ArticlePDF Available

Antiviral Activity of (+)-Rutamarin against Kaposi's Sarcoma-Associated Herpesvirus by Inhibition of the Catalytic Activity of Human Topoisomerase II

American Society for Microbiology
Antimicrobial Agents and Chemotherapy
Authors:

Abstract and Figures

Kaposi's sarcoma-associated herpesvirus (KSHV) is an etiological agent of several AIDS-associated malignancies, including Kaposi's sarcoma (KS), primary effusion lymphoma (PEL), and multicentric Castleman's disease (MCD). Its lytic replication cycle has been proven to be critical for the pathogenesis of KSHV-associated diseases. In KS lesions, lytic viral replication, production of virion particles, and reinfection of endothelial cells are essential to sustain the population of infected cells that otherwise would be quickly lost as spindle cells divide. Thus, antivirals that block KSHV replication could be a strategy in the treatment of KSHV-associated diseases. However, there is no effective anti-KSHV drug currently available. Our previous work showed that human topoisomerase II (Topo II) is indispensable for KSHV lytic replication and is suggested to be an effective target for antiviral drugs. Here, we report the discovery and characterization of a novel catalytic inhibitor of human Topo IIα, namely, (+)-rutamarin. The binding mode of (+)-rutamarin to the ATPase domain of human Topo IIα was established by docking and validated by molecular dynamics (MD) simulations. More importantly, (+)-rutamarin efficiently inhibits KSHV lytic DNA replication in BCBL-1 cells with a half-maximal inhibitory concentration (IC50) of 1.12 μM and blocks virion production with a half-maximal antiviral effective concentration (EC50) of 1.62 μM. It possesses low cytotoxicity, as indicated by the selectivity index (SI) of 84.14. This study demonstrated great potential for (+)-rutamarin to become an effective drug for treatment of human diseases associated with KSHV infection.
This content is subject to copyright. Terms and conditions apply.
Antiviral Activity of ()-Rutamarin against Kaposi’s Sarcoma-
Associated Herpesvirus by Inhibition of the Catalytic Activity of
Human Topoisomerase II
Bo Xu,
a,b
Ling Wang,
c
Lorenzo González-Molleda,
d
Yan Wang,
a,e
Jun Xu,
c
Yan Yuan
a,b,d
Institute of Human Virology
a
and Key Laboratory of Tropical Disease Control,
b
Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, China; Research
Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China
c
; Department of Microbiology, University of
Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, USA
d
; Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, China
e
Kaposi’s sarcoma-associated herpesvirus (KSHV) is an etiological agent of several AIDS-associated malignancies, including Ka-
posi’s sarcoma (KS), primary effusion lymphoma (PEL), and multicentric Castleman’s disease (MCD). Its lytic replication cycle
has been proven to be critical for the pathogenesis of KSHV-associated diseases. In KS lesions, lytic viral replication, production
of virion particles, and reinfection of endothelial cells are essential to sustain the population of infected cells that otherwise
would be quickly lost as spindle cells divide. Thus, antivirals that block KSHV replication could be a strategy in the treatment of
KSHV-associated diseases. However, there is no effective anti-KSHV drug currently available. Our previous work showed that
human topoisomerase II (Topo II) is indispensable for KSHV lytic replication and is suggested to be an effective target for antivi-
ral drugs. Here, we report the discovery and characterization of a novel catalytic inhibitor of human Topo II, namely, ()-rut-
amarin. The binding mode of ()-rutamarin to the ATPase domain of human Topo IIwas established by docking and vali-
dated by molecular dynamics (MD) simulations. More importantly, ()-rutamarin efficiently inhibits KSHV lytic DNA
replication in BCBL-1 cells with a half-maximal inhibitory concentration (IC
50
) of 1.12 M and blocks virion production with a
half-maximal antiviral effective concentration (EC
50
) of 1.62 M. It possesses low cytotoxicity, as indicated by the selectivity
index (SI) of 84.14. This study demonstrated great potential for ()-rutamarin to become an effective drug for treatment of hu-
man diseases associated with KSHV infection.
Kaposi’s sarcoma-associated herpesvirus (KSHV), also known
as human herpesvirus 8 (HHV-8), is a member of the human
gammaherpesvirus family. It has been proven to be the etiologic
agent of Kaposi’s sarcoma (KS), a multicentric malignant neo-
plasm of endothelial origin (1–5). Although classic KS is a rare
disease and in general nonfatal, it often occurs in immunocom-
promised patients who are under immunosuppression treatment
after organ transplantation. In the AIDS epidemic, KS has become
the most common AIDS-associated malignancy and has led to
significant mortality (6). Approximately 20% of AIDS patients
develop KS during the course of their disease, and AIDS-associ-
ated KS is estimated to contribute to 10% of the deaths of AIDS
patients (7). Besides KS, two B cell-associated lymphoproliferative
disorders, namely, primary effusion lymphoma (PEL) and multi-
centric Castleman’s disease (MCD), are also related to KSHV in-
fection and mainly occur in AIDS patients (6,8). The incidence of
KSHV-associated MCD has risen since the advent of highly active
antiretroviral therapy (HAART) (9).
The contemporary treatment modalities for KS and other
KSHV-associated malignancies include conventional cancer ther-
apies, such as radiation and chemotherapy, or AIDS treatment,
such as HAART (6). These cancer therapies in general have serious
side effects, and the response of tumors to them is only transient.
The success of human papillomavirus (HPV) vaccine in prevent-
ing cervical cancer has proven that antiviral-oriented therapies
have the potential to be used to treat human malignant diseases
that are caused by viruses. Thus, an antiviral targeting KSHV
could prove successful at treating KSHV-associated tumors (10).
However, currently, there is no therapy for KSHV-associated dis-
ease based on targeting of KSHV.
Like all herpesviruses, KSHV has two types of replication cycle,
latent and lytic replication. In KS lesions, most KSHV-trans-
formed spindle-shaped cells are in the latent replication phase, but
a small percentage of infected cells undergo spontaneous lytic rep-
lication (11–13). Increasing evidence suggests that the lytic repli-
cation in these cells is necessary for maintaining stable infection
and viral pathogenicity. The majority of KSHV genes, including
those responsible for malignant cell growth and alteration of the
microenvironment, are expressed only in the lytic phase (14). Fur-
thermore, lytic replication is necessary for sustaining the popula-
tion of latently infected cells that otherwise would be quickly lost
by segregation of latent viral episomes as spindle cells divide (15).
Thus, KSHV lytic replication and constant primary infection of
fresh cells are crucial, not only for viral propagation, but also for
viral pathogenesis. Blockade of the lytic replication of KSHV could
be a good strategy for the treatment of KS or other KSHV-associ-
ated human diseases.
Received 14 June 2013 Returned for modification 8 July 2013
Accepted 11 October 2013
Published ahead of print 2 December 2013
Address correspondence to Yan Yuan, yuan2@pobox.upenn.edu, or Jun Xu,
xujun9@mail.sysu.edu.cn.
B.X., L.W., and L.G.-M. contributed equally to this article.
Supplemental material for this article may be found at http://dx.doi.org/10.1128
/AAC.01259-13.
Copyright © 2014, American Society for Microbiology. All Rights Reserved.
doi:10.1128/AAC.01259-13
January 2014 Volume 58 Number 1 Antimicrobial Agents and Chemotherapy p. 563–573 aac.asm.org 563
Viral DNA replication is considered an ideal target for antivi-
rals. Our laboratory has been studying the mechanisms that con-
trol KSHV lytic DNA replication (16–19). One of our findings was
that several host cellular proteins, including topoisomerase I
(Topo I) and Topo II, MSH2/6, RecQL, and poly(ADP-ribose)
polymerase 1 (PARP-1), are involved in KSHV lytic DNA replica-
tion (19). We also demonstrated that both Topo I and Topo II are
indispensable for KSHV lytic replication and that specific inhibi-
tors of Topo I and II can effectively inhibit KSHV lytic replication.
One category of Topo II inhibitors, namely, catalytic Topo II in-
hibitor, was found to provide marked inhibition of KSHV repli-
cation with minimal cytotoxicity, as indicated by their high selec-
tivity indices (e.g., 31.6 for novobiocin) (20). As such, Topo II can
serve as an effective target for antivirals, and catalytic inhibitors of
Topo II represent promising antiviral agents for the treatment of
malignancies associated with KSHV infection.
In this study, we attempted to search for new compounds with
higher Topo II inhibition properties and lower cytotoxicities.
With a ligand-based virtual screening approach, we screened
more than 7,200 compound structures in our compound reposi-
tory and identified a set of coumarin derivatives that showed ac-
tivities in inhibition of KSHV DNA replication equal to or greater
than that of novobiocin. In particular, ()-rutamarin exhibits the
highest efficiency in blocking KSHV lytic replication. We demon-
strated that ()-rutamarin has human Topo II inhibition activity
and belongs to the category of catalytic Topo II inhibitors.
MATERIALS AND METHODS
Cells and plasmids. BCBL-1 and JSC-1 are two primary effusion lym-
phoma cell lines that are latently infected with KSHV (21,22). The BJAB
cell line is a KSHV-negative B cell line isolated from Burkitt’s lymphoma.
These B cells were grown in RPMI 1640 medium (Gibco-BRL, Gaithers-
burg, MD) supplemented with 10% fetal bovine serum (Gibco-BRL),
penicillin-streptomycin (50 units/ml), and 1.25 g/ml amphotericin
B-1.25g/ml sodium deoxycholate (Fungizone). Peripheral blood mono-
nuclear cells (PBMC) were isolated from whole blood of a healthy donor
and cultured in RPMI 1640 medium supplemented with 10% fetal bovine
serum, penicillin-streptomycin, and amphotericin B-sodium deoxy-
cholate.
Plasmid pOri-A contains an EcoRI-PstI fragment (nucleotides 22409
to 26491) of KSHV DNA in pBluescript at the EcoRI/PstI site, as previ-
ously described (16). pCR3.1-ORF50 is an RTA expression vector de-
scribed previously (16).
Chemicals and treatment of cells. The chemicals used in this study
were from the Guangdong Small Molecule Tangible Library (GSMTL)
(23). The compounds were dissolved in dimethyl sulfoxide (DMSO) and
serially diluted before being added to BCBL-1, JSC-1, or BJAB cell cultures
(4 10
5
cells/ml). The final DMSO concentration in the culture medium
was maintained at 1%. For lytic replication, BCBL-1 cells were induced by
tetradecanoylphorbol acetate (TPA) as previously reported (20). JSC-1
cells were induced by 3 mM sodium butyrate (22). Three hours postin-
duction, the compounds in dilutions were added to BCBL-1 or JSC-1 cells,
and subsequently, the antiviral effects and cytotoxicity were assayed at
different time points.
Analysis of intracellular KSHV genomic DNA content and chemical
effects. TPA-induced and uninduced BCBL-1 cells were harvested at 48 h
postinduction, and total DNA was purified using a DNeasy kit according
to the manufacturer’s protocol (Qiagen). The KSHV genomic copy num-
ber was quantified by real-time PCR on a Roche LightCycler instrument
using the LightCycler FastStart DNA MasterPlus SYBR green kit with
primers for the detection of LANA (forward, 5=-CGCGAATACCGCTAT
GTACTCA-3=; reverse, 5=-GGAACGCGCCTCATACGA-3=). The intra-
cellular viral genomic DNA in each sample was normalized to GAPDH
(glyceraldehyde-3-phosphate dehydrogenase) using primers directed to
GAPDH (forward, 5=-ACATCATCCCTGCCTCTAC-3=; reverse, 5=-TCA
AAGGTGGAGGAGTGG-3=).
The half-maximal inhibitory concentration (IC
50
) values of com-
pounds were determined from a dose-response curve of KSHV DNA con-
tent values from TPA-induced and chemical-treated cells. The viral DNA
contents with those of uninduced cells subtracted were divided by those of
the control cells with no drug treatment and then represented on the y
axes of dose-response curves: yaxis value (TPA
X
no TPA
X
)/(TPA
0
no TPA
0
), where Xis any concentration of the drug and 0 represents
nondrug treatment. The IC
50
on viral DNA synthesis for each compound
was calculated with the aid of GraphPad Prism software.
Analysis of extracellular KSHV virion production and chemical ef-
fects. Five days postinduction with TPA, BCBL-1 culture media were
collected and extracellular virions were pelleted from the medium super-
natant. To remove contaminating DNA outside viral particles, the con-
centrated viruses were treated with Turbo DNase I (Ambion) at 37°C for
1 h, followed by proteinase K digestion. Virion DNA was extracted with
phenol-chloroform, precipitated with ice-cold ethanol, and then dis-
solved in Tris-EDTA (TE) buffer. The KSHV genomic copy numbers were
determined by real-time PCR, and the values were corrected as described
above. The half-maximal antiviral effective concentration (EC
50
) values
were calculated from dose-response curves with GraphPad Prism soft-
ware.
Cytotoxicity assay. The viabilities of BCBL-1 cells treated or not with
chemicals were assessed by counting Trypan blue-stained cells 2 or 5 days
posttreatment using a light microscope. Cell viabilities were defined rela-
tive to control cells (non-drug treated). The half-maximal cytotoxic con-
centration (CC
50
) was calculated from dose-response curves with Graph-
Pad Prism software.
Cell proliferation assay. BCBL-1 and BJAB cells (both starting with
210
5
cells/ml) were treated with ()-rutamarin for 5 days at two
different concentrations: the IC
50
and 5 times the IC
50
. The cells were
stained with Trypan blue and counted every day for 5 days. To maintain
the exponential growth, fresh medium (supplemented or not with the
drug) was added to the cultures every 2 days.
Cell cycle assay. BCBL-1 and BJAB cells (both starting with 4 10
5
cells/ml) were treated with ()-rutamarin and novobiocin at two differ-
ent concentrations: the IC
50
and 5 times the IC
50
. Two days posttreat-
ment, cells were collected and fixed with cold 70% ethanol for 15 min,
permeabilized, and stained with propidium iodide (PI) solution (50 g/ml
of PI, 0.1 mg/ml of RNase A, and 0.05% Triton X-100) for 2 h. Cell cycle
progression was measured using a FACStar Plus cell sorter flow cytometer
(Becton, Dickinson).
ori-Lyt-dependent DNA replication assay. To assess the effect of ()-
rutamarin on ori-Lyt-dependent DNA replication, BCBL-1 cells were
cotransfected with plasmids pOri-A (2.5 g) and pCR3.1-ORF50 (2.5 g)
by nucleoporation (Amaxa) and cultured with different concentrations of
()-rutamarin. Seventy-two hours posttransfection, extrachromosomal
DNA was prepared from cells using the Hirt DNA extraction method as
previously described (20). The extracted extrachromosomal DNA was
treated with RNase A at 25°C for 30 min, followed by proteinase K at 50°C
for 30 min. Five micrograms of DNA was digested with KpnI/SacI or
KpnI/SacI/DpnI (New England Bio-Labs), separated on a 1% agarose gel,
and transferred onto GeneScreen membranes (PerkinElmer, Boston,
MA). The Southern blots were hybridized with a
32
P-labeled pBluescript
plasmid in 5SSC (1SSC is 0.15 M NaCl plus 0.015 M sodium citrate),
2Denhardt’s solution, 1% SDS, and 50 g/ml denatured salmon sperm
DNA at 68°C.
Kinetoplast DNA decatenation assay. The decatenation assay is de-
signed to measure type II topoisomerase activity and inhibition of the
enzyme by testing compounds. The kinetoplast DNA (kDNA) (200 ng)
was incubated in a 20-l reaction mixture containing 50 mM Tris-HCl
(pH 7.5), 85 mM KCl, 10 mM MgCl
2
, 0.5 mM Na
2
EDTA, 0.5 mM dithio-
threitol, 1 mM ATP, 30 g/ml bovine serum albumin (BSA), and 2 units
Xu et al.
564 aac.asm.org Antimicrobial Agents and Chemotherapy
of Topo IIor Topo IIfor 30 min at 37°C in the absence or the presence
of ()-rutamarin or control compounds (novobiocin and etoposide).
The reactions were analyzed by electrophoresis on agarose gels. The de-
catenated kDNAs were measured with the Gel Dox XRimaging system
(Bio-Rad).
Malachite green assay for ATPase activity of Topo II.Malachite
green reagent was prepared by mixing malachite green (0.0812% [wt/
vol]), polyvinyl alcohol (2.32% [wt/vol]), ammonium molybdate (5.72%
[wt/vol] in 6 M HCl), and ultrapure water in a ratio of 2:1:1:2. An ATP
hydrolysis reaction with a mixture (20 l) containing 50 mM Tris-HCl,
pH 7.4, 85 mM KCl, 10 mM MgCl
2
, 0.5 mM dithiothreitol (DTT), 0.5 mM
Na
2
EDTA, 30 g/ml BSA, 200 ng pBR322 DNA, 2 units Topo II, 500 M
ATP, and various concentrations of ()-rutamarin was initiated by add-
ing ATP to the reaction mixture and incubated at 37°C for 2 h. The reac-
tion was stopped by adding 80 l of the malachite green reagent to the
reaction mixture, followed by adding 10 l of 34% sodium citrate to
develop a blue-green color change. The absorbance at 620 nm was mea-
sured using a plate reader. The optical density at 620 nm (OD
620
) was used
to represent the ATP hydrolysis level.
Molecular docking study. The X-ray crystal structure of the ATPase
domain of human Topo IIin complex with ADPNP (Protein Data Bank
[PDB] ID, 1ZXM; resolution, 1.87Å) was retrieved from the Protein Data
Bank (24). Only chain A was kept for docking study. The missing loop
(residues 345 to 350) was added and optimized using Prepare Protein
Module encoded in Discovery Studio 3.5 (Accelrys Inc.) (25). The initial
structures of ()-rutamarin and ADPNP were optimized using MMFF
force field (26), and the Powell method was used for energy minimization
by default parameters in Discovery Studio 3.5.
The docking program FlexX encoded in SYBYL 7.3 (Tripos Inc.) was
applied to identify the potential binding of ()-rutamarin to the Topo II
ATPase domain. A previous study suggested that two conserved water
molecules were not neglected to distinguish the active, moderate, and
inactive ligands (27). In the present study, the receptor for docking sim-
ulation consisted of protein chain A, two conserved water molecules, and
Mg
2
. To validate the molecular-docking protocol, ADPNP was re-
docked into the crystal structure of the ATPase domain of Topo II. The
docked ADPNP has a binding pose similar to that of the cocrystallized
ligand, with a root mean square deviation (RMSD) of about 1.6 Å. The
active sites were defined as all residues within a 6.5-Å radius of the bound
ADPNP. Other FlexX parameters were set to default values. Thirty poses
were retained during the docking process. After running FlexX, all the
poses were visually inspected, and the most suitable docking pose was
selected on the basis of the score and interactions with key residues of the
active site. The complex of Topo IIand the most suitable docking con-
former of ()-rutamarin was used as the initial coordinates for the sub-
sequent molecular dynamics (MD).
MD simulations. MD simulations were performed in AMBER 12 (28)
with the AMBER ff99SB force field (29,30). The docked structure of the
Topo IIATPase domain with ()-rutamarin was used as the initial
coordinates for MD simulations. Parameters for ()-rutamarin were ob-
tained from the ANTECHAMBER module using the Generalized Amber
Force Field (GAFF) (31) with the RESP charge-fitting procedure with
input from Hartree-Fock calculations at the 6- to 31-G* level by using the
Gaussian03 program (32). All hydrogen atoms of the protein were added
using the tleap module, considering ionizable residues set at their default
protonation states at a neutral pH. The system was solvated in a truncated
octahedron box of TIP3P water molecules with a margin distance of 10 Å.
Neutralizing counterions were added to the simulation system. The de-
tailed MD simulations can be found in the supplemental material. The
molecular-mechanics Poisson-Boltzmann surface area (MM-PBSA) and
molecular-mechanics generalized born surface area (MM-GBSA) meth-
ods (33) in the AMBER 12 suite were used to calculate the binding free
energies (see the supplemental material).
RESULTS
()-Rutamarin inhibits the lytic replication of KSHV. To search
for a novel Topo II inhibitor(s) that effectively blocks KSHV lytic
DNA replication, a ligand-based virtual screening using an in-
house three-dimensional (3D) molecular superimposing algo-
rithm, WEGA (34), was conducted against the molecular struc-
tures in our compound repository, GSMTL (23). Using the
novobiocin structure as the template, we screened approximate
7,200 compounds from the GSMTL. The top 33 compounds that
are three-dimensionally most similar to novobiocin were selected
and subsequently tested for antiviral activities. Exponentially
growing BCBL-1 cells were induced into lytic replication with
TPA. Three hours after induction, the cells were exposed to each of
the compounds, as well as novobiocin, at a concentration of 20
M. Forty-eight hours postinduction, the KSHV genomic DNA
contents of the cells treated with these compounds were analyzed
by quantitative real-time PCR. Treatment with novobiocin re-
sulted in 42% inhibition of viral replication compared to the con-
trol cells treated with no drug. Seven compounds showed inhibi-
tory effects similar to or better than those of novobiocin on viral
DNA replication (Fig. 1A). Four of these, C28, C31, C32, and C33,
also displayed strong activities in blocking virion production (Fig.
1B). With the exception of C31, the compounds possess signifi-
cant cytotoxicity, as demonstrated by low cell viability rates with
the treatment (Fig. 1C). C31, which was identified as the natural
product ()-rutamarin (Fig. 2A), exhibited significant inhibition
of both viral DNA synthesis and virion production, with relatively
low cytotoxicity at a concentration of 20 M. Therefore, it was
chosen for further investigation.
The IC
50
values of ()-rutamarin were determined from the
dose-response curve of KSHV DNA content in TPA-induced
BCBL-1 cells and were found to be 1.12 M(
Fig. 2B). The effect of
()-rutamarin on progeny virion production was also deter-
mined by quantification of encapsidated viral DNA in BCBL-1 cell
culture media. The EC
50
calculated from the dose-response curve
of extracellular virions is 1.62 M(
Fig. 2B). The cytotoxicity of
()-rutamarin on BCBL-1 cells was examined in parallel with
inhibition of KSHV DNA replication and virion production. Cells
treated with ()-rutamarin at differing concentrations were
subjected to the trypan blue exclusion method to determine cell
viability in response to ()-rutamarin treatment. The CC
50
was
determined to be 94.24 M(
Fig. 2B). Low cytotoxicity of ()-
rutamarin results in an appreciable selectivity index (SI) (SI
CC
50
/IC
50
) of 84.14.
To make sure that ()-rutamarin inhibits KSHV lytic replica-
tion regardless of host cells and the means of lytic cycle induction,
we tested the effect of ()-rutamarin on KSHV replication in
JSC-1 cells induced by sodium butyrate for lytic replication. The
IC
50
,EC
50
, and CC
50
of ()-rutamarin on butyrate-induced
JSC-1 cells were found to be 2.29 M, 1.40 M, and 94.34 M,
respectively, and are very similar to those obtained with BCBL-1.
In addition, the cytotoxicity of ()-rutamarin to primary lym-
phocytes was also assessed with PBMC. The CC
50
for PBMC was
calculated to be 60.91 M (see Fig. S1 in the supplemental mate-
rial).
Evaluation of ()-rutamarin for its effects on host cell pro-
liferation and cell cycle progression. To further investigate the
potential of ()-rutamarin to become an effective and safe anti-
viral, the effect of the compound on cell proliferation was assessed.
()-Rutamarin Inhibits KSHV Replication
January 2014 Volume 58 Number 1 aac.asm.org 565
KSHV-carrying BCBL-1 and virus-free BJAB cells were cultured
in the presence and absence of ()-rutamarin, and live cells were
counted over 5 days. ()-Rutamarin did not exhibit a cell growth-
inhibitory effect in the two cell lines at the IC
50
. At an excess
concentration (5 times the IC
50
), ()-rutamarin showed an ad-
verse effect on cell growth, but the extent of inhibition was less
than that with novobioicin (Fig. 3). The effect of ()-rutamarin
on cell cycle progression was also examined. BCBL-1 cells treated
with ()-rutamarin were stained with propidium iodide and sub-
jected to flow cytometric analysis. At the IC
50
, no effect on the cell
cycle pattern was observed with either ()-rutamarin or novobio-
cin in comparison with the vehicle control. At an excess concen-
tration (5 times the IC
50
)of()-rutamarin, a slight increase in the
S phase was observed in comparison to untreated cells. However,
the influence of ()-rutamarin on cell cycle progression was ob-
served to be less than that of novobiocin (Fig. 4). Overall, these
results demonstrated that ()-rutamarin has minimal effects on
host cell proliferation and cell cycle progression in its pharmaceu-
tical dose range.
()-Rutamarin inhibits KSHV lytic replication by blocking
viral ori-Lyt-dependent DNA replication. We have demon-
strated that ()-rutamarin is capable of inhibiting KSHV DNA
synthesis and virion production. To confirm whether the inhibi-
tory effect of ()-rutamarin on KSHV lytic replication is directly
due to the blocking of ori-Lyt-dependent DNA replication, we
conducted an ori-Lyt DNA replication assay. BCBL-1 cells were
cotransfected with an ori-Lyt-containing plasmid (pOri-A) and an
RTA expression vector. Lytic DNA replication was induced by
RTA expression (35). The transfected cells were cultured in the
presence of ()-rutamarin at various concentrations, and the ef-
fect of ()-rutamarin on ori-Lyt-dependent DNA replication was
measured by a DpnI assay (16,17). In brief, DNA was isolated
from the treated cells 72 h posttransfection and digested with
KpnI/SacI and KpnI/SacI/DpnI. Replicated plasmid DNA was dis-
tinguished from input plasmids by DpnI restriction digestion,
which cleaves input DNA that has been Dam
methylated in Esch-
erichia coli but leaves intact the DNA that has been replicated in at
least one round in eukaryotic cells. Thus, only newly replicated
plasmid DNA in BCBL-1 cells, which is resistant to DpnI diges-
tion, can be detected in Southern blot analysis. Replicated pOri-A
DNA was detected as a DpnI-resistant DNA band in the cells that
were cotransfected with pOri-A and an RTA expression vector.
However, the replicated DNA decreased with elevated concentra-
tions of ()-rutamarin in the cell culture (Fig. 5). This result
FIG 1 Compound screening. (A) Thirty-three compounds (see Table S2 in the supplemental material) derived from a virtual screening were assayed for their
effects on KSHV lytic DNA replication. BCBL-1 cells were induced with TPA and treated with each of the compounds at a concentration of 20 M. Forty-eight
hours postinduction, the intracellular KSHV genomic copy numbers were determined using quantitative real-time PCR. Seven compounds (marked with
asterisks) exhibited considerable inhibitory effects on viral lytic DNA replication. (B) Effects of the compounds on KSHV virion production. Five days
postinduction, the KSHV virions in supernatants were collected and measured by quantifying encapsidated viral DNA in the preparations. (C) Effects of the
compounds on host cell viability. Uninduced BCBL-1 cells were treated with each of the compounds at 20 M for 48 h, and cell viability was assessed by trypan
blue staining as described in Materials and Methods. The compounds are shown with ascending molecular weights. The error bars indicate standard deviations.
Xu et al.
566 aac.asm.org Antimicrobial Agents and Chemotherapy
supports the idea that ()-rutamarin inhibits KSHV ori-Lyt-de-
pendent DNA replication.
To rule out the possibility that ()-rutamarin blocks KSHV
DNA replication by inhibiting RTA expression, the effect of ()-
rutamarin on RTA expression in TPA-induced BCBL-1 cells was
examined by Western analysis. The result showed no adverse ef-
fect of the compound on RTA expression up to 25 M (22-fold
excess over the IC
50
) (see Fig. S2 in the supplemental material).
The reduced levels of RTA in the treatments with 50 and 100 M
()-rutamarin may result from the cytotoxicity of the compound
in a concentration near its IC
50
(94.24 M).
()-Rutamarin is an inhibitor of human topoisomerase II.
The next question was whether ()-rutamarin is a Topo II inhib-
itor that blocks KSHV ori-Lyt-dependent DNA replication by in-
hibiting the catalytic activity of host cell Topo II. Furthermore,
there are two Topo II isoforms in mammals, Topo IIand Topo
II. We wanted to know which isoform ()-rutamarin affects if it
is a Topo II inhibitor. To accomplish this, a DNA decatenation-
based topoisomerase II assay was established. In this system,
kinetoplast DNA consisting of a large network of interlocked
DNA minicircles can be decatenated into separate minicircles in
the presence of ATP and Topo II in either isoform. ()-Rutama-
rin in a wide range of concentrations was added to the reaction
mixture, and the effect of the compound on Topo II activity was
measured by determining decatenated kDNA levels using agarose
gel electrophoresis. The results showed that ()-rutamarin is able
to inhibit the enzymatic activity of Topo IIin a dose-dependent
manner (Fig. 6A and B). The IC
50
of ()-rutamarin in Topo II
inhibition was calculated as 28.22 6.2 M(Fig. 6C and D). At a
concentration of 100 M, the inhibition rate of ()-rutamarin is
significantly higher than that of novobiocin and close to that of the
Topo II poison etoposide (Fig. 6A). In contrast, ()-rutamarin
did not exhibit any inhibitory effect on Topo IIactivity (Fig. 6E).
To confirm if ()-rutamarin is a catalytic inhibitor of Topo
IIand acts in blocking the ATPase activity of the enzyme, as
novobiocin does, we studied the inhibition effect of rutamarin on
ATP hydrolysis of Topo IIusing the malachite green assay. As
shown in Fig. 7, the ATPase activity of Topo IIwas inhibited by
()-rutamarin in a dose-dependent manner, suggesting that, like
novobiocin, ()-rutamarin inhibits human Topo IIby binding
to the ATP pocket of the enzyme and blocking its ATP hydrolysis.
A binding model of ()-rutamarin in the ATP binding
pocket of Topo II.The mode of ()-rutamarin binding in the
ATPase domain of Topo IIwas investigated by docking and fur-
ther defined by MD simulations. Figure 8A illustrates the time
dependence of the RMSD values for the X-ray reference enzyme
structure of backbone atoms (C, C
151
, N, and O) over the produc-
tion phase of simulation. The RMSD values of simulation con-
verged after 3 ns, indicating that the system is stable and equil-
ibrated. The RMSD value of ligand compared docking pose is
swinging within 1.8 Å, which illustrates that the docking pose is
reliable. Previous studies indicated that Mg
2
is necessary for the
ATPase domain to hydrolyze the ATP molecule and is stabilized
by two conserved water molecules and residues Asn91 and Glu87
(24,27). The time evolution of oxygen- and nitrogen-Mg
2
dis-
tances for these residues and the water molecules is shown in Fig.
8C. The oxygen- and nitrogen-Mg
2
distances for these residues
and the water molecules stabilized at 1.9 to 2.0 Å during the sim-
ulation, suggesting that Mg
2
is stabilized by these factors. Our
simulation results are consistent with previous experimental re-
sults, suggesting that our model is reliable and reasonable.
The details of binding of ()-rutamarin to the ATPase domain
of Topo IIat an atomic level were revealed in a conformation
clustering analysis. The results suggest that ()-rutamarin forms
a hydrogen bond with the side chain of Asn95, Ala167, and con-
served water molecule 2 and hydrophobically interacts with
Ile125, Ile141, Phe142, Ala167, Thr215, Gly166, and Lys123 in the
catalytic site of Topo II(Fig. 8D). The distance distribution anal-
ysis also showed that the hydrogen bonds between conserved wa-
ter molecule 2- and Ala167–()-rutamarin are comparatively sta-
ble, whereas the hydrogen bond of Asn95–()-rutamarin is
unstable (Fig. 8B). Solvent-accessible-surface area (SASA) analy-
sis suggests that the polar surface of ()-rutamarin is comple-
FIG 2 Effect of ()-rutamarin on KSHV replication and its associated cyto-
toxicity. (A) Chemical structure of ()-rutamarin. (B) Effect of ()-rutama-
rin on KSHV lytic replication and its associated cytotoxicity in BCBL-1 cells
that were treated with a wide range of concentrations of ()-rutamarin 3 h
after lytic replication was induced by TPA. Intracellular KSHV genomic DNA
replication (blue), extracellular virion production (green), and cell viability
(orange) were determined as described in Materials and Methods. The values
were compared to those from the control cells (nondrug treatment). The mean
values of results from three independent experiments and standard deviations
are presented on the yaxis of dose-response curves. (C) Effect of ()-rutama-
rin on KSHV lytic replication and its associated cytotoxicity in JSC-1 cells that
were induced with 3 mM sodium butyrate. Intracellular KSHV genomic DNA
replication (blue), extracellular virion production (green), and cell viability
(orange) were determined as described for panel B.
()-Rutamarin Inhibits KSHV Replication
January 2014 Volume 58 Number 1 aac.asm.org 567
mentary to the polar surface of the binding pocket of Topo II
(Fig. 8E). Superimposition of ()-rutamarin and cocrystallized
ADPNP suggests that ()-rutamarin occupies the ATP binding
pocket in a fashion similar to that of an ATP molecule (Fig. 8F).
The acetyl groups of ()-rutamarin, like the three phosphate
groups of ATP, is involved in interaction with the highly con-
served Walker A consensus motif GXXGXG at residues 161 to 166
of Topo II(Fig. 8D and F)(36). This conserved motif is impor-
tant for the binding of ATP, as well as for the catalytic inhibitors
acting on the ATPase domain (27). This notion is supported by a
site-directed mutagenesis result showing that the mutations in
Arg162Gln and Tyr165Ser in the ATP binding site lead to drug
resistance (37,38). These observations, consistent with the results
of the DNA decatenation assay and the malachite green assay,
support the idea that ()-rutamarin functions as a Topo IIcat-
alytic inhibitor.
The MM-PBSA and MM-GBSA methods were employed to
calculate the binding free energies and to gain information on
the different components of interaction energy that contribute
to ()-rutamarin binding. The results are listed in Table 1.
G
MM-PBSA
and G
MM-GBSA
for the Topo II-()-rutamarin
complex are 29.90 and 24.66 kcal/mol, respectively. G
MM-
PBSA
, accounting for the total relative binding free energy, equal to
G
gas
plus G
solv
, comes mainly from the van der Waals compo-
nent, whereas the electrostatic contribution is much less. This no-
tion is also reflected in the results of the MM-GBSA method
(Table 1), which show that ()-rutamarin is a comparatively hy-
drophobic molecule that can form favorable hydrophobic inter-
actions with residues of the Topo IIcatalytic pocket.
DISCUSSION
In the current study, we identified ()-rutamarin as an effective
antiviral agent that inhibits KSHV ori-Lyt-dependent DNA repli-
cation with low cytotoxicity. It blocks KSHV DNA replication by
FIG 3 Effects of ()-rutamarin and novobiocin on BCBL-1 cell proliferation. BCBL-1 cells (starting with 2 10
5
cells/ml) were exposed to ()-rutamarin and
novobiocin at their IC
50
s and 5 times their IC
50
s. Data were obtained from three independent determinations and are presented as means and standard
deviations.
FIG 4 Effects of ()-rutamarin and novobiocin on BCBL-1 cell cycle progression. BCBL-1 cells (starting with 2 10
5
cells/ml) were exposed to ()-rutamarin
and novobiocin at their IC
50
s and 5 times their IC
50
s for 48 h. Cell cycle progressions were measured by PI staining, followed by flow cytometric analysis. The data
are presented as means obtained from three independent experiments.
Xu et al.
568 aac.asm.org Antimicrobial Agents and Chemotherapy
inhibiting the catalytic activity of human Topo II. The salient
features and implications of this finding are as follows.
There is a need for effective drugs targeting KSHV. KSHV has
been proven to be the etiological cause of KS and other KSHV-
associated malignancies. However, the current treatment modal-
ities for KSHV-associated diseases include only traditional cancer
therapies. For classic KS, the chemotherapeutics that have been
approved by the FDA include liposomal anthracycline products
(liposomal doxorubicin or liposomal daunorubicin), paclitaxel,
and alpha interferon (39). However, the majority of these agents
have serious side effects, and the tumor responses to the chemo-
therapeutic regimens are only transient. In the current AIDS epi-
demic, even though KS has become a major AIDS-associated ma-
lignancy and remains an important cause of morbidity and
mortality in AIDS patients, little effort has been made to develop
more effective drugs for KS, as there was a general belief that
AIDS-associated KS would disappear if AIDS were under control
FIG 5 Inhibition of KSHV ori-Lyt-dependent DNA replication with ()-
rutamarin. BCBL-1 cells were transfected with a KSHV ori-Lyt-containing
plasmid (pOri-A) and an RTA expression vector (pCR3.1-ORF50). The trans-
fected cells were treated with increasing concentrations of ()-rutamarin and
incubated for 72 h. Hirt DNAs were extracted and digested with KpnI/SacI or
KpnI/SacI/DpnI. DpnI-resistant viral replicated DNA (Rep’d DNA) was de-
tected by Southern blotting with
32
P-labeled pBluescript plasmid.
FIG 6 Identification of ()-rutamarin as a catalytic inhibitor of Topo II. (A) A Topo II-mediated kinetoplast DNA decatenation assay was performed to
evaluate the effects of testing compounds for Topo IIactivity at a concentration of 100 M. The catenated (cat) and decatenated (decat) DNA positions in gels
are indicated. (A and B) The Topo IIactivities in the reactions were quantitated (A), and inhibition rates of compounds were calculated (B). (C and E) Inhibition
of Topo II(C) and Topo II(E) activities by ()-rutamarin in a wide range of concentrations. (D) The IC
50
of ()-rutamarin on Topo IIinhibition was
calculated as 28.22 6.2 M. The data are means and standard deviations for three experimental replicates. (E) ()-Rutamarin exhibits no obvious inhibitory
effect on human Topo II. Rut, ()-rutamarin; Eto, etoposide; Nov, novobiocin.
FIG 7 Effect of ()-rutamarin on ATPase activity of Topo II. The inhibitory
effect of ()-rutamarin on Topo IIATPase activity was measured using the
malachite green assay as described in Materials and Methods. The OD
620
rep-
resents the ATP hydrolysis level and reflects the inhibition of ATPase activity
by ()-rutamarin. The error bars indicate standard deviations.
()-Rutamarin Inhibits KSHV Replication
January 2014 Volume 58 Number 1 aac.asm.org 569
FIG 8 Stability properties of the simulation system and binding model of ()-rutamarin with Topo II. (A) RMSD plot for the backbone atoms and
()-rutamarin during MD simulations after equilibration. (B) Distance distributions between Asn95, Ala167, and conserved water molecule 2 and ()-
rutamarin in water at 310 K. The cutoff value for the formation of a hydrogen bond is 3.5 Å. (C) Time dependence of the distance between the carboxyl oxygen
of Glu87, the amide nitrogen of Asn91, the two conserved water molecules, and Mg
2
during the 10-ns MD simulations. (D) Detailed binding model between
()-rutamarin and the residues in the ATPase domain of Topo II. Hydrogen bonds are shown by red dashed lines, and magenta spheres represent Mg cations.
(E) Polar and hydrophobic surface profile of the ATPase domain of Topo IIwith ()-rutamarin. Red and gray represent polar and hydrophobic areas,
respectively. (F) Superposition of ()-rutamarin with ADPNP in the ligand binding pocket.
Xu et al.
570 aac.asm.org Antimicrobial Agents and Chemotherapy
with the advent of HAART. However, despite its dramatic de-
crease in frequency since the advent of HAART, KS remains the
most common AIDS-associated cancer (40). In addition, as expe-
rience with HAART has grown, a new HAART-associated syn-
drome has emerged. In a subset of HIV-seropositive individuals,
starting HAART in the setting of advanced HIV infection results
in a paradoxical clinical worsening of existing infection or the
appearance of a new condition, including KS, in a process known
as immune reconstitution inflammatory syndrome (IRIS) (41).
IRIS is thought to be the direct result of a reconstituted im-
mune system recognizing pathogens or antigens that were pre-
viously present but clinically asymptomatic. The current regi-
men for IRIS-associated KS is a combination therapy with
HAART plus chemotherapeutics, such as liposomal doxorubicin.
Since IRIS-KS is the result of responses by a recovered immune
system to the KS-causing pathogen, i.e., KSHV, treatment of
KSHV-seropositive, HIV-positive patients with a combination of
antiretroviral (HAART) and anti-KSHV therapeutics is expected
to yield positive results. However, at this time, there are no effec-
tive drugs targeting KSHV available. Earlier studies on cohorts of
HIV-seropositive subjects suggested that treatment with the anti-
herpesviral drug ganciclovir or foscarnet could reduce the inci-
dence of KS in AIDS patients (42,43). However, severe side effects
(renal impairment and bone marrow suppression), as well as
rapid development of drug resistance, have limited the use of these
drugs in the treatment of KS. Therefore, there is a need for effec-
tive drugs targeting KSHV.
()-Rutamarin is an antiviral-drug candidate for KSHV-as-
sociated diseases. Recently, we reported that host cellular Topo II
is absolutely required for KSHV DNA replication, and thus, Topo
II inhibitors effectively block KSHV DNA synthesis (20). Topo II
inhibitors are divided into two categories: Topo II poisons, which
target the topoisomerase-DNA intermediate (cleavable complex),
and Topo II catalytic inhibitors, which disrupt catalytic turnover
of the enzyme. Although both categories possess inhibitory activ-
ities against KSHV DNA replication, Topo II poisons in general
exhibit very strong cytotoxicity to host cells. In contrast, Topo II
catalytic inhibitors show less cytotoxicity (such as novobiocin,
with a CC
50
value of 871 M, and merbarone, with a CC
50
value of
212.9 M for BCBL-1 cells). Both novobiocin and merbarone are
effective in halting KSHV DNA synthesis, with IC
50
s of 27.55 M
and 19.54 M, respectively. The low cytotoxicities and high inhi-
bition rates for viral replication of catalytic Topo II inhibitors
suggest the potential for Topo II catalytic inhibitors to become
effective anti-KSHV drugs for treatment of KS and other KSHV-
associated diseases (20). In addition, given that viral polymerases
may introduce mutations which subsequently can give rise to drug
resistance, targeting host cellular proteins that viruses rely on for
their replication offers the advantage of minimizing drug resis-
tance and thus constitutes a novel therapeutic strategy.
In the current study, we attempted to search for new Topo II
catalytic inhibitors that are potent in inhibiting viral replication
and that have low cytotoxicity. This effort led to identification of
()-rutamarin as a promising lead that efficiently inhibits KSHV
lytic DNA replication in BCBL-1 cells with an IC
50
of 1.12 M and
an EC
50
of 1.62 M, 25- and 17-fold lower than those of novobio-
cin. In addition, ()-rutamarin exhibits very low cytotoxicity,
and the SI of KSHV lytic replication was calculated to be 84.14,
three times better that that of novobiocin. In addition, the molec-
ular weight of ()-rutamarin (356.42) is smaller than that of
novobiocin (634.61), and the mass concentration differences of
their IC
50
s and EC
50
s would be greater. These data demonstrate
great potential for ()-rutamarin to become an effective antiviral
agent with low cytotoxicity.
()-Rutamarin is a natural product found in plants, such as
Ruta graveolens L (common rue). It has been reported that ()-
rutamarin possesses antiproliferation and anticancer activities
against a variety of tumor cell lines (44). It has also been found to
induce the expression and translocation of glucose transporter 4
(GLUT4). As impaired translocation or decreased expression
of GLUT4 in response to insulin is one of the major patholog-
ical features of type 2 diabetes, the function of ()-rutamarin in
ameliorating glucose homeostasis suggests a potential for the
compound in anti-type 2 diabetes drug development (45). Our
study is the first to demonstrate the capability of the compound in
inhibition of viral replication with the potential to be an antiviral.
Anti-proliferation potential of ()-rutamarin. At a dose
equal to the IC
50
for inhibiting KSHV replication, ()-rutamarin
did not show a perceptible effect on cell proliferation and cell cycle
progression in BCLB-1 and BJAB cells. At an excess dose (5 times
the IC
50
,), ()-rutamarin exhibited a degree of suppression of cell
proliferation with a delay in the S phase.
The effects of ()-rutamarin on cell proliferation in a variety
of tumor cell lines were estimated, and inhibitory activities against
A549, Bel7402, HepG2, and HCT8 cell proliferation were reported
(44). The antiproliferation activity of ()-rutamarin against these
tumor cell lines appears to be similar to the effects on B cells
observed in our study. Although ()-rutamarin has an extent of
antiproliferative ability that may provide an additional benefit if it
becomes a drug to treat KSHV-associated malignancies, the mod-
erate antiproliferation ability may not be potent enough to pro-
vide antitumor activity. This may also be true of all Topo II cata-
lytic inhibitors. This notion is supported by studies and
observations of some Topo II catalytic inhibitors, including novo-
biocin and merbarone, which exhibit antiproliferation activities
in cell cultures but show no significant effects on tumor develop-
ment in clinical trials (46,47).
The moderate effect of ()-rutamarin on cell proliferation, as
well as its low cytotoxicity, may result from redundancy of Topo II
activities in mammalian cells. Mammals express two isoforms of
TABLE 1 Binding free energy for Topo II–()-rutamarin complex
and its different energy components based on MM-PBSA and MM-
GBSA methods
Energy term
Binding free energy (kcal/mol) (SEM)
MM-PBSA MM-GBSA
E
vdwa
40.12 (2.14) 40.22 (2.26)
E
eleb
0.35 (0.22) 0.42 (0.15)
E
pol,solvc
39.99 (1.93) 22.09 (1.66)
E
nonpol,solvd
29.43 (1.37) 6.11 (0.34)
G
gase
40.47 (2.41) 40.64 (2.28)
G
solvf
10.56 (1.28) 15.98 (1.14)
G
bindingg
29.91 (1.38) 24.66 (1.69)
a
Nonbonded van der Waals.
b
Nonbonded electrostatics.
c
Polar component to solvation.
d
Nonpolar component to solvation.
e
Total gas phase energy.
f
Sum of nonpolar and polar contributions to solvation.
g
Final estimated binding free energy calculated from the terms above.
()-Rutamarin Inhibits KSHV Replication
January 2014 Volume 58 Number 1 aac.asm.org 571
Topo II, alpha and beta, which are highly homologous but display
differences in expression and subcellular localization at the time of
mitosis. The similar functions and domain structures of the two
isoforms make it possible that Topo IImay be able to partially
compensate for the loss of Topo IIfunction for cell survival in
the presence of an inhibitor, such as ()-rutamarin. However,
KSHV may be dependent on only Topo IIfor its lytic replication,
explaining the sensitivity of KSHV replication to Topo IIcata-
lytic inhibitors.
()-Rutamarin is a novel human Topo IIcatalytic inhibi-
tor. Topo II is an ATPase and uses the energy derived from ATP
hydrolysis to resolve the winding problem of double-stranded
DNA. The detailed binding mode of ()-rutamarin and human
Topo IIwas identified using molecular docking and MD simu-
lation. The significance of these studies is 2-fold. First, the study
provided insight into the binding of ()-rutamarin to Topo II,
and the results confirmed that the compound binds to the ATP
pocket of Topo II. Second, the results of the study will be of great
help in our next effort to modify the compound for more potent
antiviral candidates. Energy decomposition analysis led to identi-
fication of key residues contributing to binding affinity at the ac-
tive site. As shown in Fig. 9, the major contributing residues can be
divided into three clusters, which are consistent with the three
binding pocket sites, i.e., (i) the Walker A consensus motif (resi-
dues 161 to 167), (ii) the Mg
2
binding area (residues 87, 91, and
94), and (iii) the hydrophobic chamber (key residues, Ile 125,
Ile141, Phe142, Lys123, and Thr215). All of these residues except
Glu87 positively contribute to the binding affinity for ()-
rutamarin. The incompatibility between the carboxyl group of
Glu87 and the acetyl groups of ()-rutamarin suggests that Glu87
is unfavorable for binding affinity to ()-rutamarin (Fig. 8D).
These results provide a theoretical basis for ()-rutamarin lead
optimization.
ACKNOWLEDGMENTS
We thank Manunya Nuth at the University of Pennsylvania for construc-
tive discussion, suggestions, and critical reading of the manuscript.
This work was supported by Natural Science Foundation of China
research grants (no. 81271805 and 81171575), the Guangdong Innovative
Research Team program (no. 2009010058), and a research grant from the
U.S. National Institutes of Health (R01AI052789).
REFERENCES
1. Chang Y, Cesarman E, Pessin MS, Lee F, Culpepper J, Knowles DM,
Moore PS. 1994. Identification of herpesvirus-like DNA sequences in
AIDS-associated Kaposi’s sarcoma. Science 266:1865–1869. http://dx.doi
.org/10.1126/science.7997879.
2. Moore PS, Chang Y. 1995. Detection of herpesvirus-like DNA sequences in
Kaposi’s sarcoma in patients with and without HIV infection. N. Engl. J. Med.
332:1181–1185. http://dx.doi.org/10.1056/NEJM199505043321801.
3. Dupin N, Grandadam M, Calvez V, Aubin JT, Agut H, Gorin I, Havard
S, Lamy F, Leibowitch M, Huraux JM, Escande JP. 1995. Herpesvirus-
like DNA sequences in patients with Mediterranean Kaposi’s sarcoma.
Lancet 345:761–762. http://dx.doi.org/10.1016/S0140-6736(95)90642-8.
4. Schalling M, Ekman M, Kaaya EE, Linde A, Biberfeld P. 1995. A role for
a new herpes virus (KSHV) in different forms of Kaposi’s sarcoma. Nat.
Med. 1:707–708. http://dx.doi.org/10.1038/nm0795-707.
5. Chuck S, Grant RM, Katongole-Mbidde E, Conant M, Ganem D. 1996.
Frequent presence of a novel herpesvirus genome in lesions of human
immunodeficiency virus-negative Kaposi’s sarcoma. J. Infect. Dis. 173:
248–251. http://dx.doi.org/10.1093/infdis/173.1.248.
6. Antman K, Chang Y. 2000. Kaposi’s sarcoma. N. Engl. J. Med. 342:1027–
1038. http://dx.doi.org/10.1056/NEJM200004063421407.
7. Krentz HB, Kliewer G, Gill MJ. 2005. Changing mortality rates and
causes of death for HIV-infected individuals living in Southern Alberta,
Canada from 1984 to 2003. HIV Med. 6:99–106. http://dx.doi.org/10
.1111/j.1468-1293.2005.00271.x.
8. Cesarman E, Knowles DM. 1999. The role of Kaposi’s sarcoma-
associated herpesvirus (KSHV/HHV-8) in lymphoproliferative diseases.
Semin. Cancer Biol. 9:165–174. http://dx.doi.org/10.1006/scbi.1998.0118.
9. Powles T, Stebbing J, Bazeos A, Hatzimichael E, Mandalia S, Nelson M,
Gazzard B, Bower M. 2009. The role of immune suppression and HHV-8 in
the increasing incidence of HIV-associated multicentric Castleman’s disease.
Ann. Oncol. 20:775–779. http://dx.doi.org/10.1093/annonc/mdn697.
10. Moore PS, Chang Y. 2011. KSHV: forgotten but not gone. Blood 117:
6973–6974. http://dx.doi.org/10.1182/blood-2011-05-350306.
11. Staskus KA, Zhong W, Gebhard K, Herndier B, Wang H, Renne R,
Beneke J, Pudney J, Anderson DJ, Ganem D, Haase AT. 1997. Kaposi’s
sarcoma-associated herpesvirus gene expression in endothelial (spindle)
tumor cells. J. Virol. 71:715–719.
12. Sun R, Lin SF, Staskus K, Gradoville L, Grogan E, Haase A, Miller G.
1999. Kinetics of Kaposi’s sarcoma-associated herpesvirus gene expres-
sion. J. Virol. 73:2232–2242.
13. Zhong W, Wang H, Herndier B, Ganem D. 1996. Restricted expression
of Kaposi sarcoma-associated herpesvirus (human herpesvirus 8) genes in
Kaposi sarcoma. Proc. Natl. Acad. Sci. U. S. A. 93:6641–6646. http://dx
.doi.org/10.1073/pnas.93.13.6641.
14. Cesarman E, Mesri EA, Gershengorn MC. 2000. Viral G protein-coupled
receptor and Kaposi’s sarcoma: a model of paracrine neoplasia? J. Exp.
Med. 191:417–422. http://dx.doi.org/10.1084/jem.191.3.417.
15. Grundhoff A, Ganem D. 2004. Inefficient establishment of KSHV latency
suggests an additional role for continued lytic replication in Kaposi sar-
coma pathogenesis. J. Clin. Invest. 113:124–136. http://dx.doi.org/10
.1172/JCI17803.
16. Lin CL, Li H, Wang Y, Zhu FX, Kudchodkar S, Yuan Y. 2003. Kaposi’s
sarcoma-associated herpesvirus lytic origin (ori-Lyt)-dependent DNA
replication: identification of the ori-Lyt and association of K8 bZip pro-
tein with the origin. J. Virol. 77:5578 –5588. http://dx.doi.org/10.1128/JVI
.77.10.5578-5588.2003.
17. Wang Y, Li H, Chan MY, Zhu FX, Lukac DM, Yuan Y. 2004. Kaposi’s
sarcoma-associated herpesvirus ori-Lyt-dependent DNA replication: cis-
acting requirements for replication and ori-Lyt-associated RNA transcrip-
tion. J. Virol. 78:8615–8629. http://dx.doi.org/10.1128/JVI.78.16.8615
-8629.2004.
18. Wang Y, Tang Q, Maul GG, Yuan Y. 2006. Kaposi’s sarcoma-associated
herpesvirus ori-Lyt-dependent DNA replication: dual role of replication
and transcription activator. J. Virol. 80:12171–12186. http://dx.doi.org/10
.1128/JVI.00990-06.
19. Wang Y, Li H, Tang Q, Maul GG, Yuan Y. 2008. Kaposi’s sarcoma-
associated herpesvirus ori-Lyt-dependent DNA replication: involvement
of host cellular factors. J. Virol. 82:2867–2882. http://dx.doi.org/10.1128
/JVI.01319-07.
20. Gonzalez-Molleda L, Wang Y, Yuan Y. 2012. Potent antiviral activity of
topoisomerase I and II inhibitors against Kaposi’s sarcoma-associated
FIG 9 Binding free energy decomposition results based on the MM-GBSA
method. The key residues are labeled.
Xu et al.
572 aac.asm.org Antimicrobial Agents and Chemotherapy
herpesvirus. Antimicrob. Agents Chemother. 56:893–902. http://dx.doi
.org/10.1128/AAC.05274-11.
21. Renne R, Zhong W, Herndier B, McGrath M, Abbey N, Kedes D,
Ganem D. 1996. Lytic growth of Kaposi’s sarcoma-associated herpesvirus
(human herpesvirus 8) in culture. Nat. Med. 2:342–346. http://dx.doi.org
/10.1038/nm0396-342.
22. Cannon JS, Ciufo D, Hawkins AL, Griffin CA, Borowitz MJ, Hayward
GS, Ambinder RF. 2000. A new primary effusion lymphoma-derived cell
line yields a highly infectious Kaposi’s sarcoma herpesvirus-containing
supernatant. J. Virol. 74:10187–10193. http://dx.doi.org/10.1128/JVI.74
.21.10187-10193.2000.
23. Gu Q, Xu J, Gu L. 2010. Selecting diversified compounds to build a
tangible library for biological and biochemical assays. Molecules 15:5031–
5044. http://dx.doi.org/10.3390/molecules15075031.
24. Wei H, Ruthenburg AJ, Bechis SK, Verdine GL. 2005. Nucleotide-
dependent domain movement in the ATPase domain of a human type IIA
DNA topoisomerase. J. Biol. Chem. 280:37041–37047. http://dx.doi.org
/10.1074/jbc.M506520200.
25. Spassov VZ, Flook PK, Yan L. 2008. LOOPER: a molecular mechanics-
based algorithm for protein loop prediction. Protein Eng. Des. Sel. 21:91–
100. http://dx.doi.org/10.1093/protein/gzm083.
26. Cheng A, Best SA, Merz KM, Jr, Reynolds CH. 2000. GB/SA water model
for the Merck molecular force field (MMFF). J. Mol. Graph. Model. 18:
273–282. http://dx.doi.org/10.1016/S1093-3263(00)00038-3.
27. Baviskar AT, Madaan C, Preet R, Mohapatra P, Jain V, Agarwal A,
Guchhait SK, Kundu CN, Banerjee UC, Bharatam PV. 2011. N-fused
imidazoles as novel anticancer agents that inhibit catalytic activity of
topoisomerase II alpha and induce apoptosis in G
1
/S phase. J. Med. Chem.
54:5013–5030. http://dx.doi.org/10.1021/jm200235u.
28. Amaro RE, Swift RV, Votapka L, Li WW, Walker RC, Bush RM. 2011.
Mechanism of 150-cavity formation in influenza neuraminidase. Nat.
Commun. 2:388. http://dx.doi.org/10.1038/ncomms1390.
29. Hornak V, Abel R, Okur A, Strockbine B, Roitberg A, Simmerling C.
2006. Comparison of multiple amber force fields and development of
improved protein backbone parameters. Proteins 65:712–725. http://dx
.doi.org/10.1002/prot.21123.
30. Newhouse EI, Xu D, Markwick PRL, Amaro RE, Pao HC, Wu KJ, Alam
M, McCammon JA, Li WW. 2009. Mechanism of glycan receptor recog-
nition and specificity switch for avian, swine, and human adapted influ-
enza virus hemagglutinins: a molecular dynamics perspective. J. Am.
Chem. Soc. 131:17430–17442. http://dx.doi.org/10.1021/ja904052q.
31. Zhang W, Hou TJ, Qiao XB, Xu XJ. 2003. Parameters for the generalized
born model consistent with RESP atomic partial charge assignment protocol.
J. Phys. Chem. B 107:9071–9078. http://dx.doi.org/10.1021/jp034613k.
32. Frisch MJ, Trucks GW, Schlegel HB, Scuseria GE, Robb MA, Cheese-
man JR, Montgomery JA, Jr, Vreven T, Kudin KN, Burant JC, Millam
JM, Iyengar SS, Tomasi J, Barone V, Mennucci B, Cossi M, Scalmani G,
Rega N, Petersson GA, Nakatsuji H, Hada M, Ehara M, Toyota K,
Fukuda R, Hasegawa J, Ishida M, Nakajima T, Honda Y, Kitao O, Nakai
H, Klene M, Li X, Knox JE, Hratchian HP, Cross JB, Adamo C,
Jaramillo J, Gomperts R, Stratmann RE, Yazyev O, Austin AJ, Cammi
R, Pomelli C, Ochterski JW, Ayala PY, Morokuma K, Voth GA, Salva-
dor P, Dannenberg JJ, Zakrzewski VG, Dapprich S, Daniels AD, Strain
MC, Farkas O, Malick DK, Rabuck AD, Raghavachari K, Foresman JB,
Ortiz JV, Cui Q, Baboul AG, Clifford S, Cioslowski J, Stefanov BB, Liu
G, Liashenko A, Piskorz P, Komaromi I, Martin RL, Fox DJ, Keith T,
Al-Laham MA, Peng CY, Nanayakkara A, Challacombe M, Gill PMW,
Johnson B, Chen W, Wong MW, Gonzalez C, Pople JA. 2004. Gaussian
03, revision E. 01. Gaussian Inc., Pittsburgh, PA.
33. Kollman PA, Massova I, Reyes C, Kuhn B, Huo SH, Chong L, Lee M,
Lee T, Duan Y, Wang W, Donini O, Cieplak P, Srinivasan J, Case DA,
Cheatham TE. 2000. Calculating structures and free energies of complex
molecules: Combining molecular mechanics and continuum models. Acc.
Chem. Res. 33:889897. http://dx.doi.org/10.1021/ar000033j.
34. Yan X, Li J, Liu Z, Zheng M, Ge H, Xu J. 2013. Enhancing molecular
shape comparison by weighted Gaussian functions. J. Chem. Inf. Model.
53:1967–1978. http://dx.doi.org/10.1021/ci300601q.
35. Sun R, Lin SF, Gradoville L, Yuan Y, Zhu F, Miller G. 1998. A viral gene
that activates lytic cycle expression of Kaposi’s sarcoma-associated herpes-
virus. Proc. Natl. Acad. Sci. U. S. A. 95:10866 –10871. http://dx.doi.org/10
.1073/pnas.95.18.10866.
36. Walker JE, Saraste M, Runswick MJ, Gay NJ. 1982. Distantly related
sequences in the alpha- and beta-subunits of ATP synthase, myosin, ki-
nases and other ATP-requiring enzymes and a common nucleotide bind-
ing fold. EMBO J. 1:945–951.
37. Wessel I, Jensen LH, Jensen PB, Falck J, Rose A, Roerth M, Nitiss JL,
Sehested M. 1999. Human small cell lung cancer NYH cells selected for
resistance to the bisdioxopiperazine topoisomerase II catalytic inhibitor
ICRF-187 demonstrate a functional R162Q mutation in the Walker A
consensus ATP binding domain of the alpha isoform. Cancer Res. 59:
3442–3450.
38. Wessel I, Jensen LH, Renodon-Corniere A, Sorensen TK, Nitiss JL,
Jensen PB, Sehested M. 2002. Human small cell lung cancer NYH cells
resistant to the bisdioxopiperazine ICRF-187 exhibit a functional domi-
nant Tyr165Ser mutation in the Walker A ATP binding site of topoisom-
erase II alpha. FEBS Lett. 520:161–166. http://dx.doi.org/10.1016/S0014
-5793(02)02805-3.
39. Potthoff A, Brockmeyer NH. 2007. HIV-associated Kaposi sarcoma:
pathogenesis and therapy. J. Dtsch. Dermatol. Ges. 5:1091–1094. http://dx
.doi.org/10.1111/j.1610-0387.2007.06567.x.
40. Boshoff C, Weiss R. 2002. AIDS-related malignancies. Nat. Rev. Cancer.
2:373–382. http://dx.doi.org/10.1038/nrc797.
41. Shelburne SA, III, Hamill RJ. 2003. The immune reconstitution inflam-
matory syndrome. AIDS Rev. 5:67–79.
42. Glesby MJ, Hoover DR, Weng S, Graham NM, Phair JP, Detels R, Ho
M, Saah AJ. 1996. Use of antiherpes drugs and the risk of Kaposi’s sar-
coma: data from the Multicenter AIDS Cohort Study. J. Infect. Dis. 173:
1477–1480. http://dx.doi.org/10.1093/infdis/173.6.1477.
43. Mocroft A, Youle M, Gazzard B, Morcinek J, Halai R, Phillips AN.
1996. Anti-herpesvirus treatment and risk of Kaposi’s sarcoma in HIV
infection. Royal Free/Chelsea and Westminster Hospitals Collaborative
Group. AIDS 10:1101–1105.
44. Yang QY, Tian XY, Fang WS. 2007. Bioactive coumarins from Boen-
ninghausenia sessilicarpa. J. Asian Nat. Prod. Res. 9:5965. http://dx.doi
.org/10.1080/10286020500382397.
45. Zhang Y, Zhang H, Yao XG, Shen H, Chen J, Li C, Chen L, Zheng M,
Ye J, Hu L, Shen X, Jiang H. 2012. ()-Rutamarin as a dual inducer of
both GLUT4 translocation and expression efficiently ameliorates glucose
homeostasis in insulin-resistant mice. PLoS One 7:e31811. http://dx.doi
.org/10.1371/journal.pone.0031811.
46. Eder JP, Wheeler CA, Teicher BA, Schnipper LE. 1991. A phase I clinical
trial of novobiocin, a modulator of alkylating agent cytotoxicity. Cancer
Res. 51:510–513.
47. Chang AY, Kim K, Glick J, Anderson T, Karp D, Johnson D. 1993.
Phase II study of taxol, merbarone, and piroxantrone in stage IV non-
small-cell lung cancer: The Eastern Cooperative Oncology Group Results.
J. Natl. Cancer Inst. 85:388–394. http://dx.doi.org/10.1093/jnci/85.5.388.
()-Rutamarin Inhibits KSHV Replication
January 2014 Volume 58 Number 1 aac.asm.org 573
... Coumarin scaffolds have proven to be important in the development of anticancer medications since various coumarin derivatives have shown anticancer action on a range of cell types [7][8][9]. Coumarins are becoming increasingly popular because of their broad range of pharmacological properties, including their anti-inflammatory [10], antibacterial [11], antiviral [12], antioxidant [13], antinociceptive [14], anti-cancer [15], anti-asthmatic [16], antidepressant [17], anti-HIV [18], anti-tuberculosis [19], anti-Alzheimer's [14], anti-influenza [20], and antihyperlipidemic abilities [21] (Fig. 1). Specifically interesting, 3-arylcoumarin is an important structural element of HIV-1 replication inhibitors [22], horseradish peroxidase (HRP) inhibitors [23], and selective monoamine oxidase B (MAO-B) inhibitors [24,25] (Fig. 1). ...
... The structural requirements for the compounds potency (structure activity relationship, SAR) studies, for scoparone (1) and all 3-substititued derivatives (2)(3)(4)(5)(6)(7)(8)(9)(10)(11)(12)(13)(14)(15)(16)(17)(18)(19)(20) were evaluated using in vitro and in vivo anti-inflammatory activity. the entire library of tested molecules, compounds 3 and 9 demonstrated the most potent effect by inhibiting more than 60% TNF-α. ...
... In vivo testing is generally preferred to in vitro testing as it allows for a more thorough analysis of an experiment's overall impact on a living subject. Therefore, we next investigated anti-inflammatory activity of all the compounds (1)(2)(3)(4)(5)(6)(7)(8)(9)(10)(11)(12)(13)(14)(15)(16)(17)(18)(19)(20) in vivo using carrageenan-induced paw edema model in ...
Article
Full-text available
Natural products (NPs) continue to serve as a structural model for the development of new bioactive molecules and improve the process of identifying novel medicines. The biological effects of coumarins, one of the most researched compounds among NPs, are currently being thoroughly investigated. In the present investigation, we reported the synthesis of nineteen semi-synthetic 3-substituted scoparone analogues, followed by their characterization using analytical methods such as NMR, HPLC, and HRMS. All compounds screened for in vitro and in vivo study for their ability to reduce inflammation. The SAR study worked effectively for this particular scoparone 3-substitution, as compounds 3, 4, 9, 16, 18, and 20 displayed improved in vitro results for TNF-α than the parent molecule. Similarly, compounds 3, and 17 showed a higher percentage of IL-6 inhibition. Compounds 3, 4, and 12 have also been identified by in vivo studies as promising candidates with higher percent inhibition than the parent scoparone molecule. As evident from all in vitro and in vivo studies, compound 3 showed the most potent anti-inflammatory activity among all.
... The compound is moderately cytotoxic to colorectal adenocarcinoma HT29 cells (IC50 = 5.6 μM), and induces cell cycle perturbations and caspasedependent apoptosis [73]. At the molecular level, rutamarin is believed to function as a catalytic inhibitor of human topoisomerase II (not a classical Topo II poison), binding to the ATPase domain of the enzyme, thereby blocking DNA replication [74]. This mechanism accounts for both the anticancer and antiviral activities of rutamarin, and probably for its antiprotozoal activity as well [75]. ...
... This mechanism accounts for both the anticancer and antiviral activities of rutamarin, and probably for its antiprotozoal activity as well [75]. Rutamarin inhibits DNA replication of Epstein-Barr virus (EBV) (IC50 = 7.0 μM) [76,77] and Kaposi's sarcoma-associated herpesvirus (KSHV) (IC50 = 1.12 μM), at least the (+)-enantiomer [74]. Beyond TopoII, two additional targets for rutamarin have been proposed on the basis of a molecular modeling analysis: the protein tyrosine phosphatase 1B (PTP1B) and the retinoid X receptor α (RXRα). ...
... The compound is moderately cytotoxic to colorectal adenocarcinoma HT29 cells (IC 50 = 5.6 µM), and induces cell cycle perturbations and caspase-dependent apoptosis [73]. At the molecular level, rutamarin is believed to function as a catalytic inhibitor of human topoisomerase II (not a classical Topo II poison), binding to the ATPase domain of the enzyme, thereby blocking DNA replication [74]. This mechanism accounts for both the anticancer and antiviral activities of rutamarin, and probably for its antiprotozoal activity as well [75]. ...
Article
Full-text available
The genus Ruta in the family Rutaceae includes about 40 species, such as the well-known plants R. graveolens L. (common rue) or R. chalepensis L. (fringed rue), but also much lesser-known species such as R. angustifolia Pers. (narrow-leaved fringed rue). This rue specie, originating from the Mediterranean region, is well-distributed in Southeast Asia, notably in the Indo-Chinese peninsula and other territories. In some countries, such as Malaysia, the plant is used to treat liver diseases and cancer. Extracts of R. angustifolia display antifungal, antiviral and antiparasitic effects. Diverse bioactive natural products have been isolated from the aerial parts of the plant, notably quinoline alkaloids and furocoumarins, which present noticeable anti-inflammatory, antioxidant and/or antiproliferative properties. The present review discusses the main pharmacological properties of the plant and its phytoconstituents, with a focus on the anticancer activities evidenced with diverse alkaloids and terpenoids isolated from the aerial parts of the plant. Quinoline alkaloids such as graveoline, kokusaginine, and arborinine have been characterized and their mode of action defined. Arborinine stands as a remarkable inhibitor of histone demethylase LSD1, endowed with promising anticancer activities. Other anticancer compounds, such as the furocoumarins chalepin and rutamarin, have revealed antitumor effects. Their mechanism of action is discussed together with that of other bioactive natural products, including angustifolin and moskachans. Altogether, R. angustifolia Pers. presents a rich phytochemical profile, fully consistent with the traditional use of the plant to treat cancer. This rue species, somewhat neglected, warrant further investigations as a medicinal plant and a source of inspiration for drug discovery and design.
... Later, this compound was also observed to hinder EBV lytic DNA replication with an IC 50 value of 7.0 µM by a mechanism that targets viral protein synthesis [119]. (+)-Rutamarin inhibitory activi-ties against KSHV lytic DNA replication and virion production have also been reported with an IC 50 value of 1.12 µM and an EC 50 value of 1.62 µM, respectively [120]. ...
Article
Full-text available
Herpesviruses are one of the most contagious DNA viruses that threaten human health, causing severe diseases, including, but not limited to, certain types of cancer and neurological complications. The overuse and misuse of anti-herpesvirus drugs are key factors leading to drug resistance. Therefore, targeting human herpesviruses with natural products is an attractive form of therapy, as it might improve treatment efficacy in therapy-resistant herpesviruses. Plant polyphenols are major players in the health arena as they possess diverse bioactivities. Hence, in this article, we comprehensively summarize the recent advances that have been attained in employing plant non-flavonoid polyphenols, such as phenolic acids, tannins and their derivatives, stilbenes and their derivatives, lignans, neolignans, xanthones, anthraquinones and their derivatives, curcuminoids, coumarins, furanocoumarins, and other polyphenols (phloroglucinol) as promising anti-herpesvirus drugs against various types of herpesvirus such as alpha-herpesviruses (herpes simplex virus type 1 and 2 and varicella-zoster virus), beta-herpesviruses (human cytomegalovirus), and gamma-herpesviruses (Epstein–Barr virus and Kaposi sarcoma-associated herpesvirus). The molecular mechanisms of non-flavonoid polyphenols against the reviewed herpesviruses are also documented.
... As earlier as two decades ago, substances with coumarin similar structures including saxalin, psoralen ( Figure 8C), and its methoxy derivative bergapten ( Figure 7A) were known to suppress HIV replication [89], and sometime later, such coumarins as mesoul and isomesoul were reported to prevent HIV replication in jurkat T cell [90]. A sesquiterpene coumarin kellerin; a natural furanocoumarin rutamarin, an aryl-coumarin glycycoumarin, and a simple coumarin osthole ( Figure 8D) were reported to be anti-HSV and anti-HCV agents [91,92]. Moreover, other studies have reported that some of the natural coumarins such as xanthotoxin ( Figure 7D), glycycoumarin, oxypeucedanin, pranferol, and heraclenol have anti-HIV activity [38,93]. ...
Article
Full-text available
Coumarins are a structurally varied set of 2H-chromen-2-one compounds categorized also as members of the benzopyrone group of secondary metabolites. Coumarin derivatives attract interest owing to their wide practical application and the unique reactivity of fused benzene and pyrone ring systems in molecular structure. Coumarins have their own specific fingerprints as antiviral, antimicrobial, antioxidant, anti-inflammatory, antiadipogenic, cytotoxic, apoptosis, antitumor, antitubercular, and cytotoxicity agents. Natural products have played an essential role in filling the pharmaceutical pipeline for thousands of years. Biological effects of natural coumarins have laid the basis of low-toxic and highly effective drugs. Presently, more than 1300 coumarins have been identified in plants, bacteria, and fungi. Fungi as cultivated microbes have provided many of the nature-inspired syntheses of chemically diverse drugs. Endophytic fungi bioactivities attract interest, with applications in fields as diverse as cancer and neuronal injury or degeneration, microbial and parasitic infections, and others. Fungal mycelia produce several classes of bioactive molecules, including a wide group of coumarins. Of promise are further studies of conditions and products of the natural and synthetic coumarins’ biotransformation by the fungal cultures, aimed at solving the urgent problem of searching for materials for biomedical engineering. The present review evaluates the fungal coumarins, their structure-related peculiarities, and their future therapeutic potential. Special emphasis has been placed on the coumarins successfully bioprospected from fungi, whereas an industry demand for the same coumarins earlier found in plants has faced hurdles. Considerable attention has also been paid to some aspects of the molecular mechanisms underlying the coumarins’ biological activity. The compounds are selected and grouped according to their cytotoxic, anticancer, antibacterial, antifungal, and miscellaneous effects.
... (+)-Rutamarin (30), extracted from Ruta graveolens, has been reported to inhibit EBV lytic DNA replication [175]. Later, 30 was also found to inhibit EBV lytic DNA replication with an IC 50 [130]. It also inhibited virion production with low toxicity, with EC 50 , CC 50 , and SI values of 1.62 µM, 94.24 µM, and 84.14, respectively. ...
Article
Full-text available
Herpesviruses establish long-term latent infection for the life of the host and are known to cause numerous diseases. The prevalence of viral infection is significantly increased and causes a worldwide challenge in terms of health issues due to drug resistance. Prolonged treatment with conventional antiviral drugs is more likely to develop drug-resistant strains due to mutations of thymidine nucleoside kinase or DNA polymerase. Hence, the development of alternative treatments is clearly required. Natural products and their derivatives have played a significant role in treating herpesvirus infection rather than nucleoside analogs in drug-resistant strains with minimal undesirable effects and different mechanisms of action. Numerous plants, animals, fungi, and bacteria-derived compounds have been proved to be efficient and safe for treating human herpesvirus infection. This review covers the natural antiherpetic agents with the chemical structural class of alkaloids, flavonoids, terpenoids, polyphenols, anthraquinones, anthracyclines, and miscellaneous compounds, and their antiviral mechanisms have been summarized. This review would be helpful to get a better grasp of anti-herpesvirus activity of natural products and their derivatives, and to evaluate the feasibility of natural compounds as an alternative therapy against herpesvirus infections in humans.
Article
Full-text available
Monkeypox (now Mpox), a zoonotic disease caused by the monkeypox virus (MPXV) is an emerging threat to global health. In the time span of only six months, from May to October 2022, the number of MPXV cases breached 80,000 and many of the outbreaks occurred in locations that had never previously reported MPXV. Currently there are no FDA-approved MPXV-specific vaccines or treatments, therefore, finding drugs to combat MPXV is of utmost importance. The A42R profilin-like protein of the MPXV is involved in cell development and motility making it a critical drug target. A42R protein is highly conserved across orthopoxviruses, thus A42R inhibitors may work for other family members. This study sought to identify potential A42R inhibitors for MPXV treatment using computational approaches. The energy minimized 3D structure of the A42R profilin-like protein (PDB ID: 4QWO) underwent virtual screening using a library of 36,366 compounds from Traditional Chinese Medicine (TCM), AfroDb, and PubChem databases as well as known inhibitor tecovirimat via AutoDock Vina. A total of seven compounds comprising PubChem CID: 11371962, ZINC000000899909, ZINC000001632866, ZINC000015151344, ZINC000013378519, ZINC000000086470, and ZINC000095486204, predicted to have favorable binding were shortlisted. Molecular docking suggested that all seven proposed compounds have higher binding affinities to A42R (–7.2 to –8.3 kcal/mol) than tecovirimat (–6.7 kcal/mol). This was corroborated by MM/PBSA calculations, with tecovirimat demonstrating the highest binding free energy of –68.694 kJ/mol (lowest binding affinity) compared to the seven shortlisted compounds that ranged from –73.252 to –97.140 kJ/mol. Furthermore, the 7 compounds in complex with A42R demonstrated higher stability than the A42R-tecovirimat complex when subjected to 100 ns molecular dynamics simulations. The protein-ligand interaction maps generated using LigPlot+ suggested that residues Met1, Glu3, Trp4, Ile7, Arg127, Val128, Thr131, and Asn133 are important for binding. These seven compounds were adequately profiled to be potential antivirals via PASS predictions and structural similarity searches. All seven potential lead compounds were scored Pa > Pi for antiviral activity while ZINC000001632866 and ZINC000015151344 were predicted as poxvirus inhibitors with Pa values of 0.315 and 0.215, and Pi values of 0.052 and 0.136, respectively. Further experimental validations of the identified lead compounds are required to corroborate their predicted activity. These seven identified compounds represent solid footing for development of antivirals against MPXV and other orthopoxviruses.
Article
Full-text available
Indian traditional medicinal systems are one of the oldest therapeutic systems in the world. Medicinal and aromatic plants play a dominant role in Indian traditional medicinal systems. Traditionally, many medicinal plants are used in India for their therapeutic relevance so much so that they have acquired a significant role in Indian religion as well. Many of these plants have proven antiviral effects. This review documents up-to-date information about many such medicinal herbs used in India which have got pharmacological significance in fighting viral infections. These plants surely have the potential to provide protection against Covid-19. The review presents a list of such plants along with their chemical ingredients and possible modes of action against the respective viral diseases. All information has been obtained by consulting the databases of Scopus, PubMed, Science Direct, Elsevier, Springer and relevant research papers and reports on COVID-19. The cited medicinal plants are used extensively in India as herbal remedies. The use of these plants is validated in light of research papers citing their ethnobotanical uses, important active principles and modes of action of the of medicinally important natural products. The plants listed have great potential to fight COVID-19 and other viral infections. Many of these are immunity boosters providing strength to the body to control the onset of diseases.
Chapter
Kaposi's sarcoma‐associated herpesvirus (KSHV), or human herpesvirus 8 (HHV 8), is a human γ 2 ‐herpesvirus and causes three neoplastic diseases. These are Kaposi's sarcoma (KS), a tumor derived from infected endothelial cells, as well as primary effusion lymphoma (PEL) and the plasma cell variant of multicentric Castleman's disease (MCD) . PEL is a monoclonal B cell lymphoma of post‐germinal center differentiation and MCD, an oligoclonal B cell lymphoproliferative disease of immature B cells. In addition, KSHV can cause polyclonal B‐cell lymphoproliferations, KSHV inflammatory cytokine syndrome (KICS), hemophagocytosis, and bone marrow failure, as well as rare cases of hepatitis. KSHV‐associated diseases are primarily of concern in AIDS patients, in transplant recipients from and in KSHV‐endemic countries, and in Africa. Currently used antiviral drugs with activity against KSHV are three herpesviral DNA polymerase inhibitors , ganciclovir , cidofovir , and foscarnet . These drugs are capable of inhibiting KSHV replication in tissue culture and in patients; however, when used alone, their efficacy against KSHV‐associated neoplastic disease is moderate. Other strategies to treat KSHV‐associated neoplasia therefore involve cancer chemotherapy, radiotherapy, kinase inhibitors, proteasome inhibitors, and the elimination of KSHV‐infected B cells using rituximab. Additional viral DNA polymerase inhibitors with activity against KSHV in preclinical studies exist, but have not yet been approved for clinical use. Currently ongoing efforts to develop new drugs with activity against KSHV include noncompetitive inhibitors of the DNA polymerase complex, inhibitors of the viral protease, the viral ssDNA‐binding protein, the viral terminase, viral glycoproteins as well as of the latent viral proteins LANA and vFLIP. In addition, several cellular proteins essential for viral replication, such as kinases, the proteasome, heat shock protein complexes HSP70 and HSP90, topoisomerases, metabolic regulators as well as cytokines and cytokine receptors have emerged as possible druggable targets.
Article
Full-text available
A historical perspective on the application of molecular dynamics (MD) to biological macromolecules is presented. Recent developments combining state-of-the-art force fields with continuum solvation calculations have allowed us to reach the fourth era of MD applications in which one can often derive both accurate structure and accurate relative free energies from molecular dynamics trajectories. We illustrate such applications on nucleic acid duplexes, RNA hairpins, protein folding trajectories, and protein−ligand, protein−protein, and protein−nucleic acid interactions.
Article
Full-text available
Glucose transporter 4 (GLUT4) is a principal glucose transporter in response to insulin, and impaired translocation or decreased expression of GLUT4 is believed to be one of the major pathological features of type 2 diabetes mellitus (T2DM). Therefore, induction of GLUT4 translocation or/and expression is a promising strategy for anti-T2DM drug discovery. Here we report that the natural product (+)-Rutamarin (Rut) functions as an efficient dual inducer on both insulin-induced GLUT4 translocation and expression. Rut-treated 3T3-L1 adipocytes exhibit efficiently enhanced insulin-induced glucose uptake, while diet-induced obese (DIO) mice based assays further confirm the Rut-induced improvement of glucose homeostasis and insulin sensitivity in vivo. Subsequent investigation of Rut acting targets indicates that as a specific protein tyrosine phosphatase 1B (PTP1B) inhibitor Rut induces basal GLUT4 translocation to some extent and largely enhances insulin-induced GLUT4 translocation through PI3 kinase-AKT/PKB pathway, while as an agonist of retinoid X receptor α (RXRα), Rut potently increases GLUT4 expression. Furthermore, by using molecular modeling and crystallographic approaches, the possible binding modes of Rut to these two targets have been also determined at atomic levels. All our results have thus highlighted the potential of Rut as both a valuable lead compound for anti-T2DM drug discovery and a promising chemical probe for GLUT4 associated pathways exploration.
Chapter
Research since the early 1970s has generated significant support for the concept that clinical manifestations of infectious diseases are often due more to the host immune response rather than to direct effects of a particular microbe. Perhaps the longest recognized example of the dominance of an immune response in determining a clinical presentation occurs in Mycobacterium leprae infection. However, the introduction of highly active antiretroviral therapy (HAART) for the treatment of human immunodeficiency virus (HIV) infection has markedly increased the appreciation for the dramatic interaction that can occur between microbes and a recovering immune system. The immune restoration mediated by HAART has markedly decreased the rates of opportunistic infections among HIV-infected patients, leading to dramatically lower mortality rates. However, in some patients, the recovery of immune function can lead to an inflammatory reaction aimed at either previously recognized or subclinical microbes or even autoimmune disorders. Multiple names have been given to this syndrome, including immune recovery disease, immune restoration disease, and immunoreconstitution disease. For the purposes of this chapter, we will utilize the term immune reconstitution inflammatory syndrome (IRIS) as it includes one of the defining features of these patients' presentations, i.e., inflammation.
Article
Shape comparing technologies based on Gaussian functions have been widely used in virtual screening of drug discovery. For efficiency, most of them adopt the First Order Gaussian Approximation (FOGA), in which the shape density of a molecule is represented as a simple sum of all individual atomic shape densities. In the current work, the effectiveness and error in shape similarity calculated by such an approximation are carefully analyzed. A new approach, which is called the Weighted Gaussian Algorithm (WEGA), is proposed to improve the accuracy of the first order approximation. The new approach significantly improves the accuracy of molecular volumes and reduces the error of shape similarity calculations by 37% using hard-sphere model as the reference. The new algorithm also keeps the simplicity and efficiency of the FOGA. A program based on the new method has been implemented for molecular overlay and shape-based virtual screening. With improved accuracy for shape similarity scores, the new algorithm also improves virtual screening results, particularly when shape-feature combo scoring function is used.
Article
Objective: With the recent identification of a new herpesvirus in patients with Kaposi's sarcoma (human herpesvirus-8 or Kaposi's sarcoma-associated herpesvirus), there have been several reports on the use of anti-herpesvirus therapy (foscarnet, ganciclovir and aciclovir) and risk of developing Kaposi's sarcoma. We therefore investigated the association between use of anti-herpesvirus drugs and Kaposi's sarcoma in a large unselected group of patients with AIDS. Patients and methods: We studied a group of HIV-positive patients at the Chelsea and Westminster Hospital, for whom details on all AIDS-defining diagnoses made during follow-up, treatment and regular CD4 counts were available. Cox proportional hazards models with time dependant covariates were used to assess the association between treatment with aciclovir, foscarnet and ganciclovir and risk of Kaposi's sarcoma. Results: A total of 3688 patients have been followed up for a median period of 4.2 years, during which time 598 patients (16.2%) developed Kaposi's sarcoma. After adjustments for sex, exposure category, age, treatment with antiretrovirals or Pneumocystis carinii pneumonia prophylaxis, the development of AIDS-defining conditions (including separate adjustment for the development of cytomegalovirus and herpes simplex virus) and CD4 count, there was a decreased risk of developing Kaposi's sarcoma with foscarnet (relative hazard (RH), 0.38; 95% confidence interval (CI), 0.15-0.95; P = 0.038) and with ganciclovir (RH, 0.39; 95% CI, 0.19-0.84; P = 0.015), but not with aciclovir (RH, 1.10; 95% CI, 0.88-1.38; P = 0.40). Conclusions: These results suggest that both foscarnet and ganciclovir may have some activity in preventing the occurrence of Kaposi's sarcoma, but that aciclovir has no benefit. Further studies of the effect of these drugs on the risk of Kaposi's sarcoma is warranted. (C) Lippincott-Raven Publishers.
Article
Here we report a set of new parameters for the generalized Born (GB) model consistent with the RESP atomic partial charge assignment protocol. Effective atomic radii and screen factors as parameters have been obtained through genetic algorithm optimization in the parameter space to minimize the differences between the calculated and experimental solvation free energies. Here, the calculated solvation free energies are based on a GB model using partial charges fitted from the electrostatic potentials based on the 6-31G* basis set with the nonelectrostatic contributions to the free energy of solvation modeled in terms of the solvent accessible surface area (SASA). The mean unsigned error in the solvation free energies calculated by the GB/surface area calculations using the final parameters of the 328 neutral molecules in the training set is 0.85 kcal/mol, and for the 30 charged molecules the value is 4.36 kcal/mol. The refined parameters were then applied to predict the solvation free energies of 44 neutral or charged organic molecules and 15 proteins, and reliable results were obtained for both organic molecules and proteins. For the 36 neutral organic molecules in the test set, our parameters incurred an unsigned mean error of 0.73 kcal/mol, and for the eight charged molecules in the test set, our parameters incurred an unsigned mean error of 3.65 kcal/mol. For the 44 organic molecules, the performance of the GB/SA model based on our new parameters was much better than Possion-Boltzmann (PB)/SA and GB/SA based on Jayaram's parameters. For the 15 proteins randomly selected from the Protein Data Bank, the calculated results from GB/SA based on our new parameters also gave consistent results with those from PB/SA and were much better than GB/SA based on Jayaram's parameters. This model might be widely applied in molecules dynamics, protein folding, molecular docking, free energy calculations, and conformation analysis. Moreover, we are now supplying a program to help AMBER users apply our new parameters to their MD simulations.
Article
The Kaposi's sarcoma-associated herpesvirus (KSHV), also called human herpesvirus 8 (HHV–8), has been found to be present in a limited subset of lymphoproliferative disorders. Among these are the primary effusion lymphomas, formerly designated body cavity-based lymphomas, a rare type of malignant lymphoma which possesses an unusual set of clinical and biologic features, suggesting that they represent a distinct disease entity. This virus is also present in a large proportion of cases of multicentric Castleman's disease, particularly those associated with HIV–infection. In addition, KSHV has been implicated in the pathogenesis of multiple myeloma, where it has been identified in bone marrow adherent cells but not in the neoplastic myeloma plasma cell population. However, the latter finding remains controversial. The discovery of KSHV in a subset of malignant lymphomas has allowed the development of lymphoma cell lines which now serve as biological reagents for propagating the virus, as a substrate for serologic assays, and as a model system for pathobiologic studies. This review discusses the features of KSHV–associated lymphoproliferative disorders and the evidence supporting its role in the pathogenesis of these diseases.
Article
A revised generalized Born/surface area (GB/SA) continuum solvation model has been developed for water that is compatible with the Merck molecular force field (MMFF). This model gives free energies of aqueous solvation that are comparable in accuracy to the original water model when the OPLS∗ force field is employed. The average unsigned error in aqueous ΔGsol using the new water model and MMFF is 0.62 kcal/mol for a training set of 82 solutes compared to 1.24 kcal/mol for the original GB/SA water model and MMFF. The average unsigned errors for 47 neutral solutes outside the training set and 10 ions are 0.96 and 2.32 kcal/mol, respectively. By comparison, the average errors for the test set and ions using the original GB/SA water model are 1.76 and 5.32 kcal/mol. This revised parameter set provides a more accurate representation of aqueous solvation for use with MMFF.