Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Pathog ; 18(7): e1010688, 2022 07.
Article in English | MEDLINE | ID: mdl-35793357

ABSTRACT

Herpes simplex virus type 1 (HSV-1) is a common virus of mankind and HSV-1 infections are a significant cause of blindness. The current antiviral treatment of herpes infection relies on acyclovir and related compounds. However, acyclovir resistance emerges especially in the long term prophylactic treatment that is required for prevention of recurrent herpes keratitis. Earlier we have established antiviral siRNA swarms, targeting sequences of essential genes of HSV, as effective means of silencing the replication of HSV in vitro or in vivo. In this study, we show the antiviral efficacy of 2´-fluoro modified antiviral siRNA swarms against HSV-1 in human corneal epithelial cells (HCE). We studied HCE for innate immunity responses to HSV-1, to immunostimulatory cytotoxic double stranded RNA, and to the antiviral siRNA swarms, with or without a viral challenge. The panel of studied innate responses included interferon beta, lambda 1, interferon stimulated gene 54, human myxovirus resistance protein A, human myxovirus resistance protein B, toll-like receptor 3 and interferon kappa. Our results demonstrated that HCE cells are a suitable model to study antiviral RNAi efficacy and safety in vitro. In HCE cells, the antiviral siRNA swarms targeting the HSV UL29 gene and harboring 2´-fluoro modifications, were well tolerated, induced only modest innate immunity responses, and were highly antiviral with more than 99% inhibition of viral release. The antiviral effect of the 2'-fluoro modified swarm was more apparent than that of the unmodified antiviral siRNA swarm. Our results encourage further research in vitro and in vivo on antiviral siRNA swarm therapy of corneal HSV infection, especially with modified siRNA swarms.


Subject(s)
Herpes Simplex , Herpesvirus 1, Human , Acyclovir/metabolism , Acyclovir/pharmacology , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Epithelial Cells/metabolism , Herpes Simplex/genetics , Herpes Simplex/therapy , Herpesvirus 1, Human/physiology , Humans , RNA, Double-Stranded/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Virus Replication/genetics
2.
Mol Cell ; 55(1): 111-22, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24954902

ABSTRACT

DNA damage associated with viral DNA synthesis can result in double-strand breaks that threaten genome integrity and must be repaired. Here, we establish that the cellular Fanconi anemia (FA) genomic stability pathway is exploited by herpes simplex virus 1 (HSV-1) to promote viral DNA synthesis and enable its productive growth. Potent FA pathway activation in HSV-1-infected cells resulted in monoubiquitination of FA effector proteins FANCI and FANCD2 (FANCI-D2) and required the viral DNA polymerase. FANCD2 relocalized to viral replication compartments, and FANCI-D2 interacted with a multisubunit complex containing the virus-encoded single-stranded DNA-binding protein ICP8. Significantly, whereas HSV-1 productive growth was impaired in monoubiquitination-defective FA cells, this restriction was partially surmounted by antagonizing the DNA-dependent protein kinase (DNA-PK), a critical enzyme required for nonhomologous end-joining (NHEJ). This identifies the FA-pathway as a cellular factor required for herpesvirus productive growth and suggests that FA-mediated suppression of NHEJ is a fundamental step in the viral life cycle.


Subject(s)
DNA, Viral/biosynthesis , Fanconi Anemia/genetics , Genomic Instability , Herpesvirus 1, Human/physiology , Animals , Chlorocebus aethiops , DNA Damage , DNA End-Joining Repair , DNA-Directed DNA Polymerase/physiology , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , Herpesvirus 1, Human/genetics , Ubiquitination , Vero Cells , Virus Replication
3.
J Virol ; 93(8)2019 04 15.
Article in English | MEDLINE | ID: mdl-30760568

ABSTRACT

A majority of adults in Finland are seropositive carriers of herpes simplex viruses (HSV). Infection occurs at epithelial or mucosal surfaces, after which virions enter innervating nerve endings, eventually establishing lifelong infection in neurons of the sensory or autonomic nervous system. Recent data have highlighted the genetic diversity of HSV-1 strains and demonstrated apparent geographic patterns in strain similarity. Though multiple HSV-1 genomes have been sequenced from Europe to date, there is a lack of sequenced genomes from the Nordic countries. Finland's history includes at least two major waves of human migration, suggesting the potential for diverse viruses to persist in the population. Here, we used HSV-1 clinical isolates from Finland to test the relationship between viral phylogeny, genetic variation, and phenotypic characteristics. We found that Finnish HSV-1 isolates separated into two distinct phylogenetic groups, potentially reflecting historical waves of human (and viral) migration into Finland. Each HSV-1 isolate harbored a distinct set of phenotypes in cell culture, including differences in the amount of virus production, extracellular virus release, and cell-type-specific fitness. Importantly, the phylogenetic clusters were not predictive of any detectable pattern in phenotypic differences, demonstrating that whole-genome relatedness is not a proxy for overall viral phenotype. Instead, we highlight specific gene-level differences that may contribute to observed phenotypic differences, and we note that strains from different phylogenetic groups can contain the same genetic variations.IMPORTANCE Herpes simplex viruses (HSV) infect a majority of adults. Recent data have highlighted the genetic diversity of HSV-1 strains and demonstrated apparent genomic relatedness between strains from the same geographic regions. We used HSV-1 clinical isolates from Finland to test the relationship between viral genomic and geographic relationships, differences in specific genes, and characteristics of viral infection. We found that viral isolates from Finland separated into two distinct groups of genomic and geographic relatedness, potentially reflecting historical patterns of human and viral migration into Finland. These Finnish HSV-1 isolates had distinct infection characteristics in multiple cell types tested, which were specific to each isolate and did not group according to genomic and geographic relatedness. This demonstrates that HSV-1 strain differences in specific characteristics of infection are set by a combination of host cell type and specific viral gene-level differences.


Subject(s)
Genetic Variation , Genome, Viral , Herpes Simplex/genetics , Herpesvirus 1, Human/genetics , Phylogeny , Animals , Chlorocebus aethiops , Female , Finland , Herpesvirus 1, Human/isolation & purification , Humans , Male , Vero Cells , Whole Genome Sequencing
4.
Mol Ther ; 26(9): 2315-2325, 2018 09 05.
Article in English | MEDLINE | ID: mdl-30005865

ABSTRACT

The approval of the first oncolytic virus for the treatment of metastatic melanoma and the compiling evidence that the use of oncolytic viruses can enhance cancer immunotherapies targeted against various immune checkpoint proteins has attracted great interest in the field of cancer virotherapy. We have developed a novel platform for clinically relevant enveloped viruses that can direct the virus-induced immune response against tumor antigens. By physically attaching tumor-specific peptides onto the viral envelope of vaccinia virus and herpes simplex virus 1 (HSV-1), we were able to induce a strong T cell-specific immune response toward these tumor antigens. These therapeutic peptides could be attached onto the viral envelope by using a cell-penetrating peptide sequence derived from human immunodeficiency virus Tat N-terminally fused to the tumor-specific peptides or, alternatively, therapeutic peptides could be conjugated with cholesterol for the attachment of the peptides onto the viral envelope. We used two mouse models of melanoma termed B16.OVA and B16-F10 for testing the efficacy of OVA SIINFEKL-peptide-coated viruses and gp100-Trp2-peptide-coated viruses, respectively, and show that by coating the viral envelope with therapeutic peptides, the anti-tumor immunity and the number of tumor-specific CD8+ T cells in the tumor microenvironment can be significantly enhanced.


Subject(s)
Cancer Vaccines/chemistry , Peptides/metabolism , A549 Cells , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Chlorocebus aethiops , Herpesvirus 1, Human/metabolism , Humans , Melanoma/immunology , Melanoma/therapy , Mice , Oncolytic Virotherapy/methods , Oncolytic Viruses , Peptides/immunology , Vaccinia virus/metabolism , Vero Cells , Viral Envelope Proteins/metabolism
5.
J Med Virol ; 90(3): 564-570, 2018 03.
Article in English | MEDLINE | ID: mdl-28975630

ABSTRACT

Herpes simplex virus (HSV) establishes latency in neurons and recurrent infections in oral mucosa. This prospective study analyzes HSV prevalence in oral mucosal brush samples from men with known human papillomavirus (HPV) status. We hypothesized that HSV-1-infection could facilitate HPV persistence as a cofactor. This study was a part of the Finnish Family HPV study accomplished at the University of Turku/Turku University Hospital, Finland. A total of 139 men (mean age 28.6 ± 4.9 years) were enrolled at 36+-weeks of their partner's pregnancy and thereafter followed-up for 6 years. Altogether, 722 samples, extracted from oral brush samples collected at the enrollment timepoint (baseline) and at 2-, 6-, 12-, 24-, 36-month, and 6 years, were available. HSV DNA was analyzed with quantitative PCR. HSV-1 results were compared with the known HPV data. The prevalence of oral HSV-1 shedding varied between 0-7.2% (mean 2.8%) among the men. Mean copy numbers varied between 4 and 550 genome copies/sample. A total of 18 (12.9%) men were found HSV-1-positive at least once, two of them twice. Neither smoking nor oral sex was associated with the oral HSV-1-DNA finding. HPV/HSV-1 co-infection was found in 6 (4.3%) men, all of them having persistent HPV-infection. In conclusion, HSV-1 and its coinfection with HPV in oral mucosa was rare.


Subject(s)
Coinfection/epidemiology , Herpes Simplex/epidemiology , Mouth Mucosa/virology , Papillomavirus Infections/epidemiology , Adult , Coinfection/virology , DNA, Viral/genetics , Female , Finland/epidemiology , Follow-Up Studies , Genome, Viral , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Humans , Longitudinal Studies , Male , Papillomaviridae/genetics , Papillomavirus Infections/virology , Pregnancy , Prevalence , Prospective Studies , Spouses , Virus Shedding
6.
J Med Virol ; 88(12): 2196-2205, 2016 12.
Article in English | MEDLINE | ID: mdl-27191509

ABSTRACT

Herpes simplex virus (HSV) is a common human pathogen causing severe diseases such as encephalitis, keratitis, and neonatal herpes. There is no vaccine against HSV and the current antiviral chemotherapy fails to treat certain forms of the disease. Here, we evaluated the antiviral activity of enzymatically created small interfering (si)RNA pools against various pathogenic HSV strains as potential candidates for antiviral therapies. Pools of siRNA targeting 0.5-0.8 kbp of essential HSV genes UL54, UL29, or UL27 were enzymatically synthesized. Efficacy of inhibition of each siRNA pool was evaluated against multiple clinical isolates and laboratory wild type HSV-1 strains using three cell lines representing host tissues that support HSV-1 replication: epithelial, ocular, and cells that originated from the nervous system. The siRNA pools targeting UL54, UL29, and UL27, as well as their equimolar mixture, inhibited HSV replication, with the pool targeting UL29 having the most prominent antiviral effect. In contrast, the non-specific control siRNA pool did not have such an effect. Moreover, the UL29 pool elicited only a minimal innate immune response in the HSV-infected cells, thus evidencing the safety of its potential clinical use. These results are promising for the development of a topical RNA interference approach for clinical treatment of HSV infection. J. Med. Virol. 88:2196-2205, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Antiviral Agents/pharmacology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , RNA Interference , RNA, Small Interfering/genetics , Cell Line , Drug Discovery , Herpes Simplex/virology , Humans , Immunity, Innate , Viral Plaque Assay , Virus Replication
7.
Virol J ; 11: 125, 2014 Jul 08.
Article in English | MEDLINE | ID: mdl-25005804

ABSTRACT

BACKGROUND: Oral mucosa is frequently exposed to Herpes simplex virus type 1 (HSV-1) infection and irradiation due to dental radiography. During radiotherapy for oral cancer, the surrounding clinically normal tissues are also irradiated. This prompted us to study the effects of HSV-1 infection and irradiation on viability and apoptosis of oral epithelial cells. METHODS: Immortal gingival keratinocyte (HMK) cells were infected with HSV-1 at a low multiplicity of infection (MOI) and irradiated with 2 Gy 24 hours post infection. The cells were then harvested at 24, 72 and 144 hours post irradiation for viability assays and qRT-PCR analyses for the apoptosis-related genes caspases 3, 8, and 9, bcl-2, NFκB1, and viral gene VP16. Mann-Whitney U-test was used for statistical calculations. RESULTS: Irradiation improved the cell viability at 144 hours post irradiation (P = 0.05), which was further improved by HSV-1 infection at MOI of 0.00001 (P = 0.05). Simultaneously, the combined effects of infection at MOI of 0.0001 and irradiation resulted in upregulation in NFκB1 (P = 0.05). The combined effects of irradiation and HSV infection also significantly downregulated the expression of caspases 3, 8, and 9 at 144 hours (P = 0.05) whereas caspase 3 and 8 significantly upregulated in non-irradiated, HSV-infected cells as compared to uninfected controls (P = 0.05). Infection with 0.0001 MOI downregulated bcl-2 in non-irradiated cells but was upregulated by 27% after irradiation when compared to non-irradiated infected cells (P = 0.05). Irradiation had no effect on HSV-1 shedding or HSV gene expression at 144 hours. CONCLUSIONS: HSV-1 infection may improve the viability of immortal cells after irradiation. The effect might be related to inhibition of apoptosis.


Subject(s)
Gingiva/radiation effects , Gingiva/virology , Herpesvirus 1, Human/physiology , Mouth Mucosa/radiation effects , Mouth Mucosa/virology , Apoptosis/genetics , Caspases/genetics , Cell Line, Transformed , Cell Survival/radiation effects , Gingiva/cytology , Herpes Simplex/virology , Herpes Simplex Virus Protein Vmw65/genetics , Humans , Keratinocytes/radiation effects , Keratinocytes/virology , Mouth Mucosa/cytology , Mouth Neoplasms/radiotherapy , Proto-Oncogene Proteins c-bcl-2/genetics , Radiography, Dental/adverse effects , Radiotherapy/adverse effects , Virus Replication , Virus Shedding
8.
J Biol Chem ; 287(42): 35324-35332, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-22910914

ABSTRACT

Influenza A viruses (IAVs) infect humans and cause significant morbidity and mortality. Different treatment options have been developed; however, these were insufficient during recent IAV outbreaks. Here, we conducted a targeted chemical screen in human nonmalignant cells to validate known and search for novel host-directed antivirals. The screen validated saliphenylhalamide (SaliPhe) and identified two novel anti-IAV agents, obatoclax and gemcitabine. Further experiments demonstrated that Mcl-1 (target of obatoclax) provides a novel host target for IAV treatment. Moreover, we showed that obatoclax and SaliPhe inhibited IAV uptake and gemcitabine suppressed viral RNA transcription and replication. These compounds possess broad spectrum antiviral activity, although their antiviral efficacies were virus-, cell type-, and species-specific. Altogether, our results suggest that phase II obatoclax, investigational SaliPhe, and FDA/EMEA-approved gemcitabine represent potent antiviral agents.


Subject(s)
Amides/pharmacology , Antiviral Agents/pharmacology , Deoxycytidine/analogs & derivatives , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/physiology , Influenza, Human/drug therapy , Pyrroles/pharmacology , Salicylates/pharmacology , Animals , Chlorocebus aethiops , Deoxycytidine/pharmacology , Dogs , Humans , Indoles , Influenza, Human/metabolism , Myeloid Cell Leukemia Sequence 1 Protein , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Viral/biosynthesis , Vero Cells , Virus Replication , Gemcitabine
9.
Duodecim ; 129(1): 31-40, 2013.
Article in Fi | MEDLINE | ID: mdl-23431880

ABSTRACT

Current picture and treatment of herpes simplex virus infections The contraction of primary herpes simplex infection in early childhood is becoming increasingly less frequent. A growing number of new cases of genital herpes are caused by HSV-1. Excretion of virus by carriers of genital herpes often takes place also during the asymptomatic stage and symptomless recurrences of genital herpes may be very brief in duration. Primary infections and reactivations can be treated with anti-herpes medication. Progress in drug treatment has taken place in the treatment of serious infections of the newborn and in the preventive treatment of herpes reactivations, in which fairly large single doses have proven effective.


Subject(s)
Antiviral Agents/therapeutic use , Herpes Simplex/drug therapy , Herpes Simplex/virology , Child, Preschool , Herpes Genitalis/drug therapy , Herpes Genitalis/epidemiology , Herpes Genitalis/virology , Herpes Simplex/epidemiology , Herpesvirus 1, Human , Humans , Infant , Infant, Newborn , Recurrence , Virus Shedding
10.
Microorganisms ; 11(11)2023 Oct 29.
Article in English | MEDLINE | ID: mdl-38004669

ABSTRACT

Herpes simplex virus (HSV) has proven successful in treating human cancer. Since the approval of talimogene laherparepvec (T-VEC) in 2015, HSV has been thoroughly researched to discover novel mechanisms to combat cancer and treat other diseases. Another HSV-based drug, beremagene geperpavec (B-VEC), received approval in 2023 to treat the rare genetic disease dystrophic epidermolysis bullosa, and was also the first clinically approved HSV vector carrying an extracellular matrix (ECM)-modifying transgene. The ECM is a network of macromolecules surrounding cells, which provides support and regulates cell growth and differentiation, the disruption of which is common in cancer. The naked mole rat (NMR) has a thick ECM and a unique mutation in the hyaluronan synthase 2 (HAS2) gene, which has been linked to the high cancer resistance of the species. To study the effect of this mutation in human cancer, we have developed an attenuated, replication-competent HSV vector expressing the NMR-HAS2 gene. The viral replication, transgene expression and cytotoxic effect of the novel vector was studied in glioma cells. Our results show that an attenuated, replication-competent HSV vector expressing a foreign ECM-modifying transgene, namely HAS2, provides an effective tool to study and combat cancer in humans.

12.
Microorganisms ; 10(8)2022 Jul 26.
Article in English | MEDLINE | ID: mdl-35893566

ABSTRACT

The aim was to evaluate the herpes simplex virus (HSV) seroprevalence and seroconversion among 285 pregnant women and their 120 male spouses in Finland during a six-year follow-up (FU) between 1998-2008. We also studied the effect of sexual habits, pregnancy, and other demographic factors on the acquisition of HSV infection. Combined HSV-1 and HSV-2-IgG antibodies were assessed in the first baseline serum samples with an indirect enzyme immunoassay method. The individuals with seronegative or borderline HSV serology at baseline were additionally tested using their latest FU serum sample available. The overall HSV seroprevalence during the FU was 58.9% (168/285) among the women and 53.3% (64/120) among their spouses. The seroconversion rate was 11.4% (15/132) and 12.5% (8/64) among women and their spouses, respectively. Both spouses were HSV seropositive in 39.2% (47/120). To determine the HSV-2 seroprevalence, we also tested all HSV-seropositive participants using HSV-2-specific antigen. HSV-2 seropositivity was detected in 10.9% (44/405) of the participants. The age (p = 0.006) and history of genital warts (p = 0.006) of the women were associated with combined HSV-1 and/or HSV-2 seropositivity, while a younger age was related to HSV seroconversion (p = 0.023). Among the male spouses, HSV seropositivity was associated with the practice of oral sex (p = 0.033). To conclude, women of childbearing age acquire primary HSV infections and the presence of HSV in oral epithelium is common among HSV-seropositive individuals.

13.
Pharmaceuticals (Basel) ; 15(2)2022 Feb 21.
Article in English | MEDLINE | ID: mdl-35215370

ABSTRACT

RNA molecules provide promising new possibilities for the prevention and treatment of viral infections and diseases. The rapid development of RNA biology and medicine requires advanced methods for the purification of RNA molecules, which allow fast and efficient RNA processing, preferably under non-denaturing conditions. Asymmetrical flow field-flow fractionation (AF4) enables gentle separation and purification of macromolecules based on their diffusion coefficients. The aim of the study was to develop an AF4 method for efficient purification of enzymatically produced antiviral small interfering (si)RNA molecules and to evaluate the overall potential of AF4 in the separation of short single-stranded (ss) and double-stranded (ds) RNA molecules. We show that AF4 separates monomeric ssRNA from dsRNA molecules of the same size and monomeric ssRNA from multimeric forms of the same ssRNA. The developed AF4 method enabled the separation of enzymatically produced 27-nt siRNAs from partially digested substrate dsRNA, which is potentially toxic for mammalian cells. The recovery of AF4-purified enzymatically produced siRNA molecules was about 70%, which is about 20% higher than obtained using anion-exchange chromatography. The AF4-purified siRNAs were not toxic for mammalian cells and fully retained their biological activity as confirmed by efficient inhibition of herpes simplex virus 1 replication in cell culture. Our work is the first to develop AF4 methods for the separation of short RNA molecules.

14.
Viruses ; 14(6)2022 06 13.
Article in English | MEDLINE | ID: mdl-35746761

ABSTRACT

Herpes simplex virus type 1 (HSV-1) is the only FDA- and EMA- approved oncolytic virus, and accordingly, many potential oncolytic HSVs (oHSV) are in clinical development. The utilized oHSV parental strains are, however, mostly based on laboratory reference strains, which may possess a compromised cytolytic capacity in contrast to circulating strains of HSV-1. Here, we assess the phenotype of thirty-six circulating HSV-1 strains from Finland to uncover their potential as oHSV backbones. First, we determined their capacity for cell-to-cell versus extracellular spread, to find strains with replication profiles favorable for each application. Second, to unfold the differences, we studied the genetic diversity of two relevant viral glycoproteins (gB/UL27, gI/US7). Third, we examined the oncolytic potential of the strains in cells representing glioma, lymphoma, and colorectal adenocarcinoma. Our results suggest that the phenotype of a circulating isolate, including the oncolytic potential, is highly related to the host cell type. Nevertheless, we identified isolates with increased oncolytic potential in comparison with the reference viruses across many or all of the studied cancer cell types. Our research emphasizes the need for careful selection of the backbone virus in early vector design, and it highlights the potential of clinical isolates as backbones in oHSV development.


Subject(s)
Herpes Simplex , Herpesvirus 1, Human , Oncolytic Virotherapy , Oncolytic Viruses , Finland , Herpes Simplex/genetics , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/metabolism , Humans , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics
15.
J Gen Virol ; 92(Pt 1): 173-80, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20881085

ABSTRACT

We have studied cell death and its mechanisms in herpes simplex virus type 1 (HSV-1)-infected monocytic cells. The HSV-1 ICP4 and Us3 deletion mutant, d120 caused both apoptosis and necroptosis in d120-infected monocytic cells. At a late time point of infection the number of apoptotic cells was increased significantly in d120-infected cells when compared with uninfected or parental HSV-1 (KOS)-infected cells. Necroptosis inhibitor treatment increased the number of viable cells among the d120-infected cells, indicating that cell death in d120-infected cells was, in part, because of necroptosis. Moreover, lysosomal membrane permeabilization and cathepsin B and H activities were increased significantly in d120-infected cells. Inhibition of cathepsin B and S activities with specific cathepsin inhibitors led to increased cell viability, and inhibition of cathepsin L activity resulted in a decreased number of apoptotic cells. This indicates that cathepsins B, L and S may act as cell-death mediators in d120-infected monocytic cells. In addition, caspase 3 activity was increased significantly in d120-infected cells. However, the caspase 3 inhibitor treatment did not decrease the number of apoptotic cells. In contrast, inhibition of cathepsin L activity by cathepsin L-specific inhibitor clearly decreased caspase 3 activity and the number of apoptotic cells in d120-infected cells. This might suggest that, in d120-infected monocytic cells, cathepsin L activates caspase 3 and thus mediates d120-induced apoptosis. Taken together, these findings suggest that d120-induced cell death is both apoptotic and necroptotic.


Subject(s)
Cathepsins/metabolism , Cell Death , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/pathogenicity , Immediate-Early Proteins/deficiency , Monocytes/virology , Protein Serine-Threonine Kinases/deficiency , Caspase 3/metabolism , Cell Line , Cell Survival , Gene Deletion , Humans , Lysosomes/physiology , Viral Proteins
16.
Antiviral Res ; 182: 104916, 2020 10.
Article in English | MEDLINE | ID: mdl-32798603

ABSTRACT

Chemical modifications of small interfering (si)RNAs are used to enhance their stability and potency, and to reduce possible off-target effects, including immunogenicity. We have earlier introduced highly effective antiviral siRNA swarms against herpes simplex virus (HSV), targeting 653 bp of the essential UL29 viral gene. Here, we report a method for enzymatic production and antiviral use of 2'-fluoro-modified siRNA swarms. Utilizing the RNA-dependent RNA polymerase from bacteriophage phi6, we produced 2'-F-siRNA swarms containing either all or a fraction of modified adenosine, cytidine or uridine residues in the antisense strand of the UL29 target. The siRNA containing modified pyrimidines demonstrated high resistance to RNase A and the antiviral potency of all the UL29-specific 2'-F-siRNA swarms was 100-fold in comparison with the unmodified counterpart, without additional cytotoxicity. Modest stimulation of innate immunity signaling, including induced expression of both type I and type III interferons, as well as interferon-stimulated gene 54, by 2'-F-cytidine and 2'-F-uridine modified siRNA swarms occurred at early time points after transfection while the 2'-F-adenosine-containing siRNA was similar to the unmodified antiviral siRNA swarm in this respect. The antiviral efficacy of the 2'-F-siRNA swarms and the elicited cellular innate responses did not correlate suggesting that innate immunity pathways do not significantly contribute to the observed enhanced antiviral activity of the modified siRNAs. The results support further applications of enzymatically produced siRNA molecules with incorporated adenosine nucleotides, carrying fluoro-modification on ribose C2' position, for further antiviral studies in vitro and in vivo.


Subject(s)
Antiviral Agents/pharmacology , Cell Survival , Herpesvirus 1, Human/drug effects , Immunity, Innate , RNA, Small Interfering/pharmacology , RNA-Dependent RNA Polymerase/metabolism , Adenosine/metabolism , Bacteriophage phi 6/enzymology , Cell Line , Cell Line, Tumor , Cytidine/metabolism , DNA-Binding Proteins/antagonists & inhibitors , Herpesvirus 1, Human/immunology , Humans , RNA, Small Interfering/chemical synthesis , Transfection , Uridine/metabolism , Viral Proteins/antagonists & inhibitors
17.
Viruses ; 12(12)2020 12 13.
Article in English | MEDLINE | ID: mdl-33322225

ABSTRACT

Acyclovir is the drug of choice for the treatment of herpes simplex virus (HSV) infections. Acyclovir-resistant HSV strains may emerge, especially during long-term drug use, and subsequently cause difficult-to-treat exacerbations. Previously, we set up a novel treatment approach, based on enzymatically synthesized pools of siRNAs, or siRNA swarms. These swarms can cover kilobases-long target sequences, reducing the likelihood of resistance to treatment. Swarms targeting the UL29 essential gene of HSV-1 have demonstrated high efficacy against HSV-1 in vitro and in vivo. Here, we assessed the antiviral potential of a UL29 siRNA swarm against circulating strains of HSV-1, in comparison with acyclovir. All circulating strains were sensitive to both antivirals, with the half-maximal inhibitory concentrations (IC50) in the range of 350-1911 nM for acyclovir and 0.5-3 nM for the UL29 siRNA swarm. Additionally, we showed that an acyclovir-resistant HSV-1, devoid of thymidine kinase, is highly sensitive to UL29 siRNA treatment (IC50 1.0 nM; Imax 97%). Moreover, the detected minor variations in the RNAi target of the HSV strains had no effect on the potency or efficacy of UL29 siRNA swarm treatment. Our findings support the development of siRNA swarms for the treatment of HSV-1 infections, in order to circumvent any potential acyclovir resistance.


Subject(s)
Acyclovir/pharmacology , DNA-Binding Proteins/genetics , Herpes Simplex/virology , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/genetics , RNA Interference , RNA, Small Interfering/genetics , Viral Proteins/genetics , Acyclovir/therapeutic use , Animals , Chlorocebus aethiops , Dose-Response Relationship, Drug , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , Herpes Simplex/therapy , Herpesvirus 1, Human/classification , Herpesvirus 1, Human/isolation & purification , Humans , Inhibitory Concentration 50 , Vero Cells
18.
mSphere ; 5(3)2020 06 24.
Article in English | MEDLINE | ID: mdl-32581076

ABSTRACT

Infections with the nine human herpesviruses (HHVs) are globally prevalent and characterized by lifelong persistence. Reactivations can potentially manifest as life-threatening conditions for which the demonstration of viral DNA is essential. In the present study, we developed HERQ-9, a pan-HHV quantitative PCR designed in triplex reactions to differentiate and quantify each of the HHV-DNAs: (i) herpes simplex viruses 1 and 2 and varicella-zoster virus; (ii) Epstein-Barr virus, human cytomegalovirus, and Kaposi's sarcoma-associated herpesvirus; and (iii) HHV-6A, -6B, and -7. The method was validated with prequantified reference standards as well as with mucocutaneous swabs and cerebrospinal fluid, plasma, and tonsillar tissue samples. Our findings highlight the value of multiplexing in the diagnosis of many unsuspected, yet clinically relevant, herpesviruses. In addition, we report here frequent HHV-DNA co-occurrences in clinical samples, including some previously unknown. HERQ-9 exhibited high specificity and sensitivity (LOD95s of ∼10 to ∼17 copies/reaction), with a dynamic range of 101 to 106 copies/µl. Moreover, it performed accurately in the coamplification of both high- and low-abundance targets in the same reaction. In conclusion, we demonstrated that HERQ-9 is suitable for the diagnosis of a plethora of herpesvirus-related diseases. Besides its significance to clinical management, the method is valuable for the assessment of hitherto-unexplored synergistic effects of herpesvirus coinfections. Furthermore, its high sensitivity enables studies on the human virome, often dealing with minute quantities of persisting HHVs.IMPORTANCE By adulthood, almost all humans become infected by at least one herpesvirus (HHV). The maladies inflicted by these microbes extend beyond the initial infection, as they remain inside our cells for life and can reactivate, causing severe diseases. The diagnosis of active infection by these ubiquitous pathogens includes the detection of DNA with sensitive and specific assays. We developed the first quantitative PCR assay (HERQ-9) designed to identify and quantify each of the nine human herpesviruses. The simultaneous detection of HHVs in the same sample is important since they may act together to induce life-threatening conditions. Moreover, the high sensitivity of our method is of extreme value for assessment of the effects of these viruses persisting in our body and their long-term consequences on our health.


Subject(s)
Herpesviridae Infections/diagnosis , Herpesviridae/classification , Multiplex Polymerase Chain Reaction/methods , Adolescent , Adult , Aged , Cell Line , Child , Child, Preschool , Computer Simulation , DNA Primers/genetics , DNA Probes/genetics , DNA, Viral/genetics , Herpesviridae Infections/virology , Humans , Middle Aged , Palatine Tonsil/virology , Reproducibility of Results , Sensitivity and Specificity , Young Adult
19.
Mol Ther Oncolytics ; 17: 47-60, 2020 Jun 26.
Article in English | MEDLINE | ID: mdl-32322662

ABSTRACT

Despite some promising results, the majority of patients do not benefit from T cell therapies, as tumors prevent T cells from entering the tumor, shut down their activity, or downregulate key antigens. Due to their nature and mechanism of action, oncolytic viruses have features that can help overcome many of the barriers currently facing T cell therapies of solid tumors. This study aims to understand how four different oncolytic viruses (adenovirus, vaccinia virus, herpes simplex virus, and reovirus) perform in that task. For that purpose, an immunocompetent in vivo tumor model featuring adoptive tumor-infiltrating lymphocyte (TIL) therapy was used. Tumor growth control (p < 0.001) and survival analyses suggest that adenovirus was most effective in enabling T cell therapy. The complete response rate was 62% for TILs + adenovirus versus 17.5% for TILs + PBS. Of note, TIL biodistribution did not explain efficacy differences between viruses. Instead, immunostimulatory shifts in the tumor microenvironment mirrored efficacy results. Overall, the use of oncolytic viruses can improve the utility of T cell therapies, and additional virus engineering by arming with transgenes can provide further antitumor effects. This phenomenon was seen when an unarmed oncolytic adenovirus was compared to Ad5/3-E2F-d24-hTNFa-IRES-hIL2 (TILT-123). A clinical trial is ongoing, where patients receiving TIL treatment also receive TILT-123 (ClinicalTrials.gov: NCT04217473).

20.
J Gen Virol ; 90(Pt 10): 2468-2473, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19553393

ABSTRACT

Coxsackievirus B3 (CBV3) is a member of the human enterovirus B species and a common human pathogen. Even though much is known about the enteroviral life cycle, no specific drugs are available to treat enterovirus infections. RNA interference (RNAi) has evolved to be an important tool for antiviral experimental therapies and gene function studies. We describe here a novel approach for RNAi against CBVs by using a short interfering (siRNA) pool covering 3.5 kb of CBV3 genomic sequence. The RNA-dependent RNA polymerase (RdRP) of bacteriophage phi6 was used to synthesize long double-stranded RNA (dsRNA) from a cloned region (nt 3837-7399) of the CBV3 genome. The dsRNA was cleaved using Dicer, purified and introduced to cells by transfection. The siRNA pool synthesized using the phi6 RdRP (phi6-siRNAs) was considerably more effective than single-site siRNAs. The phi6-siRNA pool also inhibited replication of other enterovirus B species, such as coxsackievirus B4 and coxsackievirus A9.


Subject(s)
Bacteriophage phi 6/enzymology , Enterovirus B, Human/physiology , RNA Interference/physiology , RNA, Small Interfering/metabolism , RNA-Dependent RNA Polymerase/metabolism , Animals , Cell Line , Mice , Virus Replication/physiology
SELECTION OF CITATIONS
SEARCH DETAIL