Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
J Biol Chem ; 295(44): 15013-15028, 2020 10 30.
Article En | MEDLINE | ID: mdl-32855235

The antimalarial agents artemisinins inhibit cytomegalovirus (CMV) in vitro and in vivo, but their target(s) has been elusive. Using a biotin-labeled artemisinin, we identified the intermediate filament protein vimentin as an artemisinin target, validated by detailed biochemical and biological assays. We provide insights into the dynamic and unique modulation of vimentin, depending on the stage of human CMV (HCMV) replication. In vitro, HCMV entry and viral progeny are reduced in vimentin-deficient fibroblasts, compared with control cells. Similarly, mouse CMV (MCMV) replication in vimentin knockout mice is significantly reduced compared with controls in vivo, confirming the requirement of vimentin for establishment of infection. Early after HCMV infection of human foreskin fibroblasts vimentin level is stable, but as infection proceeds, vimentin is destabilized, concurrent with its phosphorylation and virus-induced calpain activity. Intriguingly, in vimentin-overexpressing cells, HCMV infection is reduced compared with control cells. Binding of artesunate, an artemisinin monomer, to vimentin prevents virus-induced vimentin degradation, decreasing vimentin phosphorylation at Ser-55 and Ser-83 and resisting calpain digestion. In vimentin-deficient fibroblasts, the anti-HCMV activity of artesunate is reduced compared with controls. In summary, an intact and stable vimentin network is important for the initiation of HCMV replication but hinders its completion. Artesunate binding to vimentin early during infection stabilizes it and antagonizes subsequent HCMV-mediated vimentin destabilization, thus suppressing HCMV replication. Our target discovery should enable the identification of vimentin-binding sites and compound moieties for binding.


Antiviral Agents/pharmacology , Artemisinins/pharmacology , Cytomegalovirus/drug effects , Vimentin/antagonists & inhibitors , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Artemisinins/chemistry , Artemisinins/metabolism , Artesunate/pharmacology , Binding Sites , Calpain/metabolism , Cell Cycle/drug effects , Cells, Cultured , Cytomegalovirus/physiology , Cytomegalovirus Infections/metabolism , Drug Repositioning , Humans , Mass Spectrometry , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Vimentin/genetics , Vimentin/metabolism , Virus Replication/drug effects , Withanolides/pharmacology
2.
J Med Chem ; 63(8): 3896-3907, 2020 04 23.
Article En | MEDLINE | ID: mdl-32191456

The critical consequences of human cytomegalovirus (HCMV) infection in the transplant population and in congenitally infected infants, the limited treatment options for HCMV, and the rise of resistant mutants toward existing therapies has fueled the search for new anti-HCMV agents. A pp28-luciferase recombinant HCMV was used as a reporter system for high-throughput screening of HCMV inhibitors. Approximately 400 000 compounds from existing libraries were screened. Subsequent validation assays using resynthesized compounds, several virus strains, and detailed virology assays resulted in the identification of five structurally unique and selective HCMV inhibitors, active at sub to low micromolar concentrations. Further characterization revealed that each compound inhibited a specific stage of HCMV replication. One compound was also active against herpes simplex virus (HSV1 and HSV2), and another compound was active against Epstein-Barr virus (EBV). Drug combination studies revealed that all five compounds were additive with ganciclovir or letermovir. Future studies will focus on optimization of these new anti-HCMV compounds along with mechanistic studies.


Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cytomegalovirus/drug effects , Drug Discovery/methods , Animals , Antiviral Agents/therapeutic use , Cells, Cultured , Cytomegalovirus/physiology , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/physiopathology , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Fibroblasts/physiology , Fibroblasts/virology , Humans , Male , Mice
3.
Article En | MEDLINE | ID: mdl-30373792

Artesunate (AS), a semisynthetic artemisinin approved for malaria therapy, inhibits human cytomegalovirus (HCMV) replication in vitro, but therapeutic success in humans has been variable. We hypothesized that the short in vivo half-life of AS may contribute to the different treatment outcomes. We tested novel synthetic ozonides with longer half-lives against HCMV in vitro and mouse cytomegalovirus (MCMV) in vivo Screening of the activities of four ozonides against a pp28-luciferase-expressing HCMV Towne recombinant identified OZ418 to have the best selectivity; its effective concentration inhibiting viral growth by 50% (EC50) was 9.8 ± 0.2 µM, and cytotoxicity in noninfected human fibroblasts (the concentration inhibiting cell growth by 50% [CC50]) was 128.1 ± 8.0 µM. In plaque reduction assays, OZ418 inhibited HCMV TB40 in a concentration-dependent manner as well as a ganciclovir (GCV)-resistant HCMV isolate. The combination of OZ418 and GCV was synergistic in HCMV inhibition in vitro Virus inhibition by OZ418 occurred at an early stage and was dependent on the cell density at the time of infection. OZ418 treatment reversed HCMV-mediated cell cycle progression and correlated with the reduction of HCMV-induced expression of pRb, E2F1, and cyclin-dependent kinases 1, 2, 4, and 6. In an MCMV model, once-daily oral administration of OZ418 had significantly improved efficacy against MCMV compared to that of twice-daily oral AS. A parallel pharmacokinetic study with a single oral dose of OZ418 or AS showed a prolonged plasma half-life and higher unbound concentrations of OZ418 than unbound concentrations of AS. In summary, ozonides are proposed to be potential therapeutics, alone or in combination with GCV, for HCMV infection in humans.


Antiviral Agents/pharmacology , Cytomegalovirus Infections/drug therapy , Cytomegalovirus/drug effects , Heterocyclic Compounds, 1-Ring/pharmacology , Host-Pathogen Interactions/drug effects , Spiro Compounds/pharmacology , Virus Replication/drug effects , Animals , Antiviral Agents/blood , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Line , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Cytomegalovirus Infections/virology , Drug Administration Schedule , Drug Evaluation, Preclinical , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/pathology , Fibroblasts/virology , Ganciclovir/pharmacology , Gene Expression Regulation , Heterocyclic Compounds, 1-Ring/blood , Heterocyclic Compounds, 1-Ring/chemistry , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Signal Transduction , Spiro Compounds/blood , Spiro Compounds/chemistry , Spiro Compounds/pharmacokinetics
4.
Proc Natl Acad Sci U S A ; 113(48): E7818-E7827, 2016 11 29.
Article En | MEDLINE | ID: mdl-27856764

Induction of nucleotide-binding oligomerization domain 2 (NOD2) and downstream receptor-interacting serine/threonine-protein kinase 2 (RIPK2) by human cytomegalovirus (HCMV) is known to up-regulate antiviral responses and suppress virus replication. We investigated the role of nucleotide-binding oligomerization domain 1 (NOD1), which also signals through RIPK2, in HCMV control. NOD1 activation by Tri-DAP (NOD1 agonist) suppressed HCMV and induced IFN-ß. Mouse CMV was also inhibited through NOD1 activation. NOD1 knockdown (KD) or inhibition of its activity with small molecule ML130 enhanced HCMV replication in vitro. NOD1 mutations displayed differential effects on HCMV replication and antiviral responses. In cells overexpressing the E56K mutation in the caspase activation and recruitment domain, virus replication was enhanced, but in cells overexpressing the E266K mutation in the nucleotide-binding domain or the wild-type NOD1, HCMV was inhibited, changes that correlated with IFN-ß expression. The interaction of NOD1 and RIPK2 determined the outcome of virus replication, as evidenced by enhanced virus growth in NOD1 E56K mutant cells (which failed to interact with RIPK2). NOD1 activities were executed through IFN-ß, given that IFN-ß KD reduced the inhibitory effect of Tri-DAP on HCMV. Signaling through NOD1 resulting in HCMV suppression was IKKα-dependent and correlated with nuclear translocation and phosphorylation of IRF3. Finally, NOD1 polymorphisms were significantly associated with the risk of HCMV infection in women who were infected with HCMV during participation in a glycoprotein B vaccine trial. Collectively, our data indicate a role for NOD1 in HCMV control via RIPK2- IKKα-IRF3 and suggest that its polymorphisms predict the risk of infection.


Cytomegalovirus Infections/metabolism , Cytomegalovirus/physiology , Nod1 Signaling Adaptor Protein/physiology , Animals , Cells, Cultured , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/virology , Female , Gene Expression , Genetic Association Studies , Genetic Predisposition to Disease , Humans , I-kappa B Kinase/metabolism , Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Mice, Inbred BALB C , Nod2 Signaling Adaptor Protein/physiology , Polymorphism, Single Nucleotide , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Signal Transduction , Virus Replication
5.
Sci Rep ; 6: 20295, 2016 Feb 02.
Article En | MEDLINE | ID: mdl-26830977

We recently reported that induction of NOD2 by human Cytomegalovirus (HCMV) resulted in virus inhibition and upregulation of antiviral and inflammatory cytokines. Here we investigated the effects of muramyl dipeptide (MDP), a bacterial cell wall component that activates NOD2, on HCMV replication and antiviral responses. HCMV infection of human foreskin fibroblasts induced NOD2, the downstream receptor-interacting serine/threonine-protein kinase 2 (RIPK2), resulting in phosphorylation of TANK-binding kinase 1 (TBK1) and interferon regulatory factor 3 (IRF3). MDP treatment following infection at low multiplicity (MOI = 0.1 PFU/cell) inhibited HCMV in a dose-dependent manner and further induced phosphorylation of TBK1, IRF3 and expression of IFN-ß. None of these effects of MDP were observed following infection at multiplicity of 1. In infected NOD2 knocked-down cells MDP did not induce IFN-ß, irrespective of MOI. Treatment with MDP before infection also inhibited HCMV, an effect augmented with treatment duration. Treatment with an IFN-ß receptor blocking antibody or knockdown of IFN-ß significantly attenuated the inhibitory effect of MDP on HCMV. MDP treatment before or after infection with herpesvirus 1 did not inhibit its replication. Summarized, NOD2 activation exerts anti-HCMV activities predominantly via IFN-ß. Since MDP is a bacterial cell wall component, ongoing microbial exposure may influence HCMV replication.


Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/virology , Cytomegalovirus/drug effects , Interferon-beta/metabolism , Signal Transduction/drug effects , Virus Replication/drug effects , Cell Line , Cytokines/genetics , Cytokines/metabolism , Cytomegalovirus Infections/genetics , Fibroblasts , Humans , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/metabolism , Interferon-beta/genetics , NF-kappa B/metabolism , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , Protein Serine-Threonine Kinases/metabolism
6.
Antimicrob Agents Chemother ; 59(7): 3870-9, 2015 Jul.
Article En | MEDLINE | ID: mdl-25870074

Artemisinin-derived monomers and dimers inhibit human cytomegalovirus (CMV) replication in human foreskin fibroblasts (HFFs). The monomer artesunate (AS) inhibits CMV at micromolar concentrations, while dimers inhibit CMV replication at nanomolar concentrations, without increased toxicity in HFFs. We report on the variable anti-CMV activity of AS compared to the consistent and reproducible CMV inhibition by dimer 606 and ganciclovir (GCV). Investigation of this phenomenon revealed that the anti-CMV activity of AS correlated with HFFs synchronized to the G0/G1 stage of the cell cycle. In contact-inhibited serum-starved HFFs or cells arrested at early/late G1 with specific checkpoint regulators, AS and dimer 606 efficiently inhibited CMV replication. However, in cycling HFFs, in which CMV replication was productive, virus inhibition by AS was significantly reduced, but inhibition by dimer 606 and GCV was maintained. Cell cycle analysis in noninfected HFFs revealed that AS induced early G1 arrest, while dimer 606 partially blocked cell cycle progression. In infected HFFs, AS and dimer 606 prevented the progression of cell cycle toward the G1/S checkpoint. AS reduced the expression of cyclin-dependent kinases (CDK) 2, 4, and 6 in noninfected cycling HFFs, while the effect of dimer 606 on these CDKs was moderate. Neither compound affected CDK expression in noninfected contact-inhibited HFFs. In CMV-infected cells, AS activity correlated with reduced CDK2 levels. CMV inhibition by AS and dimer 606 also correlated with hypophosphorylation (activity) of the retinoblastoma protein (pRb). AS activity was strongly associated with pRb hypophosphorylation, while its reduced anti-CMV activity was marked by pRb phosphorylation. Roscovitine, a CDK2 inhibitor, antagonized the anti-CMV activities of AS and dimer 606. These data suggest that cell cycle modulation through CDKs and pRb might play a role in the anti-CMV activities of artemisinins. Proteins involved in this modulation may be identified and targeted for CMV inhibition.


Antimalarials/pharmacology , Antiviral Agents/pharmacology , Artemisinins/pharmacology , Cell Cycle/drug effects , Cytomegalovirus/drug effects , Cells, Cultured , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/biosynthesis , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/biosynthesis , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/biosynthesis , Drug Resistance, Viral , Enzyme Inhibitors/pharmacology , Fibroblasts/virology , G1 Phase/drug effects , Ganciclovir/pharmacology , Humans , Phosphorylation , Purines/pharmacology , Resting Phase, Cell Cycle/drug effects , Retinoblastoma Protein/metabolism , Roscovitine , Virus Replication/drug effects
7.
PLoS One ; 9(3): e92704, 2014.
Article En | MEDLINE | ID: mdl-24671169

Nucleotide-binding oligomerization domain 2 (NOD2) is an important innate immune sensor of bacterial pathogens. Its induction results in activation of the classic NF-κB pathway and alternative pathways including type I IFN and autophagy. Although the importance of NOD2 in recognizing RNA viruses has recently been identified, its role in sensing DNA viruses has not been studied. We report that infection with human cytomegalovirus (HCMV) results in significant induction of NOD2 expression, beginning as early as 2 hours post infection and increasing steadily 24 hours post infection and afterwards. Infection with human herpesvirus 1 and 2 does not induce NOD2 expression. While the HCMV-encoded glycoprotein B is not required for NOD2 induction, a replication competent virion is necessary. Lentivirus-based NOD2 knockdown in human foreskin fibroblasts (HFFs) and U373 glioma cells leads to enhanced HCMV replication along with decreased levels of interferon beta (IFN-ß) and the pro-inflammatory cytokine, IL8. NOD2 induction in HCMV-infected cells activates downstream NF-κB and interferon pathways supported by reduced nuclear localization of NF-κB and pIRF3 in NOD2 knockdown HFFs. Stable overexpression of NOD2 in HFFs restricts HCMV replication in association with increased levels of IFN-ß and IL8. Similarly, transient overexpression of NOD2 in U373 cells or its downstream kinase, RIPK2, results in decreased HCMV replication and enhanced cytokine responses. However, overexpression of a mutant NOD2, 3020insC, associated with severe Crohn's disease, results in enhanced HCMV replication and decreased levels of IFN-ß in U373 cells. These results show for the first time that NOD2 plays a significant role in HCMV replication and may provide a model for studies of HCMV recognition by the host cell and HCMV colitis in Crohn's disease.


Cytomegalovirus/physiology , Immunity, Innate , Nod2 Signaling Adaptor Protein/metabolism , Virus Replication/physiology , Cell Line , Cytokines/metabolism , Cytomegalovirus/radiation effects , Cytomegalovirus Infections/immunology , Fibroblasts/metabolism , Fibroblasts/radiation effects , Fibroblasts/virology , Gene Knockdown Techniques , Herpesviridae/physiology , Herpesviridae/radiation effects , Humans , Immunity, Innate/radiation effects , Inflammation Mediators/metabolism , Interferons/metabolism , Kinetics , Male , Mutant Proteins/metabolism , NF-kappa B/metabolism , Nod2 Signaling Adaptor Protein/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ultraviolet Rays , Viral Envelope Proteins/metabolism , Virus Inactivation/radiation effects , Virus Replication/radiation effects
8.
Antimicrob Agents Chemother ; 58(2): 986-94, 2014.
Article En | MEDLINE | ID: mdl-24277030

Conventional therapy for human cytomegalovirus (CMV) relies on inhibition of the viral DNA polymerase. Ganciclovir (GCV) is the first-line therapy, but when GCV-resistant strains emerge, alternative therapies are extremely limited and are associated with significant toxicities. Combination of anti-CMV agents that act on different targets or stages of virus replication has not been well studied, mostly because of the limited number of anti-CMV agents. We report our investigation of combinations of agents that inhibit CMV by targeting the viral DNA polymerase, cellular kinases, or other cell/virus mechanisms yet to be discovered. The selected compounds differed by the slopes of their dose-response curve: compounds with a slope of 1 (GCV) representing one target or noncooperativity and compounds with high slopes indicating positive cooperativity. Analysis of anti-CMV drug combinations using the Bliss model (which accounts for the slope parameter) distinguished between combinations with synergistic, antagonistic, and additive activities. The combination of GCV and foscarnet was slightly synergistic; strong synergism was found when GCV was used with artemisinin-derived monomers or dimers or the MEK inhibitor U0126. The combination of GCV and cardiac glycosides (digoxin, digitoxin, and ouabain) was additive. The monomeric artemisinin artesunate was synergistic when combined with U0126 or the multikinase inhibitor sunitinib. However, the combination of artemisinin-derived dimers (molecular weights, 606 and 838) and U0126 or sunitinib was antagonistic. These results demonstrate that members of a specific drug class show similar patterns of combination with GCV and that the slope parameter plays an important role in the evaluation of drug combinations. Lastly, antagonism between different classes of CMV inhibitors may assist in target identification and improve the understanding of CMV inhibition by novel compounds.


Antiviral Agents/pharmacology , Cytomegalovirus/drug effects , Fibroblasts/drug effects , Virus Replication/drug effects , Artemisinins/pharmacology , Butadienes/pharmacology , Cell Line , Cytomegalovirus/growth & development , Digitoxin/pharmacology , Digoxin/pharmacology , Drug Combinations , Drug Interactions , Fibroblasts/virology , Foscarnet/pharmacology , Ganciclovir/pharmacology , Humans , Indoles/pharmacology , Nitriles/pharmacology , Ouabain/pharmacology , Pyrroles/pharmacology , Sunitinib
9.
Antimicrob Agents Chemother ; 57(6): 2761-7, 2013 Jun.
Article En | MEDLINE | ID: mdl-23571549

Infection with human cytomegalovirus (HCMV) continues to be a threat for pregnant women and immunocompromised hosts. Although limited anti-HCMV therapies are available, development of new agents is desired. The Wnt signaling pathway plays a critical role in embryonic and cancer stem cell development and is targeted by gammaherpesviruses, Epstein-Barr virus (EBV), and Kaposi's sarcoma-associated herpesvirus (KSHV). HCMV infects stem cells, including neural progenitor cells, during embryogenesis. To investigate the role of Wnt in HCMV replication in vitro, we tested monensin, nigericin, and salinomycin, compounds that inhibit cancer stem cell growth by modulating the Wnt pathway. These compounds inhibited the replication of HCMV Towne and a clinical isolate. Inhibition occurred prior to DNA replication but persisted throughout the full replication cycle. There was a significant decrease in expression of IE2, UL44, and pp65 proteins. HCMV infection resulted in a significant and sustained decrease in expression of phosphorylated and total lipoprotein receptor-related protein 6 (pLRP6 and LRP6, respectively), Wnt 5a/b, and ß-catenin and a modest decrease in Dvl2/3, while levels of the negative regulator axin 1 were increased. Nigericin decreased the expression of pLRP6, LRP6, axin 1, and Wnt 5a/b in noninfected and HCMV-infected cells. For all three compounds, a correlation was found between expression levels of Wnt 5a/b and axin 1 and HCMV inhibition. The decrease in Wnt 5a/b and axin 1 expression was more significant in HCMV-infected cells than noninfected cells. These data illustrate the complex effects of HCMV on the Wnt pathway and the fine balance between Wnt and HCMV, resulting in abrogation of HCMV replication. Additional studies are required to elucidate how HCMV targets Wnt for its benefit.


Antiviral Agents/pharmacology , Cytomegalovirus/drug effects , Monensin/pharmacology , Nigericin/pharmacology , Pyrans/pharmacology , Virus Replication/drug effects , Wnt Signaling Pathway/drug effects , Animals , Cell Line, Tumor , Cells, Cultured , Chlorocebus aethiops , Cytomegalovirus/metabolism , Cytomegalovirus/physiology , Fibroblasts/virology , Humans , Microbial Sensitivity Tests/methods , Vero Cells , Wnt Signaling Pathway/physiology
10.
ACS Chem Neurosci ; 4(7): 1093-100, 2013 Jul 17.
Article En | MEDLINE | ID: mdl-23530929

Retinoic acid (RA)-elicited signaling has been shown to play critical roles in development, organogenesis, and the immune response. RA regulates expression of Alzheimer's disease (AD)-related genes and attenuates amyloid pathology in a transgenic mouse model. In this study, we investigated whether RA can suppress the production of amyloid-ß (Aß) through direct inhibition of γ-secretase activity. We report that RA treatment of cells results in significant inhibition of γ-secretase-mediated processing of the amyloid precursor protein C-terminal fragment APP-C99, compared with DMSO-treated controls. RA-elicited signaling was found to significantly increase accumulation of APP-C99 and decrease production of secreted Aß40. In addition, RA-induced inhibition of γ-secretase activity was found to be mediated through significant activation of extracellular signal-regulated kinases (ERK1/2). Treatment of cells with the specific ERK inhibitor PD98059 completely abolished RA-mediated inhibition of γ-secretase. Consistent with these findings, RA was observed to inhibit secretase-mediated proteolysis of full-length APP. Finally, we have established that RA inhibits γ-secretase through nuclear retinoic acid receptor-α (RARα) and retinoid X receptor-α (RXRα). Our findings provide a new mechanistic explanation for the neuroprotective role of RA in AD pathology and add to the previous data showing the importance of RA signaling as a target for AD therapy.


Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Retinoid X Receptor alpha/pharmacology , Tretinoin/pharmacology , Cells, Cultured , Humans , MAP Kinase Signaling System , Signal Transduction
11.
Antimicrob Agents Chemother ; 56(9): 4891-9, 2012 Sep.
Article En | MEDLINE | ID: mdl-22777050

Infection with human cytomegalovirus (HCMV) continues to be a major threat for pregnant women and the immunocompromised population. Although several anti-HCMV therapies are available, the development of new anti-HCMV agents is highly desired. There is growing interest in identifying compounds that might inhibit HCMV by modulating the cellular milieu. Interest in cardiac glycosides (CG), used in patients with congestive heart failure, has increased because of their established anticancer and their suggested antiviral activities. We report that the several CG--digoxin, digitoxin, and ouabain--are potent inhibitors of HCMV at nM concentrations. HCMV inhibition occurred prior to DNA replication, but following binding to its cellular receptors. The levels of immediate early, early, and late viral proteins and cellular NF-κB were significantly reduced in CG-treated cells. The activity of CG in infected cells correlated with the expression of the potassium channel gene, hERG. CMV infection upregulated hERG, whereas CG significantly downregulated its expression. Infection with mouse CMV upregulated mouse ERG (mERG), but treatment with CG did not inhibit virus replication or mERG transcription. These findings suggest that CG may inhibit HCMV by modulating human cellular targets associated with hERG and that these compounds should be studied for their antiviral activities.


Antiviral Agents/pharmacology , Cardiac Glycosides/pharmacology , Cytomegalovirus/drug effects , Ether-A-Go-Go Potassium Channels , Viral Proteins/metabolism , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Cytomegalovirus/growth & development , Digitoxin/pharmacology , Digoxin/pharmacology , Ether-A-Go-Go Potassium Channels/agonists , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/genetics , Fibroblasts/drug effects , Fibroblasts/virology , Gene Expression , Genes, Reporter , Humans , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , Ouabain/pharmacology , Transcriptional Activation , Viral Proteins/genetics , Virus Replication/drug effects
12.
Antimicrob Agents Chemother ; 56(7): 3508-15, 2012 Jul.
Article En | MEDLINE | ID: mdl-22547612

We previously reported that among a series of artemisinin-derived monomers and dimers, dimer diphenyl phosphate (838) was the most potent inhibitor of human cytomegalovirus (CMV) replication. Our continued investigation of a prototypic artemisinin monomer (artesunate [AS]) and dimer (838) now reveals that both compounds have specific activity against CMV but do not inhibit lytic replication of human herpesvirus 1 or 2 or Epstein-Barr virus. AS and 838 inhibited CMV replication during the first 24 h of the virus replication cycle, earlier than the time of ganciclovir (GCV) activities and prior to DNA synthesis. Neither compound inhibited virus entry. Quantification of DNA replication and virus yield revealed a similar level of inhibition by GCV, but AS and 838 had a 10-fold-higher inhibition of virus yield than of DNA replication, suggesting that artemisinins could inhibit CMV through multiple steps: a predominant early inhibition and possibly an additional step following DNA replication. During the strong early CMV inhibition, the transcription of immediate-early genes was not significantly downregulated, and viral protein expression was reduced only after 48 h. AS and GCV were reversible CMV inhibitors, but the inhibition of CMV replication by 838 was irreversible. Combinations of GCV and 838 as well as GCV and AS were highly synergistic. Finally, treatment with 838, but not AS, prior to CMV infection demonstrated strong anti-CMV activity. These findings illustrate the unique activities of dimer 838, including early and irreversible CMV inhibition, possibly by tight binding to its target.


Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Artemisinins/chemistry , Cytomegalovirus/drug effects , Virus Replication/drug effects , Animals , Blotting, Western , Cell Line , Chlorocebus aethiops , Cytomegalovirus/genetics , DNA Replication/drug effects , Fluorescent Antibody Technique , Humans , Vero Cells
13.
J Alzheimers Dis ; 22(2): 423-42, 2010.
Article En | MEDLINE | ID: mdl-20847442

Amyloidogenic processing of amyloid-ß precursor protein (AßPP) is associated with cholesterol- and sphingolipid-rich lipid rafts. Caveolin-1, a raft-residing protein, has been implicated in the pathogenesis of Alzheimer's disease. To determine the role of caveolin-1 in governing γ-secretase-mediated AßPP proteolysis, cellular γ-secretase activity was assessed in response to alteration in caveolin-1 expression. We demonstrated that suppression of caveolin-1 expression by RNA interference resulted in a significant increase in γ-secretase-mediated proteolysis of AßPP, generation of amyloid-ß, and cleavage of Notch. Overexpression of caveolin-1 attenuated γ-secretase-mediated proteolysis of AßPP and Notch, substantiating the negative regulation of γ-secretase by caveolin-1. Furthermore, we found that cells deficient in caveolin-1 exhibited significantly increased co-localization of γ-secretase with clathrin-coated non-caveolar endocytic vesicles, demonstrating that the partitioning of γ-secretase between caveolar and non-caveolar membranes can be modulated by caveolin-1. Our data also showed that JNK activation is essential for caveolin-1-mediated regulation of γ-secretase. Together, our results strongly suggest that caveolin-1 is an important regulator of γ-secretase activity.


Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Caveolin 1/metabolism , Cell Membrane/enzymology , Membrane Microdomains/metabolism , Amyloid Precursor Protein Secretases/genetics , Cell Line, Transformed , Cell Membrane/ultrastructure , Enzyme-Linked Immunosorbent Assay/methods , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , MAP Kinase Kinase 4/metabolism , Membrane Microdomains/drug effects , Microscopy, Confocal/methods , Mutation/genetics , RNA, Small Interfering/pharmacology , Receptors, Notch/genetics , Receptors, Notch/metabolism , Subcellular Fractions , Transfection
14.
Indian J Pharmacol ; 41(1): 47-8, 2009 Feb.
Article En | MEDLINE | ID: mdl-20177582

Quinupristin and dalfopristin combination has been advocated as a drug of choice for multi-drug resistant (MDR) gram-positive cocci (GPC). We are reporting two cases of neonatal septicemia, caused by the methicillin resistant Staphylococcus aureus (MRSA), showing primary in vitro pristinamycin resistance. The Minimum inhibitory concentrations (MIC) for pristinamycin in these two cases were 30 mug and 25 mug, respectively. Universal advocacy of pristinamycin for the therapy of MDR GPC infections should be re-evaluated.

15.
Cancer Lett ; 223(2): 275-84, 2005 Jun 08.
Article En | MEDLINE | ID: mdl-15896462

Human keratinocytes (HaCaT) were exposed to UV (A+B) (UVA-350-400 mJ/cm2 and UVB-30 mJ/cm2) which induces apoptosis as evidenced by MTT assay, DNA laddering, Bax and Fas up-regulation. UV induced apoptotic conditioned media (6 h or earlier) did not cause apoptosis in unexposed cells. However, treatment with conditioned medium collected post UV exposure (1 h) induced Bax in unexposed cells as observed by RT-PCR. The induction of cell death was initiated by conditioned medium collected 12 h after UV exposure and the extent of death was increased progressively when conditioned medium collected 24 or 72 h post UV exposure was used. Medium collected 24 h after UV exposure also increased mitochondrial membrane permeability as determined by rhodamine uptake. Conditioned medium induced apoptosis did not involve reactive oxygen species (ROS) unlike UV induced apoptosis indicating that the apoptosis pathway could be different. Interestingly, at high dilution apototic conditioned medium did not induce apoptosis but actually protected cells from UV insult. The role of nerve growth factor (NGF) in UV induced bystander effects are also discussed.


Apoptosis , Bystander Effect , DNA Damage , Keratinocytes/physiology , Ultraviolet Rays , Cell Membrane Permeability , Culture Media, Conditioned , Formazans/pharmacology , Gene Expression Regulation , Humans , Mitochondria , Reactive Oxygen Species , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tetrazolium Salts/pharmacology , Up-Regulation
...