Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

International Journal of Biological Macromolecules 163 (2020) 745–755

Contents lists available at ScienceDirect

International Journal of Biological Macromolecules

journal homepage: http://www.elsevier.com/locate/ijbiomac

Microwave-assisted rapid synthesis of silver nanoparticles using


fucoidan: Characterization with assessment of biocompatibility and
antimicrobial activity
Sneha S. Rao, K. Saptami, Jayachandran Venkatesan, P.D. Rekha ⁎
Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka 575018, India

a r t i c l e i n f o a b s t r a c t

Article history: Silver nanoparticles (AgNPs) have gained attention due to their exceptional physicochemical properties and
Received 2 April 2020 biological activities, owing to which they are extensively used in biomedical field. We synthesized AgNPs by
Received in revised form 13 June 2020 rapid microwave-assisted method using fucoidan as a reducing agent. The synthesized fucoidan-AgNPs (F-
Accepted 24 June 2020
AgNPs) were characterized for the structural and functional properties. The bactericidal effect and mode of action
Available online 26 June 2020
of F-AgNPs on the pathogenic bacteria and biofilm formation were investigated along with the cytotoxicity
Keywords:
studies. The UV–Visible spectra of the F-AgNPs showed the surface resonance peak at 419 nm. The nanoparticles
Green synthesis were spherical in shape with particle size of 59.5 ± 1.46 nm and polydispersity index (PDI) of 0.3 ± 0.01. Capping
Metal nanoparticles of fucoidan on AgNPs was confirmed by FTIR with characteristic peaks of sulfate group. Silver content of F-AgNPs
P. aeruginosa biofilm inhibition was 87% with 13% contribution from the fucoidan moieties. The F-AgNPs showed antimicrobial activity against
Oxidative stress common pathogenic bacteria and was able to inhibit biofilm formation in Pseudomonas aeruginosa at 20 μg/mL
concentration. The oxidative stress and intracellular protein leakage were observed due to the F-AgNP interaction
with the cell bringing about bactericidal effect. The results highlight the synthesis and bioactivity of AgNPs doped
with organic moieties for applications as antimicrobials.
© 2020 Elsevier B.V. All rights reserved.

1. Introduction cultures as reducing agents to form metallic nanoparticles [17].This


results in non-toxic end products making them eco-friendly, cost-
Nanoparticles are extensively studied due to their advantages in effective, stable and easy to scale up compared to the chemical methods
terms of size, stability, strength, activity, large surface area and unique [18]. The natural sources represent a diverse pool of reducing agents
chemical and biological properties. These properties extend a wide and include different groups of phytochemicals, pigments, amines,
array of application in nano medicine as antimicrobial agents, wound proteins and carbohydrates that also have stabilizing effect on the
dressing, treating diabetic wounds, diagnosis and targeted drug delivery, formed nanoparticles, thus eliminating the need to use synthetic and
and biomedical applications as biosensors [1,2]. The transitional metal chemical stabilizers [19]. The phenolic group in the Coffea arabica seed
nanoparticles such as silver [3,4], gold [5,6], zinc [7,8], copper [9,10], extracts was used as an effective bio-reductant for synthesizing silver
iron [11,12] and platinum [13] are gaining attention due to their nanoparticles (AgNPs) of 20–30 nm size at room temperature [20].
versatile properties and wide range of applications as catalytic agents Alternatively, natural polymers such as agar and carrageenan from the
and antimicrobial agents [14,15]. Among these, silver and gold remain seaweeds to form silver nano-composites have been used with
the most attracted nanoparticles. The synthesis procedure is an satisfactory results [21]. The synthesis was attributed to the sulfate
important factor for achieving the size and shape of the nanoparticles and carboxylic groups present in the polymers that readily reduced
and the latter in turn determines its functional properties. the silver to form nanoparticles with size between 37 and 54 nm. The
Nanoparticle synthesis can be achieved by physical, chemical and process duration for the synthesis of AgNPs depend on the reducing
mechanical approaches and are generally classified into top-down or capacity of the reactants used. For example, formation of AgNPs using
bottom-up methods [16]. Green synthesis is a bottom-up process that the sea weed (Sargassum wightii) extract required 24 h of stirring at
uses natural materials such as plant extractives, bacterial or fungal room temperature to form stable NPs of 15–20 nm size [22].
Among the polysaccharides of natural origin, polysaccharides from
⁎ Corresponding author at: Yenepoya Research Centre, Yenepoya (Deemed to be
seaweeds such as carrageenan, alginate, pullulan and fucoidan are
University), University Road, Deralakatte, Mangalore 575018, Karnataka, India. used for the synthesis due to the presence of functional groups.
E-mail address: rekhapd@yenepoya.edu.in (P.D. Rekha). Fucoidan is a high molecular weight anionic carbohydrate polymer

https://doi.org/10.1016/j.ijbiomac.2020.06.230
0141-8130/© 2020 Elsevier B.V. All rights reserved.
746 S.S. Rao et al. / International Journal of Biological Macromolecules 163 (2020) 745–755

commonly extracted from the sea cucumbers or seaweeds such as Kyoto, Japan). The IR spectrum was recorded in the scan range of 400 to
Fucus sp., Sargassum sp., Padina sp., etc., and exhibit properties of 4000 cm−1 and interpreted based on the literature.
pharmaceutical importance [23]. Its biological activities mainly depend
on the molecular weight, degree of sulfation, occurrence of different Dynamic Light Scattering (DLS) analysis
monosaccharide units and their linkages. Fucoidan based AgNPs (F- The particle size distribution and the zeta potential of the
AgNPs) have previously been synthesized by overnight stirring and synthesized F-AgNPs were obtained by DLS method using Malvern
heating methods [24–27]. The methods so far reported for the Zetasizer Nano ZS (Worcestershire, UK). Briefly, the F-AgNPs were
production of F-AgNPs include longer time in extreme conditions. To suspended and filtered using Whatman No. 42 filter paper. The filtered
overcome the longer time required for the process, microwave based samples were analyzed at 25 °C for a minimum of three cycles and the
synthesis are being used for the synthesis of NPs as it offers benefits in particle size was estimated.
terms of batch to batch consistency in the process with quick formation
of the NPs. Hence, here, we used microwave-assisted method for the X-ray diffraction analysis
rapid synthesis of AgNPs using the fucoidan by optimizing the reaction The crystalline structure of the F-AgNPs was evaluated using an X-
conditions. The synthesized AgNPs were characterized in detail for its ray diffractometer (Rigaku miniflex 600, Tokyo, Japan). The lyophilized
structural and functional properties. F-AgNP samples were scanned at a rate of 4°/min at wavelength of
0.15406 nm and at a 2Ɵ angle pattern. The average crystalline size of
2. Materials and methods the F-AgNPs was calculated using Debye-Scherrer formula;

Fucoidan from Fucus vesiculosus (Mol. Wt. 46.4 kDa), silver nitrate L ¼ kλ=βcosθ
(AgNO3) and dimethyl sulfoxide (DMSO), 2′,7′-Dichlorofluorescin
diacetate (DCFDA) were purchased from Sigma Aldrich (St. Louis, MO, where L is the size of the crystalline nanoparticles, k is Scherrer constant
USA). NIH3T3 (mouse embryonic fibroblast) was obtained from (0.9), λ is the wavelength and β is the angular full-width at half
National Centre for Cell Sciences, Pune, Dulbecco's modified eagle's maximum (FWHM) of the XRD peak at the diffraction angle calculated
medium (DMEM), fetal bovine serum (FBS), antibiotic-antimycotic using origin software 2017.
solution, trypsin-EDTA, methyl thiazolyltetrazolium (MTT), acridine
orange, and ethidium bromide were purchased from HiMedia, India. Scanning Electron Microscopy (SEM) coupled with Energy Dispersive X-
Ray Analysis (EDX) and Transmission electron microscopy (TEM) analysis
2.1. Preparation of fucoidan-AgNPs The surface morphology of the synthesized F-AgNPs was studied
using SEM. For this, the F-AgNP sample was coated on the surface of
Fucoidan solution (1 mg/mL) and AgNO3 solution (1 mM) were the double-sided tape, gold-sputtered and analyzed using FE-SEM
prepared in distilled water at room temperature. For the synthesis of (Carl Zeiss, Germany). Scanning electron micrographs collected were
the fucoidan mediated AgNPs (F-AgNPs), silver nitrate and fucoidan analyzed for surface details. The elemental composition and distribution
solutions were mixed in different ratio (v:v) and subjected to were analyzed by Energy-dispersive X-ray spectroscopy (EDX) (Carl
microwave irradiation (LG MS-2049UW) at 100 °C for different time Zeiss, Germany). For TEM analysis, the F-AgNPs were suspended in
duration from 0 to 4 min. Further, based on the optimum ratio and water and a drop of the sample was placed on a copper grid and dried.
time, F-AgNPs were bulk synthesized and separated by centrifugation The morphology of the prepared F-AgNPs was analyzed using TEM-
at 12,000 rpm for 20 min. The supernatant and the F-AgNPs pellet SAED (Jeol/JEM 2100, Tokyo, Japan).
were collected separately. The collected F-AgNPs were washed with
distilled water, centrifuged, frozen at −80 °C overnight and lyophilized Antimicrobial activity
(FreezeZone benchtop freeze dryers, Labconco Corporation, Missouri, The antibacterial activity was tested against both Gram-positive and
USA). The lyophilized F-AgNPs were stored at room temperature in Gram-negative bacteria using agar well diffusion method as described
desiccator for further analysis. previously [28]. The microorganisms used were; Staphylococcus aureus
(ATCC 25932), Enterococcus faecalis (ATCC 29212), Streptococcus mutants
2.2. Characterization (ATCC 25932) Pseudomonas aeruginosa (MCC 2080), P. aeruginosa
(PAO1) and Escherichia coli (ATCC 25175). The anti-fungal activity was
UV–visible spectroscopy tested against Candida albicans (ATCC 90028). Briefly, inoculum was
The formation of F-AgNPs was followed by UV–visible spectroscopy prepared from overnight cultures of bacteria (0.05-OD600) and
(Shimadzu UV – 1800, Kyoto, Japan). Briefly, the different combinations inoculated on to Muller Hinton agar plates using a sterile swab to
of AgNO3 and fucoidan solutions subjected to microwave synthesis prepare a uniform lawn of bacteria. Subsequently, 6 mm diameter
were subjected to a spectrophotometric scan in the range of wells were created in the agar plates and 20 μL of F-AgNPs solutions at
200–800 nm. Based on the absorption maxima (λ max) the formation different concentrations (2.5, 5, 7.5 and 10 μg/mL) were added. For
of F-AgNPs was inferred. C. albicans, Sabouraud's dextrose agar was used and all other procedures
remained same. Standard antibiotic discs were used as positive controls
Estimation of silver content by ICP-MS and fucoidan was tested at concentrations between 2.5 and 100 μg/mL.
The concentration of silver in the F-AgNPs formed using fucoidan was The plates loaded with samples were incubated upright at 37 °C, and
determined by the inductively coupled plasma mass spectrometry after 24 h incubation, the zone of growth inhibition was measured.
analysis (ICPMS). For this both the supernatant and the lyophilized F-
AgNPs were used. The samples were typically digested and reconstituted Effect of F-AgNPs on the Pseudomonas aeruginosa biofilms
in an aqueous matrix containing 2% conc. nitric acid. Diluted and filtered Pseudomonas aeruginosa biofilms are always a concern in health care
(Whatman No. 42 filter paper). The silver content in the filtered solutions setting; hence, the biofilm inhibitory activity of F-AgNPs was tested
was analyzed using ICP-MS (Thermo Scientific iCAP RQ ICP-MS). against this model organism. As the biofilm inhibitory concentration is
always higher than the minimum inhibitory concentration (MIC), at
Fourier Transform Infrared Spectroscopy (FT-IR) first the MIC was tested. The MIC of both fucoidan and F-AgNPs was
To determine the chemical interaction between silver and fucoidan, determined by broth dilution method. In brief, Muller Hinton broth
FT-IR analysis of the F-AgNPs was carried out. Briefly, 10 mg sample was was inoculated with 105 CFU/mL of P. aeruginosa PAO1 and P. aeruginosa
placed and analyzed directly in a FT-IR instrument (IRSpirit, Shimadzu, MCC 2080 cultures. Different concentrations of fucoidan (12.5 to
S.S. Rao et al. / International Journal of Biological Macromolecules 163 (2020) 745–755 747

200 μg/mL) and F-AgNPs (0.15–100 μg/mL) were added and incubated seeded in to 96-well plates, incubated for 24 h (37 °C, 5% CO2), treated
at 37 °C for 24 h. The bacterial growth was measured based on the with different concentrations of F-AgNPs and further incubated for 24 h.
optical density at 600 nm spectrophotometrically and the MIC was To the cells, MTT reagent (1 mg/mL) was added treated for 4 h and the
determined as the lowest concentration at which no visible growth supernatants were removed. The formazan crystals formed were
was observed. For testing the biofilm inhibition by F-AgNPs, the solubilized using DMSO and the absorbance was recorded at 570 nm
P. aeruginosa biofilm was developed by growing in Luria-Bertani (LB) using a multimode micro-plate reader (FluoSTAR Omega, BMG Labtech,
media in 96-well polystyrene plates for 24 h at 37 °C. After incubation, Germany). The percent cell viability was calculated with respect to
the planktonic cells were discarded, the biofilm was treated with untreated control using the formula
different concentrations of F-AgNPs (0.125–20 μg/mL) in LB media
and incubated further for 24 h. After the incubation, the planktonic OD of sample
Cell viability% ¼  100
cells were removed, the wells containing biofilm were washed with OD of control
sterile PBS and fixed using 95% methanol. The fixed cells were strained
with crystal violet dye (0.1% aq.) for 10 min followed by PBS wash to
remove the excess stain from the wells. The stain was solubilized 2.4. Live – dead staining
inacetic acid (33%) and the absorbance of the solution was recorded at
580 nm to quantify the biofilm. The percentage inhibition was To visualize the proportion of the live and dead cells after treatment
calculated from the untreated control. with F-AgNPs acridine orange - ethidium bromide staining was used.
The NIH3T3 cells were seeded in 24-well plates (1 × 104 cells/well),
Time kill assay allowed to adhere and treated with F-AgNPs. After 24 h of treatment,
Time kill assay was performed to study the effect of F-AgNPs on the the culture supernatants were removed; cells were washed with PBS
growth kinetics of the bacteria. Briefly, bacterial cultures were prepared and fixed with chilled methanol for 10 min. Acridine orange (2 μg/mL)
with an initial OD 600 of 0.05 from overnight culture in Tryptic soya broth and ethidium bromide (2 μg/mL) were added in equal volumes and
(TSB) and treated with F-AgNPs (10 and 20 μg/mL). The bacterial growth incubated at 37 °C for 15 min in the dark. The excess stain was removed,
was measured by recording the OD600 values spectrophotometrically washed with PBS and images were captured in red and green channels
(FluoSTAR Omega, BMG Labtech, Germany). For comparison, untreated using a fluorescence imager (ZOE, Bio-Rad, California, USA).
control was used. The growth curve was plotted using the OD600
readings recorded at different time intervals between 0 and 24 h. Also, 2.5. Statistical analysis
to test the bacterial residual cell viability, the samples were serially
diluted and plated on TSA media. The colony forming units (CFU) were All the experiments were performed at least in triplicates. The data is
enumerated using a digital colony counter and represented as CFU/mL. presented as mean ± standard deviation. Difference between the
treatments was analyzed using one-way analysis of variance (ANOVA)
Estimation of reactive oxygen species (ROS) and p b 0.05 was considered as significant.
To test whether the F-AgNPs treatment results in the oxidative stress
due to the formation of reactive oxygen species (ROS), 2′,7′- 3. Results and discussion
dichlorofluorescein diacetate (DCFDA) method was used as per the
previously described method [29]. For this, the overnight cultures of 3.1. Green synthesis of F-AgNPs
P. aeruginosa PAO1 and MCC 2080 strains were treated with F-AgNPs
(10 and 20 μg/mL) in TSB and incubated for 4 h in a shaking incubator Among the different combinations of fucoidan and silver nitrate
(120 rpm) maintained at 37 °C. For comparison untreated cells were solutions used for the synthesis, the 9:1 (v/v) fucoidan (1 mg/mL) and
used as control. After the incubation, the cells were harvested by silver nitrate (1 mM) solutions at room temperature subjected to
centrifugation (5000 rpm, 5 min, 4 °C), re-suspended in sterile PBS microwave irradiation for 4 min yielded the F-AgNPs. The reaction
containing 100 μM DCFDA and incubated at 37 °C in dark for 30 min. mixture was colorless at the initial stage, turned into pale yellow at
The cells collected by centrifugation (5000 rpm, 5 min, 4 °C) were 1 min, golden yellow at 2 min, light brown at 3 min and dark brown
lysed using alkaline lysis buffer and the lysate was used for measuring at four-minutes of microwave radiation (Fig. 1b). The formation of F-
the fluorescent intensity using fluorescence spectrophotometer (F- AgNPs was indicated by the occurrence of dark brown color at 4 min,
2700, Hitachi Tokyo, Japan) at an emission wavelength of 522 nm. due to the excitation of AgNPs surface plasma resonance. The UV–
Visible spectra of the suspension containing the F-AgNPs showed the
Intracellular protein leakage characteristic peaks due to the presence of AgNPs (Fig. 1a). At the
Further, to estimate the intracellular protein leakage due to F-AgNP baseline, there was no observed peak formation, however on
treatment, overnight cultures of P. aeruginosa PAO1 and MCC 2080 microwave irradiation clear occurrence of peaks at 419 nm was visible
strains were treated with F-AgNPs (10 and 20 μg/mL) in TSB and and the peak intensity increased to reach the highest at 4 min of
incubated for 12 h in the shaking incubator (120 rpm) at 37 °C. After microwave. Generally, the AgNPs with size ranging between 2 nm to
the treatment the cultures were centrifuged to collect the cell free 100 nm show a characteristic peak at 420 ± 5 nm, due to their surface
supernatant for estimating the intracellular protein leakage by Bradford plasmon. This coincides with the most commonly reported excitation
method [30]. Respective bacterial cultures without treatment were used wavelength of AgNPs [32,33]. Under reducing environment, the
as control. OD595 readings were recorded spectrophotometrically to reduction of silver occurs and the remains stable for months. The
estimate the protein content from the standard graph obtained using microwave induced degradation of fucoidan results in release of
bovine serum albumin (BSA). reducing sugars [34] which attributes to the efficient reduction of silver
nitrate to silver nanoparticles. Also, the degradation products of the
2.3. Assay for cytotoxicity of F-AgNPs reducing agents act as capping agents to give stable nanoparticles [35].
The relative abundance of silver ions present in the pellet and
The cytotoxicity of F-AgNPs was tested at different concentrations supernatant collected after the reaction was determined by ICP-MS.
(2.5, 5, 10, 15 20 and 50 μg/mL) in vitro using NIH3T3 cells by MTT The pellet containing F-AgNPs contained 4.6 ± 0.4 mg (in 100 mL
assay as previously described by Mosmann [31]. The NIH3T3 cells reaction mixture) of Ag making up to 44.35% of the silver used in the
were grown in DMEM containing 10% fetal bovine serum and 1% reaction, while, the supernatant contained 4.4 ± 0.3 mg (in 100 mL
antibiotic-antimycotic solution. For MTT assay, 5 × 103 cells/well were reaction mixture) of Ag making up to 41%. Thus, the 10.37 mg product
748 S.S. Rao et al. / International Journal of Biological Macromolecules 163 (2020) 745–755

Fig. 1. a. UV visible spectra for the biosynthesized F-AgNPs at different time intervals of 0, 1, 2, 3 and 4 mins. The sharp peak at 419 nm at 3rd and 4th min correspond to silver; b. Formation
of F-AgNPs at different times of microwave irradiation with gradual color change from colorless to dark brown at 4th min.

recovered from the 100 mL reaction mixture after microwave the F-AgNPs was estimated as 59.5 ± 1.46 nm with a polydispersity
irradiation (4 min) contained 9.0 mg of Ag. This shows that the index (PDI) of 0.3 ± 0.01 (Fig. 2c). The PDI indicates that the prepared
recovered product is predominantly made of Ag and small quantity of particles are monodisperse in nature with a narrow range of size
the fucoidan moieties. Such data is important to know the percentage distribution. The zeta potential of the F-AgNPs was – 22.7 ± 2.44 mV,
conversion and recovery rates during the synthesis. negative charge confirming the presence of anionic fucoidan. Such
properties are associated with higher particle stability. AgNPs
3.2. Fourier Transform –Infrared (FT-IR) spectra and functional groups synthesized using natural extracts exhibit variation in the particle
sizes between 30 and 130 nm [42].The AgNPs synthesized using
The interaction between different functional groups involved in the carboxymethylated-curdlan or fucoidan by high temperature heating
formation of F-AgNPs was studied using the FT-IR spectroscopy. The (100 °C) showed particle size in the range of 40–80 nm [26]. The size
characteristic peaks for fucoidan were observed at 835 cm−1, of the nanoparticles mainly depends on the conditions used for the
1009 cm−1, 1218 cm−1, 1652 cm−1, 1729 cm−1 and 3403 cm−1 synthesis such as temperature and pH and it plays an important role
(Fig. 2a). The peaks at 835 cm−1, 1009 cm−1, 1218 cm−1 correspond in determining the physical and biological properties [43].
to the sulfate groups (S_O) [36,37] C\\O\\S stretching of the sulfate
groups in fucoidan [24]. The peaks at 835 cm−1corresponding to the 3.5. Surface morphology and atomic distribution
sulfate groups of fucoidan are evident in the absorption spectrum of F-
AgNPs. The absorption peaks of OH stretching (3403 cm−1) and The FESEM of the F-AgNPs showed uniform spherical structures in
1729 cm−1 corresponding to the C_O of the carbohydrate group the nanometer range representing the nanoparticles (Fig. 2d). Further,
observed in the FT-IR spectra of the recovered F-AgNPs strongly indicate the EDX analysis showed the distribution of silver by an optical
the presence of fucoidan moieties in the product. absorption peak at ~3 KeV as well the peaks for carbon and oxygen
from the fucoidan contributing to capping of the AgNPs (Fig. 2e). Similar
3.3. X-ray diffraction (XRD) pattern of F-AgNPs observations were reported onearlier on the F-AgNPs and occurrence of
a strong peak at 3 KeV indicating the presence of silver as the key
The XRD patterns of the biosynthesized F-AgNPs shows the component [44]. Uniform shape and particle distribution are key
crystalline nature of silver with several Braggs peaks (Fig. 2b). The attributes of successful synthesis.
peaks at 2Ɵ 38.06, 44.32, 64.58 and 77.3 corresponds to the (111),
(200), (220) and (311) planes of face-centered cubic structure of silver 3.6. Transmission Electron Microscopy-selected area electron diffraction
indicating the signature peaks for the biosynthesized AgNPs (JCPDS 31- (TEM-SAED)
1238). Many studies have reported the presence of silver peaks at these
regions in X-ray diffractogram [38–40] and this is due to the formation The TEM observations further confirmed that the biosynthesized F-
of AgNPs by reduction of Ag+ ions using fucoidan under microwave AgNPs were highly spherical in shape (Fig. 3a). The coating of AgNPs
energy. The average crystallite size of the F-AgNP calculated from is mostly due to the reducing sugars (glucose and xylose) present in
Debye-Scherer equation is 20.41 ± 11.77 nm and this size reveal the the fucoidan which can act as capping agents [45]. The ring-like
nano-crystalline nature of synthesized F-AgNPs [41]. diffraction patterns (Fig. 3b) observed in the SAED analysis corresponds
to the (111), (200), (220), and (311) faced-centered cubic (fcc)
3.4. Size and polydispersity index of the F-AgNPs structure of silver in AgNPs [39,46].The particle size measured based
on the TEM shows the size of 36.99 ± 12.39 nm (Fig. 3c). The diffraction
The hydrodynamic radius and the surface charge of the patterns obtained from SAED and XRD analysis confirms the crystalline
biosynthesized F-AgNPs were analyzed using DLS. The average size of nature of the synthesized nanoparticles. The occurrence of these
S.S. Rao et al. / International Journal of Biological Macromolecules 163 (2020) 745–755 749

Fig. 2. Characteristics of the F-AgNPs. Fourier transform infrared spectrum of Fucoidan and F-AgNPs; b. XRD pattern; c. Dynamic light scattering analysis. d. SEM images (scale bar 200 nm)
and; e. EDX spectra showing a peak at 3 KeV for the presence of metallic silver.

patterns is a common feature of AgNPs by green synthesis methods highest zone of growth inhibition at a concentration of 10 μg/mL.
[47–49]. Among the tested microbes, Candida albicans showed the highest
zone of inhibition (21 mm), followed by Pseudomonas aeruginosa
3.7. Antimicrobial activity (19 mm), Streptococcus mutants (18 mm) Staphylococcus aureus
(16 mm), Escherichia coli (15 mm) and Enterococcus faecalis
The AgNPs are well reported for their broad range antimicrobial (10 mm) (Table 1, Fig. 4a). Interestingly, at all the tested
activity [50]. The antimicrobial efficacy of the biosynthesized F- concentrations the zone of growth inhibition was obsereved
AgNPs was tested at different concentrations and showed the showing its activity against broad range of microbes.However, the

Fig. 3. a. TEM images of F-AgNPs (scale bar 10 nm); b. SAED patterns support the crystalline F-AgNPs with the occurrence of ring like diffraction patterns; and c. Particle size distribution
pattern.
750 S.S. Rao et al. / International Journal of Biological Macromolecules 163 (2020) 745–755

Table 1 3.8. MIC and Biofilm inhibitory activity of F-AgNPs on Pseudomonas


Zone of inhibition (mm) of F-AgNPs against Staphylococcus aureus, Enterococcus faecalis, aeruginosa
Pseudomonas aeruginosa, Escherichia coli, Streptococcus mutants and Candida albicans.

Microbe Zone of inhibition (mm) Pseudomonas aeruginosa is an opportunistic pathogen causing


F-AgNPs nosocomial infections and is difficult to treat due to its ability form
2.5 μg/mL 5 μg/mL 7.5 μg/mL 10 μg/mL
biofilms. The bacteria inside the biofilm environment are resistant to
S. aureus 13 14 15 16
E. faecalis 7 8 9 10 the antibiotics due to the strong matrix developed as result of secretion
E. coli 12 13 14 15 of extracellular polymeric substances [51]. Many antimicrobials fail
P. aeruginosa 16 17 18 19 against the biofilm forming organisms and also, the biofilm inhibitory
S. mutants 15 16 17 18 concentration is several folds higher than the MIC.To establish the activity
C. albicans 16 17 18 21
of F-AgNPs on the P. aeruginosa biofilms, two strains namely, PAO1 and
MCC 2080 were used. The MIC of F-AgNPs was between 5 and 10 μg/mL
against both the strains (Fig. 4b & c). As stated earlier fucoidan alone
fucoidan alone did not display the antimicrobial activity at the tested did not show any activity up to a concentration of 100 μg/mL.The strains
concentrations of 2.5–100 μg/mL. PAO1 and MCC 2080 are strong biofilm formers with biofilm intensity
The AgNPs synthesized using fucoidan from Turbinaria conoides by (OD570) of 0.90 and 0.91 respectively in LB media under control
heating at 75 °C showed growth inhibition against gram negative conditions. The F-AgNPs treatment on the pre-developed biofilm showed
bacteria such as Escherichia coli, Klebsiella pneumoniae, Vibrio cholera, a dose dependent inhibition (Fig. 4b & c). Biofilm inhibition by 50% was
Pseudomonas aeruginosa, Salmonella typhi and Shigella sonnie [25]. achieved with 10 μg/mL concentration of F-AgNP. At the higher

Fig. 4. a. Antimicrobial activity of fucoidan and F-AgNPs at different concentrations (2.5 μg/mL, 5 μg/mL, 7.5 μg/mL and 10 μg/mL) tested on Staphylococcus aureus, Enterococcus faecalis,
Pseudomonas aeruginosa, Escherichia coli, Streptococcus mutants, Candida albicans using agar well diffusion method; b. Biofilm inhibition in P. aeruginosa strains, MCC2080 and PAO1 by
F-AgNPs treatment. The data points are mean ± S.D. c. Biofilm stained by crystal violet showing the reduced intensity of PAO1 and MCC 2080 biofilms treated with different
concentrations of F-AgNPs.
S.S. Rao et al. / International Journal of Biological Macromolecules 163 (2020) 745–755 751

Fig. 5. a & b. Growth curve for the P. aeruginosa strains PAO1 and MCC 2080 treated with 10 and 20 μg/mL of F-AgNPs at different time points at 37 °C. The untreated group was used as
control. The results are expressed as mean ± S.D. compared to the control; c & d. Bar graphs representing the CFU/mL for the P. aeruginosa strains PAO1 and MCC 2080 strains treated with
10 and 20 μg/mL of F-AgNPs at 12 and 24 h. The results are expressed as mean ± S.D. **p b 0.01, compared to the control.

concentration of 20 μg/mL, the biofilm was inhibited by 80%. Disrupting are able to bring about bactericidal effect as early as 6 h in both the
the biofilm completely is difficult due to the protection of cells in the strains. The CFU calculated from the serially diluted samples were
thick biofilm matrix. The biofilm architecture contains macromolecules consistent with the OD600 values showing rapid decrease in the number
and can retain or interact with the antimicrobial compounds preventing colonies after treatment with F-AgNPs (Fig. 5c & d). At 24 h there were a
its interaction with the bacterial cells [52]. This is one of the reasons for few colonies of residual bacteria in the treatment, that represent the
the failure of antibiotic therapy by the conventional antibiotics against cells entrapped in the biofilm. Hence, repeated treatment may be
biofilm forming pathogens. required to completely eliminate the residual bacteria.
Some of the AgNPs with additional capping have shown activity
against the bacterial biofilms. For example, silver-nanoparticle- 3.10. Mode of action of F-AgNPs on the biofilm forming P. aeruginosa
decorated quercetin showed anti-biofilm effect on a multi-drug
resistant Escherichia coli strain and it was mainly due to the biofilm Due to their small size, AgNPs have the ability to attach to the
inhibitory activity of quercetin [53]. Similarly, the carboxymethyl bacterial cell wall and penetrate inside to the cytoplasm. During this
tamarind polysaccharide capped AgNPs could inhibit the bacterial cell process, it can cause structural changes in the cell membrane such as
division and thereby exerting the lethal effect on the biofilm produced changes in the permeability of the cell membrane. It is also envisaged
by drug resistant isolates [54]. The nano-sized silver particles can get that, AgNPs have the ability of producing high levels of reactive oxygen
incorporated in the biofilm and migrate to the interior of the biofilms species (ROS) and free radical species such as hydrogen peroxide,
through the micro-channels present in the architecture [55].The superoxide radicle, hydroxyl radical and singlet oxygen that damage
conventional antibiotics fail to reach the biofilm interior due to their the DNA and the cellular processes causing the bacterial cell killing
entrapment and chemical interaction with the charged extra- [56]. To test whether the F-AgNPs have similar effect on the bacterial
polymeric substances. Materials having excellent biofilm inhibitory cells, the ROS levels were estimated based on DCFDA method. The
potential are suitable for the coating of medical devices intended for treatment with F-AgNPs induced the significantly higher (p b 0.005)
implantation or devices such as catheters. ROS production at 20 μg/mL concentration compared control and
10 mg/mL F-AgNPs for both the P. aeruginosa strains as measured by
3.9. Time kill assay the fluorescent intensity (Fig. 6a, b, c & d). Similar events have been
reported by researchers on C. albicans, S. cerevisiae and other microbes
To study the effect of F-AgNPs on the growth of P. aeruginosa strains [57]. Oxidative stress is one of the common mechanisms by which the
PAO1 and MCC 2080 time kill assay was performed. The growth of the antibiotics target the bacterial cells. Increased concentration of the
bacteria under normal culture conditions followed a typical sigmoidal free radicals interferes with the cell cycle regulation and cellular
pattern and under the F-AgNP treatment, the growth did not take metabolic pathways. The ROS can cause intercellular calcium
place showing a flattened curve (Fig. 5a & b). This shows that F-AgNPs accumulation resulting in the loss of membrane potential triggering
752 S.S. Rao et al. / International Journal of Biological Macromolecules 163 (2020) 745–755

Fig. 6. a & b. Reactive oxygen species generated in P. aeruginosa strains PAO1 and MCC 2080 treated with 10 and 20 μg/mL of F-AgNPs for 4 h at 37 °C. ROS was detected using DCFDA
method and compared with untreated group control. The results are expressed as mean ± S.D., *p b 0.05 and ***p b 0.005 compared to the control; c & d. Fluorescence spectra for the
treated groups PAO1 and MCC 2080, respectively control and treatment with 10 and 20 μg/mL of F-AgNPs at 522 nm; e & f. Intracellular protein leakage in P. aeruginosa strains PAO1
and MCC 2080 treated with 10 and 20 μg/mL of F-AgNPs for 12 h at 37 °C. The untreated group served as control. The results are expressed as mean ± S.D. **p b 0.01 and ***p b 0.0001.

apoptosis [58]. The compromised cellular pathways trigger the cellular which is made up of peptidoglycan [59]. The adhesion of AgNPs on
transcription towards apoptosis failing to survive the stress. bacterial cell wall is attributed to the positive charge in the AgNPs
Intracellular protein leakage is also another important mechanism of which assures electrostatic attraction between negatively charged
action by AgNPs. The F-AgNPs treatment at concentrations of 10 and bacterial cell membrane [60]. The anti-fungal activity exerted by
20 μg/mL released significantly higher protein to the culture AgNPs in Candida is achieved by cell membrane permeabilization by
supernatant of the bacteria compared to the untreated control the NPs, manifesting disruption of the cell leading to inhibition of the
(p b 0.01). The protein contents in the supernatants of PAO1 and MCC budding [61].
2080 cultures respectively were 54.82 ± 1.42 μg/mL and 103.26 ±
0.29 μg/mL with 20 μg/mL F-AgNPs (Fig. 6e & f). It has been shown 3.11. Biocompatibility of F-AgNPs
that the silver ions of the AgNPs damage of the bacterial membrane by
binding and blocking the thiol and amino groups [29,59].The membrane It is well documented that AgNPs are relatively cytotoxic to
integrity is the key for the bacterial cell-functioning and transport of mammalian cells also. Here, the biocompatibility of the F-AgNPs was
nutrients and metabolites as well preventing the entry of xenobiotics tested in vitro using NIH3T3 cells at a wide range of concentrations
and toxic substances. The cells with destabilized membranes undergo coveringthe doses of antibacterial and anti-biofilm effects (2.5 to
apoptosis and reflected as cell killing by the treatment with F-AgNPs. 50 μg/mL). The MTT assay results showed that at concentrations of
The anti-bacterial activity of AgNPs in Gram-negative bacteria is 2.5, 5 and 7.5 μg/mL, the cell viability remained at 97.3%, 91.4% and
stronger than in Gram-positive bacteria due to the cell wall thickness, 87.5% respectively indicating the cytocompatibility of the synthesized
S.S. Rao et al. / International Journal of Biological Macromolecules 163 (2020) 745–755 753

Fig. 7. a. Effect ofF-AgNPs on the viability of NIH3T3 cells treated for 24 h. Results are based on the MTT assay and cell viability compared to the untreated control. The results are expressed
as mean ± S.D., *p b 0.05 and **p b 0.01, compared to control group; b. Live dead staining using acridine orange and ethidium bromide of NIH3T3 cells after 24 h treatment with 2.5 μg/mL
and 50 μg/mL F-AgNPs. Green represents living cells and red the dead cells. Scale bar 100 μm.

F-AgNPs (Fig. 7a). Earlier studies on AgNPs prepared using different Acknowledgements
methods on diverse cell lines showed toxicity at higher doses. The
AgNPs prepared using the bark extracts of Toxicodendron vernicifluum Authors acknowledge the funding support from DST-SERB, Govt. of
containing alcoholic, aromatic and phenolic groups showed no toxicity India (EMR/2016/003113). Authors also acknowledge the help
on NIH3T3 cells up to 320 μg/mL [62]. It is assumed that the coating provided by Dr. Ashwini Prabhu in the cell culture experiments.
with biopolymers reduces the toxicity of AgNPs that can be suitable
the tissue engineering purposes. AgNPs with a coating of low Author contribution
molecular weight chitosan reduced its toxicity on HS27 human
fibroblast cells compared to uncoated AgNPs [63]. AgNPs synthesized Sneha S Rao: Performed experiments, analyzed data and co-wrote
using natural extracts containing phenolics and flavanoids from the manuscript
plants such as Arnebia hispidissima and Terminalia bellirica showed K Saptami: Helped in conducting antimicrobial studies and data
no toxicity against normal cells [64,65].The cytotoxicity of the analysis
AgNPs also depends on the concentration of the silver present in Jayachandran Venkatesan: Conceptualised the study, involved in the
the decorated/capped preparations. Here, the Ag content in the F- research and helped in data analysis
AgNP was 44% in the prepared material. Hence, it is also important PD Rekha: Conceptualised the study, analyzed data and co-wrote
to know the silver/metal content relative to the organics present in and approved the manuscript
the total material. All authors read and approved the manuscript.
The live dead staining of the NIH3T3 cells treated with F-AgNPs is
represented in Fig.7b. The cells showed presented viability with intact References
morphology as indicated by the intense green fluorescence due to the
[1] M. Holzinger, A. Le Goff, S. Cosnier, Nanomaterials for biosensing applications: a
acridine orange. The results indicate that the F-AgNPs at lower review, Front. Chem. 2 (2014) 63, https://doi.org/10.3389/fchem.2014.00063.
concentration were least toxic to the normal NIH3T3 cells in conformity [2] M.T. Alam, M.A. Rauf, G.A. Siddiqui, M. Owais, A. Naeem, Green synthesis of silver
with the MTT results. Considering the higher activity of the AgNPs in nanoparticles, its characterization, and chaperone-like activity in the aggregation
lower concentrations, it could be considered at lower concentrations inhibition of α-chymotrypsinogen A, Int. J. Biol. Macromol. 120 (2018)
2381–2389, https://doi.org/10.1016/j.ijbiomac.2018.09.006.
at which it is compatible to normal cells for various biomedical [3] M.J. Firdhouse, P. Lalitha, Biosynthesis of silver nanoparticles and its applications, J.
applications. Nanotechnol. 2015 (2015)https://doi.org/10.1155/2015/829526.
[4] X.-F. Zhang, Z.-G. Liu, W. Shen, S. Gurunathan, Silver nanoparticles: synthesis,
characterization, properties, applications, and therapeutic approaches, Int. J. Mol.
Sci. 17 (9) (2016) 1534, https://doi.org/10.3390/ijms17091534.
4. Conclusion [5] P. Suchomel, L. Kvitek, R. Prucek, A. Panacek, A. Halder, S. Vajda, R. Zboril, Simple
size-controlled synthesis of Au nanoparticles and their size-dependent catalytic
activity, Sci. Rep. 8 (1) (2018) 4589, https://doi.org/10.1038/s41598-018-22976-5.
The microwave mediated biosynthesis of AgNPs using fucoidan was [6] Y.-C. Yeh, B. Creran, V.M. Rotello, Gold nanoparticles: preparation, properties, and
rapid and the F-AgNPs showed characteristics and properties that can applications in bionanotechnology, Nanoscale 4 (6) (2012) 1871–1880, https://
be useful for antimicrobial applications including against biofilms. doi.org/10.1039/c1nr11188d.
Detailed characterization of the F-AgNPs confirmed size shape, [7] J. Jiang, J. Pi, J. Cai, The advancing of zinc oxide nanoparticles for biomedical
applications, Bioinorganic. Chem. Appl. 2018 (2018)https://doi.org/10.1155/2018/
crystallinity and presence of functional groups due to fucoidan. The 1062562.
synthesized F-AgNPs contained 87% Ag and remaining as organics [8] A. Ali, A.R. Phull, M. Zia, Elemental zinc to zinc nanoparticles: is ZnO NPs crucial for
contributed from the fucoidan. The F-AgNPs exhibited an effective broad life? Synthesis, toxicological and environmental concerns, Nanotechnol. Rev. (2018)
https://doi.org/10.1515/ntrev-2018-0067.
range antimicrobial activity against Staphylococcus aureus, Enterococcus
[9] N. Nazar, I. Bibi, S. Kamal, M. Iqbal, S. Nouren, K. Jilani, M. Umair, S. Ata, Cu
faecalis, Pseudomonas aeruginosa, Escherichia coli, Streptococcus mutants nanoparticles synthesis using biological molecule of P. granatum seeds extract as
and Candida albicans. The study outlined a rapid and green synthesis reducing and capping agent: growth mechanism and photo-catalytic activity, Int.
method for AgNP production using fucoidan as a reducing and stabilizing J. Biol. Macromol. 106 (2018) 1203–1210, https://doi.org/10.1016/j.ijbiomac.2017.
08.126.
agent. Its excellent antimicrobial activity, biofilm inhibition potency and
[10] P. Chawla, N. Kumar, A. Bains, S.B. Dhull, M. Kumar, R. Kaushik, S. Punia, Gum arabic
biocompatibility can make it ideal for the use as coating material in capped copper nanoparticles: synthesis, characterization, and applications, Int. J.
medical devices. Biol. Macromol. (2020)https://doi.org/10.1016/j.ijbiomac.2019.12.260.
754 S.S. Rao et al. / International Journal of Biological Macromolecules 163 (2020) 745–755

[11] S. Saif, A. Tahir, Y. Chen, Green synthesis of iron nanoparticles and their [37] L. Pereira, S.F. Gheda, P.J. Ribeiro-Claro, Analysis by vibrational spectroscopy of
environmental applications and implications, Nanomaterials 6 (11) (2016) 209, seaweed polysaccharides with potential use in food, pharmaceutical, and cosmetic
https://doi.org/10.3390/nano6110209. industries, Int. J. Carbohydr. Chem. 2013 (2013)https://doi.org/10.1155/2013/
[12] D.L. Huber, Synthesis, properties, and applications of iron nanoparticles, Small 1 (5) 537202.
(2005) 482–501, https://doi.org/10.1002/smll.200500006. [38] B. Mehta, M. Chhajlani, B. Shrivastava, Green synthesis of silver nanoparticles and
[13] S. Chen, K. Kimura, Synthesis of thiolate-stabilized platinum nanoparticles in their characterization by XRD, J. Phys. Conf. Ser. 836 (1) (2017), 012050. https://
protolytic solvents as isolable colloids, J. Phys. Chem. B 105 (23) (2001) doi.org/10.1088/1742-6596/836/1/012050.
5397–5403, https://doi.org/10.1021/jp0037798. [39] C. Yu, J. Tang, X. Liu, X. Ren, M. Zhen, L. Wang, Green biosynthesis of silver
[14] J.M. Palomo, Nanobiohybrids: a new concept for metal nanoparticles synthesis, nanoparticles using eriobotrya japonica (thunb.) leaf extract for reductive catalysis,
Chem. Commun. 55 (65) (2019) 9583–9589, https://doi.org/10.1039/C9CC04944D. Materials 12 (1) (2019) 189, https://doi.org/10.3390/ma12010189.
[15] S. Dakshayani, M. Marulasiddeshwara, S. Kumar, R. Golla, S. Devaraja, R. Hosamani, [40] M. Khan, F. Tarek, M. Nuzat, M. Momin, M. Hasan, Rapid biological synthesis of silver
Antimicrobial, anticoagulant and antiplatelet activities of green synthesized silver nanoparticles from Ocimum sanctum and their characterization, J. Nanosci. 2017
nanoparticles using Selaginella (Sanjeevini) plant extract, Int. J. Biol. Macromol. (2017)https://doi.org/10.1155/2017/1693416.
131 (2019) 787–797, https://doi.org/10.1016/j.ijbiomac.2019.01.222. [41] A.M. Awwad, N.M. Salem, Green synthesis of silver nanoparticles by mulberry leaves
[16] A.M. Ealias, M. Saravanakumar, A review on the classification, characterisation, extract, J. Nanosci. Nanotechnol. 2 (4) (2012) 125–128, https://doi.org/10.5923/j.nn.
synthesis of nanoparticles and their application, IOP Conf. Ser. Mater. Sci. Eng 263 20120204.06.
(2017), 032019. https://doi.org/10.1088/1757-899X/263/3/032019.
[42] A. Roy, O. Bulut, S. Some, A.K. Mandal, M.D. Yilmaz, Green synthesis of silver
[17] M. Rafique, I. Sadaf, M.S. Rafique, M.B. Tahir, A review on green synthesis of silver
nanoparticles: biomolecule-nanoparticle organizations targeting antimicrobial
nanoparticles and their applications, Artif. Cells Nanomed. Biotechnol. 45 (7)
activity, RSC Adv. 9 (5) (2019) 2673–2702, https://doi.org/10.1039/c8ra08982e.
(2017) 1272–1291, https://doi.org/10.1080/21691401.2016.1241792.
[43] J.K. Patra, K.-H. Baek, Green nanobiotechnology: factors affecting synthesis and
[18] K. Parveen, V. Banse, L. Ledwani, Green synthesis of nanoparticles: their advantages
characterization techniques, J. Nanomater. 2014 (2014)https://doi.org/10.1155/
and disadvantages, AIP Conf. Proc. 1724 (1) (2016), 020048. https://doi.org/10.
2014/417305.
1063/1.4945168.
[44] A. Ravichandran, P. Subramanian, V. Manoharan, T. Muthu, R. Periyannan, M.
[19] M. Manoth, K. Manzoor, M. Patra, P. Pandey, S. Vadera, N. Kumar, Dendrigraft
polymer-based synthesis of silver nanoparticles showing bright blue fluorescence, Thangapandi, K. Ponnuchamy, B. Pandi, P.N. Marimuthu, Phyto-mediated synthesis
of silver nanoparticles using fucoidan isolated from Spatoglossumasperum and
Mater. Res. Bull. 44 (3) (2009) 714–717, https://doi.org/10.1016/j.materresbull.
2008.06.033. assessment of antibacterial activities, J. Photochem. Photobiol. B 185 (2018)
117–125, https://doi.org/10.1016/j.jphotobiol.2018.05.031.
[20] V. Dhand, L. Soumya, S. Bharadwaj, S. Chakra, D. Bhatt, B. Sreedhar, Green synthesis
of silver nanoparticles using Coffea arabica seed extract and its antibacterial activity, [45] A.Y. Obaid, S.A. Al-Thabaiti, E. El-Mossalamy, L.M. Al-Harbi, Z. Khan, Extracellular
Mater. Sci. Eng. C. 58 (2016) 36–43, https://doi.org/10.1016/j.msec.2015.08.018. bio-synthesis of silver nanoparticles, Arab. J. Chem. 10 (2) (2017) 226–231,
[21] T.A. Roseline, M. Murugan, M.P. Sudhakar, K. Arunkumar, Nanopesticidal potential https://doi.org/10.1016/j.sjbs.2016.02.025.
of silver nanocomposites synthesized from the aqueous extracts of red seaweeds, [46] A. Rautela, J. Rani, M.D. Das, Green synthesis of silver nanoparticles from
Environ. Technol. Innov. 13 (2019) 82–93, https://doi.org/10.1016/j.eti.2018.10.005. Tectonagrandis seeds extract: characterization and mechanism of antimicrobial
[22] N. Shanmugam, P. Rajkamal, S. Cholan, N. Kannadasan, K. Sathishkumar, G. action on different microorganisms, J. Anal. Sci. Technol. 10 (1) (2019) 5, https://
Viruthagiri, A. Sundaramanickam, Biosynthesis of silver nanoparticles from the doi.org/10.1186/s40543-018-0163-z.
marine seaweed Sargassumwightii and their antibacterial activity against some [47] K. Jyoti, M. Baunthiyal, A. Singh, Characterization of silver nanoparticles synthesized
human pathogens, Appl. Nanosci. 4 (7) (2014) 881–888, https://doi.org/10.1007/ using Urticadioica Linn. leaves and their synergistic effects with antibiotics, J. Radiat.
s13204-013-0271-4. Res. Appl. Sci. 9 (3) (2016) 217–227, https://doi.org/10.1016/j.jrras.2015.10.002.
[23] S. Luthuli, S. Wu, Y. Cheng, X. Zheng, M. Wu, H. Tong, Therapeutic effects of [48] S. Li, Y. Shen, A. Xie, X. Yu, L. Qiu, L. Zhang, Q. Zhang, Green synthesis of silver
Fucoidan: a review on recent studies, Mar. Drugs. 17 (9) (2019) 487, https://doi. nanoparticles using Capsicum annuum L. extract, Green Chem. 9 (8) (2007)
org/10.3390/md17090487. 852–858, https://doi.org/10.1039/B615357G.
[24] J. Venkatesan, S. Singh, S. Anil, S.-K. Kim, M. Shim, Preparation, characterization and [49] J. Saha, A. Begum, A. Mukherjee, S. Kumar, A novel green synthesis of silver
biological applications of biosynthesized silver nanoparticles with chitosan-fucoidan nanoparticles and their catalytic action in reduction of Methylene Blue dye, Sustain.
coating, Molecules 23 (6) (2018) 1429, https://doi.org/10.3390/ Environ. Res. 27 (5) (2017) 245–250, https://doi.org/10.1016/j.serj.2017.04.003.
molecules23061429. [50] M. Rai, A. Yadav, A. Gade, Silver nanoparticles as a new generation of antimicrobials,
[25] K. Kala, P.K. Prashob, N. Chandramohanakumar, Analysis of antimicrobial potential Biotechnol. Adv. 27 (1) (2009) 76–83, https://doi.org/10.1016/j.biotechadv.2008.09.
of silver nanoparticles synthesized by fucoidan isolated from Turbinariaconoides, 002.
Int. J. Pharmacogn. 8 (12) (2016) 1959–1963. [51] L.R. Mulcahy, V.M. Isabella, K. Lewis, Pseudomonas aeruginosa biofilms in disease,
[26] T.C.-Y. Leung, C.K. Wong, Y. Xie, Green synthesis of silver nanoparticles using Microb. Ecol. 68 (1) (2014) 1–12, https://doi.org/10.1007/s00248-013-0297-x.
biopolymers, carboxymethylated-curdlan and fucoidan, Mater. Chem. Phys. 121 [52] C. Vipin, M. Mujeeburahiman, A.B. Arun, P. Ashwini, S.V. Mangesh, P.D. Rekha,
(3) (2010) 402–405, https://doi.org/10.1016/j.matchemphys.2010.02.026. Adaptation and diversification in virulence factors among urinary catheter-
[27] S. Rajesh kumar, Phytochemical constituents of fucoidan (Padinatetrastromatica) associated Pseudomonas aeruginosa isolates, J. Appl. Microbiol. 126 (2) (2019)
and its assisted AgNPs for enhanced antibacterial activity, IET Nanobiotechnol 11 641–650, https://doi.org/10.1111/jam.14143.
(3) (2016) 292–299, https://doi.org/10.1049/iet-nbt.2016.0099.
[53] L. Yu, F. Shang, X. Chen, J. Ni, L. Yu, M. Zhang, D. Sun, T. Xue, The anti-biofilm effect of
[28] F.A. Qais, I. Ahmad, Broad-spectrum inhibitory effect of green synthesised silver silver-nanoparticle-decorated quercetin nanoparticles on a multi-drug resistant
nanoparticles from Withaniasomnifera (L.) on microbial growth, biofilm and
Escherichia coli strain isolated from a dairy cow with mastitis, PeerJ 6 (2018),
respiration: a putative mechanistic approach, IET Nanobiotechnol 12 (3) (2017) e5711. https://doi.org/10.7717/peerj.5711.
325–335, https://doi.org/10.1049/iet-nbt.2017.0193.
[54] S. Sanyasi, R.K. Majhi, S. Kumar, M. Mishra, A. Ghosh, M. Suar, P.V. Satyam, H.
[29] S. Liao, Y. Zhang, X. Pan, F. Zhu, C. Jiang, Q. Liu, Z. Cheng, G. Dai, G. Wu, L. Wang, L.
Mohapatra, C. Goswami, L. Goswami, Polysaccharide-capped silver nanoparticles
Chen, Antibacterial activity and mechanism of silver nanoparticles against
inhibit biofilm formation and eliminate multi-drug-resistant bacteria by disrupting
multidrug-resistant Pseudomonas aeruginosa, Int. J. Nanomedicine. 14 (2019)
bacterial cytoskeleton with reduced cytotoxicity towards mammalian cells, Sci. Rep.
1469–1481, https://doi.org/10.2147/IJN.S205736.
6 (1) (2016) 1–16, https://doi.org/10.1038/srep24929.
[30] M.M. Bradford, A rapid and sensitive method for the quantitation of microgram
[55] J. Wu, F. Li, X. Hu, J. Lu, X. Sun, J. Gao, D. Ling, Responsive assembly of silver
quantities of protein utilizing the principle of protein-dye binding, Anal. Biochem.
nanoclusters with a biofilm locally amplified bactericidal effect to enhance
72 (1–2) (1976) 248–254, https://doi.org/10.1016/0003-2697(76)90527-3.
treatments against multi-drug-resistant bacterial infections, ACS Cent. Sci. 5 (8)
[31] T. Mosmann, Rapid colorimetric assay for cellular growth and survival: application
(2019) 1366–1376, https://doi.org/10.1021/acscentsci.9b00359.
to proliferation and cytotoxicity assays, J. Immunol. Methods 65 (1–2) (1983)
55–63, https://doi.org/10.1016/0022-1759(83)90303-4. [56] L.C. Yun’an Qing, R. Li, G. Liu, Y. Zhang, X. Tang, J. Wang, H. Liu, Y. Qin, Potential
[32] P. Logeswari, S. Silambarasan, J. Abraham, Synthesis of silver nanoparticles using antibacterial mechanism of silver nanoparticles and the optimization of orthopedic
plants extract and analysis of their antimicrobial property, J. Saudi Chem. Soc. 19 implants by advanced modification technologies, Int. J. Nanomedicine 13 (2018)
(3) (2015) 311–317, https://doi.org/10.1016/j.jscs.2012.04.007. 3311, https://doi.org/10.2147/IJN.S165125.
[33] D. Kumar, G. Kumar, V. Agrawal, Green synthesis of silver nanoparticles using [57] B. Lee, M.J. Lee, S.J. Yun, K. Kim, I.H. Choi, S. Park, Silver nanoparticles induce reactive
Holarrhenaantidysenterica (L.) Wall. bark extract and their larvicidal activity oxygen species-mediated cell cycle delay and synergistic cytotoxicity with 3-
against dengue and filariasis vectors, Parasitol. Res. 117 (2) (2018) 377–389, bromopyruvate in Candida albicans, but not in Saccharomyces cerevisiae, Int. J.
https://doi.org/10.1007/s00436-017-5711-8. Nanomedicine 14 (2019) 4801–4816, https://doi.org/10.2147/IJN.S205736.
[34] T. Ohshiro, Y. Ohmoto, Y. Ono, R. Ohkita, Y. Miki, H. Kawamoto, Y. Izumi, Isolation [58] W. Lee, K.J. Kim, D.G. Lee, A novel mechanism for the antibacterial effect of silver
and characterization of a novel fucoidan-degrading microorganism, Biosci. nanoparticles on Escherichia coli, Biometals 27 (6) (2014) 1191–1201, https://doi.
Biotechnol. Biochem. 74 (8) (2010) 1729–1732, https://doi.org/10.1271/bbb. org/10.1007/s10534-014-9782-z.
100327. [59] Y.G. Yuan, Q.L. Peng, S. Gurunathan, Effects of silver nanoparticles on multiple drug-
[35] P. Patel, P. Agarwal, S. Kanawaria, S. Kachhwaha, S. Kothari, Plant-based synthesis of resistant strains of Staphylococcus aureus and Pseudomonas aeruginosa from
silver nanoparticles and their characterization, Nanotechnology and Plant Sciences, mastitis-infected goats: an alternative approach for antimicrobial therapy, Int. J.
Springer 2015, pp. 271–288, https://doi.org/10.1007/978-3-319-14502-0_13. Mol. Sci. 18 (3) (2017) 569, https://doi.org/10.3390/ijms18030569.
[36] M. Mmola, M. Roes-Hill, K. Durrell, J. Bolton, N. Sibuyi, M. Meyer, D. Beukes, E. [60] A. Abbaszadegan, Y. Ghahramani, A. Gholami, B. Hemmateenejad, S. Dorostkar, M.
Antunes, Enhanced antimicrobial and anticancer activity of silver and gold Nabavizadeh, H. Sharghi, The effect of charge at the surface of silver nanoparticles
nanoparticles synthesised using Sargassumincisifolium aqueous extracts, Molecules on antimicrobial activity against gram-positive and gram-negative bacteria: a
21 (12) (2016) 1633, https://doi.org/10.3390/molecules21121633. preliminary study, J. Nanomater. (2015)https://doi.org/10.1155/2015/720654.
S.S. Rao et al. / International Journal of Biological Macromolecules 163 (2020) 745–755 755

[61] K.J. Kim, W.S. Sung, B.K. Suh, S.K. Moon, J.S. Choi, J.G. Kim, D.G. Lee, Antifungal [64] S. Nindawat, V. Agrawal, Fabrication of silver nanoparticles using
activity and mode of action of silver nano-particles on Candida albicans, Biometals Arnebiahispidissima (Lehm.) A. DC. root extract and unravelling their potential
22 (2) (2009) 235–242, https://doi.org/10.1007/s10534-008-9159-2. biomedical applications, Artif. Cells Nanomed. Biotechnol. 47 (1) (2019) 166–180,
[62] K. Saravanakumar, R. Chelliah, D. MubarakAli, D.-H. Oh, K. Kathiresan, M.-H. Wang, https://doi.org/10.1080/21691401.2018.1548469.
Unveiling the potentials of biocompatible silver nanoparticles on human lung [65] S.V. Nampoothiri, B. Suresh Kumar, T. Esakkidurai, K. Pitchumani, Green synthesis of
carcinoma A549 cells and Helicobacter pylori, Sci. Rep. 9 (1) (2019) 5787, https:// silver nanoparticles using a characterized polyphenol rich fraction from
doi.org/10.1038/s41598-019-42112-1. terminaliabellirica and the evaluation of its cytotoxicity in normal and cancer
[63] Y. Peng, C. Song, C. Yang, Q. Guo, M. Yao, Low molecular weight chitosan-coated cells, Journal of Biologically Active Products from Nature 8 (6) (2018) 352–363,
silver nanoparticles are effective for the treatment of MRSA-infected wounds, Int. https://doi.org/10.1080/22311866.2018.1540944.
J. Nanomedicine 12 (2017) 295, https://doi.org/10.2147/IJN.S122357.

You might also like