Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters

Complementary Medicines
Therapeutic Methods and Therapies TCIM
Publication year range
1.
mBio ; 12(3): e0022721, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34061599

ABSTRACT

Human cytomegalovirus (HCMV) presents a major health burden in the immunocompromised and in stem cell transplant medicine. A lack of understanding about the mechanisms of HCMV latency in undifferentiated CD34+ stem cells, and how latency is broken for the virus to enter the lytic phase of its infective cycle, has hampered the development of essential therapeutics. Using a human induced pluripotent stem cell (iPSC) model of HCMV latency and patient-derived myeloid cell progenitors, we demonstrate that bone morphogenetic protein receptor type 2 (BMPR2) is necessary for HCMV latency. In addition, we define a crucial role for the transcription factor Yin Yang 1 (YY1) in HCMV latency; high levels of YY1 are maintained in latently infected cells as a result of BMPR2 signaling through the SMAD4/SMAD6 axis. Activation of SMAD4/6, through BMPR2, inhibits TGFbeta receptor signaling, which leads to the degradation of YY1 via induction of a cellular microRNA (miRNA), hsa-miR-29a. Pharmacological targeting of BMPR2 in progenitor cells results in the degradation of YY1 and an inability to maintain latency and renders cells susceptible to T cell killing. These data argue that BMPR2 plays a role in HCMV latency and is a new potential therapeutic target for maintaining or disrupting HCMV latency in myeloid progenitors. IMPORTANCE Understanding the mechanisms which regulate HCMV latency could allow therapeutic targeting of the latent virus reservoir from where virus reactivation can cause severe disease. We show that the BMPR2/TGFbeta receptor/YY1 signaling axis is crucial to maintain HCMV latency in undifferentiated cells and that pharmacological reduction of BMPR2 in latently infected cells leads to reactivation of the viral lytic transcription program, which renders the infected cell open to immune detection and clearance in infected individuals. Therefore, this work identifies key host-virus interactions which regulate HCMV latent infection. It also demonstrates a potential new therapeutic approach to reduce HCMV reactivation-mediated disease by the treatment of donor stem cells/organs prior to transplantation, which could have a major impact in the transplant disease setting.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/metabolism , Cytomegalovirus/physiology , Host-Pathogen Interactions , Induced Pluripotent Stem Cells/virology , Myeloid Cells/virology , Signal Transduction , Virus Latency , YY1 Transcription Factor/metabolism , Bone Morphogenetic Protein Receptors, Type II/genetics , Cells, Cultured , Humans , THP-1 Cells , YY1 Transcription Factor/genetics
2.
mBio ; 11(6)2020 12 15.
Article in English | MEDLINE | ID: mdl-33323506

ABSTRACT

Nitric oxide is a versatile and critical effector molecule that can modulate many cellular functions. Although recognized as a regulator of infections, the inhibitory mechanism of nitric oxide against human cytomegalovirus (HCMV) replication remains elusive. We demonstrate that nitric oxide attenuates viral replication by interfering with HCMV-mediated modulation of several cellular processes. Nitric oxide exposure reduced HCMV genome synthesis and infectious viral progeny with cell-type-dependent differences observed. Mitochondrial respiration was severely reduced in both uninfected and HCMV-infected cells during exposure with little impact on ATP levels indicating changes in cellular metabolism. Metabolomics identified significantly altered small molecules in multiple pathways during nitric oxide exposure including nucleotide biosynthesis, tricarboxylic acid (TCA) cycle, and glutamine metabolism. Glutathione metabolites were increased coinciding with a reduction in the glutathione precursor glutamine. This shift was accompanied by increased antioxidant enzymes. Glutamine deprivation mimicked defects in HCMV replication and mitochondrial respiration observed during nitric oxide exposure. These data suggest that nitric oxide limits glutaminolysis by shuttling glutamine to glutathione synthesis. In addition, lipid intermediates were severely altered, which likely contributes to the observed increase in defective viral particles. Nitric oxide disrupts multiple cellular processes, and we had limited success in rescuing replication defects by supplementing with metabolic intermediates. Our studies indicate that nitric oxide attenuation of HCMV is multifactorial with interference in viral manipulation of cellular metabolism playing a central role.IMPORTANCE Human cytomegalovirus is a prevalent pathogen that can cause serious disease in patients with compromised immune systems, including transplant patients and during congenital infection. HCMV lytic replication likely occurs in localized sites of infection with immune cells infiltrating and releasing nitric oxide with other effector molecules. This nonspecific immune response results in both uninfected and infected cells exposed to high levels of nitric oxide. The absence of nitric oxide synthase has been associated with lethal HCMV infection. We demonstrate that nitric oxide inhibition of HCMV replication is multifactorial and cell type dependent. Our results indicate that nitric oxide controls replication by interfering with viral modulation of cellular metabolism while also affecting proliferation and mitochondrial respiration of neighboring uninfected cells. These studies identify the mechanism and contribution of nitric oxide during immune control of HCMV infection and provide insight into its role in other viral infections.


Subject(s)
Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/virology , Cytomegalovirus/physiology , Nitric Oxide/metabolism , Adenosine Triphosphate/metabolism , Cell Line , Citric Acid Cycle , Cytomegalovirus/genetics , Cytomegalovirus Infections/immunology , Glutamine/metabolism , Glutathione/metabolism , Host-Pathogen Interactions , Humans , Mitochondria/metabolism , Nitric Oxide/immunology , Virus Replication
3.
BMC Complement Altern Med ; 17(1): 428, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28851336

ABSTRACT

BACKGROUND: In immunocompromised patients, human cytomegalovirus (HCMV) infection can lead to severe, life-threatening diseases, such as pneumonitis, hepatitis, gastrointestinal tract disease, and retinitis. We previously reported that a 70% ethanol extract of Elaeocarpus sylvestris leaves (ESE) inhibits human cytomegalovirus (HCMV) replication in vitro. In the present study, we determined the solvent fraction of ESE that inhibits HCMV replication using activity-guided fractionation. METHODS: Activity-guided fractionation of ESE was performed to determine the solvent fraction that inhibits HCMV replication. Effects of solvent fractions on HCMV lytic gene expression and major immediate-early (MIE) enhancer/promoter activity were further investigated. RESULTS: Among the solvent fractions tested, the EtOAc fraction of ESE markedly reduced HCMV lytic gene expression and viral replication in vitro without exerting significant cytotoxic effects against human foreskin fibroblasts (HFF). Furthermore, the EtOAc fraction negatively affected HCMV MIE enhancer/promoter activity. CONCLUSION: Our data collectively indicate that the EtOAc fraction of ESE contains active constituents that inhibit HCMV MIE enhancer/promoter activity and viral replication. The EtOAc fraction of ESE is a good source of novel drug candidates for treatment of HCMV-associated diseases.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus/drug effects , Cytomegalovirus/physiology , Elaeocarpaceae/chemistry , Plant Extracts/pharmacology , Virus Replication/drug effects , Antiviral Agents/isolation & purification , Cytomegalovirus/genetics , Cytomegalovirus Infections/virology , Genes, Immediate-Early , Humans , Plant Extracts/isolation & purification , Viral Proteins/genetics , Viral Proteins/metabolism
4.
J Gen Virol ; 98(4): 754-768, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28100301

ABSTRACT

To identify new compounds with anti-human cytomegalovirus (HCMV) activity and new anti-HCMV targets, we developed a high-throughput strategy to screen a GlaxoSmithKline Published Kinase Inhibitor Set. This collection contains a range of extensively characterized compounds grouped into chemical families (chemotypes). From our screen, we identified compounds within chemotypes that impede HCMV protein production and identified kinase proteins associated with inhibition of HCMV protein production that are potential novel anti-HCMV targets. We focused our study on a top 'hit' in our screen, SB-734117, which we found inhibits productive replication of several HCMV strains. Kinase selectivity data indicated that SB-734117 exhibited polypharmacology and was an inhibitor of several proteins from the AGC and CMCG kinase groups. Using Western blotting, we found that SB-734711 inhibited accumulation of HCMV immediate-early proteins, phosphorylation of cellular proteins involved in immediate-early protein production (cAMP response element-binding protein and histone H3) and histone H3 lysine 36 trimethylation (H3K36me3). Therefore, we identified SB-734117 as a novel anti-HCMV compound and found that inhibition of AGC and CMCG kinase proteins during productive HCMV replication was associated with inhibition of viral protein production and prevented post-translational modification of cellular factors associated with viral protein production.


Subject(s)
Antiviral Agents/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Cytomegalovirus/drug effects , Histones/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Virus Replication/drug effects , Blotting, Western , Cytomegalovirus/physiology , Drug Evaluation, Preclinical , High-Throughput Screening Assays , Humans
5.
Bioorg Med Chem ; 23(21): 7035-44, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26443550

ABSTRACT

The need for novel therapeutic options to fight herpesvirus infections still persists. Herein we report the design, synthesis and antiviral evaluation of a new family of non-nucleoside antivirals, derived from 1-[ω-(4-bromophenoxy)alkyl]uracil derivatives--previously reported inhibitors of human cytomegalovirus (HCMV). Introduction of the N-(4-phenoxyphenyl)acetamide side chain at N(3) increased their potency and widened activity spectrum. The most active compounds in the series exhibit submicromolar activity against different viral strains of HCMV and varicella zoster virus (VZV) replication in HEL cell cultures. Inactivity against other DNA and RNA viruses, including herpes simplex virus 1/2, points to a novel mechanism of antiviral action.


Subject(s)
Acetamides/chemistry , Antiviral Agents/chemistry , Cytomegalovirus/physiology , Herpesvirus 3, Human/physiology , Uracil/chemistry , Acetamides/chemical synthesis , Acetamides/toxicity , Antiviral Agents/chemical synthesis , Antiviral Agents/toxicity , Cell Line , Cell Proliferation/drug effects , DNA Viruses/drug effects , DNA Viruses/physiology , Drug Evaluation, Preclinical , Herpesvirus 3, Human/drug effects , Humans , RNA Viruses/drug effects , RNA Viruses/physiology , Structure-Activity Relationship , Virus Replication/drug effects
6.
J Eur Acad Dermatol Venereol ; 29(12): 2451-7, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26403508

ABSTRACT

BACKGROUND: Lung transplant recipients (LTR) are at increased risk for squamous cell carcinoma of the skin (SCC), but risk factors (RF) are incompletely understood. OBJECTIVE: To assess associations between exposure to certain medications and viral infections, and subsequent SCC development. METHODS: Retrospective study examining incidence and potential RF for SCC in LTR transplanted from 1992 to 2010 followed up at one centre. Cumulative incidence and Cox proportional hazards regression models were used to evaluate RF in the first year post-transplant for SCC formation during the follow-up. RESULTS: In 205 analysed LTR, 46 patients were diagnosed with SCC during a median follow-up of 4.9 years. The cumulative incidences of first SCC were 16.7% and 34.1%, for 5 and 10 years post-transplantation respectively. Multivariable analysis identified CMV replication (HR 7.69, 95% CI 2.93-20.2, P < 0.001) and moxifloxacin exposure (HR 2.35, 95% CI 1.15-4.81, P = 0.020) during the first year post-transplantation as independent RF for SCC development during follow-up. CONCLUSION: In our cohort, moxifloxacin use and CMV replication during the first year post-transplantation were associated with increased risk for SCC. These two factors could be indicators of over-immunosuppression. Their role in SCC development requires investigations in larger cohorts and prospective studies.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Carcinoma, Squamous Cell/epidemiology , Cytomegalovirus Infections/epidemiology , Fluoroquinolones/therapeutic use , Lung Transplantation , Skin Neoplasms/epidemiology , Adult , Carcinoma, Squamous Cell/etiology , Cytomegalovirus/physiology , Female , Follow-Up Studies , Humans , Incidence , Male , Middle Aged , Moxifloxacin , Retrospective Studies , Risk Factors , Skin Neoplasms/etiology , Virus Replication
7.
Viruses ; 7(6): 2816-33, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26053925

ABSTRACT

Human cytomegalovirus (HCMV) is a major risk factor in transplantation and AIDS patients, which induces high morbidity and mortality. These patients infected with HCMV experience an imbalance of redox homeostasis that cause accumulation of reactive oxygen species (ROS) at the cellular level. H2O2, the most common reactive oxygen species, is the main byproduct of oxidative metabolism. However, the function of H2O2 on HCMV infection is not yet fully understood and the effect and mechanism of N-acetylcysteine (NAC) on H2O2-stimulated HCMV replication is unclear. We, therefore, examined the effect of NAC on H2O2-induced HCMV production in human foreskin fibroblast cells. In the present study, we found that H2O2 enhanced HCMV lytic replication through promoting major immediate early (MIE) promoter activity and immediate early (IE) gene transcription. Conversely, NAC inhibited H2O2-upregulated viral IE gene expression and viral replication. The suppressive effect of NAC on CMV in an acute CMV-infected mouse model also showed a relationship between antioxidants and viral lytic replication. Intriguingly, the enhancement of HCMV replication via supplementation with H2O2 was accompanied with the activation of the p38 mitogen-activated protein kinase pathway. Similar to NAC, the p38 inhibitor SB203580 inhibited H2O2-induced p38 phosphorylation and HCMV upregulation, while upregulation of inducible ROS was unaffected. These results directly relate HCMV replication to H2O2, suggesting that treatment with antioxidants may be an attractive preventive and therapeutic strategy for HCMV.


Subject(s)
Cytomegalovirus/drug effects , Cytomegalovirus/physiology , Hydrogen Peroxide/metabolism , Signal Transduction , Virus Replication/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Acetylcysteine/metabolism , Animals , Antioxidants/metabolism , Cells, Cultured , Cytomegalovirus Infections/virology , Disease Models, Animal , Fibroblasts/virology , Genes, Immediate-Early , Humans , Male , Mice, Inbred BALB C , Oxidants/metabolism , Promoter Regions, Genetic/drug effects , Transcription, Genetic
8.
J Med Virol ; 87(12): 2135-44, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26058558

ABSTRACT

Human cytomegalovirus (HCMV) is an opportunistic pathogen that causes severe diseases in congenitally infected newborns and immunocompromised patients. Currently, no vaccine is available to prevent HCMV infection. Anti-viral drugs are limited by their side effects and drug resistance. In this study, by performing a medium-sized, anti-HCMV chemical screening, we identified SP600125, CC-401, and the c-Jun N-terminal kinase (JNK) inhibitor VIII, three structurally different small molecule JNK inhibitors that effectively inhibited HCMV replication in cultured human fibroblasts (HFs). SP600125 showed its potential by inhibiting the viral replication of a HCMV laboratory strain in HFs and a HCMV clinical strain in human retinal pigment epithelial cells. Knockdown of JNK expression by RNA interference significantly impaired HCMV replication, mimicking the effect of the chemical inhibitors on virus infection. Mechanistically, SP600125 affects a very early step of the viral life cycle. Viral binding, entry, and the delivery of viral DNA into the cells were not inhibited by the compound. Instead, it suppressed the transcription of the immediate-early viral genes IE1/2 and the accumulation of their gene products. IE1/2 are among the first genes expressed after viral entry, and they are the master regulators of late phase viral gene expression. Consistent with this notion, the expression of other viral genes was also reduced after SP600125 treatment. We propose that JNK inhibitors have the potential to become a new class of anti-HCMV drug candidates, and JNK is a feasible target for the development of anti-HCMV drugs.


Subject(s)
Anthracenes/pharmacology , Antiviral Agents/pharmacology , Cytomegalovirus/drug effects , Enzyme Inhibitors/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Virus Replication/drug effects , Cells, Cultured , Cytomegalovirus/physiology , Drug Evaluation, Preclinical , Epithelial Cells/virology , Fibroblasts/virology , Gene Knockdown Techniques , Humans , Infant, Newborn , JNK Mitogen-Activated Protein Kinases/genetics , Microbial Sensitivity Tests
9.
PLoS One ; 10(5): e0126239, 2015.
Article in English | MEDLINE | ID: mdl-25992785

ABSTRACT

Human cytomegalovirus (HCMV) infection can lead to congenital hearing loss and mental retardation. Upon immune suppression, reactivation of latent HCMV or primary infection increases morbidity in cancer, transplantation, and late stage AIDS patients. Current treatments include nucleoside analogues, which have significant toxicities limiting their usefulness. In this study we screened a panel of synthetic heparin-binding peptides for their ability to prevent CMV infection in vitro. A peptide designated, p5+14 exhibited ~ 90% reduction in murine CMV (MCMV) infection. Because negatively charged, cell-surface heparan sulfate proteoglycans (HSPGs), serve as the attachment receptor during the adsorption phase of the CMV infection cycle, we hypothesized that p5+14 effectively competes for CMV adsorption to the cell surface resulting in the reduction in infection. Positively charged Lys residues were required for peptide binding to cell-surface HSPGs and reducing viral infection. We show that this inhibition was not due to a direct neutralizing effect on the virus itself and that the peptide blocked adsorption of the virus. The peptide also inhibited infection of other herpesviruses: HCMV and herpes simplex virus 1 and 2 in vitro, demonstrating it has broad-spectrum antiviral activity. Therefore, this peptide may offer an adjunct therapy for the treatment of herpes viral infections and other viruses that use HSPGs for entry.


Subject(s)
Heparan Sulfate Proteoglycans/metabolism , Heparan Sulfate Proteoglycans/pharmacology , Herpesviridae/drug effects , Herpesviridae/physiology , Virus Internalization/drug effects , Amino Acid Sequence , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cells, Cultured , Cytomegalovirus/drug effects , Cytomegalovirus/pathogenicity , Cytomegalovirus/physiology , Drug Evaluation, Preclinical , Heparan Sulfate Proteoglycans/chemistry , Herpesviridae/pathogenicity , Herpesviridae Infections/drug therapy , Herpesviridae Infections/prevention & control , Herpesviridae Infections/virology , Humans , In Vitro Techniques , Mice , Models, Molecular , Molecular Sequence Data , Muromegalovirus/drug effects , Muromegalovirus/pathogenicity , Muromegalovirus/physiology , Protein Structure, Secondary , Virus Attachment/drug effects
10.
Microbes Infect ; 14(12): 1086-92, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22683667

ABSTRACT

It has been reported that treatment with tricin (4',5,7-trihydroxy-3',5'-dimethoxyflavone), a derivative of Sasa albo-marginata, after human cytomegalovirus (HCMV) infection significantly suppressed both infectious virus production and HCMV replication in the human embryonic fibroblast cell line MRC-5. In this paper, we examined the mechanisms for the anti-HCMV effects of tricin in MRC-5 cells. Exposure of fibroblasts to tricin inhibited infectious HCMV production, with concomitant decreases in levels of transcripts of the CXC chemokine IFN-inducible T cell alpha chemoattractant (I-TAC or CXCL11) gene. We also found that the transcripts of the HCMV immediate early (IE) gene and replication of HCMV were lower in CXCL11 gene-knockdown cells. These results suggest that tricin is a novel compound with potential anti-HCMV activity and that CXCL11 is one of the chemokines involved in HCMV replication. In addition, it is possible that CXCL11 is the one of the targets of tricin.


Subject(s)
Antiviral Agents/pharmacology , Chemokine CXCL11/metabolism , Cytomegalovirus/drug effects , Flavonoids/pharmacology , Antiviral Agents/isolation & purification , Cell Line , Chemokine CXCL11/genetics , Cytomegalovirus/physiology , Fibroblasts/drug effects , Fibroblasts/virology , Flavonoids/isolation & purification , Gene Expression/drug effects , Gene Expression Profiling , Gene Knockout Techniques , Humans , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Sasa/chemistry , Virus Replication/drug effects
11.
Antiviral Res ; 95(1): 12-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22580129

ABSTRACT

Certain viral protein-protein interactions provide attractive targets for antiviral drug development. Recently, we described a ß-lactamase based protein fragment complementation assay (PCA) to study the core interaction of the nuclear egress complex (NEC) of different herpesviruses in cells. Now, to have a cell free assay for inhibitor screens, we expressed split ß-lactamase tagged interaction domains of the viral pUL50 and pUL53 proteins representing the NEC of human cytomegalovirus (HCMV) in bacteria. After validation and basic characterization of this NEC-PCA, we tested peptide inhibitors of the pUL50-pUL53 complex. We show that peptides resembling sequences of the first conserved region of pUL53 can inhibit the NEC-PCA. This, on one hand, indicated that the core interaction in the HCMV NEC is mediated by a linear motif. On the other hand it proved that this new pUL50-pUL53 interaction assay allows a simple cell free test for small molecular inhibitors.


Subject(s)
Cytomegalovirus/physiology , Viral Proteins/metabolism , Virology/methods , Virus Release , Antiviral Agents/pharmacology , Bacteria , Drug Evaluation, Preclinical/methods , Humans , Protein Binding , beta-Lactamases/metabolism
12.
Antiviral Res ; 89(3): 246-56, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21277329

ABSTRACT

Human cytomegalovirus (HCMV) enters cells through a complex pathway involving the interaction of multiple viral glycoproteins and cellular receptors. While HCMV clinical isolates enter a wide range of cell types, entry has historically been studied using a laboratory strain of virus that can only infect fibroblasts. Herein, we have constructed a HCMV reporter strain that contains GFP fused to the abundant tegument protein pp65 to allow for the direct visualization of virus attachment and entry. Furthermore, the UL131 gene of this strain was restored to clinical isolate sequence to expand our studies of entry into physiologically relevant epithelial cell types. Using the HCMV-GFP reporter virus, we developed an image-based assay and screened a library containing 65,000 compounds for the inhibition of virus entry into fibroblasts. In addition to assessing the effect on virus entry, automated image analysis provided information on compound toxicity and whether the compounds acted as attachment or post-attachment inhibitors. To identify therapeutically viable inhibitors capable of blocking entry in multiple cell types, the inhibitors were screened further for their ability to inhibit virus entry into epithelial cells. Compounds were identified that were able to inhibit virus entry into both cell types at either attachment or post-attachment steps.


Subject(s)
Antiviral Agents/isolation & purification , Cytomegalovirus/physiology , Drug Evaluation, Preclinical/methods , Epithelial Cells/virology , Fibroblasts/virology , Virology/methods , Virus Internalization , Antiviral Agents/pharmacology , Cytomegalovirus/drug effects , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Staining and Labeling/methods
13.
Antivir Chem Chemother ; 21(2): 85-95, 2010.
Article in English | MEDLINE | ID: mdl-21107017

ABSTRACT

BACKGROUND: Many viruses display affinity for polysaccharides presented at the surface of target cells with high biological relevance for virus attachment and entry. This raises the possibility of the application of polysaccharides, particularly their sulfated modifications, in studies of receptor binding and also in antiviral therapy. METHODS: In this study, we analysed various sulfated glucans, generated from a commercial preparation of rice bran using chemical, chromatographic, spectroscopic and virological methods. RESULTS: A number of sulfated polysaccharides with different charge densities were generated from a commercial rice bran preparation by aqueous extraction followed by chemical sulfation. The backbone of the type of glucans identified was made up mainly of α-(1→4)-linked glucopyranosyl residues. Sulfate groups were found to be located at C6 and partly at C2 or C3 of glucopyranosyl residues. Sulfation appeared to be very important for anti-cytomegaloviral activity, as desulfation experiments demonstrated an impairment of activity. Using an established cytomegalovirus replication assay with primary human fibroblasts, data demonstrated that the anti-cytomegaloviral effect was determined primarily at the stage of viral entry. CONCLUSIONS: Sulfated glucans derived from rice bran possess very promising characteristics for their potential use as entry-inhibiting anti-cytomegaloviral agents.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cytomegalovirus Infections/drug therapy , Cytomegalovirus , Glucans , Oryza , Plant Extracts/chemistry , Plant Extracts/pharmacology , Polysaccharides/chemistry , Polysaccharides/pharmacology , Sulfates/chemistry , Virus Internalization/drug effects , Antiviral Agents/isolation & purification , Cell Line , Cytomegalovirus/physiology , Cytomegalovirus Infections/virology , Glucans/chemistry , Glucans/isolation & purification , Glucans/pharmacology , Humans , Oryza/chemistry , Polysaccharides/isolation & purification , Spectrum Analysis , Virus Replication/drug effects
14.
Antibiot Khimioter ; 53(11-12): 3-10, 2008.
Article in Russian | MEDLINE | ID: mdl-19441649

ABSTRACT

Human cytomegalovirus (CMV), an agent of infection (CMVI), lethally dangerous for immune deficient neonates and adults was investigated in vitro as a target for a therapeutic effect of new membrane-active polyanionic compounds (MPC). Previous studies on the alicycle- and sulfate-modified carboxy-MPCs revealed a well-defined tendency of the anti-CMV activity amplification in parallel with increasing of the content of sulfate groups, enhancing the negative charge of the macromolecule. The dominating role of the electrostatic factor was confirmed by the highest activity of AS-688, compound with maximum sulfation among the tested MPCs. Its selectivity index (SI) of the CMVI inhibition in human diploid fibroblast cells reached 5450, 7500, 250 and 4286 in the microbicidal, viricidal, prophylactic and therapeutic schemes of the experiment respectively. The antiviral activity at the first, second and third schemes was explained by the polyanion-typical potential of electrostatic neutralization of the countercharged virions and prevention of the virus adsorption on the cell membranes (in competition with heparin sulfate, a cellular receptor of CMV), whereas the therapeutic effect required the ability of MPC to influence the intracellular stages of the CMV life cycle. The PCR and immunochemical assays revealed an inhibitory action of AS-688 on replication of the viral DNA and the following synthesis of the late viral protein gB with efficiency similar to that of gancyclovir (GCV). However, in contrast to GCV, acting as inhibitor of enzyme (viral RNA-polymerase) factor of the biosynthesis, the therapeutic activity of MPC could be interpreted by competition with viral RNA/DNA due to the specific character of the MPC molecular basis, initially constructed on the principle of nucleic acids backbone and charge adjustable imitation. This mechanism assuming reduction of the cytotoxicity risks, explained the experimentally observed fact of low cytotoxicity of MPCs and possible achievement of high SI. The MPC ability to penetrate into the cells without disruption of cellular membrane permeability was confirmed in experiments with the fluorescent-labeled derivate AS-679, structurally and functionally related to AS-688. In the light of the previously described HIV inhibiting properties of AS-688, AS-679 and MPC analogous, the results could be considered prospective in development of new highly effective agents for combined antiviral protection.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus Infections/drug therapy , Cytomegalovirus/physiology , Polymers/pharmacology , Virus Attachment/drug effects , Cell Line , Cytomegalovirus Infections/metabolism , DNA Replication/drug effects , DNA Replication/physiology , DNA, Viral/biosynthesis , Drug Evaluation, Preclinical , Fibroblasts/virology , Humans , Polyelectrolytes , Virus Replication/drug effects , Virus Replication/physiology
15.
Antivir Chem Chemother ; 17(1): 11-6, 2006.
Article in English | MEDLINE | ID: mdl-16542001

ABSTRACT

We examined the effect of Kampo on the replication of ganciclovir (GCV)-resistant human cytomegalovirus (HCMV) in the human embryonic fibroblast cell line MRC-5. Treatment of HCMV-infected cells with Sho-seiryu-to (SST; Xiao-Qing-Long-Tang in Chinese) resulted in the inhibition of viral replication without affecting the cell growth. SST treatment decreased the synthesis of viral DNA, but had no virucidal effect on cell-free HCMV. However, the inhibitory effect of SST on HCMV replication was ablated by anti-interferon-beta (IFN-beta) antibody suggesting that SST inhibits the replication of GCV-resistant HCMV through the induction of IFN-beta. These results suggest that SST is a novel compund with potential as an anti-HCMV.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus Infections/virology , Cytomegalovirus/physiology , Drugs, Chinese Herbal/pharmacology , Ganciclovir/pharmacology , Medicine, Kampo , Virus Replication/drug effects , Cytomegalovirus/drug effects , Cytomegalovirus/isolation & purification , Cytomegalovirus Infections/drug therapy , DNA, Viral/analysis , Drug Resistance, Viral , Fibroblasts , Humans , Interferon-beta/metabolism , Nucleic Acid Hybridization
17.
Zhongguo Zhong Yao Za Zhi ; 30(1): 47-9, 2005 Jan.
Article in Chinese | MEDLINE | ID: mdl-15714801

ABSTRACT

OBJECTIVE: To investigate the effect of allitridin injection on the expression of human cytomegalovirus (HCMV) immediate-early antigens (IEAs including IE72 and IE86) in human embryonic lung cells. METHOD: HCMV AD 169 Virus strain infected cell model (MOI = 2.5 and 0.25, respectively) were established, and then treated with ICm5 and MTC doses of allitridin. Western blot was used to analyze the of IE72 and IE86 expression after the treatment, ganciclovir(GCV, IC50 and 2.3 x IC50) treatment as control. RESULT: No matter what kind of MOI was used, both IE86 and IE72 antigens' expression was effectively suppressed by allitridin treatment, and the inhibitory rate of IE86 was almost twice of IE72's. Compared with GCV, allitridin had stronger inhibitory effect on IE86 expressing, although its efficacy on IE72 was weaker than GCV. CONCLUSION: Allitridin could suppress the expression of IE72 and IE86, especially for IE86 expressing, maybe it is ore of key role in the mechanism of allitridin against HCMV.


Subject(s)
Allyl Compounds/pharmacology , Cytomegalovirus/genetics , Garlic , Immediate-Early Proteins/metabolism , Sulfides/pharmacology , Trans-Activators/metabolism , Viral Proteins/metabolism , Allyl Compounds/administration & dosage , Allyl Compounds/isolation & purification , Antiviral Agents/pharmacology , Cells, Cultured , Cytomegalovirus/physiology , Fibroblasts/cytology , Fibroblasts/metabolism , Garlic/chemistry , Gene Expression Regulation, Viral/drug effects , Humans , Injections , Lung/cytology , Sulfides/administration & dosage , Sulfides/isolation & purification
18.
Antiviral Res ; 60(1): 41-9, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14516920

ABSTRACT

Cytomegalovirus (CMV) is the most common cause of congenital infection in the developed world and can lead to a life-threatening disease. We therefore developed an animal model to evaluate candidate anti-CMV drugs and to further define the pathogenesis of CMV infections. Newborn guinea pigs were infected by intraperitoneal administration of 10(6) pfu of a virulent salivary gland (SG) passaged guinea pig CMV (gpCMV) within 48 h of birth. Inoculation of animals produced 50% overall mortality. A lack of weight gain was also a hallmark of infection. By day 14 after inoculation the weight of gpCMV-infected animals was significantly less than controls (152.9+/-45 g versus 254.7+/-38.5 g, P<0.0001). The most consistent isolation and highest titers of virus were found in the liver and spleen early while lung titers were maximal at day 10. A quantitative competitive PCR (qcPCR) assay confirmed the presence of a high CMV viral load in infected organs. Antiviral treatment with cyclic HPMPC (cHPMPC) for 7 days significantly reduced mortality (1/20 versus 14/20, P<0.001) and viral replication but did not improve weight gain. This model should be useful for further evaluations of the pathogenesis of CMV infections and for evaluation of antiviral drugs and vaccines.


Subject(s)
Antiviral Agents/therapeutic use , Cytomegalovirus Infections/drug therapy , Cytomegalovirus/drug effects , Cytosine/analogs & derivatives , Cytosine/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Organophosphorus Compounds/therapeutic use , Animals , Animals, Newborn , Body Weight/drug effects , Brain/virology , Cytomegalovirus/pathogenicity , Cytomegalovirus/physiology , Cytomegalovirus Infections/pathology , Cytomegalovirus Infections/virology , Guinea Pigs , Liver/virology , Lung/virology , Spleen/virology , Virus Replication/drug effects
19.
Antivir Chem Chemother ; 12(1): 61-70, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11437323

ABSTRACT

Human cytomegalovirus (HCMV) and herpes simplex virus (HSV) can cause a wide variety of clinical manifestations in man. Ganciclovir (GCV) is effective against HCMV infection when administered by the intravenous route and may be used orally in large doses for prophylaxis of HCMV infections in organ transplantation patients and in AIDS patients. In previous studies with acyclovir (ACV), we found that covalent attachment of an alkyl glycerol phosphate moiety greatly increased oral bioavailability and increased antiviral activity against hepatitis B virus. Adducts of ACV with alkyl propanediol phosphate were more active than the alkyl glycerol phosphate analogue in vitro in 2.2.15 cells, which constitutively produce hepatitis B virus. To see if this strategy would work for two other poorly absorbed nucleoside analogues, we synthesized 1-O-hexadecylpropanediol-3-phospho-GCV (HDP-P-GCV) and 1-O-hexadecyl-propanediol-3-phospho-penciclovir (HDP-P-PCV), and evaluated the in vitro antiviral activity, selectivity and oral antiviral activity of both compounds versus GCV or PCV in mice infected with HSV-1 or HDP-P-GCV versus murine cytomegalovirus (MCMV). HDP-P-GCV is orally active in both MCMV and HSV-1 infection in mice with antiviral activity equivalent to (HSV-1) or greater than oral GCV (MCMV). Oral HDP-P-PCV was more active than PCV orally versus intranasal HSV-1 infection in mice.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus Infections/drug therapy , Cytomegalovirus/drug effects , Ganciclovir/pharmacology , Herpes Simplex/drug therapy , Simplexvirus/drug effects , Administration, Oral , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacokinetics , Antiviral Agents/therapeutic use , Biological Availability , Biotransformation , Cell Line/drug effects , Cytomegalovirus/physiology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Fibroblasts/drug effects , Fibroblasts/virology , Ganciclovir/analogs & derivatives , Ganciclovir/chemical synthesis , Ganciclovir/pharmacokinetics , Ganciclovir/therapeutic use , Humans , Lung , Mice , Mice, Inbred BALB C , Muromegalovirus/drug effects , Muromegalovirus/physiology , Simplexvirus/physiology , Viral Plaque Assay , Virus Replication/drug effects
20.
J Gen Virol ; 82(Pt 6): 1439-1450, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11369889

ABSTRACT

The UL97-encoded protein kinase (pUL97) of human cytomegalovirus (HCMV) plays a critical role in the control of virus replication. Deletion of the UL97 gene results in a drastic reduction in the replication efficiency. Although the exact function of pUL97 remains unclear and its sensitivity to specific inhibitors is speculative, protein kinase inhibitors of the indolocarbazole class are effective inhibitors of cytomegalovirus. Based on the phosphorylation of ganciclovir (GCV), a novel quantification system for pUL97 kinase activity was established: the phosphorylated form of GCV exerts an easily quantifiable cytotoxic effect in transfected cells. Importantly, the addition of indolocarbazole compounds, Gö6976 and NGIC-I, which were highly effective at nanomolar concentrations while other protein kinase inhibitors were not, led to a significant reduction of pUL97 kinase activity. It was also demonstrated that a catalytically inactive mutant of pUL97, K355M, and a GCV-resistant mutant, M460I, were both negative for GCV phosphorylation, although protein phosphorylation remained detectable for the latter mutant. In vitro kinase assays were used to confirm the levels of pUL97-mediated phosphorylation recorded. To generate a tool for screening large numbers of putative inhibitors that preferentially interfere with GCV as well as protein phosphorylation, pUL97-expressing cell clones with stable pUL97 kinase activity were selected. This study demonstrates that certain indolocarbazole compounds are potent pUL97 inhibitors and, therefore, represent novel candidates for antiviral drugs that target viral protein kinase functions.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus/drug effects , Cytomegalovirus/enzymology , Enzyme Inhibitors/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Virus Replication/drug effects , Carbazoles/pharmacology , Cell Line , Clone Cells/drug effects , Clone Cells/enzymology , Clone Cells/metabolism , Cytomegalovirus/physiology , Drug Evaluation, Preclinical , Drug Resistance, Microbial , Ganciclovir/metabolism , Ganciclovir/toxicity , Humans , Indoles/pharmacology , Mutation/genetics , Mutation, Missense/genetics , Phosphorylation/drug effects , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Kinase Inhibitors , Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL