Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters











Publication year range
1.
PLoS One ; 17(11): e0272594, 2022.
Article in English | MEDLINE | ID: mdl-36322572

ABSTRACT

With the rapid progress made in the development of vaccines to fight the SARS-CoV-2 pandemic, almost >90% of vaccine candidates under development and a 100% of the licensed vaccines are delivered intramuscularly (IM). While these vaccines are highly efficacious against COVID-19 disease, their efficacy against SARS-CoV-2 infection of upper respiratory tract and transmission is at best temporary. Development of safe and efficacious vaccines that are able to induce robust mucosal and systemic immune responses are needed to control new variants. In this study, we have used our nanoemulsion adjuvant (NE01) to intranasally (IN) deliver stabilized spike protein (S-2P) to induce immunogenicity in mouse and hamster models. Data presented demonstrate the induction of robust immunity in mice resulting in 100% seroconversion and protection against SARS-CoV-2 in a hamster challenge model. There was a significant induction of mucosal immune responses as demonstrated by IgA- and IgG-producing memory B cells in the lungs of animals that received intranasal immunizations compared to an alum adjuvanted intramuscular vaccine. The efficacy of the S-2P/NE01 vaccine was also demonstrated in an intranasal hamster challenge model with SARS-CoV-2 and conferred significant protection against weight loss, lung pathology, and viral clearance from both upper and lower respiratory tract. Our findings demonstrate that intranasal NE01-adjuvanted vaccine promotes protective immunity against SARS-CoV-2 infection and disease through activation of three arms of immune system: humoral, cellular, and mucosal, suggesting that an intranasal SARS-CoV-2 vaccine may play a role in addressing a unique public health problem and unmet medical need.


Subject(s)
COVID-19 , Immunity, Mucosal , Mice , Humans , Animals , Cricetinae , COVID-19 Vaccines , Antibodies, Viral , COVID-19/prevention & control , SARS-CoV-2 , Adjuvants, Immunologic , Administration, Intranasal , Antibodies, Neutralizing , Spike Glycoprotein, Coronavirus
2.
Sci Rep ; 12(1): 11369, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35790783

ABSTRACT

Intramuscular vaccines have greatly reduced hospitalization and death due to severe COVID-19. However, most countries are experiencing a resurgence of infection driven predominantly by the Delta and Omicron variants of SARS-CoV-2. In response, booster dosing of COVID-19 vaccines has been implemented in many countries to address waning immunity and reduced protection against the variants. However, intramuscular boosting fails to elicit mucosal immunity and therefore does not solve the problem of persistent viral carriage and transmission, even in patients protected from severe disease. In this study, two doses of stabilized prefusion SARS-CoV-2 spike (S-2P)-based intramuscular vaccine adjuvanted with Alum/CpG1018, MVC-COV1901, were used as a primary vaccination series, followed by an intranasal booster vaccination with nanoemulsion (NE01)-adjuvanted S-2P vaccine in a hamster model to demonstrate immunogenicity and protection from viral challenge. Here we report that this vaccination regimen resulted not only in the induction of robust immunity and protection against weight loss and lung pathology following challenge with SARS-CoV-2, but also led to increased viral clearance from both upper and lower respiratory tracts. Our findings showed that intramuscular MVC-COV1901 vaccine followed by a booster with intranasal NE01-adjuvanted vaccine promotes protective immunity against both viral infection and disease, suggesting that this immunization protocol may offer a solution in addressing a significant, unmet medical need for both the COVID-19 and future pandemics.


Subject(s)
COVID-19 , Viral Vaccines , Adjuvants, Immunologic , Animals , Antibodies, Viral , COVID-19/prevention & control , COVID-19 Vaccines , Cricetinae , Humans , SARS-CoV-2
3.
Infect Genet Evol ; 96: 105096, 2021 12.
Article in English | MEDLINE | ID: mdl-34601094

ABSTRACT

Human metapneumovirus (HMPV), an unsegmented negative-strand RNA virus, is the second most detected respiratory pathogen and one of the leading causes of respiratory illness in infants and immunodeficient individuals. HMPV infection of permissive cells in culture triggers a transient IFN response, which is efficiently suppressed later in infection. We report that two structural glycoproteins of the virus - namely G (Glycoprotein) and SH (Small Hydrophobic) - suppress the type I interferon (IFN) response in cell culture. This is manifested by inhibition of diverse steps of IFN induction and response, such as phosphorylation and nuclear translocation of IFN regulatory factor-3 and -7 (IRF3, IRF7), major transcription factors of the IFN gene. Furthermore, HMPV suppresses the cellular response to IFN by inhibiting the phosphorylation of STAT1 (Signal Transducer and Activator of Transcription 1), required for the induction of IFN-stimulated genes that act as antivirals. Site-directed mutagenesis revealed an important role of critical cysteine (Cys) residues in the Cys-rich carboxy terminal region of the SH protein in IFN suppression, whereas for G, the ectodomain plays a role. These results shed light on the mechanism of IFN suppression by HMPV, and may also offer avenues for new antiviral approaches in the future.


Subject(s)
Glycoproteins/metabolism , Immunity, Innate , Paramyxoviridae Infections/immunology , Viral Proteins/metabolism , A549 Cells , Humans , Metapneumovirus/physiology , Paramyxoviridae Infections/virology
4.
Front Immunol ; 10: 2411, 2019.
Article in English | MEDLINE | ID: mdl-31681300

ABSTRACT

DNA vaccines have great potential to control infectious disease, particularly those caused by intracellular organisms. They are inexpensive to produce and can be quickly modified to combat emerging infectious threats, but often fail to generate a strong immunologic response limiting enthusiasm for their use in humans and animals. To improve the immunogenic response, we developed a DNA vaccine in which the F protein ectodomain of Respiratory Syncytial Virus (RSV-F) was covalently linked to specific antigens of interest. The presence of the RSV-F ectodomain allowed secretion of the translated fusion product out of the originally transfected cells followed by its active binding to adjacent cells. This allowed the targeting of a greater number of cells than those originally transfected, enhancing both humoral and cytotoxic immune responses against the expressed antigen(s). We developed an engrafted mouse model that used antigen-expressing tumor cells to assess the in vivo cytotoxic immune response to specific antigens. We then used this model to demonstrate that a DNA vaccine in which the RSV-F ectodomain is fused to two antigens expressed by Burkholderia pseudomallei, the intracellular gram-negative organism that causes melioidosis, generated a stronger cytotoxic response than a DNA vaccine that lacked the RSV-F sequence while still generating a robust humoral response.


Subject(s)
Bacterial Proteins , Burkholderia pseudomallei , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Viruses , Vaccines, DNA , Viral Fusion Proteins , Virulence Factors , Animals , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Burkholderia pseudomallei/genetics , Burkholderia pseudomallei/immunology , Mice , Mice, Inbred BALB C , Protein Domains , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/genetics , Respiratory Syncytial Viruses/immunology , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology , Virulence Factors/genetics , Virulence Factors/immunology
6.
Vaccine ; 37(43): 6470-6477, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31515143

ABSTRACT

Genital herpes is a sexually transmitted disease representing a major global health concern. Currently, there is no approved vaccine and existing antiviral therapies exhibit limited efficacy. Herein, we describe an intranasal (IN) vaccine comprised of HSV-2 surface glycoproteins gD2 and gB2 formulated in a nanoemulsion adjuvant (NE01-gD2/gB2). Using the HSV-2 genital herpes guinea pig model, we demonstrate that IN NE01-gD2/gB2 induces higher levels of neutralizing antibody compared to a monovalent IN NE01-gD2 vaccine, but less than an intramuscular (IM) Alum/MPL-gD2 vaccine. Following intravaginal (IVag) challenge with HSV-2, the group immunized with IN NE01-gD2/gB2 exhibited significantly reduced acute and recurrent disease scores compared to placebo recipients. Significantly, latent virus was only detected in the dorsal root ganglia of 1 of 12 IN NE01-gD2/gB2-vaccinated animals compared to 11 of 12 placebo recipient. In the therapeutic model, IN NE01-gD2/gB2 immunized guinea pigs exhibited a significant reduction in the recurrent lesions scores (64%, p < 0.01), number of animal days with disease (64%, p < 0.01), number of animals with viral shedding (50%, p < 0.04) and reduction in virus positive vaginal swabs (56%, p < 0.04), These data suggests that the treatment may be effective in treating chronic disease and minimizing virus transmission. These results warrant advancing the development of IN NE01-gD2/gB2 as both a prophylactic and therapeutic vaccine against HSV-2.


Subject(s)
Antibodies, Viral/blood , Herpes Genitalis/prevention & control , Herpes Genitalis/therapy , Herpes Simplex Virus Vaccines/administration & dosage , Herpes Simplex Virus Vaccines/therapeutic use , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Disease Models, Animal , Emulsions/administration & dosage , Emulsions/chemistry , Female , Guinea Pigs , Herpes Genitalis/immunology , Herpesvirus 2, Human , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/therapeutic use , Virus Shedding
7.
Mol Ther Nucleic Acids ; 5(10): e369, 2016 Oct 04.
Article in English | MEDLINE | ID: mdl-27701401

ABSTRACT

Minimal piggyBac vectors are a modified single-plasmid version of the classical piggyBac delivery system that can be used for stable transgene integration. These vectors have a truncated terminal domain in the delivery cassette and thus, integrate significantly less flanking transposon DNA into host cell chromatin than classical piggyBac vectors. Herein, we test various characteristics of this modified transposon. The integration efficiency of minimal piggyBac vectors was inversely related to the size of both the transposon and the entire plasmid, but inserts as large as 15 kb were efficiently integrated. Open and super-coiled vectors demonstrated the same integration efficiency while DNA methylation decreased the integration efficiency and silenced the expression of previously integrated sequences in some cell types. Importantly, the incidence of plasmid backbone integration was not increased above that seen in nontransposon control vectors. In BALB/c mice, we demonstrated prolonged expression of two transgenes (intracellular mCherry and secretable Gaussia luciferase) when delivered by the minimal piggyBac that resulted in a more sustained antibody production against the immunogenic luciferase than when delivered by a transient (nontransposon) vector plasmid. We conclude that minimal piggyBac vectors are an effective alternative to other integrative systems for stable DNA delivery in vitro and in vivo.

8.
BMC Res Notes ; 8: 379, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26306622

ABSTRACT

BACKGROUND: Transient production of gamma-retroviruses, including self-inactivating (SIN) retroviruses, is a common method for rapidly generating virus capable of gene delivery. Stable (continuous) production of virus is preferable to transient production for clinical and biotechnology purposes, however, because it allows for significant quantities of a uniform virus to be generated over a prolonged period of time, thus allowing for longitudinal functional studies and quality analysis. Unfortunately, stable production of SIN retroviruses is difficult to achieve. RESULTS: We describe a novel method to rapidly and cost-effectively create packaging cells capable of continuously producing self-inactivating gamma-retroviruses. We imbedded the SIN proviral construct into a minimal piggyBac transposon vector and then integrated the hybrid vector into packaging cells that already stably expressed the viral gag-pro-pol and envelope genes. Cells that stably produced self-inactivating gamma-retroviruses could be identified (and purified) as early as 3 weeks after initial transfection; these cells produced virus for at least 9 weeks without a decline in titer. CONCLUSIONS: This viral-minimal piggyBac hybrid vector allowed for the rapid generation and purification of packaging cells capable of stably producing self-inactivated gamma-retroviruses. This method can be applied to the large-scale production of viruses for use in research, biotechnology, and potentially, clinical trials.


Subject(s)
DNA Transposable Elements , Genetic Vectors , Retroviridae/physiology , HEK293 Cells , Humans , Retroviridae/genetics
9.
Hum Vaccin Immunother ; 11(12): 2904-12, 2015.
Article in English | MEDLINE | ID: mdl-26307915

ABSTRACT

Respiratory Syncytial Virus is a leading cause of bronchiolitis and pneumonia in infants, the elderly and individuals with compromised immune systems. Despite decades of research, there is currently no available vaccine for RSV. Our group has previously demonstrated that intranasal immunization of mice with RSV inactivated by and adjuvanted with W805EC nanoemulsion elicits robust humoral and cellular immune responses, resulting in protection against RSV infection. This protection was achieved without the induction of airway hyper-reactivity or a Th2-skewed immune response. The cotton rat Sigmodon hispidus has been used for years as an excellent small animal model of RSV disease. Thus, we extended these rodent studies to the more permissive cotton rat model. Intranasal immunization of the nanoemulsion-adjuvanted RSV vaccines induced high antibody titers and a robust Th1-skewed cellular response. Importantly, vaccination provided sterilizing cross-protective immunity against a heterologous RSV challenge and did not induce marked or severe histological effects or eosinophilia in the lung after viral challenge. Overall, these data demonstrate that nanoemulsion-formulated whole RSV vaccines are both safe and effective for immunization in multiple animal models.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Emulsions/therapeutic use , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/therapeutic use , Sigmodontinae/immunology , Vaccines, Inactivated/immunology , Administration, Intranasal , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Bronchiolitis/immunology , Bronchiolitis/prevention & control , Bronchiolitis/virology , Cross Protection/immunology , Female , Lung/pathology , Lung/virology , Pneumonia, Viral/immunology , Pneumonia, Viral/prevention & control , Pneumonia, Viral/virology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Sigmodontinae/virology , Th1 Cells/immunology , Vaccination , Viral Fusion Proteins/immunology , Viral Load/immunology
10.
Hum Vaccin Immunother ; 10(3): 615-22, 2014.
Article in English | MEDLINE | ID: mdl-24326268

ABSTRACT

Respiratory Syncytial Virus (RSV) is a ubiquitous virus that infects almost all people by age two and is a major source of respiratory illness in infants, the elderly and others with compromised immune systems. Currently there is no available vaccine. Prior efforts using formalin-inactivated RSV (FI-RSV) were associated with enhanced respiratory disease upon viral exposure following clinical vaccine trials. Several researchers and pharmaceutical companies have utilized vector-associated live attenuated RSV vaccines in pre-clinical and clinical studies. Another attractive approach, however, is a subunit vaccine which would be easier to produce and quality control. Our group has previously demonstrated in a murine model of infection that intranasal immunization with nanoemulsion-inactivated and adjuvanted RSV induces humoral and cellular immune responses, resulting in protection against RSV infection. The present studies characterize the immune responses elicited by intranasal RSV F protein adjuvanted with nanoemulsion. Intranasal application of nanoemulsion adjuvanted F protein induced a rapid and robust systemic and mucosal antibody response, as well as protection against subsequent RSV challenge. Importantly, RSV challenge in immunized animals did not elicit airway hyper-reactivity, a Th2-skewed immune response or immunopathology associated with hypersensitivity reactions with formalin-inactivated vaccine. These results suggest that RSV F protein adjuvanted with nanoemulsion may be a good mucosal vaccine candidate. Formulating RSV F protein in nanoemulsion creates a well-defined and well-controlled vaccine that can be delivered intranasally to induce T cell mediated immunity without inducing enhanced disease associated with the mouse model of FI-RSV vaccination and infection.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Immunization/methods , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Viruses/immunology , Administration, Intranasal , Animals , Antibodies, Viral/analysis , Antibodies, Viral/blood , Blood/immunology , Disease Models, Animal , Emulsions/administration & dosage , Female , Immunity, Mucosal , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Viruses/isolation & purification , Th2 Cells/immunology , Treatment Outcome , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Viral Fusion Proteins/immunology
11.
Am J Physiol Lung Cell Mol Physiol ; 297(3): L538-45, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19561138

ABSTRACT

Using adapted retroviruses for gene delivery is a modern and powerful tool in biological research as well as a promising approach for gene therapy. An important limitation for the extensive use of retroviral vectors is the low infection rate in target cells such as pulmonary vascular endothelial cells due to the insufficient infectivity of standard retrovirus supernatants that can only be overcome by complicated methods of virus concentration. This paper describes two easy methods to augment target cell infectivity, first by increasing the retroviral titer in the medium collected from packaging cells by stimulation of viral propagation with dexamethasone, and second, by increasing target cell sensitivity to retroviral infection by the glucocorticoid receptor antagonist, mifepristone. Using this method, we increased the infectivity of pulmonary microvascular endothelial cells from 16% to 85%. We demonstrate that mifepristone increased the susceptibility of target cells to retroviruses without increasing the viral titer of the supernatant. Dexamethasone, but not mifepristone, increased expression of delivered proteins such as GFP that are important for early identification of infected cells. Each, or both step(s), may be included in a standard protocol for retrovirus propagation and infection of target cells.


Subject(s)
Dexamethasone/pharmacology , Gene Transfer Techniques , Mifepristone/pharmacology , Retroviridae/drug effects , Retroviridae/pathogenicity , Animals , Culture Media, Conditioned , Endothelial Cells/drug effects , Endothelial Cells/virology , Hormones/metabolism , Promoter Regions, Genetic/genetics , Rats , Receptors, Glucocorticoid/antagonists & inhibitors , Retroviridae/physiology , Serum , Subcellular Fractions/metabolism , Terminal Repeat Sequences/genetics , Viral Proteins/metabolism
12.
J Virol ; 82(16): 7977-87, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18550659

ABSTRACT

The La antigen (SS-B) associates with a wide variety of cellular and viral RNAs to affect gene expression in multiple systems. We show that La is the major cellular protein found to be associated with the abundant 44-nucleotide viral leader RNA (leRNA) early after infection with respiratory syncytial virus (RSV), a nonsegmented negative-strand RNA virus. Consistent with this, La redistributes from the nucleus to the cytoplasm in RSV-infected cells. Upon RNA interference knockdown of La, leRNA is redirected to associate with the RNA-binding protein RIG-I, a known activator of interferon (IFN) gene expression, and this is accompanied by the early induction of IFN mRNA. These results suggest that La shields leRNA from RIG-I, abrogating the early viral activation of type I IFN. We mapped the leRNA binding function to RNA recognition motif 1 of La and showed that while wild-type La greatly enhanced RSV growth, a La mutant defective in RSV leRNA binding also did not support RSV growth. Comparative studies of RSV and Sendai virus and the use of IFN-negative Vero cells indicated that La supports the growth of nonsegmented negative-strand RNA viruses by both IFN suppression and a potentially novel IFN-independent mechanism.


Subject(s)
Autoantigens/physiology , DEAD-box RNA Helicases/chemistry , RNA, Viral , Ribonucleoproteins/physiology , Amino Acid Motifs , Animals , Autoantigens/chemistry , Cell Line, Tumor , Cell Nucleus/metabolism , Chlorocebus aethiops , Cloning, Molecular , Cytoplasm/metabolism , DEAD Box Protein 58 , Humans , Models, Genetic , RNA Interference , Receptors, Immunologic , Ribonucleoproteins/chemistry , Vero Cells , Virus Replication , SS-B Antigen
13.
Methods Mol Biol ; 442: 75-82, 2008.
Article in English | MEDLINE | ID: mdl-18369779

ABSTRACT

The intranasal administration of siRNA has opened new vistas in drug delivery and respiratory therapy. In this strategy, synthetic siRNA with or without chemical modifications can be applied intranasally. Various delivery vehicles have been tested and optimized. With a few exceptions, all promote significant uptake of siRNA into the lung tissue and offer protection against respiratory viruses such as respiratory syncytial virus (RSV), parainfluenza virus (PIV), and influenza virus. No major adverse immune reaction has been encountered. Nasally applied siRNA remains within the lung and does not have systemic access, as judged by its absence in other major organs such as the lung, liver, heart, and kidney. We provide techniques for using the nose as a specific route for siRNA delivery into the lung of laboratory animals, which has enormous potential for clinical applications.


Subject(s)
Administration, Intranasal , RNA, Small Interfering/administration & dosage , Animals , Humans , Mice , Mice, Inbred BALB C , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transfection
14.
J Mol Med (Berl) ; 86(3): 313-22, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18193184

ABSTRACT

MicroRNAs (miRNAs) are endogenous noncoding RNAs that down-regulate gene expression by promoting cleavage or translational arrest of target mRNAs. While most miRNAs are transcribed from their own dedicated genes, some map to introns of 'host' transcripts, the biological significance of which remains unknown. Here, we show that prostate cells are naturally devoid of EGF-like domain 7 (Egfl7) transcripts and hence also deficient in a miRNA, miR-126*, generated from splicing and processing of its ninth intron. Use of recombinant and synthetic miRNAs or a specific antagomir established a role of miR-126* in silencing prostein in non-endothelial cells. We mapped two miR-126*-binding sites in the 3'UTR of the prostein mRNA required for translational repression. Transfection of synthetic miR-126* into prostate cancer LNCaP cells strongly reduced the translation of prostein. Interestingly, loss of prostein correlated with reduction of LNCaP cell migration and invasion. Thus, the robust expression of prostein protein in the prostate cells results from a combination of transcriptional activation of the prostein gene and absence of intronic miRNA-126* due to the prostate-specific repression of the Egfl7 gene. We conclude that intronic miRNAs from tissue-specific transcripts, or their natural absence, make cardinal contributions to cellular gene expression and phenotype. These findings also open the door to tissue-specific miRNA therapy.


Subject(s)
Endothelium, Vascular/metabolism , Introns/genetics , Membrane Proteins/genetics , MicroRNAs/genetics , Neoplasm Proteins/genetics , Prostatic Neoplasms/pathology , Protein Biosynthesis , 3' Untranslated Regions , Base Sequence , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Male , Membrane Proteins/metabolism , Models, Genetic , Molecular Sequence Data , Neoplasm Invasiveness , Organ Specificity , RNA Splicing/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering
15.
Curr Opin Mol Ther ; 9(2): 119-25, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17458164

ABSTRACT

Recent progress in the development of two antisense strategies--DNA oligonucleotides and RNA interference for the treatment of respiratory diseases are discussed in this review. Appropriate formulations and chemical modifications for improved stability should foster the clinical application of antisense drugs, but the delivery of antisense drugs to their intended target tissue remains the biggest challenge for most therapeutic applications of these compounds. The nose provides a particularly attractive route for antisense drug delivery to the respiratory system; however, significant hurdles remain.


Subject(s)
Administration, Intranasal , Biological Therapy , Drug Evaluation, Preclinical , Oligonucleotides, Antisense/administration & dosage , Respiratory Tract Diseases/therapy , Animals , Humans , RNA Interference
16.
J Mol Med (Berl) ; 85(7): 745-52, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17351763

ABSTRACT

Stable RNA interference (RNAi) is commonly achieved by recombinant expression of short hairpin RNA (shRNA). To generate virus-resistant cell lines, we cloned a shRNA cassette against the phosphoprotein gene of respiratory syncytial virus (RSV) into a polIII-driven plasmid vector. Analysis of individual stable transfectants showed a spectrum of RSV resistance correlating with the levels of shRNA expressed from different chromosomal locations. Interestingly, resistance in a minority of clones was due to mono-allelic disruption of the cellular gene for vasodilator-stimulated phosphoprotein (VASP). Thus, pure clones of chromosomally integrated DNA-directed RNAi can exhibit gene disruption phenotypes resembling but unrelated to RNAi.


Subject(s)
Cell Adhesion Molecules/physiology , Immunity , Microfilament Proteins/physiology , Oligodeoxyribonucleotides/genetics , Phosphoproteins/physiology , RNA Interference , RNA, Small Interfering/physiology , Respiratory Syncytial Viruses/immunology , Base Sequence , Cell Adhesion Molecules/genetics , Chromosomes, Human , Cloning, Molecular/methods , Genetic Vectors , HeLa Cells , Humans , Microfilament Proteins/genetics , Oligodeoxyribonucleotides/chemical synthesis , Oligodeoxyribonucleotides/pharmacology , Phosphoproteins/genetics , Respiratory Syncytial Viruses/growth & development , Transfection
17.
Drug Discov Today Ther Strateg ; 4(4): 273-276, 2007.
Article in English | MEDLINE | ID: mdl-19081824

ABSTRACT

This review summarizes recent experimental achievements in the area of the development of new RNA interference (RNAi) therapeutics for the treatment of viral respiratory diseases. Delivery of siRNA to their intended target tissue remains the biggest problem for most therapeutic applications of these compounds. Appropriate formulations and chemical modifications for improved stability will boost the probability of utilization of RNAi drugs in the clinical applications.

18.
J Virol ; 81(2): 783-90, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17050596

ABSTRACT

Respiratory syncytial virus (RSV) is the foremost respiratory pathogen in newborns and claims millions of lives annually. However, there has been no methodical study of the pathway(s) of entry of RSV or its interaction with nonrespiratory tissues. We and others have recently established a significant association between allergic conjunctivitis and the presence of RSV in the eye. Here we adopt a BALB/c mouse model and demonstrate that when instilled in the live murine eye, RSV not only replicated robustly in the eye but also migrated to the lung and produced a respiratory disease that is indistinguishable from the standard, nasally acquired RSV disease. Ocularly applied synthetic anti-RSV small interfering RNA prevented infection of the eye as well as the lung. RSV infection of the eye activated a plethora of ocular cytokines and chemokines with profound relevance to inflammation of the eye. Anticytokine treatments in the eye reduced ocular inflammation but had no effect on viral growth in both eye and lung, demonstrating a role of the cytokine response in ocular pathology. These results establish the eye as a major gateway of respiratory infection and a respiratory virus as a bona fide eye pathogen, thus offering novel intervention and treatment options.


Subject(s)
Conjunctivitis, Viral/transmission , Conjunctivitis, Viral/virology , Eye/virology , Lung/virology , Respiratory Syncytial Virus Infections/transmission , Respiratory Syncytial Virus, Human/pathogenicity , Respiratory Tract Infections/virology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Conjunctivitis, Viral/therapy , Disease Models, Animal , Eye/immunology , Female , Humans , Interleukin-1alpha/immunology , Lung/immunology , Mice , Mice, Inbred BALB C , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Respiratory Syncytial Virus Infections/therapy , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/metabolism , Respiratory Syncytial Virus, Human/physiology , Respiratory Tract Infections/therapy , Treatment Outcome , Tumor Necrosis Factor-alpha/immunology , Virus Replication
19.
J Virol ; 81(4): 1786-95, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17151097

ABSTRACT

The two nonstructural (NS) proteins NS1 and NS2 of respiratory syncytial virus (RSV) are abundantly expressed in the infected cell but are not packaged in mature progeny virions. We found that both proteins were expressed early in infection, whereas the infected cells underwent apoptosis much later. Coincident with NS protein expression, a number of cellular antiapoptotic factors were expressed or activated at early stages, which included NF-kappaB and phosphorylated forms of protein kinases AKT, phosphoinositide-dependent protein kinase, and glycogen synthase kinase. Using specific short interfering RNAs (siRNAs), we achieved significant knockdown of one or both NS proteins in the infected cell, which resulted in abrogation of the antiapoptotic functions and led to early apoptosis. NS-dependent suppression of apoptosis was observed in Vero cells that are naturally devoid of type I interferons (IFN). The siRNA-based results were confirmed by the use of NS-deleted RSV mutants. Early activation of epidermal growth factor receptor (EGFR) in the RSV-infected cell did not require NS proteins. Premature apoptosis triggered by the loss of NS or by apoptosis-promoting drugs caused a severe reduction of RSV growth. Finally, recombinantly expressed NS1 and NS2, individually and together, reduced apoptosis by tumor necrosis factor alpha, suggesting an intrinsic antiapoptotic property of both. We conclude that the early-expressed nonstructural proteins of RSV boost viral replication by delaying the apoptosis of the infected cell via a novel IFN- and EGFR-independent pathway.


Subject(s)
Apoptosis , Interferons/physiology , NF-kappa B/physiology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/pathogenicity , Viral Nonstructural Proteins/metabolism , Animals , Cell Line , Chlorocebus aethiops , Down-Regulation , ErbB Receptors/physiology , Humans , Proteasome Endopeptidase Complex , Protein Kinases/metabolism , Proteins/metabolism , Respiratory Syncytial Viruses/physiology , Vero Cells , Virus Replication
20.
Expert Opin Biol Ther ; 6(11): 1151-60, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17049013

ABSTRACT

Respiratory viruses, such as influenza, parainfluenza and respiratory syncytial virus (RSV), claim millions of lives annually. At present, there is no completely effective vaccine or drug against these highly mutable RNA viruses. Passive antibody therapies for RSV, despite their limited application and staggering cost, enjoy a virtual monopoly in a multibillion-dollar global market. Recently, however, pioneering discoveries have launched RNA interference as a novel, nucleic acid-based therapy against viral pathogens. Specifically, small interfering RNAs (siRNAs) offered protection against respiratory syncytial virus, parainfluenza and influenza. siRNA against RSV has entered Phase I clinical trials in humans, and preliminary reports are promising. If appropriately formulated for improved specificity, delivery and pharmacokinetics, siRNAs may indeed become effective antivirals in the clinics of the future. This paper provides an overview of the prospects and hurdles facing the antiviral siRNA drugs, with special emphasis on RSV.


Subject(s)
RNA Interference , RNA, Small Interfering/therapeutic use , RNA, Viral/physiology , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Viruses/drug effects , Animals , Humans , RNA Interference/drug effects , RNA, Viral/genetics , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/genetics
SELECTION OF CITATIONS
SEARCH DETAIL