351
|
Kim D, Thairu MW, Hansen AK. Novel Insights into Insect-Microbe Interactions-Role of Epigenomics and Small RNAs. FRONTIERS IN PLANT SCIENCE 2016; 7:1164. [PMID: 27540386 PMCID: PMC4972996 DOI: 10.3389/fpls.2016.01164] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/20/2016] [Indexed: 05/23/2023]
Abstract
It has become increasingly clear that microbes form close associations with the vast majority of animal species, especially insects. In fact, an array of diverse microbes is known to form shared metabolic pathways with their insect hosts. A growing area of research in insect-microbe interactions, notably for hemipteran insects and their mutualistic symbionts, is to elucidate the regulation of this inter-domain metabolism. This review examines two new emerging mechanisms of gene regulation and their importance in host-microbe interactions. Specifically, we highlight how the incipient areas of research on regulatory "dark matter" such as epigenomics and small RNAs, can play a pivotal role in the evolution of both insect and microbe gene regulation. We then propose specific models of how these dynamic forms of gene regulation can influence insect-symbiont-plant interactions. Future studies in this area of research will give us a systematic understanding of how these symbiotic microbes and animals reciprocally respond to and regulate their shared metabolic processes.
Collapse
|
352
|
microRNA-132/212 deficiency enhances Aβ production and senile plaque deposition in Alzheimer's disease triple transgenic mice. Sci Rep 2016; 6:30953. [PMID: 27484949 PMCID: PMC4971468 DOI: 10.1038/srep30953] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 07/11/2016] [Indexed: 01/02/2023] Open
Abstract
The abnormal regulation of amyloid-β (Aβ) metabolism (e.g., production, cleavage, clearance) plays a central role in Alzheimer’s disease (AD). Among endogenous factors believed to participate in AD progression are the small regulatory non-coding microRNAs (miRs). In particular, the miR-132/212 cluster is severely reduced in the AD brain. In previous studies we have shown that miR-132/212 deficiency in mice leads to impaired memory and enhanced Tau pathology as seen in AD patients. Here we demonstrate that the genetic deletion of miR-132/212 promotes Aβ production and amyloid (senile) plaque formation in triple transgenic AD (3xTg-AD) mice. Using RNA-Seq and bioinformatics, we identified genes of the miR-132/212 network with documented roles in the regulation of Aβ metabolism, including Tau, Mapk, and Sirt1. Consistent with these findings, we show that the modulation of miR-132, or its target Sirt1, can directly regulate Aβ production in cells. Finally, both miR-132 and Sirt1 levels correlated with Aβ load in humans. Overall, our results support the hypothesis that the miR-132/212 network, including Sirt1 and likely other target genes, contributes to abnormal Aβ metabolism and senile plaque deposition in AD. This study strengthens the importance of miR-dependent networks in neurodegenerative disorders, and opens the door to multifactorial drug targets of AD by targeting Aβ and Tau.
Collapse
|
353
|
Bhajun R, Guyon L, Gidrol X. MicroRNA degeneracy and pluripotentiality within a Lavallière-tie architecture confers robustness to gene expression networks. Cell Mol Life Sci 2016; 73:2821-7. [PMID: 27038488 PMCID: PMC4937071 DOI: 10.1007/s00018-016-2186-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 02/24/2016] [Accepted: 03/18/2016] [Indexed: 11/03/2022]
Abstract
Modularity, feedback control, functional redundancy and bowtie architecture have been proposed as key factors that confer robustness to complex biological systems. MicroRNAs (miRNAs) are highly conserved but functionally dispensable. These antinomic properties suggest that miRNAs fine-tune gene expression rather than act as genetic switches. We synthesize published and unpublished data and hypothesize that miRNA pluripotentiality acts to buffer gene expression, while miRNA degeneracy tunes the expression of targets, thus providing robustness to gene expression networks. Furthermore, we propose a Lavallière-tie architecture by integrating signal transduction, miRNAs and protein expression data to model complex gene expression networks.
Collapse
Affiliation(s)
- Ricky Bhajun
- CEA, BIG, BGE, 17, rue des Martyrs, 38000, Grenoble, France
- University Grenoble Alpes, BGE, 38000, Grenoble, France
- INSERM, U1038, 38000, Grenoble, France
| | - Laurent Guyon
- CEA, BIG, BGE, 17, rue des Martyrs, 38000, Grenoble, France
- University Grenoble Alpes, BGE, 38000, Grenoble, France
- INSERM, U1038, 38000, Grenoble, France
| | - Xavier Gidrol
- CEA, BIG, BGE, 17, rue des Martyrs, 38000, Grenoble, France.
- University Grenoble Alpes, BGE, 38000, Grenoble, France.
- INSERM, U1038, 38000, Grenoble, France.
| |
Collapse
|
354
|
Chang TC, Pertea M, Lee S, Salzberg SL, Mendell JT. Genome-wide annotation of microRNA primary transcript structures reveals novel regulatory mechanisms. Genome Res 2016; 25:1401-9. [PMID: 26290535 PMCID: PMC4561498 DOI: 10.1101/gr.193607.115] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Precise regulation of microRNA (miRNA) expression is critical for diverse physiologic and pathophysiologic processes. Nevertheless, elucidation of the mechanisms through which miRNA expression is regulated has been greatly hindered by the incomplete annotation of primary miRNA (pri-miRNA) transcripts. While a subset of miRNAs are hosted in protein-coding genes, the majority of pri-miRNAs are transcribed as poorly characterized noncoding RNAs that are 10's to 100's of kilobases in length and low in abundance due to efficient processing by the endoribonuclease DROSHA, which initiates miRNA biogenesis. Accordingly, these transcripts are poorly represented in existing RNA-seq data sets and exhibit limited and inaccurate annotation in current transcriptome assemblies. To overcome these challenges, we developed an experimental and computational approach that allows genome-wide detection and mapping of pri-miRNA structures. Deep RNA-seq in cells expressing dominant-negative DROSHA resulted in much greater coverage of pri-miRNA transcripts compared with standard RNA-seq. A computational pipeline was developed that produces highly accurate pri-miRNA assemblies, as confirmed by extensive validation. This approach was applied to a panel of human and mouse cell lines, providing pri-miRNA transcript structures for 1291/1871 human and 888/1181 mouse miRNAs, including 594 human and 425 mouse miRNAs that fall outside protein-coding genes. These new assemblies uncovered unanticipated features and new potential regulatory mechanisms, including links between pri-miRNAs and distant protein-coding genes, alternative pri-miRNA splicing, and transcripts carrying subsets of miRNAs encoded by polycistronic clusters. These results dramatically expand our understanding of the organization of miRNA-encoding genes and provide a valuable resource for the study of mammalian miRNA regulation.
Collapse
Affiliation(s)
- Tsung-Cheng Chang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Mihaela Pertea
- Center for Computational Biology, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | - Sungyul Lee
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Steven L Salzberg
- Center for Computational Biology, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland 21205, USA; Departments of Biomedical Engineering, Computer Science, and Biostatistics, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | - Joshua T Mendell
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
355
|
MiR-130a regulates neurite outgrowth and dendritic spine density by targeting MeCP2. Protein Cell 2016; 7:489-500. [PMID: 27245166 PMCID: PMC4930766 DOI: 10.1007/s13238-016-0272-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 04/13/2016] [Indexed: 01/03/2023] Open
Abstract
MicroRNAs (miRNAs) are critical for both development and function of the central nervous system. Significant evidence suggests that abnormal expression of miRNAs is associated with neurodevelopmental disorders. MeCP2 protein is an epigenetic regulator repressing or activating gene transcription by binding to methylated DNA. Both loss-of-function and gain-of-function mutations in the MECP2 gene lead to neurodevelopmental disorders such as Rett syndrome, autism and MECP2 duplication syndrome. In this study, we demonstrate that miR-130a inhibits neurite outgrowth and reduces dendritic spine density as well as dendritic complexity. Bioinformatics analyses, cell cultures and biochemical experiments indicate that miR-130a targets MECP2 and down-regulates MeCP2 protein expression. Furthermore, expression of the wild-type MeCP2, but not a loss-of-function mutant, rescues the miR-130a-induced phenotype. Our study uncovers the MECP2 gene as a previous unknown target for miR-130a, supporting that miR-130a may play a role in neurodevelopment by regulating MeCP2. Together with data from other groups, our work suggests that a feedback regulatory mechanism involving both miR-130a and MeCP2 may serve to ensure their appropriate expression and function in neural development.
Collapse
|
356
|
Bavia L, Mosimann ALP, Aoki MN, Duarte Dos Santos CN. A glance at subgenomic flavivirus RNAs and microRNAs in flavivirus infections. Virol J 2016; 13:84. [PMID: 27233361 PMCID: PMC4884392 DOI: 10.1186/s12985-016-0541-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/17/2016] [Indexed: 11/10/2022] Open
Abstract
The family Flaviviridae comprises a wide variety of viruses that are distributed worldwide, some of which are associated with high rates of morbidity and mortality. There are neither vaccines nor antivirals for most flavivirus infections, reinforcing the importance of research on different aspects of the viral life cycle. During infection, cytoplasmic accumulation of RNA fragments mainly originating from the 3' UTRs, which have been designated subgenomic flavivirus RNAs (sfRNAs), has been detected. It has been shown that eukaryotic exoribonucleases are involved in viral sfRNA production. Additionally, viral and human small RNAs (sRNAs) have also been found in flavivirus-infected cells, especially microRNAs (miRNAs). miRNAs were first described in eukaryotic cells and in a mature and functional state present as single-stranded 18-24 nt RNA fragments. Their main function is the repression of translation through base pairing with cellular mRNAs, besides other functions, such as mRNA degradation. Canonical miRNA biogenesis involves Drosha and Dicer, however miRNA can also be generated by alternative pathways. In the case of flaviviruses, alternative pathways have been suggested. Both sfRNAs and miRNAs are involved in viral infection and host cell response modulation, representing interesting targets of antiviral strategies. In this review, we focus on the generation and function of viral sfRNAs, sRNAs and miRNAs in West Nile, dengue, Japanese encephalitis, Murray Valley encephalitis and yellow fever infections, as well as their roles in viral replication, translation and cell immune response evasion. We also give an overview regarding other flaviviruses and the generation of cellular miRNAs during infection.
Collapse
Affiliation(s)
- Lorena Bavia
- Laboratório de Virologia Molecular, Instituto Carlos Chagas (ICC/FIOCRUZ-PR), Rua Prof. Algacyr Munhoz Mader 3775, CIC, CEP: 81350-010, Curitiba, Paraná, Brazil
| | - Ana Luiza Pamplona Mosimann
- Laboratório de Virologia Molecular, Instituto Carlos Chagas (ICC/FIOCRUZ-PR), Rua Prof. Algacyr Munhoz Mader 3775, CIC, CEP: 81350-010, Curitiba, Paraná, Brazil
| | - Mateus Nóbrega Aoki
- Laboratório de Virologia Molecular, Instituto Carlos Chagas (ICC/FIOCRUZ-PR), Rua Prof. Algacyr Munhoz Mader 3775, CIC, CEP: 81350-010, Curitiba, Paraná, Brazil
| | - Claudia Nunes Duarte Dos Santos
- Laboratório de Virologia Molecular, Instituto Carlos Chagas (ICC/FIOCRUZ-PR), Rua Prof. Algacyr Munhoz Mader 3775, CIC, CEP: 81350-010, Curitiba, Paraná, Brazil.
| |
Collapse
|
357
|
Abstract
MicroRNAs are involved in almost all biological processes and have emerged as regulators of signaling pathways. We show that miRNA target genes and pathway genes are not uniformly expressed across human tissues. To capture tissue specific effects, we developed a novel methodology for tissue specific pathway analysis of miRNAs. We incorporated the most recent and highest quality miRNA targeting data (TargetScan and StarBase), RNA-seq based gene expression data (EBI Expression Atlas) and multiple new pathway data sources to increase the biological relevance of the predicted miRNA-pathway associations. We identified new potential roles of miR-199a-3p, miR-199b-3p and the miR-200 family in hepatocellular carcinoma, involving the regulation of metastasis through MAPK and Wnt signaling. Also, an association of miR-571 and Notch signaling in liver fibrosis was proposed. To facilitate data update and future extensions of our tool, we developed a flexible database backend using the graph database neo4j. The new backend as well as the novel methodology were included in the updated miTALOS v2, a tool that provides insights into tissue specific miRNA regulation of biological pathways. miTALOS v2 is available at http://mips.helmholtz-muenchen.de/mitalos.
Collapse
|
358
|
Gjymishka A, Pi L, Oh SH, Jorgensen M, Liu C, Protopapadakis Y, Patel A, Petersen BE. miR-133b Regulation of Connective Tissue Growth Factor: A Novel Mechanism in Liver Pathology. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1092-102. [PMID: 26945106 DOI: 10.1016/j.ajpath.2015.12.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 11/30/2015] [Accepted: 12/28/2015] [Indexed: 02/07/2023]
Abstract
miRNAs are involved in liver regeneration, and their expression is dysregulated in hepatocellular carcinoma (HCC). Connective tissue growth factor (CTGF), a direct target of miR-133b, is crucial in the ductular reaction (DR)/oval cell (OC) response for generating new hepatocyte lineages during liver injury in the context of hepatotoxin-inhibited hepatocyte proliferation. Herein, we investigate whether miR-133b regulation of CTGF influences HCC cell proliferation and migration, and DR/OC response. We analyzed miR-133b expression and found it to be down-regulated in HCC patient samples and induced in the rat DR/OC activation model of 2-acetylaminofluorene with partial hepatectomy. Furthermore, overexpression of miR-133b via adenoviral system in vitro led to decreased CTGF expression and reduced proliferation and Transwell migration of both HepG2 HCC cells and WBF-344 rat OCs. In vivo, overexpression of miR-133b in DR/OC activation models of 2-acetylaminofluorene with partial hepatectomy in rats, and 3,5-diethoxycarbonyl-1,4-dihydrocollidine in mice, led to down-regulation of CTGF expression and OC proliferation. Collectively, these results show that miR-133b regulation of CTGF is a novel mechanism critical for the proliferation and migration of HCC cells and OC response.
Collapse
Affiliation(s)
- Altin Gjymishka
- Department of Pediatrics, University of Florida, Gainesville, Florida
| | - Liya Pi
- Department of Pediatrics, University of Florida, Gainesville, Florida
| | - Seh-Hoon Oh
- Department of Pediatrics, University of Florida, Gainesville, Florida
| | - Marda Jorgensen
- Department of Pediatrics, University of Florida, Gainesville, Florida
| | - Chen Liu
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida
| | | | - Ashnee Patel
- Department of Pediatrics, University of Florida, Gainesville, Florida
| | - Bryon E Petersen
- Department of Pediatrics, University of Florida, Gainesville, Florida.
| |
Collapse
|
359
|
Jeyabal P, Thandavarayan RA, Joladarashi D, Suresh Babu S, Krishnamurthy S, Bhimaraj A, Youker KA, Kishore R, Krishnamurthy P. MicroRNA-9 inhibits hyperglycemia-induced pyroptosis in human ventricular cardiomyocytes by targeting ELAVL1. Biochem Biophys Res Commun 2016; 471:423-9. [PMID: 26898797 DOI: 10.1016/j.bbrc.2016.02.065] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 02/16/2016] [Indexed: 02/04/2023]
Abstract
Diabetic cardiomyopathy is a common complication in patients with diabetes and is associated with underlying chronic inflammation and cardiac cell death, subsequently leading to heart failure (HF). ELAV-like protein 1 (ELAVL1) plays a critical role in the progression of inflammation and HF. However the role of ELAVL-1 in inflammation induced cardiac cell death (pyroptosis) under hyperglycemic condition remains elusive. Our data demonstrates that ELAVL1 expression augmented with a concomitant increase in caspase-1 and IL-1 beta expression in human hearts and human ventricular cardiomyocytes under hyperglycemic condition. Furthermore, ELAVL1 knockdown abrogates TNF-α induced canonical pyroptosis via NLRP3, caspase-1 and IL-1beta suppression. Bioinformatics analysis and target validation assays showed that miR-9 directly targets ELAVL1. Interestingly, miRNA-9 expression significantly reduced in high glucose treated cardiomyocytes and in human diabetic hearts. Inhibition of miR-9 upregulates ELAVL1 expression and activates caspase-1. Alternatively, treatment with miR-9 mimics attenuates hyperglycemia-induced ELAVL1 and inhibits cardiomyocyte pyroptosis. Taken together our study highlights the potential therapeutic implications of targeting miR-9/ELAVL1 in preventing cardiomyocyte cell loss during HF in diabetics.
Collapse
Affiliation(s)
- Prince Jeyabal
- Department of Cardiovascular Sciences, Centre for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Rajarajan A Thandavarayan
- Department of Cardiovascular Sciences, Centre for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Darukeshwara Joladarashi
- Department of Cardiovascular Sciences, Centre for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Sahana Suresh Babu
- Department of Cardiovascular Sciences, Centre for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Shashirekha Krishnamurthy
- Department of Cardiovascular Sciences, Centre for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Arvind Bhimaraj
- Houston Methodist DeBakey Heart & Vascular Center, Houston Methodist Hospital, Houston, TX, USA
| | - Keith A Youker
- Houston Methodist DeBakey Heart & Vascular Center, Houston Methodist Hospital, Houston, TX, USA
| | - Raj Kishore
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Prasanna Krishnamurthy
- Department of Cardiovascular Sciences, Centre for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; Cell and Developmental Biology, Department of Cardiothoracic Surgery, Weill Cornell Medical College of Cornell University, New York, NY, USA.
| |
Collapse
|
360
|
Liew YJ, Ryu T, Aranda M, Ravasi T. miRNA Repertoires of Demosponges Stylissa carteri and Xestospongia testudinaria. PLoS One 2016; 11:e0149080. [PMID: 26871907 PMCID: PMC4752309 DOI: 10.1371/journal.pone.0149080] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 01/26/2016] [Indexed: 11/23/2022] Open
Abstract
MicroRNAs (miRNAs) are small regulatory RNAs that are involved in many biological process in eukaryotes. They play a crucial role in modulating genetic expression of their targets, which makes them integral components of transcriptional regulatory networks. As sponges (phylum Porifera) are commonly considered the most basal metazoan, the in-depth capture of miRNAs from these organisms provides additional clues to the evolution of miRNA families in metazoans. Here, we identified the core proteins involved in the biogenesis of miRNAs, and obtained evidence for bona fide miRNA sequences for two marine sponges Stylissa carteri and Xestospongia testudinaria (11 and 19 respectively). Our analysis identified several miRNAs that are conserved amongst demosponges, and revealed that all of the novel miRNAs identified in these two species are specific to the class Demospongiae.
Collapse
Affiliation(s)
- Yi Jin Liew
- Division of Biological and Environmental Sciences & Engineering, King Abdullah University of Science and Technology, Thuwal, 23955–6900, Kingdom of Saudi Arabia
- Red Sea Research Center, King Abdullah University of Science and Technology, Thuwal, 23955–6900, Kingdom of Saudi Arabia
| | - Taewoo Ryu
- Division of Biological and Environmental Sciences & Engineering, King Abdullah University of Science and Technology, Thuwal, 23955–6900, Kingdom of Saudi Arabia
- KAUST Environmental Epigenetics Program (KEEP), King Abdullah University of Science and Technology, Thuwal, 23955–6900, Kingdom of Saudi Arabia
| | - Manuel Aranda
- Division of Biological and Environmental Sciences & Engineering, King Abdullah University of Science and Technology, Thuwal, 23955–6900, Kingdom of Saudi Arabia
- Red Sea Research Center, King Abdullah University of Science and Technology, Thuwal, 23955–6900, Kingdom of Saudi Arabia
- * E-mail: (MA); (TR)
| | - Timothy Ravasi
- Division of Biological and Environmental Sciences & Engineering, King Abdullah University of Science and Technology, Thuwal, 23955–6900, Kingdom of Saudi Arabia
- KAUST Environmental Epigenetics Program (KEEP), King Abdullah University of Science and Technology, Thuwal, 23955–6900, Kingdom of Saudi Arabia
- * E-mail: (MA); (TR)
| |
Collapse
|
361
|
Wong JJL, Au AYM, Gao D, Pinello N, Kwok CT, Thoeng A, Lau KA, Gordon JEA, Schmitz U, Feng Y, Nguyen TV, Middleton R, Bailey CG, Holst J, Rasko JEJ, Ritchie W. RBM3 regulates temperature sensitive miR-142-5p and miR-143 (thermomiRs), which target immune genes and control fever. Nucleic Acids Res 2016; 44:2888-97. [PMID: 26825461 PMCID: PMC4824108 DOI: 10.1093/nar/gkw041] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 01/13/2016] [Indexed: 12/27/2022] Open
Abstract
Fever is commonly used to diagnose disease and is consistently associated with increased mortality in critically ill patients. However, the molecular controls of elevated body temperature are poorly understood. We discovered that the expression of RNA-binding motif protein 3 (RBM3), known to respond to cold stress and to modulate microRNA (miRNA) expression, was reduced in 30 patients with fever, and in THP-1-derived macrophages maintained at a fever-like temperature (40°C). Notably, RBM3 expression is reduced during fever whether or not infection is demonstrable. Reduced RBM3 expression resulted in increased expression of RBM3-targeted temperature-sensitive miRNAs, we termed thermomiRs. ThermomiRs such as miR-142–5p and miR-143 in turn target endogenous pyrogens including IL-6, IL6ST, TLR2, PGE2 and TNF to complete a negative feedback mechanism, which may be crucial to prevent pathological hyperthermia. Using normal PBMCs that were exogenously exposed to fever-like temperature (40°C), we further demonstrate the trend by which decreased levels of RBM3 were associated with increased levels of miR-142–5p and miR-143 and vice versa over a 24 h time course. Collectively, our results indicate the existence of a negative feedback loop that regulates fever via reduced RBM3 levels and increased expression of miR-142–5p and miR-143.
Collapse
Affiliation(s)
- Justin J-L Wong
- Gene & Stem Cell Therapy Program, Centenary Institute, Camperdown 2050, Australia Sydney Medical School, University of Sydney, NSW 2006, Australia
| | - Amy Y M Au
- Gene & Stem Cell Therapy Program, Centenary Institute, Camperdown 2050, Australia Sydney Medical School, University of Sydney, NSW 2006, Australia
| | - Dadi Gao
- Gene & Stem Cell Therapy Program, Centenary Institute, Camperdown 2050, Australia Sydney Medical School, University of Sydney, NSW 2006, Australia Bioinformatics Laboratory, Centenary Institute, Camperdown 2050, Australia
| | - Natalia Pinello
- Gene & Stem Cell Therapy Program, Centenary Institute, Camperdown 2050, Australia Sydney Medical School, University of Sydney, NSW 2006, Australia
| | - Chau-To Kwok
- Gene & Stem Cell Therapy Program, Centenary Institute, Camperdown 2050, Australia Sydney Medical School, University of Sydney, NSW 2006, Australia
| | - Annora Thoeng
- Gene & Stem Cell Therapy Program, Centenary Institute, Camperdown 2050, Australia Sydney Medical School, University of Sydney, NSW 2006, Australia
| | - Katherine A Lau
- Gene & Stem Cell Therapy Program, Centenary Institute, Camperdown 2050, Australia Sydney Medical School, University of Sydney, NSW 2006, Australia
| | - Jane E A Gordon
- Gene & Stem Cell Therapy Program, Centenary Institute, Camperdown 2050, Australia Sydney Medical School, University of Sydney, NSW 2006, Australia
| | - Ulf Schmitz
- Gene & Stem Cell Therapy Program, Centenary Institute, Camperdown 2050, Australia Sydney Medical School, University of Sydney, NSW 2006, Australia
| | - Yue Feng
- Gene & Stem Cell Therapy Program, Centenary Institute, Camperdown 2050, Australia Sydney Medical School, University of Sydney, NSW 2006, Australia
| | - Trung V Nguyen
- Gene & Stem Cell Therapy Program, Centenary Institute, Camperdown 2050, Australia Sydney Medical School, University of Sydney, NSW 2006, Australia
| | - Robert Middleton
- Gene & Stem Cell Therapy Program, Centenary Institute, Camperdown 2050, Australia Sydney Medical School, University of Sydney, NSW 2006, Australia Bioinformatics Laboratory, Centenary Institute, Camperdown 2050, Australia
| | - Charles G Bailey
- Gene & Stem Cell Therapy Program, Centenary Institute, Camperdown 2050, Australia Sydney Medical School, University of Sydney, NSW 2006, Australia
| | - Jeff Holst
- Sydney Medical School, University of Sydney, NSW 2006, Australia Origins of Cancer Program, Centenary Institute, Camperdown 2050, Australia
| | - John E J Rasko
- Gene & Stem Cell Therapy Program, Centenary Institute, Camperdown 2050, Australia Sydney Medical School, University of Sydney, NSW 2006, Australia Cell and Molecular Therapies, Royal Prince Alfred Hospital, Camperdown 2050, Australia
| | - William Ritchie
- Gene & Stem Cell Therapy Program, Centenary Institute, Camperdown 2050, Australia Sydney Medical School, University of Sydney, NSW 2006, Australia Bioinformatics Laboratory, Centenary Institute, Camperdown 2050, Australia CNRS, UMR 5203, Montpellier 34094, France
| |
Collapse
|
362
|
Amin ND, Bai G, Klug JR, Bonanomi D, Pankratz MT, Gifford WD, Hinckley CA, Sternfeld MJ, Driscoll SP, Dominguez B, Lee KF, Jin X, Pfaff SL. Loss of motoneuron-specific microRNA-218 causes systemic neuromuscular failure. Science 2016; 350:1525-9. [PMID: 26680198 DOI: 10.1126/science.aad2509] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Dysfunction of microRNA (miRNA) metabolism is thought to underlie diseases affecting motoneurons. One miRNA, miR-218, is abundantly and selectively expressed by developing and mature motoneurons. Here we show that mutant mice lacking miR-218 die neonatally and exhibit neuromuscular junction defects, motoneuron hyperexcitability, and progressive motoneuron cell loss, all of which are hallmarks of motoneuron diseases such as amyotrophic lateral sclerosis and spinal muscular atrophy. Gene profiling reveals that miR-218 modestly represses a cohort of hundreds of genes that are neuronally enriched but are not specific to a single neuron subpopulation. Thus, the set of messenger RNAs targeted by miR-218, designated TARGET(218), defines a neuronal gene network that is selectively tuned down in motoneurons to prevent neuromuscular failure and neurodegeneration.
Collapse
Affiliation(s)
- Neal D Amin
- Howard Hughes Medical Institute and Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA. Medical Scientist Training Program, University of California, San Diego (UCSD), 9500 Gilman Drive, La Jolla, CA 92037, USA. Biomedical Sciences Graduate Program, UCSD, 9500 Gilman Drive, La Jolla, CA 92037, USA
| | - Ge Bai
- Howard Hughes Medical Institute and Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jason R Klug
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Dario Bonanomi
- Howard Hughes Medical Institute and Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Matthew T Pankratz
- Howard Hughes Medical Institute and Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Wesley D Gifford
- Howard Hughes Medical Institute and Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA. Medical Scientist Training Program, University of California, San Diego (UCSD), 9500 Gilman Drive, La Jolla, CA 92037, USA. Neurosciences Graduate Program, UCSD, 9500 Gilman Drive, La Jolla, CA 92037, USA
| | - Christopher A Hinckley
- Howard Hughes Medical Institute and Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Matthew J Sternfeld
- Howard Hughes Medical Institute and Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA. Biological Sciences Graduate Program, UCSD, 9500 Gilman Drive, La Jolla, CA 92037, USA
| | - Shawn P Driscoll
- Howard Hughes Medical Institute and Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Bertha Dominguez
- Peptide Biology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Kuo-Fen Lee
- Peptide Biology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Xin Jin
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Samuel L Pfaff
- Howard Hughes Medical Institute and Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
363
|
Gene targets of mouse miR-709: regulation of distinct pools. Sci Rep 2016; 6:18958. [PMID: 26743462 PMCID: PMC4705522 DOI: 10.1038/srep18958] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 12/01/2015] [Indexed: 12/03/2022] Open
Abstract
MicroRNA (miRNA) are short non-coding RNA molecules that regulate multiple cellular processes, including development, cell differentiation, proliferation and death. Nevertheless, little is known on whether miRNA control the same gene networks in different tissues. miR-709 is an abundant miRNA expressed ubiquitously. Through transcriptome analysis, we have identified targets of miR-709 in hepatocytes. miR-709 represses genes implicated in cytoskeleton organization, extracellular matrix attachment, and fatty acid metabolism. Remarkably, none of the previously identified targets in non-hepatic tissues are silenced by miR-709 in hepatocytes, even though several of these genes are abundantly expressed in liver. In addition, miR-709 is upregulated in hepatocellular carcinoma, suggesting it participates in the genetic reprogramming that takes place during cell division, when cytoskeleton remodeling requires substantial changes in gene expression. In summary, the present study shows that miR-709 does not repress the same pool of genes in separate cell types. These results underscore the need for validating gene targets in every tissue a miRNA is expressed.
Collapse
|
364
|
Marty V, Labialle S, Bortolin-Cavaillé ML, Ferreira De Medeiros G, Moisan MP, Florian C, Cavaillé J. Deletion of the miR-379/miR-410 gene cluster at the imprintedDlk1-Dio3locus enhances anxiety-related behaviour. Hum Mol Genet 2016; 25:728-39. [DOI: 10.1093/hmg/ddv510] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/08/2015] [Indexed: 12/31/2022] Open
|
365
|
Yang ZB, Li TB, Zhang Z, Ren KD, Zheng ZF, Peng J, Luo XJ. The Diagnostic Value of Circulating Brain-specific MicroRNAs for Ischemic Stroke. Intern Med 2016; 55:1279-86. [PMID: 27181533 DOI: 10.2169/internalmedicine.55.5925] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Objective Circulating microRNAs have been recognized as promising biomarkers for various diseases. The aim of the present study was to explore the potential role of circulating miR-107, miR-128b and miR-153 as non-invasive biomarkers in the diagnosis of ischemia stroke. Methods One hundred and fourteen ischemic stroke patients (61±11.3 years old) and 58 healthy volunteers (56±3.9 years old) matched for age and sex were enrolled in this study. Total RNA was isolated from plasma with TRIzol reagent. The circulating microRNAs levels were measured by quantitative real-time polymerase chain reaction. Results The circulating levels of miR-107, miR-128b and miR-153 significantly increased 2.78-, 2.13- and 1.83-fold in ischemia stroke patients in comparison to the healthy volunteers, respectively. Receiver operating characteristic (ROC) curves were analyzed using the SPSS software program and revealed the areas under the curve for circulating miR-107, miR-128b and miR-153 to be 0.97, 0.903 and 0.893 in ischemia stroke patients in comparison to healthy volunteers, respectively. The levels of circulating miR-107, miR-128b and miR-153 therefore positively correlated with the severity of stroke as defined by NIHSS classes. Conclusion Our results suggest that circulating miR-107, miR-128b and miR-153 might be used as potential novel non-invasive biomarkers for the diagnosis of ischemia stroke. However, future prospective trials in large-sized patient cohorts are needed before drawing any definitive conclusions.
Collapse
Affiliation(s)
- Zhong-Bao Yang
- Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, China
| | | | | | | | | | | | | |
Collapse
|
366
|
Edmonds MD, Boyd KL, Moyo T, Mitra R, Duszynski R, Arrate MP, Chen X, Zhao Z, Blackwell TS, Andl T, Eischen CM. MicroRNA-31 initiates lung tumorigenesis and promotes mutant KRAS-driven lung cancer. J Clin Invest 2015; 126:349-64. [PMID: 26657862 DOI: 10.1172/jci82720] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 11/06/2015] [Indexed: 01/12/2023] Open
Abstract
MicroRNA (miR) are important regulators of gene expression, and aberrant miR expression has been linked to oncogenesis; however, little is understood about their contribution to lung tumorigenesis. Here, we determined that miR-31 is overexpressed in human lung adenocarcinoma and this overexpression independently correlates with decreased patient survival. We developed a transgenic mouse model that allows for lung-specific expression of miR-31 to test the oncogenic potential of miR-31 in the lung. Using this model, we observed that miR-31 induction results in lung hyperplasia, followed by adenoma formation and later adenocarcinoma development. Moreover, induced expression of miR-31 in mice cooperated with mutant KRAS to accelerate lung tumorigenesis. We determined that miR-31 regulates lung epithelial cell growth and identified 6 negative regulators of RAS/MAPK signaling as direct targets of miR-31. Our study distinguishes miR-31 as a driver of lung tumorigenesis that promotes mutant KRAS-mediated oncogenesis and reveals that miR-31 directly targets and reduces expression of negative regulators of RAS/MAPK signaling.
Collapse
|
367
|
Liu C, Guan H, Wang Y, Chen M, Xu B, Zhang L, Lu K, Tao T, Zhang X, Huang Y. miR-195 Inhibits EMT by Targeting FGF2 in Prostate Cancer Cells. PLoS One 2015; 10:e0144073. [PMID: 26650737 PMCID: PMC4674136 DOI: 10.1371/journal.pone.0144073] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 10/14/2015] [Indexed: 11/21/2022] Open
Abstract
Prostate cancer (PCa) is one of the leading causes of deaths in America. The major cause of mortality can be attributed to metastasis. Cancer metastasis involves sequential and interrelated events. miRNAs and epithelial-mesenchymal transition (EMT) are implicated in this process. miR-195 is downregulated in many human cancers. However, the roles of miR-195 in PCa metastasis and EMT remain unclear. In this study, data from Memorial Sloan Kettering Cancer Center (MSKCC) prostate cancer database were re-analysed to detect miR-195 expression and its roles in PCa. miR-195 was then overexpressed in castration-resistant PCa cell lines, DU-145 and PC-3. The role of miR-195 in migration and invasion in vitro was also investigated, and common markers in EMT were evaluated through Western blot analysis. A luciferase reporter assay was conducted to confirm the target gene of miR-195; were validated in PCa cells. In MSKCC data re-analyses, miR-195 was poorly expressed in metastatic PCa; miR-195 could be used to diagnose metastatic PCa by measuring the corresponding expression. Area under the receiver operating characteristic curve (AUC-ROC) was 0.705 (P = 0.017). Low miR-195 expression was characterised with a shorter relapse-free survival (RFS) time. miR-195 overexpression suppressed cell migration, invasion and EMT. Fibroblast growth factor 2 (FGF2) was confirmed as a direct target of miR-195. FGF2 knockdown also suppressed migration, invasion and EMT; by contrast, increased FGF2 partially reversed the suppressive effect of miR-195. And data from ONCOMINE prostate cancer database showed that PCa patients with high FGF2 expression showed shorter RFS time (P = 0.046). Overall, this study demonstrated that miR-195 suppressed PCa cell metastasis by downregulating FGF2. miR-195 restoration may be considered as a new therapeutic method to treat metastatic PCa.
Collapse
Affiliation(s)
- Chunhui Liu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
| | - Han Guan
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
| | - Yiduo Wang
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
| | - Ming Chen
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- * E-mail:
| | - Bin Xu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
| | - Lei Zhang
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
| | - Kai Lu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
| | - Tao Tao
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
| | - Xiaowen Zhang
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
| | - Yeqing Huang
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
| |
Collapse
|
368
|
Smith PY, Hernandez-Rapp J, Jolivette F, Lecours C, Bisht K, Goupil C, Dorval V, Parsi S, Morin F, Planel E, Bennett DA, Fernandez-Gomez FJ, Sergeant N, Buée L, Tremblay MÈ, Calon F, Hébert SS. miR-132/212 deficiency impairs tau metabolism and promotes pathological aggregation in vivo. Hum Mol Genet 2015; 24:6721-35. [PMID: 26362250 PMCID: PMC4634376 DOI: 10.1093/hmg/ddv377] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 08/27/2015] [Accepted: 09/08/2015] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) and related tauopathies comprise a large group of neurodegenerative diseases associated with the pathological aggregation of tau protein. While much effort has focused on understanding the function of tau, little is known about the endogenous mechanisms regulating tau metabolism in vivo and how these contribute to disease. Previously, we have shown that the microRNA (miRNA) cluster miR-132/212 is downregulated in tauopathies such as AD. Here, we report that miR-132/212 deficiency in mice leads to increased tau expression, phosphorylation and aggregation. Using reporter assays and cell-based studies, we demonstrate that miR-132 directly targets tau mRNA to regulate its expression. We identified GSK-3β and PP2B as effectors of abnormal tau phosphorylation in vivo. Deletion of miR-132/212 induced tau aggregation in mice expressing endogenous or human mutant tau, an effect associated with autophagy dysfunction. Conversely, treatment of AD mice with miR-132 mimics restored in part memory function and tau metabolism. Finally, miR-132 and miR-212 levels correlated with insoluble tau and cognitive impairment in humans. These findings support a role for miR-132/212 in the regulation of tau pathology in mice and humans and provide new alternatives for therapeutic development.
Collapse
Affiliation(s)
- Pascal Y Smith
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Psychiatrie et Neurosciences
| | - Julia Hernandez-Rapp
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Psychiatrie et Neurosciences
| | - Francis Jolivette
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Psychiatrie et Neurosciences
| | - Cynthia Lecours
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Médecine Moléculaire
| | - Kanchan Bisht
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Médecine Moléculaire
| | - Claudia Goupil
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Psychiatrie et Neurosciences
| | - Veronique Dorval
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Psychiatrie et Neurosciences
| | - Sepideh Parsi
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Psychiatrie et Neurosciences
| | - Françoise Morin
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Psychiatrie et Neurosciences
| | - Emmanuel Planel
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Psychiatrie et Neurosciences
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Francisco-Jose Fernandez-Gomez
- Faculté de Médecine, Université de Lille, UDSL, Lille F-59045, France and UMR-S 1172, Alzheimer and Tauopathies, Inserm, Lille F-59045, France
| | - Nicolas Sergeant
- Faculté de Médecine, Université de Lille, UDSL, Lille F-59045, France and UMR-S 1172, Alzheimer and Tauopathies, Inserm, Lille F-59045, France
| | - Luc Buée
- Faculté de Médecine, Université de Lille, UDSL, Lille F-59045, France and UMR-S 1172, Alzheimer and Tauopathies, Inserm, Lille F-59045, France
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Médecine Moléculaire
| | - Frédéric Calon
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Faculté de Pharmacie, Université Laval, Québec, QC, Canada G1V 0A6
| | - Sébastien S Hébert
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Psychiatrie et Neurosciences,
| |
Collapse
|
369
|
Cosco D, Cilurzo F, Maiuolo J, Federico C, Di Martino MT, Cristiano MC, Tassone P, Fresta M, Paolino D. Delivery of miR-34a by chitosan/PLGA nanoplexes for the anticancer treatment of multiple myeloma. Sci Rep 2015; 5:17579. [PMID: 26620594 PMCID: PMC4665167 DOI: 10.1038/srep17579] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 11/02/2015] [Indexed: 01/09/2023] Open
Abstract
The encapsulation of miR-34a into chitosan/PLGA nanoparticles in order to obtain nanoplexes useful for the modulation of the biopharmaceutical features of the active compound was studied. The nanoplexes were obtained through nanoprecipitation and were characterized by a mean diameter of ~160 nm, a good size distribution and a positive surface charge. The structure of the nanoparticles allowed a high level of entrapment efficiency of the miR-34a and provided protection of the genetic material from the effects of RNase. A high degree of transfection efficiency of the nanoplexes and a significant in vitro antitumor effect against multiple myeloma cells was demonstrated. The therapeutic properties of the nanoplexes were evaluated in vivo against human multiple myeloma xenografts in NOD-SCID mice. The systemic injection of miR-34a mimic-loaded nanoparticles significantly inhibited tumor growth and translated into improved survival of the laboratory mice. RT-PCR analysis carried out on retrieved tumors demonstrated the presence of a high concentration of miR-34a mimics. The integrity of the nanoplexes remained intact and no organ toxicity was observed in treated animals.
Collapse
Affiliation(s)
- Donato Cosco
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Viale S. Venuta, Germaneto, I-88100 Catanzaro, Italy.,IRC FSH- Interregional Research Center for Food Safety &Health, University of Catanzaro "Magna Græcia", Campus Universitario "S. Venuta" - Building of BioSciences, Viale S. Venuta, I-88100 Germaneto; Catanzaro, Italy
| | - Felisa Cilurzo
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Viale S. Venuta, Germaneto, I-88100 Catanzaro, Italy
| | - Jessica Maiuolo
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Viale S. Venuta, Germaneto, I-88100 Catanzaro, Italy
| | - Cinzia Federico
- Department of Experimental and Clinical Medicine, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Viale S. Venuta, Germaneto, I-88100 Catanzaro, Italy
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Viale S. Venuta, Germaneto, I-88100 Catanzaro, Italy
| | - Maria Chiara Cristiano
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Viale S. Venuta, Germaneto, I-88100 Catanzaro, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Viale S. Venuta, Germaneto, I-88100 Catanzaro, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, US
| | - Massimo Fresta
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Viale S. Venuta, Germaneto, I-88100 Catanzaro, Italy.,IRC FSH- Interregional Research Center for Food Safety &Health, University of Catanzaro "Magna Græcia", Campus Universitario "S. Venuta" - Building of BioSciences, Viale S. Venuta, I-88100 Germaneto; Catanzaro, Italy
| | - Donatella Paolino
- Department of Experimental and Clinical Medicine, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Viale S. Venuta, Germaneto, I-88100 Catanzaro, Italy.,IRC FSH- Interregional Research Center for Food Safety &Health, University of Catanzaro "Magna Græcia", Campus Universitario "S. Venuta" - Building of BioSciences, Viale S. Venuta, I-88100 Germaneto; Catanzaro, Italy
| |
Collapse
|
370
|
A Small RNA-Based Immune System Defends Germ Cells against Mobile Genetic Elements. Stem Cells Int 2015; 2016:7595791. [PMID: 26681955 PMCID: PMC4670677 DOI: 10.1155/2016/7595791] [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: 04/14/2015] [Accepted: 06/11/2015] [Indexed: 11/17/2022] Open
Abstract
Transposons are mobile genetic elements that threaten the survival of species by destabilizing the germline genomes. Limiting the spread of these selfish elements is imperative. Germ cells employ specialized small regulatory RNA pathways to restrain transposon activity. PIWI proteins and Piwi-interacting RNAs (piRNAs) silence transposons at the transcriptional and posttranscriptional level with loss-of-function mutant animals universally exhibiting sterility often associated with germ cell defects. This short review aims to illustrate basic strategies of piRNA-guided defense against transposons. Mechanisms of piRNA silencing are most readily studied in Drosophila melanogaster, which serves as a model to delineate molecular concepts and as a reference for mammalian piRNA systems. PiRNA pathways utilize two major strategies to handle the challenges of transposon control: (1) the hard-wired molecular memory of prior transpositions enables recognition of mobile genetic elements and discriminates transposons from host genes; (2) a feed-forward adaptation mechanism shapes piRNA populations to selectively combat the immediate threat of transposon transcripts. In flies, maternally contributed PIWI-piRNA complexes bolster both of these lines of defense and ensure transgenerational immunity. While recent studies have provided a conceptual framework of what could be viewed as an ancient immune system, we are just beginning to appreciate its many molecular innovations.
Collapse
|
371
|
Maalouf SW, Liu WS, Pate JL. MicroRNA in ovarian function. Cell Tissue Res 2015; 363:7-18. [PMID: 26558383 DOI: 10.1007/s00441-015-2307-4] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 09/29/2015] [Indexed: 01/14/2023]
Abstract
The mammalian ovary is a dynamic organ. The coordination of follicle recruitment, selection, and ovulation and the timely development and regression of the corpus luteum are essential for a functional ovary and fertility. Deregulation of any of these processes results in ovarian dysfunction and potential infertility. MicroRNA (miRNA) are short noncoding RNA that regulate developmental processes and time-sensitive functions. The expression of miRNA in the ovary varies with cell type, function, and stage of the estrous cycle. miRNA are involved in the formation of primordial follicles, follicular recruitment and selection, follicular atresia, oocyte-cumulus cell interaction, granulosal cell function, and luteinization. miRNA are differentially expressed in luteal cells at the various stages of the estrous cycle and during maternal recognition of pregnancy, suggesting a role in luteal development, maintenance, and regression. An understanding of the patterns of expression and functions of miRNA in the ovary will lead to novel therapeutics to treat ovarian dysfunction and improve fertility and, potentially, to the development of better contraceptives.
Collapse
Affiliation(s)
- S W Maalouf
- Department of Animal Science, Center for Reproductive Biology and Health, The Pennsylvania State University, 324 Henning Building, University Park, PA 16802, USA
| | - W S Liu
- Department of Animal Science, Center for Reproductive Biology and Health, The Pennsylvania State University, 324 Henning Building, University Park, PA 16802, USA
| | - J L Pate
- Department of Animal Science, Center for Reproductive Biology and Health, The Pennsylvania State University, 324 Henning Building, University Park, PA 16802, USA.
| |
Collapse
|
372
|
Koturbash I, Tolleson WH, Guo L, Yu D, Chen S, Hong H, Mattes W, Ning B. microRNAs as pharmacogenomic biomarkers for drug efficacy and drug safety assessment. Biomark Med 2015; 9:1153-76. [PMID: 26501795 PMCID: PMC5712454 DOI: 10.2217/bmm.15.89] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Much evidence has documented that microRNAs (miRNAs) play an important role in the modulation of interindividual variability in the production of drug metabolizing enzymes and transporters (DMETs) and nuclear receptors (NRs) through multidirectional interactions involving environmental stimuli/stressors, the expression of miRNA molecules and genetic polymorphisms. MiRNA expression has been reported to be affected by drugs and miRNAs themselves may affect drug metabolism and toxicity. In cancer research, miRNA biomarkers have been identified to mediate intrinsic and acquired resistance to cancer therapies. In drug safety assessment, miRNAs have been found associated with cardiotoxicity, hepatotoxicity and nephrotoxicity. This review article summarizes published studies to show that miRNAs can serve as early biomarkers for the evaluation of drug efficacy and drug safety.
Collapse
Affiliation(s)
- Igor Koturbash
- Department of Environmental & Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - William H Tolleson
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR 72079, USA
| | - Lei Guo
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR 72079, USA
| | - Dianke Yu
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR 72079, USA
| | - Si Chen
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR 72079, USA
| | - Huixiao Hong
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR 72079, USA
| | - William Mattes
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR 72079, USA
| | - Baitang Ning
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR 72079, USA
| |
Collapse
|
373
|
Hecht PM, Ballesteros-Yanez I, Grepo N, Knowles JA, Campbell DB. Noncoding RNA in the transcriptional landscape of human neural progenitor cell differentiation. Front Neurosci 2015; 9:392. [PMID: 26557050 PMCID: PMC4615820 DOI: 10.3389/fnins.2015.00392] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 10/06/2015] [Indexed: 01/01/2023] Open
Abstract
Increasing evidence suggests that noncoding RNAs play key roles in cellular processes, particularly in the brain. The present study used RNA sequencing to identify the transcriptional landscape of two human neural progenitor cell lines, SK-N-SH and ReNcell CX, as they differentiate into human cortical projection neurons. Protein coding genes were found to account for 54.8 and 57.0% of expressed genes, respectively, and alignment of RNA sequencing reads revealed that only 25.5-28.1% mapped to exonic regions of the genome. Differential expression analysis in the two cell lines identified altered gene expression in both protein coding and noncoding RNAs as they undergo neural differentiation with 222 differentially expressed genes observed in SK-N-SH cells and 19 differentially expressed genes in ReNcell CX. Interestingly, genes showing differential expression in SK-N-SH cells are enriched in genes implicated in autism spectrum disorder, but not in gene sets related to cancer or Alzheimer's disease. Weighted gene co-expression network analysis (WGCNA) was used to detect modules of co-expressed protein coding and noncoding RNAs in SK-N-SH cells and found four modules to be associated with neural differentiation. These modules contain varying levels of noncoding RNAs ranging from 10.7 to 49.7% with gene ontology suggesting roles in numerous cellular processes important for differentiation. These results indicate that noncoding RNAs are highly expressed in human neural progenitor cells and likely hold key regulatory roles in gene networks underlying neural differentiation and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Patrick M Hecht
- Keck School of Medicine, Zilkha Neurogenetic Institute, University of Southern California Los Angeles, CA, USA
| | - Inmaculada Ballesteros-Yanez
- Department of Inorganic, Organic Chemistry and Biochemistry, Faculty of Medicine, CRIB, University of Castile-La Mancha Ciudad Real, Spain
| | - Nicole Grepo
- Keck School of Medicine, Zilkha Neurogenetic Institute, University of Southern California Los Angeles, CA, USA
| | - James A Knowles
- Keck School of Medicine, Zilkha Neurogenetic Institute, University of Southern California Los Angeles, CA, USA ; Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California Los Angeles, CA, USA
| | - Daniel B Campbell
- Keck School of Medicine, Zilkha Neurogenetic Institute, University of Southern California Los Angeles, CA, USA ; Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California Los Angeles, CA, USA
| |
Collapse
|
374
|
Yang SL, Yang M, Herrlinger S, Liang C, Lai F, Chen JF. MiR-302/367 regulate neural progenitor proliferation, differentiation timing, and survival in neurulation. Dev Biol 2015; 408:140-50. [PMID: 26441343 DOI: 10.1016/j.ydbio.2015.09.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 09/26/2015] [Accepted: 09/26/2015] [Indexed: 11/16/2022]
Abstract
How neural progenitor cell (NPC) behaviors are temporally controlled in early developing embryos remains undefined. The in vivo functions of microRNAs (miRNAs) in early mammalian development remain largely unknown. Mir-302/367 is a miRNA cluster that encodes miR-367 and four miR-302 members (miR302a-d). We show that miR-302b is highly expressed in early neuroepithelium and its expression decline as development progresses. We generated a mir-302/367 knockout mouse model and found that deletion of mir-302/367 results in an early embryonic lethality and open neural tube defect (NTD). NPCs exhibit enhanced proliferation, precocious differentiation, and decreased cell survival in mutant embryos. Furthermore, we identified Fgf15, Cyclin D1, and D2 as direct targets of miR-302 in NPCs in vivo, and their expression is enhanced in mutant NPCs. Ectopic expression of Cyclin D1 and D2 increases NPC proliferation, while FGF19 (human ortholog of Fgf15) overexpression leads to an increase of NPC differentiation. Thus, these findings reveal essential roles of miR-302/367 in orchestrating gene expression and NPC behaviors in neurulation; they also point to miRNAs as critical genetic components associated with neural tube formation.
Collapse
Affiliation(s)
- Si-Lu Yang
- Department of Genetics, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Mei Yang
- Department of Genetics, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Stephanie Herrlinger
- Department of Genetics, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Chen Liang
- Department of Genetics, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Fan Lai
- Biochemistry & Molecular Biology, University of Miami, Miami, FL 33136, USA
| | - Jian-Fu Chen
- Department of Genetics, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
375
|
Parsi S, Smith PY, Goupil C, Dorval V, Hébert SS. Preclinical Evaluation of miR-15/107 Family Members as Multifactorial Drug Targets for Alzheimer's Disease. MOLECULAR THERAPY. NUCLEIC ACIDS 2015; 4:e256. [PMID: 26440600 PMCID: PMC4881761 DOI: 10.1038/mtna.2015.33] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 08/20/2015] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial, fatal neurodegenerative disorder characterized by the abnormal accumulation of Aβ and Tau deposits in the brain. There is no cure for AD, and failure at different clinical trials emphasizes the need for new treatments. In recent years, significant progress has been made toward the development of miRNA-based therapeutics for human disorders. This study was designed to evaluate the efficiency and potential safety of miRNA replacement therapy in AD, using miR-15/107 paralogues as candidate drug targets. We identified miR-16 as a potent inhibitor of amyloid precursor protein (APP) and BACE1 expression, Aβ peptide production, and Tau phosphorylation in cells. Brain delivery of miR-16 mimics in mice resulted in a reduction of AD-related genes APP, BACE1, and Tau in a region-dependent manner. We further identified Nicastrin, a γ-secretase component involved in Aβ generation, as a target of miR-16. Proteomics analysis identified a number of additional putative miR-16 targets in vivo, including α-Synuclein and Transferrin receptor 1. Top-ranking biological networks associated with miR-16 delivery included AD and oxidative stress. Collectively, our data suggest that miR-16 is a good candidate for future drug development by targeting simultaneously endogenous regulators of AD biomarkers (i.e., Aβ and Tau), inflammation, and oxidative stress.
Collapse
Affiliation(s)
- Sepideh Parsi
- Centre de recherche du CHU de Québec, CHUL, Axe Neurosciences, Québec, Québec City, Canada.,Département de psychiatrie et neurosciences, Université Laval, Québec, Québec City, Canada
| | - Pascal Y Smith
- Centre de recherche du CHU de Québec, CHUL, Axe Neurosciences, Québec, Québec City, Canada.,Département de psychiatrie et neurosciences, Université Laval, Québec, Québec City, Canada
| | - Claudia Goupil
- Centre de recherche du CHU de Québec, CHUL, Axe Neurosciences, Québec, Québec City, Canada.,Département de psychiatrie et neurosciences, Université Laval, Québec, Québec City, Canada
| | - Véronique Dorval
- Centre de recherche du CHU de Québec, CHUL, Axe Neurosciences, Québec, Québec City, Canada.,Département de psychiatrie et neurosciences, Université Laval, Québec, Québec City, Canada
| | - Sébastien S Hébert
- Centre de recherche du CHU de Québec, CHUL, Axe Neurosciences, Québec, Québec City, Canada.,Département de psychiatrie et neurosciences, Université Laval, Québec, Québec City, Canada
| |
Collapse
|
376
|
Tang Y, Lin Y, Li C, Hu X, Liu Y, He M, Luo J, Sun G, Wang T, Li W, Guo M. MicroRNA-720 promotes in vitro cell migration by targeting Rab35 expression in cervical cancer cells. Cell Biosci 2015; 5:56. [PMID: 26413265 PMCID: PMC4583841 DOI: 10.1186/s13578-015-0047-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 09/22/2015] [Indexed: 12/18/2022] Open
Abstract
Background MicroRNA-720 (miR-720), a nonclassical miRNA, is involved in the initiation and progression of several tumors. In our previous studies, miR-720 was shown to be significantly upregulated in cervical cancer tissues compared with normal cervical tissues. However, the precise biological functions of miR-720, and its molecular mechanisms of action, are still unknown. Results Microarray expression profiles, luciferase reporter assays, and western blot assays were used to validate Rab35 as a target gene of miR-720 in HEK293T and HeLa cells. The regulation of Rab35 expression by miR-720 was assessed using qRT-PCR and western blot assays, and the effects of exogenous miR-720 and Rab35 on cell migration were evaluated in vitro using Transwell® assay, wound healing assay, and real-time analyses in HeLa cells. The influences of exogenous miR-720 on cell proliferation were evaluated in vitro by the MTT assay in HeLa cells. In addition, expression of E-cadherin and vimentin associated with epithelial-mesenchymal transition were also assessed using western blot analyses after transfection of miR-720 mimics and Rab35 expression vectors. The results showed that the small GTPase, Rab35, is a direct functional target of miR-720 in cervical cancer HeLa cells. By targeting Rab35, overexpression of miR-720 resulted in a decrease in E-cadherin expression and an increase in vimentin expression and finally led to promotion of HeLa cell migration. Furthermore, reintroduction of Rab35 3′-UTR(−) markedly reversed the induction of cell migration in miR-720-expressing HeLa cells. Conclusions The miR-720 promotes cell migration of HeLa cells by downregulating Rab35. The results show that miR-720 is a novel cell migration-associated gene in cervical cancer cells. Electronic supplementary material The online version of this article (doi:10.1186/s13578-015-0047-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yunlan Tang
- College of Life Sciences and State Key Laboratory of Virology, Wuhan University, 430072 Wuhan, People's Republic of China
| | - Yi Lin
- College of Life Sciences and State Key Laboratory of Virology, Wuhan University, 430072 Wuhan, People's Republic of China
| | - Chuang Li
- College of Life Sciences and State Key Laboratory of Virology, Wuhan University, 430072 Wuhan, People's Republic of China
| | - Xunwu Hu
- College of Life Sciences and State Key Laboratory of Virology, Wuhan University, 430072 Wuhan, People's Republic of China
| | - Yi Liu
- College of Life Sciences and State Key Laboratory of Virology, Wuhan University, 430072 Wuhan, People's Republic of China
| | - Mingyang He
- College of Life Sciences and State Key Laboratory of Virology, Wuhan University, 430072 Wuhan, People's Republic of China
| | - Jun Luo
- Department of Pathology, Zhongnan Hospital, Wuhan University, 430071 Wuhan, People's Republic of China
| | - Guihong Sun
- School of Basic Medical Sciences, Wuhan University, 430071 Wuhan, People's Republic of China
| | - Tao Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Wenxin Li
- College of Life Sciences and State Key Laboratory of Virology, Wuhan University, 430072 Wuhan, People's Republic of China
| | - Mingxiong Guo
- College of Life Sciences and State Key Laboratory of Virology, Wuhan University, 430072 Wuhan, People's Republic of China
| |
Collapse
|
377
|
Sundermeier TR, Palczewski K. The impact of microRNA gene regulation on the survival and function of mature cell types in the eye. FASEB J 2015; 30:23-33. [PMID: 26399786 DOI: 10.1096/fj.15-279745] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/14/2015] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) regulate multiple genes, often within the same pathway, fine-tuning expression of key factors and stabilizing gene networks against aberrant fluctuations. The demanding physiologic functions of photoreceptor cells and the retinal pigmented epithelium necessitate precise gene regulation to maintain their homeostasis and function, thus rendering these postmitotic cells vulnerable to premature death in retinal degenerative disorders. Recent studies of the physiologic impact of miRNAs in these cells clearly demonstrate that miRNAs are an essential component of that gene regulation. These important advances provide the foundation for future exploration of miRNA-regulated gene networks in the eye to facilitate the development of miRNA-targeted therapeutics to combat blinding diseases.
Collapse
Affiliation(s)
- Thomas R Sundermeier
- Department of Pharmacology, Case Western Reserve University, School of Medicine, Cleveland, Ohio, USA
| | - Krzysztof Palczewski
- Department of Pharmacology, Case Western Reserve University, School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
378
|
Coarfa C, Fiskus W, Eedunuri VK, Rajapakshe K, Foley C, Chew SA, Shah SS, Geng C, Shou J, Mohamed JS, O'Malley BW, Mitsiades N. Comprehensive proteomic profiling identifies the androgen receptor axis and other signaling pathways as targets of microRNAs suppressed in metastatic prostate cancer. Oncogene 2015; 35:2345-56. [PMID: 26364608 DOI: 10.1038/onc.2015.295] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 06/08/2015] [Accepted: 07/05/2015] [Indexed: 12/19/2022]
Abstract
MicroRNAs are important epigenetic regulators of protein expression by triggering degradation of target mRNAs and/or inhibiting their translation. Dysregulation of microRNA expression has been reported in several cancers, including prostate cancer (PC). We comprehensively characterized the proteomic footprint of a panel of 12 microRNAs that are potently suppressed in metastatic PC (SiM-miRNAs: miR-1, miR-133a, miR-133b, miR-135a, miR-143-3p, miR-145-3p, miR-205, miR-221-3p, miR-221-5p, miR-222-3p, miR-24-1-5p, and miR-31) using reverse-phase proteomic arrays. Re-expression of these SiM-miRNAs in PC cells suppressed cell proliferation and targeted key oncogenic pathways, including cell cycle, apoptosis, Akt/mammalian target of rapamycin signaling, metastasis and the androgen receptor (AR) axis. However, only 12%, at most, of these observed protein expression changes could be explained by predicted direct binding of miRNAs to corresponding mRNAs, suggesting that the majority of these proteomic effects result indirectly. AR and its steroid receptor coactivators (SRCs; SRC-1, -2 and -3) were recurrently affected by these SiM-miRNAs. In agreement, we identified inverse correlations between expression of these SiM-miRNAs and early clinical recurrence, as well as with AR transcriptional activity in human PC tissues. We also identified robust induction of miR-135a by androgen and strong direct binding of AR to the miR-135a locus. As miR-135a potently suppresses AR expression, this results in a negative feedback loop that suppresses AR protein expression in an androgen-dependent manner, while de-repressing AR expression upon androgen deprivation. Our results demonstrate that epigenetic silencing of these SiM-miRNAs can result in increased AR axis activity and cell proliferation, thus contributing to disease progression. We further demonstrate that a negative feedback loop involving miR-135a can restore AR expression under androgen-deprivation conditions, thus contributing to the upregulation of AR protein expression in castration-resistant PC. Finally, our unbiased proteomic profiling demonstrates that the majority of actual protein expression changes induced by SiM-miRNAs cannot be explained based on predicted direct interactions.
Collapse
Affiliation(s)
- C Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - W Fiskus
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - V K Eedunuri
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, USA
| | - K Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - C Foley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - S A Chew
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - S S Shah
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - C Geng
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - J Shou
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - J S Mohamed
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - B W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - N Mitsiades
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
379
|
A miR-130a-YAP positive feedback loop promotes organ size and tumorigenesis. Cell Res 2015; 25:997-1012. [PMID: 26272168 PMCID: PMC4559818 DOI: 10.1038/cr.2015.98] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Revised: 06/24/2015] [Accepted: 06/30/2015] [Indexed: 12/19/2022] Open
Abstract
Organ size determination is one of the most intriguing unsolved mysteries in biology. Aberrant activation of the major effector and transcription co-activator YAP in the Hippo pathway causes drastic organ enlargement in development and underlies tumorigenesis in many human cancers. However, how robust YAP activation is achieved during organ size control remains elusive. Here we report that the YAP signaling is sustained through a novel microRNA-dependent positive feedback loop. miR-130a, which is directly induced by YAP, could effectively repress VGLL4, an inhibitor of YAP activity, thereby amplifying the YAP signals. Inhibition of miR-130a reversed liver size enlargement induced by Hippo pathway inactivation and blocked YAP-induced tumorigenesis. Furthermore, the Drosophila Hippo pathway target bantam functionally mimics miR-130a by repressing the VGLL4 homolog SdBP/Tgi. These findings reveal an evolutionarily conserved positive feedback mechanism underlying robustness of the Hippo pathway in size control and tumorigenesis.
Collapse
|
380
|
Khan IS, Park CY, Mavropoulos A, Shariat N, Pollack JL, Barczak AJ, Erle DJ, McManus MT, Anderson MS, Jeker LT. Identification of MiR-205 As a MicroRNA That Is Highly Expressed in Medullary Thymic Epithelial Cells. PLoS One 2015; 10:e0135440. [PMID: 26270036 PMCID: PMC4535774 DOI: 10.1371/journal.pone.0135440] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 07/22/2015] [Indexed: 11/30/2022] Open
Abstract
Thymic epithelial cells (TECs) support T cell development in the thymus. Cortical thymic epithelial cells (cTECs) facilitate positive selection of developing thymocytes whereas medullary thymic epithelial cells (mTECs) facilitate the deletion of self-reactive thymocytes in order to prevent autoimmunity. The mTEC compartment is highly dynamic with continuous maturation and turnover, but the genetic regulation of these processes remains poorly understood. MicroRNAs (miRNAs) are important regulators of TEC genetic programs since miRNA-deficient TECs are severely defective. However, the individual miRNAs important for TEC maintenance and function and their mechanisms of action remain unknown. Here, we demonstrate that miR-205 is highly and preferentially expressed in mTECs during both thymic ontogeny and in the postnatal thymus. This distinct expression is suggestive of functional importance for TEC biology. Genetic ablation of miR-205 in TECs, however, neither revealed a role for miR-205 in TEC function during homeostatic conditions nor during recovery from thymic stress conditions. Thus, despite its distinct expression, miR-205 on its own is largely dispensable for mTEC biology.
Collapse
Affiliation(s)
- Imran S. Khan
- UCSF Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
| | - Chong Y. Park
- UCSF Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- WM Keck Center for Noncoding RNAs, University of California San Francisco, San Francisco, California, United States of America
| | - Anastasia Mavropoulos
- UCSF Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Nikki Shariat
- UCSF Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- WM Keck Center for Noncoding RNAs, University of California San Francisco, San Francisco, California, United States of America
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California, United States of America
| | - Joshua L. Pollack
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Andrea J. Barczak
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - David J. Erle
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Michael T. McManus
- UCSF Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- WM Keck Center for Noncoding RNAs, University of California San Francisco, San Francisco, California, United States of America
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California, United States of America
| | - Mark S. Anderson
- UCSF Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- * E-mail: (MSA); (LTJ)
| | - Lukas T. Jeker
- UCSF Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
- * E-mail: (MSA); (LTJ)
| |
Collapse
|
381
|
Parchem RJ, Moore N, Fish JL, Parchem JG, Braga TT, Shenoy A, Oldham MC, Rubenstein JLR, Schneider RA, Blelloch R. miR-302 Is Required for Timing of Neural Differentiation, Neural Tube Closure, and Embryonic Viability. Cell Rep 2015. [PMID: 26212322 PMCID: PMC4741278 DOI: 10.1016/j.celrep.2015.06.074] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The evolutionarily conserved miR-302 family of microRNAs is expressed during early mammalian embryonic development. Here, we report that deletion of miR-302a-d in mice results in a fully penetrant late embryonic lethal phenotype. Knockout embryos have an anterior neural tube closure defect associated with a thickened neuroepithelium. The neuroepithelium shows increased progenitor proliferation, decreased cell death, and precocious neuronal differentiation. mRNA profiling at multiple time points during neurulation uncovers a complex pattern of changing targets over time. Overexpression of one of these targets, Fgf15, in the neuroepithelium of the chick embryo induces precocious neuronal differentiation. Compound mutants between mir-302 and the related mir-290 locus have a synthetic lethal phenotype prior to neurulation. Our results show that mir-302 helps regulate neurulation by suppressing neural progenitor expansion and precocious differentiation. Furthermore, these results uncover redundant roles for mir-290 and mir-302 early in development.
Collapse
Affiliation(s)
- Ronald J Parchem
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Urology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nicole Moore
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Urology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jennifer L Fish
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jacqueline G Parchem
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tarcio T Braga
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Urology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Archana Shenoy
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Urology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael C Oldham
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - John L R Rubenstein
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Richard A Schneider
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Robert Blelloch
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Urology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
382
|
Abstract
The discovery of the first microRNA (miRNA) over 20 years ago has ushered in a new era in molecular biology. There are now over 2000 miRNAs that have been discovered in humans and it is believed that they collectively regulate one third of the genes in the genome. miRNAs have been linked to many human diseases and are being pursued as clinical diagnostics and as therapeutic targets. This review presents an overview of the miRNA pathway, including biogenesis routes, biological roles, and clinical approaches.
Collapse
Affiliation(s)
- Scott M Hammond
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
383
|
Jeker LT, Marone R. Targeting microRNAs for immunomodulation. Curr Opin Pharmacol 2015; 23:25-31. [PMID: 26021286 DOI: 10.1016/j.coph.2015.05.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 05/04/2015] [Accepted: 05/06/2015] [Indexed: 12/29/2022]
Abstract
microRNAs (miRNA) are small regulatory RNAs exerting pleiotropic functions in virtually any immune cell-type. Dozens of miRNAs with a known function in the immune system constitute interesting drug targets for immunomodulation. Chemical modifications of nucleic acid-based miRNA mimics and inhibitors largely solved instability issues but delivery to immune cells remains a major challenge. However, recent success targeting the acidic tumor microenvironment is very promising for inflammatory diseases. Moreover, small molecules are being explored as an interesting alternative. Although RNA is often considered 'undruggable' by small molecules recent progress modulating miRNA function through small molecules is encouraging. Computational approaches even allow predictions about specific small molecule/RNA interactions. Finally, recent clinical success demonstrates that drugs targeting RNAs work in humans.
Collapse
Affiliation(s)
- Lukas T Jeker
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.
| | - Romina Marone
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| |
Collapse
|
384
|
The microRNA-200 family regulates pancreatic beta cell survival in type 2 diabetes. Nat Med 2015; 21:619-27. [PMID: 25985365 DOI: 10.1038/nm.3862] [Citation(s) in RCA: 222] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/15/2015] [Indexed: 02/07/2023]
Abstract
Pancreatic beta cell death is a hallmark of type 1 (T1D) and type 2 (T2D) diabetes, but the molecular mechanisms underlying this aspect of diabetic pathology are poorly understood. Here we report that expression of the microRNA (miR)-200 family is strongly induced in islets of diabetic mice and that beta cell-specific overexpression of miR-200 in mice is sufficient to induce beta cell apoptosis and lethal T2D. Conversely, mir-200 ablation in mice reduces beta cell apoptosis and ameliorates T2D. We show that miR-200 negatively regulates a conserved anti-apoptotic and stress-resistance network that includes the essential beta cell chaperone Dnajc3 (also known as p58IPK) and the caspase inhibitor Xiap. We also observed that mir-200 dosage positively controls activation of the tumor suppressor Trp53 and thereby creates a pro-apoptotic gene-expression signature found in islets of diabetic mice. Consequently, miR-200-induced T2D is suppressed by interfering with the signaling of Trp53 and Bax, a proapoptotic member of the B cell lymphoma 2 protein family. Our results reveal a crucial role for the miR-200 family in beta cell survival and the pathophysiology of diabetes.
Collapse
|
385
|
Taki FA, Pan X, Zhang B. Revisiting Chaos Theorem to Understand the Nature of miRNAs in Response to Drugs of Abuse. J Cell Physiol 2015; 230:2857-68. [PMID: 25966899 DOI: 10.1002/jcp.25037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 05/05/2015] [Indexed: 11/08/2022]
Abstract
Just like Matryoshka dolls, biological systems follow a hierarchical order that is based on dynamic bidirectional communication among its components. In addition to the convoluted inter-relationships, the complexity of each component spans several folds. Therefore, it becomes rather challenging to investigate phenotypes resulting from these networks as it requires the integration of reductionistic and holistic approaches. One dynamic system is the transcriptome which comprises a variety of RNA species. Some, like microRNAs, have recently received a lot of attention. miRNAs are very pleiotropic and have been considered as therapeutic and diagnostic candidates in the biomedical fields. In this review, we survey miRNA profiles in response to drugs of abuse (DA) using 118 studies. After providing a summary of miRNAs related to substance use disorders (SUD), general patterns of miRNA signatures are compared among studies for single or multiple drugs of abuse. Then, current challenges and drawbacks in the field are discussed. Finally, we provide support for considering miRNAs as a chaotic system in normal versus disrupted states particularly in SUD and propose an integrative approach for studying and analyzing miRNA data.
Collapse
Affiliation(s)
- Faten A Taki
- Department of Biology, East Carolina University, Greenville, North Carolina
| | - Xiaoping Pan
- Department of Biology, East Carolina University, Greenville, North Carolina
| | - Baohong Zhang
- Department of Biology, East Carolina University, Greenville, North Carolina
| |
Collapse
|
386
|
Memory formation and retention are affected in adult miR-132/212 knockout mice. Behav Brain Res 2015; 287:15-26. [PMID: 25813747 DOI: 10.1016/j.bbr.2015.03.032] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/11/2015] [Accepted: 03/16/2015] [Indexed: 01/16/2023]
Abstract
The miR-132/212 family is thought to play an important role in neural function and plasticity, while its misregulation has been observed in various neurodegenerative disorders. In this study, we analyzed 6-month-old miR-132/212 knockout mice in a battery of cognitive and non-cognitive behavioral tests. No significant changes were observed in reflexes and basic sensorimotor functions as determined by the SHIRPA primary screen. Accordingly, miR-132/212 knockout mice did not differ from wild-type controls in general locomotor activity in an open-field test. Furthermore, no significant changes of anxiety were measured in an elevated plus maze task. However, the mutant mice showed retention phase defects in a novel object recognition test and in the T-water maze. Moreover, the learning and probe phases in the Barnes maze were clearly altered in knockout mice when compared to controls. Finally, changes in BDNF, CREB, and MeCP2 were identified in the miR-132/212-deficient mice, providing a potential mechanism for promoting memory loss. Taken together, these results further strengthen the role of miR-132/212 in memory formation and retention, and shed light on the potential consequences of its deregulation in neurodegenerative diseases.
Collapse
|
387
|
Olive V, Minella AC, He L. Outside the coding genome, mammalian microRNAs confer structural and functional complexity. Sci Signal 2015; 8:re2. [PMID: 25783159 PMCID: PMC4425368 DOI: 10.1126/scisignal.2005813] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) comprise a class of small, regulatory noncoding RNAs (ncRNAs) with pivotal roles in posttranscriptional gene regulation. Since their initial discovery in 1993, numerous miRNAs have been identified in mammalian genomes, many of which play important roles in diverse cellular processes in development and disease. These small ncRNAs regulate the expression of many protein-coding genes posttranscriptionally, thus adding a substantial complexity to the molecular networks underlying physiological development and disease. In part, this complexity arises from the distinct gene structures, the extensive genomic redundancy, and the complex regulation of the expression and biogenesis of miRNAs. These characteristics contribute to the functional robustness and versatility of miRNAs and provide important clues to the functional significance of these small ncRNAs. The unique structure and function of miRNAs will continue to inspire many to explore the vast noncoding genome and to elucidate the molecular basis for the functional complexity of mammalian genomes.
Collapse
Affiliation(s)
- Virginie Olive
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94705, USA
| | - Alex C Minella
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI 53226, USA
| | - Lin He
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94705, USA.
| |
Collapse
|
388
|
Zhang C, Zhao H, Li J, Liu H, Wang F, Wei Y, Su J, Zhang D, Liu T, Zhang Y. The identification of specific methylation patterns across different cancers. PLoS One 2015; 10:e0120361. [PMID: 25774687 PMCID: PMC4361543 DOI: 10.1371/journal.pone.0120361] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 01/20/2015] [Indexed: 12/16/2022] Open
Abstract
Abnormal DNA methylation is known as playing an important role in the tumorgenesis. It is helpful for distinguishing the specificity of diagnosis and therapeutic targets for cancers based on characteristics of DNA methylation patterns across cancers. High throughput DNA methylation analysis provides the possibility to comprehensively filter the epigenetics diversity across various cancers. We integrated whole-genome methylation data detected in 798 samples from seven cancers. The hierarchical clustering revealed the existence of cancer-specific methylation pattern. Then we identified 331 differentially methylated genes across these cancers, most of which (266) were specifically differential methylation in unique cancer. A DNA methylation correlation network (DMCN) was built based on the methylation correlation between these genes. It was shown the hubs in the DMCN were inclined to cancer-specific genes in seven cancers. Further survival analysis using the part of genes in the DMCN revealed high-risk group and low-risk group were distinguished by seven biomarkers (PCDHB15, WBSCR17, IGF1, GYPC, CYGB, ACTG2, and PRRT1) in breast cancer and eight biomarkers (ZBTB32, OR51B4, CCL8, TMEFF2, SALL3, GPSM1, MAGEA8, and SALL1) in colon cancer, respectively. At last, a protein-protein interaction network was introduced to verify the biological function of differentially methylated genes. It was shown that MAP3K14, PTN, ACVR1 and HCK sharing different DNA methylation and gene expression across cancers were relatively high degree distribution in PPI network. The study suggested that not only the identified cancer-specific genes provided reference for individual treatment but also the relationship across cancers could be explained by differential DNA methylation.
Collapse
Affiliation(s)
- Chunlong Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Hongyan Zhao
- Department of Gastroenterology, The fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jie Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Hongbo Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Fang Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yanjun Wei
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Jianzhong Su
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Dongwei Zhang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tiefu Liu
- Department of Gastroenterology, The fourth Affiliated Hospital of Harbin Medical University, Harbin, China
- * E-mail: (YZ); (TL)
| | - Yan Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- * E-mail: (YZ); (TL)
| |
Collapse
|
389
|
Introduction to microRNAs: Biogenesis, Action, Relevance of Tissue microRNAs in Disease Pathogenesis, Diagnosis and Therapy-The Concept of Circulating microRNAs. EXPERIENTIA SUPPLEMENTUM (2012) 2015; 106:3-30. [PMID: 26608197 DOI: 10.1007/978-3-0348-0955-9_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
MicroRNAs as the endogenous mediators of RNA interference have principal roles in gene expression regulation. Since their discovery in the early 1990s, their number has steadily grown to approximately 2500 known microRNAs at present in humans. MicroRNAs encoded by distinct genes regulate the expression of about 30-60 % of human protein coding genes by targeting their messenger RNAs (mRNAs) and induce mostly posttranscriptional inhibition, or in some cases enhancement. MicroRNAs, as fine regulators of the gene expression, have important roles in development, the physiological functioning of the organism, e.g. organogenesis, immune functioning, vascular system, etc. The deregulation of microRNA expression has been observed in many disorders, such as in carcinogenesis. Given their tissue specificity and stability, and specific disease-related alterations, tissue microRNAs can be exploited as excellent biomarkers in the diagnosis. Moreover, microRNAs might also be envisaged as novel therapeutic targets. Beside tissue microRNAs, novel data show that microRNAs are also present in body fluids that could further extend their diagnostic utility as minimally invasive biomarkers of various diseases, but also raises intriguing questions regarding their biological relevance. In this introductory chapter, we summarise the most relevant features of microRNAs including their biogenesis, action, the biological, pathological, diagnostic and potential therapeutical relevance of tissue microRNAs.
Collapse
|