Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Proc Natl Acad Sci U S A ; 121(21): e2401748121, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38739789

ABSTRACT

Potyviridae, the largest family of plant RNA viruses, includes many important pathogens that significantly reduce the yields of many crops worldwide. In this study, we report that the 6-kilodalton peptide 1 (6K1), one of the least characterized potyviral proteins, is an endoplasmic reticulum-localized protein. AI-assisted structure modeling and biochemical assays suggest that 6K1 forms pentamers with a central hydrophobic tunnel, can increase the cell membrane permeability of Escherichia coli and Nicotiana benthamiana, and can conduct potassium in Saccharomyces cerevisiae. An infectivity assay showed that viral proliferation is inhibited by mutations that affect 6K1 multimerization. Moreover, the 6K1 or its homologous 7K proteins from other viruses of the Potyviridae family also have the ability to increase cell membrane permeability and transmembrane potassium conductance. Taken together, these data reveal that 6K1 and its homologous 7K proteins function as viroporins in viral infected cells.


Subject(s)
Nicotiana , Nicotiana/virology , Nicotiana/metabolism , Potyviridae/genetics , Potyviridae/metabolism , Viral Proteins/metabolism , Viral Proteins/genetics , Cell Membrane Permeability , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Viroporin Proteins/metabolism , Viroporin Proteins/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Plant Viruses/genetics , Plant Viruses/physiology , Plant Diseases/virology , Potassium/metabolism
2.
mBio ; 15(1): e0303023, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38078754

ABSTRACT

IMPORTANCE: The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has tragically claimed millions of lives through coronavirus disease 2019 (COVID-19), and there remains a critical gap in our understanding of the precise molecular mechanisms responsible for the associated fatality. One key viral factor of interest is the SARS-CoV-2 ORF3a protein, which has been identified as a potent inducer of host cellular proinflammatory responses capable of triggering the catastrophic cytokine storm, a primary contributor to COVID-19-related deaths. Moreover, ORF3a, much like the spike protein, exhibits a propensity for frequent mutations, with certain variants linked to the severity of COVID-19. Our previous research unveiled two distinct types of ORF3a mutant proteins, categorized by their subcellular localizations, setting the stage for a comparative investigation into the functional and mechanistic disparities between these two types of ORF3a variants. Given the clinical significance and functional implications of the natural ORF3a mutations, the findings of this study promise to provide invaluable insights into the potential roles undertaken by these mutant ORF3a proteins in the pathogenesis of COVID-19.


Subject(s)
COVID-19 , Endoplasmic Reticulum , SARS-CoV-2 , Viroporin Proteins , Humans , COVID-19/virology , Cytokine Release Syndrome/pathology , Cytokine Release Syndrome/virology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Endoplasmic Reticulum-Associated Degradation , Mutant Proteins , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Viroporin Proteins/genetics , Viroporin Proteins/metabolism
3.
Curr HIV Res ; 21(2): 99-108, 2023.
Article in English | MEDLINE | ID: mdl-36809950

ABSTRACT

BACKGROUND: The genetic diversity in HIV-1 genes affects viral pathogenesis in HIV-1 positive patients. Accessory genes of HIV-1, including vpu, are reported to play a critical role in HIV pathogenesis and disease progression. Vpu has a crucial role in CD4 degradation and virus release. The sequence heterogeneity in the vpu gene may affect disease progression in patients, therefore, the current study was undertaken to identify the role of vpu in patients defined as rapid progressors. OBJECTIVE: The objective of the study was to identify the viral determinants present on vpu that may be important in disease progression in rapid progressors. METHODS: Blood samples were collected from 13 rapid progressors. DNA was isolated from PBMCs and vpu was amplified using nested PCR. Both strands of the gene were sequenced using an automated DNA Sequencer. The characterization and analysis of vpu was done using various bioinformatics tools. RESULTS: The analysis revealed that all sequences had intact ORF and sequence heterogeneity was present across all sequences and distributed all over the gene. The synonymous substitutions, however, were higher than nonsynonymous substitutions. The phylogenetic tree analysis showed an evolutionary relationship with previously published Indian subtype C sequences. Comparatively, the cytoplasmic tail(77 - 86) showed the highest degree of variability in these sequences as determined by Entropy- one tool. CONCLUSION: The study showed that due to the robust nature of the protein, the biological activity of the protein was intact and sequence heterogeneity may promote disease progression in the study population.


Subject(s)
HIV Infections , HIV Seropositivity , HIV-1 , Humans , Amino Acid Sequence , Phylogeny , Disease Progression , Genetic Variation , Human Immunodeficiency Virus Proteins/genetics , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism , Viroporin Proteins/genetics , Viroporin Proteins/metabolism
4.
Protein Sci ; 32(1): e4528, 2023 01.
Article in English | MEDLINE | ID: mdl-36468608

ABSTRACT

Detailed knowledge of a protein's key residues may assist in understanding its function and designing inhibitors against it. Consequently, such knowledge of one of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)'s proteins is advantageous since the virus is the etiological agent behind one of the biggest health crises of recent times. To that end, we constructed an exhaustive library of bacteria differing from each other by the mutated version of the virus's ORF3a viroporin they harbor. Since the protein is harmful to bacterial growth due to its channel activity, genetic selection followed by deep sequencing could readily identify mutations that abolish the protein's function. Our results have yielded numerous mutations dispersed throughout the sequence that counteract ORF3a's ability to slow bacterial growth. Comparing these data with the conservation pattern of ORF3a within the coronavirinae provided interesting insights: Deleterious mutations obtained in our study corresponded to conserved residues in the protein. However, despite the comprehensive nature of our mutagenesis coverage (108 average mutations per site), we could not reveal all of the protein's conserved residues. Therefore, it is tempting to speculate that our study unearthed positions in the protein pertinent to channel activity, while other conserved residues may correspond to different functionalities of ORF3a. In conclusion, our study provides important information on a key component of SARS-CoV-2 and establishes a procedure to analyze other viroporins comprehensively.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Amino Acid Sequence , Mutagenesis , Mutation , SARS-CoV-2/genetics , Viroporin Proteins/genetics , Open Reading Frames
5.
J Med Virol ; 94(9): 4565-4575, 2022 09.
Article in English | MEDLINE | ID: mdl-35509176

ABSTRACT

The high-risk alpha human papillomaviruses (HPVs) are responsible for 99% of cervical cancers. While the biological functions of the HPV E6 and E7 oncoproteins are well-characterized, the function of E5 has remained elusive. Here, we examined gene expression changes induced by E5 proteins from high-risk HPV-16 and low-risk HPV-6b in multiple pools of primary human keratinocytes. Surprisingly, microarray analysis revealed that over 700 genes were significantly regulated by HPV-6b E5, while only 25 genes were consistently and significantly regulated by HPV-16 E5 in three biological replicates. However, we observed that more than thousand genes were altered in individual sample compared with vector. The gene expression profile induced by 16E5 in primary genital keratinocytes was very different from what has been previously published using immortalized HaCaT cells. Genes altered by HPV-16 E5 were unaffected by HPV-6b E5. Our data demonstrate that E5 proteins from the high- and low-risk HPVs have different functions in the HPV-host cell. Interestingly, conversion of two amino acids in HPV-16 E5 to the low-risk HPV-6b sequence eliminated the induction of high-risk related cellular genes.


Subject(s)
Oncogene Proteins, Viral , Papillomavirus Infections , Viroporin Proteins , Amino Acids , Female , Human papillomavirus 16/genetics , Humans , Oncogene Proteins, Viral/genetics , Viroporin Proteins/genetics
6.
Int J Mol Sci ; 23(4)2022 Feb 09.
Article in English | MEDLINE | ID: mdl-35216056

ABSTRACT

The development of prophylactic agents against the SARS-CoV-2 virus is a public health priority in the search for new surrogate markers of active virus replication. Early detection markers are needed to follow disease progression and foresee patient negativization. Subgenomic RNA transcripts (with a focus on sgN) were evaluated in oro/nasopharyngeal swabs from COVID-19-affected patients with an analysis of 315 positive samples using qPCR technology. Cut-off Cq values for sgN (Cq < 33.15) and sgE (Cq < 34.06) showed correlations to high viral loads. The specific loss of sgN in home-isolated and hospitalized COVID-19-positive patients indicated negativization of patient condition, 3-7 days from the first swab, respectively. A new detection kit for sgN, gene E, gene ORF1ab, and gene RNAse P was developed recently. In addition, in vitro studies have shown that 2'-O-methyl antisense RNA (related to the sgN sequence) can impair SARS-CoV-2 N protein synthesis, viral replication, and syncytia formation in human cells (i.e., HEK-293T cells overexpressing ACE2) upon infection with VOC Alpha (B.1.1.7)-SARS-CoV-2 variant, defining the use that this procedure might have for future therapeutic actions against SARS-CoV-2.


Subject(s)
COVID-19/virology , Coronavirus Nucleocapsid Proteins/genetics , SARS-CoV-2/physiology , Virus Replication/physiology , Coronavirus Nucleocapsid Proteins/analysis , Giant Cells/drug effects , Giant Cells/virology , HEK293 Cells , Humans , Limit of Detection , Nasopharynx/virology , Phosphoproteins/analysis , Phosphoproteins/genetics , RNA, Antisense/pharmacology , RNA, Viral , Ribonuclease P/genetics , SARS-CoV-2/drug effects , SARS-CoV-2/genetics , Sensitivity and Specificity , Social Isolation , Viral Load , Viroporin Proteins/genetics , Virus Replication/drug effects
7.
Sci Rep ; 12(1): 1005, 2022 01 19.
Article in English | MEDLINE | ID: mdl-35046461

ABSTRACT

The pandemic of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has caused a public health emergency, and research on the development of various types of vaccines is rapidly progressing at an unprecedented development speed internationally. Some vaccines have already been approved for emergency use and are being supplied to people around the world, but there are still many ongoing efforts to create new vaccines. Virus-like particles (VLPs) enable the construction of promising platforms in the field of vaccine development. Here, we demonstrate that non-infectious SARS-CoV-2 VLPs can be successfully assembled by co-expressing three important viral proteins membrane (M), envelop (E) and nucleocapsid (N) in plants. Plant-derived VLPs were purified by sedimentation through a sucrose cushion. The shape and size of plant-derived VLPs are similar to native SARS-CoV-2 VLPs without spike. Although the assembled VLPs do not have S protein spikes, they could be developed as formulations that can improve the immunogenicity of vaccines including S antigens, and further could be used as platforms that can carry S antigens of concern for various mutations.


Subject(s)
COVID-19 Vaccines/immunology , COVID-19/immunology , Coronavirus M Proteins/immunology , Coronavirus Nucleocapsid Proteins/immunology , SARS-CoV-2/immunology , Vaccines, Virus-Like Particle/immunology , Viroporin Proteins/immunology , COVID-19/prevention & control , COVID-19/virology , COVID-19 Vaccines/administration & dosage , Coronavirus M Proteins/genetics , Coronavirus M Proteins/metabolism , Coronavirus Nucleocapsid Proteins/genetics , Coronavirus Nucleocapsid Proteins/metabolism , Humans , Nicotiana/immunology , Nicotiana/metabolism , Nicotiana/virology , Vaccines, Virus-Like Particle/genetics , Vaccines, Virus-Like Particle/metabolism , Viroporin Proteins/genetics , Viroporin Proteins/metabolism
8.
Viruses ; 14(1)2022 01 05.
Article in English | MEDLINE | ID: mdl-35062292

ABSTRACT

The NLRP3 inflammasome consists of NLRP3, ASC, and pro-caspase-1 and is an important arm of the innate immune response against influenza A virus (IAV) infection. Upon infection, the inflammasome is activated, resulting in the production of IL-1ß and IL-18, which recruits other immune cells to the site of infection. It has been suggested that in the presence of stress molecules such as nigericin, the trans-Golgi network (TGN) disperses into small puncta-like structures where NLRP3 is recruited and activated. Here, we investigated whether IAV infection could lead to TGN dispersion, whether dispersed TGN (dTGN) is responsible for NLRP3 inflammasome activation, and which viral protein is involved in this process. We showed that the IAV causes dTGN formation, which serves as one of the mechanisms of NLRP3 inflammasome activation in response to IAV infection. Furthermore, we generated a series of mutant IAVs that carry mutations in the M2 protein. We demonstrated the M2 proton channel activity, specifically His37 and Trp41 are pivotal for the dispersion of TGN, NLRP3 conformational change, and IL-1ß induction. The results revealed a novel mechanism behind the activation and regulation of the NLRP3 inflammasome in IAV infection.


Subject(s)
Inflammasomes/immunology , Influenza A Virus, H1N1 Subtype/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , trans-Golgi Network/physiology , Animals , CARD Signaling Adaptor Proteins/metabolism , Caspase 1/metabolism , Cell Line , Cells, Cultured , Dogs , Humans , Immunity, Innate , Influenza A Virus, H1N1 Subtype/immunology , Interleukin-1beta/biosynthesis , Mutation , Swine , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Viroporin Proteins/chemistry , Viroporin Proteins/genetics , Viroporin Proteins/metabolism , trans-Golgi Network/ultrastructure
9.
J Am Chem Soc ; 144(2): 769-776, 2022 01 19.
Article in English | MEDLINE | ID: mdl-34985907

ABSTRACT

The influenza A M2 channel, a prototype for viroporins, is an acid-activated viroporin that conducts protons across the viral membrane, a critical step in the viral life cycle. Four central His37 residues control channel activation by binding subsequent protons from the viral exterior, which opens the Trp41 gate and allows proton flux to the interior. Asp44 is essential for maintaining the Trp41 gate in a closed state at high pH, resulting in asymmetric conduction. The prevalent D44N mutant disrupts this gate and opens the C-terminal end of the channel, resulting in increased conduction and a loss of this asymmetric conduction. Here, we use extensive Multiscale Reactive Molecular Dynamics (MS-RMD) and quantum mechanics/molecular mechanics (QM/MM) molecular dynamics simulations with an explicit, reactive excess proton to calculate the free energy of proton transport in this M2 mutant and to study the dynamic molecular-level behavior of D44N M2. We find that this mutation significantly lowers the barrier of His37 deprotonation in the activated state and shifts the barrier for entry to the Val27 tetrad. These free energy changes are reflected in structural shifts. Additionally, we show that the increased hydration around the His37 tetrad diminishes the effect of the His37 charge on the channel's water structure, facilitating proton transport and enabling activation from the viral interior. Altogether, this work provides key insight into the fundamental characteristics of PT in WT M2 and how the D44N mutation alters this PT mechanism, and it expands understanding of the role of emergent mutations in viroporins.


Subject(s)
Influenza A virus/metabolism , Viral Matrix Proteins/metabolism , Viroporin Proteins/metabolism , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protons , Quantum Theory , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/genetics , Viroporin Proteins/chemistry , Viroporin Proteins/genetics , Water/chemistry
10.
Virology ; 568: 13-22, 2022 03.
Article in English | MEDLINE | ID: mdl-35066302

ABSTRACT

Heightened inflammatory response is a prominent feature of severe COVID-19 disease. We report that the SARS-CoV-2 ORF3a viroporin activates the NLRP3 inflammasome, the most promiscuous of known inflammasomes. Ectopically expressed ORF3a triggers IL-1ß expression via NFκB, thus priming the inflammasome. ORF3a also activates the NLRP3 inflammasome but not NLRP1 or NLRC4, resulting in maturation of IL-1ß and cleavage/activation of Gasdermin. Notably, ORF3a activates the NLRP3 inflammasome via both ASC-dependent and -independent modes. This inflammasome activation requires efflux of potassium ions and oligomerization between the kinase NEK7 and NLRP3. Importantly, infection of epithelial cells with SARS-CoV-2 similarly activates the NLRP3 inflammasome. With the NLRP3 inhibitor MCC950 and select FDA-approved oral drugs able to block ORF3a-mediated inflammasome activation, as well as key ORF3a amino acid residues needed for virus release and inflammasome activation conserved in the new variants of SARS-CoV-2 isolates across continents, ORF3a and NLRP3 present prime targets for intervention.


Subject(s)
COVID-19/metabolism , COVID-19/virology , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , SARS-CoV-2/physiology , Signal Transduction , Viroporin Proteins/genetics , Amino Acid Sequence , Antiviral Agents/pharmacology , Cell Death , Cell Line , Host-Pathogen Interactions , Humans , Models, Biological , Open Reading Frames , Potassium/metabolism , Signal Transduction/drug effects , Viroporin Proteins/chemistry , Viroporin Proteins/metabolism
11.
J Virol ; 96(6): e0192921, 2022 03 23.
Article in English | MEDLINE | ID: mdl-35080425

ABSTRACT

The HIV-1 Nef and Vpu accessory proteins are known to protect infected cells from antibody-dependent cellular cytotoxicity (ADCC) responses by limiting exposure of CD4-induced (CD4i) envelope (Env) epitopes at the cell surface. Although both proteins target the host receptor CD4 for degradation, the extent of their functional redundancy is unknown. Here, we developed an intracellular staining technique that permits the intracellular detection of both Nef and Vpu in primary CD4+ T cells by flow cytometry. Using this method, we show that the combined expression of Nef and Vpu predicts the susceptibility of HIV-1-infected primary CD4+ T cells to ADCC by HIV+ plasma. We also show that Vpu cannot compensate for the absence of Nef, thus providing an explanation for why some infectious molecular clones that carry a LucR reporter gene upstream of Nef render infected cells more susceptible to ADCC responses. Our method thus represents a new tool to dissect the biological activity of Nef and Vpu in the context of other host and viral proteins within single infected CD4+ T cells. IMPORTANCE HIV-1 Nef and Vpu exert several biological functions that are important for viral immune evasion, release, and replication. Here, we developed a new method allowing simultaneous detection of these accessory proteins in their native form together with some of their cellular substrates. This allowed us to show that Vpu cannot compensate for the lack of a functional Nef, which has implications for studies that use Nef-defective viruses to study ADCC responses.


Subject(s)
CD4-Positive T-Lymphocytes , HIV Infections , HIV-1 , Human Immunodeficiency Virus Proteins , Viral Regulatory and Accessory Proteins , Viroporin Proteins , nef Gene Products, Human Immunodeficiency Virus , Antibody-Dependent Cell Cytotoxicity/physiology , CD4 Antigens/metabolism , CD4-Positive T-Lymphocytes/virology , Flow Cytometry , HIV Infections/physiopathology , HIV-1/genetics , HIV-1/metabolism , Human Immunodeficiency Virus Proteins/genetics , Human Immunodeficiency Virus Proteins/isolation & purification , Human Immunodeficiency Virus Proteins/metabolism , Humans , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/isolation & purification , Viral Regulatory and Accessory Proteins/metabolism , Viroporin Proteins/genetics , Viroporin Proteins/isolation & purification , Viroporin Proteins/metabolism , nef Gene Products, Human Immunodeficiency Virus/genetics , nef Gene Products, Human Immunodeficiency Virus/isolation & purification , nef Gene Products, Human Immunodeficiency Virus/metabolism
12.
Arch Biochem Biophys ; 717: 109124, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35085577

ABSTRACT

The coronavirus disease 2019 (COVID-19) is caused by the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS- CoV-2) with an estimated fatality rate of less than 1%. The SARS-CoV-2 accessory proteins ORF3a, ORF6, ORF7a, ORF7b, ORF8, and ORF10 possess putative functions to manipulate host immune mechanisms. These involve interferons, which appear as a consensus function, immune signaling receptor NLRP3 (NLR family pyrin domain-containing 3) inflammasome, and inflammatory cytokines such as interleukin 1ß (IL-1ß) and are critical in COVID-19 pathology. Outspread variations of each of the six accessory proteins were observed across six continents of all complete SARS-CoV-2 proteomes based on the data reported before November 2020. A decreasing order of percentage of unique variations in the accessory proteins was determined as ORF3a > ORF8 > ORF7a > ORF6 > ORF10 > ORF7b across all continents. The highest and lowest unique variations of ORF3a were observed in South America and Oceania, respectively. These findings suggest that the wide variations in accessory proteins seem to affect the pathogenicity of SARS-CoV-2.


Subject(s)
COVID-19/virology , SARS-CoV-2/genetics , Viral Proteins/genetics , Viroporin Proteins/genetics , COVID-19/pathology , Genetic Variation , Humans , Phylogeny , SARS-CoV-2/pathogenicity
13.
J Biomol Struct Dyn ; 40(20): 10454-10469, 2022.
Article in English | MEDLINE | ID: mdl-34229570

ABSTRACT

One major obstacle in designing a successful therapeutic regimen to combat COVID-19 pandemic is the frequent occurrence of mutations in the SARS-CoV-2 resulting in patient to patient variations. Out of the four structural proteins of SARS-CoV-2 namely, spike, envelope, nucleocapsid and membrane, envelope protein governs the virus pathogenicity and induction of acute-respiratory-distress-syndrome which is the major cause of death in COVID-19 patients. These effects are facilitated by the viroporin (ion-channel) like activities of the envelope protein. Our current work reports metagenomic analysis of envelope protein at the amino acid sequence level through mining all the available SARS-CoV-2 genomes from the GISAID and coronapp servers. We found majority of mutations in envelope protein were localized at or near PDZ binding motif. Our analysis also demonstrates that the acquired mutations might have important implications on its structure and ion-channel activity. A statistical correlation between specific mutations (e.g. F4F, R69I, P71L, L73F) with patient mortalities were also observed, based on the patient data available for 18,691 SARS-CoV-2-genomes in the GISAID database till 30 April 2021. Albeit, whether these mutations exist as the cause or the effect of co-infections and/or co-morbid disorders within COVID-19 patients is still unclear. Moreover, most of the current vaccine and therapeutic interventions are revolving around spike protein. However, emphasizing on envelope protein's (1) conserved epitopes, (2) pathogenicity attenuating mutations, and (3) mutations present in the deceased patients, as reported in our present study, new directions to the ongoing efforts of therapeutic developments against COVID-19 can be achieved by targeting envelope viroporin.


Subject(s)
COVID-19 , SARS-CoV-2 , Viroporin Proteins , Humans , COVID-19/mortality , COVID-19/virology , Mutation , SARS-CoV-2/genetics , Viroporin Proteins/genetics
14.
Mol Immunol ; 142: 11-21, 2022 02.
Article in English | MEDLINE | ID: mdl-34959069

ABSTRACT

Senecavirus A (SVA) is an oncolytic virus, which can propagate in human tumor cells and has been used as an oncolytic virotherapy candidate in humans. Besides, SVA circulates in pigs and causes vesicles and coalescing erosions on the snouts and coronary bands in infected pigs and results in neonatal morbidity. SVA has evolved the ability to suppress host innate immune response to benefit viral replication. SVA 3Cpro and 2C protein inhibit the production of host type I interferon (IFN) by degradation of several components of innate immune pathway. In this study, for the first time, we determined that SVA 2B antagonized host innate immune response in both human and porcine cells. SVA 2B protein degraded mitochondrial antiviral-signaling protein (MAVS), a key host molecule in the innate immune pathway, and a colocalization and interaction between 2B and MAVS was observed in the context of viral infection. Further study showed that the 1-48 and 100-128 regions of 2B were essential for inhibition of type I IFN expression. In addition, we determined that 2B degraded MAVS depending on caspase-9 and caspase-3. In conclusion, our results revealed a novel strategy for SVA 2B protein to antagonize host innate immune response, which will help for clarification of the pathogenesis of SVA and provide an insight for oncolytic virotherapy of SVA.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Immunity, Innate/immunology , Interferon Type I/biosynthesis , Picornaviridae/metabolism , Viral Nonstructural Proteins/immunology , Animals , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Interferon Type I/immunology , Oncolytic Virotherapy/methods , Picornaviridae Infections/pathology , Picornaviridae Infections/virology , Swine , Viral Nonstructural Proteins/genetics , Viroporin Proteins/genetics , Viroporin Proteins/immunology , Virus Release/physiology , Virus Replication/physiology
15.
CRISPR J ; 4(6): 854-871, 2021 12.
Article in English | MEDLINE | ID: mdl-34847745

ABSTRACT

The lack of efficient tools to label multiple endogenous targets in cell lines without staining or fixation has limited our ability to track physiological and pathological changes in cells over time via live-cell studies. Here, we outline the FAST-HDR vector system to be used in combination with CRISPR-Cas9 to allow visual live-cell studies of up to three endogenous proteins within the same cell line. Our approach utilizes a novel set of advanced donor plasmids for homology-directed repair and a streamlined workflow optimized for microscopy-based cell screening to create genetically modified cell lines that do not require staining or fixation to accommodate microscopy-based studies. We validated this new methodology by developing two advanced cell lines with three fluorescent-labeled endogenous proteins that support high-content imaging without using antibodies or exogenous staining. We applied this technology to study seven severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2/COVID-19) viral proteins to understand better their effects on autophagy, mitochondrial dynamics, and cell growth. Using these two cell lines, we were able to identify the protein ORF3a successfully as a potent inhibitor of autophagy, inducer of mitochondrial relocalization, and a growth inhibitor, which highlights the effectiveness of live-cell studies using this technology.


Subject(s)
Autophagy , COVID-19 , CRISPR-Cas Systems , Gene Targeting , Mitochondrial Dynamics , SARS-CoV-2 , Viroporin Proteins , COVID-19/genetics , COVID-19/metabolism , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Microscopy , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Viroporin Proteins/genetics , Viroporin Proteins/metabolism
16.
PLoS Pathog ; 17(11): e1009409, 2021 11.
Article in English | MEDLINE | ID: mdl-34843601

ABSTRACT

The HIV-1 accessory protein Vpu modulates membrane protein trafficking and degradation to provide evasion of immune surveillance. Targets of Vpu include CD4, HLAs, and BST-2. Several cellular pathways co-opted by Vpu have been identified, but the picture of Vpu's itinerary and activities within membrane systems remains incomplete. Here, we used fusion proteins of Vpu and the enzyme ascorbate peroxidase (APEX2) to compare the ultrastructural locations and the proximal proteomes of wild type Vpu and Vpu-mutants. The proximity-omes of the proteins correlated with their ultrastructural locations and placed wild type Vpu near both retromer and ESCRT-0 complexes. Hierarchical clustering of protein abundances across the mutants was essential to interpreting the data and identified Vpu degradation-targets including CD4, HLA-C, and SEC12 as well as Vpu-cofactors including HGS, STAM, clathrin, and PTPN23, an ALIX-like protein. The Vpu-directed degradation of BST-2 was supported by STAM and PTPN23 and to a much lesser extent by the retromer subunits Vps35 and SNX3. PTPN23 also supported the Vpu-directed decrease in CD4 at the cell surface. These data suggest that Vpu directs targets from sorting endosomes to degradation at multi-vesicular bodies via ESCRT-0 and PTPN23.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , HIV Infections/virology , Human Immunodeficiency Virus Proteins/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Proteome/metabolism , Sorting Nexins/metabolism , Vesicular Transport Proteins/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Viroporin Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , HIV Infections/genetics , HIV Infections/metabolism , HIV-1/physiology , HeLa Cells , Human Immunodeficiency Virus Proteins/genetics , Humans , Microscopy, Electron , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Transport , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Proteome/analysis , Sorting Nexins/chemistry , Sorting Nexins/genetics , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics , Viral Regulatory and Accessory Proteins/genetics , Viroporin Proteins/genetics
17.
Appl Biochem Biotechnol ; 193(12): 4214-4227, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34632548

ABSTRACT

T7 expression system (T7 RNA polymerase / T7 promoter), derived from T7 bacteriophage, is one of the most extensively used protein expression systems, which is also an enabling tool in synthetic biology. However, in eukaryote, most of T7 expression system is transient expression system. This is mainly due to the absence of post-transcriptional processing of mRNAs transcribed by T7RNAP in eukaryotic cells, so they cannot effectively pass through nuclear membrane and enter cytoplasm. In this study, Saccharomyces cerevisiae was selected as host to construct stable T7 expression system, in which HIV-1 viroporin (Vpu) was used to improve the permeability of nuclear membrane. Results of NanoLuc® (Nluc) luciferase expression indicated that Vpu could effectively promote the transport of T7 transcripts and increase the amount of protein synthesized. The method of using viroporin to improve permeability of the nuclear membrane provides an effective tool for constructing a stable T7 expression system in eukaryote.


Subject(s)
Cell Membrane Permeability , Cell Membrane , DNA-Directed RNA Polymerases , HIV-1/genetics , Human Immunodeficiency Virus Proteins , Saccharomyces cerevisiae , Viral Proteins , Viral Regulatory and Accessory Proteins , Viroporin Proteins , Cell Membrane/genetics , Cell Membrane/metabolism , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Human Immunodeficiency Virus Proteins/biosynthesis , Human Immunodeficiency Virus Proteins/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Regulatory and Accessory Proteins/biosynthesis , Viral Regulatory and Accessory Proteins/genetics , Viroporin Proteins/biosynthesis , Viroporin Proteins/genetics
18.
Virology ; 564: 46-52, 2021 12.
Article in English | MEDLINE | ID: mdl-34653774

ABSTRACT

Antiviral short-hairpin RNAs (shRNAs) delivered by recombinant adeno-associated virus (rAAV) were investigated for their potential prophylactic and therapeutic applications related to the influenza A virus (IAV). To express shRNAs efficiently, an H1 promoter was inserted into the commercial rAAV2 system. The modified rAAV2 system could express shRNAs, and the purified rAAV was obtained at levels over 1013 viral genomes/ml and 1010 viral infection units/ml. The shNP-1496-n and shM2-925 delivered by rAAV could inhibit the replication of the H1N1 and H5N1 virus by targeting the conserved regions of the IAV nucleoprotein and matrix 2 genes in MDCK cells. The shNP-1496-n and shM2-925 expressed by rAAV could provide potent and long-term anti-H5N1 virus effects in rAAV-shRNA-enriched MDCK cells. Our findings provide a rational basis for developing RNA interference for the prevention and therapy of IAV infection.


Subject(s)
Antiviral Agents/pharmacology , Dependovirus/genetics , Influenza A virus/drug effects , RNA, Small Interfering/pharmacology , Virus Replication/drug effects , Animals , Dogs , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/drug effects , Madin Darby Canine Kidney Cells , Nucleocapsid Proteins/genetics , Promoter Regions, Genetic/genetics , RNA Interference , RNA, Small Interfering/genetics , Viral Matrix Proteins/genetics , Viroporin Proteins/genetics
19.
Front Immunol ; 12: 745625, 2021.
Article in English | MEDLINE | ID: mdl-34712234

ABSTRACT

Developing influenza vaccines that protect against a broad range of viruses is a global health priority. Several conserved viral proteins or domains have been identified as promising targets for such vaccine development. However, none of the targets is sufficiently immunogenic to elicit complete protection, and vaccine platforms that can enhance immunogenicity and deliver multiple antigens are desperately needed. Here, we report proof-of-concept studies for the development of next-generation influenza vaccines using the bacteriophage T4 virus-like particle (VLP) platform. Using the extracellular domain of influenza matrix protein 2 (M2e) as a readout, we demonstrate that up to ~1,281 M2e molecules can be assembled on a 120 x 86 nanometer phage capsid to generate M2e-T4 VLPs. These M2e-decorated nanoparticles, without any adjuvant, are highly immunogenic, stimulate robust humoral as well as cellular immune responses, and conferred complete protection against lethal influenza virus challenge. Potentially, additional conserved antigens could be incorporated into the M2e-T4 VLPs and mass-produced in E. coli in a short amount of time to deal with an emerging influenza pandemic.


Subject(s)
Bacteriophage T4/immunology , Capsid Proteins/immunology , Influenza Vaccines , Vaccine Development/methods , Viral Matrix Proteins/immunology , Viroporin Proteins/immunology , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/blood , Bronchoalveolar Lavage Fluid/immunology , Capsid Proteins/genetics , Female , Humans , Immunogenicity, Vaccine , Influenza A virus/immunology , Influenza, Human/prevention & control , Influenza, Human/virology , Mice , Mice, Inbred BALB C , Nanoparticle Drug Delivery System , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Peptide Library , Proof of Concept Study , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Viral Matrix Proteins/genetics , Viroporin Proteins/genetics
20.
Cell Rep ; 37(4): 109899, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34706226

ABSTRACT

Although commonly associated with autophagosomes, LC3 can also be recruited to membranes by covalent lipidation in a variety of non-canonical contexts. These include responses to ionophores such as the M2 proton channel of influenza A virus. We report a subtractive CRISPR screen that identifies factors required for non-canonical LC3 lipidation. As well as the enzyme complexes directly responsible for LC3 lipidation in all contexts, we show the RALGAP complex is important for M2-induced, but not ionophore drug-induced, LC3 lipidation. In contrast, ATG4D is responsible for LC3 recycling in M2-induced and basal LC3 lipidation. Identification of a vacuolar ATPase subunit in the screen suggests a common mechanism for non-canonical LC3 recruitment. Influenza-induced and ionophore drug-induced LC3 lipidation lead to association of the vacuolar ATPase and ATG16L1 and can be antagonized by Salmonella SopF. LC3 recruitment to erroneously neutral compartments may therefore represent a response to damage caused by diverse invasive pathogens.


Subject(s)
Autophagy-Related Proteins , Lipoylation , Microtubule-Associated Proteins , Autophagosomes/genetics , Autophagosomes/metabolism , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , CRISPR-Cas Systems , HCT116 Cells , HEK293 Cells , Humans , Influenza A virus/genetics , Influenza A virus/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Salmonella/genetics , Salmonella/metabolism , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Viroporin Proteins/genetics , Viroporin Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...