Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
PLoS One ; 18(10): e0293263, 2023.
Article in English | MEDLINE | ID: mdl-37874836

ABSTRACT

The COVID-19 pandemic has created an urgent need for effective therapeutic and diagnostic strategies to manage the disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, the emergence of numerous variants of concern (VOCs) has made it challenging to develop targeted therapies that are broadly specific in neutralizing the virus. In this study, we aimed to develop neutralizing nanobodies (Nbs) using computational techniques that can effectively neutralize the receptor-binding domain (RBD) of SARS-CoV-2 VOCs. We evaluated the performance of different protein-protein docking programs and identified HDOCK as the most suitable program for Nb/RBD docking with high accuracy. Using this approach, we designed 14 novel Nbs with high binding affinity to the VOC RBDs. The Nbs were engineered with mutated amino acids that interacted with key amino acids of the RBDs, resulting in higher binding affinity than human angiotensin-converting enzyme 2 (ACE2) and other viral RBDs or haemagglutinins (HAs). The successful development of these Nbs demonstrates the potential of molecular modeling as a low-cost and time-efficient method for engineering effective Nbs against SARS-CoV-2. The engineered Nbs have the potential to be employed in RBD-neutralizing assays, facilitating the identification of novel treatment, prevention, and diagnostic strategies against SARS-CoV-2.


Subject(s)
COVID-19 , Single-Domain Antibodies , Humans , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Antibodies, Neutralizing/metabolism , Single-Domain Antibodies/genetics , Single-Domain Antibodies/metabolism , Antibodies, Viral/metabolism , Pandemics , Protein Binding , Amino Acids/metabolism , Spike Glycoprotein, Coronavirus/chemistry
2.
Nature ; 619(7970): 585-594, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37468583

ABSTRACT

Understanding kidney disease relies on defining the complexity of cell types and states, their associated molecular profiles and interactions within tissue neighbourhoods1. Here we applied multiple single-cell and single-nucleus assays (>400,000 nuclei or cells) and spatial imaging technologies to a broad spectrum of healthy reference kidneys (45 donors) and diseased kidneys (48 patients). This has provided a high-resolution cellular atlas of 51 main cell types, which include rare and previously undescribed cell populations. The multi-omic approach provides detailed transcriptomic profiles, regulatory factors and spatial localizations spanning the entire kidney. We also define 28 cellular states across nephron segments and interstitium that were altered in kidney injury, encompassing cycling, adaptive (successful or maladaptive repair), transitioning and degenerative states. Molecular signatures permitted the localization of these states within injury neighbourhoods using spatial transcriptomics, while large-scale 3D imaging analysis (around 1.2 million neighbourhoods) provided corresponding linkages to active immune responses. These analyses defined biological pathways that are relevant to injury time-course and niches, including signatures underlying epithelial repair that predicted maladaptive states associated with a decline in kidney function. This integrated multimodal spatial cell atlas of healthy and diseased human kidneys represents a comprehensive benchmark of cellular states, neighbourhoods, outcome-associated signatures and publicly available interactive visualizations.


Subject(s)
Gene Expression Profiling , Kidney Diseases , Kidney , Single-Cell Analysis , Transcriptome , Humans , Cell Nucleus/genetics , Kidney/cytology , Kidney/injuries , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Transcriptome/genetics , Case-Control Studies , Imaging, Three-Dimensional
3.
Sci Rep ; 13(1): 7678, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37169829

ABSTRACT

Cell-cycle control is accomplished by cyclin-dependent kinases (CDKs), motivating extensive research into CDK targeting small-molecule drugs as cancer therapeutics. Here we use combinatorial CRISPR/Cas9 perturbations to uncover an extensive network of functional interdependencies among CDKs and related factors, identifying 43 synthetic-lethal and 12 synergistic interactions. We dissect CDK perturbations using single-cell RNAseq, for which we develop a novel computational framework to precisely quantify cell-cycle effects and diverse cell states orchestrated by specific CDKs. While pairwise disruption of CDK4/6 is synthetic-lethal, only CDK6 is required for normal cell-cycle progression and transcriptional activation. Multiple CDKs (CDK1/7/9/12) are synthetic-lethal in combination with PRMT5, independent of cell-cycle control. In-depth analysis of mRNA expression and splicing patterns provides multiple lines of evidence that the CDK-PRMT5 dependency is due to aberrant transcriptional regulation resulting in premature termination. These inter-dependencies translate to drug-drug synergies, with therapeutic implications in cancer and other diseases.


Subject(s)
Neoplasms , Humans , Cell Cycle Checkpoints , Cell Cycle/genetics , Neoplasms/drug therapy , Protein-Arginine N-Methyltransferases/pharmacology
4.
Mol Biotechnol ; 65(4): 598-611, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36103078

ABSTRACT

The ongoing COVID-19 pandemic has resulted from widespread infection by the SARS-CoV-2 virus. As new variants of concern continue to emerge, understanding the correlation between the level of neutralizing antibodies (NAb) and clinical protection from SAR-CoV-2 infection could be critical in planning the next steps in COVID-19 vaccine programs. This study explored the potential usefulness of E. coli as an alternative expression system that can be used to produce a SARS-CoV-2 receptor-binding domain (RBD) for the development of an affordable and flexible NAb detection assay. We expressed the RBD of Beta, Delta, and Omicron variants in the E. coli BL21(DE3) strain and purified them from whole bacterial cells using His-tag-mediated affinity chromatography and urea-assisted refolding. Next, we conducted a head-to-head comparison of the binding activity of our E. coli-produced RBD (E-RBD) with commercial HEK293-produced RBD (H-RBD). The results of a direct binding assay revealed E-RBD and H-RBD binding with ACE2-hFc in similar signal strengths. Furthermore, in the NAb detection assay, % inhibition obtained from both E-RBD and H-RBD demonstrated comparable results in all the investigated assays, suggesting that non-glycosylated RBD produced from E. coli may offer a cost-effective alternative to the use of more expensive glycosylated RBD produced from human cells in the development of such an assay.


Subject(s)
COVID-19 , Spike Glycoprotein, Coronavirus , Humans , Spike Glycoprotein, Coronavirus/genetics , COVID-19 Vaccines , Escherichia coli/genetics , HEK293 Cells , Pandemics , SARS-CoV-2/genetics , Antibodies, Neutralizing , Antibodies, Viral
5.
Nat Protoc ; 16(11): 4992-5029, 2021 11.
Article in English | MEDLINE | ID: mdl-34650278

ABSTRACT

Comprehensive characterization of cellular heterogeneity and the underlying regulatory landscapes of tissues and organs requires a highly robust and scalable method to acquire matched RNA and chromatin accessibility profiles on the same cells. Here, we describe a single-nucleus chromatin accessibility and mRNA expression sequencing 2 (SNARE-seq2) assay, implemented with cellular combinatorial indexing. This method involves tagmentation within permeabilized and fixed single-nucleus isolates to capture accessible chromatin (AC) regions, followed by the capture and reverse transcription of RNA transcripts. Through combinatorial split pool ligations, cDNA and AC within each single nucleus become appended with a common cell barcode combination. The captured cDNA and AC are then co-amplified before splitting and enrichment into single-nucleus RNA and single-nucleus AC sequencing libraries. This protocol is compatible with both nuclei and whole cells and can be completed in 3.5 d. SNARE-seq2 permits robust generation of high-quality, joint single-cell RNA and AC sequencing libraries from hundreds of thousands of single cells per experiment.


Subject(s)
Chromatin , SNARE Proteins , Cell Nucleus , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , RNA, Messenger/genetics , Single-Cell Analysis
6.
Nat Commun ; 10(1): 2832, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31249312

ABSTRACT

Defining cellular and molecular identities within the kidney is necessary to understand its organization and function in health and disease. Here we demonstrate a reproducible method with minimal artifacts for single-nucleus Droplet-based RNA sequencing (snDrop-Seq) that we use to resolve thirty distinct cell populations in human adult kidney. We define molecular transition states along more than ten nephron segments spanning two major kidney regions. We further delineate cell type-specific expression of genes associated with chronic kidney disease, diabetes and hypertension, providing insight into possible targeted therapies. This includes expression of a hypertension-associated mechano-sensory ion channel in mesangial cells, and identification of proximal tubule cell populations defined by pathogenic expression signatures. Our fully optimized, quality-controlled transcriptomic profiling pipeline constitutes a tool for the generation of healthy and diseased molecular atlases applicable to clinical samples.


Subject(s)
Cell Nucleus/genetics , Kidney Diseases/genetics , Kidney/metabolism , Kidney/pathology , Sequence Analysis, RNA/methods , Aged , Cell Nucleus/metabolism , Female , Gene Expression Profiling , Humans , Kidney Diseases/diagnosis , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Mesangial Cells/metabolism , Middle Aged , Single-Cell Analysis/methods
7.
Methods Mol Biol ; 1708: 365-382, 2018.
Article in English | MEDLINE | ID: mdl-29224154

ABSTRACT

Bisulfite padlock probes (BSPP) are a method for the targeted quantification of DNA methylation in mammalian genomes. They can simultaneously characterize the level of methylcytosine modification in a large number of targeted regions at single-base resolution. A major advantage of BSPP is that it allows the flexible capture of an arbitrary subset of genomic regions (hundreds to hundreds of thousands of genomic loci) in single-tube reactions. Large number of samples can be processed efficiently and converted into multiplexed sequencing libraries with only three enzymatic steps, without the conventional library preparation procedures. BSPP are applicable to clinical studies, screening cell lines, and for quantifying low abundance regions using deep sequencing.


Subject(s)
DNA Methylation , Sequence Analysis, DNA/methods , CpG Islands , Epigenesis, Genetic , Genome, Human , Genomic Library , High-Throughput Nucleotide Sequencing , Humans , Sulfites
8.
Nat Genet ; 49(4): 635-642, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28263317

ABSTRACT

Adjacent CpG sites in mammalian genomes can be co-methylated owing to the processivity of methyltransferases or demethylases, yet discordant methylation patterns have also been observed, which are related to stochastic or uncoordinated molecular processes. We focused on a systematic search and investigation of regions in the full human genome that show highly coordinated methylation. We defined 147,888 blocks of tightly coupled CpG sites, called methylation haplotype blocks, after analysis of 61 whole-genome bisulfite sequencing data sets and validation with 101 reduced-representation bisulfite sequencing data sets and 637 methylation array data sets. Using a metric called methylation haplotype load, we performed tissue-specific methylation analysis at the block level. Subsets of informative blocks were further identified for deconvolution of heterogeneous samples. Finally, using methylation haplotypes we demonstrated quantitative estimation of tumor load and tissue-of-origin mapping in the circulating cell-free DNA of 59 patients with lung or colorectal cancer.


Subject(s)
DNA Methylation/genetics , DNA/genetics , Haplotypes/genetics , Chromosome Mapping/methods , CpG Islands/genetics , Genome, Human/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Sequence Analysis, DNA/methods
9.
Nat Rev Genet ; 15(10): 647-61, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25159599

ABSTRACT

Chemical modifications of DNA have been recognized as key epigenetic mechanisms for maintenance of the cellular state and memory. Such DNA modifications include canonical 5-methylcytosine (5mC), 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC) and 5-carboxycytosine (5caC). Recent advances in detection and quantification of DNA modifications have enabled epigenetic variation to be connected to phenotypic consequences on an unprecedented scale. These methods may use chemical or enzymatic DNA treatment, may be targeted or non-targeted and may utilize array-based hybridization or sequencing. Key considerations in the choice of assay are cost, minimum sample input requirements, accuracy and throughput. This Review discusses the principles behind recently developed techniques, compares their respective strengths and limitations and provides general guidelines for selecting appropriate methods for specific experimental contexts.


Subject(s)
Cytosine/metabolism , DNA Methylation , Gene Expression Profiling/trends , Algorithms , Animals , Chromosome Mapping/methods , Chromosome Mapping/trends , Epigenesis, Genetic/physiology , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing , Humans
10.
PLoS One ; 9(7): e99313, 2014.
Article in English | MEDLINE | ID: mdl-25019935

ABSTRACT

Genetic polymorphisms can shape the global landscape of DNA methylation, by either changing substrates for DNA methyltransferases or altering the DNA binding affinity of cis-regulatory proteins. The interactions between CpG methylation and genetic polymorphisms have been previously investigated by methylation quantitative trait loci (mQTL) and allele-specific methylation (ASM) analysis. However, it remains unclear whether these approaches can effectively and comprehensively identify all genetic variants that contribute to the inter-individual variation of DNA methylation levels. Here we used three independent approaches to systematically investigate the influence of genetic polymorphisms on variability in DNA methylation by characterizing the methylation state of 96 whole blood samples in 52 parent-child trios from 22 nuclear pedigrees. We performed targeted bisulfite sequencing with padlock probes to quantify the absolute DNA methylation levels at a set of 411,800 CpG sites in the human genome. With mid-parent offspring analysis (MPO), we identified 10,593 CpG sites that exhibited heritable methylation patterns, among which 70.1% were SNPs directly present in methylated CpG dinucleotides. We determined the mQTL analysis identified 49.9% of heritable CpG sites for which regulation occurred in a distal cis-regulatory manner, and that ASM analysis was only able to identify 5%. Finally, we identified hundreds of clusters in the human genome for which the degree of variation of CpG methylation, as opposed to whether or not CpG sites were methylated, was associated with genetic polymorphisms, supporting a recent hypothesis on the genetic influence of phenotypic plasticity. These results show that cis-regulatory SNPs identified by mQTL do not comprise the full extent of heritable CpG methylation, and that ASM appears overall unreliable. Overall, the extent of genome-methylome interactions is well beyond what is detectible with the commonly used mQTL and ASM approaches, and is likely to include effects on plasticity.


Subject(s)
DNA Methylation , Genome, Human/genetics , Nuclear Family , Pedigree , CpG Islands/genetics , Female , Genomics , Humans , Male , Polymorphism, Single Nucleotide , Quantitative Trait Loci/genetics
11.
Nat Commun ; 5: 4330, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-24999918

ABSTRACT

Fanconi anaemia (FA) is a recessive disorder characterized by genomic instability, congenital abnormalities, cancer predisposition and bone marrow (BM) failure. However, the pathogenesis of FA is not fully understood partly due to the limitations of current disease models. Here, we derive integration free-induced pluripotent stem cells (iPSCs) from an FA patient without genetic complementation and report in situ gene correction in FA-iPSCs as well as the generation of isogenic FANCA-deficient human embryonic stem cell (ESC) lines. FA cellular phenotypes are recapitulated in iPSCs/ESCs and their adult stem/progenitor cell derivatives. By using isogenic pathogenic mutation-free controls as well as cellular and genomic tools, our model serves to facilitate the discovery of novel disease features. We validate our model as a drug-screening platform by identifying several compounds that improve hematopoietic differentiation of FA-iPSCs. These compounds are also able to rescue the hematopoietic phenotype of FA patient BM cells.


Subject(s)
Drug Evaluation, Preclinical/methods , Fanconi Anemia/etiology , Fanconi Anemia/pathology , Models, Biological , Stem Cells/pathology , Cell Differentiation , Epigenesis, Genetic , Fanconi Anemia/drug therapy , Fanconi Anemia Complementation Group A Protein/genetics , Humans , Induced Pluripotent Stem Cells , Male , Young Adult
12.
Protein Cell ; 5(1): 59-68, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24474197

ABSTRACT

With defined culture protocol, human embryonic stem cells (hESCs) are able to generate cardiomyocytes in vitro, therefore providing a great model for human heart development, and holding great potential for cardiac disease therapies. In this study, we successfully generated a highly pure population of human cardiomyocytes (hCMs) (>95% cTnT(+)) from hESC line, which enabled us to identify and characterize an hCM-specific signature, at both the gene expression and DNA methylation levels. Gene functional association network and gene-disease network analyses of these hCM-enriched genes provide new insights into the mechanisms of hCM transcriptional regulation, and stand as an informative and rich resource for investigating cardiac gene functions and disease mechanisms. Moreover, we show that cardiac-structural genes and cardiac-transcription factors have distinct epigenetic mechanisms to regulate their gene expression, providing a better understanding of how the epigenetic machinery coordinates to regulate gene expression in different cell types.


Subject(s)
DNA Methylation , Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , Cell Differentiation , Cell Line , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Humans , Myocytes, Cardiac/metabolism , Transcription, Genetic
13.
Protein Cell ; 2013 Aug 27.
Article in English | MEDLINE | ID: mdl-23982742

ABSTRACT

With defined culture protocol, human embryonic stem cells (hESCs) are able to generate cardiomyocytes in vitro, therefore providing a great model for human heart development, and holding great potential for cardiac disease therapies. In this study, we successfully generated a highly pure population of human cardiomyocytes (hCMs) (>95% cTnT+) from hESC line, which enabled us to identify and characterize an hCM-specific signature, at both the gene expression and DNA methylation levels. Gene functional association network and gene-disease network analyses of these hCM-enriched genes provide new insights into the mechanisms of hCM transcriptional regulation, and stand as an informative and rich resource for investigating cardiac gene functions and disease mechanisms. Moreover, we show that cardiac-structural genes and cardiac-transcription factors have distinct epigenetic mechanisms to regulate their gene expression, providing a better understanding of how the epigenetic machinery coordinates to regulate gene expression in different cell types.

14.
Proc Natl Acad Sci U S A ; 109(40): 16196-201, 2012 Oct 02.
Article in English | MEDLINE | ID: mdl-22991473

ABSTRACT

Generation of human induced pluripotent stem cells (hiPSCs) by the expression of specific transcription factors depends on successful epigenetic reprogramming to a pluripotent state. Although hiPSCs and human embryonic stem cells (hESCs) display a similar epigenome, recent reports demonstrated the persistence of specific epigenetic marks from the somatic cell type of origin and aberrant methylation patterns in hiPSCs. However, it remains unknown whether the use of different somatic cell sources, encompassing variable levels of selection pressure during reprogramming, influences the level of epigenetic aberrations in hiPSCs. In this work, we characterized the epigenomic integrity of 17 hiPSC lines derived from six different cell types with varied reprogramming efficiencies. We demonstrate that epigenetic aberrations are a general feature of the hiPSC state and are independent of the somatic cell source. Interestingly, we observe that the reprogramming efficiency of somatic cell lines inversely correlates with the amount of methylation change needed to acquire pluripotency. Additionally, we determine that both shared and line-specific epigenetic aberrations in hiPSCs can directly translate into changes in gene expression in both the pluripotent and differentiated states. Significantly, our analysis of different hiPSC lines from multiple cell types of origin allow us to identify a reprogramming-specific epigenetic signature comprised of nine aberrantly methylated genes that is able to segregate hESC and hiPSC lines regardless of the somatic cell source or differentiation state.


Subject(s)
Cellular Reprogramming/physiology , DNA Methylation/genetics , Epigenesis, Genetic/physiology , Induced Pluripotent Stem Cells/physiology , Cell Line , Cellular Reprogramming/genetics , CpG Islands/genetics , Epigenesis, Genetic/genetics , Epigenomics , Fluorescent Antibody Technique , Gene Library , Humans , Microarray Analysis , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA
15.
Nat Methods ; 9(3): 270-2, 2012 Feb 05.
Article in English | MEDLINE | ID: mdl-22306810

ABSTRACT

Targeted quantification of DNA methylation allows for interrogation of the most informative loci across many samples quickly and cost-effectively. Here we report improved bisulfite padlock probes (BSPPs) with a design algorithm to generate efficient padlock probes, a library-free protocol that dramatically reduces sample-preparation cost and time and is compatible with automation, and an efficient bioinformatics pipeline to accurately obtain both methylation levels and genotypes from sequencing of bisulfite-converted DNA.


Subject(s)
DNA Probes/chemistry , DNA Probes/genetics , DNA/chemistry , DNA/genetics , Polymerase Chain Reaction/methods , Sequence Analysis, DNA/methods , Sulfites/chemistry , Base Sequence , Gene Library , Molecular Sequence Data
16.
Cell Res ; 22(1): 168-77, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22064701

ABSTRACT

Metabolism is vital to every aspect of cell function, yet the metabolome of induced pluripotent stem cells (iPSCs) remains largely unexplored. Here we report, using an untargeted metabolomics approach, that human iPSCs share a pluripotent metabolomic signature with embryonic stem cells (ESCs) that is distinct from their parental cells, and that is characterized by changes in metabolites involved in cellular respiration. Examination of cellular bioenergetics corroborated with our metabolomic analysis, and demonstrated that somatic cells convert from an oxidative state to a glycolytic state in pluripotency. Interestingly, the bioenergetics of various somatic cells correlated with their reprogramming efficiencies. We further identified metabolites that differ between iPSCs and ESCs, which revealed novel metabolic pathways that play a critical role in regulating somatic cell reprogramming. Our findings are the first to globally analyze the metabolome of iPSCs, and provide mechanistic insight into a new layer of regulation involved in inducing pluripotency, and in evaluating iPSC and ESC equivalence.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells/metabolism , Metabolome , DNA Methylation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Energy Metabolism , Gene Expression Regulation , Glycolysis , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Induced Pluripotent Stem Cells/cytology , Oxidation-Reduction , Oxidative Phosphorylation , Plasmids/genetics , Plasmids/metabolism , Retroviridae/genetics , Retroviridae/metabolism
17.
Cell Stem Cell ; 8(6): 688-94, 2011 Jun 03.
Article in English | MEDLINE | ID: mdl-21596650

ABSTRACT

Combination of stem cell-based approaches with gene-editing technologies represents an attractive strategy for studying human disease and developing therapies. However, gene-editing methodologies described to date for human cells suffer from technical limitations including limited target gene size, low targeting efficiency at transcriptionally inactive loci, and off-target genetic effects that could hamper broad clinical application. To address these limitations, and as a proof of principle, we focused on homologous recombination-based gene correction of multiple mutations on lamin A (LMNA), which are associated with various degenerative diseases. We show that helper-dependent adenoviral vectors (HDAdVs) provide a highly efficient and safe method for correcting mutations in large genomic regions in human induced pluripotent stem cells and can also be effective in adult human mesenchymal stem cells. This type of approach could be used to generate genotype-matched cell lines for disease modeling and drug discovery and potentially also in therapeutics.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Lamin Type A/genetics , Mutation , Cell Line , Genotype , Humans , Induced Pluripotent Stem Cells/pathology
18.
Biochem Biophys Res Commun ; 354(3): 789-94, 2007 Mar 16.
Article in English | MEDLINE | ID: mdl-17266933

ABSTRACT

The ability of cells to survive and recover from deteriorating effects of endoplasmic reticulum (ER) stress relies on the unfolded protein response (UPR). The signaling pathway of Ire1p mediate mRNA splicing plays diverge role in UPR response in different organisms from yeast to mammals. Here, we report that Ire1p mediated XBP1 mRNA splicing mechanism is extremely conserved and exerts a critical role for modulating Xbp1 protein synthesis in Drosophila melanogaster. This system is operative in Drosophila S2 cells as a prominent mechanism to mediate transcriptional activation of UPR responsive genes during ER stress.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Endoribonucleases/physiology , Molecular Chaperones/metabolism , RNA Splicing , Saccharomyces cerevisiae Proteins/metabolism , Animals , Base Sequence , Cells, Cultured , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster , Endoplasmic Reticulum/metabolism , Oxidative Stress/physiology
19.
Antiviral Res ; 74(2): 150-5, 2007 May.
Article in English | MEDLINE | ID: mdl-17166601

ABSTRACT

Yellow head virus (YHV) is an invertebrate nidovirus which causes a severe mortality in cultured Penaeus monodon. The mortality may be prevented by prior treatment of shrimps with YHV-protease dsRNA. Whether the YHV infected shrimp might be cured by the dsRNA remains to be investigated. P. monodon injected with 10(-6) YHV showed a high virus replication and mortality within 2 days. Injection of 25 microg YHV-protease dsRNA at 3, 6, 12 or 24 h post YHV infection showed a strong inhibition of YHV replication up to 12 h. Unrelated dsRNA-GFP showed no inhibition, indicating that the inhibition was nucleic acid sequence specific through RNAi pathway. Shrimp mortality could be prevented at 3h post YHV infection by the dsRNA, but not at 24 h. These results demonstrate that YHV-protease dsRNA gives therapeutic effect and pave the way to develop a cure for YHV-infected shrimps.


Subject(s)
Antiviral Agents/pharmacology , Penaeidae/virology , RNA, Double-Stranded/pharmacology , RNA, Viral/antagonists & inhibitors , Roniviridae/drug effects , Viral Proteins/biosynthesis , Animals , Disease Models, Animal , Peptide Hydrolases/biosynthesis , Peptide Hydrolases/genetics , RNA Interference , RNA, Double-Stranded/genetics , RNA, Viral/genetics , Roniviridae/genetics , Roniviridae/physiology , Viral Proteins/genetics , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL