Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
PLoS Pathog ; 18(9): e1010874, 2022 09.
Article in English | MEDLINE | ID: mdl-36121891

ABSTRACT

Influenza A virus (IAV) infection causes acute respiratory disease with potential severe and deadly complications. Viral pathogenesis is not only due to the direct cytopathic effect of viral infections but also to the exacerbated host inflammatory responses. Influenza viral infection can activate various host signaling pathways that function to activate or inhibit viral replication. Our previous studies have shown that a receptor tyrosine kinase TrkA plays an important role in the replication of influenza viruses in vitro, but its biological roles and functional mechanisms in influenza viral infection have not been characterized. Here we show that IAV infection strongly activates TrkA in vitro and in vivo. Using a chemical-genetic approach to specifically control TrkA kinase activity through a small molecule compound 1NMPP1 in a TrkA knock-in (TrkA KI) mouse model, we show that 1NMPP1-mediated TrkA inhibition completely protected mice from a lethal IAV infection by significantly reducing viral loads and lung inflammation. Using primary lung cells isolated from the TrkA KI mice, we show that specific TrkA inhibition reduced IAV viral RNA synthesis in airway epithelial cells (AECs) but not in alveolar macrophages (AMs). Transcriptomic analysis confirmed the cell-type-specific role of TrkA in viral RNA synthesis, and identified distinct gene expression patterns under the TrkA regulation in IAV-infected AECs and AMs. Among the TrkA-activated targets are various proinflammatory cytokines and chemokines such as IL6, IL-1ß, IFNs, CCL-5, and CXCL9, supporting the role of TrkA in mediating lung inflammation. Indeed, while TrkA inhibitor 1NMPP1 administered after the peak of IAV replication had no effect on viral load, it was able to decrease lung inflammation and provided partial protection in mice. Taken together, our results have demonstrated for the first time an important biological role of TrkA signaling in IAV infection, identified its cell-type-specific contribution to viral replication, and revealed its functional mechanism in virus-induced lung inflammation. This study suggests TrkA as a novel host target for therapeutic development against influenza viral disease.


Subject(s)
Influenza A virus , Influenza, Human , Orthomyxoviridae Infections , Pneumonia , Animals , Cytokines/metabolism , Humans , Influenza A virus/genetics , Interleukin-6/metabolism , Lung/pathology , Mice , Protein-Tyrosine Kinases/metabolism , RNA, Viral/metabolism , Receptor, trkA/metabolism , Tropomyosin/metabolism , Tropomyosin/pharmacology , Virus Replication/physiology
2.
J Virol ; 96(16): e0075422, 2022 08 24.
Article in English | MEDLINE | ID: mdl-35913216

ABSTRACT

Lassa virus (LASV) is a mammarenavirus that can cause lethal Lassa fever disease with no FDA-approved vaccine and limited treatment options. Fatal LASV infections are associated with innate immune suppression. We have previously shown that the small matrix Z protein of LASV, but not of a nonpathogenic arenavirus Pichinde virus (PICV), can inhibit the cellular RIG-I-like receptors (RLRs), but its biological significance has not been evaluated in an infectious virus due to the multiple essential functions of the Z protein required for the viral life cycle. In this study, we developed a stable HeLa cell line (HeLa-iRIGN) that could be rapidly and robustly induced by doxycycline (Dox) treatment to express RIG-I N-terminal effector, with concomitant production of type I interferons (IFN-Is). We also generated recombinant tri-segmented PICVs, rP18tri-LZ, and rP18tri-PZ, which encode LASV Z and PICV Z, respectively, as an extra mScarlet fusion protein that is nonessential for the viral life cycle. Upon infection, rP18tri-LZ consistently expressed viral genes at a higher level than rP18tri-PZ. rP18tri-LZ also showed a higher level of a viral infection than rP18tri-PZ did in HeLa-iRIGN cells, especially upon Dox induction. The heterologous Z gene did not alter viral growth in Vero and A549 cells by growth curve analysis, while LASV Z strongly increased and prolonged viral gene expression, especially in IFN-competent A549 cells. Our study provides important insights into the biological role of LASV Z-mediated RIG-I inhibition and implicates LASV Z as a potential virulence factor. IMPORTANCE Lassa virus (LASV) can cause lethal hemorrhagic fever disease in humans but other arenaviruses, such as Pichinde virus (PICV), do not cause obvious disease. We have previously shown that the Z protein of LASV but not of PICV can inhibit RIG-I, a cytosolic innate immune receptor. In this study, we developed a stable HeLa cell line that can be induced to express the RIG-I N-terminal effector domain, which allows for timely control of RIG-I activation. We also generated recombinant PICVs encoding LASV Z or PICV Z as an extra gene that is nonessential for the viral life cycle. Compared to PICV Z, LASV Z could increase viral gene expression and viral infection in an infectious arenavirus system, especially when RIG-I signaling is activated. Our study presented a convenient cell system to characterize RIG-I signaling and its antagonists and revealed LASV Z as a possible virulence factor and a potential antiviral target.


Subject(s)
Lassa virus , Viral Proteins/metabolism , HeLa Cells , Humans , Lassa Fever/virology , Lassa virus/pathogenicity , Lassa virus/physiology , Pichinde virus/genetics , Virulence Factors
3.
Article in English | MEDLINE | ID: mdl-33468464

ABSTRACT

Neglected diseases caused by arenaviruses such as Lassa virus (LASV) and filoviruses like Ebola virus (EBOV) primarily afflict resource-limited countries, where antiviral drug development is often minimal. Previous studies have shown that many approved drugs developed for other clinical indications inhibit EBOV and LASV and that combinations of these drugs provide synergistic suppression of EBOV, often by blocking discrete steps in virus entry. We hypothesize that repurposing of combinations of orally administered approved drugs provides effective suppression of arenaviruses. In this report, we demonstrate that arbidol, an approved influenza antiviral previously shown to inhibit EBOV, LASV, and many other viruses, inhibits murine leukemia virus (MLV) reporter viruses pseudotyped with the fusion glycoproteins (GPs) of other arenaviruses (Junin virus [JUNV], lymphocytic choriomeningitis virus [LCMV], and Pichinde virus [PICV]). Arbidol and other approved drugs, including aripiprazole, amodiaquine, sertraline, and niclosamide, also inhibit infection of cells by infectious PICV, and arbidol, sertraline, and niclosamide inhibit infectious LASV. Combining arbidol with aripiprazole or sertraline results in the synergistic suppression of LASV and JUNV GP-bearing pseudoviruses. This proof-of-concept study shows that arenavirus infection in vitro can be synergistically inhibited by combinations of approved drugs. This approach may lead to a proactive strategy with which to prepare for and control known and new arenavirus outbreaks.


Subject(s)
Antiviral Agents/therapeutic use , Arenaviridae Infections/drug therapy , Arenavirus/drug effects , Administration, Oral , Animals , Arenaviridae Infections/virology , Cell Line , Chlorocebus aethiops , Drug Synergism , Drug Therapy, Combination/methods , HEK293 Cells , Humans , Mice , Proof of Concept Study , Vero Cells
4.
J Virol ; 93(22)2019 11 15.
Article in English | MEDLINE | ID: mdl-31462569

ABSTRACT

Several mammarenaviruses can cause deadly hemorrhagic fever infections in humans, with limited preventative and therapeutic measures available. Arenavirus cell entry is mediated by the viral glycoprotein (GP) complex, which consists of the stable signal peptide (SSP), the receptor-binding subunit GP1, and the transmembrane subunit GP2. The GP2 cytoplasmic tail (CT) is relatively conserved among arenaviruses and is known to interact with the SSP to regulate GP processing and membrane fusion, but its biological role in the context of an infectious virus has not been fully characterized. Using a Pichinde virus (PICV) GP expression vector and a PICV reverse genetics system, we systematically characterized the functional roles of 12 conserved residues within the GP2 CT in GP processing, trafficking, assembly, and fusion, as well as in viral replication. Except for P478A and K505A R508A, alanine substitutions at conserved residues abolished GP processing and membrane fusion in plasmid-transfected cells. Six invariant H and C residues and W503 are essential for viral replication, as evidenced by the fact that their mutant viruses could not be rescued. Both P480A and R482A mutant viruses were rescued, grew similarly to wild-type (WT) virus, and produced evidently processed GP1 and GP2 subunits in virus-infected cells, despite the fact that the same mutations abolished GP processing and membrane fusion in a plasmid-based protein expression system, illustrating the importance of using an infectious-virus system for analyzing viral glycoprotein function. In summary, our results demonstrate an essential biological role of the GP2 CT in arenavirus replication and suggest it as a potential novel target for developing antivirals and/or attenuated viral vaccine candidates.IMPORTANCE Several arenaviruses, such as Lassa virus (LASV), can cause severe and lethal hemorrhagic fever diseases with high mortality and morbidity, for which no FDA-approved vaccines or therapeutics are available. Viral entry is mediated by the arenavirus GP complex, which consists of the stable signal peptide (SSP), the receptor-binding subunit GP1, and the transmembrane subunit GP2. The cytoplasmic tail (CT) of GP2 is highly conserved among arenaviruses, but its functional role in viral replication is not completely understood. Using a reverse genetics system of a prototypic arenavirus, Pichinde virus (PICV), we show that the GP2 CT contains certain conserved residues that are essential for virus replication, implicating it as a potentially good target for developing antivirals and live-attenuated viral vaccines against deadly arenavirus pathogens.


Subject(s)
Glycoproteins/metabolism , Pichinde virus/genetics , Viral Envelope Proteins/genetics , A549 Cells , Amino Acid Substitution/genetics , Animals , Arenaviridae , Arenaviridae Infections/genetics , Arenaviridae Infections/metabolism , Arenavirus/genetics , Arenavirus/metabolism , Cell Line , Chlorocebus aethiops , Glycoproteins/genetics , HEK293 Cells , Humans , Membrane Fusion/genetics , Mutation/genetics , Pichinde virus/metabolism , Protein Sorting Signals/genetics , Vero Cells , Viral Envelope Proteins/metabolism , Virus Internalization , Virus Replication
5.
Cell Mol Biol (Noisy-le-grand) ; 66(5): 49-53, 2020 Jul 31.
Article in English | MEDLINE | ID: mdl-33040813

ABSTRACT

Objective of current research was to investigate the effect of sevoflurane inhalation anesthesia on hemodynamics and inflammatory response in elderly patients with lung cancer lobectomy. Methods: A total of 168 patients with lung cancer who underwent lobectomy in our hospital from January 2019 to December 2019 were selected as the study subjects. The patients were divided into an observation group and control group according to the anesthesia program. In the control group, 1 mg/kg propofol intravenous pump induced anesthesia was maintained at 6mg/kg/h. In the observation group, 8% sevoflurane was used to induce anesthesia and 2% sevoflurane was used to maintain anesthesia. Mean artery pressure (MAP), heart rate (HR) and blood oxygen saturation (SpO2) were monitored at the beginning of single-lung ventilation (t1), when single-lung ventilation was changed to double-lung ventilation (t2), and at 30 minutes after double-lung ventilation (t3), respectively. Serum levels of interleukin-6 (IL-6), tumor necrosis factor (TNF-α), and matrix metalloproteinases (MMP-9) were measured using an enzyme-linked immunosorbent assay (ELISA) kit. Assess the patient's adverse reactions. Results: At time t1 and time t2, there was no significant difference in the three hemodynamic indicators between the two groups (P>0.05). However, at t3, both MAP and HR in the observation group were significantly lower than those in the control group, while SpO2 was significantly higher than those in the control group (P<0.05). At t1 and t2, there was no significant difference in IL-6 and TNF- levels between the two groups, but at t3, IL-6 and TNF-α levels in the observation group were significantly lower than those in the control group (P<0.05). Compared with the control group, serum MMP-9 level was significantly decreased in the whole t1 to t3 stage (P<0.05). The incidence of complications in the observation group was significantly higher than that in the control group. It was calculated that Sevoflurane can significantly improve hemodynamics and inflammatory response in elderly patients with lung cancer lobectomy, but the incidence of complications is high.


Subject(s)
Hemodynamics/drug effects , Interleukin-6/metabolism , Lung Neoplasms/metabolism , Matrix Metalloproteinase 9/metabolism , Sevoflurane/therapeutic use , Tumor Necrosis Factor-alpha/metabolism , Aged , Anesthesia, Inhalation/methods , Blood Pressure/drug effects , Female , Heart Rate/drug effects , Humans , Inflammation/metabolism , Lung/metabolism , Lung/surgery , Lung Neoplasms/surgery , Male , Middle Aged
6.
J Virol ; 92(13)2018 07 01.
Article in English | MEDLINE | ID: mdl-29669840

ABSTRACT

RIG-I is a major cytoplasmic sensor of viral pathogen-associated molecular pattern (PAMP) RNA and induces type I interferon (IFN) production upon viral infection. A double-stranded RNA (dsRNA)-binding protein, PACT, plays an important role in potentiating RIG-I function. We have shown previously that arenaviral nucleoproteins (NPs) suppress type I IFN production via their RNase activity to degrade PAMP RNA. We report here that NPs of arenaviruses block the PACT-induced enhancement of RIG-I function to mediate type I IFN production and that this inhibition is dependent on the RNase function of NPs, which is different from that of a known mechanism of other viral proteins to abolish the interaction between PACT and RIG-I. To understand the biological roles of PACT and RIG-I in authentic arenavirus infection, we analyze growth kinetics of recombinant Pichinde virus (PICV), a prototypical arenavirus, in RIG-I knockout (KO) and PACT KO mouse embryonic fibroblast (MEF) cells. Wild-type (WT) PICV grew at higher titers in both KO MEF lines than in normal MEFs, suggesting the important roles of these cellular proteins in restricting virus replication. PICV carrying the NP RNase catalytically inactive mutation could not grow in normal MEFs but could replicate to some extent in both KO MEF lines. The level of virus growth was inversely correlated with the amount of type I IFNs produced. These results suggest that PACT plays an important role in potentiating RIG-I function to produce type I IFNs in order to restrict arenavirus replication and that viral NP RNase activity is essential for optimal viral replication by suppressing PACT-induced RIG-I activation.IMPORTANCE We report here a new role of the nucleoproteins of arenaviruses that can block type I IFN production via their specific inhibition of the cellular protein sensors of virus infection (RIG-I and PACT). Our results suggest that PACT plays an important role in potentiating RIG-I function to produce type I IFNs in order to restrict arenavirus replication. This new knowledge can be exploited for the development of novel antiviral treatments and/or vaccines against some arenaviruses that can cause severe and lethal hemorrhagic fever diseases in humans.


Subject(s)
Arenavirus/pathogenicity , Host-Pathogen Interactions , Interferon Type I/metabolism , Nucleoproteins/metabolism , RNA-Binding Proteins/metabolism , Receptors, Retinoic Acid/metabolism , Viral Proteins/metabolism , Arenaviridae Infections/genetics , Arenaviridae Infections/metabolism , Arenaviridae Infections/virology , HEK293 Cells , Humans , Nucleoproteins/genetics , Pichinde virus/physiology , RNA-Binding Proteins/genetics , Receptors, Retinoic Acid/genetics , Viral Proteins/genetics , Virus Replication
8.
J Virol ; 90(5): 2551-60, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26676795

ABSTRACT

UNLABELLED: Pichinde virus (PICV) is a bisegmented enveloped RNA virus that targets macrophages and dendritic cells (DCs) early in infection and induces strong innate and adaptive immunity in mice. We have developed a reverse genetics system to produce live recombinant PICV (strain P18) with a trisegmented RNA genome (rP18tri), which encodes all four PICV gene products and as many as two foreign genes. We have engineered the vector to express the green fluorescent protein (GFP) reporter gene (abbreviated as G in virus designations) and either the hemagglutination (HA [H]) or the nucleoprotein (NP [P]) gene of the influenza A/PR8 virus. The trisegmented viruses rP18tri-G/H and rP18tri-G/P showed slightly reduced growth in vitro and expressed HA and NP, respectively. Mice immunized with rP18tri-G/H were completely protected against lethal influenza virus challenge even 120 days after immunization. These rP18tri-based vectors could efficiently induce both neutralizing antibodies and antigen-specific T cell responses via different immunization routes. Interestingly, the immune responses were significantly increased upon a booster dose and remained at high levels even after three booster doses. In summary, we have developed a novel PICV-based live vaccine vector that can express foreign antigens to induce strong humoral and cell-mediated immunity and is ideal for a prime-and-boost vaccination strategy. IMPORTANCE: We have developed a novel Pichinde virus (PICV)-based live viral vector, rP18tri, that packages three RNA segments and encodes as many as two foreign genes. Using the influenza virus HA and NP genes as model antigens, we show that this rP18tri vector can induce strong humoral and cellular immunity via different immunization routes and can lead to protection in mice. Interestingly, a booster dose further enhances the immune responses, a feature that distinguishes this from other known live viral vectors. In summary, our study demonstrates a unique feature of this live rP18tri vector to be used as a novel vaccine platform for a prime-and-boost vaccination strategy.


Subject(s)
Drug Carriers , Genetic Vectors , Influenza Vaccines/immunology , Pichinde virus/genetics , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Disease Models, Animal , Female , Gene Expression , Genes, Reporter , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Guinea Pigs , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Male , Mice, Inbred C57BL , Nucleocapsid Proteins , Orthomyxoviridae Infections/prevention & control , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Survival Analysis , T-Lymphocytes/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Core Proteins/genetics , Viral Core Proteins/immunology
9.
J Virol ; 89(13): 6595-607, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25878103

ABSTRACT

UNLABELLED: Arenaviruses cause severe hemorrhagic fever diseases in humans, and there are limited preventative and therapeutic measures against these diseases. Previous structural and functional analyses of arenavirus nucleoproteins (NPs) revealed a conserved DEDDH exoribonuclease (RNase) domain that is important for type I interferon (IFN) suppression, but the biological roles of the NP RNase in viral replication and host immune suppression have not been well characterized. Infection of guinea pigs with Pichinde virus (PICV), a prototype arenavirus, can serve as a surrogate small animal model for arenavirus hemorrhagic fevers. In this report, we show that mutation of each of the five RNase catalytic residues of PICV NP diminishes the IFN suppression activity and slightly reduces the viral RNA replication activity. Recombinant PICVs with RNase catalytic mutations can induce high levels of IFNs and barely grow in IFN-competent A549 cells, in sharp contrast to the wild-type (WT) virus, while in IFN-deficient Vero cells, both WT and mutant viruses can replicate at relatively high levels. Upon infection of guinea pigs, the RNase mutant viruses stimulate strong IFN responses, fail to replicate productively, and can become WT revertants. Serial passages of the RNase mutants in vitro can also generate WT revertants. Thus, the NP RNase function is essential for the innate immune suppression that allows the establishment of a productive early viral infection, and it may be partly involved in the process of viral RNA replication. IMPORTANCE: Arenaviruses, such as Lassa, Lujo, and Machupo viruses, can cause severe and deadly hemorrhagic fever diseases in humans, and there are limited preventative and treatment options against these diseases. Development of broad-spectrum antiviral drugs depends on a better mechanistic understanding of the conserved arenavirus proteins in viral infection. The nucleoprotein (NPs) of all arenaviruses carry a unique exoribonuclease (RNase) domain that has been shown to be critical for the suppression of type I interferons. However, the functional roles of the NP RNase in arenavirus replication and host immune suppression have not been characterized systematically. Using a prototype arenavirus, Pichinde virus (PICV), we characterized the viral growth and innate immune suppression of recombinant RNase-defective mutants in both cell culture and guinea pig models. Our study suggests that the NP RNase plays an essential role in the suppression of host innate immunity, and possibly in viral RNA replication, and that it can serve as a novel target for developing antiviral drugs against arenavirus pathogens.


Subject(s)
Exoribonucleases/metabolism , Host-Pathogen Interactions , Immune Evasion , Nucleoproteins/metabolism , Pichinde virus/enzymology , Pichinde virus/physiology , Virus Replication , Amino Acid Motifs , Amino Acid Substitution , Animals , Arenaviridae Infections/immunology , Arenaviridae Infections/virology , Cell Line , DNA Mutational Analysis , Disease Models, Animal , Exoribonucleases/genetics , Guinea Pigs , Humans , Male , Nucleoproteins/genetics , Pichinde virus/genetics , Pichinde virus/immunology
10.
J Biol Chem ; 288(23): 16949-16959, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23615902

ABSTRACT

A hallmark of severe Lassa fever is the generalized immune suppression, the mechanism of which is poorly understood. Lassa virus (LASV) nucleoprotein (NP) is the only known 3'-5' exoribonuclease that can suppress type I interferon (IFN) production possibly by degrading immune-stimulatory RNAs. How this unique enzymatic activity of LASV NP recognizes and processes RNA substrates is unknown. We provide an atomic view of a catalytically active exoribonuclease domain of LASV NP (LASV NP-C) in the process of degrading a 5' triphosphate double-stranded (ds) RNA substrate, a typical pathogen-associated molecular pattern molecule, to induce type I IFN production. Additionally, we provide for the first time a high-resolution crystal structure of an active exoribonuclease domain of Tacaribe arenavirus (TCRV) NP. Coupled with the in vitro enzymatic and cell-based interferon suppression assays, these structural analyses strongly support a unified model of an exoribonuclease-dependent IFN suppression mechanism shared by all known arenaviruses. New knowledge learned from these studies should aid the development of therapeutics against pathogenic arenaviruses that can infect hundreds of thousands of individuals and kill thousands annually.


Subject(s)
Arenaviruses, New World , Exoribonucleases , Immune Tolerance , Interferon Type I , Lassa Fever , Lassa virus , Nucleoproteins , RNA, Double-Stranded , RNA, Viral , Viral Proteins , Arenaviruses, New World/enzymology , Arenaviruses, New World/genetics , Arenaviruses, New World/immunology , Cell Line , Crystallography, X-Ray , Exoribonucleases/chemistry , Exoribonucleases/genetics , Exoribonucleases/immunology , Exoribonucleases/metabolism , Humans , Interferon Type I/immunology , Interferon Type I/metabolism , Lassa Fever/genetics , Lassa Fever/immunology , Lassa Fever/metabolism , Lassa virus/enzymology , Lassa virus/genetics , Lassa virus/immunology , Nucleoproteins/chemistry , Nucleoproteins/genetics , Nucleoproteins/immunology , Nucleoproteins/metabolism , Protein Structure, Tertiary , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/genetics , RNA, Double-Stranded/immunology , RNA, Double-Stranded/metabolism , RNA, Viral/chemistry , RNA, Viral/genetics , RNA, Viral/immunology , RNA, Viral/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism
11.
PLoS Pathog ; 8(8): e1002899, 2012.
Article in English | MEDLINE | ID: mdl-22956907

ABSTRACT

Human bocavirus 1 (HBoV1) has been identified as one of the etiological agents of wheezing in young children with acute respiratory-tract infections. In this study, we have obtained the sequence of a full-length HBoV1 genome (including both termini) using viral DNA extracted from a nasopharyngeal aspirate of an infected patient, cloned the full-length HBoV1 genome, and demonstrated DNA replication, encapsidation of the ssDNA genome, and release of the HBoV1 virions from human embryonic kidney 293 cells. The HBoV1 virions generated from this cell line-based production system exhibits a typical icosahedral structure of approximately 26 nm in diameter, and is capable of productively infecting polarized primary human airway epithelia (HAE) from the apical surface. Infected HAE showed hallmarks of lung airway-tract injury, including disruption of the tight junction barrier, loss of cilia and epithelial cell hypertrophy. Notably, polarized HAE cultured from an immortalized airway epithelial cell line, CuFi-8 (originally derived from a cystic fibrosis patient), also supported productive infection of HBoV1. Thus, we have established a reverse genetics system and generated the first cell line-based culture system for the study of HBoV1 infection, which will significantly advance the study of HBoV1 replication and pathogenesis.


Subject(s)
Epithelial Cells/virology , Human bocavirus/physiology , Parvoviridae Infections/virology , Respiratory Tract Infections/virology , Virus Replication , Base Sequence , Cell Line , DNA, Viral/chemistry , DNA, Viral/genetics , Epithelium/virology , Female , Genome, Viral/genetics , Human bocavirus/genetics , Human bocavirus/isolation & purification , Humans , Inverted Repeat Sequences , Molecular Sequence Data , Respiratory System/virology , Reverse Genetics , Sequence Analysis, DNA , Transfection
12.
J Virol ; 86(19): 10748-58, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22837195

ABSTRACT

Human parvovirus B19 (B19V) infection is highly restricted to human erythroid progenitor cells, in which it induces a DNA damage response (DDR). The DDR signaling is mainly mediated by the ATR (ataxia telangiectasia-mutated and Rad3-related) pathway, which promotes replication of the viral genome; however, the exact mechanisms employed by B19V to take advantage of the DDR for virus replication remain unclear. In this study, we focused on the initiators of the DDR and the role of the DDR in cell cycle arrest during B19V infection. We examined the role of individual viral proteins, which were delivered by lentiviruses, in triggering a DDR in ex vivo-expanded primary human erythroid progenitor cells and the role of DNA replication of the B19V double-stranded DNA (dsDNA) genome in a human megakaryoblastoid cell line, UT7/Epo-S1 (S1). All the cells were cultured under hypoxic conditions. The results showed that none of the viral proteins induced phosphorylation of H2AX or replication protein A32 (RPA32), both hallmarks of a DDR. However, replication of the B19V dsDNA genome was capable of inducing the DDR. Moreover, the DDR per se did not arrest the cell cycle at the G(2)/M phase in cells with replicating B19V dsDNA genomes. Instead, the B19V nonstructural 1 (NS1) protein was the key factor in disrupting the cell cycle via a putative transactivation domain operating through a p53-independent pathway. Taken together, the results suggest that the replication of the B19V genome is largely responsible for triggering a DDR, which does not perturb cell cycle progression at G(2)/M significantly, during B19V infection.


Subject(s)
DNA Damage , DNA Replication , Parvovirus B19, Human/genetics , Parvovirus B19, Human/metabolism , Virus Replication , Antigens, CD34/biosynthesis , Cell Cycle Checkpoints , Cell Division , G2 Phase , Genome, Viral , Histones/metabolism , Humans , Hypoxia , Lentivirus/genetics , Mutation , Phosphorylation , Promoter Regions, Genetic
13.
Vaccines (Basel) ; 11(12)2023 Dec 02.
Article in English | MEDLINE | ID: mdl-38140210

ABSTRACT

Pichinde virus (PICV) can infect several animal species and has been developed as a safe and effective vaccine vector. Our previous study showed that pigs vaccinated with a recombinant PICV-vectored vaccine expressing the hemagglutinin (HA) gene of an H3N2 influenza A virus of swine (IAV-S) developed virus-neutralizing antibodies and were protected against infection by the homologous H3N2 strain. The objective of the current study was to evaluate the immunogenicity and protective efficacy of a trivalent PICV-vectored vaccine expressing HA antigens from the three co-circulating IAV-S subtypes: H1N1, H1N2, and H3N2. Pigs immunized with the trivalent PICV vaccine developed virus-neutralizing (VN) and hemagglutination inhibition (HI) antibodies against all three matching IAV-S. Following challenge infection with the H1N1 strain, five of the six pigs vaccinated with the trivalent vaccine had no evidence of IAV-S RNA genomes in nasal swabs and bronchoalveolar lavage fluid, while all non-vaccinated control pigs showed high number of copies of IAV-S genomic RNA in these two types of samples. Overall, our results demonstrate that the trivalent PICV-vectored vaccine elicits antibody responses against the three targeted IAV-S strains and provides protection against homologous virus challenges in pigs. Therefore, PICV exhibits the potential to be explored as a viral vector for delivering multiple vaccine antigens in swine.

14.
Front Immunol ; 14: 1127515, 2023.
Article in English | MEDLINE | ID: mdl-36845108

ABSTRACT

Introduction: Tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb) remains a major global health threat. The only available vaccine Bacille Calmette-Guérin (BCG) does not prevent adult pulmonary TB. New effective TB vaccines should aim to stimulate robust T cell responses in the lung mucosa to achieve high protective efficacy. We have previously developed a novel viral vaccine vector based on recombinant Pichinde virus (PICV), a non-pathogenic arenavirus with low seroprevalence in humans, and have demonstrated its efficacy to induce strong vaccine immunity with undetectable anti-vector neutralization activity. Methods: Using this tri-segmented PICV vector (rP18tri), we have generated viral vectored TB vaccines (TBvac-1, TBvac-2, and TBvac-10) encoding several known TB immunogens (Ag85B, EsxH, and ESAT-6/EsxA). A P2A linker sequence was used to allow for the expression of two proteins from one open-reading-frame (ORF) on the viral RNA segments. The immunogenicity of TBvac-2 and TBvac-10 and the protective efficacy of TBvac-1 and TBvac-2 were evaluated in mice. Results: Both viral vectored vaccines elicited strong antigen-specific CD4 and CD8 T cells through intramuscular (IM) and intranasal (IN) routes as evaluated by MHC-I and MHC-II tetramer analyses, respectively. The IN inoculation route helped to elicit strong lung T cell responses. The vaccine-induced antigen-specific CD4 T cells are functional, expressing multiple cytokines as detected by intracellular cytokine staining. Finally, immunization with TBvac-1 or TBvac-2, both expressing the same trivalent antigens (Ag85B, EsxH, ESAT6/EsxA), reduced Mtb lung tissue burden and dissemination in an aerosol challenge mouse model. Conclusions: The novel PICV vector-based TB vaccine candidates can express more than two antigens via the use of P2A linker sequence and elicit strong systemic and lung T cell immunity with protective efficacy. Our study suggests the PICV vector as an attractive vaccine platform for the development of new and effective TB vaccine candidates.


Subject(s)
Tuberculosis Vaccines , Tuberculosis , Animals , Humans , Mice , Antigens, Bacterial/genetics , Antigens, Viral , Bacterial Proteins/genetics , Cytokines/metabolism , Seroepidemiologic Studies , Tuberculosis Vaccines/genetics , Vaccines, Synthetic/genetics , T-Lymphocytes/immunology
15.
J Biol Chem ; 286(28): 24793-805, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21622561

ABSTRACT

Alternative processing of parvovirus B19 (B19V) pre-mRNA is critical to generating appropriate levels of B19V mRNA transcripts encoding capsid proteins and small nonstructural proteins. Polyadenylation of the B19V pre-mRNA at the proximal polyadenylation site ((pA)p), which prevents generation of full-length capsid proteins encoding mRNA transcripts, has been suggested as a step that blocks B19V permissiveness. We report here that efficient splicing of the B19V pre-mRNA within the first intron (upstream of the (pA)p site) stimulated the polyadenylation; in contrast, splicing of the B19V pre-mRNA within the second intron (in which the (pA)p site resides) interfered with the polyadenylation, leading to the generation of a sufficient number of B19V mRNA transcripts polyadenylated at the distal polyadenylation site ((pA)d). We also found that splicing within the second intron and polyadenylation at the (pA)p site compete during processing of the B19V pre-mRNA. Furthermore, we discovered that the U1 RNA that binds to the 5' splice donor site of the second intron is fully responsible for inhibiting polyadenylation at the (pA)p site, whereas actual splicing, and perhaps assembly of the functional spliceosome, is not required. Finally, we demonstrated that inhibition of B19V pre-mRNA splicing within the second intron by targeting an intronic splicing enhancer using a Morpholino antisense oligonucleotide prevented B19V mRNA transcripts polyadenylated at the (pA)d site during B19V infection of human erythroid progenitors. Thus, our study reveals the mechanism by which alternative splicing coordinates alternative polyadenylation to generate full-length B19V mRNA transcripts at levels sufficient to support productive B19V infection.


Subject(s)
Alternative Splicing/physiology , Parvovirus B19, Human/metabolism , RNA 3' Polyadenylation Signals/physiology , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional/physiology , RNA Splice Sites/physiology , RNA, Viral/metabolism , Animals , COS Cells , Chlorocebus aethiops , Humans , Introns/physiology , Morpholinos/metabolism , Morpholinos/pharmacology , Parvovirus B19, Human/genetics , RNA Precursors/genetics , RNA Processing, Post-Transcriptional/drug effects , RNA, Viral/genetics
16.
J Virol ; 85(5): 2463-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21159861

ABSTRACT

Alternative splicing of the precursor mRNA (pre-mRNA) of human parvovirus B19 (B19V) plays a key role in posttranscriptional regulation of B19V gene expression. We report that the central exon of the B19V pre-mRNA is defined by three GAA motif-containing exonic splicing enhancers and a G/GU-rich intronic splicing enhancer that lies adjacent to the second donor site. Moreover, targeting of morpholino antisense oligonucleotides to the two splicing enhancers surrounding the second donor site led to a significant reduction in splicing at this donor site during B19V infection of permissive CD36(+) erythroid progenitor cells.


Subject(s)
Alternative Splicing , Enhancer Elements, Genetic , Exons , Introns , Parvovirus B19, Human/genetics , RNA Precursors/genetics , RNA, Viral/genetics , Base Sequence , Erythema Infectiosum/virology , Erythroid Precursor Cells/virology , Humans , Molecular Sequence Data , RNA, Messenger/genetics
17.
Virulence ; 13(1): 1049-1061, 2022 12.
Article in English | MEDLINE | ID: mdl-35758052

ABSTRACT

Lassa fever (LF) is a neglected tropical disease that is caused by Lassa virus (LASV), a human hemorrhagic fever-causing mammarenavirus. A notable sequela of LF is sensorineural hearing loss (SNHL) that can develop in about 33% of the patients. Animal models of LF-associated SNHL have been limited in size and scope because LASV is a biosafety level 4 (BSL4) pathogen that requires its handling in a high biocontainment laboratory. In this report, we describe the development of an alternative arenavirus hearing loss model by infecting outbred Hartley guinea pigs with a virulent strain (rP18) of the Pichinde virus (PICV), which is a guinea pig-adapted mammarenavirus that has been used as a surrogate model of mammarenaviral hemorrhagic fevers in a conventional (BSL2) laboratory. By measuring auditory brainstem response (ABR) throughout the course of the virulent rP18 PICV infection, we noticed that some of the animals experienced an acute but transient level of hearing loss. Cochleae of hearing-impaired animals, but not of controls, had demonstrable viral RNA by quantitative RT-PCR, indicating the presence of virus in the affected inner ear with no overt histopathological changes. In contrast, neither the outbred Hartley guinea pigs infected with a known avirulent strain (rP2) of PICV nor those that were mock-infected showed any evidence of hearing loss or viral infection of the inner ear. This is the first report of an immunocompetent small animal model of mammarenavirus-induced hearing loss that can be used to evaluate potential therapeutics against virus-induced hearing impairment under a conventional laboratory setting.


Subject(s)
Hearing Loss , Lassa Fever , Pichinde virus , Animals , Disease Models, Animal , Fever , Guinea Pigs , Hearing Loss/complications , Humans , Lassa virus , Pichinde virus/genetics
18.
Vaccines (Basel) ; 10(9)2022 Aug 26.
Article in English | MEDLINE | ID: mdl-36146478

ABSTRACT

Influenza A virus of swine (IAV-S) is an economically important swine pathogen. The IAV-S hemagglutinin (HA) surface protein is the main target for vaccine development. In this study, we evaluated the feasibility of using the recombinant tri-segmented Pichinde virus (rPICV) as a viral vector to deliver HA antigen to protect pigs against IAV-S challenge. Four groups of weaned pigs (T01-T04) were included in the study. T01 was injected with PBS to serve as a non-vaccinated control. T02 was inoculated with rPICV expressing green fluorescence protein (rPICV-GFP). T03 was vaccinated with rPICV expressing the HA antigen of the IAV-S H3N2 strain (rPICV-H3). T04 was vaccinated with the recombinant HA protein antigen of the same H3N2 strain. Pigs were vaccinated twice at day 0 and day 21 and challenged at day 43 by intra-tracheal inoculation with the homologous H3N2 IAV-S strain. After vaccination, all pigs in T03 and T04 groups were seroconverted and exhibited high titers of plasma neutralizing antibodies. After challenge, high levels of IAV-S RNA were detected in the nasal swabs and bronchioalveolar lavage fluid of pigs in T01 and T02 but not in the T03 and T04 groups. Similarly, lung lesions were observed in T01 and T02, but not in the T03 and T04 groups. No significant difference in terms of protection was observed between the T03 and T04 group. Collectively, our results demonstrate that the rPICV-H3 vectored vaccine elicited protective immunity against IAV-S challenge. This study shows that rPICV is a promising viral vector for the development of vaccines against IAV-S.

20.
Front Med (Lausanne) ; 9: 920062, 2022.
Article in English | MEDLINE | ID: mdl-36314011

ABSTRACT

Background: Hepatic fibrosis is a health challenge due to the absence of satisfactory therapy, especially at the cirrhosis stage. Dahuang Zhechong pill (DHZCP)-based therapy is reportedly a successful treatment for hepatic fibrosis and is even beneficial for the treatment of cirrhosis. Hence, a systematic review and clinical evidence assessment of DHZCP-based therapy should be performed, and clinical recommendations based on its efficacy for the treatment of hepatic fibrosis should be generated. With respect to potential indicators, the comparative value of the hepatic function, spleen thickness, and portal vein internal diameter should be evaluated. Materials and methods: PubMed, the Excerpta Medica Database, the Cochrane Library, the Web of Science, the WanFang Database, the Chinese Scientific Journal Database, and the Chinese National Knowledge Infrastructure database were searched to identify clinical trials. Three subgroup analyses were performed based on the stage of disease, medication use, and the course of treatment. Statistical analyses were performed using Review Manager 5.4. Results: A total of 18 studies including 1,494 patients were evaluated. The DHZCP-based therapy was effective in reducing the plasma levels of hyaluronic acid, and laminin, procollagen III, and IV collagen were also reduced irrespective of the hepatitis stage or the presence of hepatic cirrhosis. Abnormalities in alanine aminotransferase, aspartate aminotransferase, albumin, and total bilirubin were reversed. A 6-month course of treatment was the most beneficial DHZCP-based therapy regimen. Alanine aminotransferase improvement was more obvious in patients with cirrhosis, and alanine aminotransferase was reduced significantly in patients with hepatic cirrhosis. With respect to pharmacological mechanisms, DHZCP-based therapy could inhibit hepatic stellate cell growth and activation, reduce inflammation, and prevent extracellular matrix formation. Hepatic portal hypertension and splenomegaly were ameliorated significantly in the DHZCP-based therapy group. Conclusion: Dahuang Zhechong pill-based therapy has demonstrated efficacy as a treatment for hepatic fibrosis and cirrhosis. A 6-month course of treatment is the recommended option for DHZCP-based therapy in clinical practice. The combination of DHZCP-based therapy and entecavir is a favorable treatment for hepatic cirrhosis.

SELECTION OF CITATIONS
SEARCH DETAIL