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










Publication year range
1.
J Mol Biol ; 434(5): 167403, 2022 03 15.
Article in English | MEDLINE | ID: mdl-34914966

ABSTRACT

COVID-19 caused by SARS-CoV-2 is the latest pandemic which has thrown the world into an unprecedented social and economic uncertainties along with huge loss to humanity. Identification of the host factors regulating the replication of SARS-CoV-2 in human host may help in the development of novel anti-viral therapies to combat the viral infection and spread. Recently, some research groups used genome-wide CRISPR/Cas screening to identify the host factors critical for the SARS-CoV-2 replication and infection. A comparative analysis of these significant host factors (p < 0.05) identified fifteen proteins common in these studies. Apart from ACE2 (receptor for SARS-CoV-2 attachment), other common host factors were CSNK2B, GDI2, SLC35B2, DDX51, VPS26A, ARPP-19, C1QTNF7, ALG6, LIMA1, COG3, COG8, BCOR, LRRN2 and TLR9. Additionally, viral interactome of these host factors revealed that many of them were associated with several SARS-CoV-2 proteins as well. Interestingly, some of these host factors have already been shown to be critical for the pathogenesis of other viruses suggesting their crucial role in virus-host interactions. Here, we review the functions of these host factors and their role in other diseases with special emphasis on viral diseases.


Subject(s)
COVID-19/virology , Host Microbial Interactions , Host-Derived Cellular Factors/metabolism , Pandemics , SARS-CoV-2/physiology , COVID-19/epidemiology , Clustered Regularly Interspaced Short Palindromic Repeats , Host-Derived Cellular Factors/genetics , Humans , SARS-CoV-2/genetics
2.
Clin Microbiol Rev ; 33(3)2020 06 17.
Article in English | MEDLINE | ID: mdl-32404434

ABSTRACT

Antiviral drugs have traditionally been developed by directly targeting essential viral components. However, this strategy often fails due to the rapid generation of drug-resistant viruses. Recent genome-wide approaches, such as those employing small interfering RNA (siRNA) or clustered regularly interspaced short palindromic repeats (CRISPR) or those using small molecule chemical inhibitors targeting the cellular "kinome," have been used successfully to identify cellular factors that can support virus replication. Since some of these cellular factors are critical for virus replication, but are dispensable for the host, they can serve as novel targets for antiviral drug development. In addition, potentiation of immune responses, regulation of cytokine storms, and modulation of epigenetic changes upon virus infections are also feasible approaches to control infections. Because it is less likely that viruses will mutate to replace missing cellular functions, the chance of generating drug-resistant mutants with host-targeted inhibitor approaches is minimized. However, drug resistance against some host-directed agents can, in fact, occur under certain circumstances, such as long-term selection pressure of a host-directed antiviral agent that can allow the virus the opportunity to adapt to use an alternate host factor or to alter its affinity toward the target that confers resistance. This review describes novel approaches for antiviral drug development with a focus on host-directed therapies and the potential mechanisms that may account for the acquisition of antiviral drug resistance against host-directed agents.


Subject(s)
CRISPR-Cas Systems , Drug Development , Host-Derived Cellular Factors/antagonists & inhibitors , RNA, Small Interfering , Virus Replication/genetics , Animals , Gene Targeting , Host-Derived Cellular Factors/genetics , Host-Pathogen Interactions/genetics , Humans , Mice , Viruses/genetics
3.
mBio ; 10(5)2019 10 08.
Article in English | MEDLINE | ID: mdl-31594818

ABSTRACT

A genome-scale CRISPR knockout library screen of THP-1 human macrophages was performed to identify loss-of-function mutations conferring resistance to Salmonella uptake. The screen identified 183 candidate genes, from which 14 representative genes involved in actin dynamics (ACTR3, ARPC4, CAPZB, TOR3A, CYFIP2, CTTN, and NHLRC2), glycosaminoglycan metabolism (B3GNT1), receptor signaling (PDGFB and CD27), lipid raft formation (CLTCL1), calcium transport (ATP2A2 and ITPR3), and cholesterol metabolism (HMGCR) were analyzed further. For some of these pathways, known chemical inhibitors could replicate the Salmonella resistance phenotype, indicating their potential as targets for host-directed therapy. The screen indicated a role for the relatively uncharacterized gene NHLRC2 in both Salmonella invasion and macrophage differentiation. Upon differentiation, NHLRC2 mutant macrophages were hyperinflammatory and did not exhibit characteristics typical of macrophages, including atypical morphology and inability to interact and phagocytose bacteria/particles. Immunoprecipitation confirmed an interaction of NHLRC2 with FRYL, EIF2AK2, and KLHL13.IMPORTANCESalmonella exploits macrophages to gain access to the lymphatic system and bloodstream to lead to local and potentially systemic infections. With an increasing number of antibiotic-resistant isolates identified in humans, Salmonella infections have become major threats to public health. Therefore, there is an urgent need to identify alternative approaches to anti-infective therapy, including host-directed therapies. In this study, we used a simple genome-wide screen to identify 183 candidate host factors in macrophages that can confer resistance to Salmonella infection. These factors may be potential therapeutic targets against Salmonella infections.


Subject(s)
Disease Resistance , Gene Knockout Techniques , Genetic Testing , Host-Derived Cellular Factors/immunology , Macrophages/immunology , Salmonella/immunology , Endocytosis , Host-Derived Cellular Factors/genetics , Humans , Macrophages/microbiology , Models, Theoretical , Salmonella/growth & development , Salmonella Infections/immunology , THP-1 Cells
4.
Nature ; 574(7777): 259-263, 2019 10.
Article in English | MEDLINE | ID: mdl-31554973

ABSTRACT

Chikungunya virus (CHIKV) is a re-emerging alphavirus that is transmitted to humans by mosquito bites and causes musculoskeletal and joint pain1,2. Despite intensive investigations, the human cellular factors that are critical for CHIKV infection remain unknown, hampering the understanding of viral pathogenesis and the development of anti-CHIKV therapies. Here we identified the four-and-a-half LIM domain protein 1 (FHL1)3 as a host factor that is required for CHIKV permissiveness and pathogenesis in humans and mice. Ablation of FHL1 expression results in the inhibition of infection by several CHIKV strains and o'nyong-nyong virus, but not by other alphaviruses and flaviviruses. Conversely, expression of FHL1 promotes CHIKV infection in cells that do not normally express it. FHL1 interacts directly with the hypervariable domain of the nsP3 protein of CHIKV and is essential for the replication of viral RNA. FHL1 is highly expressed in CHIKV-target cells and is particularly abundant in muscles3,4. Dermal fibroblasts and muscle cells derived from patients with Emery-Dreifuss muscular dystrophy that lack functional FHL15 are resistant to CHIKV infection. Furthermore,  CHIKV infection  is undetectable in Fhl1-knockout mice. Overall, this study shows that FHL1 is a key factor expressed by the host that enables CHIKV infection and identifies the interaction between nsP3 and FHL1 as a promising target for the development of anti-CHIKV therapies.


Subject(s)
Chikungunya Fever/virology , Chikungunya virus/pathogenicity , Host-Derived Cellular Factors/metabolism , Host-Pathogen Interactions , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Muscle Proteins/metabolism , Animals , Cells, Cultured , Chikungunya Fever/drug therapy , Chikungunya virus/drug effects , Chikungunya virus/genetics , Chikungunya virus/growth & development , Female , Fibroblasts/virology , HEK293 Cells , Host-Derived Cellular Factors/genetics , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/deficiency , LIM Domain Proteins/genetics , Male , Mice , Muscle Proteins/deficiency , Muscle Proteins/genetics , Myoblasts/virology , O'nyong-nyong Virus/growth & development , O'nyong-nyong Virus/pathogenicity , Protein Binding , RNA, Viral/biosynthesis , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication
5.
Trends Microbiol ; 25(4): 243-245, 2017 04.
Article in English | MEDLINE | ID: mdl-28233621

ABSTRACT

A new study employs genome-wide loss-of-function CRISPR/Cas9 screening to identify three novel factors for HIV-1 entry. The factors represent promising targets for therapeutics as they are essential for HIV-1 infection, but dispensable for cell survival. The involved pathways were validated in primary CD4+ T cells, target cells for HIV-1.


Subject(s)
CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , HIV-1/growth & development , Host-Derived Cellular Factors/genetics , Virus Internalization , Virus Replication/genetics , CD4-Positive T-Lymphocytes/virology , HIV Infections/virology , HIV-1/genetics , Humans
6.
Nature ; 541(7637): 412-416, 2017 01 19.
Article in English | MEDLINE | ID: mdl-28077878

ABSTRACT

Picornaviruses are a leading cause of human and veterinary infections that result in various diseases, including polio and the common cold. As archetypical non-enveloped viruses, their biology has been extensively studied. Although a range of different cell-surface receptors are bound by different picornaviruses, it is unclear whether common host factors are needed for them to reach the cytoplasm. Using genome-wide haploid genetic screens, here we identify the lipid-modifying enzyme PLA2G16 (refs 8, 9, 10, 11) as a picornavirus host factor that is required for a previously unknown event in the viral life cycle. We find that PLA2G16 functions early during infection, enabling virion-mediated genome delivery into the cytoplasm, but not in any virion-assigned step, such as cell binding, endosomal trafficking or pore formation. To resolve this paradox, we screened for suppressors of the ΔPLA2G16 phenotype and identified a mechanism previously implicated in the clearance of intracellular bacteria. The sensor of this mechanism, galectin-8 (encoded by LGALS8), detects permeated endosomes and marks them for autophagic degradation, whereas PLA2G16 facilitates viral genome translocation and prevents clearance. This study uncovers two competing processes triggered by virus entry: activation of a pore-activated clearance pathway and recruitment of a phospholipase to enable genome release.


Subject(s)
Cytoplasm/virology , Genome, Viral , Host-Derived Cellular Factors/metabolism , Phospholipases A2, Calcium-Independent/metabolism , Picornaviridae/genetics , Picornaviridae/physiology , Tumor Suppressor Proteins/metabolism , Virus Internalization , Animals , Autophagy , Biological Transport , Cell Line , Cytoplasm/genetics , Endosomes/metabolism , Female , Galectins/genetics , Galectins/metabolism , Host-Derived Cellular Factors/deficiency , Host-Derived Cellular Factors/genetics , Humans , Male , Mice , Mutation , Phenotype , Phospholipases A2, Calcium-Independent/deficiency , Phospholipases A2, Calcium-Independent/genetics , Suppression, Genetic , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Virion/genetics , Virion/metabolism , Virus Replication
7.
Nature ; 535(7610): 159-63, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27383987

ABSTRACT

The Flaviviridae are a family of viruses that cause severe human diseases. For example, dengue virus (DENV) is a rapidly emerging pathogen causing an estimated 100 million symptomatic infections annually worldwide. No approved antivirals are available to date and clinical trials with a tetravalent dengue vaccine showed disappointingly low protection rates. Hepatitis C virus (HCV) also remains a major medical problem, with 160 million chronically infected patients worldwide and only expensive treatments available. Despite distinct differences in their pathogenesis and modes of transmission, the two viruses share common replication strategies. A detailed understanding of the host functions that determine viral infection is lacking. Here we use a pooled CRISPR genetic screening strategy to comprehensively dissect host factors required for these two highly important Flaviviridae members. For DENV, we identified endoplasmic-reticulum (ER)-associated multi-protein complexes involved in signal sequence recognition, N-linked glycosylation and ER-associated degradation. DENV replication was nearly completely abrogated in cells deficient in the oligosaccharyltransferase (OST) complex. Mechanistic studies pinpointed viral RNA replication and not entry or translation as the crucial step requiring the OST complex. Moreover, we show that viral non-structural proteins bind to the OST complex. The identified ER-associated protein complexes were also important for infection by other mosquito-borne flaviviruses including Zika virus, an emerging pathogen causing severe birth defects. By contrast, the most significant genes identified in the HCV screen were distinct and included viral receptors, RNA-binding proteins and enzymes involved in metabolism. We found an unexpected link between intracellular flavin adenine dinucleotide (FAD) levels and HCV replication. This study shows notable divergence in host-depenency factors between DENV and HCV, and illuminates new host targets for antiviral therapy.


Subject(s)
CRISPR-Cas Systems/genetics , Dengue Virus/physiology , Genome, Human/genetics , Hepacivirus/physiology , Host-Derived Cellular Factors/genetics , Host-Pathogen Interactions/genetics , Dengue Virus/genetics , Dengue Virus/growth & development , Drug Discovery , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum-Associated Degradation , Flavin-Adenine Dinucleotide/biosynthesis , Flavin-Adenine Dinucleotide/metabolism , Flavivirus Infections/genetics , Flavivirus Infections/virology , Glycosylation , Hexosyltransferases/deficiency , Hexosyltransferases/genetics , Hexosyltransferases/metabolism , Humans , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Targeted Therapy , Protein Binding , Protein Sorting Signals , RNA-Binding Proteins/genetics , Receptors, Virus/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication , Zika Virus/metabolism
8.
Nature ; 535(7610): 164-8, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27383988

ABSTRACT

Flaviviruses infect hundreds of millions of people annually, and no antiviral therapy is available. We performed a genome-wide CRISPR/Cas9-based screen to identify host genes that, when edited, resulted in reduced flavivirus infection. Here, we validated nine human genes required for flavivirus infectivity, and these were associated with endoplasmic reticulum functions including translocation, protein degradation, and N-linked glycosylation. In particular, a subset of endoplasmic reticulum-associated signal peptidase complex (SPCS) proteins was necessary for proper cleavage of the flavivirus structural proteins (prM and E) and secretion of viral particles. Loss of SPCS1 expression resulted in markedly reduced yield of all Flaviviridae family members tested (West Nile, Dengue, Zika, yellow fever, Japanese encephalitis, and hepatitis C viruses), but had little impact on alphavirus, bunyavirus, or rhabdovirus infection or the surface expression or secretion of diverse host proteins. We found that SPCS1 dependence could be bypassed by replacing the native prM protein leader sequences with a class I major histocompatibility complex (MHC) antigen leader sequence. Thus, SPCS1, either directly or indirectly via its interactions with unknown host proteins, preferentially promotes the processing of specific protein cargo, and Flaviviridae have a unique dependence on this signal peptide processing pathway. SPCS1 and other signal processing pathway members could represent pharmacological targets for inhibiting infection by the expanding number of flaviviruses of medical concern.


Subject(s)
CRISPR-Cas Systems/genetics , Flavivirus/physiology , Genome, Human/genetics , Host-Derived Cellular Factors/genetics , Protein Sorting Signals/physiology , Animals , Cell Line , Drosophila/cytology , Drosophila/genetics , Drosophila/virology , Drug Discovery , Endoplasmic Reticulum/metabolism , Female , Flavivirus/metabolism , Flavivirus Infections/genetics , Flavivirus Infections/virology , Glycosylation , Host-Pathogen Interactions/genetics , Humans , Membrane Proteins/genetics , Molecular Targeted Therapy , Protein Transport/genetics , Proteolysis , Reproducibility of Results , Serine Endopeptidases/genetics , Species Specificity , Viral Proteins/chemistry , Viral Proteins/metabolism , Viral Structural Proteins/metabolism
9.
Nature ; 524(7566): 471-5, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26266980

ABSTRACT

Since its discovery in 1989, efforts to grow clinical isolates of the hepatitis C virus (HCV) in cell culture have met with limited success. Only the JFH-1 isolate has the capacity to replicate efficiently in cultured hepatoma cells without cell culture-adaptive mutations. We hypothesized that cultured cells lack one or more factors required for the replication of clinical isolates. To identify the missing factors, we transduced Huh-7.5 human hepatoma cells with a pooled lentivirus-based human complementary DNA (cDNA) library, transfected the cells with HCV subgenomic replicons lacking adaptive mutations, and selected for stable replicon colonies. This led to the identification of a single cDNA, SEC14L2, that enabled RNA replication of diverse HCV genotypes in several hepatoma cell lines. This effect was dose-dependent, and required the continuous presence of SEC14L2. Full-length HCV genomes also replicated and produced low levels of infectious virus. Remarkably, SEC14L2-expressing Huh-7.5 cells also supported HCV replication following inoculation with patient sera. Mechanistic studies suggest that SEC14L2 promotes HCV infection by enhancing vitamin E-mediated protection against lipid peroxidation. This provides a foundation for development of in vitro replication systems for all HCV isolates, creating a useful platform to dissect the mechanisms by which cell culture-adaptive mutations act.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/virology , Carrier Proteins/metabolism , Cell Culture Techniques , Genotype , Hepacivirus/growth & development , Hepacivirus/genetics , Host-Derived Cellular Factors/metabolism , Lipoproteins/metabolism , Trans-Activators/metabolism , Virus Replication , Antioxidants/metabolism , Carcinoma, Hepatocellular/genetics , Carrier Proteins/genetics , Cell Line, Tumor , Cells, Cultured , Gene Library , Genome, Viral/genetics , Hepacivirus/physiology , Host-Derived Cellular Factors/genetics , Humans , Lentivirus/genetics , Lipid Peroxidation , Lipoproteins/genetics , Mutation/genetics , RNA, Viral/biosynthesis , RNA, Viral/genetics , Replicon/genetics , Serum/virology , Trans-Activators/genetics , Transduction, Genetic , Virus Replication/genetics , Vitamin E/metabolism
10.
BMC Syst Biol ; 8: 129, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25409737

ABSTRACT

BACKGROUND: One of the central interests of Virology is the identification of host factors that contribute to virus infection. Despite tremendous efforts, the list of factors identified remains limited. With omics techniques, the focus has changed from identifying and thoroughly characterizing individual host factors to the simultaneous analysis of thousands of interactions, framing them on the context of protein-protein interaction networks and of transcriptional regulatory networks. This new perspective is allowing the identification of direct and indirect viral targets. Such information is available for several members of the Potyviridae family, one of the largest and more important families of plant viruses. RESULTS: After collecting information on virus protein-protein interactions from different potyviruses, we have processed it and used it for inferring a protein-protein interaction network. All proteins are connected into a single network component. Some proteins show a high degree and are highly connected while others are much less connected, with the network showing a significant degree of dissortativeness. We have attempted to integrate this virus protein-protein interaction network into the largest protein-protein interaction network of Arabidopsis thaliana, a susceptible laboratory host. To make the interpretation of data and results easier, we have developed a new approach for visualizing and analyzing the dynamic spread on the host network of the local perturbations induced by viral proteins. We found that local perturbations can reach the entire host protein-protein interaction network, although the efficiency of this spread depends on the particular viral proteins. By comparing the spread dynamics among viral proteins, we found that some proteins spread their effects fast and efficiently by attacking hubs in the host network while other proteins exert more local effects. CONCLUSIONS: Our findings confirm that potyvirus protein-protein interaction networks are highly connected, with some proteins playing the role of hubs. Several topological parameters depend linearly on the protein degree. Some viral proteins focus their effect in only host hubs while others diversify its effect among several proteins at the first step. Future new data will help to refine our model and to improve our predictions.


Subject(s)
Arabidopsis/metabolism , Host-Derived Cellular Factors/genetics , Models, Biological , Potyvirus/genetics , Protein Interaction Maps/physiology , Arabidopsis/virology , Host-Derived Cellular Factors/metabolism , Potyvirus/metabolism , Protein Interaction Maps/genetics
11.
Med Sci Monit ; 20: 321-8, 2014 Feb 26.
Article in English | MEDLINE | ID: mdl-24569300

ABSTRACT

BACKGROUND: Success in treating hepatitis B virus (HBV) infection with nucleoside analogues drugs is limited by the emergence of drug-resistant viral strains upon prolonged therapy. In addition to mutation patterns in the viral polymerase gene, host factors are assumed to contribute to failure of treatment in chronic HBV infections. The aim of this study was to analyze the correlation between efficacy of antiviral therapy and the prevalence of HBV pretreatment drug-resistant variants. We also analyzed the role of heterogeneity in the promoter region of the IL-10 on the HBV pol/s gene polymorphisms and efficacy of analogues-driven therapy. MATERIAL AND METHODS: HBV DNA was extracted from 54 serum samples from chronic hepatitis B (CHB) patients. Drug-resistance mutations were analyzed using MALDI-TOF mass spectrometry technology (MALDI-TOF MS) and Multi-temperature single-strand conformation polymorphism (MSSCP). IL-10 gene promoter region polymorphisms at positions -1082, -819, and -592 were determined in allele-specific PCR reactions (AS-PCR). RESULTS: Drug-resistance mutations were detected in 74% of naïve and 93% of experienced patients, but the effect of pre-existence of drug-resistant HBV variants on antiviral therapy was not statistically significant (p=0.86). The role of polymorphisms at positions -1082 (p=0.88), -819 (p=0.26), and -592 (p=0.26) of IL-10 promoter region polymorphisms was excluded from the response-predicting factors. The main host factors predicting successful response to antiviral therapy were female sex (p=0.007) and young age (p=0.013). CONCLUSIONS: The presence of drug-resistant HBV variants in baseline is not a viral predictor of good response to nucleoside/nucleotide analogues therapy. Only low HBV viral load predicted positive response to antiviral therapy. The ideal candidate for antiviral therapy is an immunocompetent, young female with low HBV viral load and elevated ALT activity.


Subject(s)
Antiviral Agents/pharmacology , Drug Resistance, Viral/genetics , Hepatitis B, Chronic/drug therapy , Host-Derived Cellular Factors/genetics , Interleukin-10/genetics , Adenine/analogs & derivatives , Adenine/pharmacology , Adult , Age Factors , Drug Therapy, Combination , Female , Hepatitis B, Chronic/virology , Humans , Lamivudine/pharmacology , Middle Aged , Mutation/genetics , Nucleosides/pharmacology , Organophosphonates/pharmacology , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Promoter Regions, Genetic/genetics , Sex Factors , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tenofovir , Viral Load
12.
Cell Death Dis ; 4: e769, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23949218

ABSTRACT

Cell death is a characteristic consequence of cellular infection by influenza virus. Mounting evidence indicates the critical involvement of host-mediated cellular death pathways in promoting efficient influenza virus replication. Furthermore, it appears that many signaling pathways, such as NF-κB, formerly suspected to solely promote cell survival, can also be manipulated to induce cell death. Current understanding of the cell death pathways involved in influenza virus-mediated cytopathology and in virus replication is limited. This study was designed to identify host genes that are required for influenza-induced cell death. The approach was to perform genome-wide lentiviral-mediated human gene silencing in A549 cells and determine which genes could be silenced to provide resistance to influenza-induced cell death. The assay proved to be highly reproducible with 138 genes being identified in independent screens. The results were independently validated using siRNA to each of these candidates. Graded protection was observed in this screen with the silencing of any of 19 genes, each providing > 85% protection. Three gene products, TNFSF13 (APRIL), TNFSF12-TNFSF13 (TWE-PRIL) and USP47, were selected because of the high levels of protection conferred by their silencing. Protein and mRNA silencing and protection from influenza-induced cell death was confirmed using multiple shRNA clones and siRNA, indicating the specificity of the effects. USP47 knockdown prevented proper viral entry into the host cell, whereas TNFSF12-13/TNFSF13 knockdown blocked a late stage in viral replication. This screening approach offers the means to identify a large number of potential candidates for the analysis of viral-induced cell death. These results may also have much broader applicability in defining regulatory mechanisms involved in cell survival.


Subject(s)
Cytoprotection/genetics , Gene Knockdown Techniques , Host-Derived Cellular Factors/genetics , Orthomyxoviridae/physiology , Cell Death/genetics , Cell Line, Tumor , Genetic Association Studies , Host-Derived Cellular Factors/metabolism , Humans , Multiprotein Complexes/metabolism , Poly(ADP-ribose) Polymerases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Reproducibility of Results , Virus Internalization , Virus Replication/genetics
13.
Methods ; 59(2): 217-24, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23036328

ABSTRACT

The use of genome wide RNA interference (RNAi) screens to investigate host-virals interactions has revealed unexpected connections that have improved our understanding of viral pathogenesis and cell biology. This work describes the use of an RNAi screening method employing an immunofluorescence image-based strategy and influenza A virus. We find this approach to be readily implemented, scalable and amenable to the direct evaluation of a variety of viral lifecycles.


Subject(s)
Genome-Wide Association Study/methods , High-Throughput Screening Assays/methods , RNA Interference , Virus Replication/genetics , HeLa Cells , Host-Derived Cellular Factors/genetics , Humans , Influenza A virus , RNA, Small Interfering/genetics , Tissue Culture Techniques
14.
Pharmacol Ther ; 137(2): 266-82, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23103333

ABSTRACT

Dengue virus (DV) is the most widespread arbovirus, being endemic in over 100 countries, and is estimated to cause 50 million infections annually. Viral factors, such as the genetic composition of the virus strain can play a role in determining the virus virulence and subsequent clinical disease severity. Virus vector competence plays an integral role in virus transmission and is a critical factor in determining the severity and impact of DV outbreaks. Host genetic variations in immune-related genes, including the human leukocyte antigen, have also been shown to correlate with clinical disease and thus may play a role in regulating disease severity. The host's immune system, however, appears to be the primary factor in DV pathogenesis with the delicate interplay of innate and acquired immunity playing a crucial role. Although current research of DV pathogenesis has been limited by the lack of an appropriate animal model, the development of DV therapeutics has been a primary focus of research groups around the world. In the past decade advances in both the development of vaccines and anti-virals have increased in dramatically. This review summarises the current understanding of viral, vector and host factors which contribute to dengue virus pathogenesis and how this knowledge is critically important in the development of pharmaceutical interventions.


Subject(s)
Dengue Virus/pathogenicity , Dengue/etiology , Genetic Predisposition to Disease , Host-Derived Cellular Factors , Insect Vectors , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Dengue/immunology , Dengue/prevention & control , Dengue/virology , Dengue Virus/genetics , Dengue Virus/metabolism , Host-Derived Cellular Factors/genetics , Host-Derived Cellular Factors/immunology , Humans , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Vaccines/immunology , Viral Vaccines/pharmacology
15.
PLoS One ; 7(6): e38097, 2012.
Article in English | MEDLINE | ID: mdl-22701604

ABSTRACT

Salmonella enterica is a bacterial pathogen of humans that can proliferate within epithelial cells as well as professional phagocytes of the immune system. While much has been learned about the microbial genes that influence the infectious process through decades of intensive research, relatively little is known about the host factors that affect infection. We performed a genome-wide siRNA screen to identify host genes that Salmonella enterica serovar Typhimurium (S. typhimurium) utilizes to facilitate growth within human epithelial cells. In this screen, with siRNAs targeting every predicted gene in the human genome, we identified 252 new human-host-susceptibility factors (HSFs) for S. typhimurium. We also identified 39 genes whose silencing results in increased intracellular growth of S. typhimurium. The HSFs identified are regulated most centrally by NFκB and associate with each other through an extremely dense network of interactions that center around a group of kinases. Most genes identified were not previously appreciated as playing roles in the intracellular lifecycle of S. enterica. Numerous HSFs identified with interesting characteristics that could play plausible roles in mediating intracellular microbial growth are discussed. Importantly, this study reveals significant overlap between the host network that supports S. typhimurium growth within human epithelial cells and the one that promotes the growth of Mycobacterium tuberculosis within human macrophages. In addition to providing much new information about the molecular mechanisms underlying S. enterica-host cell interplay, all 252 HSFs identified are candidates for new anti-microbial targets for controlling S. enterica infections, and some may provide broad-spectrum anti-microbial activity.


Subject(s)
Genome, Human/genetics , Host-Derived Cellular Factors/genetics , Host-Pathogen Interactions/genetics , Mycobacterium tuberculosis/growth & development , Salmonella Infections/prevention & control , Salmonella typhimurium/growth & development , Epithelial Cells/metabolism , Gene Regulatory Networks/genetics , Humans , Mycobacterium tuberculosis/metabolism , RNA Interference , RNA, Small Interfering/genetics , Salmonella typhimurium/metabolism
16.
Proc Natl Acad Sci U S A ; 108(47): 19036-41, 2011 Nov 22.
Article in English | MEDLINE | ID: mdl-22065774

ABSTRACT

Negative-strand (NS) RNA viruses comprise many pathogens that cause serious diseases in humans and animals. Despite their clinical importance, little is known about the host factors required for their infection. Using vesicular stomatitis virus (VSV), a prototypic NS RNA virus in the family Rhabdoviridae, we conducted a human genome-wide siRNA screen and identified 72 host genes required for viral infection. Many of these identified genes were also required for infection by two other NS RNA viruses, the lymphocytic choriomeningitis virus of the Arenaviridae family and human parainfluenza virus type 3 of the Paramyxoviridae family. Genes affecting different stages of VSV infection, such as entry/uncoating, gene expression, and assembly/release, were identified. Depletion of the proteins of the coatomer complex I or its upstream effectors ARF1 or GBF1 led to detection of reduced levels of VSV RNA. Coatomer complex I was also required for infection of lymphocytic choriomeningitis virus and human parainfluenza virus type 3. These results highlight the evolutionarily conserved requirements for gene expression of diverse families of NS RNA viruses and demonstrate the involvement of host cell secretory pathway in the process.


Subject(s)
Host-Derived Cellular Factors/genetics , Secretory Pathway/genetics , Vesicular stomatitis Indiana virus/physiology , Virus Integration/genetics , Animals , Cell Line , Dogs , Electrophoresis, Polyacrylamide Gel , Gene Expression Profiling , Humans , Immunoblotting , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/physiology , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 3, Human/physiology , RNA Interference , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Vesicular stomatitis Indiana virus/genetics
17.
Amino Acids ; 41(5): 1137-45, 2011 Nov.
Article in English | MEDLINE | ID: mdl-20016921

ABSTRACT

The viral protein integrase (IN) catalyzes the integration of the HIV-1 cDNA into the host cellular genome. We have recently demonstrated that IN is acetylated by a cellular histone acetyltransferase, p300, which modifies three lysines located in the C-terminus of the viral factor (Cereseto et al. in EMBO J 24:3070-3081, 2005). This modification enhances IN catalytic activity, as demonstrated by in vitro assays. Consistently, mutations introduced in the targeted lysines greatly decrease the efficiency of HIV-1 integration. Acetylation was proven to regulate protein functions by modulating protein-protein interactions. HIV-1 to efficiently complete its replication steps, including the integration reaction, requires interacting with numerous cellular factors. Therefore, we sought to investigate whether acetylation might modulate the interaction between IN and the cellular factors. To this aim we performed a yeast two-hybrid screening that differs from the screenings so far performed (Rain et al. in Methods 47:291-297, 2009; Studamire and Goff in Retrovirology 5:48, 2008) for using as bait IN constitutively acetylated. From this analysis we have identified thirteen cellular factors involved in transcription, chromatin remodeling, nuclear transport, RNA binding, protein synthesis regulation and microtubule organization. To validate these interactions, binding assays were performed showing that acetylation increases the affinity of IN with specific factors. Nevertheless, few two-hybrid hits bind with the same affinity the acetylated and the unmodified IN. These results further underlie the relevance of IN post-translational modification by acetylation in HIV-1 replication cycle.


Subject(s)
HIV Infections/metabolism , HIV Integrase/metabolism , HIV-1/enzymology , Host-Derived Cellular Factors/metabolism , Proteins/metabolism , Acetylation , Cell Line , HIV Infections/genetics , HIV Infections/virology , HIV Integrase/genetics , HIV-1/genetics , Host-Derived Cellular Factors/genetics , Humans , Protein Binding , Proteins/genetics , Two-Hybrid System Techniques
18.
Methods ; 53(1): 3-12, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20654720

ABSTRACT

Advances in the application of RNA interference (RNAi) have facilitated the establishment of systematic cell-based loss-of-function screening platforms. Widespread implementation of this technology has enabled genome-wide genetic analysis of a diverse array of cellular phenotypes, including the identification of host cell factors involved in viral replication. Four recent studies employed whole-genome RNAi technologies to elucidate cellular genes important for the replication of HIV-1. While these four genome-scale screens shared a common objective, they differ in their scope and experimental design. In this review we explore alternative strategies for developing RNAi screens, and discuss potential pitfalls of the technology. Important technical considerations include the choice of silencing reagents, experimental systems, assay readout and analysis methods. We focus on experimental and computational parameters that can impact the outcome of high-throughput genetic screens, and provide guidelines for the development of reliable cell-based RNAi screens.


Subject(s)
HIV Infections/metabolism , HIV-1/physiology , Host-Derived Cellular Factors/genetics , RNA Interference , Data Interpretation, Statistical , Genome-Wide Association Study , HEK293 Cells , HIV Infections/virology , HIV-1/genetics , High-Throughput Screening Assays , Host-Derived Cellular Factors/metabolism , Humans , RNA, Small Interfering/genetics , Transfection/methods , Virus Replication
19.
AIDS Res Hum Retroviruses ; 26(10): 1155-61, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20925576

ABSTRACT

Human immunodeficiency virus 1 (HIV-1) and host cell factors show important mutual interactions. We found that HIV-1 infection induced expression of a likely ortholog of mouse immediate early response erythropoietin 4 (LEREPO4) in vitro. When LEREPO4 expression was suppressed by siRNA in P4-CCR5 cells, HIV-1 replication showed significantly reduced HIV-1 transcript and p24 protein levels as measured by quantitative PCR and ELISA, respectively. The LEREPO4 knockdown also had an inhibitory effect on HIV-1-LTR-driven reporter plasmid expression of ß-galactosidase. Furthermore, the inhibitory effect of LEREPO4 silencing on HIV-1 replication was confirmed in Jurkat T cells. The up-regulation of LEREPO4 by HIV-1 and the inhibition of HIV-1 replication mediated by knockdown of LEREPO4 may point to an important functional role of LEREPO4 as a novel HIV-1 dependency factor.


Subject(s)
Gene Knockdown Techniques , HIV Infections/genetics , HIV-1/physiology , Virus Replication , HIV Infections/virology , Host-Derived Cellular Factors/genetics , Host-Derived Cellular Factors/metabolism , Host-Pathogen Interactions , Humans , Jurkat Cells , RNA, Small Interfering , Receptors, CCR5/metabolism , Up-Regulation , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL
...