SlideShare a Scribd company logo
1 of 8
Download to read offline
BIOCOMPATIBILITY STUDIES Original Research
Nanosilver based anionic linear globular dendrimer with a special
significant antiretroviral activity
Mehdi Shafiee Ardestani1 • Alireza Salehi Fordoei2,3 • Asghar Abdoli2 •
Reza Ahangari Cohan4 • Golnaz Bahramali2 • Seyed Mehdi Sadat2 •
Seyed Davar Siadat5 • Hamid Moloudian6 • Nasser Nassiri Koopaei7 •
Azam Bolhasani2 • Pooneh Rahimi2 • Soheila Hekmat2 • Mehdi Davari2 •
Mohammad Reza Aghasadeghi2
Received: 10 January 2015 / Accepted: 30 March 2015
Ó Springer Science+Business Media New York 2015
Abstract HIV is commonly caused to a very complicated
disease which has not any recognized vaccine, so designing
and development of novel antiretroviral agents with
specific application of nanomedicine is a globally inter-
ested research subject worldwide. In the current study, a
novel structure of silver complexes with anionic linear
globular dendrimer was synthesized, characterized and
then assessed against HIV replication pathway in vitro as
well. The results showed a very good yield of synthesis (up
to 70 %) for the nano-complex as well as a very potent
significant (P  0.05) antiretroviral activity with non-sev-
ere toxic effects in comparison with the Nevirapine as
standard drug in positive control group. According to the
present data, silver anionic linear globular dendrimers
complex may have a promising future to inhibit replication
of HIV viruse in clinical practice.
Abbreviations
HIV Human immunodeficiency virus
RSV Respiratory syncytial virus
PEG-600 Polyethylene glycole 600 Da
DCC Dicyclohexyl carbodiimide
DLS Dynamic light scattering
DMSO Dimethyl sulfoxide
FTIR Fourier transforms spectroscopy
XTT 2,3-Bis(2-methoxy-4-nitro-5-sulfophenyl)-2H-
tetrazolium-5-carboxanilide
MPPI Mannosylated poly (propyleneimine)
dendrimer
1 Introduction
Globally, more than 40 million people suffer from the
human immunodeficiency virus (HIV) infection. The high
activity antiretroviral therapy (HAART) combining three
or more of antiretroviral (ARV) drugs administered for
lifelong renders acquired immunodeficiency syndrome
(AIDS) as a controlled chronic disease [1]. Limitations of
the current ARV include long lasting medications with
remarkable adverse effects, poor patient-medication ad-
herence, multi-drug resistance (MDR) due to mutations,
drug–drug interactions, poor drug pharmacokinetics, viral
load rise very quickly following medication discontinued
and affordability as well as establishing a latent infection
[2]. Unique properties of dendrimers as novel and exciting
& Mohammad Reza Aghasadeghi
mr_sadeqi@yahoo.com; mrasadeghi@pasteur.ac.ir
1
Department of Radiopharmacy, Faculty of Pharmacy, Tehran
University of Medical Sciences, Tehran, Iran
2
Department of Hepatitis and AIDS, Pasteur Institute of Iran,
13164 Tehran, Iran
3
Department of Biology, College of Basic Sciences, Tehran
Science and Research Branch, Islamic Azad University,
Tehran, Iran
4
Virology Research Group, Department of Rabies, Pasteur
Institute of Iran, Tehran, Iran
5
Department of Bacteriology, Pasteur Institute of Iran, Tehran,
Iran
6
Faculty of Pharmacy, Pharmaceutical Quality Assurance
Research Center, Tehran University of Medical Sciences,
Tehran, Iran
7
Department of Pharmaceutical Biotechnology, Faculty of
Pharmacy, Tehran University of Medical Sciences, Tehran,
Iran
123
J Mater Sci: Mater Med (2015) 26:179
DOI 10.1007/s10856-015-5510-7
polymers such as uniform size, water solubility, available
internal cavities, multivalency, well-defined molecular
weight, among others, make them attractive for drug de-
livery purposes since past decades [3–5]. These features are
caused by highly branched, spherical and nanometer di-
mension of dendrimers [4]. Furthermore, attempts have
been made to apply dendrimers to other areas such as
immunology and development of vaccines, antimicrobials
and antivirals [3]. Biocompatibility and release of drugs
from dendrimers are controversial issues [3]. Selection of
polymers has always been affected by issues such as
biodegradability, biocompatibility, bioadhesiveness while
respecting physical and biochemical properties [5]. Spheric
shape of dendrimers makes their structures much more
accurately controllable. They are indeed nano-sized mole-
cules with sizes between 10 and 100 nm [5].
Dendrimers due to aforementioned characteristics have
emerged as an important class of antiretroviral carriers [6].
Dendrimers as organic nano-platforms can act in
therapeutic and diagnostic applications [7]. To deliver a
therapeutic and non-toxic dose of drug to diseases, nano
and micro-carriers are efficient tools [8]. Increased effi-
ciency and decreased toxicity of anti-retroviral drug loaded
dendrimers were investigated in previous studies. Dutta
and Jain studied targeting and anti-HIV potential of lami-
vudine loaded mannosylated poly (propyleneimine)
(MPPI) dendrimer and concluded that the drug release
profile and cellular uptake of drug were improved sig-
nificantly. MPPI is a surface modified version of poly
(propyleneimine) (PPI) [9]. Passive targeting of drug
loaded nano-carriers is limited to tumors, the reticular en-
dothelial system and lymph nodes. Attractive features of
dendrimers like narrow molecular weight distribution,
small size and easy incorporation of targeting ligand ren-
ders them remarkable applicability in antiretroviral therapy
[10]. Dendrimers are also advantageous with respect to
their drug interactions [11]. Dendrimers could be widely
used in targeted and controlled release drug delivery sys-
tems; however, toxicity problems still exist and need some
modifications. Release profile of dendrimers is easily
modifiable and controllable [12]. Citric acid–polyethylene
glycol–citric acid (CPEGC) dendrimers were mentioned as
biocompatible compounds and investigated as drug deliv-
ery systems. Some small and hydrophobic molecules and
drugs were incorporated into the above dendrimer. These
dendrimers had good water solubility, biocompatibility and
low toxicity [13]. Synthesis of dendrimers has recently
been facilitated by the divergent ‘lego’ and the convergent
‘click’ approaches. Drugs could be physically loaded
within the dendrimer or chemically attached to the surface.
Ester-terminated dendrimers are more bioavailable than
amino-terminated ones. Increase of dendrimer sizes makes
them more soluble. Nonetheless, cytotoxic effects of
dendrimers increase with their generation number. Cyto-
toxicity could be reduced by dendrimer functionalization
(particularly with PEG). Dendrimers can inherently act
against tumors, bacteria and viruses [14].
AgNPs have been used for their antibiotic effects,
noteworthy that no cross-resistance with other antibiotics is
evident and efficacy against multi drug resistant strains [15,
16]. AgNPs show wide spectrum against different bacteria
and fungi [17–22] commercially used in health and im-
plantable biomaterial, general utilities and home appli-
ances. Moreover, antiviral effects of AgNPs have attracted
attention recently. AgNPs present remarkable antiviral
activity against HIV, hepatitis B, HSV, RSV, monkeypox
virus, tacaribe virus (TCRV), RNA viruses and adenovirus
type 3 [21, 23–26]. The supporting mechanism for AgNPs
antiretroviral effect is through glycoprotein 120 (gp120)
binding and prevention following the penetration and in-
terfering with intracellular stages. With the characteristics
mentioned, AgNPs are applied in a vaginal product to
prevent sexually transmitted diseases like HIV. However,
higher efficacy has been proposed with the concomitant
use of neutralizing antibodies [23]. Wide anti-infectious
properties of AgNPs as well as wound healing effects
makes silver suitable candidate for topical formulations
[20, 22, 27, 28].
Limitations of current antiretroviral drugs such as ex-
tensive first pass metabolism, astrointestinal degradation,
poor solubility, etc. lead investigation of novel drug de-
livery systems (NDDS) such as sustained and targeted drug
delivery systems. So, in the present study, a novel structure
of silver complexes with anionic linear globular dendrimer
was synthesized, characterized and then assessed against
HIV replication pathway in vitro.
2 Materials and methods
2.1 Materials
PEG 600 was purchased (from Merck,Germany). Dicy-
clohexylcarbodiimide, dimethylsulfoxide, citric acid and
silver were obtained (from Merck,Germany). Besides
pSPAX.2, pmzNL4-3 and pMD2.G plasmids were used for
transfection. In cellular studies, HEK293T cell line and
DMEM primary culture medium were used. HEK is an
abbreviation for human embryonic kidney cells. This cell
line was chosen for this research because it has superior
survival and proliferation compared to adult cells.
2.2 Synthesis of dendrimers G1 and G2
Synthesis of compound G1 of dendrimer was initiated
through the reaction of PEG 600 (as the core of dendrimer)
179 Page 2 of 8 J Mater Sci: Mater Med (2015) 26:179
123
and dicyclohexylcarbodiimide (DCC) in anhydrous DMSO
in the condition of darkness and vacuum. DCC activated
PEG by substitution with hydrogen atom of carboxylic acid
groups. Then, DCC was replaced with citric acid, and so
color of the tick solution changed to orange with stench.
Reaction was terminated with adding water and after fil-
tration; yellow-brown solution of dendrimer was obtained.
Primarily, synthesis of compound G1 was confirmed by
thin layer chromatography (TLC); stationary phase: silica
gel, mobile phase: 65 % CCl4/35 % MeOH. Then, the
product of G1 was separated and purified by dialysis.
Volume and concentration of dialysis buffer was 1600 fold
and half of the contents of dialysis bag, respectively. Pore
size of dialysis bag was 500–1000 Da. PEG was used as
limiting substrate of reaction and didn’t leave any residue.
Other substrates were excess. Mass of dendrimer was
transferred to several vials and lyophilized in yield 91 %
after freezing in liquid nitrogen. Procedures were severally
repeated to obtain the optimum procedure. To synthesize
dendrimer G2, DCC and citric acid were added to solution
of dendrimer G1 in DMSO and the solution became ticker
and thereafter dark cream color was appeared. In this stage,
compound G1, was rate limiting of reaction. As synthesis of
dendrimer G1, reaction was terminated with water addition,
solution was filtrated, dialyzed, and powder was lyophi-
lized. Structure of dendrimer G2 (in yield 73 %) was
confirmed by FT-IR. Size and electric charge of G1 and G2
dendrimers was determined by zetasizer (dynamic light
scattering, DLS).
Briefly, 2 mg of plasmid mixture was dissolved in
150 ll of deficient culture medium. Then, 40 ll of lipo-
fectin was added to the tube containing DNA and incubated
in room temperature for 10 min. Following that, 3 mL of
cell medium.
2.3 Preparation of Ag-dendrimer conjugates
Tendency of nano-silver particles to the oxygen atoms of
dendrimer leads to the reaction and incorporation of silver
into the dendrimers. In the isolated and light protected
condition, the mixture of dendrimer and silver was stirred
for 24 h. Yellow-brown color of mixture changed to gray.
Then, the mixture was filtered and brown solution of Ag-
dendrimer conjugated was separated from insoluble free
silver. Final product (liquid or lyophilized) was perma-
nently preserved from light and air, during the analyses.
Structure of product was assessed by FT-IR and compared
with G1 and G2 dendrimers. Size and charge of the final
product was determined and compared as well. Imaging of
Ag-dendrimer conjugated was made by AFM. Concentra-
tion of conjugated silver was determined by ICP-Mass
technique. Maximum incorporation of silver into the den-
drimer was calculated by ICP-Mass and divided by total
mass of G2 dendrimer and multiplied by 100; therefore
percentage of loading complex was obtained (52 %).
2.4 Cell culture and antiviral assessments
2.4.1 Cells, transfection and virus production
To produce HIV-1 SCR virions pseudotyped by VSVG,
Qiagen sample was used. First HEK293T cells were cul-
tured and maintained in Dulbecco’s modified Eagle’s cul-
ture medium (DMEM, Sigma, USA) supplemented with
FBS (15 %), penicillin (100 U/ml), streptomycin (100 lg/
ml), and glutamine (2 mmol/l); incubated in 5 % CO2,
37 °C inside a 6-well plate until they reach a total number
of approximately 1.2 9 106
. Transfection was carried out
using standard Lipofectamine 2000 according to Qiagen
protocol. Plasmids of pSPAX.2, pmzNL4-3 and pMD2.G
were simultaneously transfected into HEK293T cells to
produce proviruses. Next, cell culture plates were incu-
bated for minimum time of 24 h in 37 °C and 5 % CO2.
HEPES (4-(2-hydroxyethyl)-1-piperazineethanesulfonic
acid) (25 mM) was used to adjust the pH of the cell culture
medium in transfection stage. To prepare virus stock, cul-
ture supernatants of the transfected cells were harvested at
24, 48, and 72 h post transfection, and after filtration were
concentrated for 2 h in 60,0009g in 4 °C by ultracen-
trifugation [29]. The pelleted virions were solved in 1/20
volume of RPMI 1640 by mixing gently overnight at 4 °C
to prevent virus aggregation, quantified by P24 ELISA
assay kit (Biorad) and stored at -70 °C until use [30].
HEK cells were used to determine P24 titer of SCR HIV-1
virions pseudotyped by stomatitis virus surface VSVG
vesicular glycoprotein. A 60 9 103
count of HEK cells
were inoculated in each well of a 24 well plate containing
250 ll of culture medium and then 600 ng of P24 was
added to each well. Next, the cells were washed every 5 h
using 10 % FBS. Every time, the supernatants were col-
lected and replaced with new culture medium. Finally, P24
concentration in the supernatant was determined according
to the ELISA capture protocol (Biomerieux) developed by
the manufacturing company. All the components were
dissolved in dimethylsulfoxide (DMSO) with concentra-
tions ranging from 10 lM to 10 mM. Nevirapine (NVP)
was dissolved in DMSO with 50 lM concentration, ex-
tracted from commercial preparations was used as positive
control to inhibit viral growth and DMSO with final con-
centration of 1 % as the negative control. All the ex-
periments were triplicate. Toxicity of compounds was
separately estimated on the HEK cells. CC50 (cytotoxicity
capacity 50 %) was calculated. Dividing CC50 by IC50
provided TI (therapeutic index) and/or SI (selectivity in-
dex) (TI or SI = CC50/IC50). Antiviral effects of different
compounds were compared through TI or SI indices.
J Mater Sci: Mater Med (2015) 26:179 Page 3 of 8 179
123
2.5 Evaluation of the new nano-drug effectiveness
on HIV replication
A HEK cells count of 60 9 103
in each well of a 24 well
plate containing 250 ll of culture medium was prepared.
SCR HIV virions and 600 ng P24 were added to each well.
The compounds were added in four concentrations (20, 40,
80 and 120 lM) to the wells as triplicate.
After 5 h, the cells were washed twice with DMEM/FBS
10 % [31]. Again, the compounds and fresh medium with
the same amounts were added. At 72 h after inoculation,
the supernatant was collected. The P24 level of the su-
pernatant was determined with ELISA capture (Biorad).
Related ELISA test was performed and finally, the con-
centration was calculated. To examine cellular prolif-
eration, XTT method (Roche) was used [30]. Medium of
phenol red was used to diminish test errors in proliferation
assay.
3 Results
3.1 Time relation of the SCR viral replication
Replication of the SCR HIV-1 virions psuedotyped by
VSVG on the HEK cells was determined after viral
inoculation. P24 secretion was started at the first day and
increased to a maximum level at 48 h after SCR virions
inoculation. Then, P24 level was gradually decreased from
the fourth day after inoculation. As the results denote,
maximum cumulative amount of P24 in the supernatant
was 72 h after inoculation. Therefore, P24 determination in
the supernatant was used to assess the viral replication at
72 h after inoculation (Fig. 1).
3.2 SCR HIV-1 replication regarding the primary
MOI
HEK cells were infected by different multiplicity of in-
fection (MOI) of pseudotype SCR virions. The supernatant
of the infected cells was assessed through P24 level 3 days
after inoculation. An inverse linear relationship between
MOI and P24 production makes this test a precise method
to investigate replication. Based on the assessments, the
adequate viral count for replication start is 600 ng P24
(Fig. 2).
3.3 Nevirapine efficacy as the control positive
Based on the results, suitable viral count for replication
(600 ng P24) and time (72 h) for P24 determination was
selected. To assess nevirapine function that is a NRTI drug,
SCR virions replication in the presence of its different
concentrations was measured. The results showed that in-
creasing nevirapine level decreased P24 production and
50,000 nM concentration inhibited P24 production. These
results revealed the correct function of the drug in the
primary test conditions and replication assessment based
on SCR HIV-1 suitably shows drug function in different
concentrations (Fig. 3).
3.4 Inhibitory effects of silver dendrimer
nanoconjugate
The ability of Iron chelating compounds to inhibit HIV
virus replication was investigated by assessing HIV repli-
cation in the presence of various compounds and calculated
based on the light absorption in P24 ELISA OD.
Nanoconjugated silver dendrimer effects in 20, 40, 80 and
120 lM concentrations on HEK cells was assessed. The
best antiviral effects were observed in 120 lM that inhib-
ited viral contamination up to 87 %. Generally speaking,
higher levels of drug produced higher level of viral
Fig. 1 The effects of nanosilver on the P24 levels at different time
exposures
Fig. 2 The effects of nanosilver on the different amounts of starter
virus
179 Page 4 of 8 J Mater Sci: Mater Med (2015) 26:179
123
inhibitory effects. This was also evident for control (silver
nitrate) (see more detailed data at Table 1, Fig. 4).
3.5 Cytotoxicity of nanoconjugated silver
dendrimers in different concentrations
Based on ODs from ELISA proliferation test, cytotoxicity
was assessed through XTT method after 72 h. Although,
the compounds showed potent antiviral effects but they had
cytotoxicity as well. Cell survival in concentrations of 40,
80 and 120 lM was 23, 36 and 48 %, respectively and was
50 % in 80 and 120 lM concentration of silver nitrate as
the control. DMSO was used as the positive control. In-
hibitory effect was low at 20 lM concentration while low
cytotoxicity was observed as well (see more detailed data
at Table 2, Fig. 5).
3.6 TI indexing
According to the calculated IC50 and CC50, TI index was
determined for different compounds. Comparing this index
for various concentrations of nanoconjugated silver den-
drimers provides a method to identify concentrations with
higher antiretroviral effects and lower cytotoxicity.
Therapeutic index (TI) in 20 lM concentration was 5.3, the
best among all concentrations. TI for 40, 80 and 120 lM
concentrations was 0.96, 0.54 and 0.26, respectively.
Dendrimeric seizes for G1 and G2 products were 70 and
90 nm, respectively; dendrimeric charges were -3.5 and
-11.8 millivolt/Ampere (mv A-1
), respectively. More
Fig. 3 The effects of
Nevirapine on the P24 at
different time exposures
Table 1 Depiction of some
details on HIV inhibitory effects
of nanosilver and the control
drug
P24 OD Mean P24 OD HIV inhibitory %
Uninfected cells 0.04, 0.03 0.035 –
Virus control (without drug) 0.8, 0.9, 0.7 0.8 0
Nevirapine (anti-retroviral drug) 0.02, 0.05 0.03 100
Ag-dendrimer (120 lM) 0.08, 0.2 0.14 87
Ag-dendrimer (80 lM) 0.21, 0.32 0.26 67
Ag-dendrimer (40 lM) 0.42, 0.34 0.38 52
Ag-dendrimer (20 lM) 0.48, 0.88 0.68 15
AgNO3 (control) (120 lM) 0.04, 0.1 0.07 96
AgNO3 (control) (80 lM) 0.32, 0.42 0.37 53
AgNO3 (control) (40 lM) 0.55, 0.4 0.47 41
AgNO3 (control) (20 lM) 0.62, 0.92 0.77 4
Fig. 4 Antiretroviral effects of AgNO3 and silver based dendrimer
nanoparticle
J Mater Sci: Mater Med (2015) 26:179 Page 5 of 8 179
123
citric acid moieties at the surface of compound G2 leads to
its larger size and more negative charge compared with
compound G1. Atomic force microscopy (AFM) imaging
of dendrimer G2 was performed and compared with G1.
The important absorption bonds in FTIR spectrum for G1
are: 3000–3500 cm-1
for hydroxyl group of citric acid
attached to PEG, 2800–2900 cm-1
for aliphatic carbon
chain of citric acid and PEG, and shifted peak of carbonyl
group of citric acid from 1700 to 1600 cm-1
that confirms
low concentration of citric acid in the structure of den-
drimer and conjugation of citric acid and PEG. The ab-
sorbed bonds in FTIR spectrum [OH group of citric acid
attached to PEG (broad, 3500–3000 cm-1
), C=O
(1600 cm-1
)] for G2 are: wide and strong peak from 3000
to 3500 cm-1
for hydroxyl group of citric acid, strength-
ened peak from 2800 to 2900 cm-1
for aliphatic carbon of
citric acid and PEG and a strong peak about 1700 cm-1
is
related to strengthened steric bond of citric acid addition
[OH group of citric acid (broad and strong,
3500–3000 cm-1
), steric bond of citric acid (1700 cm-1
)].
Last mentioned peak implies high concentration of citric
acid in the structure of compound G2 and conjugation of
citric acid and PEG. Considering significant increase in
aliphatic and hydroxyl peaks of compound G2 compared
with G1; thus it can be concluded that IR spectra approved
the synthesis of both dendrimer generations.
4 Discussion
HIV transcripts are RNA to DNA by RT and introduce
provirus into the host cell DNA by integrase. CD4?
T cells
are the major hosts and with proceeding decrease of their
reservoir, opportunistic infections and cancers lead death.
HIV/AIDS has imposed heavy economic and health costs
to world health. Because of loss of vaccine, antiviral drugs
have been the best confronting approach. Kim et al. ex-
amined oral toxicity of AgNPs for 3 months finding the
target organ to be the liver with biliary tract hyperplasia,
possible necrosis, fibrosis and pigmentation. Finally, the
LOAEL and NOAEL were determined to be 125 and
30 mg/kg, respectively, while AgNPs could accumulate in
tissues investigated [32]. AgNPs exert bactericidal activity
through concentrations that no remarkable acute human
toxicity would emerge [22, 33]. But toxicity of available
antiviral drugs and resistance of virus increased the im-
portance of development of new antiretroviral drugs [34,
35]. Inhibition of virus replication via interaction with
cellular pathways is the current strategy [36–38].
Combination of nanotechnology and antimicrobial prop-
erties of Cr, Ag, Ti, and Zn presents effective anti-HIV ap-
proaches. Some studies have shown antiviral activities of
nano-silver (against HIV-1, HBV, HSV, RSV, and monkey-
pox virus) [39]. Some other studies showed nano-silver par-
ticles inhibit HIV fusion to the host cell via interaction with gp
120 [23, 40]. Furthermore nano-silver particles can interact
with HIV through other ways such as inhibition of reverse
transcriptase, protein activity and inhibition of host cell cycle.
Dendrimers can trap different molecules within their
branches and protect them from environmental influences
and controllably release in targeted positions. Also den-
drimers increase solubility of drugs by trapping them.
Table 2 Illustration of some
details on cell viability effects
of nanosilver and the control
drug
ELISA OD (450 nm) Mean OD Cell viability %
Un-treated (1 ll DMSO) cells 0.65, 0.61, 0.55 0.6 100
Ag-dendrimer (120 lg) 0.14, 0.14 0.14 23
Ag-dendrimer (80 lg) 0.22, 0.22 0.22 36
Ag-dendrimer (40 lg) 0.31, 0.28 0.29 48
Ag-dendrimer (20 lg) 0.49, 0.51 0.5 83
AgNO3 (control) (120 lg) 0.31, 0.29 0.3 50
AgNO3 (control) (80 lg) 0.32, 0.28 0.3 50
AgNO3 (control) (40 lg) 0.43, 0.45 0.44 73
AgNO3 (control) (20 lg) 0.53, 0.43 0.48 80
Fig. 5 Effects of different concentrations of Ag-dendrimer and
AgNO3 exposures on cell viability percentage results of
179 Page 6 of 8 J Mater Sci: Mater Med (2015) 26:179
123
Dendrimers can carry molecules on their surface via in-
teraction with functional groups; a property that enables
specific cell or tissue targeting.
In this study we investigated several concentrations of
Ag-dendrimer nano-conjugated compounds against repli-
cation of HIV and determined cytotoxicity of the new
compound in different concentrations. Researches showed
the Ag-dendrimer conjugated reduces expression of P24
antigen on the surface of monocytes and lymphocytes. It
seems virus inhibition is due to the reduction of cellular
proliferation. Inhibition of cellular proliferation can lead
inhibition of P24 production in lymphocytes. As a result, it
seems Ag-dendrimer conjugated don’t target virus directly
and indeed attack host cell of replicating virus. But some
other drugs such as nevirapine inhibit replication, directly
without any influence on cellular proliferation. HIV repli-
cation rate and survival was assessed after application of
several concentrations (20, 40, 80, and 120 lM) of new
compound. Inhibition of cellular proliferation cause to in-
hibit replication of virus but clinical researches have shown
nano-silver particles decrease P24 level with 50 % without
any effect on cellular proliferation. In a mathematical
model for anti CD4?
therapy, it has been showed that 25 %
decrease in CD4?
cell counts can reduce function of virus
and rate of mutations [41]. With this respect current
compound by killing proliferating CD4?
T cells and de-
pleting host cell from dATP, inhibit P24 production and
HIV-1 replication [42–45]. Our previous studies have
demonstrated that biocompatible range of Anionic linear-
globular dendrimer up to the concentration of 0.5 mg/ml
and less of the Anionic linear-globular dendrimer-cis-
platinum(II) conjugates would be required to obtain a
similar therapeutic effect against different cancer cell lines
when compared with cisplatin treatment alone [46, 47].
Decreasing dose of drug usage by dendrimers may result in
less drug resistance and drug side effects.
Results of this study showed that all concentrations of
Ag-dendrimer nano-conjugated could inhibit HIV effec-
tively. Inhibition percent of HIV for concentrations 120,
80, 40, and 20 lM is 78, 67, 52, and 15, respectively. First
three concentrations have highest inhibition rates but show
high level of cytotoxicity as well (77, 64, and 52 %, re-
spectively). Although inhibition rate of concentration
20 lM was only 15 %; but its cytotoxicity was 17 % and
this concentration of new compound is totally more ap-
propriate. AgNO3 was used as control and concentration of
120 lM showed better inhibition and lower toxicity (96
and 50 %, respectively) than conjugated compound but not
appropriate for clinical application. At the concentration of
80 lM, the new compound is better than AgNO3 (53 %
inhibition), but its cytotoxicity is high (50 %). In this
concentration both compounds had remarkable cyto-
toxicity, too. At the concentration of 40 lM,
nanoconjugated compound had a stronger inhibition and
lower cell survival profile (48 % in front of 73 %) than
AgNO3. In this concentration none of compounds couldn’t
be selected as an antiretroviral drug. At the concentration
of 20 lM, AgNO3 has a very low inhibition rate (4 %) and
a considerable cytotoxicity (20 %), while Ag-dendrimer at
concentration of 20 % had a stronger inhibition and lower
cytotoxicity profile.
5 Conclusion
Finally, we conclude that the concentration of 20 lM of
nanoconjugated compound has the best therapeutic profile
(stronger inhibition and lower toxicity). Silver saturated
dendrimers were used in this study and highly toxic com-
pounds were obtained. Lower silver loaded dendrimers or
other generations of dendrimer such as G1 can lead to more
applicable and different results. Also we propose that the
combination of concentration of 20 lM dendrimer with
other antiretroviral drugs to be investigated for better
therapeutic profile.
Acknowledgments We thank all members of the Hepatitis and
AIDS Department and Department of Radiopharmacy, Faculty of
Pharmacy, Tehran University of Medical Sciences of Pasteur Institute
of Iran and for their advice and assistance with the experiments. This
work was supported by Grant No. 519 from Pasteur Institute of Iran,
Tehran, Iran.
Conflict of interest The authors disclose no potential conflicts of
interest.
References
1. Sosnik A, Chiappetta DA, Carcaboso AM. Drug delivery systems
in HIV pharmacotherapy: what has been done and the challenges
standing ahead. J Control Release. 2009;138(1):2–15.
2. Gupta U, Jain NK. Non-polymeric nano-carriers in HIV/AIDS
drug delivery and targeting. Adv Drug Deliv Rev.
2010;62(4–5):478–90.
3. Gillies ER, Frechet JM. Dendrimers and dendritic polymers in
drug delivery. Drug Discov Today. 2005;10(1):35–43.
4. Patri AK, Majoros IJ, Baker JR. Dendritic polymer macro-
molecular carriers for drug delivery. Curr Opin Chem Biol.
2002;6(4):466–71.
5. Pillai O, Panchagnula R. Polymers in drug delivery. Curr Opin
Chem Biol. 2001;5(4):447–51.
6. Ojewole E, Mackraj I, Naidoo P, Govender T. Exploring the use
of novel drug delivery systems for antiretroviral drugs. Eur J
Pharm Biopharm. 2008;70(3):697–710.
7. Jabr-Milane L, van Vlerken L, Devalapally H, Shenoy D, Ko-
mareddy S, Bhavsar M, Amiji M. Multi-functional nanocarriers
for targeted delivery of drugs and genes. J Control Release.
2008;130(2):121–8.
8. Kumar M. Nano and microparticles as controlled drug delivery
devices. Int J Pharm Pharm Sci. 2000;3(2):234–58.
9. Dutta T, Jain NK. Targeting potential and anti-HIV activity of
lamivudine loaded mannosylated poly (propyleneimine) den-
drimer. Biochim Biophys Acta. 2007;1770(4):681–6.
J Mater Sci: Mater Med (2015) 26:179 Page 7 of 8 179
123
10. Gunaseelan S, Gunaseelan K, Deshmukh M, Zhang X, Sinko PJ.
Surface modifications of nanocarriers for effective intracellular de-
livery of anti-HIV drugs. Adv Drug Deliv Rev. 2010;62(4–5):518–31.
11. D’Emanuele A, Attwood D. Dendrimer–drug interactions. Adv
Drug Deliv Rev. 2005;57(15):2147–62.
12. Nanjwade BK, Bechra HM, Derkar GK, Manvi FV, Nanjwade
VK. Dendrimers: emerging polymers for drug-delivery systems.
Eur J Pharm Sci. 2009;38(3):185–96.
13. Namazi H, Adeli M. Dendrimers of citric acid and poly (ethylene
glycol) as the new drug-delivery agents. Biomaterials. 2005;26(10):
1175–83.
14. Goldberg M, Langer R, Jia X. Nanostructured materials for ap-
plications in drug delivery and tissue engineering. J Biomater Sci
Polym Ed. 2007;18(3):241–68.
15. Kalishwaralal K, BarathManiKanth S, Pandian SR, Deepak V,
Gurunathan S. Silver nanoparticles impede the biofilm formation
by Pseudomonas aeruginosa and Staphylococcus epidermidis.
Colloids Surf B. 2010;79(2):340–4.
16. Rai MK, Deshmukh SD, Ingle AP, Gade AK. Silver nanoparti-
cles: the powerful nanoweapon against multidrug-resistant bac-
teria. J Appl Microbiol. 2012;112(5):841–52.
17. He Y, Du Z, Lv H, Jia Q, Tang Z, Zheng X, Zhang K, Zhao F.
Green synthesis of silver nanoparticles by Chrysanthemum
morifolium Ramat. extract and their application in clinical ul-
trasound gel. Int J Nanomed. 2013;8:1809.
18. Kim KJ, Sung WS, Moon SK, Choi JS, Kim JG, Lee DG. An-
tifungal effect of silver nanoparticles on dermatophytes. J Mi-
crobiol Biotechnol. 2008;18(8):1482–4.
19. Murugan E, Vimala G. Effective functionalization of multiwalled
carbon nanotube with amphiphilic poly(propyleneimine) dendrimer
carrying silver nanoparticles for better dispersability and antimi-
crobial activity. J Colloid Interface Sci. 2011;357(2):354–65.
20. You C, Han C, Wang X, Zheng Y, Li Q, Hu X, Sun H. The progress
of silver nanoparticles in the antibacterial mechanism, clinical ap-
plication and cytotoxicity. Mol Biol Rep. 2012;39(9):9193–201.
21. Lu L, Sun RW, Chen R, Hui CK, Ho CM, Luk JM, Lau GK, Che
CM. Silver nanoparticles inhibit hepatitis B virus replication.
Antivir Ther. 2008;13(2):253.
22. Morones JR, Elechiguerra JL, Camacho A, Holt K, Kouri JB,
Ramı´rez JT, Yacaman MJ. The bactericidal effect of silver
nanoparticles. Nanotechnology. 2005;16(10):2346.
23. Lara HH, Ixtepan-Turrent L, Garza Trevin˜o EN, Singh DK. Use
of silver nanoparticles increased inhibition of cell-associated
HIV-1 infection by neutralizing antibodies developed against
HIV-1 envelope proteins. J Nanobiotechnol. 2011;9:38.
24. Lai T, Hou Q, Yang H, Luo X, Xi M. Clinical application of a
novel sliver nanoparticles biosensor based on localized surface
plasmon resonance for detecting the microalbuminuria. Acta
Biochim Biophys Sin. 2010;42(11):787–92.
25. Mohammed Fayaz A, Ao Z, Girilal M, Chen L, Xiao X,
Kalaichelvan P, Yao X. Inactivation of microbial infectiousness by
silver nanoparticles-coated condom: a new approach to inhibit
HIV-and HSV-transmitted infection. Int J Nanomed. 2012;7:5007.
26. Butkus MA, Labare MP, Starke JA, Moon K, Talbot M. Use of
aqueous silver to enhance inactivation of coliphage MS-2 by UV
disinfection. Appl Environ Microbiol. 2004;70(5):2848–53.
27. Rezaei A, Zabihollahi R, Salehi M, Moghim S, Tamizifar H,
Yazdanpanahi N, Amini G. Designing a non-virulent HIV-1
strain: potential implications for vaccine and experimental re-
search. J Res Med Sci. 2007;12(5):227–34.
28. Gunasekaran T, Nigusse T, Dhanaraju MD. Silver nanoparticles
as real topical bullets for wound healing. J Am Coll Clin Wound
Spec. 2011;3(4):82–96.
29. Cavrois M, Neidleman J, Yonemoto W, Fenard D, Greene WC.
HIV-1 virion fusion assay: uncoating not required and no effect
of Nef on fusion. Virology. 2004;328(1):36–44.
30. Svarovskaia ES, Barr R, Zhang X, Pais GC, Marchand C, Pommier
Y, Burke TR Jr, Pathak VK. Azido-containing diketo acid
derivatives inhibit human immunodeficiency virus type 1 integrase
in vivo and influence the frequency of deletions at two-long-ter-
minal-repeat-circle junctions. J Virol. 2004;78(7):3210–22.
31. Morgan JR, LeDoux JM, Snow RG, Tompkins RG, Yarmush ML.
Retrovirus infection: effect of time and target cell number.
J Virol. 1995;69(11):6994–7000.
32. Kim YS, Song MY, Park JD, Song KS, Ryu HR, Chung YH,
Chang HK, Lee JH, Oh KH, Kelman BJ, Hwang IK, Yu IJ.
Subchronic oral toxicity of silver nanoparticles. Particle Fibre
Toxicol. 2010;7(1):20.
33. Pana´cek A, Kola´r M, Vecerova´ R, Prucek R, Soukupova´ J, Krystof V,
Hamal P, Zboril R, Kvı´tek L. Antifungal activity of silver nanoparti-
cles against Candida spp. Biomaterials. 2009;30(31):6333–40.
34. Siliciano JD, Kajdas J, Finzi D, Quinn TC, Chadwick K, Mar-
golick JB, Kovacs C, Gange SJ, Siliciano RF. Long-term follow-
up studies confirm the stability of the latent reservoir for HIV-1 in
resting CD4? T cells. Nat Med. 2003;9(6):727–8.
35. Grant RM, Hecht FM, Warmerdam M, et al. Time trends in
primary HIV-1 drug resistance among recently infected persons.
JAMA. 2002;288(2):181–8.
36. Baba M. Recent status of HIV-1 gene expression inhibitors.
Antiviral Res. 2006;71(2):301–6.
37. Emerman M, Malim MH. HIV-1 regulatory/accessory genes:
keys to unraveling viral and host cell biology. Science.
1998;280(5371):1880–4.
38. Lau A, Swinbank KM, Ahmed PS, et al. Suppression of HIV-1
infection by a small molecule inhibitor of the ATM kinase. Nat
Cell Biol. 2005;7(5):493–500.
39. Galdiero S, Falanga A, Vitiello M, Cantisani M, Marra V, Gal-
diero M. Silver nanoparticles as potential antiviral agents.
Molecules. 2011;16(10):8894–918.
40. Elechiguerra JL, Burt JL, Morones JR, Camacho-Bragado A,
et al. Interaction of silver nanoparticles with HIV-1.
J Nanobiotechnol. 2005;3(6):1–10.
41. De Boer RJ, Boucher CA. Anti-CD4 therapy for AIDS suggested
by mathematical models. Proc R Soc Lond B.
1996;263(1372):899–905.
42. Lori F, Jessen H, Lieberman J, Clerici M, Tinelli C, Lisziewicz J.
Immune restoration by combination of a cytostatic drug (hy-
droxyurea) and anti-HIV drugs (didanosine and indinavir). AIDS
Res Hum Retroviruses. 1999;15(7):619–24.
43. Lori F, Jessen H, Foli A, Lisziewicz J, Matteo PS. Long-term
suppression of HIV-1 by hydroxyurea and didanosine. JAMA.
1997;277(18):1437–8.
44. Lori F, Jessen H, Lieberman J, et al. Treatment of human im-
munodeficiency virus infection with hydroxyurea, didanosine,
and a protease inhibitor before seroconversion is associated with
normalized immune parameters and limited viral reservoir. J In-
fect Dis. 1999;180(6):1827–32.
45. Georgiou NA, van der Bruggen T, Oudshoorn M, Nottet HS,
Marx JJ, van Asbeck BS. Inhibition of human immunodeficiency
virus type 1 replication in human mononuclear blood cells by the
iron chelators deferoxamine, deferiprone, and bleomycin. J Infect
Dis. 2000;181(2):484–90.
46. Alavidjeh MS, Haririan I, Khorramizadeh MR, Ghane ZZ,
Ardestani MS, Namazi H. Anionic linear-globular dendrimers:
biocompatible hybrid materials with potential uses in nanome-
dicine. J Mater Sci. 2010;21(4):1121–33.
47. Haririan I, Alavidjeh MS, Khorramizadeh MR, Ardestani MS,
Ghane ZZ, Namazi H. Anionic linear-globular dendrimer-cis-
platinum (II) conjugates promote cytotoxicity in vitro against
different cancer cell lines. Int J Nanomed. 2010;5:63–75.
179 Page 8 of 8 J Mater Sci: Mater Med (2015) 26:179
123

More Related Content

What's hot

Project Report Winter 2016
Project Report Winter 2016Project Report Winter 2016
Project Report Winter 2016
Theresa Rizk
 
Evaluation of infection course in mice induced by L. major in presence of pos...
Evaluation of infection course in mice induced by L. major in presence of pos...Evaluation of infection course in mice induced by L. major in presence of pos...
Evaluation of infection course in mice induced by L. major in presence of pos...
Nanomedicine Journal (NMJ)
 
Biomonitoring: Its Expanding Role in Public Health Evaluations and Litigation
Biomonitoring: Its Expanding Role in Public Health Evaluations and LitigationBiomonitoring: Its Expanding Role in Public Health Evaluations and Litigation
Biomonitoring: Its Expanding Role in Public Health Evaluations and Litigation
kurfirst
 
Pesticide Handbook
Pesticide HandbookPesticide Handbook
Pesticide Handbook
kk 555888
 
Synthesis and Evaluation of the Cytotoxic and Anti-Proliferative Properties o...
Synthesis and Evaluation of the Cytotoxic and Anti-Proliferative Properties o...Synthesis and Evaluation of the Cytotoxic and Anti-Proliferative Properties o...
Synthesis and Evaluation of the Cytotoxic and Anti-Proliferative Properties o...
IJEAB
 
Honours presentation (ed)
Honours presentation (ed)Honours presentation (ed)
Honours presentation (ed)
Manoka Marageni
 
Drv r and ral interactions fabbiani 2011
Drv r and ral interactions fabbiani 2011Drv r and ral interactions fabbiani 2011
Drv r and ral interactions fabbiani 2011
Heverton Zambrini
 

What's hot (17)

Project Report Winter 2016
Project Report Winter 2016Project Report Winter 2016
Project Report Winter 2016
 
Evaluation of infection course in mice induced by L. major in presence of pos...
Evaluation of infection course in mice induced by L. major in presence of pos...Evaluation of infection course in mice induced by L. major in presence of pos...
Evaluation of infection course in mice induced by L. major in presence of pos...
 
Nano medicine
Nano medicineNano medicine
Nano medicine
 
Biomonitoring: Its Expanding Role in Public Health Evaluations and Litigation
Biomonitoring: Its Expanding Role in Public Health Evaluations and LitigationBiomonitoring: Its Expanding Role in Public Health Evaluations and Litigation
Biomonitoring: Its Expanding Role in Public Health Evaluations and Litigation
 
Phenotyping and Genotyping Characteristics of Serratia Marcescens
Phenotyping and Genotyping Characteristics of Serratia MarcescensPhenotyping and Genotyping Characteristics of Serratia Marcescens
Phenotyping and Genotyping Characteristics of Serratia Marcescens
 
Pesticide poisoning cognitive brain and issues
Pesticide poisoning  cognitive brain and issuesPesticide poisoning  cognitive brain and issues
Pesticide poisoning cognitive brain and issues
 
Design, synthesis and anti-tumour activity of new pyrimidine-pyrrole appended...
Design, synthesis and anti-tumour activity of new pyrimidine-pyrrole appended...Design, synthesis and anti-tumour activity of new pyrimidine-pyrrole appended...
Design, synthesis and anti-tumour activity of new pyrimidine-pyrrole appended...
 
Pesticide Handbook
Pesticide HandbookPesticide Handbook
Pesticide Handbook
 
Open Journal of Chemistry
Open Journal of ChemistryOpen Journal of Chemistry
Open Journal of Chemistry
 
POLISH JOURNAL OF SCIENCE №40 (2021)
POLISH JOURNAL OF SCIENCE №40  (2021)POLISH JOURNAL OF SCIENCE №40  (2021)
POLISH JOURNAL OF SCIENCE №40 (2021)
 
Immunotoxicity
ImmunotoxicityImmunotoxicity
Immunotoxicity
 
Jurnal proceeding
Jurnal proceedingJurnal proceeding
Jurnal proceeding
 
Synthesis and Evaluation of the Cytotoxic and Anti-Proliferative Properties o...
Synthesis and Evaluation of the Cytotoxic and Anti-Proliferative Properties o...Synthesis and Evaluation of the Cytotoxic and Anti-Proliferative Properties o...
Synthesis and Evaluation of the Cytotoxic and Anti-Proliferative Properties o...
 
Example academic CV
Example academic CVExample academic CV
Example academic CV
 
Spectroscopic Characterization of Chloramphenicol and Tetracycline: An Impact...
Spectroscopic Characterization of Chloramphenicol and Tetracycline: An Impact...Spectroscopic Characterization of Chloramphenicol and Tetracycline: An Impact...
Spectroscopic Characterization of Chloramphenicol and Tetracycline: An Impact...
 
Honours presentation (ed)
Honours presentation (ed)Honours presentation (ed)
Honours presentation (ed)
 
Drv r and ral interactions fabbiani 2011
Drv r and ral interactions fabbiani 2011Drv r and ral interactions fabbiani 2011
Drv r and ral interactions fabbiani 2011
 

Viewers also liked

В.С.Крикоров-Единый Космос 2
В.С.Крикоров-Единый Космос 2В.С.Крикоров-Единый Космос 2
В.С.Крикоров-Единый Космос 2
Yury Podusov
 
В.С.Крикоров-Единый Космос 5
В.С.Крикоров-Единый Космос 5В.С.Крикоров-Единый Космос 5
В.С.Крикоров-Единый Космос 5
Yury Podusov
 

Viewers also liked (18)

6 Dr Nevin presentation Townsville Health Forum March 2016
6 Dr Nevin presentation Townsville Health Forum March 20166 Dr Nevin presentation Townsville Health Forum March 2016
6 Dr Nevin presentation Townsville Health Forum March 2016
 
Studiare i profili di rischio per costruire un business model vincente.
Studiare i profili di rischio per costruire un business model vincente.Studiare i profili di rischio per costruire un business model vincente.
Studiare i profili di rischio per costruire un business model vincente.
 
تقرير السجون مارس 2017
تقرير السجون مارس 2017  تقرير السجون مارس 2017
تقرير السجون مارس 2017
 
Step By Step Guide to Learn R
Step By Step Guide to Learn RStep By Step Guide to Learn R
Step By Step Guide to Learn R
 
JavaOne 2016: Code Generation with JavaCompiler for Fun, Speed and Business P...
JavaOne 2016: Code Generation with JavaCompiler for Fun, Speed and Business P...JavaOne 2016: Code Generation with JavaCompiler for Fun, Speed and Business P...
JavaOne 2016: Code Generation with JavaCompiler for Fun, Speed and Business P...
 
Light electric vehicles for urban freight Barcelona sept 2016
Light electric vehicles for urban freight Barcelona sept 2016Light electric vehicles for urban freight Barcelona sept 2016
Light electric vehicles for urban freight Barcelona sept 2016
 
NUMA World Summit #1 - The future of innovation in fast growing markets
NUMA World Summit #1 - The future of innovation in fast growing marketsNUMA World Summit #1 - The future of innovation in fast growing markets
NUMA World Summit #1 - The future of innovation in fast growing markets
 
Casos de uso de fuentes big data en las estadísticas económicas de Canarias
Casos de uso de fuentes big data en las estadísticas económicas de CanariasCasos de uso de fuentes big data en las estadísticas económicas de Canarias
Casos de uso de fuentes big data en las estadísticas económicas de Canarias
 
How Cloud Enhances Agile Software Development
How Cloud Enhances Agile Software DevelopmentHow Cloud Enhances Agile Software Development
How Cloud Enhances Agile Software Development
 
Inbound-myynti
Inbound-myynti Inbound-myynti
Inbound-myynti
 
e-Book Web Analytics Uma Visão Brasileira - 2a. edição
e-Book Web Analytics Uma Visão Brasileira - 2a. ediçãoe-Book Web Analytics Uma Visão Brasileira - 2a. edição
e-Book Web Analytics Uma Visão Brasileira - 2a. edição
 
How to Hook Customers with Habit-forming Products
How to Hook Customers with Habit-forming ProductsHow to Hook Customers with Habit-forming Products
How to Hook Customers with Habit-forming Products
 
31 Best Growth Hacking Resources
31 Best Growth Hacking Resources31 Best Growth Hacking Resources
31 Best Growth Hacking Resources
 
Future Social: 10 Key Trends in Social Media
Future Social: 10 Key Trends in Social MediaFuture Social: 10 Key Trends in Social Media
Future Social: 10 Key Trends in Social Media
 
Equivalence
Equivalence Equivalence
Equivalence
 
В.С.Крикоров-Единый Космос 2
В.С.Крикоров-Единый Космос 2В.С.Крикоров-Единый Космос 2
В.С.Крикоров-Единый Космос 2
 
В.С.Крикоров-Единый Космос 5
В.С.Крикоров-Единый Космос 5В.С.Крикоров-Единый Космос 5
В.С.Крикоров-Единый Космос 5
 
Finanças dos clubes brasileiros em 2015- Amir Somoggi
Finanças dos clubes brasileiros em 2015- Amir SomoggiFinanças dos clubes brasileiros em 2015- Amir Somoggi
Finanças dos clubes brasileiros em 2015- Amir Somoggi
 

Similar to Nanosilver based anionic linear globular dendrimer with a special significant antiretroviral activity

Antimicrobial Susceptibility, Biochemical Characterization and Molecular Typi...
Antimicrobial Susceptibility, Biochemical Characterization and Molecular Typi...Antimicrobial Susceptibility, Biochemical Characterization and Molecular Typi...
Antimicrobial Susceptibility, Biochemical Characterization and Molecular Typi...
wilhelm mendel
 
Antimicrobial Susceptibility, Biochemical Characterization and Molecular Typi...
Antimicrobial Susceptibility, Biochemical Characterization and Molecular Typi...Antimicrobial Susceptibility, Biochemical Characterization and Molecular Typi...
Antimicrobial Susceptibility, Biochemical Characterization and Molecular Typi...
rachelsalk
 
An Evaluation of Biofield Treatment on Susceptibility Pattern of Multidrug Re...
An Evaluation of Biofield Treatment on Susceptibility Pattern of Multidrug Re...An Evaluation of Biofield Treatment on Susceptibility Pattern of Multidrug Re...
An Evaluation of Biofield Treatment on Susceptibility Pattern of Multidrug Re...
Mahendra Kumar Trivedi
 
Advancements in Nucleic Acid Based Therapeutics against Respiratory Viral Inf...
Advancements in Nucleic Acid Based Therapeutics against Respiratory Viral Inf...Advancements in Nucleic Acid Based Therapeutics against Respiratory Viral Inf...
Advancements in Nucleic Acid Based Therapeutics against Respiratory Viral Inf...
Dr Binod Kumar
 
Green synthesis of silver nanoparticles: The reasons for and against Aspergil...
Green synthesis of silver nanoparticles: The reasons for and against Aspergil...Green synthesis of silver nanoparticles: The reasons for and against Aspergil...
Green synthesis of silver nanoparticles: The reasons for and against Aspergil...
Nanomedicine Journal (NMJ)
 
Green synthesis of silver nanoparticles: The reasons for and against Aspergil...
Green synthesis of silver nanoparticles: The reasons for and against Aspergil...Green synthesis of silver nanoparticles: The reasons for and against Aspergil...
Green synthesis of silver nanoparticles: The reasons for and against Aspergil...
Nanomedicine Journal (NMJ)
 

Similar to Nanosilver based anionic linear globular dendrimer with a special significant antiretroviral activity (20)

Antimicrobial Susceptibility, Biochemical Characterization and Molecular Typi...
Antimicrobial Susceptibility, Biochemical Characterization and Molecular Typi...Antimicrobial Susceptibility, Biochemical Characterization and Molecular Typi...
Antimicrobial Susceptibility, Biochemical Characterization and Molecular Typi...
 
Antimicrobial Susceptibility, Biochemical Characterization and Molecular Typi...
Antimicrobial Susceptibility, Biochemical Characterization and Molecular Typi...Antimicrobial Susceptibility, Biochemical Characterization and Molecular Typi...
Antimicrobial Susceptibility, Biochemical Characterization and Molecular Typi...
 
Short-term improvement of clinical parameters and microbial diversity in peri...
Short-term improvement of clinical parameters and microbial diversity in peri...Short-term improvement of clinical parameters and microbial diversity in peri...
Short-term improvement of clinical parameters and microbial diversity in peri...
 
An Impact of Biofield Treatment: Antimycobacterial Susceptibility Potential U...
An Impact of Biofield Treatment: Antimycobacterial Susceptibility Potential U...An Impact of Biofield Treatment: Antimycobacterial Susceptibility Potential U...
An Impact of Biofield Treatment: Antimycobacterial Susceptibility Potential U...
 
Nanotechnology in Targeted Drug Delivery.pdf
Nanotechnology in Targeted Drug Delivery.pdfNanotechnology in Targeted Drug Delivery.pdf
Nanotechnology in Targeted Drug Delivery.pdf
 
1641217636Pharmacogenomics__1_.pdf
1641217636Pharmacogenomics__1_.pdf1641217636Pharmacogenomics__1_.pdf
1641217636Pharmacogenomics__1_.pdf
 
Dendrimers for Target Drug Delivery In Treatment of Cancer
Dendrimers for Target Drug Delivery In Treatment of CancerDendrimers for Target Drug Delivery In Treatment of Cancer
Dendrimers for Target Drug Delivery In Treatment of Cancer
 
Oral cancer and Gene therapy
Oral cancer and Gene therapyOral cancer and Gene therapy
Oral cancer and Gene therapy
 
An Evaluation of Biofield Treatment on Susceptibility Pattern of Multidrug Re...
An Evaluation of Biofield Treatment on Susceptibility Pattern of Multidrug Re...An Evaluation of Biofield Treatment on Susceptibility Pattern of Multidrug Re...
An Evaluation of Biofield Treatment on Susceptibility Pattern of Multidrug Re...
 
An Evaluation of Biofield Treatment on Susceptibility Pattern of Multidrug Re...
An Evaluation of Biofield Treatment on Susceptibility Pattern of Multidrug Re...An Evaluation of Biofield Treatment on Susceptibility Pattern of Multidrug Re...
An Evaluation of Biofield Treatment on Susceptibility Pattern of Multidrug Re...
 
An Evaluation of Biofield Treatment on Susceptibility Pattern of Multidrug Re...
An Evaluation of Biofield Treatment on Susceptibility Pattern of Multidrug Re...An Evaluation of Biofield Treatment on Susceptibility Pattern of Multidrug Re...
An Evaluation of Biofield Treatment on Susceptibility Pattern of Multidrug Re...
 
Advancements in Nucleic Acid Based Therapeutics against Respiratory Viral Inf...
Advancements in Nucleic Acid Based Therapeutics against Respiratory Viral Inf...Advancements in Nucleic Acid Based Therapeutics against Respiratory Viral Inf...
Advancements in Nucleic Acid Based Therapeutics against Respiratory Viral Inf...
 
Non-biological gene carriers designed for overcoming the major extra- and int...
Non-biological gene carriers designed for overcoming the major extra- and int...Non-biological gene carriers designed for overcoming the major extra- and int...
Non-biological gene carriers designed for overcoming the major extra- and int...
 
Cancer therapy
Cancer therapyCancer therapy
Cancer therapy
 
Evaluation of cutaneous adverse drug reactions due to antimicrobial agents: A...
Evaluation of cutaneous adverse drug reactions due to antimicrobial agents: A...Evaluation of cutaneous adverse drug reactions due to antimicrobial agents: A...
Evaluation of cutaneous adverse drug reactions due to antimicrobial agents: A...
 
New coronavirus inhibitor exhibits antiviral activity
New coronavirus inhibitor exhibits antiviral activity New coronavirus inhibitor exhibits antiviral activity
New coronavirus inhibitor exhibits antiviral activity
 
Green synthesis of silver nanoparticles: The reasons for and against Aspergil...
Green synthesis of silver nanoparticles: The reasons for and against Aspergil...Green synthesis of silver nanoparticles: The reasons for and against Aspergil...
Green synthesis of silver nanoparticles: The reasons for and against Aspergil...
 
Green synthesis of silver nanoparticles: The reasons for and against Aspergil...
Green synthesis of silver nanoparticles: The reasons for and against Aspergil...Green synthesis of silver nanoparticles: The reasons for and against Aspergil...
Green synthesis of silver nanoparticles: The reasons for and against Aspergil...
 
Physical, Thermal and Spectroscopic Characterization of m-Toluic Acid: an Imp...
Physical, Thermal and Spectroscopic Characterization of m-Toluic Acid: an Imp...Physical, Thermal and Spectroscopic Characterization of m-Toluic Acid: an Imp...
Physical, Thermal and Spectroscopic Characterization of m-Toluic Acid: an Imp...
 
Biofield Treatment: An Alternative Approach to Combat Multidrug-Resistant Sus...
Biofield Treatment: An Alternative Approach to Combat Multidrug-Resistant Sus...Biofield Treatment: An Alternative Approach to Combat Multidrug-Resistant Sus...
Biofield Treatment: An Alternative Approach to Combat Multidrug-Resistant Sus...
 

Nanosilver based anionic linear globular dendrimer with a special significant antiretroviral activity

  • 1. BIOCOMPATIBILITY STUDIES Original Research Nanosilver based anionic linear globular dendrimer with a special significant antiretroviral activity Mehdi Shafiee Ardestani1 • Alireza Salehi Fordoei2,3 • Asghar Abdoli2 • Reza Ahangari Cohan4 • Golnaz Bahramali2 • Seyed Mehdi Sadat2 • Seyed Davar Siadat5 • Hamid Moloudian6 • Nasser Nassiri Koopaei7 • Azam Bolhasani2 • Pooneh Rahimi2 • Soheila Hekmat2 • Mehdi Davari2 • Mohammad Reza Aghasadeghi2 Received: 10 January 2015 / Accepted: 30 March 2015 Ó Springer Science+Business Media New York 2015 Abstract HIV is commonly caused to a very complicated disease which has not any recognized vaccine, so designing and development of novel antiretroviral agents with specific application of nanomedicine is a globally inter- ested research subject worldwide. In the current study, a novel structure of silver complexes with anionic linear globular dendrimer was synthesized, characterized and then assessed against HIV replication pathway in vitro as well. The results showed a very good yield of synthesis (up to 70 %) for the nano-complex as well as a very potent significant (P 0.05) antiretroviral activity with non-sev- ere toxic effects in comparison with the Nevirapine as standard drug in positive control group. According to the present data, silver anionic linear globular dendrimers complex may have a promising future to inhibit replication of HIV viruse in clinical practice. Abbreviations HIV Human immunodeficiency virus RSV Respiratory syncytial virus PEG-600 Polyethylene glycole 600 Da DCC Dicyclohexyl carbodiimide DLS Dynamic light scattering DMSO Dimethyl sulfoxide FTIR Fourier transforms spectroscopy XTT 2,3-Bis(2-methoxy-4-nitro-5-sulfophenyl)-2H- tetrazolium-5-carboxanilide MPPI Mannosylated poly (propyleneimine) dendrimer 1 Introduction Globally, more than 40 million people suffer from the human immunodeficiency virus (HIV) infection. The high activity antiretroviral therapy (HAART) combining three or more of antiretroviral (ARV) drugs administered for lifelong renders acquired immunodeficiency syndrome (AIDS) as a controlled chronic disease [1]. Limitations of the current ARV include long lasting medications with remarkable adverse effects, poor patient-medication ad- herence, multi-drug resistance (MDR) due to mutations, drug–drug interactions, poor drug pharmacokinetics, viral load rise very quickly following medication discontinued and affordability as well as establishing a latent infection [2]. Unique properties of dendrimers as novel and exciting & Mohammad Reza Aghasadeghi mr_sadeqi@yahoo.com; mrasadeghi@pasteur.ac.ir 1 Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran 2 Department of Hepatitis and AIDS, Pasteur Institute of Iran, 13164 Tehran, Iran 3 Department of Biology, College of Basic Sciences, Tehran Science and Research Branch, Islamic Azad University, Tehran, Iran 4 Virology Research Group, Department of Rabies, Pasteur Institute of Iran, Tehran, Iran 5 Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran 6 Faculty of Pharmacy, Pharmaceutical Quality Assurance Research Center, Tehran University of Medical Sciences, Tehran, Iran 7 Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran 123 J Mater Sci: Mater Med (2015) 26:179 DOI 10.1007/s10856-015-5510-7
  • 2. polymers such as uniform size, water solubility, available internal cavities, multivalency, well-defined molecular weight, among others, make them attractive for drug de- livery purposes since past decades [3–5]. These features are caused by highly branched, spherical and nanometer di- mension of dendrimers [4]. Furthermore, attempts have been made to apply dendrimers to other areas such as immunology and development of vaccines, antimicrobials and antivirals [3]. Biocompatibility and release of drugs from dendrimers are controversial issues [3]. Selection of polymers has always been affected by issues such as biodegradability, biocompatibility, bioadhesiveness while respecting physical and biochemical properties [5]. Spheric shape of dendrimers makes their structures much more accurately controllable. They are indeed nano-sized mole- cules with sizes between 10 and 100 nm [5]. Dendrimers due to aforementioned characteristics have emerged as an important class of antiretroviral carriers [6]. Dendrimers as organic nano-platforms can act in therapeutic and diagnostic applications [7]. To deliver a therapeutic and non-toxic dose of drug to diseases, nano and micro-carriers are efficient tools [8]. Increased effi- ciency and decreased toxicity of anti-retroviral drug loaded dendrimers were investigated in previous studies. Dutta and Jain studied targeting and anti-HIV potential of lami- vudine loaded mannosylated poly (propyleneimine) (MPPI) dendrimer and concluded that the drug release profile and cellular uptake of drug were improved sig- nificantly. MPPI is a surface modified version of poly (propyleneimine) (PPI) [9]. Passive targeting of drug loaded nano-carriers is limited to tumors, the reticular en- dothelial system and lymph nodes. Attractive features of dendrimers like narrow molecular weight distribution, small size and easy incorporation of targeting ligand ren- ders them remarkable applicability in antiretroviral therapy [10]. Dendrimers are also advantageous with respect to their drug interactions [11]. Dendrimers could be widely used in targeted and controlled release drug delivery sys- tems; however, toxicity problems still exist and need some modifications. Release profile of dendrimers is easily modifiable and controllable [12]. Citric acid–polyethylene glycol–citric acid (CPEGC) dendrimers were mentioned as biocompatible compounds and investigated as drug deliv- ery systems. Some small and hydrophobic molecules and drugs were incorporated into the above dendrimer. These dendrimers had good water solubility, biocompatibility and low toxicity [13]. Synthesis of dendrimers has recently been facilitated by the divergent ‘lego’ and the convergent ‘click’ approaches. Drugs could be physically loaded within the dendrimer or chemically attached to the surface. Ester-terminated dendrimers are more bioavailable than amino-terminated ones. Increase of dendrimer sizes makes them more soluble. Nonetheless, cytotoxic effects of dendrimers increase with their generation number. Cyto- toxicity could be reduced by dendrimer functionalization (particularly with PEG). Dendrimers can inherently act against tumors, bacteria and viruses [14]. AgNPs have been used for their antibiotic effects, noteworthy that no cross-resistance with other antibiotics is evident and efficacy against multi drug resistant strains [15, 16]. AgNPs show wide spectrum against different bacteria and fungi [17–22] commercially used in health and im- plantable biomaterial, general utilities and home appli- ances. Moreover, antiviral effects of AgNPs have attracted attention recently. AgNPs present remarkable antiviral activity against HIV, hepatitis B, HSV, RSV, monkeypox virus, tacaribe virus (TCRV), RNA viruses and adenovirus type 3 [21, 23–26]. The supporting mechanism for AgNPs antiretroviral effect is through glycoprotein 120 (gp120) binding and prevention following the penetration and in- terfering with intracellular stages. With the characteristics mentioned, AgNPs are applied in a vaginal product to prevent sexually transmitted diseases like HIV. However, higher efficacy has been proposed with the concomitant use of neutralizing antibodies [23]. Wide anti-infectious properties of AgNPs as well as wound healing effects makes silver suitable candidate for topical formulations [20, 22, 27, 28]. Limitations of current antiretroviral drugs such as ex- tensive first pass metabolism, astrointestinal degradation, poor solubility, etc. lead investigation of novel drug de- livery systems (NDDS) such as sustained and targeted drug delivery systems. So, in the present study, a novel structure of silver complexes with anionic linear globular dendrimer was synthesized, characterized and then assessed against HIV replication pathway in vitro. 2 Materials and methods 2.1 Materials PEG 600 was purchased (from Merck,Germany). Dicy- clohexylcarbodiimide, dimethylsulfoxide, citric acid and silver were obtained (from Merck,Germany). Besides pSPAX.2, pmzNL4-3 and pMD2.G plasmids were used for transfection. In cellular studies, HEK293T cell line and DMEM primary culture medium were used. HEK is an abbreviation for human embryonic kidney cells. This cell line was chosen for this research because it has superior survival and proliferation compared to adult cells. 2.2 Synthesis of dendrimers G1 and G2 Synthesis of compound G1 of dendrimer was initiated through the reaction of PEG 600 (as the core of dendrimer) 179 Page 2 of 8 J Mater Sci: Mater Med (2015) 26:179 123
  • 3. and dicyclohexylcarbodiimide (DCC) in anhydrous DMSO in the condition of darkness and vacuum. DCC activated PEG by substitution with hydrogen atom of carboxylic acid groups. Then, DCC was replaced with citric acid, and so color of the tick solution changed to orange with stench. Reaction was terminated with adding water and after fil- tration; yellow-brown solution of dendrimer was obtained. Primarily, synthesis of compound G1 was confirmed by thin layer chromatography (TLC); stationary phase: silica gel, mobile phase: 65 % CCl4/35 % MeOH. Then, the product of G1 was separated and purified by dialysis. Volume and concentration of dialysis buffer was 1600 fold and half of the contents of dialysis bag, respectively. Pore size of dialysis bag was 500–1000 Da. PEG was used as limiting substrate of reaction and didn’t leave any residue. Other substrates were excess. Mass of dendrimer was transferred to several vials and lyophilized in yield 91 % after freezing in liquid nitrogen. Procedures were severally repeated to obtain the optimum procedure. To synthesize dendrimer G2, DCC and citric acid were added to solution of dendrimer G1 in DMSO and the solution became ticker and thereafter dark cream color was appeared. In this stage, compound G1, was rate limiting of reaction. As synthesis of dendrimer G1, reaction was terminated with water addition, solution was filtrated, dialyzed, and powder was lyophi- lized. Structure of dendrimer G2 (in yield 73 %) was confirmed by FT-IR. Size and electric charge of G1 and G2 dendrimers was determined by zetasizer (dynamic light scattering, DLS). Briefly, 2 mg of plasmid mixture was dissolved in 150 ll of deficient culture medium. Then, 40 ll of lipo- fectin was added to the tube containing DNA and incubated in room temperature for 10 min. Following that, 3 mL of cell medium. 2.3 Preparation of Ag-dendrimer conjugates Tendency of nano-silver particles to the oxygen atoms of dendrimer leads to the reaction and incorporation of silver into the dendrimers. In the isolated and light protected condition, the mixture of dendrimer and silver was stirred for 24 h. Yellow-brown color of mixture changed to gray. Then, the mixture was filtered and brown solution of Ag- dendrimer conjugated was separated from insoluble free silver. Final product (liquid or lyophilized) was perma- nently preserved from light and air, during the analyses. Structure of product was assessed by FT-IR and compared with G1 and G2 dendrimers. Size and charge of the final product was determined and compared as well. Imaging of Ag-dendrimer conjugated was made by AFM. Concentra- tion of conjugated silver was determined by ICP-Mass technique. Maximum incorporation of silver into the den- drimer was calculated by ICP-Mass and divided by total mass of G2 dendrimer and multiplied by 100; therefore percentage of loading complex was obtained (52 %). 2.4 Cell culture and antiviral assessments 2.4.1 Cells, transfection and virus production To produce HIV-1 SCR virions pseudotyped by VSVG, Qiagen sample was used. First HEK293T cells were cul- tured and maintained in Dulbecco’s modified Eagle’s cul- ture medium (DMEM, Sigma, USA) supplemented with FBS (15 %), penicillin (100 U/ml), streptomycin (100 lg/ ml), and glutamine (2 mmol/l); incubated in 5 % CO2, 37 °C inside a 6-well plate until they reach a total number of approximately 1.2 9 106 . Transfection was carried out using standard Lipofectamine 2000 according to Qiagen protocol. Plasmids of pSPAX.2, pmzNL4-3 and pMD2.G were simultaneously transfected into HEK293T cells to produce proviruses. Next, cell culture plates were incu- bated for minimum time of 24 h in 37 °C and 5 % CO2. HEPES (4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid) (25 mM) was used to adjust the pH of the cell culture medium in transfection stage. To prepare virus stock, cul- ture supernatants of the transfected cells were harvested at 24, 48, and 72 h post transfection, and after filtration were concentrated for 2 h in 60,0009g in 4 °C by ultracen- trifugation [29]. The pelleted virions were solved in 1/20 volume of RPMI 1640 by mixing gently overnight at 4 °C to prevent virus aggregation, quantified by P24 ELISA assay kit (Biorad) and stored at -70 °C until use [30]. HEK cells were used to determine P24 titer of SCR HIV-1 virions pseudotyped by stomatitis virus surface VSVG vesicular glycoprotein. A 60 9 103 count of HEK cells were inoculated in each well of a 24 well plate containing 250 ll of culture medium and then 600 ng of P24 was added to each well. Next, the cells were washed every 5 h using 10 % FBS. Every time, the supernatants were col- lected and replaced with new culture medium. Finally, P24 concentration in the supernatant was determined according to the ELISA capture protocol (Biomerieux) developed by the manufacturing company. All the components were dissolved in dimethylsulfoxide (DMSO) with concentra- tions ranging from 10 lM to 10 mM. Nevirapine (NVP) was dissolved in DMSO with 50 lM concentration, ex- tracted from commercial preparations was used as positive control to inhibit viral growth and DMSO with final con- centration of 1 % as the negative control. All the ex- periments were triplicate. Toxicity of compounds was separately estimated on the HEK cells. CC50 (cytotoxicity capacity 50 %) was calculated. Dividing CC50 by IC50 provided TI (therapeutic index) and/or SI (selectivity in- dex) (TI or SI = CC50/IC50). Antiviral effects of different compounds were compared through TI or SI indices. J Mater Sci: Mater Med (2015) 26:179 Page 3 of 8 179 123
  • 4. 2.5 Evaluation of the new nano-drug effectiveness on HIV replication A HEK cells count of 60 9 103 in each well of a 24 well plate containing 250 ll of culture medium was prepared. SCR HIV virions and 600 ng P24 were added to each well. The compounds were added in four concentrations (20, 40, 80 and 120 lM) to the wells as triplicate. After 5 h, the cells were washed twice with DMEM/FBS 10 % [31]. Again, the compounds and fresh medium with the same amounts were added. At 72 h after inoculation, the supernatant was collected. The P24 level of the su- pernatant was determined with ELISA capture (Biorad). Related ELISA test was performed and finally, the con- centration was calculated. To examine cellular prolif- eration, XTT method (Roche) was used [30]. Medium of phenol red was used to diminish test errors in proliferation assay. 3 Results 3.1 Time relation of the SCR viral replication Replication of the SCR HIV-1 virions psuedotyped by VSVG on the HEK cells was determined after viral inoculation. P24 secretion was started at the first day and increased to a maximum level at 48 h after SCR virions inoculation. Then, P24 level was gradually decreased from the fourth day after inoculation. As the results denote, maximum cumulative amount of P24 in the supernatant was 72 h after inoculation. Therefore, P24 determination in the supernatant was used to assess the viral replication at 72 h after inoculation (Fig. 1). 3.2 SCR HIV-1 replication regarding the primary MOI HEK cells were infected by different multiplicity of in- fection (MOI) of pseudotype SCR virions. The supernatant of the infected cells was assessed through P24 level 3 days after inoculation. An inverse linear relationship between MOI and P24 production makes this test a precise method to investigate replication. Based on the assessments, the adequate viral count for replication start is 600 ng P24 (Fig. 2). 3.3 Nevirapine efficacy as the control positive Based on the results, suitable viral count for replication (600 ng P24) and time (72 h) for P24 determination was selected. To assess nevirapine function that is a NRTI drug, SCR virions replication in the presence of its different concentrations was measured. The results showed that in- creasing nevirapine level decreased P24 production and 50,000 nM concentration inhibited P24 production. These results revealed the correct function of the drug in the primary test conditions and replication assessment based on SCR HIV-1 suitably shows drug function in different concentrations (Fig. 3). 3.4 Inhibitory effects of silver dendrimer nanoconjugate The ability of Iron chelating compounds to inhibit HIV virus replication was investigated by assessing HIV repli- cation in the presence of various compounds and calculated based on the light absorption in P24 ELISA OD. Nanoconjugated silver dendrimer effects in 20, 40, 80 and 120 lM concentrations on HEK cells was assessed. The best antiviral effects were observed in 120 lM that inhib- ited viral contamination up to 87 %. Generally speaking, higher levels of drug produced higher level of viral Fig. 1 The effects of nanosilver on the P24 levels at different time exposures Fig. 2 The effects of nanosilver on the different amounts of starter virus 179 Page 4 of 8 J Mater Sci: Mater Med (2015) 26:179 123
  • 5. inhibitory effects. This was also evident for control (silver nitrate) (see more detailed data at Table 1, Fig. 4). 3.5 Cytotoxicity of nanoconjugated silver dendrimers in different concentrations Based on ODs from ELISA proliferation test, cytotoxicity was assessed through XTT method after 72 h. Although, the compounds showed potent antiviral effects but they had cytotoxicity as well. Cell survival in concentrations of 40, 80 and 120 lM was 23, 36 and 48 %, respectively and was 50 % in 80 and 120 lM concentration of silver nitrate as the control. DMSO was used as the positive control. In- hibitory effect was low at 20 lM concentration while low cytotoxicity was observed as well (see more detailed data at Table 2, Fig. 5). 3.6 TI indexing According to the calculated IC50 and CC50, TI index was determined for different compounds. Comparing this index for various concentrations of nanoconjugated silver den- drimers provides a method to identify concentrations with higher antiretroviral effects and lower cytotoxicity. Therapeutic index (TI) in 20 lM concentration was 5.3, the best among all concentrations. TI for 40, 80 and 120 lM concentrations was 0.96, 0.54 and 0.26, respectively. Dendrimeric seizes for G1 and G2 products were 70 and 90 nm, respectively; dendrimeric charges were -3.5 and -11.8 millivolt/Ampere (mv A-1 ), respectively. More Fig. 3 The effects of Nevirapine on the P24 at different time exposures Table 1 Depiction of some details on HIV inhibitory effects of nanosilver and the control drug P24 OD Mean P24 OD HIV inhibitory % Uninfected cells 0.04, 0.03 0.035 – Virus control (without drug) 0.8, 0.9, 0.7 0.8 0 Nevirapine (anti-retroviral drug) 0.02, 0.05 0.03 100 Ag-dendrimer (120 lM) 0.08, 0.2 0.14 87 Ag-dendrimer (80 lM) 0.21, 0.32 0.26 67 Ag-dendrimer (40 lM) 0.42, 0.34 0.38 52 Ag-dendrimer (20 lM) 0.48, 0.88 0.68 15 AgNO3 (control) (120 lM) 0.04, 0.1 0.07 96 AgNO3 (control) (80 lM) 0.32, 0.42 0.37 53 AgNO3 (control) (40 lM) 0.55, 0.4 0.47 41 AgNO3 (control) (20 lM) 0.62, 0.92 0.77 4 Fig. 4 Antiretroviral effects of AgNO3 and silver based dendrimer nanoparticle J Mater Sci: Mater Med (2015) 26:179 Page 5 of 8 179 123
  • 6. citric acid moieties at the surface of compound G2 leads to its larger size and more negative charge compared with compound G1. Atomic force microscopy (AFM) imaging of dendrimer G2 was performed and compared with G1. The important absorption bonds in FTIR spectrum for G1 are: 3000–3500 cm-1 for hydroxyl group of citric acid attached to PEG, 2800–2900 cm-1 for aliphatic carbon chain of citric acid and PEG, and shifted peak of carbonyl group of citric acid from 1700 to 1600 cm-1 that confirms low concentration of citric acid in the structure of den- drimer and conjugation of citric acid and PEG. The ab- sorbed bonds in FTIR spectrum [OH group of citric acid attached to PEG (broad, 3500–3000 cm-1 ), C=O (1600 cm-1 )] for G2 are: wide and strong peak from 3000 to 3500 cm-1 for hydroxyl group of citric acid, strength- ened peak from 2800 to 2900 cm-1 for aliphatic carbon of citric acid and PEG and a strong peak about 1700 cm-1 is related to strengthened steric bond of citric acid addition [OH group of citric acid (broad and strong, 3500–3000 cm-1 ), steric bond of citric acid (1700 cm-1 )]. Last mentioned peak implies high concentration of citric acid in the structure of compound G2 and conjugation of citric acid and PEG. Considering significant increase in aliphatic and hydroxyl peaks of compound G2 compared with G1; thus it can be concluded that IR spectra approved the synthesis of both dendrimer generations. 4 Discussion HIV transcripts are RNA to DNA by RT and introduce provirus into the host cell DNA by integrase. CD4? T cells are the major hosts and with proceeding decrease of their reservoir, opportunistic infections and cancers lead death. HIV/AIDS has imposed heavy economic and health costs to world health. Because of loss of vaccine, antiviral drugs have been the best confronting approach. Kim et al. ex- amined oral toxicity of AgNPs for 3 months finding the target organ to be the liver with biliary tract hyperplasia, possible necrosis, fibrosis and pigmentation. Finally, the LOAEL and NOAEL were determined to be 125 and 30 mg/kg, respectively, while AgNPs could accumulate in tissues investigated [32]. AgNPs exert bactericidal activity through concentrations that no remarkable acute human toxicity would emerge [22, 33]. But toxicity of available antiviral drugs and resistance of virus increased the im- portance of development of new antiretroviral drugs [34, 35]. Inhibition of virus replication via interaction with cellular pathways is the current strategy [36–38]. Combination of nanotechnology and antimicrobial prop- erties of Cr, Ag, Ti, and Zn presents effective anti-HIV ap- proaches. Some studies have shown antiviral activities of nano-silver (against HIV-1, HBV, HSV, RSV, and monkey- pox virus) [39]. Some other studies showed nano-silver par- ticles inhibit HIV fusion to the host cell via interaction with gp 120 [23, 40]. Furthermore nano-silver particles can interact with HIV through other ways such as inhibition of reverse transcriptase, protein activity and inhibition of host cell cycle. Dendrimers can trap different molecules within their branches and protect them from environmental influences and controllably release in targeted positions. Also den- drimers increase solubility of drugs by trapping them. Table 2 Illustration of some details on cell viability effects of nanosilver and the control drug ELISA OD (450 nm) Mean OD Cell viability % Un-treated (1 ll DMSO) cells 0.65, 0.61, 0.55 0.6 100 Ag-dendrimer (120 lg) 0.14, 0.14 0.14 23 Ag-dendrimer (80 lg) 0.22, 0.22 0.22 36 Ag-dendrimer (40 lg) 0.31, 0.28 0.29 48 Ag-dendrimer (20 lg) 0.49, 0.51 0.5 83 AgNO3 (control) (120 lg) 0.31, 0.29 0.3 50 AgNO3 (control) (80 lg) 0.32, 0.28 0.3 50 AgNO3 (control) (40 lg) 0.43, 0.45 0.44 73 AgNO3 (control) (20 lg) 0.53, 0.43 0.48 80 Fig. 5 Effects of different concentrations of Ag-dendrimer and AgNO3 exposures on cell viability percentage results of 179 Page 6 of 8 J Mater Sci: Mater Med (2015) 26:179 123
  • 7. Dendrimers can carry molecules on their surface via in- teraction with functional groups; a property that enables specific cell or tissue targeting. In this study we investigated several concentrations of Ag-dendrimer nano-conjugated compounds against repli- cation of HIV and determined cytotoxicity of the new compound in different concentrations. Researches showed the Ag-dendrimer conjugated reduces expression of P24 antigen on the surface of monocytes and lymphocytes. It seems virus inhibition is due to the reduction of cellular proliferation. Inhibition of cellular proliferation can lead inhibition of P24 production in lymphocytes. As a result, it seems Ag-dendrimer conjugated don’t target virus directly and indeed attack host cell of replicating virus. But some other drugs such as nevirapine inhibit replication, directly without any influence on cellular proliferation. HIV repli- cation rate and survival was assessed after application of several concentrations (20, 40, 80, and 120 lM) of new compound. Inhibition of cellular proliferation cause to in- hibit replication of virus but clinical researches have shown nano-silver particles decrease P24 level with 50 % without any effect on cellular proliferation. In a mathematical model for anti CD4? therapy, it has been showed that 25 % decrease in CD4? cell counts can reduce function of virus and rate of mutations [41]. With this respect current compound by killing proliferating CD4? T cells and de- pleting host cell from dATP, inhibit P24 production and HIV-1 replication [42–45]. Our previous studies have demonstrated that biocompatible range of Anionic linear- globular dendrimer up to the concentration of 0.5 mg/ml and less of the Anionic linear-globular dendrimer-cis- platinum(II) conjugates would be required to obtain a similar therapeutic effect against different cancer cell lines when compared with cisplatin treatment alone [46, 47]. Decreasing dose of drug usage by dendrimers may result in less drug resistance and drug side effects. Results of this study showed that all concentrations of Ag-dendrimer nano-conjugated could inhibit HIV effec- tively. Inhibition percent of HIV for concentrations 120, 80, 40, and 20 lM is 78, 67, 52, and 15, respectively. First three concentrations have highest inhibition rates but show high level of cytotoxicity as well (77, 64, and 52 %, re- spectively). Although inhibition rate of concentration 20 lM was only 15 %; but its cytotoxicity was 17 % and this concentration of new compound is totally more ap- propriate. AgNO3 was used as control and concentration of 120 lM showed better inhibition and lower toxicity (96 and 50 %, respectively) than conjugated compound but not appropriate for clinical application. At the concentration of 80 lM, the new compound is better than AgNO3 (53 % inhibition), but its cytotoxicity is high (50 %). In this concentration both compounds had remarkable cyto- toxicity, too. At the concentration of 40 lM, nanoconjugated compound had a stronger inhibition and lower cell survival profile (48 % in front of 73 %) than AgNO3. In this concentration none of compounds couldn’t be selected as an antiretroviral drug. At the concentration of 20 lM, AgNO3 has a very low inhibition rate (4 %) and a considerable cytotoxicity (20 %), while Ag-dendrimer at concentration of 20 % had a stronger inhibition and lower cytotoxicity profile. 5 Conclusion Finally, we conclude that the concentration of 20 lM of nanoconjugated compound has the best therapeutic profile (stronger inhibition and lower toxicity). Silver saturated dendrimers were used in this study and highly toxic com- pounds were obtained. Lower silver loaded dendrimers or other generations of dendrimer such as G1 can lead to more applicable and different results. Also we propose that the combination of concentration of 20 lM dendrimer with other antiretroviral drugs to be investigated for better therapeutic profile. Acknowledgments We thank all members of the Hepatitis and AIDS Department and Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences of Pasteur Institute of Iran and for their advice and assistance with the experiments. This work was supported by Grant No. 519 from Pasteur Institute of Iran, Tehran, Iran. Conflict of interest The authors disclose no potential conflicts of interest. References 1. Sosnik A, Chiappetta DA, Carcaboso AM. Drug delivery systems in HIV pharmacotherapy: what has been done and the challenges standing ahead. J Control Release. 2009;138(1):2–15. 2. Gupta U, Jain NK. Non-polymeric nano-carriers in HIV/AIDS drug delivery and targeting. Adv Drug Deliv Rev. 2010;62(4–5):478–90. 3. Gillies ER, Frechet JM. Dendrimers and dendritic polymers in drug delivery. Drug Discov Today. 2005;10(1):35–43. 4. Patri AK, Majoros IJ, Baker JR. Dendritic polymer macro- molecular carriers for drug delivery. Curr Opin Chem Biol. 2002;6(4):466–71. 5. Pillai O, Panchagnula R. Polymers in drug delivery. Curr Opin Chem Biol. 2001;5(4):447–51. 6. Ojewole E, Mackraj I, Naidoo P, Govender T. Exploring the use of novel drug delivery systems for antiretroviral drugs. Eur J Pharm Biopharm. 2008;70(3):697–710. 7. Jabr-Milane L, van Vlerken L, Devalapally H, Shenoy D, Ko- mareddy S, Bhavsar M, Amiji M. Multi-functional nanocarriers for targeted delivery of drugs and genes. J Control Release. 2008;130(2):121–8. 8. Kumar M. Nano and microparticles as controlled drug delivery devices. Int J Pharm Pharm Sci. 2000;3(2):234–58. 9. Dutta T, Jain NK. Targeting potential and anti-HIV activity of lamivudine loaded mannosylated poly (propyleneimine) den- drimer. Biochim Biophys Acta. 2007;1770(4):681–6. J Mater Sci: Mater Med (2015) 26:179 Page 7 of 8 179 123
  • 8. 10. Gunaseelan S, Gunaseelan K, Deshmukh M, Zhang X, Sinko PJ. Surface modifications of nanocarriers for effective intracellular de- livery of anti-HIV drugs. Adv Drug Deliv Rev. 2010;62(4–5):518–31. 11. D’Emanuele A, Attwood D. Dendrimer–drug interactions. Adv Drug Deliv Rev. 2005;57(15):2147–62. 12. Nanjwade BK, Bechra HM, Derkar GK, Manvi FV, Nanjwade VK. Dendrimers: emerging polymers for drug-delivery systems. Eur J Pharm Sci. 2009;38(3):185–96. 13. Namazi H, Adeli M. Dendrimers of citric acid and poly (ethylene glycol) as the new drug-delivery agents. Biomaterials. 2005;26(10): 1175–83. 14. Goldberg M, Langer R, Jia X. Nanostructured materials for ap- plications in drug delivery and tissue engineering. J Biomater Sci Polym Ed. 2007;18(3):241–68. 15. Kalishwaralal K, BarathManiKanth S, Pandian SR, Deepak V, Gurunathan S. Silver nanoparticles impede the biofilm formation by Pseudomonas aeruginosa and Staphylococcus epidermidis. Colloids Surf B. 2010;79(2):340–4. 16. Rai MK, Deshmukh SD, Ingle AP, Gade AK. Silver nanoparti- cles: the powerful nanoweapon against multidrug-resistant bac- teria. J Appl Microbiol. 2012;112(5):841–52. 17. He Y, Du Z, Lv H, Jia Q, Tang Z, Zheng X, Zhang K, Zhao F. Green synthesis of silver nanoparticles by Chrysanthemum morifolium Ramat. extract and their application in clinical ul- trasound gel. Int J Nanomed. 2013;8:1809. 18. Kim KJ, Sung WS, Moon SK, Choi JS, Kim JG, Lee DG. An- tifungal effect of silver nanoparticles on dermatophytes. J Mi- crobiol Biotechnol. 2008;18(8):1482–4. 19. Murugan E, Vimala G. Effective functionalization of multiwalled carbon nanotube with amphiphilic poly(propyleneimine) dendrimer carrying silver nanoparticles for better dispersability and antimi- crobial activity. J Colloid Interface Sci. 2011;357(2):354–65. 20. You C, Han C, Wang X, Zheng Y, Li Q, Hu X, Sun H. The progress of silver nanoparticles in the antibacterial mechanism, clinical ap- plication and cytotoxicity. Mol Biol Rep. 2012;39(9):9193–201. 21. Lu L, Sun RW, Chen R, Hui CK, Ho CM, Luk JM, Lau GK, Che CM. Silver nanoparticles inhibit hepatitis B virus replication. Antivir Ther. 2008;13(2):253. 22. Morones JR, Elechiguerra JL, Camacho A, Holt K, Kouri JB, Ramı´rez JT, Yacaman MJ. The bactericidal effect of silver nanoparticles. Nanotechnology. 2005;16(10):2346. 23. Lara HH, Ixtepan-Turrent L, Garza Trevin˜o EN, Singh DK. Use of silver nanoparticles increased inhibition of cell-associated HIV-1 infection by neutralizing antibodies developed against HIV-1 envelope proteins. J Nanobiotechnol. 2011;9:38. 24. Lai T, Hou Q, Yang H, Luo X, Xi M. Clinical application of a novel sliver nanoparticles biosensor based on localized surface plasmon resonance for detecting the microalbuminuria. Acta Biochim Biophys Sin. 2010;42(11):787–92. 25. Mohammed Fayaz A, Ao Z, Girilal M, Chen L, Xiao X, Kalaichelvan P, Yao X. Inactivation of microbial infectiousness by silver nanoparticles-coated condom: a new approach to inhibit HIV-and HSV-transmitted infection. Int J Nanomed. 2012;7:5007. 26. Butkus MA, Labare MP, Starke JA, Moon K, Talbot M. Use of aqueous silver to enhance inactivation of coliphage MS-2 by UV disinfection. Appl Environ Microbiol. 2004;70(5):2848–53. 27. Rezaei A, Zabihollahi R, Salehi M, Moghim S, Tamizifar H, Yazdanpanahi N, Amini G. Designing a non-virulent HIV-1 strain: potential implications for vaccine and experimental re- search. J Res Med Sci. 2007;12(5):227–34. 28. Gunasekaran T, Nigusse T, Dhanaraju MD. Silver nanoparticles as real topical bullets for wound healing. J Am Coll Clin Wound Spec. 2011;3(4):82–96. 29. Cavrois M, Neidleman J, Yonemoto W, Fenard D, Greene WC. HIV-1 virion fusion assay: uncoating not required and no effect of Nef on fusion. Virology. 2004;328(1):36–44. 30. Svarovskaia ES, Barr R, Zhang X, Pais GC, Marchand C, Pommier Y, Burke TR Jr, Pathak VK. Azido-containing diketo acid derivatives inhibit human immunodeficiency virus type 1 integrase in vivo and influence the frequency of deletions at two-long-ter- minal-repeat-circle junctions. J Virol. 2004;78(7):3210–22. 31. Morgan JR, LeDoux JM, Snow RG, Tompkins RG, Yarmush ML. Retrovirus infection: effect of time and target cell number. J Virol. 1995;69(11):6994–7000. 32. Kim YS, Song MY, Park JD, Song KS, Ryu HR, Chung YH, Chang HK, Lee JH, Oh KH, Kelman BJ, Hwang IK, Yu IJ. Subchronic oral toxicity of silver nanoparticles. Particle Fibre Toxicol. 2010;7(1):20. 33. Pana´cek A, Kola´r M, Vecerova´ R, Prucek R, Soukupova´ J, Krystof V, Hamal P, Zboril R, Kvı´tek L. Antifungal activity of silver nanoparti- cles against Candida spp. Biomaterials. 2009;30(31):6333–40. 34. Siliciano JD, Kajdas J, Finzi D, Quinn TC, Chadwick K, Mar- golick JB, Kovacs C, Gange SJ, Siliciano RF. Long-term follow- up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4? T cells. Nat Med. 2003;9(6):727–8. 35. Grant RM, Hecht FM, Warmerdam M, et al. Time trends in primary HIV-1 drug resistance among recently infected persons. JAMA. 2002;288(2):181–8. 36. Baba M. Recent status of HIV-1 gene expression inhibitors. Antiviral Res. 2006;71(2):301–6. 37. Emerman M, Malim MH. HIV-1 regulatory/accessory genes: keys to unraveling viral and host cell biology. Science. 1998;280(5371):1880–4. 38. Lau A, Swinbank KM, Ahmed PS, et al. Suppression of HIV-1 infection by a small molecule inhibitor of the ATM kinase. Nat Cell Biol. 2005;7(5):493–500. 39. Galdiero S, Falanga A, Vitiello M, Cantisani M, Marra V, Gal- diero M. Silver nanoparticles as potential antiviral agents. Molecules. 2011;16(10):8894–918. 40. Elechiguerra JL, Burt JL, Morones JR, Camacho-Bragado A, et al. Interaction of silver nanoparticles with HIV-1. J Nanobiotechnol. 2005;3(6):1–10. 41. De Boer RJ, Boucher CA. Anti-CD4 therapy for AIDS suggested by mathematical models. Proc R Soc Lond B. 1996;263(1372):899–905. 42. Lori F, Jessen H, Lieberman J, Clerici M, Tinelli C, Lisziewicz J. Immune restoration by combination of a cytostatic drug (hy- droxyurea) and anti-HIV drugs (didanosine and indinavir). AIDS Res Hum Retroviruses. 1999;15(7):619–24. 43. Lori F, Jessen H, Foli A, Lisziewicz J, Matteo PS. Long-term suppression of HIV-1 by hydroxyurea and didanosine. JAMA. 1997;277(18):1437–8. 44. Lori F, Jessen H, Lieberman J, et al. Treatment of human im- munodeficiency virus infection with hydroxyurea, didanosine, and a protease inhibitor before seroconversion is associated with normalized immune parameters and limited viral reservoir. J In- fect Dis. 1999;180(6):1827–32. 45. Georgiou NA, van der Bruggen T, Oudshoorn M, Nottet HS, Marx JJ, van Asbeck BS. Inhibition of human immunodeficiency virus type 1 replication in human mononuclear blood cells by the iron chelators deferoxamine, deferiprone, and bleomycin. J Infect Dis. 2000;181(2):484–90. 46. Alavidjeh MS, Haririan I, Khorramizadeh MR, Ghane ZZ, Ardestani MS, Namazi H. Anionic linear-globular dendrimers: biocompatible hybrid materials with potential uses in nanome- dicine. J Mater Sci. 2010;21(4):1121–33. 47. Haririan I, Alavidjeh MS, Khorramizadeh MR, Ardestani MS, Ghane ZZ, Namazi H. Anionic linear-globular dendrimer-cis- platinum (II) conjugates promote cytotoxicity in vitro against different cancer cell lines. Int J Nanomed. 2010;5:63–75. 179 Page 8 of 8 J Mater Sci: Mater Med (2015) 26:179 123