Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Res Sq ; 2024 Feb 02.
Article in English | MEDLINE | ID: mdl-38352529

ABSTRACT

The interaction of host and Ebola virus (EBOV) proteins is required for establishing infection of the cell. To identify protein binding partners, a proximity-dependent protein interaction screen was performed for six EBOV proteins. Hits were computationally mapped onto a human protein-protein interactome and then annotated with viral proteins to reveal known and previously undescribed EBOV-host protein interactions and processes. Importantly, this approach efficiently arranged proteins into functional complexes associated with single viral proteins. Focused characterization of interactions between EBOV VP35 and the mRNA decapping complex demonstrated that VP35 binds the scaffold protein EDC4 through the C-terminal subdomain, with each protein found associated in EBOV-infected cells. Mechanistically, depletion of three components of the complex each similarly inhibited viral replication by reducing early viral RNA synthesis. Overall, we demonstrate successful identification of EBOV protein interaction with entire cellular machines, providing a deeper understanding of replication mechanism for therapeutic intervention.

2.
J Mol Biol ; 435(20): 168241, 2023 10 15.
Article in English | MEDLINE | ID: mdl-37598728

ABSTRACT

Viral inclusion bodies (IBs) are potential sites of viral replication and assembly. How viral IBs form remains poorly defined. Here we describe a combined biophysical and cellular approach to identify the components necessary for IB formation during Ebola virus (EBOV) infection. We find that the eNP0VP35 complex containing Ebola nucleoprotein (eNP) and viral protein 35 (eVP35), the functional equivalents of nucleoprotein (N) and phosphoprotein (P) in non-segmented negative strand viruses (NNSVs), phase separates to form inclusion bodies. Phase separation of eNP0VP35 is reversible and modulated by ionic strength. The multivalency of eVP35, and not eNP, is also critical for phase separation. Furthermore, overexpression of an eVP35 peptide disrupts eNP0VP35 complex formation, leading to reduced frequency of IB formation and limited viral infection. Together, our results show that upon EBOV infection, the eNP0VP35 complex forms the minimum unit to drive IB formation and viral replication.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Inclusion Bodies , Nucleoproteins , Virus Replication , Humans , Ebolavirus/metabolism , Ebolavirus/physiology , Hemorrhagic Fever, Ebola/virology , Inclusion Bodies/virology , Nucleoproteins/metabolism , Viral Regulatory and Accessory Proteins/metabolism
6.
iScience ; 25(9): 104925, 2022 Sep 16.
Article in English | MEDLINE | ID: mdl-35992305

ABSTRACT

Pharmacologically active compounds with known biological targets were evaluated for inhibition of SARS-CoV-2 infection in cell and tissue models to help identify potent classes of active small molecules and to better understand host-virus interactions. We evaluated 6,710 clinical and preclinical compounds targeting 2,183 host proteins by immunocytofluorescence-based screening to identify SARS-CoV-2 infection inhibitors. Computationally integrating relationships between small molecule structure, dose-response antiviral activity, host target, and cell interactome produced cellular networks important for infection. This analysis revealed 389 small molecules with micromolar to low nanomolar activities, representing >12 scaffold classes and 813 host targets. Representatives were evaluated for mechanism of action in stable and primary human cell models with SARS-CoV-2 variants and MERS-CoV. One promising candidate, obatoclax, significantly reduced SARS-CoV-2 viral lung load in mice. Ultimately, this work establishes a rigorous approach for future pharmacological and computational identification of host factor dependencies and treatments for viral diseases.

7.
Proc Natl Acad Sci U S A ; 119(33): e2204706119, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35939689

ABSTRACT

Oropouche orthobunyavirus (OROV; Peribunyaviridae) is a mosquito-transmitted virus that causes widespread human febrile illness in South America, with occasional progression to neurologic effects. Host factors mediating the cellular entry of OROV are undefined. Here, we show that OROV uses the host protein low-density lipoprotein-related protein 1 (Lrp1) for efficient cellular infection. Cells from evolutionarily distinct species lacking Lrp1 were less permissive to OROV infection than cells with Lrp1. Treatment of cells with either the high-affinity Lrp1 ligand receptor-associated protein (RAP) or recombinant ectodomain truncations of Lrp1 significantly reduced OROV infection. In addition, chimeric vesicular stomatitis virus (VSV) expressing OROV glycoproteins (VSV-OROV) bound to the Lrp1 ectodomain in vitro. Furthermore, we demonstrate the biological relevance of the OROV-Lrp1 interaction in a proof-of-concept mouse study in which treatment of mice with RAP at the time of infection reduced tissue viral load and promoted survival from an otherwise lethal infection. These results with OROV, along with the recent finding of Lrp1 as an entry factor for Rift Valley fever virus, highlight the broader significance of Lrp1 in cellular infection by diverse bunyaviruses. Shared strategies for entry, such as the critical function of Lrp1 defined here, provide a foundation for the development of pan-bunyaviral therapeutics.


Subject(s)
Bunyaviridae Infections , Low Density Lipoprotein Receptor-Related Protein-1 , Orthobunyavirus , Virus Internalization , Animals , Bunyaviridae Infections/metabolism , Bunyaviridae Infections/virology , Gene Knockout Techniques , Humans , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Mice , Orthobunyavirus/physiology , South America
10.
Proc Natl Acad Sci U S A ; 119(10): e2119676119, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35235462

ABSTRACT

Lymphocytic choriomeningitis virus (LCMV) is a rodent-borne zoonotic arenavirus that causes congenital abnormalities and can be fatal for transplant recipients. Using a genome-wide loss-of-function screen, we identify host factors required for LCMV entry into cells. We identify the lysosomal mucin CD164, glycosylation factors, the heparan sulfate biosynthesis machinery, and the known receptor alpha-dystroglycan (α-DG). Biochemical analysis revealed that the LCMV glycoprotein binds CD164 at acidic pH and requires a sialylated glycan at residue N104. We demonstrate that LCMV entry proceeds by the virus switching binding from heparan sulfate or α-DG at the plasma membrane to CD164 prior to membrane fusion, thus identifying additional potential targets for therapeutic intervention.


Subject(s)
Lymphocytic choriomeningitis virus/physiology , Virus Internalization , A549 Cells , CRISPR-Cas Systems , Endolyn/physiology , Gene Editing , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions , Humans , Hydrogen-Ion Concentration , Lymphocytic choriomeningitis virus/pathogenicity , Membrane Fusion , Virulence Factors
11.
bioRxiv ; 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-33907750

ABSTRACT

Identification of host factors contributing to replication of viruses and resulting disease progression remains a promising approach for development of new therapeutics. Here, we evaluated 6710 clinical and preclinical compounds targeting 2183 host proteins by immunocytofluorescence-based screening to identify SARS-CoV-2 infection inhibitors. Computationally integrating relationships between small molecule structure, dose-response antiviral activity, host target and cell interactome networking produced cellular networks important for infection. This analysis revealed 389 small molecules, >12 scaffold classes and 813 host targets with micromolar to low nanomolar activities. From these classes, representatives were extensively evaluated for mechanism of action in stable and primary human cell models, and additionally against Beta and Delta SARS-CoV-2 variants and MERS-CoV. One promising candidate, obatoclax, significantly reduced SARS-CoV-2 viral lung load in mice. Ultimately, this work establishes a rigorous approach for future pharmacological and computational identification of novel host factor dependencies and treatments for viral diseases.

12.
Cell ; 184(20): 5163-5178.e24, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34559985

ABSTRACT

Rift Valley fever virus (RVFV) is a zoonotic pathogen with pandemic potential. RVFV entry is mediated by the viral glycoprotein (Gn), but host entry factors remain poorly defined. Our genome-wide CRISPR screen identified low-density lipoprotein receptor-related protein 1 (mouse Lrp1/human LRP1), heat shock protein (Grp94), and receptor-associated protein (RAP) as critical host factors for RVFV infection. RVFV Gn directly binds to specific Lrp1 clusters and is glycosylation independent. Exogenous addition of murine RAP domain 3 (mRAPD3) and anti-Lrp1 antibodies neutralizes RVFV infection in taxonomically diverse cell lines. Mice treated with mRAPD3 and infected with pathogenic RVFV are protected from disease and death. A mutant mRAPD3 that binds Lrp1 weakly failed to protect from RVFV infection. Together, these data support Lrp1 as a host entry factor for RVFV infection and define a new target to limit RVFV infections.


Subject(s)
Host-Pathogen Interactions , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Rift Valley fever virus/physiology , Virus Internalization , Animals , Antibody Specificity/immunology , Base Sequence , Brain/pathology , Brain/virology , CRISPR-Cas Systems/genetics , Cell Membrane/metabolism , Cells, Cultured , Glycoproteins/metabolism , Glycosaminoglycans/metabolism , Glycosylation , Humans , LDL-Receptor Related Protein-Associated Protein/metabolism , Ligands , Low Density Lipoprotein Receptor-Related Protein-1/deficiency , Membrane Glycoproteins/metabolism , Mice , Protein Binding , Protein Denaturation , Rift Valley Fever/pathology , Rift Valley Fever/prevention & control , Rift Valley Fever/virology , Rift Valley fever virus/immunology
13.
mBio ; 12(4): e0046321, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34340542

ABSTRACT

Oropouche virus (OROV) infection of humans is associated with a debilitating febrile illness that can progress to meningitis or encephalitis. First isolated from a forest worker in Trinidad and Tobago in 1955, the arbovirus OROV has since been detected throughout the Amazon basin with an estimated 500,000 human infections over 60 years. Like other members of the family Peribunyaviridae, the viral genome exists as 3 single-stranded negative-sense RNA segments. The medium-sized segment encodes a viral glycoprotein complex (GPC) that is proteolytically processed into two viral envelope proteins, Gn and Gc, responsible for attachment and membrane fusion. There are no therapeutics or vaccines to combat OROV infection, and we have little understanding of protective immunity to infection. Here, we generated a replication competent chimeric vesicular stomatitis virus (VSV), in which the endogenous glycoprotein was replaced by the GPC of OROV. Serum from mice immunized by intramuscular injection with VSV-OROV specifically neutralized wild-type OROV, and using peptide arrays we mapped multiple epitopes within an N-terminal variable region of Gc recognized by the immune sera. VSV-OROV lacking this variable region of Gc was also immunogenic in mice producing neutralizing sera that recognize additional regions of Gc. Challenge of both sets of immunized mice with wild-type OROV shows that the VSV-OROV chimeras reduce wild-type viral infection and suggest that antibodies that recognize the variable N terminus of Gc afford less protection than those that target more conserved regions of Gc. IMPORTANCE Oropouche virus (OROV), an orthobunyavirus found in Central and South America, is an emerging public health challenge that causes debilitating febrile illness. OROV is transmitted by arthropods, and increasing mobilization has the potential to significantly increase the spread of OROV globally. Despite this, no therapeutics or vaccines have been developed to combat infection. Using vesicular stomatitis (VSV) as a backbone, we developed a chimeric virus bearing the OROV glycoproteins (VSV-OROV) and tested its ability to elicit a neutralizing antibody response. Our results demonstrate that VSV-OROV produces a strong neutralizing antibody response that is at least partially targeted to the N-terminal region of Gc. Importantly, vaccination with VSV-OROV reduces viral loads in mice challenged with wild-type virus. These data provide novel evidence that targeting the OROV glycoproteins may be an effective vaccination strategy to combat OROV infection.


Subject(s)
Bunyaviridae Infections/prevention & control , Genome, Viral , Orthobunyavirus/genetics , Vesiculovirus/genetics , Vesiculovirus/immunology , Viral Envelope Proteins/genetics , Animals , Antibodies, Neutralizing , Bunyaviridae Infections/immunology , Male , Mice , Mice, Inbred C57BL , Vesicular Stomatitis/virology , Virus Replication
14.
PLoS Pathog ; 14(8): e1007123, 2018 08.
Article in English | MEDLINE | ID: mdl-30080900

ABSTRACT

Endogenous retroviruses (ERVs), remnants of ancient germline infections, comprise 8% of the human genome. The most recently integrated includes human ERV-K (HERV-K) where several envelope (env) sequences remain intact. Viral pseudotypes decorated with one of those Envs are infectious. Using a recombinant vesicular stomatitis virus encoding HERV-K Env as its sole attachment and fusion protein (VSV-HERVK) we conducted a genome-wide haploid genetic screen to interrogate the host requirements for infection. This screen identified 11 genes involved in heparan sulfate biosynthesis. Genetic inhibition or chemical removal of heparan sulfate and addition of excess soluble heparan sulfate inhibit infection. Direct binding of heparin to soluble HERV-K Env and purified VSV-HERVK defines it as critical for viral attachment. Cell surface bound VSV-HERVK particles are triggered to infect on exposure to acidic pH, whereas acid pH pretreatment of virions blocks infection. Testing of additional endogenous HERV-K env sequences reveals they bind heparin and mediate acid pH triggered fusion. This work reconstructs and defines key steps in the infectious entry pathway of an extinct virus.


Subject(s)
Endogenous Retroviruses/physiology , Heparitin Sulfate/metabolism , Viral Envelope Proteins/metabolism , Viral Tropism/physiology , Virus Internalization , Humans
15.
Cell Host Microbe ; 22(5): 688-696.e5, 2017 Nov 08.
Article in English | MEDLINE | ID: mdl-29120745

ABSTRACT

Arenaviruses cause fatal hemorrhagic disease in humans. Old World arenavirus glycoproteins (GPs) mainly engage α-dystroglycan as a cell-surface receptor, while New World arenaviruses hijack transferrin receptor. However, the Lujo virus (LUJV) GP does not cluster with New or Old World arenaviruses. Using a recombinant vesicular stomatitis virus containing LUJV GP as its sole attachment and fusion protein (VSV-LUJV), we demonstrate that infection is independent of known arenavirus receptor genes. A genome-wide haploid genetic screen identified the transmembrane protein neuropilin 2 (NRP2) and tetraspanin CD63 as factors for LUJV GP-mediated infection. LUJV GP binds the N-terminal domain of NRP2, while CD63 stimulates pH-activated LUJV GP-mediated membrane fusion. Overexpression of NRP2 or its N-terminal domain enhances VSV-LUJV infection, and cells lacking NRP2 are deficient in wild-type LUJV infection. These findings uncover this distinct set of host cell entry factors in LUJV infection and are attractive focus points for therapeutic intervention.


Subject(s)
Lujo virus/physiology , Neuropilin-2/metabolism , Tetraspanin 30/metabolism , Viral Fusion Proteins/metabolism , Viral Proteins/metabolism , Virus Internalization , Carrier Proteins , Cell Line , Host-Pathogen Interactions/physiology , Human Umbilical Vein Endothelial Cells , Humans , Lujo virus/genetics , Lujo virus/pathogenicity , Protein Interaction Domains and Motifs , Receptors, Cell Surface/metabolism , Receptors, Transferrin , Viral Fusion Proteins/genetics , Viral Proteins/genetics
16.
J Virol ; 90(9): 4494-4510, 2016 May.
Article in English | MEDLINE | ID: mdl-26912630

ABSTRACT

UNLABELLED: Virus entry into cells is a multistep process that often requires the subversion of subcellular machineries. A more complete understanding of these steps is necessary to develop new antiviral strategies. While studying the potential role of the actin network and one of its master regulators, the small GTPase Cdc42, during Junin virus (JUNV) entry, we serendipitously uncovered the small molecule ZCL278, reported to inhibit Cdc42 function as an entry inhibitor for JUNV and for vesicular stomatitis virus, lymphocytic choriomeningitis virus, and dengue virus but not for the nonenveloped poliovirus. Although ZCL278 did not interfere with JUNV attachment to the cell surface or virus particle internalization into host cells, it prevented the release of JUNV ribonucleoprotein cores into the cytosol and decreased pH-mediated viral fusion with host membranes. We also identified SVG-A astroglial cell-derived cells to be highly permissive for JUNV infection and generated new cell lines expressing fluorescently tagged Rab5c or Rab7a or lacking Cdc42 using clustered regularly interspaced short palindromic repeat (CRISPR)-caspase 9 (Cas9) gene-editing strategies. Aided by these tools, we uncovered that perturbations in the actin cytoskeleton or Cdc42 activity minimally affect JUNV entry, suggesting that the inhibitory effect of ZCL278 is not mediated by ZCL278 interfering with the activity of Cdc42. Instead, ZCL278 appears to redistribute viral particles from endosomal to lysosomal compartments. ZCL278 also inhibited JUNV replication in a mouse model, and no toxicity was detected. Together, our data suggest the unexpected antiviral activity of ZCL278 and highlight its potential for use in the development of valuable new tools to study the intracellular trafficking of pathogens. IMPORTANCE: The Junin virus is responsible for outbreaks of Argentine hemorrhagic fever in South America, where 5 million people are at risk. Limited options are currently available to treat infections by Junin virus or other viruses of the Arenaviridae, making the identification of additional tools, including small-molecule inhibitors, of great importance. How Junin virus enters cells is not yet fully understood. Here we describe new cell culture models in which the cells are susceptible to Junin virus infection and to which we applied CRISPR-Cas9 genome engineering strategies to help characterize early steps during virus entry. We also uncovered ZCL278 to be a new antiviral small molecule that potently inhibits the cellular entry of the Junin virus and other enveloped viruses. Moreover, we show that ZCL278 also functions in vivo, thereby preventing Junin virus replication in a mouse model, opening the possibility for the discovery of ZCL278 derivatives of therapeutic potential.


Subject(s)
Antiviral Agents/pharmacology , Benzamides/pharmacology , Drug Discovery , Thiourea/analogs & derivatives , Virus Internalization/drug effects , Actins/metabolism , Animals , Cell Line , Cells, Cultured , Clathrin/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Endocytosis/drug effects , Endosomes/drug effects , Endosomes/virology , Gene Knockout Techniques , Hemorrhagic Fever, American/genetics , Hemorrhagic Fever, American/metabolism , Hemorrhagic Fever, American/virology , Humans , Junin virus/drug effects , Junin virus/physiology , Mice , Protein Binding , Protein Transport , Proteolysis , Ribonucleoproteins/metabolism , Thiourea/pharmacology , Viral Load , Viral Proteins/metabolism , Virus Attachment/drug effects , Virus Replication/drug effects , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism
17.
Nucleic Acids Res ; 43(1): 504-19, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25477387

ABSTRACT

Pre-mRNA processing is mechanistically linked to transcription with RNA pol II serving as a platform to recruit RNA processing factors to nascent transcripts. The TREX complex member, REF/Aly, has been suggested to play roles in transcription and nuclear RNA stability in addition to its more broadly characterized role in mRNA export. We employed RNA-seq to identify a subset of transcripts with decreased expression in both nuclear and cytoplasmic fractions upon REF/Aly knockdown, which implies that REF/Aly affects their expression upstream of its role in mRNA export. Transcription inhibition experiments and metabolic labeling assays argue that REF/Aly does not affect stability of selected candidate transcripts. Instead, ChIP assays and nuclear run-on analysis reveal that REF/Aly depletion diminishes the transcription of these candidate genes. Furthermore, we determined that REF/Aly binds directly to candidate transcripts, supporting a direct effect of REF/Aly on candidate gene transcription. Taken together, our data suggest that the importance of REF/Aly is not limited to RNA export, but that REF/Aly is also critical for gene expression at the level of transcription. Our data are consistent with the model that REF/Aly is involved in linking splicing with transcription efficiency.


Subject(s)
Nuclear Proteins/metabolism , RNA Polymerase II/metabolism , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Cell Line , Down-Regulation , Gene Knockdown Techniques , Humans , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , RNA Stability , RNA, Messenger/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
18.
Anal Chem ; 86(20): 10397-405, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25286390

ABSTRACT

This work describes a new specific, sensitive, and rapid stable isotope dilution method for the simultaneous detection of the organophosphorus nerve agents (OPNAs) tabun (GA), sarin (GB), soman (GD), cyclosarin (GF), VR, VX, and VM adducts to tyrosine (Tyr). Serum, plasma, and lysed whole blood samples (50 µL) were prepared by protein precipitation followed by digestion with Pronase. Specific Tyr adducts were isolated from the digest by a single solid phase extraction (SPE) step, and the analytes were separated by reversed-phase ultra high performance liquid chromatography (UHPLC) gradient elution in less than 2 min. Detection was performed on a triple quadrupole tandem mass spectrometer using time-triggered selected reaction monitoring (SRM) in positive electrospray ionization (ESI) mode. The calibration range was characterized from 0.100-50.0 ng/mL for GB- and VR-Tyr and 0.250-50.0 ng/mL for GA-, GD-, GF-, and VX/VM-Tyr (R(2) ≥ 0.995). Inter- and intra-assay precision had coefficients of variation of ≤17 and ≤10%, respectively, and the measured concentration accuracies of spiked samples were within 15% of the targeted value for multiple spiking levels. The limit of detection was calculated to be 0.097, 0.027, 0.018, 0.074, 0.023, and 0.083 ng/mL for GA-, GB-, GD-, GF-, VR-, and VX/VM-Tyr, respectively. A convenience set of 96 serum samples with no known nerve agent exposure was screened and revealed no baseline values or potential interferences. This method provides a simple and highly specific diagnostic tool that may extend the time postevent that a confirmation of nerve agent exposure can be made with confidence.


Subject(s)
Blood Chemical Analysis/methods , Chemical Warfare Agents/analysis , Chromatography, High Pressure Liquid , Spectrometry, Mass, Electrospray Ionization , Blood Chemical Analysis/instrumentation , Humans , Organophosphorus Compounds/blood , Organophosphorus Compounds/chemistry , Organothiophosphorus Compounds/blood , Reproducibility of Results , Sarin/blood , Sarin/chemistry , Soman/blood , Soman/chemistry , Time Factors , Tyrosine/blood , Tyrosine/chemistry
19.
Science ; 344(6191): 1506-10, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24970085

ABSTRACT

Lassa virus spreads from a rodent to humans and can lead to lethal hemorrhagic fever. Despite its broad tropism, chicken cells were reported 30 years ago to resist infection. We found that Lassa virus readily engaged its cell-surface receptor α-dystroglycan in avian cells, but virus entry in susceptible species involved a pH-dependent switch to an intracellular receptor, the lysosome-resident protein LAMP1. Iterative haploid screens revealed that the sialyltransferase ST3GAL4 was required for the interaction of the virus glycoprotein with LAMP1. A single glycosylated residue in LAMP1, present in susceptible species but absent in birds, was essential for interaction with the Lassa virus envelope protein and subsequent infection. The resistance of Lamp1-deficient mice to Lassa virus highlights the relevance of this receptor switch in vivo.


Subject(s)
Lassa virus/physiology , Lysosomal-Associated Membrane Protein 1/metabolism , Receptors, Virus/metabolism , Viral Envelope Proteins/metabolism , Virus Internalization , Amino Acid Sequence , Animals , Cell Line , Cell Membrane/metabolism , Cell Membrane/virology , Cells, Cultured , Chickens , Dystroglycans/genetics , Dystroglycans/metabolism , Glycosylation , Humans , Hydrogen-Ion Concentration , Lassa Fever/virology , Lysosomal-Associated Membrane Protein 1/chemistry , Lysosomes/metabolism , Lysosomes/virology , Mice , Mice, Knockout , Molecular Sequence Data , Protein Binding , Sialyltransferases/metabolism , beta-Galactoside alpha-2,3-Sialyltransferase
20.
Methods Enzymol ; 539: 67-80, 2014.
Article in English | MEDLINE | ID: mdl-24581439

ABSTRACT

RNA-protein complexes are critical for almost all aspects of gene expression. Analysis of RNA-protein interactions can be complicated by the disruption of native complexes and the formation of new, reassorted complexes upon cell lysis. Before concluding that a specific RNA and protein interact in vivo, cell-mixing experiments can be performed to ensure that observed RNA-protein complexes are not formed after lysis of cells.


Subject(s)
RNA/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Animals , Blotting, Northern , Blotting, Western , Cell Extracts/chemistry , Cell Fractionation , Cell-Free System , Cells, Cultured , Humans , Immunoprecipitation , RNA/chemistry , Ribonucleoproteins, Small Nuclear/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...