Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Genome Res ; 30(7): 1073-1081, 2020 07.
Article in English | MEDLINE | ID: mdl-32079618

ABSTRACT

Long noncoding RNAs (lncRNAs) have emerged as key coordinators of biological and cellular processes. Characterizing lncRNA expression across cells and tissues is key to understanding their role in determining phenotypes, including human diseases. We present here FC-R2, a comprehensive expression atlas across a broadly defined human transcriptome, inclusive of over 109,000 coding and noncoding genes, as described in the FANTOM CAGE-Associated Transcriptome (FANTOM-CAT) study. This atlas greatly extends the gene annotation used in the original recount2 resource. We demonstrate the utility of the FC-R2 atlas by reproducing key findings from published large studies and by generating new results across normal and diseased human samples. In particular, we (a) identify tissue-specific transcription profiles for distinct classes of coding and noncoding genes, (b) perform differential expression analysis across thirteen cancer types, identifying novel noncoding genes potentially involved in tumor pathogenesis and progression, and (c) confirm the prognostic value for several enhancer lncRNAs expression in cancer. Our resource is instrumental for the systematic molecular characterization of lncRNA by the FANTOM6 Consortium. In conclusion, comprised of over 70,000 samples, the FC-R2 atlas will empower other researchers to investigate functions and biological roles of both known coding genes and novel lncRNAs.


Subject(s)
Transcriptome , Databases, Genetic , Enhancer Elements, Genetic , Gene Expression Profiling , Genome, Human , Humans , Neoplasms/genetics , Organ Specificity , Prognosis , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/metabolism
2.
Genome Res ; 30(7): 1060-1072, 2020 07.
Article in English | MEDLINE | ID: mdl-32718982

ABSTRACT

Long noncoding RNAs (lncRNAs) constitute the majority of transcripts in the mammalian genomes, and yet, their functions remain largely unknown. As part of the FANTOM6 project, we systematically knocked down the expression of 285 lncRNAs in human dermal fibroblasts and quantified cellular growth, morphological changes, and transcriptomic responses using Capped Analysis of Gene Expression (CAGE). Antisense oligonucleotides targeting the same lncRNAs exhibited global concordance, and the molecular phenotype, measured by CAGE, recapitulated the observed cellular phenotypes while providing additional insights on the affected genes and pathways. Here, we disseminate the largest-to-date lncRNA knockdown data set with molecular phenotyping (over 1000 CAGE deep-sequencing libraries) for further exploration and highlight functional roles for ZNF213-AS1 and lnc-KHDC3L-2.


Subject(s)
RNA, Long Noncoding/physiology , Cell Growth Processes/genetics , Cell Movement/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , KCNQ Potassium Channels/metabolism , Molecular Sequence Annotation , Oligonucleotides, Antisense , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/metabolism , RNA, Small Interfering
3.
Nature ; 543(7644): 199-204, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28241135

ABSTRACT

Long non-coding RNAs (lncRNAs) are largely heterogeneous and functionally uncharacterized. Here, using FANTOM5 cap analysis of gene expression (CAGE) data, we integrate multiple transcript collections to generate a comprehensive atlas of 27,919 human lncRNA genes with high-confidence 5' ends and expression profiles across 1,829 samples from the major human primary cell types and tissues. Genomic and epigenomic classification of these lncRNAs reveals that most intergenic lncRNAs originate from enhancers rather than from promoters. Incorporating genetic and expression data, we show that lncRNAs overlapping trait-associated single nucleotide polymorphisms are specifically expressed in cell types relevant to the traits, implicating these lncRNAs in multiple diseases. We further demonstrate that lncRNAs overlapping expression quantitative trait loci (eQTL)-associated single nucleotide polymorphisms of messenger RNAs are co-expressed with the corresponding messenger RNAs, suggesting their potential roles in transcriptional regulation. Combining these findings with conservation data, we identify 19,175 potentially functional lncRNAs in the human genome.


Subject(s)
Databases, Genetic , RNA, Long Noncoding/chemistry , RNA, Long Noncoding/genetics , Transcriptome/genetics , Cells, Cultured , Conserved Sequence/genetics , Datasets as Topic , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation , Genome, Human/genetics , Genome-Wide Association Study , Genomics , Humans , Internet , Molecular Sequence Annotation , Organ Specificity/genetics , Polymorphism, Single Nucleotide , Promoter Regions, Genetic/genetics , Quantitative Trait Loci/genetics , RNA Stability , RNA, Messenger/genetics
4.
Nucleic Acids Res ; 49(D1): D892-D898, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33211864

ABSTRACT

The Functional ANnoTation Of the Mammalian genome (FANTOM) Consortium has continued to provide extensive resources in the pursuit of understanding the transcriptome, and transcriptional regulation, of mammalian genomes for the last 20 years. To share these resources with the research community, the FANTOM web-interfaces and databases are being regularly updated, enhanced and expanded with new data types. In recent years, the FANTOM Consortium's efforts have been mainly focused on creating new non-coding RNA datasets and resources. The existing FANTOM5 human and mouse miRNA atlas was supplemented with rat, dog, and chicken datasets. The sixth (latest) edition of the FANTOM project was launched to assess the function of human long non-coding RNAs (lncRNAs). From its creation until 2020, FANTOM6 has contributed to the research community a large dataset generated from the knock-down of 285 lncRNAs in human dermal fibroblasts; this is followed with extensive expression profiling and cellular phenotyping. Other updates to the FANTOM resource includes the reprocessing of the miRNA and promoter atlases of human, mouse and chicken with the latest reference genome assemblies. To facilitate the use and accessibility of all above resources we further enhanced FANTOM data viewers and web interfaces. The updated FANTOM web resource is publicly available at https://fantom.gsc.riken.jp/.


Subject(s)
Molecular Sequence Annotation , RNA, Long Noncoding/genetics , Transcriptome/genetics , Animals , Binding Sites , Chromatin/metabolism , Drosophila/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Genome , Humans , Metadata , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Promoter Regions, Genetic , RNA, Long Noncoding/metabolism , Transcription Factors/metabolism , User-Computer Interface
5.
BMC Cancer ; 17(1): 409, 2017 Jun 10.
Article in English | MEDLINE | ID: mdl-28601093

ABSTRACT

BACKGROUND: Granulin-epithelin precursor (GEP) is a secretory growth factor, which has been demonstrated to control cancer growth, invasion, drug resistance and immune escape. Our previous studies and others also demonstrated its potential in targeted therapy. Comprehensive characterization of GEP partner on cancer cells are warranted. We have previously shown that GEP interacted with heparan sulfate on the surface of liver cancer cells and the interaction is crucial for GEP-mediated signaling transduction. This study aims to characterize GEP protein partner at the cell membrane with the co-immunoprecipitation and mass spectrometry approach. METHODS: The membrane fraction from liver cancer model Hep3B was used for capturing binding partner with the specific monoclonal antibody against GEP. The precipitated proteins were analyzed by mass spectrometry. After identifying the GEP binding partner, this specific interaction was validated in additional liver cancer cell line HepG2 by co-immunoprecipitation using GRP78 and GEP antibodies, respectively, as the bait. GRP78 transcript levels in hepatocellular carcinoma (HCC) clinical samples (n = 77 pairs) were examined by real-time quantitative RT-PCR. GEP and GRP78 protein expressions were investigated by immunohistochemistry on paraffin sections. RESULTS: We identified the GEP-binding protein as 78-kDa glucose-regulated protein (GRP78, also named heat shock 70-kDa protein 5, HSPA5). This interaction was validated in independent HCC cell lines. Increased GRP78 mRNA levels were demonstrated in liver cancer tissues compared with the paralleled liver tissues (t-test, P = 0.002). GRP78 and GEP transcript levels were significantly correlated (Spearman's correlation, P = 0.001), and the proteins were also detectable in the cytoplasm of liver cancer cells by immunohistochemical staining. CONCLUSIONS: GRP78 and GEP are interacting protein partners in liver cancer cells and may play a role in GEP-mediated cancer progression in HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Heat-Shock Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Liver Neoplasms/metabolism , Signal Transduction , Cell Line, Tumor , Endoplasmic Reticulum Chaperone BiP , Female , Gene Expression Regulation, Neoplastic , Heat-Shock Proteins/genetics , Hep G2 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Male , Middle Aged , Progranulins , Protein Binding
6.
Cancer Cell Int ; 16: 41, 2016.
Article in English | MEDLINE | ID: mdl-27279800

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is an aggressive cancer with high mortality and morbidity worldwide. The limited clinically relevant model has impeded the development of effective HCC treatment strategy. Patient-derived xenograft (PDX) models retain most of the characteristics of original tumors and were shown to be highly predictive for clinical outcomes. Notably, primary cell line models allow in-depth molecular characterization and high-throughput analysis. Combined usage of the two models would provide an excellent tool for systematic study of therapeutic strategies. Here, we comprehensively characterized the novel PDX and the paralleled primary HCC cell line model. METHODS: Tumor tissues were collected from HCC surgical specimens. HCC cells were sorted for in vivo PDX and in vitro cell line establishment by the expression of hepatic cancer stem cell marker to enhance cell viability and the rate of success on subsequent culture. The PDX and its matching primary cell line were authenticated and characterized in vitro and in vivo. RESULTS: Among the successful cases for generating PDXs and primary cells, HCC40 is capable for both PDX and primary cell line establishment, which were then further characterized. The novel HCC40-PDX and HCC40-CL exhibited consistent phenotypic characteristics as the original tumor in terms of HBV protein and AFP expressions. In common with HCC40-PDX, HCC40-CL was tumorigenic in immunocompromised mice. The migration ability in vitro and metastatic properties in vivo echoed the clinical feature of venous infiltration. Genetic profiling by short tandem repeat analysis and p53 mutation pattern consolidated that both the HCC40-PDX and HCC40-CL models were derived from the HCC40 clinical specimen. CONCLUSIONS: The paralleled establishment of PDX and primary cell line would serve as useful models in comprehensive studies for HCC pathogenesis and therapeutics development for personalized treatment.

7.
BMC Cancer ; 15: 264, 2015 Apr 11.
Article in English | MEDLINE | ID: mdl-25885205

ABSTRACT

BACKGROUND: Granulin-epithelin precursor (GEP), a secretory growth factor, demonstrated overexpression in various human cancers, however, mechanism remain elusive. Primary liver cancer, hepatocellular carcinoma (HCC), ranks the second in cancer-related death globally. GEP controlled growth, invasion, metastasis and chemo-resistance in liver cancer. Noted that GEP gene locates at 17q21 and the region has been frequently reported to be amplified in subset of HCC. The study aims to investigate if copy number gain would associate with GEP overexpression. METHODS: Quantitative Microsatellite Analysis (QuMA) was used to quantify the GEP DNA copy number, and fluorescent in situ hybridization (FISH) was performed to consolidate the amplification status. GEP gene copy number, mRNA expression level and clinico-pathological features were analyzed. RESULTS: GEP DNA copy number determined by QuMA corroborated well with the FISH data, and the gene copy number correlated with the expression levels (n = 60, r = 0.331, P = 0.010). Gain of GEP copy number was observed in 20% (12/60) HCC and associated with hepatitis B virus infection status (P = 0.015). In HCC with increased GEP copy number, tight association between GEP DNA and mRNA levels were observed (n = 12, r = 0.664, P = 0.019). CONCLUSIONS: Gain of the GEP gene copy number was observed in 20% HCC and the frequency comparable to literatures reported on the chromosome region 17q. Increased gene copy number contributed to GEP overexpression in subset of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Dosage/genetics , Intercellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/genetics , Aged , Carcinoma, Hepatocellular/pathology , Chromosomes, Human, Pair 17/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization, Fluorescence , Intercellular Signaling Peptides and Proteins/biosynthesis , Liver Neoplasms/pathology , Male , Microsatellite Repeats/genetics , Middle Aged , Neoplasm Staging , Progranulins
8.
Carcinogenesis ; 35(11): 2485-94, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25115442

ABSTRACT

Granulin-epithelin precursor (GEP) is a pluripotent secretory growth factor which promotes cancer progression in a number of human cancers. However, how cancer cells interact with GEP remains unknown. In this study, we aimed to identify the cell surface-binding partner of GEP on liver cancer cells. Human recombinant GEP (rGEP) was expressed and purified to homogeneity. The rGEP was shown to trigger phosphorylation of AKT and ERK1/2 in liver cancer cells. We demonstrated cell surface attachment of rGEP, which was blocked by prebinding of platelet-derived growth factor-AA, platelet-derived growth factor-BB and fibroblast growth factor-2. Therefore, heparan sulfate (HS) had been reasoned as the binding partner of rGEP. Heparinase digestion validated the role of HS on supporting the attachment. The heparin-binding domain of GEP was mapped to RRH(555-557) in the C-terminal region. Suppression of the HS polymerase exostosin-1 reduced the rGEP binding and rGEP-mediated signaling transduction. Suppression of a specific HS proteoglycan, glypican-3, also showed a partial reduction of rGEP binding and an inhibition on rGEP-mediated activation of AKT. Furthermore, glypican-3 was shown to correlate with the expressions of GEP in clinical samples (Spearman's ρ = 0.363, P = 0.001). This study identified HS, partly through glypican-3, as a novel binding partner of GEP on the surface of liver cancer cells.


Subject(s)
Carcinoma, Hepatocellular/genetics , Glypicans/metabolism , Heparitin Sulfate/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/genetics , Carcinoma, Hepatocellular/pathology , Fibroblast Growth Factor 2/biosynthesis , Fibroblast Growth Factor 2/genetics , Gene Expression Regulation, Neoplastic , Glypicans/antagonists & inhibitors , Hep G2 Cells , Heparitin Sulfate/genetics , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Liver Neoplasms/metabolism , MAP Kinase Signaling System/genetics , Oncogene Protein v-akt/genetics , Progranulins , Protein Binding
9.
Cancer Cell Int ; 14(1): 103, 2014.
Article in English | MEDLINE | ID: mdl-25349534

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is a highly aggressive and heterogeneous disease. HCC cell lines established from different patients would be useful in elucidating the molecular pathogenesis. However, success of HCC primary culture establishment remains at low rate. We aim to establish and characterize HCC primary culture and the derived cell line. METHODS: Fresh tumor tissues were collected from 30 HCC patients. Culture conditions were optimized for the attachment and growth of the isolated hepatocytes. Granulin-epithelin precursor (GEP), a growth factor reported to associate with cancer stem cell properties, was examined by flow cytometry to elucidate its role on primary culture establishment. The primary cell line was characterized in detail. RESULTS: Cells isolated from 16 out of 30 HCC cases (53%) had viability more than 70% and were subject to subsequent in vitro culture. 7 out of 16 cases (44%) could give rise to cells that were able to attach and grow in culture. GEP expression levels significantly correlated with the viability of isolated hepatocytes and success rate of subsequent primary culture establishment. Cells from HCC patient 21 grew and expanded rapidly in vitro and was selected to be further characterized. The line, designated HCC21, derived from a Hong Kong Chinese female patient with HCC at Stage II. The cells exhibited typical epithelial morphology and expressed albumin, AFP and HBV antigens. The cell line was authenticated by short tandem repeat analysis. Comparative genome hybridization analysis revealed chromosomal loss at 1p35-p36, 1q44, 2q11.2-q24.3, 2q37, 4q12-q13.3, 4q21.21-q35.2, 8p12-p23, 15q11.2-q14, 15q24-q26, 16p12.1-p13.3, 16q, 17p, 22q and gain at 1q21-q43 in both HCC21 cells and the original clinical tumor specimen. Sequence analysis revealed p53 gene mutation. Subcutaneous injection of HCC21 cells into immunodeficient mice showed that the cells were able to form tumors at the primary injection sites and metastatic tumors in the peritoneal cavity. CONCLUSIONS: The newly established cell line could serve as useful in vitro and in vivo models for studying primary HCC that possess metastasis ability.

10.
PLoS One ; 19(5): e0295971, 2024.
Article in English | MEDLINE | ID: mdl-38709794

ABSTRACT

The human genome is pervasively transcribed and produces a wide variety of long non-coding RNAs (lncRNAs), constituting the majority of transcripts across human cell types. Some specific nuclear lncRNAs have been shown to be important regulatory components acting locally. As RNA-chromatin interaction and Hi-C chromatin conformation data showed that chromatin interactions of nuclear lncRNAs are determined by the local chromatin 3D conformation, we used Hi-C data to identify potential target genes of lncRNAs. RNA-protein interaction data suggested that nuclear lncRNAs act as scaffolds to recruit regulatory proteins to target promoters and enhancers. Nuclear lncRNAs may therefore play a role in directing regulatory factors to locations spatially close to the lncRNA gene. We provide the analysis results through an interactive visualization web portal at https://fantom.gsc.riken.jp/zenbu/reports/#F6_3D_lncRNA.


Subject(s)
Chromatin , RNA, Long Noncoding , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Chromatin/metabolism , Chromatin/genetics , Humans , Molecular Sequence Annotation , Cell Nucleus/metabolism , Cell Nucleus/genetics , Genome, Human , Promoter Regions, Genetic
11.
Cell Rep ; 41(13): 111893, 2022 12 27.
Article in English | MEDLINE | ID: mdl-36577377

ABSTRACT

Within the scope of the FANTOM6 consortium, we perform a large-scale knockdown of 200 long non-coding RNAs (lncRNAs) in human induced pluripotent stem cells (iPSCs) and systematically characterize their roles in self-renewal and pluripotency. We find 36 lncRNAs (18%) exhibiting cell growth inhibition. From the knockdown of 123 lncRNAs with transcriptome profiling, 36 lncRNAs (29.3%) show molecular phenotypes. Integrating the molecular phenotypes with chromatin-interaction assays further reveals cis- and trans-interacting partners as potential primary targets. Additionally, cell-type enrichment analysis identifies lncRNAs associated with pluripotency, while the knockdown of LINC02595, CATG00000090305.1, and RP11-148B6.2 modulates colony formation of iPSCs. We compare our results with previously published fibroblasts phenotyping data and find that 2.9% of the lncRNAs exhibit a consistent cell growth phenotype, whereas we observe 58.3% agreement in molecular phenotypes. This highlights that molecular phenotyping is more comprehensive in revealing affected pathways.


Subject(s)
Induced Pluripotent Stem Cells , RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Induced Pluripotent Stem Cells/metabolism , Oligonucleotides, Antisense , Gene Expression Profiling/methods , Embryonic Stem Cells/metabolism
12.
Nat Commun ; 13(1): 156, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013174

ABSTRACT

Immune evasion is indispensable for cancer initiation and progression, although its underlying mechanisms in pancreatic ductal adenocarcinoma (PDAC) are not fully known. Here, we characterize the function of tumor-derived PGRN in promoting immune evasion in primary PDAC. Tumor- but not macrophage-derived PGRN is associated with poor overall survival in PDAC. Multiplex immunohistochemistry shows low MHC class I (MHCI) expression and lack of CD8+ T cell infiltration in PGRN-high tumors. Inhibition of PGRN abrogates autophagy-dependent MHCI degradation and restores MHCI expression on PDAC cells. Antibody-based blockade of PGRN in a PDAC mouse model remarkably decelerates tumor initiation and progression. Notably, tumors expressing LCMV-gp33 as a model antigen are sensitized to gp33-TCR transgenic T cell-mediated cytotoxicity upon PGRN blockade. Overall, our study shows a crucial function of tumor-derived PGRN in regulating immunogenicity of primary PDAC.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Pancreatic Ductal/genetics , Histocompatibility Antigens Class I/genetics , Pancreatic Neoplasms/genetics , Progranulins/genetics , Tumor Escape/genetics , Adenocarcinoma/immunology , Adenocarcinoma/mortality , Adenocarcinoma/therapy , Animals , Antibodies, Neutralizing/pharmacology , Antigens, Viral/genetics , Antigens, Viral/immunology , Autophagy/drug effects , Autophagy/genetics , Autophagy/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/therapy , Cell Line, Tumor , Cell Movement/drug effects , Cohort Studies , Cytotoxicity, Immunologic , Gene Expression , Glycoproteins/genetics , Glycoproteins/immunology , Histocompatibility Antigens Class I/immunology , Humans , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/immunology , Mice , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/therapy , Peptide Fragments/genetics , Peptide Fragments/immunology , Progranulins/antagonists & inhibitors , Progranulins/immunology , Proteolysis , Survival Analysis , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Xenograft Model Antitumor Assays
13.
J Virol ; 84(17): 8700-11, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20554771

ABSTRACT

Type 2 (or North American-like) porcine reproductive and respiratory syndrome virus (PRRSV) was first recorded in 1987 in the United States and now occurs in most commercial swine industries throughout the world. In this study, we investigated the epidemiological and evolutionary behaviors of type 2 PRRSV. Based on phylogenetic analyses of 8,624 ORF5 sequences, we described a comprehensive picture of the diversity of type 2 PRRSVs and systematically classified all available sequences into lineages and sublineages, including a number of previously undescribed lineages. With the rapid growth of sequence deposition into the databases, it would be technically difficult for veterinary researchers to genotype their sequences by reanalyzing all sequences in the databases. To this end, a set of reference sequences was established based on our classification system, which represents the principal diversity of all available sequences and can readily be used for further genotyping studies. In addition, we further investigated the demographic histories of these lineages and sublineages by using Bayesian coalescence analyses, providing evolutionary insights into several important epidemiological events of type 2 PRRSV. Moreover, by using a phylogeographic approach, we were able to estimate the transmission frequencies between the pig-producing states in the United States and identified several states as the major sources of viral spread, i.e., "transmission centers." In summary, this study represents the most extensive phylogenetic analyses of type 2 PRRSV to date, providing a basis for future genotyping studies and dissecting the epidemiology of type 2 PRRSV from phylogenetic perspectives.


Subject(s)
Evolution, Molecular , Phylogeny , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/classification , Animals , Genetic Variation , Geography , Molecular Sequence Data , Porcine Reproductive and Respiratory Syndrome/transmission , Porcine respiratory and reproductive syndrome virus/genetics , Porcine respiratory and reproductive syndrome virus/isolation & purification , Swine
14.
Essays Biochem ; 65(4): 761-773, 2021 10 27.
Article in English | MEDLINE | ID: mdl-33835127

ABSTRACT

Recent efforts on the characterization of long non-coding RNAs (lncRNAs) revealed their functional roles in modulating diverse cellular processes. These include pluripotency maintenance, lineage commitment, carcinogenesis, and pathogenesis of various diseases. By interacting with DNA, RNA and protein, lncRNAs mediate multifaceted mechanisms to regulate transcription, RNA processing, RNA interference and translation. Of more than 173000 discovered lncRNAs, the majority remain functionally unknown. The cell type-specific expression and localization of the lncRNA also suggest potential distinct functions of lncRNAs across different cell types. This highlights the niche of identifying functional lncRNAs in different biological processes and diseases through high-throughput (HTP) screening. This review summarizes the current work performed and perspectives on HTP screening of functional lncRNAs where different technologies, platforms, cellular responses and the downstream analyses are discussed. We hope to provide a better picture in applying different technologies to facilitate functional annotation of lncRNA efficiently.


Subject(s)
RNA, Long Noncoding , High-Throughput Screening Assays , RNA, Long Noncoding/genetics
15.
PLoS Pathog ; 4(8): e1000130, 2008 Aug 22.
Article in English | MEDLINE | ID: mdl-18725937

ABSTRACT

H5N1 highly pathogenic avian influenza (HPAI) viruses have seriously affected the Asian poultry industry since their recurrence in 2003. The viruses pose a threat of emergence of a global pandemic influenza through point mutation or reassortment leading to a strain that can effectively transmit among humans. In this study, we present phylogenetic evidences for the interlineage reassortment among H5N1 HPAI viruses isolated from humans, cats, and birds in Indonesia, and identify the potential genetic parents of the reassorted genome segments. Parsimony analyses of viral phylogeography suggest that the reassortant viruses may have originated from greater Jakarta and surroundings, and subsequently spread to other regions in the West Java province. In addition, Bayesian methods were used to elucidate the genetic diversity dynamics of the reassortant strain and one of its genetic parents, which revealed a more rapid initial growth of genetic diversity in the reassortant viruses relative to their genetic parent. These results demonstrate that interlineage exchange of genetic information may play a pivotal role in determining viral genetic diversity in a focal population. Moreover, our study also revealed significantly stronger diversifying selection on the M1 and PB2 genes in the lineages preceding and subsequent to the emergence of the reassortant viruses, respectively. We discuss how the corresponding mutations might drive the adaptation and onward transmission of the newly formed reassortant viruses.


Subject(s)
Evolution, Molecular , Influenza A Virus, H5N1 Subtype/genetics , Influenza in Birds/genetics , Influenza in Birds/transmission , Poultry Diseases/genetics , Poultry Diseases/transmission , Animals , Genes, Viral , Humans , Indonesia/epidemiology , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza in Birds/epidemiology , Poultry , Poultry Diseases/epidemiology
16.
J Virol ; 82(4): 1819-26, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18057240

ABSTRACT

Bats have been identified as the natural reservoir of severe acute respiratory syndrome (SARS)-like and SARS coronaviruses (SLCoV and SCoV). However, previous studies suggested that none of the currently sampled bat SLCoVs is the descendant of the direct ancestor of SCoV, based on their relatively distant phylogenetic relationship. In this study, evidence of the recombinant origin of the genome of a bat SLCoV is demonstrated. We identified a potential recombination breakpoint immediately after the consensus intergenic sequence between open reading frame 1 and the S coding region, suggesting the replication intermediates may participate in the recombination event, as previously speculated for other CoVs. Phylogenetic analysis of its parental regions suggests the presence of an uncharacterized SLCoV lineage that is phylogenetically closer to SCoVs than any of the currently sampled bat SLCoVs. Using various Bayesian molecular-clock models, interspecies transfer of this SLCoV lineage from bats to the amplifying host (e.g., civets) was estimated to have happened a median of 4.08 years before the SARS outbreak. Based on this relatively short window period, we speculate that this uncharacterized SLCoV lineage may contain the direct ancestor of SCoV. This study sheds light on the possible host bat species of the direct ancestor of SCoV, providing valuable information on the scope and focus of surveillance for the origin of SCoV.


Subject(s)
Chiroptera/virology , Genome, Viral/genetics , Models, Genetic , Severe acute respiratory syndrome-related coronavirus/classification , Severe acute respiratory syndrome-related coronavirus/genetics , Animals , Base Sequence , Bayes Theorem , Molecular Sequence Data , Phylogeny , Recombination, Genetic , Severe acute respiratory syndrome-related coronavirus/isolation & purification
17.
Cancer Lett ; 457: 98-109, 2019 08 10.
Article in English | MEDLINE | ID: mdl-31100412

ABSTRACT

ATP-binding cassette (ABC) transporters mediate multidrug resistance and cancer stem cell properties in various model systems. Yet, their biological significance in cancers, especially in hepatocellular carcinoma (HCC), remains unclear. In this study, we investigated the function of ABCF1 in HCC and explored its potential as a therapeutic target. ABCF1 was highly expressed in fetal mouse livers but not in normal adult livers. ABCF1 expression was upregulated in HCCs. These results demonstrate that ABCF1 functions as a hepatic oncofetal protein. We further demonstrated elevated ABCF1 expression in HCC cells upon acquiring chemoresistance. Suppression of ABCF1 by siRNA sensitized both parental cells and chemoresistant cells to chemotherapeutic agents. Reversely, ABCF1 overexpression promoted chemoresistance and drug efflux. In addition, overexpression of ABCF1 enhanced migration, spheroid and colony formation and epithelial-mesenchymal transition (EMT) induction. RNA sequencing analysis revealed EMT inducers HIF1α/IL8 and Sox4 as potential mediators for the oncogenic effect of ABCF1 in HCC progression. Together, this study illustrates that ABCF1 is a novel potential therapeutic target for HCC treatment.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Carcinoma, Hepatocellular/metabolism , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Liver Neoplasms/metabolism , Neoplastic Stem Cells/drug effects , ATP-Binding Cassette Transporters/genetics , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Movement , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice, Inbred ICR , Mice, Nude , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism , Signal Transduction , Xenograft Model Antitumor Assays
18.
Methods Mol Biol ; 1806: 131-144, 2018.
Article in English | MEDLINE | ID: mdl-29956274

ABSTRACT

The use of monoclonal antibody (mAb) has become a unique means of targeted therapy for human cancers. mAb-based therapies have shown survival benefits by applying alone or in combination with chemotherapeutics. Being a humanized biomolecule with exquisite target specificity, mAb demonstrated effects in a relatively lower dose range with limited off-target harm to the patients. Nowadays, novel targets involved in tumorigenic mechanisms and biomarkers expressed exclusively on cancer cell surface are being constantly discovered. The potential effects of their specific mAb could be investigated in the preclinical cancer model. In this chapter, we outlined our experimental procedures in determining the feasibility of novel mAb in the preclinical cancer model, with an example of progranulin (PGRN/GEP) mAb against hepatocellular carcinoma (HCC) tumor in mouse model. This chapter included the establishment of subcutaneous and orthotopic HCC tumor in mouse model, the injection of the mouse monoclonal antibody in combination with cytotoxic chemotherapeutics, the assessment of tumor development, and the analyses of the molecular changes of the tumor cells.


Subject(s)
Antibodies, Monoclonal/metabolism , Progranulins/metabolism , Animals , Apoptosis , Cell Line, Tumor , Disease Models, Animal , Flow Cytometry , Male , Mice, Inbred BALB C , Mice, Nude , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spheroids, Cellular/pathology , Tumor Stem Cell Assay
19.
Virus Res ; 123(1): 50-6, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16965830

ABSTRACT

Tissue culture adaptation of infectious bursal disease virus (IBDV) results in alternation of three residues on its major capsid protein VP2 and these residues may engage in receptor binding. Although the key of successful infection of tissue culture adapted IBDV in tissue cultures was defined as the virus entering steps, mechanism of the adaptation is poorly understood. In this study, recombinant VP2s of an attenuated strain (D78) and a very virulent strain (HK46) of IBDV tagged with rabbit immunoglobulin G heavy chain were expressed in mammalian cells, generating RAVP2 and RVVP2, respectively, in high purity. Using flow cytometry, both RAVP2 and RVVP2 were demonstrated to bind with Vero cells while these bindings were blocked by D78 viral particles, implying both very virulent IBDVs (vvIBDVs) and attenuated IBDVs bind to Vero cells through the same receptor(s). Since vvIBDVs cannot be propagated directly in tissue cultures, the specific binding between RVVP2 and Vero cells suggests the barrier for tissue culture adaptation may be beyond the virus attachment process.


Subject(s)
Birnaviridae Infections/virology , Capsid Proteins/metabolism , Infectious bursal disease virus/chemistry , Infectious bursal disease virus/physiology , Receptors, Virus/metabolism , Adaptation, Physiological , Animals , Chlorocebus aethiops , Vero Cells , Virus Replication
20.
FEBS Lett ; 580(24): 5612-20, 2006 Oct 16.
Article in English | MEDLINE | ID: mdl-16989815

ABSTRACT

Neutralizing effects of antibodies targeting the C-terminal stalk (S2) subunit of the spike protein of severe acute respiratory syndrome coronavirus have previously been reported, although its mechanism remained elusive. In this study, high titered mouse antisera against the N-terminal globular (S1) and S2 subunits of the S protein were generated and total immunoglobulin G (IgG) was purified from these antisera. The efficiency of these purified IgGs in virus neutralization and blocking of receptor binding were compared quantitatively using virus neutralization assay and a previously developed cell-based receptor binding assay, respectively. We demonstrated that anti-S1 IgG neutralizes the virus and binds to the membrane associated S protein more efficiently than anti-S2 IgG does. Moreover, both anti-S1 and anti-S2 IgGs were able to abolish the binding between S protein and its cellular receptor(s), although anti-S1 IgG showed a significantly higher blocking efficiency. The unexpected blocking ability of anti-S2 IgG towards the receptor binding implied a possible role of the S2 subunit in virus docking process and argues against the current hypothesis of viral entry. On the other hand, the functional roles of the previously reported neutralizing epitopes within S2 subunit were investigated using an antigen specific antibody depletion assay. Depletion of antibodies against these regions significantly diminished, though not completely abolished, the neutralizing effects of anti-S2 IgG. It suggests the absence of a major neutralizing domain on S2 protein. The possible ways of anti-S2 IgGs to abolish the receptor binding and the factors restricting anti-S2 IgGs to neutralize the virus are discussed.


Subject(s)
Antibodies, Viral/immunology , Antibodies, Viral/pharmacology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Severe acute respiratory syndrome-related coronavirus/drug effects , Severe acute respiratory syndrome-related coronavirus/immunology , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , Epitopes/immunology , Mice , Protein Binding , Protein Subunits/immunology , Protein Subunits/metabolism , Spike Glycoprotein, Coronavirus
SELECTION OF CITATIONS
SEARCH DETAIL