Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
PLoS Pathog ; 15(2): e1007536, 2019 02.
Article in English | MEDLINE | ID: mdl-30716130

ABSTRACT

Extracellular signaling is a mechanism that higher eukaryotes have evolved to facilitate organismal homeostasis. Recent years have seen an emerging interest in the role of secreted microvesicles, termed extracellular vesicles (EV) or exosomes in this signaling network. EV contents can be modified by the cell in response to stimuli, allowing them to relay information to neighboring cells, influencing their physiology. Here we show that the tumor virus Kaposi's Sarcoma-associated herpesvirus (KSHV) hijacks this signaling pathway to induce cell proliferation, migration, and transcriptome reprogramming in cells not infected with the virus. KSHV-EV activates the canonical MEK/ERK pathway, while not alerting innate immune regulators, allowing the virus to exert these changes without cellular pathogen recognition. Collectively, we propose that KSHV establishes a niche favorable for viral spread and cell transformation through cell-derived vesicles, all while avoiding detection.


Subject(s)
Cellular Reprogramming/physiology , Extracellular Vesicles/physiology , Herpesvirus 8, Human/metabolism , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cellular Reprogramming/genetics , Endothelial Cells/physiology , Herpesvirus 8, Human/genetics , Host-Pathogen Interactions , Human Umbilical Vein Endothelial Cells , Humans , Lymphoma/genetics , Lymphoma/metabolism , Sarcoma, Kaposi/metabolism , Sarcoma, Kaposi/virology , Signal Transduction , Transcriptome/genetics , Viral Proteins , Virus Latency
2.
J Virol ; 88(2): 1389-93, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24227849

ABSTRACT

Latent infection with Epstein-Barr virus (EBV) is responsible for multiple types of malignancies, including 10% of all gastric carcinomas. The microRNA (miRNA) expression in several EBV-infected AGS gastric carcinoma cell lines was determined. Infected cells expressed the viral BamHI A rightward transcript (BART) miRNAs at high levels and had consistently decreased expression of a small fraction of cellular miRNAs with specific downregulation of tumor suppressor miRNAs. These changes likely reflect expression of the viral noncoding RNAs and not latent protein expression.


Subject(s)
Carcinoma/genetics , Epstein-Barr Virus Infections/genetics , Herpesvirus 4, Human/genetics , MicroRNAs/genetics , RNA, Viral/genetics , Stomach Neoplasms/genetics , Carcinoma/metabolism , Carcinoma/virology , Cell Line, Tumor , Down-Regulation , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/physiology , Humans , MicroRNAs/metabolism , RNA, Viral/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/virology , Transcriptome
3.
J Virol ; 88(10): 5778-87, 2014 May.
Article in English | MEDLINE | ID: mdl-24623417

ABSTRACT

UNLABELLED: Kaposi's sarcoma-associated herpesvirus (KSHV) has been shown to be recognized by two families of pattern recognition receptors (PRRs), Toll-like receptors (TLRs) and NOD-like receptors (NLRs). Here we show that MAVS and RIG-I (retinoic acid-inducible gene 1), an RLR family member, also have a role in suppressing KSHV replication and production. In the context of primary infection, we show that in cells with depleted levels of MAVS or RIG-I, KSHV transcription is increased, while beta interferon (IFN-ß) induction is attenuated. We also observed that MAVS and RIG-I are critical during the process of reactivation. Depletion of MAVS and RIG-I prior to reactivation led to increased viral load and production of infectious virus. Finally, MAVS depletion in latent KSHV-infected B cells leads to increased viral gene transcription. Overall, this study suggests a role for MAVS and RIG-I signaling during different stages of the KSHV life cycle. IMPORTANCE: We show that RIG-I and its adaptor protein, MAVS, can sense KSHV infection and that these proteins can suppress KSHV replication following primary infection and/or viral reactivation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DEAD-box RNA Helicases/metabolism , Herpesvirus 8, Human/immunology , Herpesvirus 8, Human/physiology , Host-Pathogen Interactions , Virus Replication , Cell Line , DEAD Box Protein 58 , Humans , Receptors, Immunologic , Signal Transduction , Virus Activation
4.
PLoS Pathog ; 9(7): e1003484, 2013.
Article in English | MEDLINE | ID: mdl-23874201

ABSTRACT

MicroRNAs (miRNAs) are stable, small non-coding RNAs that modulate many downstream target genes. Recently, circulating miRNAs have been detected in various body fluids and within exosomes, prompting their evaluation as candidate biomarkers of diseases, especially cancer. Kaposi's sarcoma (KS) is the most common AIDS-associated cancer and remains prevalent despite Highly Active Anti-Retroviral Therapy (HAART). KS is caused by KS-associated herpesvirus (KSHV), a gamma herpesvirus also associated with Primary Effusion Lymphoma (PEL). We sought to determine the host and viral circulating miRNAs in plasma, pleural fluid or serum from patients with the KSHV-associated malignancies KS and PEL and from two mouse models of KS. Both KSHV-encoded miRNAs and host miRNAs, including members of the miR-17-92 cluster, were detectable within patient exosomes and circulating miRNA profiles from KSHV mouse models. Further characterization revealed a subset of miRNAs that seemed to be preferentially incorporated into exosomes. Gene ontology analysis of signature exosomal miRNA targets revealed several signaling pathways that are known to be important in KSHV pathogenesis. Functional analysis of endothelial cells exposed to patient-derived exosomes demonstrated enhanced cell migration and IL-6 secretion. This suggests that exosomes derived from KSHV-associated malignancies are functional and contain a distinct subset of miRNAs. These could represent candidate biomarkers of disease and may contribute to the paracrine phenotypes that are a characteristic of KS.


Subject(s)
Herpesvirus 8, Human/isolation & purification , MicroRNAs/blood , RNA, Neoplasm/blood , RNA, Viral/blood , Sarcoma, Kaposi/diagnosis , Animals , Biomarkers/blood , Biomarkers/metabolism , Body Fluids/metabolism , Body Fluids/virology , Cell Line , Cell Movement , Cells, Cultured , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/virology , Exosomes/metabolism , Exosomes/ultrastructure , Exosomes/virology , Gene Expression Profiling , Herpesvirus 8, Human/metabolism , Humans , Interleukin-6/metabolism , Mice , MicroRNAs/metabolism , Pleural Cavity , Pleural Effusion, Malignant/etiology , RNA, Neoplasm/metabolism , RNA, Viral/metabolism , Sarcoma, Kaposi/metabolism , Sarcoma, Kaposi/physiopathology , Sarcoma, Kaposi/virology , Up-Regulation , Viral Load
5.
Bioinformatics ; 29(9): 1105-11, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23428639

ABSTRACT

MOTIVATION: Next-generation (NextGen) sequencing is becoming increasingly popular as an alternative for transcriptional profiling, as is the case for micro RNAs (miRNA) profiling and classification. miRNAs are a new class of molecules that are regulated in response to differentiation, tumorigenesis or infection. Our primary motivating application is to identify different viral infections based on the induced change in the host miRNA profile. Statistical challenges are encountered because of special features of NextGen sequencing data: the data are read counts that are extremely skewed and non-negative; the total number of reads varies dramatically across samples that require appropriate normalization. Statistical tools developed for microarray expression data, such as principal component analysis, are sub-optimal for analyzing NextGen sequencing data. RESULTS: We propose a family of Poisson factor models that explicitly takes into account the count nature of sequencing data and automatically incorporates sample normalization through the use of offsets. We develop an efficient algorithm for estimating the Poisson factor model, entitled Poisson Singular Value Decomposition with Offset (PSVDOS). The method is shown to outperform several other normalization and dimension reduction methods in a simulation study. Through analysis of an miRNA profiling experiment, we further illustrate that our model achieves insightful dimension reduction of the miRNA profiles of 18 samples: the extracted factors lead to more accurate and meaningful clustering of the cell lines. AVAILABILITY: The PSVDOS software is available on request.


Subject(s)
Algorithms , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing , MicroRNAs/metabolism , Cluster Analysis , Humans , Models, Statistical , Poisson Distribution , Software
6.
Am J Respir Crit Care Med ; 183(10): 1391-401, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21169470

ABSTRACT

RATIONALE: Neutrophils are usually the first circulating leukocytes to respond during bacterial pneumonia. Their expression of oxidants, proteases, and other mediators present in granules is well documented, but their ability to produce mediators through transcription and translation after migration to an inflammatory site has been appreciated only more recently. Interferon (IFN)-γ is a cytokine with many functions important in host defense and immunity. OBJECTIVES: To examine the expression and function of IFN-γ in bacterial pneumonias. METHODS: IFN-γ mRNA and protein were measured in digests of mouse lungs with 24-hour bacterial pneumonia. Bacterial clearance was studied with IFN-γ-deficient mice. MEASUREMENTS AND MAIN RESULTS: Streptococcus pneumoniae and Staphylococcus aureus each induce expression of IFN-γ mRNA and protein by neutrophils by 24 hours. Only neutrophils that have migrated into pneumonic tissue produce IFN-γ. Deficiency of Hck/Fgr/Lyn, Rac2, or gp91(phox) prevents IFN-γ production. IFN-γ enhances bacterial clearance and is required for formation of neutrophil extracellular traps. In contrast, Pseudomonas aeruginosa and Escherichia coli induce production of IFN-γ mRNA but not protein. During pneumonia induced by E. coli but not S. pneumoniae, neutrophils produce microRNAs that target the 3' untranslated region of the IFN-γ gene. CONCLUSIONS: S. pneumoniae and S. aureus, but not P. aeruginosa and E. coli, induce emigrated neutrophils to produce IFN-γ within 24 hours. Hck/Fgr/Lyn, Rac2, and NADPH oxidase are required for IFN-γ production. IFN-γ facilitates bacterial clearance at least in part through regulating formation of neutrophil extracellular traps. Differential expression by neutrophils of microRNAs that target the 3' untranslated region of the IFN-γ gene may contribute to the pathogen-specific regulation of translation.


Subject(s)
Interferon-gamma/immunology , Neutrophils/immunology , Pneumonia, Bacterial/immunology , Animals , Bronchoalveolar Lavage Fluid/immunology , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Escherichia coli/immunology , Immunity, Innate/immunology , Lung/immunology , Mice , Mice, Inbred C57BL , Pseudomonas aeruginosa/immunology , Reverse Transcriptase Polymerase Chain Reaction , Staphylococcus aureus/immunology , Streptococcus pneumoniae/immunology
7.
PLoS Pathog ; 5(4): e1000389, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19381257

ABSTRACT

MicroRNAs (miRNA) have emerged as key regulators of cell lineage differentiation and cancer. We used precursor miRNA profiling by a novel real-time QPCR method (i) to define progressive stages of endothelial cell transformation cumulating in Kaposi sarcoma (KS) and (ii) to identify specific miRNAs that serve as biomarkers for tumor progression. We were able to compare primary patient biopsies to well-established culture and mouse tumor models. Loss of mir-221 and gain of mir-15 expression demarked the transition from merely immortalized to fully tumorigenic endothelial cells. Mir-140 and Kaposi sarcoma-associated herpesvirus viral miRNAs increased linearly with the degree of transformation. Mir-24 emerged as a biomarker specific for KS.


Subject(s)
Cell Transformation, Neoplastic/pathology , Endothelial Cells/pathology , MicroRNAs/genetics , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/pathology , Animals , Cell Line, Tumor , Humans , Mice , MicroRNAs/physiology , Polymerase Chain Reaction
8.
Retrovirology ; 6: 57, 2009 Jun 08.
Article in English | MEDLINE | ID: mdl-19505314

ABSTRACT

BACKGROUND: The acute phase of immunodeficiency virus infection plays a crucial role in determining steady-state virus load and subsequent progression of disease in both humans and nonhuman primates. The acute period is also the time when vaccine-mediated effects on host immunity are likely to exert their major effects on virus infection. Recently we developed a Monte-Carlo (MC) simulation with mathematical analysis of viral evolution during primary HIV-1 infection that enables classification of new HIV-1 infections originating from multiple versus single transmitted viral strains and the estimation of time elapsed following infection. RESULTS: A total of 322 SIV nef SIV sequences, collected during the first 3 weeks following experimental infection of two rhesus macaques with the SIVmac239 clone, were analyzed and found to display a comparable level of genetic diversity, 0.015% to 0.052%, with that of env sequences from acute HIV-1 infection, 0.005% to 0.127%. We confirmed that the acute HIV-1 infection model correctly identified the experimental SIV infections in rhesus macaques as "homogenous" infections, initiated by a single founder strain. The consensus sequence of the sampled strains corresponded to the transmitted sequence as the model predicted. However, measured sequential decrease in diversity at day 7, 11, and 18 post infection violated the model assumption, neutral evolution without any selection. CONCLUSION: While nef gene evolution over the first 3 weeks of SIV infection originating from a single transmitted strain showed a comparable rate of sequence evolution to that observed during acute HIV-1 infection, a purifying selection for the founder nef gene was observed during the early phase of experimental infection of a nonhuman primate.


Subject(s)
Evolution, Molecular , Sequence Analysis, DNA , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Viral Regulatory and Accessory Proteins/genetics , Acute Disease , Animals , Genetic Variation , HIV Infections/virology , HIV-1/genetics , Macaca mulatta , Models, Genetic , Monte Carlo Method , Point Mutation , RNA, Viral/analysis , RNA, Viral/genetics , Simian Acquired Immunodeficiency Syndrome/physiopathology , env Gene Products, Human Immunodeficiency Virus/genetics
9.
Retrovirology ; 5: 11, 2008 Jan 31.
Article in English | MEDLINE | ID: mdl-18237430

ABSTRACT

BACKGROUND: Unlike CD4+ T cells, HIV-1 infected macrophages exhibit extended life span even upon stress, consistent with their in vivo role as long-lived HIV-1 reservoirs. RESULTS: Here, we demonstrate that PI3K/Akt inhibitors, including clinically available Miltefosine, dramatically reduced HIV-1 production from long-living virus-infected macrophages. These PI3K/Akt inhibitors hyper-sensitize infected macrophages to extracellular stresses that they are normally exposed to, and eventually lead to cell death of infected macrophages without harming uninfected cells. Based on the data from these Akt inhibitors, we were able to further investigate how HIV-1 infection utilizes the PI3K/Akt pathway to establish the cytoprotective effect of HIV-1 infection, which extends the lifespan of infected macrophages, a key viral reservoir. First, we found that HIV-1 infection activates the well characterized pro-survival PI3K/Akt pathway in primary human macrophages, as reflected by decreased PTEN protein expression and increased Akt kinase activity. Interestingly, the expression of HIV-1 or SIV Tat is sufficient to mediate this cytoprotective effect, which is dependent on the basic domain of Tat - a region that has previously been shown to bind p53. Next, we observed that this interaction appears to contribute to the downregulation of PTEN expression, since HIV-1 Tat was found to compete with PTEN for p53 binding; this is known to result in p53 destabilization, with a consequent reduction in PTEN protein production. CONCLUSION: Since HIV-1 infected macrophages display highly elevated Akt activity, our results collectively show that PI3K/Akt inhibitors may be a novel therapy for interfering with the establishment of long-living HIV-1 infected reservoirs.


Subject(s)
HIV Infections/virology , HIV-1/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/pharmacology , Cells, Cultured , Down-Regulation , HIV-1/physiology , Humans , Macrophages , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Virus Replication/drug effects
10.
J Mol Biol ; 366(1): 67-81, 2007 Feb 09.
Article in English | MEDLINE | ID: mdl-17157319

ABSTRACT

The interaction of human immunodeficiency virus type 1 (HIV-1) with CD4+ T lymphocytes is well studied and typically results in virally induced cytolysis. In contrast, relatively little is known concerning the interplay between HIV-1 and microglia. Recent findings suggest that, counter-intuitively, HIV-1 infection may extend the lifespan of microglia. We developed a novel cell line model system to confirm and mechanistically study this phenomenon. We found that transduction of a human microglial cell line with an HIV-1 vector results in a powerful cytoprotective effect following apoptotic challenge. This effect was reproduced by ectopic expression of a single virus-encoded protein, Tat. Subsequent studies showed that the pro-survival effects of intracellular Tat could be attributed to activation of the PI-3-kinase (PI3K)/Akt pathway in the microglial cell line. Furthermore, we found that expression of Tat led to decreased expression of PTEN, a negative regulator of the PI-3-K pathway. Consistent with this, decreased p53 activity and increased E2F activity were observed. Based on these findings, a model of possible regulatory circuits that intracellular Tat and HIV-1 infection engage during the cytoprotective event in microglia has been suggested. We propose that the expression of Tat may enable HIV-1 infected microglia to survive throughout the course of infection, leading to persistent HIV-1 production and infection in the central nervous system.


Subject(s)
Gene Products, tat/physiology , HIV-1/physiology , Microglia/virology , Cell Line , Cell Proliferation , Cell Survival , Cells, Cultured , Gene Products, tat/metabolism , HIV-1/genetics , Humans , Macrophages/physiology , Macrophages/virology , Microglia/cytology , Microglia/physiology , Models, Biological , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transduction, Genetic , Transfection , tat Gene Products, Human Immunodeficiency Virus
11.
PLoS One ; 9(8): e104770, 2014.
Article in English | MEDLINE | ID: mdl-25127040

ABSTRACT

The innate immune response to West Nile virus (WNV) infection involves recognition through toll-like receptors (TLRs) and RIG-I-like receptors (RLRs), leading to establishment of an antiviral state. MiRNAs (miRNAs) have been shown to be reliable biomarkers of TLR activation. Here, we sought to evaluate the contribution of TLR3 and miRNAs to the host response to WNV infection. We first analyzed HEK293-NULL and HEK293-TLR3 cells for changes in the innate immune response to infection. The presence of TLR3 did not seem to affect WNV load, infectivity or phosphorylation of IRF3. Analysis of experimentally validated NFκB-responsive genes revealed a WNV-induced signature largely independent of TLR3. Since miRNAs are involved in viral pathogenesis and the innate response to infection, we sought to identify changes in miRNA expression upon infection in the presence or absence of TLR3. MiRNA profiling revealed 70 miRNAs induced following WNV infection in a TLR3-independent manner. Further analysis of predicted gene targets of WNV signature miRNAs revealed genes highly associated with pathways regulating cell death, viral pathogenesis and immune cell trafficking.


Subject(s)
Interferon Regulatory Factor-3/metabolism , MicroRNAs/immunology , NF-kappa B/metabolism , Toll-Like Receptor 3/metabolism , West Nile virus/immunology , Cell Line , DEAD Box Protein 58 , DEAD-box RNA Helicases/metabolism , Gene Expression Profiling , HEK293 Cells , Humans , Immunity, Innate/immunology , Interferon Regulatory Factor-3/genetics , Interferon-Induced Helicase, IFIH1 , MicroRNAs/genetics , NF-kappa B/genetics , RNA, Messenger/genetics , Receptors, Immunologic , Signal Transduction , Toll-Like Receptor 3/genetics , Viral Load , West Nile Fever/immunology
12.
Nat Med ; 20(8): 927-35, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25064127

ABSTRACT

Oxidative tissue injury often accompanies viral infection, yet there is little understanding of how it influences virus replication. We show that multiple hepatitis C virus (HCV) genotypes are exquisitely sensitive to oxidative membrane damage, a property distinguishing them from other pathogenic RNA viruses. Lipid peroxidation, regulated in part through sphingosine kinase-2, severely restricts HCV replication in Huh-7 cells and primary human hepatoblasts. Endogenous oxidative membrane damage lowers the 50% effective concentration of direct-acting antivirals in vitro, suggesting critical regulation of the conformation of the NS3-4A protease and the NS5B polymerase, membrane-bound HCV replicase components. Resistance to lipid peroxidation maps genetically to transmembrane and membrane-proximal residues within these proteins and is essential for robust replication in cell culture, as exemplified by the atypical JFH1 strain of HCV. Thus, the typical, wild-type HCV replicase is uniquely regulated by lipid peroxidation, providing a mechanism for attenuating replication in stressed tissue and possibly facilitating long-term viral persistence.


Subject(s)
Hepacivirus/enzymology , Lipid Peroxidation , Oxidative Stress , RNA-Dependent RNA Polymerase/metabolism , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects , Adaptor Proteins, Signal Transducing/genetics , Antiviral Agents/pharmacology , Cell Line , Cell Membrane/pathology , Hepacivirus/drug effects , Hepacivirus/genetics , Hepatitis C/drug therapy , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , RNA Interference , RNA, Small Interfering/genetics , Viral Nonstructural Proteins/genetics
13.
Wiley Interdiscip Rev RNA ; 3(5): 601-16, 2012.
Article in English | MEDLINE | ID: mdl-22566380

ABSTRACT

MicroRNAs (miRNAs) are small, noncoding RNAs that post-transcriptionally influence a wide range of cellular processes such as the host response to viral infection, innate immunity, cell cycle progression, migration, and apoptosis through the inhibition of target mRNA translation. Owing to the growing number of miRNAs and identification of their functional roles, miRNA profiling of many different sample types has become more expansive, especially with relevance to disease signatures. In this review, we address some of the advantages and potential pitfalls of the currently available methods for miRNA expression profiling. Some of the topics discussed include isomiRNAs, comparison of different profiling platforms, normalization strategies, and issues with regard to sample preparation and experimental analyses.


Subject(s)
Gene Expression Profiling/methods , MicroRNAs/analysis , MicroRNAs/genetics , Animals , Gene Expression Profiling/economics , Gene Expression Profiling/standards , Humans , Neoplasms/genetics , RNA Processing, Post-Transcriptional , Specimen Handling/methods , Virus Diseases/genetics , Virus Diseases/virology
14.
J Vis Exp ; (46)2010 Dec 03.
Article in English | MEDLINE | ID: mdl-21178961

ABSTRACT

Quantitative real-time PCR (QPCR) has emerged as an accurate and valuable tool in profiling gene expression levels. One of its many advantages is a lower detection limit compared to other methods of gene expression profiling while using smaller amounts of input for each assay. Automated qPCR setup has improved this field by allowing for greater reproducibility. Its convenient and rapid setup allows for high-throughput experiments, enabling the profiling of many different genes simultaneously in each experiment. This method along with internal plate controls also reduces experimental variables common to other techniques. We recently developed a qPCR assay for profiling of pre-microRNAs (pre-miRNAs) using a set of 186 primer pairs. MicroRNAs have emerged as a novel class of small, non-coding RNAs with the ability to regulate many mRNA targets at the post-transcriptional level. These small RNAs are first transcribed by RNA polymerase II as a primary miRNA (pri-miRNA) transcript, which is then cleaved into the precursor miRNA (pre-miRNA). Pre-miRNAs are exported to the cytoplasm where Dicer cleaves the hairpin loop to yield mature miRNAs. Increases in miRNA levels can be observed at both the precursor and mature miRNA levels and profiling of both of these forms can be useful. There are several commercially available assays for mature miRNAs; however, their high cost may deter researchers from this profiling technique. Here, we discuss a cost-effective, reliable, SYBR-based qPCR method of profiling pre-miRNAs. Changes in pre-miRNA levels often reflect mature miRNA changes and can be a useful indicator of mature miRNA expression. However, simultaneous profiling of both pre-miRNAs and mature miRNAs may be optimal as they can contribute nonredundant information and provide insight into microRNA processing. Furthermore, the technique described here can be expanded to encompass the profiling of other library sets for specific pathways or pathogens.


Subject(s)
Gene Expression Profiling/methods , MicroRNAs/analysis , Polymerase Chain Reaction/methods , Humans , MicroRNAs/genetics
15.
J Biol Chem ; 283(14): 9206-16, 2008 Apr 04.
Article in English | MEDLINE | ID: mdl-18218633

ABSTRACT

We characterized HIV-1 reverse transcriptase (RT) variants either with or without the (-)-2',3'-deoxy-3'-thiacytidine-resistant M184I mutation isolated from a single HIV-1 infected patient. First, unlike variants with wild-type M184, M184I RT variants displayed significantly reduced DNA polymerase activity at low dNTP concentrations, which is indicative of reduced dNTP binding affinity. Second, the M184I variant displayed a approximately 10- to 13-fold reduction in dNTP binding affinity, compared with the Met-184 variant. However, the k(pol) values of these two RTs were similar. Third, unlike HIV-1 vectors with wild-type RT, the HIV-1 vector harboring M184I RT failed to transduce cell types containing low dNTP concentrations, such as human macrophage, likely due to the reduced DNA polymerization activity of the M184I RT under low cellular dNTP concentration conditions. Finally, we compared the binary complex structures of wild-type and M184I RTs. The Ile mutation at position 184 with a longer and more rigid beta-branched side chain, which was previously known to alter the RT-template interaction, also appears to deform the shape of the dNTP binding pocket. This can restrict ground state dNTP binding and lead to inefficient DNA synthesis particularly at low dNTP concentrations, ultimately contributing to viral replication failure in macrophage and instability in vivo of the M184I mutation.


Subject(s)
Deoxyribonucleotides/metabolism , Drug Resistance, Viral , HIV Infections/enzymology , HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , Macrophages/metabolism , Mutation, Missense , Amino Acid Substitution , Cell Line , DNA, Viral/biosynthesis , DNA, Viral/genetics , Drug Resistance, Viral/genetics , HIV Infections/genetics , HIV Reverse Transcriptase/genetics , HIV-1/genetics , Humans , Lamivudine/pharmacology , Macrophages/virology , Protein Structure, Quaternary/genetics , Protein Structure, Secondary/genetics , Reverse Transcriptase Inhibitors/pharmacology
16.
J Biol Chem ; 281(19): 13388-13395, 2006 May 12.
Article in English | MEDLINE | ID: mdl-16497663

ABSTRACT

Retroviruses and DNA viruses utilize cellular dNTPs as substrates for their DNA polymerases during viral replication in infected cells. However, because of S phase-dependent dNTP biosynthesis, the availability of cellular dNTPs significantly varies among cell types (e.g. dividing versus nondividing cells and normal versus tumor cells). Here we tested whether alterations in the dNTP utilization efficiency and dNTP binding affinity of viral DNA polymerases can switch viral infection specificity to cell types with different dNTP concentrations. We employed an HIV-1 reverse transcriptase (RT) mutant (Q151N), which is catalytically active only at high dNTP concentrations because of its reduced dNTP binding affinity. Indeed, the modified HIV-1 vector harboring the Q151N mutant RT preferentially transduced tumor cells containing higher cellular dNTP concentrations than primary cells (e.g. human lung fibroblasts (HLFs) and human keratinocytes). Although the wild type HIV-1 vector transduced both HLFs and tumor cells, the Q151N vector failed to transduce HLFs and keratinocytes but efficiently transduced tumor cells. Pretreatment of HLFs with deoxynucleosides, which increase cellular dNTP pools, enabled the mutant vector to transduce HLFs, suggesting that the transduction failure of the RT mutant vector to primary cells is because of inefficient reverse transcription in low cellular dNTP environments. We also observed that the Q151N vector expressing herpes simplex virus-thymidine kinase renders tumor cells sensitive to gancyclovir. This study validates a novel strategy in which modifications of viral DNA polymerases in various vector systems allow the delivery of target genes exclusively to tumor cells exploiting elevated cellular dNTP concentration as a tumor cell-specific host factor.


Subject(s)
HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , Nucleosides/metabolism , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/virology , Gene Expression Regulation, Viral , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/genetics , HIV-1/genetics , Humans , Keratinocytes/metabolism , Keratinocytes/virology , Macrophages/metabolism , Macrophages/virology , Mutation , Organ Specificity
SELECTION OF CITATIONS
SEARCH DETAIL