Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Mol Microbiol ; 118(6): 731-743, 2022 12.
Article in English | MEDLINE | ID: mdl-36308071

ABSTRACT

Acute respiratory infection by influenza virus is a persistent and pervasive public health problem. Antiviral innate immunity initiated by type I interferon (IFN) is the first responder to pathogen invasion and provides the first line of defense. We discovered that Axin1, a scaffold protein, was reduced during influenza virus infection. We also found that overexpression of Axin1 and the chemical stabilizer of Axin1, XAV939, reduced influenza virus replication in lung epithelial cells. This effect was also observed with respiratory syncytial virus and vesicular stomatitis virus. Axin1 boosted type I IFN response to influenza virus infection and activated JNK/c-Jun and Smad3 signaling. XAV939 protected mice from influenza virus infection. Thus, our studies provide new mechanistic insights into the regulation of the type I IFN response and present a new potential therapeutic of targeting Axin1 against influenza virus infection.


Subject(s)
Axin Protein , Influenza, Human , Interferons , Animals , Humans , Mice , Axin Protein/metabolism , Epithelial Cells , Immunity, Innate , Influenza, Human/immunology , Influenza, Human/metabolism , Interferons/metabolism , Virus Replication
2.
J Virol ; 91(2)2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27847354

ABSTRACT

Neuraminidase (NA) is a sialidase expressed on the surface of influenza A viruses that releases progeny viruses from the surface of infected cells and prevents viruses becoming trapped in mucus. It is a homotetramer, with each monomer consisting of a transmembrane region, a stalk, and a globular head with sialidase activity. We recently characterized two swine viruses of the pandemic H1N1 lineage, A/swine/Virginia/1814-1/2012 (pH1N1low-1) and A/swine/Virginia/1814-2/2012 (pH1N1low-2), with almost undetectable NA enzymatic activity compared to that of the highly homologous A/swine/Pennsylvania/2436/2012 (pH1N1-1) and A/swine/Minnesota/2499/2012 (pH1N1-2) viruses. pH1N1-1 transmitted to aerosol contact ferrets, but pH1N1low-1 did not. The aim of this study was to identify the molecular determinants associated with low NA activity as potential markers of aerosol transmission. We identified the shared unique substitutions M19V, A232V, D248N, and I436V (N1 numbering) in pH1N1low-1 and pH1N1low-2. pH1N1low-1 also had the unique Y66D substitution in the stalk domain, where 66Y was highly conserved in N1 NAs. Restoration of 66Y was critical for the NA activity of pH1N1low-1 NA, although 19M or 248D in conjunction with 66Y was required to recover the level of activity to that of pH1N1 viruses. Studies of NA stability and molecular modeling revealed that 66Y likely stabilized the NA homotetramer. Therefore, 66Y in the stalk domain of N1 NA was critical for the stability of the NA tetramer and, subsequently, for NA enzymatic activity. IMPORTANCE: Neuraminidase (NA) is a sialidase that is one of the major surface glycoproteins of influenza A viruses and the target for the influenza drugs oseltamivir and zanamivir. NA is important as it releases progeny viruses from the surface of infected cells and prevents viruses becoming trapped in mucus. Mutations in the globular head domain that decrease enzymatic activity but confer resistance to NA inhibitors have been characterized; however, the importance of specific mutations in the stalk domain is unknown. We identified 66Y (N1 numbering), a highly conserved amino acid that was critical for the stability of the NA tetramer and, subsequently, for NA enzymatic activity.


Subject(s)
Amino Acids/genetics , Neuraminidase/genetics , Neuraminidase/metabolism , Protein Domains/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Amino Acid Substitution , Amino Acids/chemistry , Animals , Cell Line , Dogs , Enzyme Activation , Enzyme Stability , Humans , Influenza A Virus, H1N1 Subtype/enzymology , Influenza A Virus, H1N1 Subtype/genetics , Models, Molecular , Mutation , Mutation Rate , Neuraminidase/chemistry , Protein Conformation , Structure-Activity Relationship , Viral Proteins/chemistry , Virus Replication
3.
J Virol ; 90(23): 10612-10628, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27654298

ABSTRACT

Virus-like particles (VLPs) are attractive as a vaccine concept. For human respiratory syncytial virus (hRSV), VLP assembly is poorly understood and appears inefficient. Hence, hRSV antigens are often incorporated into foreign VLP systems to generate anti-RSV vaccine candidates. To better understand the assembly, and ultimately to enable efficient production, of authentic hRSV VLPs, we examined the associated requirements and mechanisms. In a previous analysis in HEp-2 cells, the nucleoprotein (N), phosphoprotein (P), matrix protein (M), and fusion protein (F) were required for formation of filamentous VLPs, which, similar to those of wild-type virus, were associated with the cell surface. Using fluorescence and electron microscopy combined with immunogold labeling, we examined the surfaces of transfected HEp-2 cells and further dissected the process of filamentous VLP formation. Our results show that N is not required. Coexpression of P plus M plus F, but not P plus M, M plus F, or P plus F, induced both viral protein coalescence and formation of filamentous VLPs that resembled wild-type virions. Despite suboptimal coalescence in the absence of P, the M and F proteins, when coexpressed, formed cell surface-associated filaments with abnormal morphology, appearing longer and thinner than wild-type virions. For F, only the carboxy terminus (Fstem) was required, and addition of foreign protein sequences to Fstem allowed incorporation into VLPs. Together, the data show that P, M, and the F carboxy terminus are sufficient for robust viral protein coalescence and filamentous VLP formation and suggest that M-F interaction drives viral filament formation, with P acting as a type of cofactor facilitating the process and exerting control over particle morphology. IMPORTANCE: hRSV is responsible for >100,000 deaths in children worldwide, and a vaccine is not available. Among the potential anti-hRSV approaches are virus-like particle (VLP) vaccines, which, based on resemblance to virus or viral components, can induce protective immunity. For hRSV, few reports are available concerning authentic VLP production or testing, in large part because VLP production is inefficient and the mechanisms underlying particle assembly are poorly understood. Here, we took advantage of the cell-associated nature of RSV particles and used high-resolution microscopy analyses to examine the viral proteins required for formation of wild-type-virus-resembling VLPs, the contributions of these proteins to morphology, and the domains involved in incorporation of the antigenically important viral F protein. The results provide new insights that will facilitate future production of hRSV VLPs with defined shapes and compositions and may translate into improved manufacture of live-attenuated hRSV vaccines.


Subject(s)
Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Vaccines, Virus-Like Particle/immunology , Viral Proteins/immunology , Antigens, Viral/chemistry , Antigens, Viral/genetics , Cell Line , Humans , Microscopy, Electron, Scanning , Phosphoproteins/chemistry , Phosphoproteins/genetics , Phosphoproteins/immunology , Protein Domains , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Respiratory Syncytial Virus Vaccines/chemistry , Respiratory Syncytial Virus Vaccines/genetics , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/physiology , Vaccines, Virus-Like Particle/genetics , Vaccines, Virus-Like Particle/ultrastructure , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Assembly
4.
J Virol ; 87(19): 10730-41, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23903836

ABSTRACT

The human respiratory syncytial virus (HRSV) fusion (F) protein cytoplasmic tail (CT) and matrix (M) protein are key mediators of viral assembly, but the underlying mechanisms are poorly understood. A complementation assay was developed to systematically examine the role of the F protein CT in infectious virus production. The ability of F mutants with alanine substitutions in the CT to complement an F-null virus in generating infectious progeny was quantitated by flow cytometry. Two CT regions with impact on infectious progeny production were identified: residues 557 to 566 (CT-R1) and 569 to 572 (CT-R2). Substitutions in CT-R1 decreased infectivity by 40 to 85% and increased the level of F-induced cell-cell fusion but had little impact on assembly of viral surface filaments, which are believed to be virions. Substitutions in CT-R2, as well as deletion of the entire CT, abrogated infectious progeny production and impaired viral filament formation. However, CT-R2 mutations did not block but rather delayed the formation of viral filaments, which continued to form at a low rate and contained the viral M protein and nucleoprotein (N). Microscopy analysis revealed that substitutions in CT-R2 but not CT-R1 led to accumulation of M and F proteins within and at the perimeter of viral inclusion bodies (IBs), respectively. The accumulation of M and F at IBs and coincident strong decrease in filament formation and infectivity upon CT-R2 mutations suggest that F interaction with IBs is an important step in the virion assembly process and that CT residues 569 to 572 act to facilitate release of M-ribonucleoprotein complexes from IBs.


Subject(s)
Epithelial Cells/virology , Inclusion Bodies, Viral/metabolism , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/pathogenicity , Viral Fusion Proteins/metabolism , Animals , Cell Fusion , Cells, Cultured , Chlorocebus aethiops , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Membrane Fusion , Mutation/genetics , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/metabolism , Vero Cells , Viral Fusion Proteins/genetics , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism
5.
J Virol ; 86(8): 4432-43, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22318136

ABSTRACT

An experimental system was developed to generate infectious human respiratory syncytial virus (HRSV) lacking matrix (M) protein expression (M-null virus) from cDNA. The role of the M protein in virus assembly was then examined by infecting HEp-2 and Vero cells with the M-null virus and assessing the impact on infectious virus production and viral protein trafficking. In the absence of M, the production of infectious progeny was strongly impaired. Immunofluorescence (IF) microscopy analysis using antibodies against the nucleoprotein (N), attachment protein (G), and fusion protein (F) failed to detect the characteristic virus-induced cell surface filaments, which are believed to represent infectious virions. In addition, a large proportion of the N protein was detected in viral replication factories termed inclusion bodies (IBs). High-resolution analysis of the surface of M-null virus-infected cells by field emission scanning electron microscopy (SEM) revealed the presence of large areas with densely packed, uniformly short filaments. Although unusually short, these filaments were otherwise similar to those induced by an M-containing control virus, including the presence of the viral G and F proteins. The abundance of the short, stunted filaments in the absence of M indicates that M is not required for the initial stages of filament formation but plays an important role in the maturation or elongation of these structures. In addition, the absence of mature viral filaments and the simultaneous increase in the level of the N protein within IBs suggest that the M protein is involved in the transport of viral ribonucleoprotein (RNP) complexes from cytoplasmic IBs to sites of budding.


Subject(s)
Respiratory Syncytial Virus, Human/metabolism , Viral Matrix Proteins/metabolism , Animals , Cell Line , Cell Surface Extensions/ultrastructure , Chlorocebus aethiops , Codon , Gene Expression , Gene Order , Humans , Mutation , Open Reading Frames/genetics , Protein Transport , Respiratory Syncytial Virus, Human/genetics , Viral Matrix Proteins/genetics , Virus Replication/genetics
6.
Blood Adv ; 7(18): 5608-5623, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37522715

ABSTRACT

ETS variant 6 (ETV6) encodes a transcriptional repressor expressed in hematopoietic stem and progenitor cells (HSPCs), where it is required for adult hematopoiesis. Heterozygous pathogenic germline ETV6 variants are associated with thrombocytopenia 5 (T5), a poorly understood genetic condition resulting in thrombocytopenia and predisposition to hematologic malignancies. To elucidate how germline ETV6 variants affect HSPCs and contribute to disease, we generated a mouse model harboring an Etv6R355X loss-of-function variant, equivalent to the T5-associated variant ETV6R359X. Under homeostatic conditions, all HSPC subpopulations are present in the bone marrow (BM) of Etv6R355X/+ mice; however, these animals display shifts in the proportions and/or numbers of progenitor subtypes. To examine whether the Etv6R355X/+ mutation affects HSPC function, we performed serial competitive transplantation and observed that Etv6R355X/+ lineage-sca1+cKit+ (LSK) cells exhibit impaired reconstitution, with near complete failure to repopulate irradiated recipients by the tertiary transplant. Mechanistic studies incorporating cleavage under target and release under nuclease assay, assay for transposase accessible chromatin sequencing, and high-throughput chromosome conformation capture identify ETV6 binding at inflammatory gene loci, including multiple genes within the tumor necrosis factor (TNF) signaling pathway in ETV6-sufficient mouse and human HSPCs. Furthermore, single-cell RNA sequencing of BM cells isolated after transplantation reveals upregulation of inflammatory genes in Etv6R355X/+ progenitors when compared to Etv6+/+ counterparts. Corroborating these findings, Etv6R355X/+ HSPCs produce significantly more TNF than Etv6+/+ cells post-transplantation. We conclude that ETV6 is required to repress inflammatory gene expression in HSPCs under conditions of hematopoietic stress, and this mechanism may be critical to sustain HSPC function.


Subject(s)
Hematopoietic Stem Cells , Thrombocytopenia , Animals , Humans , Mice , Bone Marrow , Bone Marrow Cells/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Thrombocytopenia/metabolism , ETS Translocation Variant 6 Protein
7.
bioRxiv ; 2023 Dec 09.
Article in English | MEDLINE | ID: mdl-38106088

ABSTRACT

Sequencing of bulk tumor populations has improved genetic classification and risk assessment of B-ALL, but does not directly examine intratumor heterogeneity or infer leukemia cellular origins. We profiled 89 B-ALL samples by single-cell RNA-seq (scRNA-seq) and compared them to a reference map of normal human B-cell development established using both functional and molecular assays. Intra-sample heterogeneity was driven by cell cycle, metabolism, differentiation, and inflammation transcriptional programs. By inference of B lineage developmental state composition, nearly all samples possessed a high abundance of pro-B cells, with variation between samples mainly driven by sub-populations. However, ZNF384- r and DUX4- r B-ALL showed composition enrichment of hematopoietic stem cells, BCR::ABL1 and KMT2A -r ALL of Early Lymphoid progenitors, MEF2D -r and TCF3::PBX1 of Pre-B cells. Enrichment of Early Lymphoid progenitors correlated with high-risk clinical features. Understanding variation in transcriptional programs and developmental states of B-ALL by scRNA-seq refines existing clinical and genomic classifications and improves prediction of treatment outcome.

8.
Front Mol Biosci ; 8: 633283, 2021.
Article in English | MEDLINE | ID: mdl-34970593

ABSTRACT

Avian influenza is a disease with every possibility to evolve as a human-to-human pandemic arising out of frequent mutations and genetic reassortment or recombination of avian influenza (AI) virus. The greatest concern is that till date, no satisfactory medicine or vaccines are available, leading to massive culling of poultry birds, causing huge economic loss and ban on export of chicken products, which emphasizes the need to develop an alternative strategy for control of AI. In the current study, we attempt to explore the molecular mechanism of innate immune potential of ducks against avian influenza. In the present study, we have characterized immune response molecules such as duck TLR3, TLR7, and RIGI that are predicted to have potent antiviral activities against the identified strain of avian influenza through in silico studies (molecular docking) followed by experimental validation with differential mRNA expression analysis. Future exploitation may include immunomodulation with the recombinant protein, and transgenic or gene-edited chicken resistant to bird flu.

9.
Physiol Genomics ; 37(3): 268-78, 2009 May 13.
Article in English | MEDLINE | ID: mdl-19439715

ABSTRACT

MicroRNAs (miRNAs) are small endogenous RNAs and are widely regarded as one of the most important regulators of gene expression in both plants and animals. To define the roles of miRNAs in fetal lung development, we profiled the miRNA expression pattern during lung development with a miRNA microarray. We identified 21 miRNAs that showed significant changes in expression during lung development. These miRNAs were grouped into four distinct clusters based on their expression pattern. Cluster 1 contained miRNAs whose expression increased as development progressed, while clusters 2 and 3 showed the opposite trend of expression. miRNAs in cluster 4 including miRNA-127 (miR-127) had the highest expression at the late stage of fetal lung development. Quantitative real-time PCR validated the microarray results of six selected miRNAs. In situ hybridization demonstrated that miR-127 expression gradually shifted from mesenchymal cells to epithelial cells as development progressed. Overexpression of miR-127 in fetal lung organ culture significantly decreased the terminal bud count, increased terminal and internal bud sizes, and caused unevenness in bud sizes, indicating improper development. These findings suggest that miR-127 may have an important role in fetal lung development.


Subject(s)
Lung/metabolism , MicroRNAs/genetics , Animals , Animals, Newborn , Cluster Analysis , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , In Situ Hybridization , Lung/embryology , Lung/growth & development , Male , MicroRNAs/classification , Oligonucleotide Array Sequence Analysis/methods , Organ Culture Techniques , Pregnancy , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
10.
Gene Expr Patterns ; 8(6): 397-403, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18539546

ABSTRACT

Cl(-) transport is essential for lung development. Because gamma-aminobutyric acid (GABA) receptors allow the flow of negatively-charged Cl(-) ions across the cell membrane, we hypothesized that the expression of ionotropic GABA receptors are regulated in the lungs during development. We identified 17 GABA receptor subunits in the lungs by real-time PCR. These subunits were categorized into four groups: Group 1 had high mRNA expression during fetal stages and low in adults; Group 2 had steady expression to adult stages with a slight up-regulation at birth; Group 3 showed an increasing expression from fetal to adult lungs; and Group 4 displayed irregular mRNA fluctuations. The protein levels of selected subunits were also determined by Western blots and some subunits had protein levels that corresponded to mRNA levels. Further studied subunits were primarily localized in epithelial cells in the developing lung with differential mRNA expression between isolated cells and whole lung tissues. Our results add to the knowledge of GABA receptor expression in the lung during development.


Subject(s)
Lung/embryology , Lung/metabolism , Receptors, GABA/metabolism , Animals , Epithelial Cells/metabolism , Female , Gene Expression , Gene Expression Regulation, Developmental , Lung/cytology , Male , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, GABA/analysis , Receptors, GABA/genetics
11.
Physiol Genomics ; 31(3): 554-62, 2007 Nov 14.
Article in English | MEDLINE | ID: mdl-17804605

ABSTRACT

RNA interference (RNAi) has become a powerful technique for studying gene function, biological pathways, and the physiology of diseases. Typically, the RNAi response in mammalian cells is mediated by small interfering RNA (siRNA). The use of synthesized siRNA to silence gene is relatively quick and easy, but it is costly with transient effects. A short hairpin RNA (shRNA) with complementary sense and antisense sequences of a target gene separated by a loop structure results in gene silencing that is as effective as chemically synthesized siRNA with fewer limitations. However, current methods for constructing shRNA vectors require the synthesis of long oligonucleotides, which is costly and often suffers from mutation problems during synthesis. Here, we report an alternative approach to generate a shRNA expression vector with high efficacy. We utilized shorter (

Subject(s)
Genetic Vectors , MicroRNAs/genetics , RNA Interference , RNA, Small Interfering/genetics , Animals , Base Sequence , DNA Primers , Mutation
12.
Cell Host Microbe ; 19(2): 159-68, 2016 Feb 10.
Article in English | MEDLINE | ID: mdl-26867175

ABSTRACT

The interaction between respiratory pathogens and their hosts is complex and incompletely understood. This is particularly true when pathogens encounter the mucus layer covering the respiratory tract. The mucus layer provides an essential first host barrier to inhaled pathogens that can prevent pathogen invasion and subsequent infection. Respiratory mucus has numerous functions and interactions, both with the host and with pathogens. This review summarizes the current understanding of respiratory mucus and its interactions with the respiratory pathogens Pseudomonas aeruginosa, respiratory syncytial virus and influenza viruses, with particular focus on influenza virus transmissibility and host-range specificity. Based on current findings we propose that respiratory mucus represents an understudied host-restriction factor for influenza virus.


Subject(s)
Mucus/microbiology , Mucus/virology , Pseudomonas aeruginosa/physiology , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/virology , Animals , Host-Pathogen Interactions , Humans , Respiratory Tract Infections/physiopathology , Virus Physiological Phenomena
13.
Anim Reprod Sci ; 133(3-4): 159-68, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22871329

ABSTRACT

Interferon stimulated gene 15 (ISG15), one of the several proteins induced by conceptus derived Type I and/or a Type II interferon (IFN), is implicated as an important factor in determining the uterine receptivity and conceptus development. However, presence as well as specific role of the ISG15 in buffalo (Bubalus bubalis) reproduction is yet to be elucidated. In the present study, both genomic and cDNA sequences of bubaline (bu) ISG15 were cloned and investigated for its expression in different tissues of female reproductive tract of buffalo. Sequence analysis revealed 100% identity among the genomic sequences (1014 bp) of buISG15 from three different breeds of buffalo (viz., Murrah: Acc. No. DQ118137, Mehsana: Acc. No. DQ118138, and Nagpuri: Acc. No. DQ118136) and cDNAs (Acc. Nos. HM543268-HM543270). As in cattle, the buISG15 was comprised of two exons of 57 bp and 520 bp encoding a peptide of 154 amino acids. Moreover, the buISG15 cDNA sequence exhibited 98.3% and 98.5% identity with that of taurine and indicine cattle, respectively. Subsequent reverse transcription PCR analysis revealed expression of the buISG15 in the uterine endometrium, corpus luteum (CL), corpus hemorrhagicum and oviduct. Quantitative Real Time PCR (RTqPCR) analysis also confirmed the constitutive expression of the buISG15 in the uterine endometrium during different stages (i.e. estrus, diestrus and proestrus) of estrous cycle and also during early (∼d 30-40) pregnancy. Western blot analysis of the endometrial extract from both estrous cyclic as well as pregnant buffalo demonstrated the presence of only conjugated ISG15 which was >40 kDa. ISG15 mRNA and immune-reactive proteins were localized in the stromal as well as glandular epithelial cells of the uterine endometrium of estrous cyclic as well as pregnant buffalo. However, there was no significant difference in amount of ISG15 mRNA across the different reproductive phases. To conclude, this study will be helpful for the further understanding of the roles of the ISG15 in pregnancy of buffalo cows.


Subject(s)
Buffaloes/physiology , Endometrium/physiology , Transcriptome/physiology , Ubiquitins/metabolism , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , DNA, Complementary/metabolism , Female , Immunohistochemistry , In Situ Hybridization , Molecular Sequence Data , Phylogeny , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ubiquitins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL