nguyen LH, Zhou K, de Soysa TY, Li L, Miller JB, Tian J, Locker J, Zhang S, Shinoda G, Seligson MT, Zeitels LR, Acharya A, Wang SC, Mendell JT, He X, Nishino J, Morrison SJ, Siegwart DJ, Daley GQ, Shyh-Chang N, Zhu H. Precise let-7 expression levels balance organ regeneration against tumor suppression. eLife 2015; 4:e09431. [PMID: 26445246 PMCID: PMC4716837 DOI: 10.7554/elife.09431] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/05/2015] [Indexed: 02/06/2023] Open
Abstract
The in vivo roles for even the most intensely studied microRNAs remain poorly defined. Here, analysis of mouse models revealed that let-7, a large and ancient microRNA family, performs tumor suppressive roles at the expense of regeneration. Too little or too much let-7 resulted in compromised protection against cancer or tissue damage, respectively. Modest let-7 overexpression abrogated MYC-driven liver cancer by antagonizing multiple let-7 sensitive oncogenes. However, the same level of overexpression blocked liver regeneration, while let-7 deletion enhanced it, demonstrating that distinct let-7 levels can mediate desirable phenotypes. let-7 dependent regeneration phenotypes resulted from influences on the insulin-PI3K-mTOR pathway. We found that chronic high-dose let-7 overexpression caused liver damage and degeneration, paradoxically leading to tumorigenesis. These dose-dependent roles for let-7 in tissue repair and tumorigenesis rationalize the tight regulation of this microRNA in development, and have important implications for let-7 based therapeutics. DOI:http://dx.doi.org/10.7554/eLife.09431.001 The development of animals is guided by the expression of certain genes at critical moments. Many different mechanisms control development; in one of them, the expression of genes can be decreased by molecules called microRNAs. In particular, the group of microRNAs called let-7 has been intensively studied in roundworms and fruit flies. Although mammals have extremely similar let-7 microRNAs they seem to be more important during adulthood. Previous studies using cells grown in the laboratory have shown that mammalian let-7 microRNAs decrease cell proliferation and cell growth. Furthermore, in mouse models of various cancers, let-7 microRNAs often reduce tumour growth when they are supplied to adult mice. Therefore, overall the let-7 group has been classified as genes that act to suppress tumors, and thus protect mice (and most likely humans too) from cancers. However, in-depth analysis of let-7 microRNAs was still missing. Wu and Nguyen et al. have now studied mice with liver cancer using strains where they were able to regulate the levels of let-7. These mice overproduce a strong cancer-inducing gene in the liver; half were used as controls and the other half were further engineered to have moderately elevated levels of let-7 expression. Most of the control mice got large cancerous tumors, but only a few mice in the other group developed cancers and the tumors were smaller. This confirmed that let-7 hinders tumor formation. Wu and Nguyen et al. also observed that the protected mice were less able to regenerate their liver tissues. Further experiments showed that deleting just two out of ten let-7 microRNAs enhanced the mice’s ability to regenerate liver tissue after injury. These findings indicate that let-7 microRNAs slow down the growth of both cancerous and normal cells. Lastly, when let-7 levels were raised to very high levels for a prolonged amount of time this actually led to liver damage and subsequent tumor formation. This last observation may have important consequences for possible cancer therapies. Some scientists have shown that providing extra let-7 can slow or even reverse tumour growth, but the findings here clearly point out that too much let-7 could actually worsen the situation. Since the let-7 family comprises a handful of microRNAs in mammals, in the future it will also be important to find out to what extent these molecules play overlapping roles and how much they differ. DOI:http://dx.doi.org/10.7554/eLife.09431.002
Collapse
Affiliation(s)
- Linwei Wu
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States.,Organ Transplant Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Liem H Nguyen
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States.,Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Kejin Zhou
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States
| | - T Yvanka de Soysa
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, United States.,Harvard Stem Cell Institute, Harvard University, Boston, United States.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, United States.,The Manton Center for Orphan Disease Research, Howard Hughes Medical Institute, Boston Children's Hospital, Boston, United States
| | - Lin Li
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States.,Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Jason B Miller
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States
| | - Jianmin Tian
- Department of Pathology, University of Pittsburg, Pittsburg, United States
| | - Joseph Locker
- Department of Pathology, University of Pittsburg, Pittsburg, United States
| | - Shuyuan Zhang
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States.,Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Gen Shinoda
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, United States.,Harvard Stem Cell Institute, Harvard University, Boston, United States.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, United States.,The Manton Center for Orphan Disease Research, Howard Hughes Medical Institute, Boston Children's Hospital, Boston, United States
| | - Marc T Seligson
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, United States.,Harvard Stem Cell Institute, Harvard University, Boston, United States.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, United States.,The Manton Center for Orphan Disease Research, Howard Hughes Medical Institute, Boston Children's Hospital, Boston, United States
| | - Lauren R Zeitels
- Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Asha Acharya
- Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Sam C Wang
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States.,Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Surgery, University of Texas Southwestern Medical Center, Dallas, United States
| | - Joshua T Mendell
- Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Xiaoshun He
- Organ Transplant Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jinsuke Nishino
- Howard Hughes Medical Institute, Children's Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, United States
| | - Sean J Morrison
- Howard Hughes Medical Institute, Children's Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, United States
| | - Daniel J Siegwart
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States
| | - George Q Daley
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, United States.,Harvard Stem Cell Institute, Harvard University, Boston, United States.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, United States.,The Manton Center for Orphan Disease Research, Howard Hughes Medical Institute, Boston Children's Hospital, Boston, United States
| | - Ng Shyh-Chang
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, United States.,Harvard Stem Cell Institute, Harvard University, Boston, United States.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, United States.,The Manton Center for Orphan Disease Research, Howard Hughes Medical Institute, Boston Children's Hospital, Boston, United States.,Stem cell and Regenerative Biology, Genome Institute of Singapore, Singapore, Singapore
| | - Hao Zhu
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States.,Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
408
|
Liu CY, Stücker I, Chen C, Goodman G, McHugh MK, D'Amelio AM, Etzel CJ, Li S, Lin X, Christiani DC. Genome-wide Gene-Asbestos Exposure Interaction Association Study Identifies a Common Susceptibility Variant on 22q13.31 Associated with Lung Cancer Risk. Cancer Epidemiol Biomarkers Prev 2015; 24:1564-73. [PMID: 26199339 PMCID: PMC4592421 DOI: 10.1158/1055-9965.epi-15-0021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 06/30/2015] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Occupational asbestos exposure has been found to increase lung cancer risk in epidemiologic studies. METHODS We conducted an asbestos exposure-gene interaction analyses among several Caucasian populations who were current or ex-smokers. The discovery phase included 833 Caucasian cases and 739 Caucasian controls, and used a genome-wide association study (GWAS) to identify single-nucleotide polymorphisms (SNP) with gene-asbestos interaction effects. The top ranked SNPs from the discovery phase were replicated within the International Lung and Cancer Consortium (ILCCO). First, in silico replication was conducted in those groups that had GWAS and asbestos exposure data, including 1,548 cases and 1,527 controls. This step was followed by de novo genotyping to replicate the results from the in silico replication, and included 1,539 cases and 1,761 controls. Multiple logistic regression was used to assess the SNP-asbestos exposure interaction effects on lung cancer risk. RESULTS We observed significantly increased lung cancer risk among MIRLET7BHG (MIRLET7B host gene located at 22q13.31) polymorphisms rs13053856, rs11090910, rs11703832, and rs12170325 heterozygous and homozygous variant allele(s) carriers (P < 5 × 10(-7) by likelihood ratio test; df = 1). Among the heterozygous and homozygous variant allele(s) carriers of polymorphisms rs13053856, rs11090910, rs11703832, and rs12170325, each unit increase in the natural log-transformed asbestos exposure score was associated with age-, sex-, smoking status, and center-adjusted ORs of 1.34 [95% confidence interval (CI), 1.18-1.51], 1.24 (95% CI, 1.14-1.35), 1.28 (95% CI, 1.17-1.40), and 1.26 (95% CI, 1.15-1.38), respectively, for lung cancer risk. CONCLUSION Our findings suggest that MIRLET7BHG polymorphisms may be important predictive markers for asbestos exposure-related lung cancer. IMPACT To our knowledge, our study is the first report using a systematic genome-wide analysis in combination with detailed asbestos exposure data and replication to evaluate asbestos-associated lung cancer risk.
Collapse
Affiliation(s)
- Chen-Yu Liu
- Environmental and Occupational Medicine and Epidemiology Program, Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts. Institute of Environmental Health, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Isabelle Stücker
- Institut National de la Santé et de la Recherche Médicale Unit U754, Villejuif, France
| | - Chu Chen
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington. Department of Epidemiology, School of Public Health, University of Washington, Seattle, Washington. Department of Otolaryngology, Head and Neck Surgery, School of Medicine, University of Washington, Seattle, Washington
| | - Gary Goodman
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Michelle K McHugh
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anthony M D'Amelio
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Biomath/Biostatistics Program, The University of Texas Graduate School for Biomedical Sciences, Houston, Texas
| | - Carol J Etzel
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Su Li
- Environmental and Occupational Medicine and Epidemiology Program, Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts
| | - Xihong Lin
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts
| | - David C Christiani
- Environmental and Occupational Medicine and Epidemiology Program, Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts. Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
409
|
Sanders AP, Burris HH, Just AC, Motta V, Amarasiriwardena C, Svensson K, Oken E, Solano-Gonzalez M, Mercado-Garcia A, Pantic I, Schwartz J, Tellez-Rojo MM, Baccarelli AA, Wright RO. Altered miRNA expression in the cervix during pregnancy associated with lead and mercury exposure. Epigenomics 2015; 7:885-96. [PMID: 26418635 DOI: 10.2217/epi.15.54] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
AIM Toxic metals including lead and mercury are associated with adverse pregnancy outcomes. This study aimed to assess the association between miRNA expression in the cervix during pregnancy with lead and mercury levels. MATERIALS & METHODS We obtained cervical swabs from pregnant women (n = 60) and quantified cervical miRNA expression. Women's blood lead, bone lead and toenail mercury levels were analyzed. We performed linear regression to examine the association between metal levels and expression of 74 miRNAs adjusting for covariates. RESULTS Seventeen miRNAs were negatively associated with toenail mercury levels, and tibial bone lead levels were associated with decreased expression of miR-575 and miR-4286. CONCLUSION The findings highlight miRNAs in the human cervix as novel responders to maternal chemical exposure during pregnancy.
Collapse
Affiliation(s)
- Alison P Sanders
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Heather H Burris
- Department of Neonatology, Beth Israel Deaconess Medical Center & Division of Newborn Medicine, Boston Children's Hospital & Harvard Medical School, 330 Brookline Ave, RO 318, Boston, MA 02215, USA
| | - Allan C Just
- Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| | - Valeria Motta
- Laboratory of Environmental Epigenetics, Exposure Epidemiology & Risk Program, Harvard TH Chan School of Public Health, Boston, MA 02115, USA.,Department of Clinical Sciences & Community Health University of Milan - Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Chitra Amarasiriwardena
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Laboratory of Environmental Epigenetics, Exposure Epidemiology & Risk Program, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| | - Katherine Svensson
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Emily Oken
- Department of Population Medicine, Harvard Medical School & Harvard Pilgrim Health Care Institute, Boston, MA 02215, USA
| | - Maritsa Solano-Gonzalez
- Center for Nutrition & Health Research, National Institute of Public Health, Cuernavaca, Morelos, Mexico
| | - Adriana Mercado-Garcia
- Center for Nutrition & Health Research, National Institute of Public Health, Cuernavaca, Morelos, Mexico
| | - Ivan Pantic
- Laboratory of Environmental Epigenetics, Exposure Epidemiology & Risk Program, Harvard TH Chan School of Public Health, Boston, MA 02115, USA.,Division of Research in Public Health, National Institute of Perinatology, Mexico City, Mexico
| | - Joel Schwartz
- Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| | - Martha M Tellez-Rojo
- Center for Nutrition & Health Research, National Institute of Public Health, Cuernavaca, Morelos, Mexico
| | - Andrea A Baccarelli
- Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA 02115, USA.,Laboratory of Environmental Epigenetics, Exposure Epidemiology & Risk Program, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| | - Robert O Wright
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
410
|
Qi W, Chen J, Cheng X, Huang J, Xiang T, Li Q, Long H, Zhu B. Targeting the Wnt-Regulatory Protein CTNNBIP1 by microRNA-214 Enhances the Stemness and Self-Renewal of Cancer Stem-Like Cells in Lung Adenocarcinomas. Stem Cells 2015; 33:3423-36. [PMID: 26299367 DOI: 10.1002/stem.2188] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 07/29/2015] [Accepted: 08/07/2015] [Indexed: 01/07/2023]
Abstract
A novel hypothesis in cancer biology proposes that cancer growth is driven by cancer stem-like cells (CSLCs), also called tumor-initiating cells, which can self-renew and differentiate into multilineage progeny in a fashion similar to stem cells. However, the impact and underlying mechanisms of this process in lung adenocarcinoma (LAC) remain to be elucidated. Here, we report that microRNA-214 (miR-214) contributes to cell self-renewal by directly targeting catenin beta interacting protein 1 (CTNNBIP1), a member of the Wnt signaling pathway. We demonstrate that miR-214 overexpression enhances stem-like properties in LAC cells and that miR-214 shows increased expression in CSLCs derived from primary tumor tissue and from two LAC cell lines (A549 and NCI-H1650). Strikingly, downregulation of miR-214 expression in CSLCs resulted in a significant decrease in spheroid formation and the expression of the stem-cell markers Nanog, Oct-4, and Sox-2. Finally, CTNNBIP1 was identified as a target of miR-214. miR-214 expression in LAC was negatively correlated with CTNNBIP1 expression and positively correlated with differentiated cellular states. Moreover, CTNNBIP1 expression correlated with longer overall survival in LAC patients. This study reveals that miR-214 plays a critical role in CSLC self-renewal and stemness by targeting CTNNBIP1. The identification of this functional miR-214-CTNNBIP1 interaction that regulates self-renewal in CSLCs has the potential to direct the development of novel therapeutic strategies for LAC.
Collapse
Affiliation(s)
- Wei Qi
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Junying Chen
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Xiaoming Cheng
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Jiani Huang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Tong Xiang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Qijing Li
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Haixia Long
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, People's Republic of China.,Biomedical Analysis Center, Third Military Medical University, Chongqing, People's Republic of China
| |
Collapse
|
411
|
Zamanian M, Fraser LM, Agbedanu PN, Harischandra H, Moorhead AR, Day TA, Bartholomay LC, Kimber MJ. Release of Small RNA-containing Exosome-like Vesicles from the Human Filarial Parasite Brugia malayi. PLoS Negl Trop Dis 2015; 9:e0004069. [PMID: 26401956 PMCID: PMC4581865 DOI: 10.1371/journal.pntd.0004069] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 08/18/2015] [Indexed: 01/28/2023] Open
Abstract
Lymphatic filariasis (LF) is a socio-economically devastating mosquito-borne Neglected Tropical Disease caused by parasitic filarial nematodes. The interaction between the parasite and host, both mosquito and human, during infection, development and persistence is dynamic and delicately balanced. Manipulation of this interface to the detriment of the parasite is a promising potential avenue to develop disease therapies but is prevented by our very limited understanding of the host-parasite relationship. Exosomes are bioactive small vesicles (30-120 nm) secreted by a wide range of cell types and involved in a wide range of physiological processes. Here, we report the identification and partial characterization of exosome-like vesicles (ELVs) released from the infective L3 stage of the human filarial parasite Brugia malayi. Exosome-like vesicles were isolated from parasites in culture media and electron microscopy and nanoparticle tracking analysis were used to confirm that vesicles produced by juvenile B. malayi are exosome-like based on size and morphology. We show that loss of parasite viability correlates with a time-dependent decay in vesicle size specificity and rate of release. The protein cargo of these vesicles is shown to include common exosomal protein markers and putative effector proteins. These Brugia-derived vesicles contain small RNA species that include microRNAs with host homology, suggesting a potential role in host manipulation. Confocal microscopy shows J774A.1, a murine macrophage cell line, internalize purified ELVs, and we demonstrate that these ELVs effectively stimulate a classically activated macrophage phenotype in J774A.1. To our knowledge, this is the first report of exosome-like vesicle release by a human parasitic nematode and our data suggest a novel mechanism by which human parasitic nematodes may actively direct the host responses to infection. Further interrogation of the makeup and function of these bioactive vesicles could seed new therapeutic strategies and unearth stage-specific diagnostic biomarkers.
Collapse
Affiliation(s)
- Mostafa Zamanian
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, United States of America
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Lisa M Fraser
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, United States of America
| | - Prince N Agbedanu
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, United States of America
| | - Hiruni Harischandra
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, United States of America
| | - Andrew R Moorhead
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, United States of America
| | - Tim A Day
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, United States of America
| | - Lyric C Bartholomay
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Michael J Kimber
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, United States of America
- * E-mail:
| |
Collapse
|
412
|
Hardy RR, Hayakawa K. Perspectives on fetal derived CD5+ B1 B cells. Eur J Immunol 2015; 45:2978-84. [PMID: 26339791 DOI: 10.1002/eji.201445146] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 08/21/2015] [Accepted: 08/31/2015] [Indexed: 01/01/2023]
Abstract
CD5(+) B-cell origins and their predisposition to lymphoma are long-standing issues. Transfer of fetal and adult liver BM Pro-B cells generates B cells with distinct phenotypes: fetal cells generate IgM(high) IgD(low) CD5(+) , whereas adult cells IgM(low) IgD(high) CD5(-) . This suggests a developmental switch in B lymphopoiesis, similar to the switch in erythropoiesis. Comparison of mRNA and miRNA expression in fetal and adult Pro-B cells revealed differential expression of Lin28b mRNA and Let-7 miRNA, providing evidence that this regulatory axis functions in the switch. Recent work has shown that Arid3a is a key transcription factor mediating fetal-type B-cell development. Lin28b-promoted fetal development generates CD5(+) B cells as a consequence of positively selected self-reactivity. CD5(+) B cells play important roles in clearance of apoptotic cells and in protective immune responses, but also pose a risk of progression to leukemia/lymphoma. Differential Lin28b expression in fetal and adult human B-cell precursors showed that human B-cell development may resemble mouse, with self-reactive "innate-like" B cells generated early in life. It remains to be determined whether such human B cells have a higher propensity to leukemic progression. This review describes our recent research with CD5(+) B cells and presents our perspective on their role in disease.
Collapse
|
413
|
Lee H, Han S, Kwon CS, Lee D. Biogenesis and regulation of the let-7 miRNAs and their functional implications. Protein Cell 2015; 7:100-13. [PMID: 26399619 PMCID: PMC4742387 DOI: 10.1007/s13238-015-0212-y] [Citation(s) in RCA: 247] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 08/17/2015] [Indexed: 12/11/2022] Open
Abstract
The let-7 miRNA was one of the first miRNAs discovered in the nematode, Caenorhabditis elegans, and its biological functions show a high level of evolutionary conservation from the nematode to the human. Unlike in C. elegans, higher animals have multiple isoforms of let-7 miRNAs; these isoforms share a consensus sequence called the ‘seed sequence’ and these isoforms are categorized into let-7 miRNA family. The expression of let-7 family is required for developmental timing and tumor suppressor function, but must be suppressed for the self-renewal of stem cells. Therefore, let-7 miRNA biogenesis must be carefully controlled. To generate a let-7 miRNA, a primary transcript is produced by RNA polymerase II and then subsequently processed by Drosha/DGCR8, TUTase, and Dicer. Because dysregulation of let-7 processing is deleterious, biogenesis of let-7 is tightly regulated by cellular factors, such as the RNA binding proteins, LIN28A/B and DIS3L2. In this review, we discuss the biological functions and biogenesis of let-7 miRNAs, focusing on the molecular mechanisms of regulation of let-7 biogenesis in vertebrates, such as the mouse and the human.
Collapse
Affiliation(s)
- Hosuk Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 305-701, Korea
| | - Sungwook Han
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 305-701, Korea
| | - Chang Seob Kwon
- Department of Chemistry and Biology, Korea Science Academy of KAIST, Busan, 614-822, Korea
| | - Daeyoup Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 305-701, Korea.
| |
Collapse
|
414
|
Wen F, Li B, Huang C, Wei Z, Zhou Y, Liu J, Zhang H. MiR-34a is Involved in the Decrease of ATP Contents Induced by Resistin Through Target on ATP5S in HepG2 Cells. Biochem Genet 2015; 53:301-9. [PMID: 26385595 DOI: 10.1007/s10528-015-9693-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 09/05/2015] [Indexed: 01/22/2023]
Abstract
Resistin is associated with metabolic syndrome and deciphering its developmental and molecular mechanisms may help the development of new treatments. MiRNAs serve as negative regulators in many physiological and pathological processes. Here, miRNA microarrays were used to detect differences in expression between resistin-treated and control mice, and results showed miR-34a to be upregulated by resistin. The purpose of this study was to determine whether miR-34a played a role in resistin-induced decrease of ATP contents. Transient transfection of miR-34a mimics was used to overexpress miR-34a and quantitative RT-PCR was used to detect its expression. Western blot analysis was used to determine the rate of expression at the protein level. ATP content was measured using an ATP assay kit. The target gene of miR-34a was analyzed using bioinformatics and confirmed with dual-luciferase report system. MiR-34a was upregulated by resistin in HepG2 cells, and overexpression of miR-34a was found to diminish ATP levels significantly. This study is the first to show that ATP5S is one of the target genes of miR-34a. Resistin diminishes ATP content through the targeting of ATP5S mRNA 3'UTR by miR-34a.
Collapse
Affiliation(s)
- Fengyun Wen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471003, Henan, People's Republic of China.
| | - Bin Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Chunyan Huang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Zhiguo Wei
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471003, Henan, People's Republic of China
| | - Yingying Zhou
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471003, Henan, People's Republic of China
| | - Jianyu Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471003, Henan, People's Republic of China
| | - Haiwei Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471003, Henan, People's Republic of China
| |
Collapse
|
415
|
Gupta S, Verma S, Mantri S, Berman NE, Sandhir R. Targeting MicroRNAs in Prevention and Treatment of Neurodegenerative Disorders. Drug Dev Res 2015; 76:397-418. [PMID: 26359796 DOI: 10.1002/ddr.21277] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Preclinical Research microRNAs (miRNAs) are small noncoding RNAs (ncRNAs) that are key regulators of gene expression. They act on wide range of targets by binding to mRNA via imperfect complementarity at 3' UTR. Evidence suggests that miRNAs regulate many biological processes including neuronal development, differentiation, and disease. Altered expression of several miRNAs has been reported in many neurodegenerative disorders (NDDs). Many miRNAs are altered in these diseases, but miRNA 15, miRNA 21, and miRNA 146a have been shown to play critical role in many neurodegenerative conditions. As these miRNAs regulate many genes, miRNA targeted approaches would allow concurrently targeting of multiple effectors of pathways that regulate disease progression. In this review, we describe the role of miRNAs in various NDDs and their potential as therapeutic tools in prevention and treatment of neurological conditions.
Collapse
Affiliation(s)
- Smriti Gupta
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India
| | - Savita Verma
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India
| | - Shrikant Mantri
- Computational Biology Laboratory, National Agri-Food Biotechnology Institute, Mohali, Punjab, 160071, India
| | - Nancy E Berman
- Department of Anatomy & Cell Biology, Kansas University Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Rajat Sandhir
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India
| |
Collapse
|
416
|
Wan XX, Yi H, Qu JQ, He QY, Xiao ZQ. Integrated analysis of the differential cellular and EBV miRNA expression profiles in microdissected nasopharyngeal carcinoma and non-cancerous nasopharyngeal tissues. Oncol Rep 2015; 34:2585-601. [PMID: 26330189 DOI: 10.3892/or.2015.4237] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 06/26/2015] [Indexed: 01/17/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is commonly diagnosed in southern Asia. MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression post-transcriptionally. Increasing evidence suggests that the dysregulation of miRNAs promotes NPC tumorigenesis. Epstein-Barr virus (EBV) infection and EBV-encoded miRNAs are also associated with the development of NPC. However, it is unclear how cellular and EBV miRNAs jointly regulate target genes and signaling pathways in NPC. In the present study, we analyzed the differential cellular and EBV miRNA expression profiles in 20 pooled NPC tissues using microarrays. We found that 19 cellular miRNAs and 9 EBV miRNAs were upregulated and 31 cellular miRNAs were downregulated in NPC tissues. Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis indicated that the 19 upregulated miRNAs target mainly the p53 signaling pathway in cancer, whereas the downregulated miRNAs regulate pathways related to cancer, focal adhesion and Erb, and MAPK signaling. In contrast, the upregulated EBV miRNAs target primarily the TGF-β and Wnt signaling pathways. Data also suggested that cellular miR-34b, miR-34c, miR-18a, miR‑200a/b, miR-449a, miR-31 and let-7 may be dysregulated in NPCs, and that the aberrant activation of their target genes in the p53 pathway and cell cycle enhance NPC cell survival and proliferation. In addition, EBV-miRNAs such as BART3 and BART5 target genes in the p53, TGF-β and Wnt signaling pathways to modulate NPC apoptosis and transformation. To better elucidate the interaction between miRNAs and target genes, we constructed an anti-correlated cellular and EBV miRNA/target gene regulatory network. The current findings may help dissect the roles played by cellular and EBV miRNAs during NPC tumorigenesis, and also provide useful biomarkers for the diagnosis and treatment of NPCs.
Collapse
Affiliation(s)
- Xun-Xun Wan
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Hong Yi
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jia-Quan Qu
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Qiu-Yan He
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Zhi-Qiang Xiao
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
417
|
Jefferson A, Cadet VE, Hielscher A. The mechanisms of genetically modified vaccinia viruses for the treatment of cancer. Crit Rev Oncol Hematol 2015; 95:407-16. [PMID: 25900073 DOI: 10.1016/j.critrevonc.2015.04.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 12/18/2014] [Accepted: 04/02/2015] [Indexed: 02/06/2023] Open
Abstract
The use of oncolytic viruses for the treatment of cancer is an emerging field of cancer research and therapy. Oncolytic viruses are designed to induce tumor specific immunity while replicating selectively within cancer cells to cause lysis of the tumor cells. While there are several forms of oncolytic viruses, the use of vaccinia viruses for oncolysis may be more beneficial than other forms of oncolytic viruses. For example, vaccinia viruses have been shown to exert their anti-tumor effects through genetic engineering strategies which enhance their therapeutic efficacy. This paper will address some of the most common forms of genetically modified vaccinia viruses and will explore the mechanisms whereby they selectively target, enter and destroy cancer cells. Furthermore, this review will highlight how vaccinia viruses activate host immune responses against cancer cells and will address clinical trials evaluating the tumor-directed and killing efficacy of these viruses against solid tumors.
Collapse
Affiliation(s)
- Artrish Jefferson
- Department of Biomedical Sciences, Georgia-Philadelphia College of Osteopathic Medicine, Suwanee, GA 30024, United States
| | - Valerie E Cadet
- Department of Biomedical Sciences, Georgia-Philadelphia College of Osteopathic Medicine, Suwanee, GA 30024, United States
| | - Abigail Hielscher
- Department of Biomedical Sciences, Georgia-Philadelphia College of Osteopathic Medicine, Suwanee, GA 30024, United States.
| |
Collapse
|
418
|
Sclafani F, Chau I, Cunningham D, Peckitt C, Lampis A, Hahne JC, Braconi C, Tabernero J, Glimelius B, Cervantes A, Begum R, Gonzalez De Castro D, Hulkki Wilson S, Eltahir Z, Wotherspoon A, Tait D, Brown G, Oates J, Valeri N. Prognostic role of the LCS6 KRAS variant in locally advanced rectal cancer: results of the EXPERT-C trial. Ann Oncol 2015; 26:1936-1941. [PMID: 26162609 PMCID: PMC4551162 DOI: 10.1093/annonc/mdv285] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 05/29/2015] [Accepted: 06/26/2015] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Lethal-7 (let-7) is a tumour suppressor miRNA which acts by down-regulating several oncogenes including KRAS. A single-nucleotide polymorphism (rs61764370, T > G base substitution) in the let-7 complementary site 6 (LCS-6) of KRAS mRNA has been shown to predict prognosis in early-stage colorectal cancer (CRC) and benefit from anti-epidermal growth factor receptor monoclonal antibodies in metastatic CRC. PATIENTS AND METHODS We analysed rs61764370 in EXPERT-C, a randomised phase II trial of neoadjuvant CAPOX followed by chemoradiotherapy, surgery and adjuvant CAPOX plus or minus cetuximab in locally advanced rectal cancer. DNA was isolated from formalin-fixed paraffin-embedded tumour tissue and genotyped using a PCR-based commercially available assay. Kaplan-Meier method and Cox regression analysis were used to calculate survival estimates and compare treatment arms. RESULTS A total of 155/164 (94.5%) patients were successfully analysed, of whom 123 (79.4%) and 32 (20.6%) had the LCS-6 TT and LCS-6 TG genotype, respectively. Carriers of the G allele were found to have a statistically significantly higher rate of complete response (CR) after neoadjuvant therapy (28.1% versus 10.6%; P = 0.020) and a trend for better 5-year progression-free survival (PFS) [77.4% versus 64.5%: hazard ratio (HR) 0.56; P = 0.152] and overall survival (OS) rates (80.3% versus 71.9%: HR 0.59; P = 0.234). Both CR and survival outcomes were independent of the use of cetuximab. The negative prognostic effect associated with KRAS mutation appeared to be stronger in patients with the LCS-6 TT genotype (HR PFS 1.70, P = 0.078; HR OS 1.79, P = 0.082) compared with those with the LCS-6 TG genotype (HR PFS 1.33, P = 0.713; HR OS 1.01, P = 0.995). CONCLUSION This analysis suggests that rs61764370 may be a biomarker of response to neoadjuvant treatment and an indicator of favourable outcome in locally advanced rectal cancer possibly by mitigating the poor prognosis of KRAS mutation. In this setting, however, this polymorphism does not appear to predict cetuximab benefit.
Collapse
Affiliation(s)
- F Sclafani
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey
| | - I Chau
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey
| | - D Cunningham
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey
| | - C Peckitt
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey
| | - A Lampis
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton
| | - J C Hahne
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton
| | - C Braconi
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey; Division of Cancer Therapeutics, The Institute of Cancer Research, London and Sutton, UK
| | - J Tabernero
- Department of Medical Oncology, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - B Glimelius
- Department of Immunology, Genetics and Pathology, University of Uppsala, Uppsala, Sweden
| | - A Cervantes
- Department of Hematology and Medical Oncology, Biomedical Research Institute INCLIVA, University of Valencia, Valencia, Spain
| | - R Begum
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey
| | - D Gonzalez De Castro
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey
| | - S Hulkki Wilson
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey
| | - Z Eltahir
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey
| | - A Wotherspoon
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey
| | - D Tait
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey
| | - G Brown
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey
| | - J Oates
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey
| | - N Valeri
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey; Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton.
| |
Collapse
|
419
|
Chen Z, Wang D, Gu C, Liu X, Pei W, Li J, Cao Y, Jiao Y, Tong J, Nie J. Down-regulation of let-7 microRNA increased K-ras expression in lung damage induced by radon. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 40:541-548. [PMID: 26318567 DOI: 10.1016/j.etap.2015.08.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 08/06/2015] [Accepted: 08/08/2015] [Indexed: 06/04/2023]
Abstract
Radon has long been recognized as a human carcinogen leading to lung cancer, but the underlying mechanisms remain obscure. Recent studies have shown that the let-7 microRNA and K-ras play an important role in the development of various cancers. However, the exact role between let-7 and K-ras in radon induced lung damage has not been explored so far. In the present study, wistar rats and human bronchial epithelial (HBE) cells were long-term exposed to radon, and then alterations in histological pathology of rat lung tissue, ROS, antioxidant enzymes activities and clonogenic formation in HBE cells, as well as changes in let-7 and K-ras expression were determined to observe the adverse effects induced by radon. The results showed that long-term exposure to radon produced severe lung damage in rats, significantly increased ROS production and clonogenic formation ratios and decreased SOD activities in HBE cells. In addition, an obvious down-regulation of let-7 and up-regulation of K-ras were also revealed both in mRNA and in protein level in lung tissue of rats and HBE cells exposed to radon. Furthermore, a significant down-regulation of K-ras was then confirmed in both let-7b-3p and let-7a-2-3p transfected HBE cells. Taken together, the present results propose an involvement of let-7 microRNA and K-ras in radon induced lung damage both in vivo and in vitro, which may thus be of potential value in early diagnosis and therapy of radon-induced lung tumorgenesis.
Collapse
Affiliation(s)
- Zhihai Chen
- Department of Toxicology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou 215123, China
| | - Dapeng Wang
- Department of Toxicology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou 215123, China
| | - Chao Gu
- Department of Toxicology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou 215123, China
| | - Xing Liu
- Department of Toxicology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou 215123, China
| | - Weiwei Pei
- Department of Toxicology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou 215123, China
| | - Jianxiang Li
- Department of Toxicology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou 215123, China
| | - Yi Cao
- Department of Toxicology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou 215123, China
| | - Yang Jiao
- School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China
| | - Jian Tong
- Department of Toxicology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou 215123, China.
| | - Jihua Nie
- Department of Toxicology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou 215123, China.
| |
Collapse
|
420
|
Sharma SB, Ruppert JM. MicroRNA-Based Therapeutic Strategies for Targeting Mutant and Wild Type RAS in Cancer. Drug Dev Res 2015; 76:328-42. [PMID: 26284568 DOI: 10.1002/ddr.21270] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRs) have been causally implicated in the progression and development of a wide variety of cancers. miRs modulate the activity of key cell signaling networks by regulating the translation of pathway component proteins. Thus, the pharmacological targeting of miRs that regulate cancer cell signaling networks, either by promoting (using miR-supplementation) or by suppressing (using antisense oligonucleotide-based strategies) miR activity is an area of intense research. The RAS-extracellular signal regulated kinase (ERK) pathway represents a major miR-regulated signaling network that endows cells with some of the classical hallmarks of cancer, and is often inappropriately activated in malignancies by somatic genetic alteration through point mutation or alteration of gene copy number. In addition, recent progress indicates that many tumors may be deficient in GTPase activating proteins (GAPs) due to the collaborative action of oncogenic miRs. Recent studies also suggest that in tumors harboring a mutant RAS allele there is a critical role for wild type RAS proteins in determining overall RAS-ERK pathway activity. Together, these two advances comprise a new opportunity for therapeutic intervention. In this review, we evaluate miR-based therapeutic strategies for modulating RAS-ERK signaling in cancers; in particular for more direct modulation of RAS-GTP levels, with the potential to complement current strategies to yield more durable treatment responses. To this end, we discuss the potential for miR-based therapies focused on three prominent miRs including the pan-RAS regulator let-7 and the GAP regulator comprised of miR-206 and miR-21 (miR-206/21).
Collapse
Affiliation(s)
- Sriganesh B Sharma
- Department of Biochemistry, West Virginia University, Morgantown, WV, 26506, USA.,Program in Cancer Cell Biology, West Virginia University, Morgantown, WV, 26506, USA
| | - John Michael Ruppert
- Department of Biochemistry, West Virginia University, Morgantown, WV, 26506, USA.,The Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV, 26506, USA
| |
Collapse
|
421
|
Kwon MS, Kim Y, Lee S, Namkung J, Yun T, Yi SG, Han S, Kang M, Kim SW, Jang JY, Park T. Integrative analysis of multi-omics data for identifying multi-markers for diagnosing pancreatic cancer. BMC Genomics 2015; 16 Suppl 9:S4. [PMID: 26328610 PMCID: PMC4547403 DOI: 10.1186/1471-2164-16-s9-s4] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background microRNA (miRNA) expression plays an influential role in cancer classification and malignancy, and miRNAs are feasible as alternative diagnostic markers for pancreatic cancer, a highly aggressive neoplasm with silent early symptoms, high metastatic potential, and resistance to conventional therapies. Methods In this study, we evaluated the benefits of multi-omics data analysis by integrating miRNA and mRNA expression data in pancreatic cancer. Using support vector machine (SVM) modelling and leave-one-out cross validation (LOOCV), we evaluated the diagnostic performance of single- or multi-markers based on miRNA and mRNA expression profiles from 104 PDAC tissues and 17 benign pancreatic tissues. For selecting even more reliable and robust markers, we performed validation by independent datasets from the Gene Expression Omnibus (GEO) and the Cancer Genome Atlas (TCGA) data depositories. For validation, miRNA activity was estimated by miRNA-target gene interaction and mRNA expression datasets in pancreatic cancer. Results Using a comprehensive identification approach, we successfully identified 705 multi-markers having powerful diagnostic performance for PDAC. In addition, these marker candidates annotated with cancer pathways using gene ontology analysis. Conclusions Our prediction models have strong potential for the diagnosis of pancreatic cancer.
Collapse
|
422
|
Next Generation Sequencing Analysis Reveals Segmental Patterns of microRNA Expression in Mouse Epididymal Epithelial Cells. PLoS One 2015; 10:e0135605. [PMID: 26270822 PMCID: PMC4535982 DOI: 10.1371/journal.pone.0135605] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 07/23/2015] [Indexed: 02/04/2023] Open
Abstract
The functional maturation of mammalian spermatozoa is accomplished as the cells descend through the highly specialized microenvironment of the epididymis. This dynamic environment is, in turn, created by the combined secretory and absorptive activity of the surrounding epithelium and displays an extraordinary level of regionalization. Although the regulatory network responsible for spatial coordination of epididymal function remains unclear, recent evidence has highlighted a novel role for the RNA interference pathway. Indeed, as noncanonical regulators of gene expression, small noncoding RNAs have emerged as key elements of the circuitry involved in regulating epididymal function and hence sperm maturation. Herein we have employed next generation sequencing technology to profile the genome-wide miRNA signatures of mouse epididymal cells and characterize segmental patterns of expression. An impressive profile of some 370 miRNAs were detected in the mouse epididymis, with a subset of these specifically identified within the epithelial cells that line the tubule (218). A majority of the latter miRNAs (75%) were detected at equivalent levels along the entire length of the mouse epididymis. We did however identify a small cohort of miRNAs that displayed highly regionalized patterns of expression, including miR-204-5p and miR-196b-5p, which were down- and up-regulated by approximately 39- and 45-fold between the caput/caudal regions, respectively. In addition we identified 79 miRNAs (representing ~ 21% of all miRNAs) as displaying conserved expression within all regions of the mouse, rat and human epididymal tissue. These included 8/14 members of let-7 family of miRNAs that have been widely implicated in the control of androgen signaling and the repression of cell proliferation and oncogenic pathways. Overall these data provide novel insights into the sophistication of the miRNA network that regulates the function of the male reproductive tract.
Collapse
|
423
|
Madison BB, Jeganathan AN, Mizuno R, Winslow MM, Castells A, Cuatrecasas M, Rustgi AK. Let-7 Represses Carcinogenesis and a Stem Cell Phenotype in the Intestine via Regulation of Hmga2. PLoS Genet 2015; 11:e1005408. [PMID: 26244988 PMCID: PMC4526516 DOI: 10.1371/journal.pgen.1005408] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 07/01/2015] [Indexed: 11/18/2022] Open
Abstract
Let-7 miRNAs comprise one of the largest and most highly expressed family of miRNAs among vertebrates, and is critical for promoting differentiation, regulating metabolism, inhibiting cellular proliferation, and repressing carcinogenesis in a variety of tissues. The large size of the Let-7 family of miRNAs has complicated the development of mutant animal models. Here we describe the comprehensive repression of all Let-7 miRNAs in the intestinal epithelium via low-level tissue-specific expression of the Lin28b RNA-binding protein and a conditional knockout of the MirLet7c-2/Mirlet7b locus. This ablation of Let-7 triggers the development of intestinal adenocarcinomas concomitant with reduced survival. Analysis of both mouse and human intestinal cancer specimens reveals that stem cell markers were significantly associated with loss of Let-7 miRNA expression, and that a number of Let-7 targets were elevated, including Hmga1 and Hmga2. Functional studies in 3-D enteroids revealed that Hmga2 is necessary and sufficient to mediate many characteristics of Let-7 depletion, namely accelerating cell cycle progression and enhancing a stem cell phenotype. In addition, inactivation of a single Hmga2 allele in the mouse intestine epithelium significantly represses tumorigenesis driven by Lin28b. In aggregate, we conclude that Let-7 depletion drives a stem cell phenotype and the development of intestinal cancer, primarily via Hmga2. Cancer develops following multiple genetic mutations (i.e. in tumor suppressors and oncogenes), and mutations that cooperate or synergize are often advantageous to cancer cell growth. To study how multiple genes might cooperate, it is usually informative to generate candidate mutations in cells or in mice. Large gene families, such as the Let-7 family, are difficult to silence or mutate because of the large amount of redundancy that exists between similar copies of the same gene; the mutation of one will often be masked or compensated by the continued function of others. In the mouse intestine we have achieved comprehensive depletion of all Let-7 miRNAs in this large multi-genic family through use of an inhibitory protein, called LIN28B, that specifically represses Let-7, and genetic inactivation of another gene cluster called MirLet7c-2/Mirlet7b. Mice with this comprehensive depletion of Let-7 develop intestinal cancers that resemble human colon cancers. Our further analysis identified another gene, HMGA2, downstream of this pathway that is critical to this outcome.
Collapse
Affiliation(s)
- Blair B. Madison
- Division of Gastroenterology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Arjun N. Jeganathan
- Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Rei Mizuno
- Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Monte M. Winslow
- Department of Genetics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Antoni Castells
- Gastroenterology Department, Hospital Clínic, CIBERehd, IDIBAPS, Barcelona, Catalonia, Spain
| | - Miriam Cuatrecasas
- Department of Pathology, Pharmacology and Microbiology, Hospital Clínic, CDB, University of Barcelona, Barcelona, Catalonia, Spain
| | - Anil K. Rustgi
- Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
424
|
Histone Deacetylase 10 Regulates the Cell Cycle G2/M Phase Transition via a Novel Let-7-HMGA2-Cyclin A2 Pathway. Mol Cell Biol 2015; 35:3547-65. [PMID: 26240284 DOI: 10.1128/mcb.00400-15] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 07/30/2015] [Indexed: 12/21/2022] Open
Abstract
Histone deacetylase (HDAC) inhibition leads to cell cycle arrest in G1 and G2, suggesting HDACs as therapeutic targets for cancer and diseases linked to abnormal cell growth and proliferation. Many HDACs are transcriptional repressors. Some may alter cell cycle progression by deacetylating histones and repressing transcription of key cell cycle regulatory genes. Here, we report that HDAC10 regulates the cell cycle via modulation of cyclin A2 expression, and cyclin A2 overexpression rescues HDAC10 knockdown-induced G2/M transition arrest. HDAC10 regulates cyclin A2 expression by deacetylating histones near the let-7 promoter, thereby repressing transcription. In HDAC10 knockdown cells, let-7f and microRNA 98 (miR-98) were upregulated and the let-7 family target, HMGA2, was downregulated. HMGA2 loss resulted in enrichment of the transcriptional repressor E4F at the cyclin A2 promoter. These findings support a role for HDACs in cell cycle regulation, reveal a novel mechanism of HDAC10 action, and extend the potential of HDACs as targets in diseases of cell cycle dysregulation.
Collapse
|
425
|
|
426
|
Song J, Cho KJ, Oh Y, Lee JE. Let7a involves in neural stem cell differentiation relating with TLX level. Biochem Biophys Res Commun 2015; 462:396-401. [PMID: 25976670 DOI: 10.1016/j.bbrc.2015.05.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 05/01/2015] [Indexed: 02/04/2023]
Abstract
Neural stem cells (NSCs) have the potential for differentiation into neurons known as a groundbreaking therapeutic solution for central nervous system (CNS) diseases. To resolve the therapeutic efficiency of NSCs, recent researchers have focused on the study on microRNA's role in CNS. Some micro RNAs have been reported significant functions in NSC self-renewal and differentiation through the post-transcriptional regulation of neurogenesis genes. MicroRNA-Let7a (Let7a) has known as the regulator of diverse cellular mechanisms including cell differentiation and proliferation. In present study, we investigated whether Let7a regulates NSC differentiation by targeting the nuclear receptor TLX, which is an essential regulator of NSC self-renewal, proliferation and differentiation. We performed the following experiments: western blot analysis, TaqMan assay, RT-PCR, and immunocytochemistry to confirm the alteration of NSCs. Our data showed that let7a play important roles in controlling NSC fate determination. Thus, manipulating Let-7A and TLX could be a novel strategy to enhance the efficiency of NSC's neuronal differentiation for CNS disorders.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
| | - Kyoung Joo Cho
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea; BK21 Plus Project for Medical Sciences, and Brain Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Yumi Oh
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea; BK21 Plus Project for Medical Sciences, and Brain Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea; BK21 Plus Project for Medical Sciences, and Brain Research Institute, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
427
|
Chen J, Wang Y, McMonechy MK, Anglesio MS, Yang W, Senz J, Maines-Bandiera S, Rosner J, Trigo-Gonzalez G, Grace Cheng SW, Kim J, Matzuk MM, Morin GB, Huntsman DG. Recurrent DICER1 hotspot mutations in endometrial tumours and their impact on microRNA biogenesis. J Pathol 2015; 237:215-25. [PMID: 26033159 DOI: 10.1002/path.4569] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 03/15/2015] [Accepted: 05/22/2015] [Indexed: 12/18/2022]
Abstract
DICER1 plays a critical role in microRNA (miRNA) biogenesis. Recurrent somatic 'hotspot' mutations at the four metal-binding sites within the RNase IIIb domain of DICER1 were identified in ovarian sex cord-stromal tumours and have since been described in other paediatric tumours. In this study, we screened the RNase IIIb domain of DICER1 in 290 endometrial tumours and identified six cases with hotspot mutations, including two cases affected by an atypical G1809R mutation directly adjacent to a metal-binding site. Using Illumina and Sanger targeted resequencing, we observed and validated biallelic DICER1 mutations in several cases with hotspot mutations. Through in vitro DICER1 cleavage assays, small RNA deep sequencing and real-time PCR, we demonstrated that mutations adding a positively charged side chain to residue 1809 have similar detrimental effects on 5p miRNA production to mutations at the metal-binding sites. As expected, 5p miRNAs were globally reduced in tumours and cell lines with hotspot mutations. Pathway analysis of gene expression profiles indicated that genes de-repressed due to loss of 5p miRNAs are strongly associated with pathways regulating the cell cycle. Using a Dicer1-null mouse cell line model, we found that expression of DICER1 hotspot mutants promoted cell proliferation, whereas wild-type (WT) DICER1 inhibited cell proliferation. Furthermore, targets of let-7 family miRNAs are enriched among the up-regulated genes, suggesting that loss of let-7 may be impacting downstream pathways. Our results reveal that DICER1 hotspot mutations are implicated in common malignancies and may constitute a unique oncogenic pathway.
Collapse
Affiliation(s)
- Jiamin Chen
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Center for Translational and Applied Genomics, BC Cancer Agency, Vancouver, BC, Canada
| | - Yemin Wang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Center for Translational and Applied Genomics, BC Cancer Agency, Vancouver, BC, Canada
| | - Melissa K McMonechy
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Center for Translational and Applied Genomics, BC Cancer Agency, Vancouver, BC, Canada
| | - Michael S Anglesio
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Winnie Yang
- Center for Translational and Applied Genomics, BC Cancer Agency, Vancouver, BC, Canada
| | - Janine Senz
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Sarah Maines-Bandiera
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Jamie Rosner
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC, Canada
| | | | - S W Grace Cheng
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC, Canada
| | - Jaeyeon Kim
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Martin M Matzuk
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Gregg B Morin
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC, Canada
| | - David G Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada.,Center for Translational and Applied Genomics, BC Cancer Agency, Vancouver, BC, Canada
| |
Collapse
|
428
|
Khan IN, Al-Karim S, Bora RS, Chaudhary AG, Saini KS. Cancer stem cells: a challenging paradigm for designing targeted drug therapies. Drug Discov Today 2015; 20:1205-16. [PMID: 26143148 DOI: 10.1016/j.drudis.2015.06.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 06/19/2015] [Accepted: 06/24/2015] [Indexed: 02/06/2023]
Abstract
Despite earlier controversies about their role and existence within tumors, cancer stem cells (CSCs) are now emerging as a plausible target for new drug discovery. Research and development (R&D) efforts are being directed against key gene(s) driving initiation, growth, and metastatic pathways in CSCs and the tumor microenvironment (TME). However, the niche signals that enable these pluripotent CSCs to evade radio- and chemotherapy, and to travel to secondary tissues remain enigmatic. Small-molecule drugs, biologics, miRNA, RNA interference (RNAi), and vaccines, among others, are under active investigation. Here, we examine the feasibility of leveraging current knowhow of the molecular biology of CSCs and their cellular milieu to design futuristic, targeted drugs with potentially lower toxicity that can override the multiple drug-resistance issues currently observed with existing therapeutics.
Collapse
Affiliation(s)
- Ishaq N Khan
- Embryonic & Cancer Stem Cell Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Centre of Innovation for Personalized Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Saleh Al-Karim
- Embryonic & Cancer Stem Cell Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Embryonic Stem Cell Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Roop S Bora
- Embryonic & Cancer Stem Cell Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Biotechnology Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; School of Biotechnology, Eternal University, Baru Sahib 173 101, Himachal Pradesh, India
| | - Adeel G Chaudhary
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Centre of Innovation for Personalized Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Kulvinder S Saini
- Embryonic & Cancer Stem Cell Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Biotechnology Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; School of Biotechnology, Eternal University, Baru Sahib 173 101, Himachal Pradesh, India.
| |
Collapse
|
429
|
Ong SG, Lee WH, Kodo K, Wu JC. MicroRNA-mediated regulation of differentiation and trans-differentiation in stem cells. Adv Drug Deliv Rev 2015; 88:3-15. [PMID: 25887992 DOI: 10.1016/j.addr.2015.04.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 03/26/2015] [Accepted: 04/06/2015] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNAs) are key components of a broadly conserved post-transcriptional mechanism that controls gene expression by targeting mRNAs. miRNAs regulate diverse biological processes, including the growth and differentiation of stem cells as well as the regulation of both endogenous tissue repair that has critical implications in the development of regenerative medicine approaches. In this review, we first describe key features of miRNA biogenesis and their role in regulating self-renewal, and then discuss the involvement of miRNAs in the determination of cell fate decisions. We highlight the role of miRNAs in the emergent field of reprogramming and trans-differentiation of somatic cells that could further our understanding of miRNA biology and regenerative medicine applications. Finally, we describe potential techniques for proper delivery of miRNAs in target cells.
Collapse
Affiliation(s)
- Sang-Ging Ong
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States; Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Won Hee Lee
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States; Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Kazuki Kodo
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States; Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States; Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA, United States; Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States; Institute of Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States.
| |
Collapse
|
430
|
Pontes TB, Moreira-Nunes CDFA, Maués JHDS, Lamarão LM, de Lemos JAR, Montenegro RC, Burbano RMR. The miRNA Profile of Platelets Stored in a Blood Bank and Its Relation to Cellular Damage from Storage. PLoS One 2015; 10:e0129399. [PMID: 26121269 PMCID: PMC4486185 DOI: 10.1371/journal.pone.0129399] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 05/07/2015] [Indexed: 12/19/2022] Open
Abstract
Millions of blood products are transfused each year, and many lives are directly affected by transfusion. Platelet concentrate (PC) is one of the main products derived from blood. Even under good storage conditions, PC is likely to suffer cell damage. The shape of platelets changes after 5 to 7 days of storage at 22°C. Taking into consideration that some platelet proteins undergo changes in their shape and functionality during PC storage. Sixteen PC bags were collected and each PC bag tube was cut into six equal pieces to perform experiments with platelets from six different days of storage. Thus, on the first day of storage, 1/6 of the tube was used for miRNA extraction, and the remaining 5/6 was stored under the same conditions until extraction of miRNAs on each the following five days. Samples were sequenced on an Illumina Platform to demonstrate the most highly expressed miRNAs. Three miRNAs, mir127, mir191 and mir320a were validated by real-time quantitative PCR (RQ-PCR) in 100 PC bags tubes. Our method suggests, the use of the miRNAs mir127 and mir320a as biomarkers to assess the "validity period" of PC bags stored in blood banks for long periods. Thus, bags can be tested on the 5th day of storage for the relative expression levels of mir127 and mir320a. Thus, we highlight candidate miRNAs as biomarkers of storage damage that can be used as tools to evaluate the quality of stored PC. The use of miRNAs as biomarkers of damage is unprecedented and will contribute to improved quality of blood products for transfusions.
Collapse
Affiliation(s)
- Thaís Brilhante Pontes
- Laboratory of Human Cytogenetics, Institute of Biological Sciences, Federal University of Pará, Belém, PA, 66075110, Brazil
| | | | - Jersey Heitor da Silva Maués
- Laboratory of Human Cytogenetics, Institute of Biological Sciences, Federal University of Pará, Belém, PA, 66075110, Brazil
| | - Letícia Martins Lamarão
- Laboratory of Genetics and Molecular Biology, Foundation Center for Hemotherapy and Hematology of Pará (HEMOPA), Belém, PA, 66033–000, Brazil
| | - José Alexandre Rodrigues de Lemos
- Laboratory of Genetics and Molecular Biology, Foundation Center for Hemotherapy and Hematology of Pará (HEMOPA), Belém, PA, 66033–000, Brazil
- Institute of Biological Sciences, Federal University of Pará, Belém, PA, 66075110, Brazil
| | - Raquel Carvalho Montenegro
- Laboratory of Human Cytogenetics, Institute of Biological Sciences, Federal University of Pará, Belém, PA, 66075110, Brazil
| | - Rommel Mário Rodriguez Burbano
- Laboratory of Human Cytogenetics, Institute of Biological Sciences, Federal University of Pará, Belém, PA, 66075110, Brazil
- * E-mail:
| |
Collapse
|
431
|
Beckers A, Van Peer G, Carter DR, Gartlgruber M, Herrmann C, Agarwal S, Helsmoortel HH, Althoff K, Molenaar JJ, Cheung BB, Schulte JH, Benoit Y, Shohet JM, Westermann F, Marshall GM, Vandesompele J, De Preter K, Speleman F. MYCN-driven regulatory mechanisms controlling LIN28B in neuroblastoma. Cancer Lett 2015; 366:123-32. [PMID: 26123663 DOI: 10.1016/j.canlet.2015.06.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 05/30/2015] [Accepted: 06/18/2015] [Indexed: 12/14/2022]
Abstract
LIN28B has been identified as an oncogene in various tumor entities, including neuroblastoma, a childhood cancer that originates from neural crest-derived cells, and is characterized by amplification of the MYCN oncogene. Recently, elevated LIN28B expression levels were shown to contribute to neuroblastoma tumorigenesis via let-7 dependent de-repression of MYCN. However, additional insight in the regulation of LIN28B in neuroblastoma is lacking. Therefore, we have performed a comprehensive analysis of the regulation of LIN28B in neuroblastoma, with a specific focus on the contribution of miRNAs. We show that MYCN regulates LIN28B expression in neuroblastoma tumors via two distinct parallel mechanisms. First, through an unbiased LIN28B-3'UTR reporter screen, we found that miR-26a-5p and miR-26b-5p regulate LIN28B expression. Next, we demonstrated that MYCN indirectly affects the expression of miR-26a-5p, and hence regulates LIN28B, therefore establishing an MYCN-miR-26a-5p-LIN28B regulatory axis. Second, we provide evidence that MYCN regulates LIN28B expression via interaction with the LIN28B promoter, establishing a direct MYCN-LIN28B regulatory axis. We believe that these findings mark LIN28B as an important effector of the MYCN oncogenic phenotype and underline the importance of MYCN-regulated miRNAs in establishing the MYCN-driven oncogenic process.
Collapse
Affiliation(s)
- Anneleen Beckers
- Center for Medical Genetics (CMGG), Ghent University, Medical Research Building (MRB1), De Pintelaan 185, 9000 Ghent, Belgium
| | - Gert Van Peer
- Center for Medical Genetics (CMGG), Ghent University, Medical Research Building (MRB1), De Pintelaan 185, 9000 Ghent, Belgium
| | - Daniel R Carter
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW, PO Box 81, Randwick, NSW 2031, Australia
| | - Moritz Gartlgruber
- Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Carl Herrmann
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany; Institute of Pharmacy and Molecular Biotechnology, and Bioquant Center, University of Heidelberg, Im Neuenheimer Feld 267, Heidelberg 69120, Germany
| | - Saurabh Agarwal
- Division of Pediatric Hematology-Oncology and Center for Cell and Gene Therapy, Department of Pediatrics, Baylor College of Medicine, 1102 Bates Street, Houston, TX 77030, USA
| | - Hetty H Helsmoortel
- Center for Medical Genetics (CMGG), Ghent University, Medical Research Building (MRB1), De Pintelaan 185, 9000 Ghent, Belgium; Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Kristina Althoff
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany; German Consortium for Translational Cancer Research (DKTK), Partner Site Essen/Duesseldorf, Essen, Germany
| | - Jan J Molenaar
- Department of Oncogenomics, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Belamy B Cheung
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW, PO Box 81, Randwick, NSW 2031, Australia
| | - Johannes H Schulte
- Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany; Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany; German Consortium for Translational Cancer Research (DKTK), Partner Site Essen/Duesseldorf, Essen, Germany; Translational Neuro-Oncology, West German Cancer Center (WTZ), University Hospital Essen, University Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; Centre for Medical Biotechnology, University Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Yves Benoit
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Jason M Shohet
- Division of Pediatric Hematology-Oncology and Center for Cell and Gene Therapy, Department of Pediatrics, Baylor College of Medicine, 1102 Bates Street, Houston, TX 77030, USA
| | - Frank Westermann
- Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Glenn M Marshall
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW, PO Box 81, Randwick, NSW 2031, Australia
| | - Jo Vandesompele
- Center for Medical Genetics (CMGG), Ghent University, Medical Research Building (MRB1), De Pintelaan 185, 9000 Ghent, Belgium
| | - Katleen De Preter
- Center for Medical Genetics (CMGG), Ghent University, Medical Research Building (MRB1), De Pintelaan 185, 9000 Ghent, Belgium
| | - Frank Speleman
- Center for Medical Genetics (CMGG), Ghent University, Medical Research Building (MRB1), De Pintelaan 185, 9000 Ghent, Belgium.
| |
Collapse
|
432
|
Li K, Liu Y, Zhou Y, Zhang R, Zhao N, Yan Z, Zhang Q, Zhang S, Qiu F, Xu Y. An integrated approach to reveal miRNAs' impacts on the functional consequence of copy number alterations in cancer. Sci Rep 2015; 5:11567. [PMID: 26099552 PMCID: PMC4477324 DOI: 10.1038/srep11567] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 05/29/2015] [Indexed: 12/21/2022] Open
Abstract
Copy number alteration (CNA) is known to induce gene expression changes mainly through dosage effect, and therefore affect the initiation and progression of tumor. However, tumor samples exhibit heterogeneity in gene dosage sensitivity due to the complicated mechanisms of transcriptional regulation. Currently, no high-throughput method has been available for identifying the regulatory factors affecting the functional consequences of CNA, and determining their effects on cancer. In view of the important regulatory role of miRNA, we investigated the influence of miRNAs on the dosage sensitivities of genes within the CNA regions. By integrating copy number, mRNA expression, miRNA expression profiles of three kinds of cancer, we observed a tendency for high dosage-sensitivity genes to be more targeted by miRNAs in cancer, and identified the miRNAs regulating the dosage sensitivity of amplified/deleted target genes. The results show that miRNAs can modulate oncogenic biological functions by regulating the genes within the CNA regions, and thus play a role as a trigger or balancer in cancer, affecting cancer processes, even survival. This work provided a framework for analyzing the regulation of dosage effect, which will shed a light on understanding the oncogenic and tumor suppressive mechanisms of CNA. Besides, new cancer-related miRNAs were identified.
Collapse
Affiliation(s)
- Kening Li
- 1] College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China [2] School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yongjing Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yuanshuai Zhou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Rui Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Ning Zhao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Zichuang Yan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Qiang Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Shujuan Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Fujun Qiu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
433
|
Izumiya Y, Jinnn M, Kimura Y, Wang Z, Onoue Y, Hanatani S, Araki S, Ihn H, Ogawa H. Expression of Let-7 family microRNAs in skin correlates negatively with severity of pulmonary hypertension in patients with systemic scleroderma. IJC HEART & VASCULATURE 2015; 8:98-102. [PMID: 28785688 PMCID: PMC5497286 DOI: 10.1016/j.ijcha.2015.06.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 06/15/2015] [Accepted: 06/16/2015] [Indexed: 02/02/2023]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a serious complication in patients with systemic scleroderma (SSc), therefore it is important to identify the factors that could predict the presence and progression of PH. Skin biopsy is performed in patients with SSc to examine the type and severity of the disease. MicroRNAs (miRNAs) are potential biomarkers for various cardiovascular diseases including PH. METHODS AND RESULTS We determined the skin miRNA expression profile in 15 SSc patients with (n = 6) and without PH (n = 9). A mixture of equal amounts of miRNAs from PH and non-PH patients were prepared and used for miRNA PCR array analysis. The analysis identified 591 upregulated miRNAs and 57 downregulated miRNAs in the PH group. Of these, only miRNAs with a Ct value of less than 35 were subjected to further analysis. When a 1.5-fold difference was considered meaningful, 32 miRNAs were upregulated and 14 miRNAs were downregulated in the PH group. Interestingly, 5 out of 14 downregulated miRNAs belonged to the let-7 family. The results were validated by quantitative real-time PCR with specific primer for each miRNA, which showed significant downregulation of five let-7 family members (let-7a, -7d, -7e, -7f, -7g) in 6 PH compared with 9 non-PH skin samples. The expression levels of let-7d and 7b correlated negatively with pulmonary arterial pressure measured by echocardiography. CONCLUSIONS The results suggest that skin miRNA is a potentially useful marker for the presence and severity of PH in patients with SSc.
Collapse
Affiliation(s)
- Yasuhiro Izumiya
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masatoshi Jinnn
- Department of Dermatology and Plastic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuichi Kimura
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Zhongzhi Wang
- Department of Dermatology and Plastic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshiro Onoue
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Shinsuke Hanatani
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoshi Araki
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hironobu Ihn
- Department of Dermatology and Plastic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hisao Ogawa
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
434
|
Let-7c is a Candidate Biomarker for Cervical Intraepithelial Lesions: A Pilot Study. Mol Diagn Ther 2015; 19:191-6. [DOI: 10.1007/s40291-015-0145-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
435
|
Matsushita R, Seki N, Chiyomaru T, Inoguchi S, Ishihara T, Goto Y, Nishikawa R, Mataki H, Tatarano S, Itesako T, Nakagawa M, Enokida H. Tumour-suppressive microRNA-144-5p directly targets CCNE1/2 as potential prognostic markers in bladder cancer. Br J Cancer 2015; 113:282-9. [PMID: 26057453 PMCID: PMC4506384 DOI: 10.1038/bjc.2015.195] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 03/30/2015] [Accepted: 05/06/2015] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Analysis of a microRNA (miRNA) expression signature of bladder cancer (BC) by deep-sequencing revealed that clustered miRNAs microRNA (miR)-451a, miR-144-3p, and miR-144-5p were significantly downregulated in BC tissues. We hypothesised that these miRNAs function as tumour suppressors in BC. The aim of this study was to investigate the functional roles of these miRNAs and their modulation of cancer networks in BC cells. METHODS The functional studies of BC cells were performed using transfection of mature miRNAs. Genome-wide gene expression analysis, in silico analysis, and dual-luciferase reporter assays were applied to identify miRNA targets. The association between miR-144-5p levels and expression of the target genes was determined, and overall patient survival as a function of target gene expression was estimated by the Kaplan-Meier method. RESULTS Gain-of-function studies showed that miR-144-5p significantly inhibited cell proliferation by BC cells. Four cell cycle-related genes (CCNE1, CCNE2, CDC25A, and PKMYT1) were identified as direct targets of miR-144-5p. The patients with high CCNE1 or CCNE2 expression had lower overall survival probabilities than those with low expression (P=0.025 and P=0.032). CONCLUSION miR-144-5p functions as tumour suppressor in BC cells. CCNE1 and CCNE2 were directly regulated by miR-144-5p and might be good prognostic markers for survival of BC patients.
Collapse
Affiliation(s)
- R Matsushita
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan
| | - N Seki
- Department of Functional Genomics, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - T Chiyomaru
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan
| | - S Inoguchi
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan
| | - T Ishihara
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan
| | - Y Goto
- Department of Functional Genomics, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - R Nishikawa
- Department of Functional Genomics, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - H Mataki
- Department of Pulmonary Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan
| | - S Tatarano
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan
| | - T Itesako
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan
| | - M Nakagawa
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan
| | - H Enokida
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan
| |
Collapse
|
436
|
Correlation of miRNA expression profiling in surgical pathology materials, with Ki-67, HER2, ER and PR in breast cancer patients. Int J Biol Markers 2015; 30:e190-9. [PMID: 25907662 DOI: 10.5301/jbm.5000141] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND New molecular markers related to prognosis and/or clinical outcome have been extensively studied in breast cancer. In particular, microRNA (miRNA) has attracted the interest of both basic and clinical investigators as one of the promising molecular markers of breast cancer patients. MiRNAs are a class of short noncoding RNAs that regulate mRNAs at posttranscriptional level and are deregulated in various human malignancies. Previous studies have reported that miRNAs were stably conserved in 10% formalin-fixed paraffin-embedded tissues without significant degradation, in contrast to more fragile RNA. METHODS Therefore, in this study, we examined 21 surgical breast cancer specimens using the Human Cancer microRNA PCR Array system (QIAGEN) to explore potential molecular targets of miRNAs. RESULTS Profiling of miRNA expression in archival materials demonstrated that a group of deregulated miRNAs was associated with clinicopathological parameters of the patients, such as Ki-67, HER2, ER and PR. For instance, an abundant expression of multiple let-7 miRNA family, also known as tumor suppressor, was detected in low Ki-67 and HER2 groups. Elevated expression of 8 miRNAs overlapped between Ki-67+/HER2+/ER+/PR+ groups, including several known oncogenic miRNAs such as miR-148b, miR-15b, miR-200c, miR-150, miR-191, miR-96, miR-25 and miR-21. CONCLUSIONS These results all indicated that when analyzing miRNAs in surgical pathology specimens of breast cancer as a biomarker, they should be examined as a cluster through miRNA profiling, rather than relying on the analysis of a single miRNA.
Collapse
|
437
|
Varshney M, Chandra A, Jain R, Ahmad R, Bihari V, Chandran CK, Mudiam MKR, Patnaik S, Goel SK. Occupational health hazards of trichloroethylene among workers in relation to altered mRNA expression of cell cycle regulating genes (p53, p21, bax and bcl-2) and PPARA. Toxicol Rep 2015; 2:748-757. [PMID: 28962410 PMCID: PMC5598265 DOI: 10.1016/j.toxrep.2015.04.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 04/21/2015] [Accepted: 04/21/2015] [Indexed: 11/30/2022] Open
Abstract
Trichloroethylene (TCE) is widely used as a metal degreaser in industrial processes. The present study reports on the effects of TCE exposure on workers employed in the lock industries. To ensure exposure of the workers to TCE, its toxic metabolites, trichloroacetic acid (TCA), dichloroacetic acid (DCA) and trichloroethanol (TCEOH) were detected in the plasma of the subjects through solid phase microextraction-gas chromatography-electron capture detection. TCA, DCA and TCEOH were detected in the range of 0.004–2.494 μg/mL, 0.01–3.612 μg/mL and 0.002–0.617 μg/mL, respectively. Quantitative reverse transcription polymerase chain reaction analysis revealed up-regulated expression of p53 (2.4-fold; p < 0.05), p21 (2-fold; p < 0.01), bax (2.9-fold; p < 0.01) mRNAs and down-regulated expression of bcl-2 (67%; p < 0.05) mRNAs, indicating DNA damaging potential of these metabolites. No effects were observed on the levels of p16 and c-myc mRNAs. Further, as TCA and DCA, the ligand of peroxisome proliferator activated receptor alpha (PPARA), are involved in the process of hepatocarcinogenesis in rodents, we examined expression of PPARA mRNA and let-7c miRNA in the workers. No statistically significant differences in expression of PPARA mRNA and let-7c miRNA in patients were observed as compared to values in controls. Dehydroepiandosterone sulfate (DHEAS) is a reported endogenous ligand of PPARA so its competitive role was also studied. We observed decreased levels of DHEAS hormone in the subjects. Hence, its involvement in mediation of the observed changes in the levels of various mRNAs analyzed in this study appears unlikely.
Collapse
Affiliation(s)
- Meenu Varshney
- Petroleum Toxicology Division, Indian Institute of Toxicology Research (Formerly - Industrial Toxicology Research Centre), M.G. Marg, Post Box No. 80, Lucknow 226001, India
| | - Abhijit Chandra
- Surgical Gastroenterology Division, Chhatrapati Shahuji Maharaj Medical University, Chowk, Lucknow 226003, India
| | - Rajeev Jain
- Analytical Chemistry Section, Indian Institute of Toxicology Research, (Formerly - Industrial Toxicology Research Centre), M.G. Marg, Post Box No. 80, Lucknow 226001, India
| | - Riaz Ahmad
- Biochemical and Clinical Genetics Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh 202002, UP, India
| | - Vipin Bihari
- Epidemiology Division, Indian Institute of Toxicology Research, (Formerly - Industrial Toxicology Research Centre), M.G. Marg, Post Box No. 80, Lucknow 226001, India
| | - C Keshava Chandran
- Epidemiology Division, Indian Institute of Toxicology Research, (Formerly - Industrial Toxicology Research Centre), M.G. Marg, Post Box No. 80, Lucknow 226001, India
| | - Mohana K R Mudiam
- Analytical Chemistry Section, Indian Institute of Toxicology Research, (Formerly - Industrial Toxicology Research Centre), M.G. Marg, Post Box No. 80, Lucknow 226001, India
| | - Satykam Patnaik
- Analytical Chemistry Section, Indian Institute of Toxicology Research, (Formerly - Industrial Toxicology Research Centre), M.G. Marg, Post Box No. 80, Lucknow 226001, India
| | - S K Goel
- Petroleum Toxicology Division, Indian Institute of Toxicology Research (Formerly - Industrial Toxicology Research Centre), M.G. Marg, Post Box No. 80, Lucknow 226001, India
| |
Collapse
|
438
|
Wen F, Zhang H, Bao C, Yang M, Wang N, Zhang J, Hu Y, Yang X, Geng J, Yang Z. Resistin Increases Ectopic Deposition of Lipids Through miR-696 in C2C12 Cells. Biochem Genet 2015; 53:63-71. [DOI: 10.1007/s10528-015-9672-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Accepted: 04/04/2015] [Indexed: 12/19/2022]
|
439
|
Wang B, Ricardo S. Role of microRNA machinery in kidney fibrosis. Clin Exp Pharmacol Physiol 2015; 41:543-50. [PMID: 24798583 DOI: 10.1111/1440-1681.12249] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 04/10/2014] [Accepted: 04/25/2014] [Indexed: 01/01/2023]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that are critical regulators of gene expression at the post-transcriptional level. The miRNAs constitute an abundant class of RNAs conserved from plants to animals and, as such, play key roles in diverse biological processes, including inflammation, development, differentiation and apoptosis. More recently, it has become apparent that changes in miRNA expression contribute to a wide spectrum of human pathologies, including heart and kidney disease, organ developmental abnormalities and neuronal degeneration. Moreover, inflammation and the development of kidney fibrosis is accompanied by changes in miRNA expression. This review summarizes the emerging field deciphering the complex connections between human miRNA biology and different aspects of kidney injury, focusing on kidney fibrosis. The miRNA-regulated fibrosis is discussed based on the classification of pivotal mechanisms, notably involving the transforming growth factor-β1 signalling pathway. In addition, the challenge of miRNA delivery vehicles as mechanisms of cellular transfer are reviewed, as is the use of miRNA as a potential biomarker for disease.
Collapse
Affiliation(s)
- Bo Wang
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | | |
Collapse
|
440
|
Wan SM, Yi SK, Zhong J, Nie CH, Guan NN, Chen BX, Gao ZX. Identification of MicroRNA for Intermuscular Bone Development in Blunt Snout Bream (Megalobrama amblycephala). Int J Mol Sci 2015; 16:10686-703. [PMID: 25970753 PMCID: PMC4463670 DOI: 10.3390/ijms160510686] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 04/17/2015] [Accepted: 05/05/2015] [Indexed: 01/08/2023] Open
Abstract
Intermuscular bone (IB), which occurs only in the myosepta of the lower teleosts, is attracting more attention of researchers due to its particular development and lack of genetic information. MicroRNAs (miRNAs) are emerging as important regulators for biological processes. In the present study, miRNAs from IBs and connective tissue (CT; encircled IBs) from six-month-old Megalobrama amblycephala were characterized and compared. The results revealed the sequences and expression levels of 218 known miRNA genes (belonging to 97 families). Of these miRNAs, 44 known microRNA sequences exhibited significant expression differences between the two libraries, with 24 and 20 differentially-expressed miRNAs exhibiting higher expression in the CT and IBs libraries, respectively. The expressions of 11 miRNAs were selected to validate in nine tissues. Among the high-ranked predicted gene targets, differentiation, cell cycle, metabolism, signal transduction and transcriptional regulation were implicated. The pathway analysis of differentially-expressed miRNAs indicated that they were abundantly involved in regulating the development and differentiation of IBs and CT. This study characterized the miRNA for IBs of teleosts for the first time, which provides an opportunity for further understanding of miRNA function in the regulation of IB development.
Collapse
Affiliation(s)
- Shi-Ming Wan
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China.
- Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan 430070, China.
| | - Shao-Kui Yi
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China.
- Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan 430070, China.
| | - Jia Zhong
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China.
- Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan 430070, China.
| | - Chun-Hong Nie
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China.
| | - Ning-Nan Guan
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China.
| | - Bo-Xiang Chen
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China.
- Animal Husbandry and Fisheries Research Center of Haid Group Co., Ltd., Guangzhou 511400, China.
| | - Ze-Xia Gao
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China.
- Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan 430070, China.
| |
Collapse
|
441
|
Dai X, Jiang Y, Tan C. Let-7 Sensitizes KRAS Mutant Tumor Cells to Chemotherapy. PLoS One 2015; 10:e0126653. [PMID: 25946136 PMCID: PMC4422443 DOI: 10.1371/journal.pone.0126653] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 04/04/2015] [Indexed: 12/30/2022] Open
Abstract
KRAS is the most commonly mutated oncogene in human cancers and is associated with poor prognosis and drug resistance. Let-7 is a family of tumor suppressor microRNAs that are frequently suppressed in solid tumors, where KRAS mutations are highly prevalent. In this study, we investigated the potential use of let-7 as a chemosensitizer. We found that let-7b repletion selectively sensitized KRAS mutant tumor cells to the cytotoxicity of paclitaxel and gemcitabine. Transfection of let-7b mimic downregulated the expression of mutant but not wild-type KRAS. Combination of let-7b mimic with paclitaxel or gemcitabine diminished MEK/ERK and PI3K/AKT signaling concurrently, triggered the onset of apoptosis, and reverted the epithelial-mesenchymal transition in KRAS mutant tumor cells. In addition, let-7b repletion downregulated the expression of β-tubulin III and ribonucleotide reductase subunit M2, two proteins known to mediate tumor resistance to paclitaxel and gemcitabine, respectively. Let-7 may represent a new class of chemosensitizer for the treatment of KRAS mutant tumors.
Collapse
Affiliation(s)
- Xin Dai
- Department of Pharmaceutical Sciences, Mercer University, Atlanta, Georgia, United States of America
| | - Ying Jiang
- Department of Pharmaceutical Sciences, Mercer University, Atlanta, Georgia, United States of America
| | - Chalet Tan
- Department of Pharmaceutical Sciences, Mercer University, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
442
|
Wei J, Li F, Yang J, Liu X, Cho WC. MicroRNAs as regulators of airborne pollution-induced lung inflammation and carcinogenesis. Arch Toxicol 2015; 89:677-685. [PMID: 25667014 DOI: 10.1007/s00204-015-1462-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 01/08/2015] [Indexed: 12/14/2022]
Abstract
The increasing incidence of pulmonary inflammation and lung cancer, as well as exacerbation of pre-existing chronic lung diseases by exposure to airborne pollutants, e.g., particulate matter and cigarette smoke, is becoming a major public health concern in the world. However, the exact mechanisms of pulmonary injury from exposure to these airborne insults have not been fully elucidated. Nevertheless, accumulating evidence suggests that microRNAs (miRNAs) may play a unique role in the regulation of airborne agent-induced lung inflammation and carcinogenesis. Since epigenetic modifications are heritable and reversible, this may provide a new insight into the relationship of miRNAs and environmental pollution-related lung disorders. The aim of this review was to update our existing knowledge regarding the mechanisms by which airborne pollutants altering miRNA profiles in the lung, specifically for cigarette smoke and airborne particulate matter, and the potential biological roles of miRNAs in the initiation of pulmonary inflammation and lung cancer, as well as the regulation of underlying genetic susceptibility to these environmental stressors.
Collapse
Affiliation(s)
- Jun Wei
- Center of Medical Research, General Hospital, Ningxia Medical University, Yinchuan, Ningxia, 750004, People's Republic of China
| | | | | | | | | |
Collapse
|
443
|
Boeri M, Sestini S, Fortunato O, Verri C, Suatoni P, Pastorino U, Sozzi G. Recent advances of microRNA-based molecular diagnostics to reduce false-positive lung cancer imaging. Expert Rev Mol Diagn 2015; 15:801-13. [PMID: 25924864 DOI: 10.1586/14737159.2015.1041377] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Lung cancer is the leading cause of cancer deaths in the world. Advances in early detection crucial to enable timely curative surgery have been made in recent years. Cost-benefit profiles of lung cancer screening in smokers by low-dose computed tomography are still under evaluation. In particular, the high false-positive rates of low-dose computed tomography, together with the issue of overdiagnosis and the overall costs of screening, prompted a focus on the development of noninvasive complementary biomarkers to implement lung cancer screening. MicroRNA are a new class of blood-based biomarkers useful for early lung cancer detection and prognosis definition. Here, we discuss the seminal publications that reported circulating microRNA signatures with the greatest potential to impact clinical activity and patient care.
Collapse
Affiliation(s)
- Mattia Boeri
- Tumor Genomics Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, Milan 20133, Italy
| | | | | | | | | | | | | |
Collapse
|
444
|
Song J, Lee JE. miR-155 is involved in Alzheimer's disease by regulating T lymphocyte function. Front Aging Neurosci 2015; 7:61. [PMID: 25983691 PMCID: PMC4415416 DOI: 10.3389/fnagi.2015.00061] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 04/09/2015] [Indexed: 12/28/2022] Open
Abstract
Alzheimer’s disease (AD) is considered the most common cause of sporadic dementia. In AD, adaptive and innate immune responses play a crucial role in clearance of amyloid beta and maintenance of cognitive functions. In addition to other changes in the immune system, AD alters the T-cell responses that affect activation of glial cells, neuronal cells, macrophages, and secretion of pro-inflammatory cytokines. These changes in the immune system influence AD pathogenesis. Micro-RNA (miRNA)-155 is a multifunctional miRNA with a distinct expression profile. It is involved in diverse physiological and pathological mechanisms, such as immunity and inflammation. Recent studies indicate that miR-155 regulates T-cell functions during inflammation. In this article, we summarize recent studies describing the therapeutic potential of miR-155 via regulation of T cells in AD. Further, we propose that regulation of miR-155 might be a new protective approach against AD pathogenesis.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Yonsei University College of Medicine Seoul, South Korea
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine Seoul, South Korea ; Brain Korea 21 Plus Project for Medical Science and Brain Research Institute, Yonsei University College of Medicine Seoul, South Korea
| |
Collapse
|
445
|
Kan Q, Ding S, Yang Y, Zhou X. Expression profile of plasma microRNAs in premature infants with respiratory distress syndrome. Mol Med Rep 2015; 12:2858-64. [PMID: 25936520 DOI: 10.3892/mmr.2015.3699] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 03/20/2015] [Indexed: 11/05/2022] Open
Abstract
As well-known regulators of gene expression, microRNAs (miRNAs) are important not only in cell proliferation and differentiation, but also in tumorigenesis and organ development. It has been estimated that miRNAs may be responsible for regulating the expression of almost one third of the human genome. Simultaneously, with advances in neonatal care in the clinic, an increased number of premature infants are being saved and, thus, respiratory distress syndrome (RDS) has become more common. However, previous non-miRNA studies have suggested their connection with RDS. In the present study, a miRNA microarray, including >1,891 capture probes was used to compared the expression profiles of plasma miRNAs between RDS and control groups. miRNAs, which were observed to have consistent fold-changes (fold-change ≥ 1.3) between the two groups were selected and validated using reverse transcription-quantitative polymerase chain reaction. As a result, 171 differentially expressed miRNAs were identified, including two upregulated and seven downregulated miRNAs. Of these miRNAs, four were selected as having higher fold-changes between the two groups. This is the first time, to the best of our knowledge, that these nine miRNAs have been reported in RDS. It was hypothesized that these novel miRNAs may be important in RDS, and may provide meaningful biomarkers for the diagnosis of RDS.
Collapse
Affiliation(s)
- Qing Kan
- Department of Neonatology, Nanjing Children's Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China
| | - Sufang Ding
- Department of Neonatology, Huaian Maternity and Child Health Care Hospital, Huaian, Jiangsu 223002, P.R. China
| | - Yang Yang
- Department of Neonatology, Nanjing Children's Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China
| | - Xiaoyu Zhou
- Department of Neonatology, Nanjing Children's Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
446
|
Tang BB, Liu SY, Zhan YU, Wei LQ, Mao XL, Wang J, Li LI, Lu ZX. microRNA-218 expression and its association with the clinicopathological characteristics of patients with cervical cancer. Exp Ther Med 2015; 10:269-274. [PMID: 26170947 DOI: 10.3892/etm.2015.2455] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 04/13/2015] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the expression of microRNA-218 (miRNA-218) in the serum and cervical tissue and its association with the clinicopathological features of cervical cancer (CC). The expression of miRNA-218 was detected in the serum and cervical tissue of 112 patients with CC and 50 age-matched hysteromyoma patients via the reverse transcription-quantitative polymerase chain reaction. The clinical data were collected and the association between the expression of miRNA-218 and the clinicopathological characteristics of the patients was analyzed. The expression of miRNA-218 in the cancer group was significantly decreased in the cervical tissue and serum compared with that in the control group (P<0.001). The decreased expression of miRNA-218 was associated with a later International Federation of Gynecology and Obstetrics stage, a more invasive pathological type and lymphatic node metastasis but not with age, age at menarche, menopausal status, number of pregnancies and deliveries, family history of cancer or tumor size. In conclusion, miRNA-218 was found to be downregulated in the cancer tissue and serum of the patients with CC. The decreased expression of miRNA-218 in CC was associated with the invasiveness of the tumor.
Collapse
Affiliation(s)
- Bei-Bei Tang
- Department of Clinical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Shui-Yi Liu
- Department of Clinical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Y U Zhan
- Department of Clinical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Li-Qing Wei
- Department of Clinical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Xiao-Lu Mao
- Department of Clinical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Jin Wang
- Department of Clinical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - L I Li
- Department of Gynecology and Obstetrics, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Zhong-Xin Lu
- Department of Clinical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| |
Collapse
|
447
|
Zhu X, Wu L, Yao J, Jiang H, Wang Q, Yang Z, Wu F. MicroRNA let-7c Inhibits Cell Proliferation and Induces Cell Cycle Arrest by Targeting CDC25A in Human Hepatocellular Carcinoma. PLoS One 2015; 10:e0124266. [PMID: 25909324 PMCID: PMC4409336 DOI: 10.1371/journal.pone.0124266] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 03/12/2015] [Indexed: 01/07/2023] Open
Abstract
Down-regulation of the microRNA let-7c plays an important role in the pathogenesis of human hepatocellular carcinoma (HCC). The aim of the present study was to determine whether the cell cycle regulator CDC25A is involved in the antitumor effect of let-7c in HCC. The expression levels of let-7c in HCC cell lines were examined by quantitative real-time PCR, and a let-7c agomir was transfected into HCC cells to overexpress let-7c. The effects of let-7c on HCC proliferation, apoptosis and cell cycle were analyzed. The in vivo tumor-inhibitory efficacy of let-7c was evaluated in a xenograft mouse model of HCC. Luciferase reporter assays and western blotting were conducted to identify the targets of let-7c and to determine the effects of let-7c on CDC25A, CyclinD1, CDK6, pRb and E2F2 expression. The results showed that the expression levels of let-7c were significantly decreased in HCC cell lines. Overexpression of let-7c repressed cell growth, induced cell apoptosis, led to G1 cell cycle arrest in vitro, and suppressed tumor growth in a HepG2 xenograft model in vivo. The luciferase reporter assay showed that CDC25A was a direct target of let-7c, and that let-7c inhibited the expression of CDC25A protein by directly targeting its 3' UTR. Restoration of CDC25A induced a let-7c-mediated G1-to-S phase transition. Western blot analysis demonstrated that overexpression of let-7c decreased CyclinD1, CDK6, pRb and E2F2 protein levels. In conclusion, this study indicates that let-7c suppresses HCC progression, possibly by directly targeting the cell cycle regulator CDC25A and indirectly affecting its downstream target molecules. Let-7c may therefore be an effective therapeutic target for HCC.
Collapse
Affiliation(s)
- Xiuming Zhu
- Department of Surgical Oncology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Department of Oncology, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Lingjiao Wu
- State key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jian Yao
- State key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Han Jiang
- Department of Surgical Oncology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Qiangfeng Wang
- Department of Surgical Oncology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | | | - Fusheng Wu
- Department of Surgical Oncology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- * E-mail:
| |
Collapse
|
448
|
Hou J, Wang F, Kong P, Yu PKN, Wang H, Han W. Gene profiling characteristics of radioadaptive response in AG01522 normal human fibroblasts. PLoS One 2015; 10:e0123316. [PMID: 25886619 PMCID: PMC4401551 DOI: 10.1371/journal.pone.0123316] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 03/02/2015] [Indexed: 12/20/2022] Open
Abstract
Radioadaptive response (RAR) in mammalian cells refers to the phenomenon where a low-dose ionizing irradiation alters the gene expression profiles, and protects the cells from the detrimental effects of a subsequent high dose exposure. Despite the completion of numerous experimental studies on RAR, the underlying mechanism has remained unclear. In this study, we aimed to have a comprehensive investigation on the RAR induced in the AG01522 human fibroblasts first exposed to 5 cGy (priming dose) and then followed by 2 Gy (challenge dose) of X-ray through comparisons to those cells that had only received a single 2 Gy dose. We studied how the priming dose affected the expression of gene transcripts, and to identify transcripts or pathways that were associated with the reduced chromosomal damages (in terms of the number of micronuclei) after application of the challenging dose. Through the mRNA and microRNA microarray analyses, the transcriptome alteration in AG01522 cells was examined, and the significantly altered genes were identified for different irradiation procedures using bioinformatics approaches. We observed that a low-dose X-ray exposure produced an alert, triggering and altering cellular responses to defend against subsequent high dose-induced damages, and accelerating the cell repair process. Moreover, the p53 signaling pathway was found to play critial roles in regulating DNA damage responses at the early stage after application of the challenging dose, particularly in the RAR group. Furthermore, microRNA analyses also revealed that cell communication and intercellular signaling transduction played important roles after low-dose irradiation. We conclude that RAR benefits from the alarm mechanisms triggered by a low-dose priming radation dose.
Collapse
Affiliation(s)
- Jue Hou
- Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Fan Wang
- Department of Radiation Oncology, First Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Peizhong Kong
- Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Peter K. N. Yu
- Department of Physics and Materials Science, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong
| | - Hongzhi Wang
- Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
- Cancer Hospital, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Wei Han
- Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
- Cancer Hospital, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| |
Collapse
|
449
|
Liu K, Zhang C, Li T, Ding Y, Tu T, Zhou F, Qi W, Chen H, Sun X. Let-7a inhibits growth and migration of breast cancer cells by targeting HMGA1. Int J Oncol 2015; 46:2526-34. [PMID: 25846193 DOI: 10.3892/ijo.2015.2949] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 03/05/2015] [Indexed: 11/06/2022] Open
Abstract
Let-7 is one of the earliest discovered microRNAs (miRNAs) and has been reported to regulate self renewal and tumorigenicity of breast cancer cells. Let-7a is a member of this family and its function has not been fully characterized in breast cancer. First, total RNAs of breast cancer cells (MDA-MB-231, MCF-7), breast cancer tissues and corresponding adjacent normal tissues were extracted and used to detect let-7a expression by qRT-PCR. Secondly, the effects of let-7a on proliferation, colony formation, migration and invasion of breast cancer cells were assessed by in vitro cell culture experiments. Finally, western blotting was performed to demonstrate how let-7a regulated HMGA1 expression. We found that let-7a expression was significantly lower in breast cancer cells and breast cancer tissues compared to corresponding adjacent normal tissues. Cell proliferation, colony formation, migration and invasion were decreased after overexpression of let-7a in breast cancer cells and vice versa. Furthermore, we identified the high mobility group A1 (HMGA1) as a potential target gene of let-7a. Protein expression of the target gene was significantly downregulated in let-7a mimic transfected breast cancer cells and significantly upregulated in let-7a inhibitor transfected breast cancer cells. Our data suggest that let-7a plays an important role as a tumor suppressor gene by targeting HMGA1, which may open novel perspectives for clinical treatments against breast cancer.
Collapse
Affiliation(s)
- Kui Liu
- School of Medicine, Jiangsu University, Jiangsu Key Laboratory of Clinical Laboratory Medicine, Zhenjiang, Jiangsu 212013, P.R. China
| | - Chunfu Zhang
- The Second People's Hospital of Kunshan, Kunshan, Jiangsu 215300, P.R. China
| | - Tao Li
- School of Medicine, Jiangsu University, Jiangsu Key Laboratory of Clinical Laboratory Medicine, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yanling Ding
- School of Medicine, Jiangsu University, Jiangsu Key Laboratory of Clinical Laboratory Medicine, Zhenjiang, Jiangsu 212013, P.R. China
| | - Tao Tu
- School of Medicine, Jiangsu University, Jiangsu Key Laboratory of Clinical Laboratory Medicine, Zhenjiang, Jiangsu 212013, P.R. China
| | - Fangfang Zhou
- School of Medicine, Jiangsu University, Jiangsu Key Laboratory of Clinical Laboratory Medicine, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wenkai Qi
- School of Medicine, Jiangsu University, Jiangsu Key Laboratory of Clinical Laboratory Medicine, Zhenjiang, Jiangsu 212013, P.R. China
| | - Huabiao Chen
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Xiaochun Sun
- School of Medicine, Jiangsu University, Jiangsu Key Laboratory of Clinical Laboratory Medicine, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
450
|
Inhibition of RAC1-GEF DOCK3 by miR-512-3p contributes to suppression of metastasis in non-small cell lung cancer. Int J Biochem Cell Biol 2015; 61:103-14. [DOI: 10.1016/j.biocel.2015.02.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 01/26/2015] [Accepted: 02/06/2015] [Indexed: 12/11/2022]
|