Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Infect Dis ; 226(3): 374-385, 2022 08 26.
Article in English | MEDLINE | ID: mdl-35668702

ABSTRACT

BACKGROUND: The true burden of lower respiratory tract infections (LRTIs) due to respiratory syncytial virus (RSV) remains unclear. This study aimed to provide more robust, multinational data on RSV-LRTI incidence and burden in the first 2 years of life. METHODS: This prospective, observational cohort study was conducted in Argentina, Bangladesh, Canada, Finland, Honduras, South Africa, Thailand, and United States. Children were followed for 24 months from birth. Suspected LRTIs were detected via active (through regular contacts) and passive surveillance. RSV and other viruses were detected from nasopharyngeal swabs using PCR-based methods. RESULTS: Of 2401 children, 206 (8.6%) had 227 episodes of RSV-LRTI. Incidence rates (IRs) of first episode of RSV-LRTI were 7.35 (95% confidence interval [CI], 5.88-9.08), 5.50 (95% CI, 4.21-7.07), and 2.87 (95% CI, 2.18-3.70) cases/100 person-years in children aged 0-5, 6-11, and 12-23 months. IRs for RSV-LRTI, severe RSV-LRTI, and RSV hospitalization tended to be higher among 0-5 month olds and in lower-income settings. RSV was detected for 40% of LRTIs in 0-2 month olds and for approximately 20% of LRTIs in older children. Other viruses were codetected in 29.2% of RSV-positive nasopharyngeal swabs. CONCLUSIONS: A substantial burden of RSV-LRTI was observed across diverse settings, impacting the youngest infants the most. Clinical Trials Registration. NCT01995175.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Respiratory Tract Infections , Viruses , Child , Hospitalization , Humans , Incidence , Infant , Prospective Studies
2.
J Infect Dis ; 225(12): 2067-2076, 2022 06 15.
Article in English | MEDLINE | ID: mdl-34146100

ABSTRACT

BACKGROUND: Respiratory syncytial virus (RSV) causes respiratory tract infections, which may require hospitalization especially in early infancy. Transplacental transfer of RSV antibodies could confer protection to infants in their first months of life. METHODS: In this first-in-human, placebo-controlled study, 502 healthy nonpregnant women were randomized 1:1:1:1 to receive a single dose of unadjuvanted vaccine containing 30/60/120 µg of RSV fusion (F) protein stabilized in the prefusion conformation (RSVPreF3) or placebo. RESULTS: Solicited local adverse events (AEs) were more frequently reported in the RSVPreF3 groups (4%-53.2%) versus placebo (0%-15.9%); most were mild/moderate. Unsolicited AEs were comparably reported among groups. Three serious AEs were reported; none was vaccination-related. Compared with prevaccination values, anti-RSV A neutralizing antibody geometric mean titers and anti-RSVPreF3 immunoglobulin G geometric mean concentrations increased 8- to 14-fold and 12- to 21-fold at day 8 and persisted 5- to 6-fold and 6- to 8-fold higher until day 91 in the RSVPreF3 groups versus 1-fold in placebo. Comparisons at day 8 and day 31 showed that the higher dose levels were significantly more immunogenic than the lowest one. CONCLUSIONS: The RSVPreF3 vaccine was well tolerated and immunogenic. The 60 and 120 µg dose levels were selected for further investigation in pregnant women. CLINICAL TRIALS REGISTRATION: NCT03674177.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , Antibodies, Neutralizing , Antibodies, Viral , Female , Humans , Infant , Pregnancy , Viral Fusion Proteins
3.
Cent Eur J Public Health ; 29(3): 167-176, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34623114

ABSTRACT

OBJECTIVES: Due to immunosenescence and presence of comorbidities, respiratory syncytial virus (RSV) disease burden is a major health concern in older adults, which is expected to increase with the life expectancy rise. Data on RSV burden are scarce in older adults residing in long-term care facilities, a vulnerable population living in crowded settings. Therefore, two independent prospective studies were conducted during the 2003-2004 and 2004-2005 RSV seasons to assess RSV acute respiratory illnesses (ARIs) and lower respiratory tract infections (LRTIs) in ≥ 65-year-old adults residing in long-term care facilities in the Czech Republic. METHODS: RSV ARI episodes were confirmed by polymerase chain reaction in nasal swabs collected within 3 days of symptoms onset. The mortality and morbidity of RSV-confirmed ARIs, as well as the risk factors associated with RSV-confirmed ARIs were evaluated. RESULTS: Among 1,251 participants in the 2003-2004 season (ARI surveillance between October and March), there were no RSV-positive cases in 255 ARI and 105 LRTI episodes. Among 1,280 participants in the 2004-2005 season (ARI surveillance between October and April), there were 39 and 26 RSV-positive cases in 335 ARI and 217 LRTI episodes, respectively, and RSV-positive ARI and LRTI episode incidence rates were 45.82 and 30.40 per 1,000 person-years. Among 290 RSV-negative and 39 RSV-positive ARI cases in the 2004-2005 season, 15 and 4 hospitalizations, 188 and 26 LRTIs, and 11 and 3 deaths were reported. Risk factors associated with RSV-positive ARI were female gender (odds ratio: 4.98), chronic heart failure class II (odds ratio: 2.31) and diabetes requiring insulin treatment (odds ratio: 9.82). CONCLUSIONS: These studies showed that RSV was an important cause of ARI in older adults living in long-term care facilities in the 2004-2005 season, with fluctuating yearly incidences.


Subject(s)
Respiratory Syncytial Virus, Human , Respiratory Tract Infections , Aged , Czech Republic/epidemiology , Female , Humans , Long-Term Care , Prospective Studies , Respiratory Tract Infections/epidemiology
4.
J Infect Dis ; 222(Suppl 7): S658-S665, 2020 10 07.
Article in English | MEDLINE | ID: mdl-32794560

ABSTRACT

Respiratory syncytial virus (RSV) is the leading viral pathogen associated with acute lower respiratory tract infection and hospitalization in children < 5 years of age worldwide. While there are known clinical risk factors for severe RSV infection, the majority of those hospitalized are previously healthy infants. There is consequently an unmet need to identify biomarkers that predict host response, disease severity, and sequelae. The primary objective is to identify biomarkers of severe RSV acute respiratory tract infection (ARTI) in infants. Secondary objectives include establishing biomarkers associated with respiratory sequelae following RSV infection and characterizing the viral load, RSV whole-genome sequencing, host immune response, and transcriptomic, proteomic, metabolomic and epigenetic signatures associated with RSV disease severity. Six hundred thirty infants will be recruited across 3 European countries: the Netherlands, Spain, and the United Kingdom. Participants will be recruited into 2 groups: (1) infants with confirmed RSV ARTI (includes upper and lower respiratory tract infections), 500 without and 50 with comorbidities; and (2) 80 healthy controls. At baseline, participants will have nasopharyngeal, blood, buccal, stool, and urine samples collected, plus complete a questionnaire and 14-day symptom diary. At convalescence (7 weeks ± 1 week post-ARTI), specimen collection will be repeated. Laboratory measures will be correlated with symptom severity scores to identify corresponding biomarkers of disease severity. CLINICAL TRIALS REGISTRATION: NCT03756766.


Subject(s)
Disease Progression , Respiratory Syncytial Virus Infections/diagnosis , Respiratory Syncytial Virus Infections/virology , Severity of Illness Index , Biomarkers , Case-Control Studies , Epigenomics , Europe , Female , Humans , Infant , Male , Metabolomics , Nasopharynx/virology , Netherlands , Proteomics , Respiratory Syncytial Virus, Human/isolation & purification , Respiratory Tract Infections/diagnosis , Respiratory Tract Infections/virology , Risk Factors , Spain , Surveys and Questionnaires , Transcriptome , United Kingdom , Viral Load
5.
J Infect Dis ; 220(11): 1816-1825, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31418022

ABSTRACT

BACKGROUND: Respiratory syncytial virus (RSV) is a common cause of respiratory tract illness and hospitalization in neonates and infants. RSV vaccination during pregnancy may protect offspring in their first months of life. METHODS: This randomized, observer-blind, multicenter, phase 2 study evaluated the immunogenicity and safety of an RSV candidate vaccine in healthy nonpregnant women aged 18-45 years. Four hundred participants were randomized (1:1:1:1) to receive a single intramuscular dose of vaccine containing 30 µg, 60 µg, or 120 µg of RSV fusion protein engineered to preferentially maintain a prefusion conformation (RSV-PreF vaccine) or placebo. RESULTS: Thirty days postvaccination, RSV-A neutralizing antibody geometric mean titers (GMTs) increased 3.75-, 4.42- and 4.36-fold; RSV-B neutralizing antibody GMTs 2.36-, 2.54- and 2.76-fold; and palivizumab competing antibody (PCA) concentrations 11.69-, 14.38- and 14.24-fold compared with baseline levels in the 30 µg, 60 µg, and 120 µg RSV-PreF groups, respectively. Antibody titers and PCA concentrations at day 30 were significantly higher with the 120 µg compared to the 30 µg RSV-PreF vaccine. All RSV-PreF vaccine formulations and the placebo had similar reactogenicity profiles. No serious adverse events were considered to be related to the RSV-PreF vaccine. CONCLUSIONS: The 3 formulations of the investigational RSV-PreF vaccine were well-tolerated and induced RSV-A and RSV-B neutralizing antibodies and PCAs in healthy, nonpregnant women. CLINICAL TRIALS REGISTRATION: NCT02956837.


Subject(s)
Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/adverse effects , Respiratory Syncytial Virus Vaccines/immunology , Viral Fusion Proteins/immunology , Adolescent , Adult , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Healthy Volunteers , Humans , Injections, Intramuscular , Middle Aged , Placebos/administration & dosage , Respiratory Syncytial Virus Vaccines/administration & dosage , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/adverse effects , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/immunology , Young Adult
6.
Vaccine ; 37(20): 2694-2703, 2019 05 06.
Article in English | MEDLINE | ID: mdl-30987852

ABSTRACT

INTRODUCTION: Respiratory syncytial virus (RSV) is a leading cause of acute lower respiratory tract infections in infants. An investigational vaccine using an engineered recombinant RSV fusion glycoprotein in its post-fusion conformation (RSV F subunit vaccine) has been developed to protect young infants via maternal immunization. This first-in-human, phase I, observer-blind study (NCT02298179) evaluated the safety and immunogenicity of different dosages and formulations of RSV F subunit vaccine in healthy non-pregnant women and men aged 18-45 years. METHODS: Participants were enrolled (1:1:1) in a stepwise dosage-escalation manner into three cohorts to receive RSV F subunit vaccine containing 45 µg, 90 µg and 135 µg of RSV F glycoprotein. Within each cohort, participants were randomized (1:1:1:1) to receive two doses of RSV F subunit vaccine with (aluminum hydroxide or MF59) or without adjuvant, or placebo, ≥28 days apart. Safety (until day 365 post-dose 2), anti-RSV neutralizing antibodies (NAbs) and serum total binding antibodies to RSV F protein (until day 181 post-dose 1) were evaluated. RESULTS: All formulations were well-tolerated. No vaccine-related serious adverse events were reported. All participants were seropositive for anti-RSV NAbs at baseline, with geometric mean titers (GMTs) ranging from 184 (95% confidence interval [CI]: 127-266) to 380 (95% CI: 272-531). At 28 days post-dose 1, anti-RSV NAb GMTs in vaccine recipients ranged from 893 (95% CI: 702-1,136) to 1,602 (95% CI: 1,243-2,064). No booster effect was observed, but immune responses were maintained above pre-vaccination levels for six months post-dose 1. Ratios of RSV F total binding antibodies fold changes to NAb fold changes ranged from 2.79 to 4.12 at 28 days post-dose 1. The impact of the adjuvant was limited. CONCLUSIONS: A single dose of each formulation of RSV subunit F vaccine was well-tolerated and enhanced preexisting NAb titers through six months of follow-up.


Subject(s)
Immunogenicity, Vaccine , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus, Human/immunology , Vaccines, Subunit/immunology , Viral Fusion Proteins/immunology , Viral Vaccines/immunology , Adolescent , Adult , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Belgium/epidemiology , Female , Humans , Incidence , Male , Middle Aged , Time Factors , Vaccination , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/adverse effects , Viral Vaccines/administration & dosage , Viral Vaccines/adverse effects , Young Adult
7.
Nat Commun ; 8(1): 1085, 2017 10 20.
Article in English | MEDLINE | ID: mdl-29057917

ABSTRACT

Human respiratory syncytial virus (hRSV) is responsible for serious lower respiratory tract disease in infants and in older adults, and remains an important vaccine need. RSV fusion (F) glycoprotein is a key target for neutralizing antibodies. RSV F stabilized in its pre-fusion conformation (DS-Cav1 F) induces high neutralizing antibody titers in naïve animals, but it remains unknown to what extent pre-fusion F can boost pre-existing neutralizing responses in RSV seropositive adults. We here assess DS-Cav1 F immunogenicity in seropositive cattle pre-exposed to bovine RSV, a virus closely related to hRSV. A single immunization with non-adjuvanted DS-Cav1 F strongly boosts RSV neutralizing responses, directed towards pre-fusion F-specific epitopes, whereas a post-fusion F is unable to do so. Vaccination with pre-fusion F thus represents a promising strategy for maternal immunization and for other RSV vaccine target populations such as older adults.


Subject(s)
Antibodies, Neutralizing/immunology , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Viral Fusion Proteins/immunology , Animals , CHO Cells , Cattle , Cricetulus , Enzyme-Linked Immunosorbent Assay , Female , Humans , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control
8.
J Virol ; 88(5): 2927-40, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24371063

ABSTRACT

Vesicular stomatitis virus (VSV) is an oncolytic virus that induces cancer cell death through activation of the apoptotic pathway. Intrinsic resistance to oncolysis is found in some cell lines and many primary tumors as a consequence of residual innate immunity to VSV. In resistant-tumor models, VSV oncolytic potential can be reversibly stimulated by combination with epigenetic modulators, such as the histone deacetylase inhibitor vorinostat. Based on this reversible effect of vorinostat, we reasoned that critical host genes involved in oncolysis may likewise be reversibly regulated by vorinostat. A transcriptome analysis in prostate cancer PC3 cells identified a subset of NF-κB target genes reversibly regulated by vorinostat, as well as a group of interferon (IFN)-stimulated genes (ISGs). Consistent with the induction of NF-κB target genes, vorinostat-mediated enhancement of VSV oncolysis increased hyperacetylation of NF-κB RELA/p65. Additional bioinformatics analysis revealed that NF-κB signaling also increased the expression of several autophagy-related genes. Kinetically, autophagy preceded apoptosis, and apoptosis was observed only when cells were treated with both VSV and vorinostat. VSV replication and cell killing were suppressed when NF-κB signaling was inhibited using pharmacological or genetic approaches. Inhibition of autophagy by 3-methyladenine (3-MA) enhanced expression of ISGs, and either 3-MA treatment or genetic ablation of the autophagic marker Atg5 decreased VSV replication and oncolysis. Together, these data demonstrate that vorinostat stimulates NF-κB activity in a reversible manner via modulation of RELA/p65 signaling, leading to induction of autophagy, suppression of the IFN-mediated response, and subsequent enhancement of VSV replication and apoptosis.


Subject(s)
Autophagy , Histone Deacetylase Inhibitors/pharmacology , NF-kappa B/metabolism , Oncolytic Viruses/drug effects , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Vesicular stomatitis Indiana virus/drug effects , Acetylation , Animals , Autophagy/drug effects , Cell Line, Tumor , Chromatin/metabolism , Cluster Analysis , Gene Knockdown Techniques , Humans , Hydroxamic Acids/pharmacology , Male , Mice , NF-kappa B/antagonists & inhibitors , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Prostatic Neoplasms/therapy , Protein Binding , Protein Transport/drug effects , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Transcriptome , Vesicular stomatitis Indiana virus/genetics , Virus Replication , Vorinostat
9.
Cell Res ; 21(6): 895-910, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21200404

ABSTRACT

Recognition of viral RNA structures by the cytosolic sensor retinoic acid-inducible gene-I (RIG-I) results in the activation of signaling cascades that culminate with the generation of the type I interferon (IFN) antiviral response. Onset of antiviral and inflammatory responses to viral pathogens necessitates the regulated spatiotemporal recruitment of signaling adapters, kinases and transcriptional proteins to the mitochondrial antiviral signaling protein (MAVS). We previously demonstrated that the serine/threonine kinase IKKε is recruited to the C-terminal region of MAVS following Sendai or vesicular stomatitis virus (VSV) infection, mediated by Lys63-linked polyubiquitination of MAVS at Lys500, resulting in inhibition of downstream IFN signaling (Paz et al, Mol Cell Biol, 2009). In this study, we demonstrate that C-terminus of MAVS harbors a novel TRAF3-binding site in the aa450-468 region of MAVS. A consensus TRAF-interacting motif (TIM), 455-PEENEY-460, within this site is required for TRAF3 binding and activation of IFN antiviral response genes, whereas mutation of the TIM eliminates TRAF3 binding and the downstream IFN response. Reconstitution of MAVS(-/-) mouse embryo fibroblasts with a construct expressing a TIM-mutated version of MAVS failed to restore the antiviral response or block VSV replication, whereas wild-type MAVS reconstituted antiviral inhibition of VSV replication. Furthermore, recruitment of IKKε to an adjacent C-terminal site (aa 468-540) in MAVS via Lys500 ubiquitination decreased TRAF3 binding and protein stability, thus contributing to IKKε-mediated shutdown of the IFN response. This study demonstrates that MAVS harbors a functional C-terminal TRAF3-binding site that participates in positive and negative regulation of the IFN antiviral response.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Feedback, Physiological , Immunity, Innate , Interferon Type I/metabolism , TNF Receptor-Associated Factor 3/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Gene Knockout Techniques , Humans , I-kappa B Kinase/metabolism , Interferon Type I/immunology , Mice , Mutagenesis, Site-Directed , Mutation, Missense , Peptide Fragments/metabolism , Protein Binding , Protein Stability , Protein Structure, Tertiary , Respirovirus Infections/immunology , Sendai virus/immunology , TNF Receptor-Associated Factor 3/immunology , Vesicular Stomatitis/immunology , Vesiculovirus/immunology
10.
PLoS Pathog ; 6(11): e1001177, 2010 Nov 04.
Article in English | MEDLINE | ID: mdl-21079688

ABSTRACT

Human T cell leukemia virus type 1 (HTLV-1) is the etiologic agent of Adult T cell Leukemia (ATL) and the neurological disorder HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). Although the majority of HTLV-1-infected individuals remain asymptomatic carriers (AC) during their lifetime, 2-5% will develop either ATL or HAM/TSP, but never both. To better understand the gene expression changes in HTLV-1-associated diseases, we examined the mRNA profiles of CD4+ T cells isolated from 7 ATL, 12 HAM/TSP, 11 AC and 8 non-infected controls. Using genomic approaches followed by bioinformatic analysis, we identified gene expression pattern characteristic of HTLV-1 infected individuals and particular disease states. Of particular interest, the suppressor of cytokine signaling 1--SOCS1--was upregulated in HAM/TSP and AC patients but not in ATL. Moreover, SOCS1 was positively correlated with the expression of HTLV-1 mRNA in HAM/TSP patient samples. In primary PBMCs transfected with a HTLV-1 proviral clone and in HTLV-1-transformed MT-2 cells, HTLV-1 replication correlated with induction of SOCS1 and inhibition of IFN-α/ß and IFN-stimulated gene expression. Targeting SOCS1 with siRNA restored type I IFN production and reduced HTLV-1 replication in MT-2 cells. Conversely, exogenous expression of SOCS1 resulted in enhanced HTLV-1 mRNA synthesis. In addition to inhibiting signaling downstream of the IFN receptor, SOCS1 inhibited IFN-ß production by targeting IRF3 for ubiquitination and proteasomal degradation. These observations identify a novel SOCS1 driven mechanism of evasion of the type I IFN antiviral response against HTLV-1.


Subject(s)
Antiviral Agents/pharmacology , Biomarkers, Tumor/genetics , CD4-Positive T-Lymphocytes/physiology , Human T-lymphotropic virus 1/drug effects , Interferon Type I/pharmacology , Signal Transduction/drug effects , Suppressor of Cytokine Signaling Proteins/metabolism , Adult , Biomarkers, Tumor/metabolism , Blotting, Western , CD4-Positive T-Lymphocytes/drug effects , Case-Control Studies , Cells, Cultured , Gene Expression Profiling , HTLV-I Infections/genetics , HTLV-I Infections/metabolism , HTLV-I Infections/virology , Human T-lymphotropic virus 1/genetics , Humans , Immunoblotting , Immunoprecipitation , Oligonucleotide Array Sequence Analysis , Paraparesis, Tropical Spastic/genetics , Paraparesis, Tropical Spastic/metabolism , Paraparesis, Tropical Spastic/virology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/antagonists & inhibitors , Suppressor of Cytokine Signaling Proteins/genetics , Viral Load
11.
Mol Ther ; 18(12): 2094-103, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20842105

ABSTRACT

In chronic lymphocytic leukemia (CLL), overexpression of antiapoptotic B-cell leukemia/lymphoma 2 (BCL-2) family members contributes to leukemogenesis by interfering with apoptosis; BCL-2 expression also impairs vesicular stomatitis virus (VSV)-mediated oncolysis of primary CLL cells. In the effort to reverse resistance to VSV-mediated oncolysis, we combined VSV with obatoclax (GX15-070)-a small-molecule BCL-2 inhibitor currently in phase 2 clinical trials-and examined the molecular mechanisms governing the in vitro and in vivo antitumor efficiency of combining the two agents. In combination with VSV, obatoclax synergistically induced cell death in primary CLL samples and reduced tumor growth in severe combined immunodeficient (SCID) mice-bearing A20 lymphoma tumors. Mechanistically, the combination stimulated the mitochondrial apoptotic pathway, as reflected by caspase-3 and -9 cleavage, cytochrome c release and BAX translocation. Combination treatment triggered the release of BAX from BCL-2 and myeloid cell leukemia-1 (MCL-1) from BAK, whereas VSV infection induced NOXA expression and increased the formation of a novel BAX-NOXA heterodimer. Finally, NOXA was identified as an important inducer of VSV-obatoclax driven apoptosis via knockdown and overexpression of NOXA. These studies offer insight into the synergy between small-molecule BCL-2 inhibitors such as obatoclax and VSV as a combination strategy to overcome apoptosis resistance in CLL.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Pyrroles/pharmacology , Vesiculovirus , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Female , Genetic Therapy , Humans , Indoles , Mice , Mice, SCID , Reverse Transcriptase Polymerase Chain Reaction , Vesiculovirus/physiology
12.
Cytokine Growth Factor Rev ; 21(2-3): 153-9, 2010.
Article in English | MEDLINE | ID: mdl-20395162

ABSTRACT

Oncolytic viruses (OVs) have shown promise as cancer therapeutics in pre-clinical and clinical testing; however, it is unlikely that OVs will constitute a stand-alone treatment. Histone deacetylase inhibitors (HDIs) represent a class of anticancer agents known to influence epigenetic modifications of chromatin, alter gene expression and manipulate a variety of signaling pathways, in some cases blunting the cellular antiviral response. Recent studies have shown that combining OV therapy with HDI treatment enhances viral replication and synergistically induces the killing of cancer cells in vitro and in vivo, an effect that has now been demonstrated in variety of virus/HDI combinations. This review discusses the results obtained with the different OV/HDI combinations, the rationale supporting these combinations and the advantages for oncolytic virus therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Clinical Trials as Topic , Combined Modality Therapy , Epigenesis, Genetic , Humans , Immunity, Innate , Neoplasms/genetics , Virus Replication
13.
J Biol Chem ; 285(25): 19434-49, 2010 Jun 18.
Article in English | MEDLINE | ID: mdl-20413592

ABSTRACT

Bovine leukemia virus (BLV) proviral latency represents a viral strategy to escape the host immune system and allow tumor development. Besides the previously demonstrated role of histone deacetylation in the epigenetic repression of BLV expression, we showed here that BLV promoter activity was induced by several DNA methylation inhibitors (such as 5-aza-2'-deoxycytidine) and that overexpressed DNMT1 and DNMT3A, but not DNMT3B, down-regulated BLV promoter activity. Importantly, cytosine hypermethylation in the 5'-long terminal repeat (LTR) U3 and R regions was associated with true latency in the lymphoma-derived B-cell line L267 but not with defective latency in YR2 cells. Moreover, the virus-encoded transactivator Tax(BLV) decreased DNA methyltransferase expression levels, which could explain the lower level of cytosine methylation observed in the L267(LTaxSN) 5'-LTR compared with the L267 5'-LTR. Interestingly, DNA methylation inhibitors and Tax(BLV) synergistically activated BLV promoter transcriptional activity in a cAMP-responsive element (CRE)-dependent manner. Mechanistically, methylation at the -154 or -129 CpG position (relative to the transcription start site) impaired in vitro binding of CRE-binding protein (CREB) transcription factors to their respective CRE sites. Methylation at -129 CpG alone was sufficient to decrease BLV promoter-driven reporter gene expression by 2-fold. We demonstrated in vivo the recruitment of CREB/CRE modulator (CREM) and to a lesser extent activating transcription factor-1 (ATF-1) to the hypomethylated CRE region of the YR2 5'-LTR, whereas we detected no CREB/CREM/ATF recruitment to the hypermethylated corresponding region in the L267 cells. Altogether, these findings suggest that site-specific DNA methylation of the BLV promoter represses viral transcription by directly inhibiting transcription factor binding, thereby contributing to true proviral latency.


Subject(s)
Activating Transcription Factor 1/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Cyclic AMP Response Element Modulator/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cytosine/metabolism , DNA Methylation , DNA/genetics , Leukemia Virus, Bovine/genetics , Lymphoma/metabolism , Promoter Regions, Genetic , Chromatin/chemistry , Cyclic AMP/metabolism , Cytosine/chemistry , Enzyme-Linked Immunosorbent Assay/methods , Humans , Plasmids/metabolism , Sulfites/chemistry
14.
Cell Microbiol ; 11(6): 889-97, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19388908

ABSTRACT

Oncolytic viruses (OVs) represent an exciting new biological approach to cancer therapy. In particular, RNA viruses have emerged as potent agents for oncolytic virotherapy because of their capacity to specifically target and destroy tumour cells while sparing normal cells and tissues. Several barriers remain in the development of OV therapy, including poor penetration into the tumour mass, inefficient virus replication in primary cancers, and tumour-specific resistance to OV-mediated killing. The combination of OVs with cytotoxic agents, such as small molecule inhibitors of signalling or immunomodulators, as well as stealth delivery of therapeutic viruses have shown promise as novel experimental strategies to overcome resistance to viral oncolysis. These agents complement OV therapy by unblocking host pathways, delivering viruses with greater efficiency and/or increasing virus proliferation at the tumour site. In this review, we summarize recent development of these concepts, the potential obstacles, and future prospects for the clinical utilization of RNA OVs in cancer therapy.


Subject(s)
Neoplasms/therapy , Oncolytic Virotherapy/methods , RNA Viruses/growth & development , Antineoplastic Agents/therapeutic use , Drug Therapy, Combination , Humans
15.
Mol Cell Biol ; 29(12): 3401-12, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19380491

ABSTRACT

Induction of the antiviral interferon response is initiated upon recognition of viral RNA structures by the RIG-I or Mda-5 DEX(D/H) helicases. A complex signaling cascade then converges at the mitochondrial adapter MAVS, culminating in the activation of the IRF and NF-kappaB transcription factors and the induction of interferon gene expression. We have previously shown that MAVS recruits IkappaB kinase epsilon (IKKepsilon) but not TBK-1 to the mitochondria following viral infection. Here we map the interaction of MAVS and IKKepsilon to the C-terminal region of MAVS and demonstrate that this interaction is ubiquitin dependent. MAVS is ubiquitinated following Sendai virus infection, and K63-linked ubiquitination of lysine 500 (K500) of MAVS mediates recruitment of IKKepsilon to the mitochondria. Real-time PCR analysis reveals that a K500R mutant of MAVS increases the mRNA level of several interferon-stimulated genes and correlates with increased NF-kappaB activation. Thus, recruitment of IKKepsilon to the mitochondria upon MAVS K500 ubiquitination plays a modulatory role in the cascade leading to NF-kappaB activation and expression of inflammatory and antiviral genes. These results provide further support for the differential role of IKKepsilon and TBK-1 in the RIG-I/Mda5 pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , I-kappa B Kinase/metabolism , Ubiquitin/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Animals , COS Cells , Cell Line , Chlorocebus aethiops , HeLa Cells , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/genetics , Interferon-beta/metabolism , Lysine/chemistry , Mitochondria/metabolism , Mutagenesis, Site-Directed , NF-kappa B/metabolism , Protein Interaction Mapping , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sendai virus/pathogenicity , Signal Transduction
16.
Proc Natl Acad Sci U S A ; 105(39): 14981-6, 2008 Sep 30.
Article in English | MEDLINE | ID: mdl-18815361

ABSTRACT

Intratumoral innate immunity can play a significant role in blocking the effective therapeutic spread of a number of oncolytic viruses (OVs). Histone deacetylase inhibitors (HDIs) are known to influence epigenetic modifications of chromatin and can blunt the cellular antiviral response. We reasoned that pretreatment of tumors with HDIs could enhance the replication and spread of OVs within malignancies. Here, we show that HDIs markedly enhance the spread of vesicular stomatitis virus (VSV) in a variety of cancer cells in vitro, in primary tumor tissue explants and in multiple animal models. This increased oncolytic activity correlated with a dampening of cellular IFN responses and augmentation of virus-induced apoptosis. These results illustrate the general utility of HDIs as chemical switches to regulate cellular innate antiviral responses and to provide controlled growth of therapeutic viruses within malignancies. HDIs could have a profoundly positive impact on the clinical implementation of OV therapeutics.


Subject(s)
Enzyme Inhibitors/therapeutic use , Histone Deacetylase Inhibitors , Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/drug effects , Animals , Benzamides/therapeutic use , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Immunity, Innate/drug effects , Interferons/administration & dosage , Male , Mice , Mice, Inbred Strains , Neoplasms/drug therapy , Neoplasms/virology , Oncolytic Viruses/immunology , Oncolytic Viruses/physiology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/therapy , Prostatic Neoplasms/virology , Pyridines/therapeutic use , Vesiculovirus/drug effects , Vesiculovirus/immunology , Vesiculovirus/physiology , Virus Replication/drug effects , Xenograft Model Antitumor Assays
17.
J Biol Chem ; 282(29): 20854-67, 2007 Jul 20.
Article in English | MEDLINE | ID: mdl-17526487

ABSTRACT

Bovine leukemia virus (BLV) expression is controlled at the transcriptional level through three Tax(BLV)-responsive elements (TxREs) responsive to the viral transactivator Tax(BLV). The cAMP-responsive element (CRE)-binding protein (CREB) has been shown to interact with CRE-like sequences present in the middle of each of these TxREs and to play critical transcriptional roles in both basal and Tax(BLV)-transactivated BLV promoter activity. In this study, we have investigated the potential involvement of the cAMP-response element modulator (CREM) in BLV transcriptional regulation, and we have demonstrated that CREM proteins were expressed in BLV-infected cells and bound to the three BLV TxREs in vitro. Chromatin immunoprecipitation assays using BLV-infected cell lines demonstrated in the context of chromatin that CREM proteins were recruited to the BLV promoter TxRE region in vivo. Functional studies, in the absence of Tax(BLV), indicated that ectopic CREMtau protein had a CRE-dependent stimulatory effect on BLV promoter transcriptional activity. Cross-link of the B-cell receptor potentiated CREMtau transactivation of the viral promoter. Further experiments supported the notion that this potentiation involved CREMtau Ser-117 phosphorylation and recruitment of CBP/p300 to the BLV promoter. Although CREB and Tax(BLV) synergistically transactivated the BLV promoter, CREMtau repressed this Tax(BLV)/CREB synergism, suggesting that a modulation of the level of Tax(BLV) transactivation through opposite actions of CREB and CREMtau could facilitate immune escape and allow tumor development.


Subject(s)
Cyclic AMP Response Element Modulator/genetics , Leukemia Virus, Bovine/genetics , Transcription, Genetic , Animals , Cell Nucleus/metabolism , Cloning, Molecular , Cross-Linking Reagents/pharmacology , Cyclic AMP/metabolism , Cyclic AMP Response Element Modulator/chemistry , Gene Products, tax/metabolism , Leukemia Virus, Bovine/metabolism , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Phosphorylation , Promoter Regions, Genetic , Protein Isoforms
18.
J Virol ; 78(24): 13848-64, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15564493

ABSTRACT

Bovine leukemia virus (BLV) infection is characterized by viral latency in a large proportion of cells containing an integrated provirus. In this study, we postulated that mechanisms directing the recruitment of deacetylases to the BLV 5' long terminal repeat (LTR) could explain the transcriptional repression of viral expression in vivo. Accordingly, we showed that BLV promoter activity was induced by several deacetylase inhibitors (such as trichostatin A [TSA]) in the context of episomal LTR constructs and in the context of an integrated BLV provirus. Moreover, treatment of BLV-infected cells with TSA increased H4 acetylation at the viral promoter, showing a close correlation between the level of histone acetylation and transcriptional activation of the BLV LTR. Among the known cis-regulatory DNA elements located in the 5' LTR, three E box motifs overlapping cyclic AMP responsive elements (CREs) in U3 were shown to be involved in transcriptional repression of BLV basal gene expression. Importantly, the combined mutations of these three E box motifs markedly reduced the inducibility of the BLV promoter by TSA. E boxes are susceptible to recognition by transcriptional repressors such as Max-Mad-mSin3 complexes that repress transcription by recruiting deacetylases. However, our in vitro binding studies failed to reveal the presence of Mad-Max proteins in the BLV LTR E box-specific complexes. Remarkably, TSA increased the occupancy of the CREs by CREB/ATF. Therefore, we postulated that the E box-specific complexes exerted their negative cooperative effect on BLV transcription by steric hindrance with the activators CREB/ATF and/or their transcriptional coactivators possessing acetyltransferase activities. Our results thus suggest that the overlapping CRE and E box elements in the BLV LTR were selected during evolution as a novel strategy for BLV to allow better silencing of viral transcription and to escape from the host immune response.


Subject(s)
Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Viral , Leukemia Virus, Bovine/metabolism , Promoter Regions, Genetic/genetics , Terminal Repeat Sequences/genetics , Transcription, Genetic , 5' Untranslated Regions/genetics , Acetylation , Activating Transcription Factors , Animals , Base Sequence , Blood Proteins/metabolism , Cattle , Cell Line , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Humans , Leukemia Virus, Bovine/genetics , Leukocytes, Mononuclear , Molecular Sequence Data , Sheep , Transcription Factors/metabolism , Virus Latency
19.
J Biol Chem ; 279(33): 35025-36, 2004 Aug 13.
Article in English | MEDLINE | ID: mdl-15163662

ABSTRACT

Efficient bovine leukemia virus (BLV) transcription requires the virus-encoded transactivator Tax(BLV), which acts through three Tax(BLV)-responsive elements located in the 5' long terminal repeat. It has been proposed that the binding of the CRE-binding protein (CREB) and the activating transcription factor (ATF) to the three imperfect cAMP-responsive elements (CREs) located in each Tax(BLV)-responsive element mediates Tax(BLV) transactivation. Here we demonstrated that deacetylase inhibitors (HDACis) synergistically enhanced the transcriptional activation of the BLV promoter by Tax(BLV) in a CRE-dependent manner. Tax(BLV) was acetylated in vivo at its N(alpha) terminus but not at internal lysine residues. Rather, HDACi potentiation of Tax(BLV) transactivation was mediated by an HDACi indirect action that requires new protein synthesis. Mechanistically, using a dominant-negative form of CREB, we showed that Tax(BLV) and HDACi synergistically activated BLV gene expression via a CREB-dependent mechanism. Moreover, electrophoretic mobility shift assay and Western blot experiments revealed that HDACi increased the in vitro DNA binding activity of CREB/ATF but did not alter CREB/ATF intranuclear presence. Remarkably, chromatin immunoprecipitation assays demonstrated that HDACi treatment increased the level of CREB bound to the BLV promoter in vivo. Our results together suggest that an increase in CREB/ATF occupancy of the viral CREs in response to HDACi potentiates Tax(BLV) transactivation of the BLV promoter.


Subject(s)
Cyclic AMP/metabolism , Gene Expression Regulation, Viral , Gene Products, tax/genetics , Hydrolases/antagonists & inhibitors , Leukemia Virus, Bovine/genetics , Animals , Blotting, Western , COS Cells , Carrier Proteins , Cell Line , Cell Nucleus/metabolism , Chromatin/metabolism , Chromosomes/metabolism , Cyclic AMP Receptor Protein/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Genes, Dominant , Genetic Vectors , Humans , Luciferases/metabolism , Mutation , Plasmids/metabolism , Precipitin Tests , Promoter Regions, Genetic , Protein Structure, Tertiary , RNA, Messenger/metabolism , Response Elements , Ribonucleases/metabolism , Transcriptional Activation , Transfection
20.
Mol Cell Biol ; 23(17): 6200-9, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12917341

ABSTRACT

Previous studies have implicated acetylases and deacetylases in regulating the transcriptional activity of NF-kappa B. Here, we show that inhibitors of deacetylases such as trichostatin A (TSA) and sodium butyrate (NaBut) potentiated TNF-induced expression of several natural NF-kappa B-driven promoters. This transcriptional synergism observed between TNF and TSA (or NaBut) required intact kappa B sites in all promoters tested and was biologically relevant as demonstrated by RNase protection on two instances of endogenous NF-kappa B-regulated gene transcription. Importantly, TSA prolonged both TNF-induced DNA-binding activity and the presence of NF-kappa B in the nucleus. We showed that the p65 subunit of NF-kappa B was acetylated in vivo. However, this acetylation was weak, suggesting that other mechanisms could be implicated in the potentiated binding and transactivation activities of NF-kappa B after TNF plus TSA versus TNF treatment. Western blot and immunofluorescence confocal microscopy experiments revealed a delay in the cytoplasmic reappearance of the I kappa B alpha inhibitor that correlated temporally with the prolonged intranuclear binding and presence of NF-kappa B. This delay was due neither to a defect in I kappa B alpha mRNA production nor to a nuclear retention of I kappa B alpha but was rather due to a persistent proteasome-mediated degradation of I kappa B alpha. A prolongation of I kappa B kinase activity could explain, at least partially, the delayed I kappa B alpha cytoplasmic reappearance observed in presence of TNF plus TSA.


Subject(s)
Cytoplasm/metabolism , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , I-kappa B Proteins/metabolism , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Butyric Acid/pharmacology , Cysteine Endopeptidases , HeLa Cells , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , I-kappa B Kinase , I-kappa B Proteins/drug effects , I-kappa B Proteins/genetics , Leupeptins/pharmacology , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Multienzyme Complexes/antagonists & inhibitors , NF-KappaB Inhibitor alpha , NF-kappa B/drug effects , NF-kappa B/genetics , Proteasome Endopeptidase Complex , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Transport/drug effects , Transcription Factor RelA
SELECTION OF CITATIONS
SEARCH DETAIL
...