You are on page 1of 15

Carbon 105 (2016) 362e376

Contents lists available at ScienceDirect

Carbon
journal homepage: www.elsevier.com/locate/carbon

Review article

The controversial antibacterial activity of graphene-based materials


Hanaa M. Hegab a, b, Ahmed ElMekawy c, d, Linda Zou a, e, Dennis Mulcahy a,
Christopher P. Saint a, Milena Ginic-Markovic f, *
a
Centre for Water Management and Reuse, University of South Australia, Adelaide, SA 5095, Australia
b
Institute of Advanced Technology and New Materials, City of Scientific Research and Technological Applications, Borg Elarab, Alexandria, Egypt
c
Genetic Engineering and Biotechnology Research Institute, University of Sadat City (USC), Sadat City, Egypt
d
School of Chemical Engineering, University of Adelaide, Adelaide, SA 5005, Australia
e
Department of Chemical and Environmental Engineering, Masdar Institute of Science and Technology, Abu Dhabi, United Arab Emirates
f
Future Industries Institute, University of South Australia, Adelaide, SA 5095, Australia

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

Article history: Graphene (Gr)-based materials are a promising nanomaterial for the development of antibacterial sur-
Received 11 January 2016 faces owing to their biocidal activity. However, the effect of the physicochemical features of these ma-
Received in revised form terials on their antibacterial activity has yet to be clarified. Gr-based nanomaterials can interact with
18 April 2016
cellular components, e.g. membranes, proteins and DNA, and initiate a sequence of nanomaterials/bac-
Accepted 19 April 2016
Available online 23 April 2016
terial interactions that rely on colloidal energies and active bio-physicochemical interfaces. Analyzing
these different interfaces permits the development of anticipated relations between physical/chemical
structure and bactericidal activity depending on Gr-based nanomaterial features such as shape, size,
hydrophilicity, roughness and functionality. Realizing how nanomaterials are interacting with bacterial
cell membranes is correlated to how they affect bactericidal activity and is thus critical for obtaining
benign applications. This review analytically discusses specific Gr-based material features related to
bacterial interactions, with special focus on the different modes of interaction between Gr-based ma-
terials and cell membranes, nucleic acids and lipid bilayers.
© 2016 Elsevier Ltd. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 363
2. Antibacterial activities of Gr-based materials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 365
2.1. Gr-based nanocomposite dispersions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 365
2.2. Gr-based nanocomposite surfaces . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 366
2.3. Gr-based hydrogels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 366
2.4. Bacterial reduction of GO . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 367
3. Gr-based materials features related to biocidal activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 367
3.1. Sheet size and concentration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 367
3.2. Surface area . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 367
3.3. Surface roughness . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 368
3.4. Hydrophilicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 368
3.5. Dispersibility . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 368
3.6. Functionalization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 369
4. Antibacterial mechanisms of Gr-based materials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 369
4.1. General mechanisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 369
4.1.1. Bacterial membrane stress . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 369
4.1.2. Oxidative stress . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 369
4.1.3. Wrapping isolation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 371

* Corresponding author.

http://dx.doi.org/10.1016/j.carbon.2016.04.046
0008-6223/© 2016 Elsevier Ltd. All rights reserved.
H.M. Hegab et al. / Carbon 105 (2016) 362e376 363

4.2. Gr-based suspension/membrane antibacterial capabilities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 371


5. Potential of Gr antiviral ability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 373
6. Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 373
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 373
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 373

1. Introduction adherence of Gr to biological cells [10].


When the toxicity of Gr nanosheets against bacteria and fungi
Biocidal nanomaterials is an attractive field of science that has was investigated, it was observed that these nanosheets can
the capability to, amongst other things, enhance medical devices possibly be applied as antimicrobial products, in a way similar to
[1], wastewater treatment [2], food packaging [3], synthetic textiles the most commonly used antimicrobial nanoscale materials like
[4] and dentistry [5] (Fig. 1). Development of novel and efficient silver nanoparticles (Ag NPs) [11]. The widespread use of Gr
antibacterial agents is urgently required due to the growing anti- nanosheets in biological applications is a result of their outstanding
bacterial resistance spectrum of bacterial infections. The antibac- bactericidal activity on a number of Gram negative and positive
terial action of nanoparticles (NPs) is generally assessed against the bacteria. This has increased the attraction towards the synthesis of
typical group of pathogenic bacteria, e.g. Staphylococcus aureus, GO along with its derivatives and rGO [12]. It has been observed
Escherichia coli, Listeria monocytogenes, Streptococcus mutans and that the degree of antibacterial effect of nanomaterials is deter-
Pseudomonas aeruginosa, which are responsible for different hu- mined by their shape, surface functionalization, size, stability and
man infections related to weakened immunity and are a significant size distribution [6]. Consequently, several GO nanosheets cova-
threat to public health [6]. Accordingly, developing nanomaterial lently and non-covalently functionalised were developed in order
surface coatings with effective antibacterial abilities is vital for to improve their antibacterial effect, stability and decrease their
human health and wellbeing. One of the most recently developed negative ecophysiological toxic side effects [13e15].
biocidal nanomaterials is graphene (Gr), which has been inten- The bactericidal effect of Gr-based materials is controversial and
sively studied as a carbon based nanomaterial. It can be considered requires further analysis. For example, in one study the antibacte-
as a single atom sheet of graphite (Gt). Gr oxide GO nanosheet is the rial property of four kinds of carbon-based materials against E. coli
oxidized form of Gr. It is a two-dimensional, one atom-thick sheet was compared [16]. GO was observed to display the maximum
with sp2-carbon atoms organized hexagonally and ornamented antibacterial activity towards E. coli as compared to Gt, Gr and rGO.
with several oxygen functional groups. Likely, it is the slimmest Another study was conducted to investigate the antimicrobial ac-
alignment of carbon molecules and is a key element for other tivity of GO [17], and results revealed faster bacterial growth, when
graphitic derivatives, Fullerenes and carbon nanotubes (CNTs) [7]. the culture medium was supplemented with GO. This was
Gr has exceptional physicochemical features, which include robust explained by the stimulation of bacterial proliferation via GO,
mechanical strength, great surface area and resistance to degra- which acts as a surface for cellular attachment and growth. Such
dation [7,8], due to its superior surface-to-volume ratio [9]. The significant outcomes for the application of Gr-based materials as
active segments, edges and surfaces, enable the attachment and antibacterial agents are a direct consequence of the intensive
attention these nanomaterials have attracted over the last six years,
as apparent from the number of publications on this topic. Fig. 2
demonstrates that searching ‘ScienceDirect’, using the keywords
‘‘graphene and antibacterial”, showed 96% growth in the total sci-
entific publications over the previous six years (2009e2015). More
than half of this research was conducted in the last two years.
Accordingly, in the present review the most contemporary

Fig. 1. Potential applications of antibacterial graphene in different fields including Fig. 2. The scientific research progress in biocidal activity of graphene based on the
water purification, energy and medicine. (A colour version of this figure can be viewed number of articles in the “ScienceDirect” database till November 2015. (A colour
online.) version of this figure can be viewed online.)
364 H.M. Hegab et al. / Carbon 105 (2016) 362e376

Table 1
The features and antibacterial efficiency of different freestanding and nanocomposite Gr-based materials. (A colour version of this figure can be viewed online.)

Gr-based materials features Antibacterial evaluation

Type Hybrid material Fabrication method Concentration Bacterial strains Incubation Method Inhibition Reference
(mg/ml) time (h) (%)

Gra-based Nanocomposite GOb Hummers and Offeman 175 P.hh aeruginosa 2 PCnn 100 [22]
Dispersions rGOc Hummers and Offeman 102 E.ii coli 4 PC 88 [138]
GO Hummers and Offeman 85 E. coli 2 PC 98.5 [20]
GO Hummers and Offeman 40 E. coli 2 PC 69.3 [16]
GO Hummers and Offeman 40 E. coli 2 PC 97.7 [21]
Gr QDsd Electrochemical 200 E. coli/S.jj aureus 0.25 PC 80/92 [23]
Gelatine/Age Green-Synthesis 10 E. coli 24 PC 99.9 [139]
nanoprisms
Ag NPsf Turkevich method with GO 5 P. aeruginosa 0.5 PC 100 [24]
dispersion
2
Ag NPs Mixing 10 E. coli 4 PC 100 [25]
Ag NPs Electrostatic interactions 3.2/6.4 E. coli 24 PC 99.9 [26]
Ag NPs Solution-based N/Agg E. coli/P. aeruginosa 24 ADoo 18/26 mm [27]
Ag NPs Chemical deposition N/A S. aureus/B.kk subtilis 24 AD 100 [28]
Ag NPs Sequential repetitive chemical N/A E. coli 0.3 AD 20 mm [29]
reduction
Ag NPs Supercritical CO2 10 E. coli/S. aureus 24 PC 100 [30]
Ag NPs Mixing 45 E. coli/S. aureus 4 PC 100 [31]
Ag NPs/PDDAg One-pot hydrothermal 50 E. coli 24 PC 100 [32]
Ag NPs/PEIh Mixing 958  103 E. coli/S. aureus 6 PC 14.8/20.5 [33]
Ag NPs/PDAi In situ reduction 25 E. coli 24 AD 23.7 mm [34]
Ag NPs/PAAj In situ grafting N/A E. coli/S. aureus 24 AD 9.9/ [35]
11.4 mm
Ag NPs/ Nucleophilic substitution 500 E. coli/S. aureus 8 PC 100 [140]
aminophenol
Ag/Fe3O4k Hydrothermal N/A E. coli 24 AD N/A [37]
Ag/TiO2l Two-phase 102 E. coli 2 PC 67 [36]
Fe3O4 NPs Mixing 3  102 E. coli 2 PC 91.5 [40]
ZnOm NPs Hydrothermal 6.6  105 Sal.ll typhi/E. coli 24 AD 13/11 mm [41]
ZnO NPs Mixing 3  103 E. coli 12 PC 100 [38]
MnOxn QDs/TiO2 UV excitation/microwave 31.25/15.62 E. coli/S. aureus 18 AD 10.9/ [43]
10.5 mm
MnFe2O4 Solvo-thermal 102 E. coli N/A PC 82 [44]
TiO2 Mixing N/A E. coli 0.5 PC 99.6 [39]
CuNPso/PLLp Mixing 50 E. coli 24 Tpp 99 [45]
Bi2WO6 Hydrothermal 250 Mixed culture 18 PC 100 [141]
Ag/CDq Mixing 0.05 B. subtilis 24 AD N/A [47]
CdSr Solvo-thermal 200 E. coli 1 PC 99.9 [48]
PEGs/PHGCt Mixing N/A E. coli/S. aureus 1 PC 100 [142]
Csu Drop-casting 3  103 P. aeruginosa 24 PC 100 [115]
Dithiothreitol Solvo-thermal 102 E. coli 4 PC 86 [138]
Sand Mixing N/A E. coli 24 AD 20 mm [143]
Ramizol Mixing N/A S. aureus 20 T 100 [144]
Gr-based Nanocomposite Ag NPs/PA Wet chemical reduction 103 E. coli 1 PC 98 [145]
Surfaces Ag/PESv Immersion phase inversion N/A E. coli/S. aureus/P. 24 AD N/A [59]
aeruginosa
Ag NPs/CAw Flow-directed assembly N/A E. coli 2 PC 86 [60]
Ag NPs/PSx Wet-phase inversion N/A E. coli 18 FESEMqq 100 [61]
Cs Solution-casting 6  104 S. aureus 3 PC 77 [50]
PVAy/Cs Electrospinning 4  103 E. coli 24 AD 8.6 mm [51]
PES Phase inversion N/A E. coli 4 PC 71 [55]
PA Surface functionalization N/A E. coli 1 PC 65 [146]
PAz Interfacial polymerization N/A E. coli 24 PC 65 [56]
PPaa LbLff- deposition N/A E. coli 12 PC 64 [57]
PLL/HAbb LbL- deposition 105 E. coli 4 PC 66 [52]
PEcc resin Casting N/A P. aeruginosa 24 AD 15 mm [54]
PA/PLL Hybrid grafting 80 Mixed culture 24 ATPrr 99 [53]
PES Phase inversion N/A E. coli 24 PC 74.88 [58]
ZnO NWs Drop-casting 103 E. coli 1 PC 95 [147]
ZnO NWs Electrophoretic deposition 103 E. coli 1 PC 99.5 [147]
SSdd/rGO Electrophoretic deposition N/A E. coli/S. aureus 1 PC 84/95 [63]
Ti Electrophoretic deposition N/A E. coli/S. aureus 24 PC 68.4/72.9 [62]
TiO2 Deposition N/A E. coli 0.5 PC 60 [49]
Cu Chemical vapor deposition N/A E. coli/S. aureus 24 PC 56/34 [64]
Gr-based Hydrogels BKBee One-pot tube inversion 4  103 E. coli/List.mm 48 T 99.3/91 [66]
hydrogel
BKB/PDA 4  103 E. coli/List. 48 T 99.3/91 [66]
Agarose Mixing N/A E. coli/S. aureus N/A PC 100 [65]
Ag Crosslinking 2.5  103 E. coli/S. aureus 0.5 PC 100 [68]
Ag/PVA Electrochemical reduction N/A E. coli/S. aureus 24 AD 100 [67]
a
Gr: Graphene.
b
GO: Graphene oxide.
H.M. Hegab et al. / Carbon 105 (2016) 362e376 365

c
rGO: Reduced graphene oxide.
d
QDs; Quantum dots.
e
Ag: Silver.
f
NPs: Nanoparticles.
g
PDDA: Poly (diallyl dimethyl ammonium chloride).
h
PEI: Polyethyleneimine.
i
PDA: Polydopamine.
j
PAA: Poly(acrylic acid).
k
Fe3O4: Iron oxide.
l
TiO2: Titanium dioxide.
m
ZnO: Zinc oxide.
n
MnO: Manganese oxide.
o
CuNPs: Copper nanoparticles.
p
PLL: Poly-L-lysine.
q
CD: Cyclodextrin.
r
CdS: Cadmium sulphide.
s
PEG: Polyethylene glycol.
t
PHGC: Polyhexamethylene guanidine hydrochloride.
u
Cs: Chitosan.
v
PES: Polyethersulfone.
w
CA: Cellulose acetate.
x
PS: Polysulfone.
y
PVA: Polyvinyl alcohol.
z
PA: Polyamide.
aa
PP: Polypropylene.
bb
HA: Hyaluronic acid.
cc
PE: Polyester.
dd
SS: Stainless steel.
ee
BKB: Benzalkonium bromide.
ff
LbL: Layer by layer.
gg
N/A: Not applicable.
hh
P.: Pseudomonas.
ii
E.: Escherichia.
jj
S.: Staphylococcus.
kk
B.: Bacillus.
ll
Sal.: Salmonella.
mm
List.: Listeria.
nn
PC: Plate count.
oo
AD: Agar diffusion.
pp
T: Turbidity.
qq
FESEM: Field-emission scanning electron microscopy.
rr
ATP: Adenosine triphosphate.

reports on the antibacterial activities of Gr-based materials are solvents [18]. GO Solutions in ethylene glycol (EG), water and N-
addressed, in order to resolve the argument about their antibac- methyl-2-pyrrolidone (NMP) offer a long lasting stability with high
terial activity. Moreover, their mechanisms of interaction with solubility values, e.g. 8.7 mg/mL for NMP [19]. Accordingly, several
bacterial cells are deliberated based on their forms and various workers investigated the antibacterial effects of Gr-based disper-
influential factors, e.g. sheet size, surface area and functionality, so sions against different Gram positive and negative bacterial cells.
that the contradictory subject of the antibacterial effect of Gr-based The first trial was conducted in 2010 when the bactericidal ac-
materials is revealed. tivities of two Gr-based derivatives in water dispersions; GO and
rGO nanosheets, were reported. It was shown that these Gr-based
nanomaterials can effectively inhibit the growth of E. coli bacte-
2. Antibacterial activities of Gr-based materials
ria, with even complete viability loss (98.5%) when GO was applied
[20]. These positive results opened the door for further studies to
In its comprehensive definition, an antibacterial agent is any
evaluate the Gr-based materials’ toxicity against bacterial cells. The
mediator that impedes the proliferation and growth of bacterial
bactericidal activity of four derivatives of carbon-based materials
cells. The Gr-based materials could follow the same concept.
(Gt, GtO, GO and rGO) against E. coli were evaluated [16]. A GO
Generally, Gr-based materials are chemically (i.e. chemical depo-
dispersion showed the uppermost activity (69.3%), consecutively
sition), electrochemically (i.e. electrophoretic deposition) or hy-
followed by rGO, Gt, and GtO with the same concentration and
drothermally (i.e. solvo-thermal) hybridized with different
under the same incubation conditions. Alternatively, the superior
functional materials (i.e. silver nanoparticles (Ag NPs), metal oxides
bactericidal activity of GO (25 mg/mL) was disproved when a bac-
and polymers) before being applied as antibacterial agents. The
terial culture was supplemented with it [17]. Faster bacterial
features of most recent antibacterial Gr-based dispersions, mem-
growth, through higher optical density, was observed compared to
branes and hydrogels are listed in Table 1 and summarized in the
cultures without GO. According to this study, it was obvious that GO
following sections.
lacks antibacterial capabilities. The contradictory findings extended
to the effect of the GO sheet size on its antibacterial property, in
2.1. Gr-based nanocomposite dispersions which E. coli was used for the evaluation of the bactericidal activity
of homogenous GO sheets dispersion, in which their lateral size
Exfoliated GO is the ultimate option for the synthesis of Gr diverges by more than 100 times [21]. The bactericidal activity of
applicable dispersion, as it can be produced in bulk from cheap Gt GO sheets against E. coli cells depended on lateral size, where larger
powder and can easily generate stable dispersions in different
366 H.M. Hegab et al. / Carbon 105 (2016) 362e376

GO sheets showed stronger bactericidal activity compared to metals (Lanthanum (La), Ag and Cd) to increase their antibacterial
smaller ones, with exposure time and concentration considered as ability [46e48]. For instance, G-CdS metallic composites with ho-
most effective aspects on bactericidal activities. The larger GO mogenous distribution of CdS NPs on GO nanosheets displayed
sheets bring about the majority of cell viability loss after 1 h of higher bactericidal activity towards E. coli than that with the pure
exposure time, and their concentration effectively impacts bacte- CdS NPs.
ricidal activity at relatively small concentration (<10 mg/mL).
Otherwise, when bacterial cells were incubated with smaller GO 2.2. Gr-based nanocomposite surfaces
sheets up to 4 h, the inhibition rate of bacterial cells increased
continuously. The threshold antibacterial concentration of GO was Membranes may be one of the most significant marketable
suggested to be 150e175 mg/mL in which no P. aeruginosa cells were products for Gr. Several industries, e.g. water desalination and gas
observed after 4 h incubation with these concentrations [22]. separation, could find application of Gr as a medium for different
Moreover, the antibacterial effect of electrochemically produced Gr membranes. Pristine or functionalized Gr membranes can have
quantum dots were investigated on S. aureus and E. coli. Bacterial outstanding potential in selective uptake and transportation of
cells lost their viability (80e92%) by the effect of generated reactive ionic or molecular species. One of the first trials to assess the
oxygen species (ROS) upon the photoexcitation of Gr quantum dots antibacterial property of Gr-based surfaces was conducted by
[23]. Akhavan and Ghaderi (2009), where GO nanosheets were depos-
In order to obtain the synergistic antibacterial effect of some ited on TiO2 thin films and displayed 60% bacterial inhibition [49].
nanomaterials along with Gr-based materials, an alternative strat- Another common route for the hybridized Gr membranes is the
egy was followed by hybridizing Ag NPs with Gr-based materials. polymer-GO, with various polymers (chitosan (CS) [50], poly vinyl
Several approaches were applied to assemble Ag NPs on GO sheets, alcohol (PVA) [51], PLL [52,53], polyester (PE) [54], poly-
i.e. one-pot hydrothermal, electrostatic interactions, simple mixing, ethersulfone (PES) [55], polyamide (PA) [56], polypropylene (PP)
chemical deposition, sequential repetitive chemical reduction and [57] and polyethylenimine (PEI) [58]) embedded via several tech-
supercritical CO2. The resultant nanocomposites were tested niques, viz. layer by layer (LbL)-deposition, casting, hybrid grafting,
against several bacterial strains (E. coli, P. aeruginosa, S. aureus, and phase inversion and electrospinning. The antibacterial efficiency of
Bacillus subtilis) in order to assess their antibacterial capabilities, these polymeric Gr-based membranes ranged from 64 to 99%
with a wide range of concentrations (3.2e102 mg/mL) [24e31]. Even [53,57]. The bactericidal activity of the membranes improved by
at low concentration, a complete inactivation rate of the tested increasing the Gr content. These results demonstrate that the
bacterial cells was observed after incubation with the GO-Ag NPs antibacterial and antifouling properties can be enhanced by bind-
for 30 min. Similarly, the smaller Ag NPs modified GO sheets ing Gr materials to the membrane surface.
showed more effective bactericidal ability compared to large Ag These promising results drove the research towards producing
NPs modified GO sheets [26]. The antibacterial activities of GO-Ag more efficient polymeric Gr-based membranes with improved
NPs against both E. coli and B. subtilis were improved more than antibacterial properties. In view of that, several Ag NPs - polymeric
one fold as compared to that of Ag NPs. Besides, this the Gram Gr based membranes were studied. The GO-Ag NPs nanocomposite
positive bacteria displayed higher resistance than the Gram nega- was used to modify PES [59], cellulose acetate (CA) [60] or poly-
tive ones to the GO-Ag NPs [30]. sulfone (PS) [61] membranes. The maximum antibacterial effi-
Polymeric modified rGO was applied as a platform for Ag NPs to ciency of these membranes was 100%, due to the synergistic effect
enhance the stability of Ag NPs. Based on this strategy, several of GO and Ag NPs.
polymers (poly diallyldimethyl ammonium chloride (PDDA), poly- Alternatively, the Gr-based composite coatings were produced
ethyleneimine (PEI), poly-dopamine (PDA) and poly acrylic acid by deposition on metal surfaces, like Ti [62], stainless steel (SS) [63]
(PAA)) were applied to obtain such composites via different syn- or copper (Cu) [64]. It was clear that the type of coated metal
thesis routes (one-pot hydrothermal, in situ reduction and in situ significantly affected the antibacterial ability of the Gr-coated sur-
grafting) [32e35]. The average bactericidal activity of Ag NPs was face. For example, when SS was coated with Gr, the biocidal activity
maintained in the polymeric rGO composites (100% inhibition), reached 95%, while this activity decreased to only 34% when Cu was
with extended time of exposure (6 h) compared to the non- coated.
polymeric Ag NPs-GO [33]. Moreover, the multifunctional GO-Ag
NPs nanocomposites were synthesized by integration with metal 2.3. Gr-based hydrogels
oxides, e.g. titanium dioxide (TiO2) [36] and iron oxide [37], for new
applications. The rapid inhibition effect (67%) against E. coli in just The three-dimensional GO-based hydrogels have recently
2 h evidently shows the synergistic influence of GO and Ag NPs on attracted more attention due to their valuable features. Neverthe-
the bactericidal features of the resultant nanocomposite particles. less, it is rather challenging to fabricate cheap, efficient and wide
On the other hand, metal oxides (MnO2, TiO2, Fe2O4 and ZnO) spectrum bactericidal hydrogels that can be easily recycled.
were solely applied to hybridize with Gr-based materials in order to Therefore, research was driven towards fabricating new Gr-based
enhance photocatalytic activity and synergistic antibacterial prop- composite hydrogels with high antibacterial capacity. One of the
erties of the resultant composites [38e44]. Different techniques early trials in this field was carried out by applying a simple and
were developed to homogenously decorate Gr-based materials eco-friendly technique to produce a multifunctional Gr-based
with metal oxide nanoparticles, viz. hydrothermal and solvo- hydrogel employing a biocompatible agarose polysaccharide
thermal, and the obtained hybrids were tested against several which simultaneously functions as a physical cross-linking agent
bacterial species. The lowest bactericidal effect (82%) was obtained and a stabilizer [65]. The newly fabricated hydrogel resulted in the
with 100 mg/mL of Gr manganese ferrite (MnFe2O4) as compared to full reduction of bacterial growth. Moreover, some commercial
Gr (37%) [44], while bacterial cells were totally inhibited upon the preservatives, e.g. benzalkonium bromide cationic surfactant, were
application of GO-ZnO or GO-TiO2 nanocomposites [38,39]. hybridized with GO to gain the dual antibacterial benefits of both
Furthermore, the triple antibacterial effect of poly-L-lysine (PLL)- materials. The benzalkonium bromide/GO hydrogel displayed
rGO-CuNPs was evaluated against E. coli and S. aureus, and the re- strong bactericidal activities toward both Gram positive (91%) and
sults revealed that there is a long-term antibacterial effect (99%) Gram negative (99%) bacteria [66]. The same “synergistic effect”
[45]. In addition, Gr-based materials were functionalized with strategy was investigated by fabricating Ag/Gr hydrogels
H.M. Hegab et al. / Carbon 105 (2016) 362e376 367

composites for enhanced levels of antibacterial efficacy. Several 3.1. Sheet size and concentration
hydrogels, like Ag/PVC/Gr [67] and Ag/Gr [68], were tested for their
antibacterial abilities, and the results revealed the complete The antibacterial activity of Gr-based materials was observed to
reduction of S. aureus and E. coli cellular growth. be influenced by the size of the GO sheet. Smaller sheet size
resulted in enhanced antibacterial activity for Gr-based surface
coatings. This concept was studied via investigating the simulating
2.4. Bacterial reduction of GO
effect of Gr sheets on bacterial cell membrane [79]. It was observed
that small Gr sheets rupture the phospholipid layer of the bacterial
One of the most confusing topics is the bacterial reduction of
membrane, while large Gr sheets stay flat on the top of this layer,
GO, where it contradicts the known bactericidal effect of Gr species.
where they destruct the membrane and turn over part of the
Some bacterial cells are capable of reducing GO nanomaterial by a
phospholipids. When the sheet area was decreased from 0.65 to
specific mechanism. This mechanism is determined by the direct or
0.01 mm2, the bactericidal activity of Gr surface coatings improved
indirect ability of bacterial cells to hydrolyze acidic groups com-
4-fold [80]. The greater antibacterial effect of smaller sheets is
bined with carbonic nanosheets in particular oxygen atoms. GO can
ascribed an oxidative stress mechanisms accompanied by the
act as a final electron acceptor in the bacterial respiration process
excessive defect density of these sheets. Moreover, GO sheet area
[69]. The first trial to bacterially reduce GO was conducted in 2010
hinders bacterial growth, in cell suspensions, by a cell wrapping
by using Shewanella with GO acting as a terminal electron acceptor,
mechanism. Under this environment, the bactericidal activity of GO
in which electron shuttling to GO is mediated by cytochromes.
is amplified as the sheet area increases, with evidence of complete
Subsequently, Shewanella was utilized for GO reduction in number
inhibition observed for sheets sized 0.65 mm2, after 3 h of incuba-
of studies by external electron shuttling intervened by the
tion. The same correlation was observed when the bactericidal
membrane-bound c-type heme cytochromes, and via self-
activity of GO sheets with various lateral sizes was assessed with
produced electron mediators [70,71].
E. coli cells [21]. GO sheets with greater lateral size showed a
Various types of bacteria were employed to assist in the
temporary powerful bactericidal activity compared to smaller ones.
reduction of GO. B. subtilis was applied to reduce GO and develop an
Nevertheless, cell wrapping was observed to be reversible, showing
enhanced supercapacitor [72]. The bacterial cells not only
that the noticeable effect of GO does not encompass bacterial in-
contribute towards the GO reduction, but also in producing the final
hibition and can be relatively designated as bacteriostatic [80]. The
composite through entrapment within the rGO. Extremophiles
size of Gr nanosheets is related to cellular uptake and several bio-
cells were also applied for GO reduction to produce an extremely
logical phenomena that rely on lateral dimension [81]. In general,
conductive Gr that can be employed in the manufacturing of mi-
the sharp edges, small size and rough surfaces of Gr nanosheets
croelectronics [73]. The extremophiles have the ability to reduce
facilitate their internalization into the bacterial cells compared to
GO through a particular mechanism that enables them to degrade
larger and smooth sheets [82]. The Gr nanosheets lateral sizes
toxic materials so as to survive under severe environmental con-
range from nano-GO (10 nm) to more than 20 mm, which is bigger
ditions. A different study was performed by Avinav et al. [74], in
than bacterial cells [83]. Additionally, lateral dimension and layer
which GO was produced from a cellulose precursor and was sub-
number have an impact on deformability, and also affect the spin
sequently reduced in situ by means of Gluconacetobacter xylinus. In
that can be employed by cellular forces, where small plates are less
two contradictory studies, E. coli was applied for the reduction of
deformable than large ones with the same layer number [84].
chemically exfoliated GO nanosheets [75,76]. In the first one, GO
Furthermore, the effect of Gr-based materials concentration on
nanosheets represent biocompatible spots for adsorption and
bactericidal activities was investigated in several studies. When GO
growth of the bacterial cells on their surfaces, whereas the resulted
suspensions, with different concentrations (5, 10, 20, 40 and 80 mg/
rGO nanosheets from the bacterial metabolism displayed inhibitory
mL), were exposed to E. coli cells, they resulted in the loss of bac-
effect for the bacterial growth on their surfaces [75]. However, the
terial viability as the GO concentration increased, until it reached
same bacterial cells have been used in the second study for GO
more than 90% with a GO concentration of 80 mg/mL [16,20,21].
reduction to Gr nanosheets that were dispersible in water [76].
Similarly, rGO dispersion with a concentration of 80 mg/mL eradi-
The bio-reduction of GO has several advantages in terms of
cates 76.8% of bacterial cells [16]. In addition, further higher con-
occurrence under ambient conditions in short time [77]. The
centrations (25e200 mg/mL) of Gr-based materials were studied in
bacterially-rGO showed outstanding electrochemical features [71],
terms of their effects on bactericidal activities [22]. P. aeruginosa
with the developed electroactive biofilm delivering 25-fold in-
gradually lost its viability with elevated concentrations of GO or
crease in the outward current in terms of electron release from
rGO, 75 and 100 mg/mL, respectively. These results suggested that
bacterial cells to electrodes [70]. The inward current was also
80 mg/mL is the threshold GO concentration that can lead to a
increased (74-fold), in terms of electron release from electrodes to
significant antibacterial activity (>90%), while this critical concen-
bacterial cells, by the developed biofilm compared to the naturally
tration is higher when rGO is used (100 mg/mL).
developed biofilms. Moreover, the bacterially-rGO has considerable
biocompatibility with fibroblast cells, even when applied at higher
3.2. Surface area
concentrations, e.g. 100 mg/ml. Hence, the green approach of the
bacterial reduction of GO confers a great applicable potential in the
Bacterial interactions of nanomaterials are greatly influenced by
biomedical field [78].
the nature of the surfaces [85]. Small NPs, less than 10 nm, usually
have a considerable portion of atomic surface exposure. Every atom
3. Gr-based materials features related to biocidal activity occurs on the surface in the case of monolayer Gr, in which each
atom is subjected to the contiguous medium on dual sides, leading
Gr-based nanomaterials vary in sheet size, surface area, rough- to increase in the speculative maximum surface area of the sp2-
ness, hydrophilicity, dispersibility and functionalization; and all of carbon sheet (2600 m2/g) [86]. This is superior to the surface area
these features can individually or interactively impact the interac- of other nanomaterials which have been investigated in microbial
tion of Gr with bacterial cells. The separate effect of each factor on systems. The GO single layer has a comparable area, however its
the viability of bacterial cells is illustrated in Fig. 3 and detailed in surface is controlled by roughness at atomic-scale. The number of
the following sections. layers in Gr nanosheets is vital as it determines bending stiffness
368 H.M. Hegab et al. / Carbon 105 (2016) 362e376

Fig. 3. The physiochemical properties of graphene nanomaterial relevant to its contradictory antibacterial effect. (A colour version of this figure can be viewed online.)

and specific surface area [87]. The adsorptive capacity for biotic contact angle 90 ). Normally, bacterial cells have higher affinity for
molecules is assumed to significantly increase as layer number hydrophobic surfaces due to hydrophobic interactions, which de-
decreases. pends on the type of bacteria tested [91]. Therefore, hydrophilic
surface modification is an effective approach for the establishment
of bactericidal surfaces by using polymeric materials, e.g. highly
3.3. Surface roughness
hydrated polyethylene glycol (PEG). However, some polymeric
chains can dehydrate at high ionic strengths, resulting in a lowering
The physical structure of the surface is a crucial factor affecting
of the efficiency for preventing bacterial adhesion on such surfaces
interaction with bacterial cells. Increased surface roughness im-
[95]. It is generally considered that the fast adsorption taking place
proves bacterial cell adhesion. Both high surface area and deep
on hydrophobic surfaces is accompanied by strong binding forces,
terrains in the surface result in greater colonization [88]. The sur-
in contrast to hydrophilic surface adhesion which follows the
face morphology features include peaks and valleys in the mem-
proposed model of reversible/irreversible adhesion [96]. A vulner-
brane surface resulting from the fabrication process [89]. Therefore
able and reversible initial bacterial adhesion stage was observed,
there is a difference between bacteriostatic surfaces that inhibit
split-up at distances of several nanometers, which could be de-
attachment and bactericidal surfaces that efficiently eradicate
tached by shear forces or spontaneous desorption. Subsequently,
bacterial cells on contact [90], although both surfaces are usually
production of extracellular biopolymers or stabilization into exist-
categorized as antibacterial. Some functionalities displayed
ing polymers results in the conversion of this adhesion to be irre-
powerful antibacterial action by generating membrane perme-
versible. The adsorbed water is replaced by these bridging
ability, due to the surface morphology, which can be employed to
polymers (separation spaces of <1 nm) along with the neutraliza-
destroy bacterial cells. Normally, such functionalized surfaces have
tion of the electrostatic repulsion [91]. Bacterial surface adhesion in
considerable roughness or ridges with high frequency, which can
aquatic environments typically encompasses hydrophobic groups
cause cracking of bacterial cell walls during contact [90]. Although
in the cell walls (nonpolar surface proteins), in which they help
the Gr sheet edges follow the latter mechanism, their basal planes
cells to make contact with the substratum, providing alterations in
are believed to be smooth at the atomic level. Thus, the interaction
surface polymers. These results in different functional groups
of the bacterial cells with Gr is expected to be greatly dependent on
approaching the surface for the creation of polymeric interactions
the alignment of the surface in relation to the bacteria [91].
[89]. Commonly, the adhesion ability of hydrophobic bacteria on
hydrophobic surfaces is greater than that of hydrophilic bacterial
3.4. Hydrophilicity cells [53,97].

The adhesion of bacterial cells on any substrate surface and their


ability to accumulate biofilm on it are influenced by its chemistry. 3.5. Dispersibility
GO sheets have amphiphilic nature with partial hydrophobic sur-
faces with some hydrophilic spots (contact angle 40e50 ) [92e94], The effective size of spherical NPs increases whenever they
while the Gr sheets have a hydrophobic surface (approximately aggregate in certain media, however, they can maintain the
H.M. Hegab et al. / Carbon 105 (2016) 362e376 369

majority of their surface area [98]. They are able to attach via any destroyed after a direct exposure to Gr-based materials [22].
contact point throughout sphere to sphere aggregation, except in A novel explanation for this antibacterial mechanism has been
the case of dissolution and reprecipitation of NPs, which may lead discovered. It revolves around the damage caused by the extraction
to partial fusion [99]. In contrast, sheets usually stack face to face, of phospholipids from the E. coli cell membrane (Fig. 4) [113]. This
which contributes to the loss of part of the surface area upon extraction was monitored by transmission electron microscope
irreversible aggregation during filtration or centrifugation pro- (TEM) after the exposure of E. coli cells to GO nanosheets. The
cesses. Regarding the GO dispersion, it comprises two-dimensional bacterial cells were initially tolerant to GO for a short time, after
carbon sheets [16], which are stable due to the great number of which a partial loss of cell membranes integrity occurred, with
different functional groups with hydrophilic nature, e.g. hydroxyl, some cells showing a lower density of surface phospholipid. Finally,
carboxyl, and epoxy groups [100]. This is one reason why GO has a the cell membranes were exposed to severe damage and some
higher bactericidal activity compared with rGO. The rGO sheets are completely losing their cytoplasm (Fig. 5A, B) [113]. This sequence
formed by the reduction of GO nanosheets, and the resultant size is of destruction was confirmed by a molecular dynamic simulation,
approximately nine times bigger than that of GO nanosheets. illustrating more details about the damaging mechanism. The Gr
Therefore they easily aggregate, leading to a lower antibacterial nanosheet starts the swing mode, in which it vibrates around the
activity of rGO particles. Dynamic simulation was applied to atom for a time of ten to a hundred nanoseconds. This is followed
investigate the interaction of Gr nanosheets with a lipid bilayer in by the insertion mode, where the sheet edge moves in and pierces
cell membrane, in terms of Gr thicknesses, e.g. single versus multi- the cell membranes as a result of the strong van der Waals in-
layered Gr [101]. It was found that single and few-layered Gr teractions with the membrane lipids and hydrophobic interface.
nanosheets can unexpectedly penetrate the cellular bilayer and Finally, the nanosheets strongly extract the phospholipids from the
orient themselves in parallel within the mid of the bilayer. lipid bilayers located on the cell surface during the extraction step.
These observations confirm that the insertion and lipid extrac-
3.6. Functionalization tion destructive mechanisms of Gr-based materials result in crucial
membrane stress, leading to a reduction in cell viability [113]. The
Adhesion for some bacterial species may be related to surface peak perturbation of the lipid bilayer by Gr sheets was observed for
chemistry [102], in which a surface charge is required owing to unoxidized and large nanosheets, in which the lipid bilayer is
ionization of the acidic/basic functional groups in their cell wall penetrated through the contact with the Gr sheet edges. These
[103]. Commonly, the bacterial cell wall has a negative charge in atomically thin sharp edges of the Gr nanosheets spontaneously
aqueous suspensions with neutral pH. Nevertheless, the level of the pierce the membrane, resulting in the reduction of the energy
charge differs from one species to another and is affected by several barrier required for membrane penetration. The negative charges of
factors, viz. the culture age, nutritional medium, ionic strength, cell the bacterial cells also play an important role in their interaction
surface structure and pH. The increased electronegativity of the cell with the sheet edges which act as good electron acceptors, and
wall causes the reduction of cellular adhesion on to the surface. therefore contribute toward bacterial cell membrane damage. In
Accordingly, several treatment or surface modification methods contrast, the hydrophilic nature of GO sheets result in them mostly
have recently been developed for the fabrication of altered bacte- attaching to the membrane surface without piercing the lipid
ricidal charged surfaces. These techniques involve functionalization bilayer [80]. The level of the cell membrane destruction by rGO is
[53,97,104e106], derivatization [107] and surface polymerization higher than that caused by the GO nanosheets, due to the abun-
[108,109], in which chemicals are used to change the surface fea- dance of sharp edges along with better charge transfer in the rGO
tures. The partially hydrophobic nature of GO surfaces along with nanosheets leading to further destruction of the bacterial cell
some hydrophilic regions [92e94] permit these surfaces to be membrane [63].
engaged in metal ion complexing and hydrogen bonding [110]. Furthermore, the effect of Gr nanosheets on bacterial cell
Moreover, GO sheets contain negatively charged edges accompa- membrane damage was studied by measuring the efflux of the
nied with carboxylate groups [111]. In contrast, only the defect sites intracellular components of the bacterial cells upon exposure to the
or edges of the hydrophobic Gr surface are able to biochemically nanosheets [63]. This was achieved by quantifying the RNA con-
react. rGO has an intermediary hydrophilic nature and reactivity, as centration in the medium. It was found to be high when the bac-
it contains basic defective spots due to oxygen removal during its terial cells were exposed to GO or rGO compared to the control
production [112]. sample. The interaction of the bacterial cells with the sharp edges of
the nanosheets resulted in higher destruction of the cell membrane
4. Antibacterial mechanisms of Gr-based materials of the Gram-positive bacteria, deficient in the external membrane,
compared to the Gram-negative ones having it [22]. Some bacterial
4.1. General mechanisms cells, e.g. P. aeruginosa, comprise of four layers of shielding mem-
branes [114], with an external membrane of a surface strongly
Bacterial cells could be eventually destroyed over time, when cross-linked lipopolysaccharide [115]. This leads to low absorbency
they come into contact with Gr-based materials, due to several of hydrophilic solutes, causing the resistance of the bacterial cells to
mechanisms, viz. membrane stress, oxidative stress, and/or wrap- several bactericidal agents.
ping isolation. These mechanisms can act separately or together to In the case of the stable suspension of Gr-based nano-
either bactericidally or bacteriostatically inhibit bacterial growth. composites, e.g. GO-Ag, GO sheets interact with bacterial cells, for
The three common antibacterial mechanisms of Gr-based materials instance, the resulting bactericidal activity of the GO-Ag nano-
are schematized in Fig. 4 and elucidated in the following sections. composites is observed via the interaction between bacteria and Ag
NPs supported on GO sheets. The bactericidal effect of Ag-GO
4.1.1. Bacterial membrane stress nanocomposites appears to be initiated by the “capturing-killing”
Cellular injury might occur from the impact of physical mem- mechanism that plays a role in the deposition of bacterial cells and
brane disruption that has been detected as the biological effects of increases their contact with the fabricated Ag NPs [116].
CNTs and fullerene [20]. Most E. coli cells lose cellular integrity and
tend to be flattened after incubation with rGO or GO dispersions 4.1.2. Oxidative stress
[16]. This demonstrates that bacterial cells can be irreversibly There are additional mechanisms related to the physicochemical
370 H.M. Hegab et al. / Carbon 105 (2016) 362e376

Fig. 4. Permanent and temporary bacterial cell growth inhibition mechanisms by Gr-based materials. (A colour version of this figure can be viewed online.)

Fig. 5. GO nanomaterial cellular destructive effect against E. coli cells; (A): TEM images of untreated cells, (B): Cells treated with GO nanosheets, (C): DNA fragmentation of cells
exposed to GO, rGO and Ag NPs. Reprinted with permission from Refs. [20,138].

interactions of Gr-based materials with bacteria which may have a cell membrane destruction and cellular growth inhibition [80]. This
significant role in their antibacterial activity. The key role of inhibition occurs via ROS generation, which might change the
oxidative stress has been proposed in the antibacterial activity of cellular redox condition. Oxidative stress as well as ROS production
GO. The action of oxidative stress on bacteria results in the oxida- were observed to be some of the main mechanisms in cellular
tion of lipids, nucleic acid and proteins which can eventually lead to defense when particles are absorbed. The oxidative stress could be
H.M. Hegab et al. / Carbon 105 (2016) 362e376 371

accelerated by NPs through the disturbance of the balance between biological simulation has verified that small Gr sheets
the intracellular oxidant and antioxidant processes [117,118], (~5.9  6.2 nm2) can be locked in bacterial cell membranes
occurring as a result of the disturbance of bacterial antioxidant comprising phospholipid components [125], with the possibility of
defense reactions (ROS-independent oxidative stress) [16,119]. The the some sheet internalization by bacterial cells. The different ac-
ROS usually includes the hydroxyl radical (OH), superoxide radical tivities of rGO and GO bring up the important role of Gr-based
(O2) and hydrogen peroxide (H2O2), that destroy cellular compo- materials dispersion/aggregation in their bactericidal effects [16].
nents, particularly proteins and DNA [119,120]. The ROS damaging
effect was confirmed by the irreversible DNA laddering occurring
upon the exposure of bacterial cells to GO (24 h) (Fig. 5C), while rGO 4.2. Gr-based suspension/membrane antibacterial capabilities
did not display the same effect [22]. This proposes that bacterial
cells need longer exposure time to rGO in order for DNA to be When GO nanosheets are immobilized on a coated surface, their
fragmented or otherwise the cell death mechanism by rGO could be interaction with bacteria may be considerably different than that in
unlike GO. This was further verified by a PDA-based experimental suspension form. The alterations in the GO nanosheets physico-
procedure, in which an atomic force microscope (AFM) probe was chemical features, e.g. sheet size, also may have a diverse effect
functionalized with GO nanosheets, and employed to screen the when employed on a surface [80]. This divergence proposes that
interaction between the bacterial cell membranes and GO [121]. the antibacterial effect of GO includes different mechanisms in the
The results showed that this interaction is mainly repulsive, with suspension phase compared to a GO-based coated surface. The
only infrequent attachment forces being monitored during probe inactivation of cell growth by huge nanosheets of GO suspension is
pull-off, which was attributed to lipopolysaccharide bridging. reversible, which elucidates that the GO suspensions/bacterial cell
The oxidative capacity of smaller GO sheets increase due to their interaction will not lead to cell growth inhibition as noticed for a
greater defect densities. Lipid peroxidation is thought to be the GO-based surface coating [80]. The reason behind this is the high
principal oxidative pathway resulting in the development of number of viable cells interacting with GO nanosheets in the bac-
oxidative stress in bacteria incubated with GO, owing to the adja- terial cell-GO aggregates. The effect of aggregation form on this
cent contact between GO nanosheets and bacterial cell membranes interaction is widely suggested to be orientation reliant [126e129].
[122]. Lipid peroxidation takes place as a chain reaction that is Cellular membrane destruction initiated by phospholipid drawing
started by the lipid molecules oxidation via ROS (Fig. 4), resulting in out or nanosheet piercing by GO is primarily caused by orthogonal
the formation of lipid peroxide radicals which initiate the cell interaction with GO nanosheet edges [127,128]. Alternatively, con-
membrane oxidative destruction [123]. tact with basal planes is required, through a quite anonymous
mechanism, for the GO antibacterial activity [126,129]. GO-coated
surfaces lessen orthogonal cell interaction with nanosheet edges
4.1.3. Wrapping isolation and improve contact with basal planes owing to flat sheet
Bacterial cells can be biologically isolated from their growth assembling.
medium when Gr sheets enclose them [124], and therefore they This was further confirmed by studies on the effect of the
cannot consume nutrients to grow, as permeability via the cell availability of the basal planes, compared to sheet edges, on the
membrane is blocked, leading to cell death [21]. GO nanosheet GO antibacterial features [126,129]. When the sheets are entirely
layers can also quarantine bacterial cells, even though they may flat on the surface and cells slightly interact with the sheet edges,
have greater permeability compared to Gr sheets (Fig. 4). It was GO nanosheets were still observed to inhibit bacteria that
revealed that the majority of E. coli cells were separately wrapped approach the surface [129]. These conclusions suggested that the
with GO nanosheets small layers, while they were typically interaction with the sheet edges, followed by direct penetration of
entrapped in aggregates of large rGO sheets [16]. Nevertheless, cell the cell membrane by GO nanosheets, is not necessary for the
inhibition by GO embedding was reversible and all the originally antibacterial effect of GO. Furthermore, the direct interaction be-
viable cells may possibly be recovered by their separation from GO tween GO and bacterial cells is required for efficient bactericidal
nanosheets via several techniques, e.g. sonication [80]. A molecular activity, as no effect on cell viability was detected for planktonic

Fig. 6. Schematic diagram illustrating the antibacterial strength of different Gr-based suspensions. (A colour version of this figure can be viewed online.)
372 H.M. Hegab et al. / Carbon 105 (2016) 362e376

growth not in direct contact with GO nanosheets, compared to or GO sheets in suspension may facilitate or impede the contact
bacterial cells adhered to all GO surfaces [16,130]. Briefly, bacterial with bacterial cells in a way of edgewise interaction. From Table 1
cell toxicity differs according to the orientation of the sheets the biocidal strength order of Gr-based nanomaterials dispersions
assembled on the surface. The sharp edges of rGO and GO nano- (Fig. 6) could be considered as follows: Gr hybrid NPs, GO and Gr.
sheets that are exposed to interaction with the bacterial cells While the antibacterial strength of Gr-based nanomaterial surfaces
induce membrane rupture, while the interfacial basal planes at the (Fig. 7) could be summarized in the order of Gr hybrid NPs, Gr and
surface showed no bactericidal activity. The orientation of the rGO GO.

Fig. 7. Schematic diagram illustrating the antibacterial strength of different Gr-based surfaces. (A colour version of this figure can be viewed online.)
H.M. Hegab et al. / Carbon 105 (2016) 362e376 373

5. Potential of Gr antiviral ability and inconsistent interpretations of the required Gr material fea-
tures for efficient antibacterial activity. The contradictory expla-
Despite the availability of attenuated and inactivated vaccines nations regarding the bactericidal properties of Gr-based materials
for some viral strains, e.g. pseudorabies virus (PRV) and porcine can be ascribed to the variations in the physicochemical features for
epidemic diarrhea virus (PEDV), these viral infections are still often each individual sample. The structural chemical and physical
detected, due to the development of new modified strains. The characteristics, viz. nanosheets thickness and size, dispersibility
absence of effective specific antiviral therapy for such viral diseases, and oxidation capacity, are distinctive, depending upon the Gt
highlights the urgent need to develop new antimicrobial agents. source and the preparation method (oxidation/exfoliation). Thus,
The antimicrobial ability of Gr material derivatives has been an accurate and comprehensive evaluation with regard to these
extended to cover viruses due to their great surface area and their factors is essential for prospective studies on the bactericidal
exceptional physical and chemical features, with less debatable properties of Gr-based materials. Moreover, the inconsistent
mode of action compared to that in bacteria [131,132]. The obvi- bactericidal outcomes of the suspension form and coated surfaces
ously low toxicity allowed Gr derivatives to be potential candidates highlight the need for a precise assessment of the antibacterial
for the future generation of antimicrobial compounds. activity of the different forms of Gr-based materials.
Researchers had been interested in the antiviral ability of Gr
derivatives, following different approaches, since 2012. A recent Acknowledgments
trial was performed by Sametband et al. (2014) who observed that
partially reduced sulfonated GO (rGO-SO3) and GO could deactivate Hanaa Hegab appreciatively acknowledges the supportive
herpes simplex virus (HSV-1) infection via cellular attachment at funding from the Australian government and University of South
small concentrations [133]. Both GO materials carry several Australia through an IPRS scholarship. Additionally, the authors
negative-charged groups, e.g. sulfonate and carboxyl, which are would like to acknowledge the support of Water Research Australia.
identical to heparan sulfate (HS) occurring on the cell surface,
resulting in the competition with the HS to bind HSV-1. Another
antiviral strategy was applied by Akhavan et al. (2012) in which Gr- References
tungsten oxide composite thin films were applied in the photo- [1] L. Zhao, H. Wang, K. Huo, L. Cui, W. Zhang, H. Ni, et al., Antibacterial nano-
inhibition of viruses under visible light irradiation [134]. Simi- structured titania coating incorporated with silver nanoparticles, Bio-
larly, Hu et al. (2012) employed a new photocatalyst, namely GO materials 32 (2011) 5706e5716, http://dx.doi.org/10.1016/
j.biomaterials.2011.04.040.
functioned aptamer, in which this photocatalyst was synthesized
[2] M. Zhang, K. Zhang, B. De Gusseme, W. Verstraete, Biogenic silver nano-
using the target recognition aptamer and the photosensitive GO. particles (bio-Ag 0) decrease biofouling of bio-Ag 0/PES nanocomposite
This photocatalyst efficiently inhibited viruses, and destructed their membranes, Water Res. 46 (2012) 2077e2087, http://dx.doi.org/10.1016/
proteins capsid along with their nucleic acids under irradiation. j.watres.2012.01.015.
[3] P. Prombutara, Y. Kulwatthanasal, N. Supaka, I. Sramala,
This resulted in the carbonylation of the proteins and oxidation of S. Chareonpornwattana, Production of nisin-loaded solid lipid nanoparticles
the nucleic acid bases, particularly guanosine, along with the for sustained antimicrobial activity, Food Control 24 (2012) 184e190, http://
reduction of GO [135]. Moreover, a GO-based material for the dx.doi.org/10.1016/j.foodcont.2011.09.025.
[4] M. Montazer, A. Shamei, F. Alimohammadi, Stabilized nanosilver loaded
intracellular transport of deoxyribozyme (Dz) was reported, in nylon knitted fabric using BTCA without yellowing, Prog. Org. Coatings 74
which it targets the mRNA of hepatitis C virus (HCV) to block the (2012) 270e276, http://dx.doi.org/10.1016/j.porgcoat.2012.01.003.
replication of HCV gene [136]. [5] L. Cheng, M.D. Weir, H.H.K. Xu, J.M. Antonucci, A.M. Kraigsley, N.J. Lin, et al.,
Antibacterial amorphous calcium phosphate nanocomposites with a qua-
The effective antiviral ability of both rGO and GO is due to the ternary ammonium dimethacrylate and silver nanoparticles, Dent. Mater. 28
distinctive single-layer configuration and negative charge, (2012) 561e572, http://dx.doi.org/10.1016/j.dental.2012.01.005.
compared to Gt and GtO which showed lesser antiviral activity, [6] M. Moritz, M. Geszke-Moritz, The newest achievements in synthesis,
immobilization and practical applications of antibacterial nanoparticles,
concluding that the nanosheet structure is vital for antiviral ability.
Chem. Eng. J. 228 (2013) 596e613, http://dx.doi.org/10.1016/
Moreover, GO inhibited viruses by mechanical destruction before j.cej.2013.05.046.
entering the viral structure. Nevertheless, the potential viral [7] A.K. Geim, K.S. Novoselov, The rise of graphene, Nat. Mater 6 (2007)
183e191, http://dx.doi.org/10.1038/nmat1849.
deactivation effects of Gr derivatives on different types of viruses
[8] D. Li, M.B. Müller, S. Gilje, R.B. Kaner, G.G. Wallace, Processable aqueous
have not been studied, along with their mode of action by which dispersions of graphene nanosheets, Nat. Nanotechnol. 3 (2008) 101e105,
they influence viruses [137]. http://dx.doi.org/10.1038/nnano.2007.451.
[9] S. Stankovich, D.A. Dikin, G.H.B. Dommett, K.M. Kohlhaas, E.J. Zimney,
E.A. Stach, et al., Graphene-based composite materials, Nature 442 (2006)
6. Concluding remarks 282e286, http://dx.doi.org/10.1038/nature04969.
[10] S. Jaworski, E. Sawosz, M. Grodzik, A. Winnicka, M. Prasek, M. Wierzbicki, et
The strong antibacterial activity of Gr-based materials against a al., In vitro evaluation of the effects of graphene platelets on glioblastoma
multiforme cells, Int. J. Nanomedicine 8 (2013) 413e420, http://dx.doi.org/
wide range of bacterial cells, including Gram negative and positive 10.2147/IJN.S39456.
strains, were demonstrated. These antibacterial properties are [11] K. Chaloupka, Y. Malam, A.M. Seifalian, Nanosilver as a new generation of
believed to be caused by chemical and physical interactions upon nanoproduct in biomedical applications, Trends Biotechnol. 28 (2010)
580e588, http://dx.doi.org/10.1016/j.tibtech.2010.07.006.
the direct contact of sheets with bacteria, in which the bacterial cell [12] V.C. Sanchez, A. Jachak, R.H. Hurt, A.B. Kane, Biological interactions of
membrane seems to be the main target of this activity. The resul- graphene-family nanomaterials: an interdisciplinary review, Chem. Res.
tant bacterial membrane destruction was recognized by morpho- Toxicol. 25 (2012) 15e34, http://dx.doi.org/10.1021/tx200339h.
[13] J. An, Y. Gou, C. Yang, F. Hu, C. Wang, Synthesis of a biocompatible gelatin
logical alterations in the cell structure, RNA leakage, release of functionalized graphene nanosheets and its application for drug delivery,
intracellular components and variations in the transmembrane Mater. Sci. Eng. C. Mater. Biol. Appl. 33 (2013) 2827e2837, http://dx.doi.org/
capacity. This damage is attributed to the atomic-based Gr sharp 10.1016/j.msec.2013.03.008.
[14] Y. Liu, D. Yu, C. Zeng, Z. Miao, L. Dai, Biocompatible graphene oxide-based
edges, which could pierce the bacterial cell membrane leading to a
glucose biosensors, Langmuir 26 (2010) 6158e6160, http://dx.doi.org/
physical disruption in its integrity. Cell membrane destruction 10.1021/la100886x.
could also be chemically mediated by lipid peroxidation initiated by [15] Y. Pan, N.G. Sahoo, L. Li, The application of graphene oxide in drug delivery,
the oxidative ability of GO. On the other hand, in spite of the sci- Expert Opin. Drug Deliv. 9 (2012) 1365e1376, http://dx.doi.org/10.1517/
17425247.2012.729575.
entific awareness for the importance of Gr-based materials [16] S. Liu, T.H. Zeng, M. Hofmann, E. Burcombe, J. Wei, R. Jiang, et al., Antibac-
employment in antibacterial surfaces, there is a limited knowledge terial activity of graphite, graphite oxide, graphene oxide, and reduced
374 H.M. Hegab et al. / Carbon 105 (2016) 362e376

graphene oxide: membrane and oxidative stress, ACS Nano 5 (2011) dx.doi.org/10.1016/j.apsusc.2015.03.082.
6971e6980, http://dx.doi.org/10.1021/nn202451x. [40] C.-H. Deng, J.-L. Gong, G.-M. Zeng, C.-G. Niu, Q.-Y. Niu, W. Zhang, et al.,
[17] O.N. Ruiz, K.A.S. Fernando, B. Wang, N.A. Brown, P.G. Luo, N.D. McNamara, et Inactivation performance and mechanism of Escherichia coli in aqueous
al., Graphene oxide: a nonspecific enhancer of cellular growth, ACS Nano 5 system exposed to iron oxide loaded graphene nanocomposites, J. Hazard.
(2011) 8100e8107, http://dx.doi.org/10.1021/nn202699t. Mater 276 (2014) 66e76, http://dx.doi.org/10.1016/j.jhazmat.2014.05.011.
[18] J.I. Paredes, S. Villar-Rodil, A. Martínez-Alonso, J.M.D. Tasco n, Graphene oxide [41] S. Bykkam, S. Narsingam, M. Ahmadipour, T. Dayakar, K. Venkateswara Rao,
dispersions in organic solvents, Langmuir 24 (2008) 10560e10564, http:// C. Shilpa Chakra, et al., Few layered graphene Sheet decorated by ZnO
dx.doi.org/10.1021/la801744a. Nanoparticles for anti-bacterial application, Superlattices Microstruct. 83
[19] D. Konios, M.M. Stylianakis, E. Stratakis, E. Kymakis, Dispersion behaviour of (2015) 776e784, http://dx.doi.org/10.1016/j.spmi.2015.03.063.
graphene oxide and reduced graphene oxide, J. Colloid Interface Sci. 430 [42] D. Wu, T. An, G. Li, W. Wang, Y. Cai, H.Y. Yip, et al., Mechanistic study of the
(2014) 108e112, http://dx.doi.org/10.1016/j.jcis.2014.05.033. visible-light-driven photocatalytic inactivation of bacteria by graphene oxi-
[20] W. Hu, C. Peng, W. Luo, M. Lv, X. Li, D. Li, et al., Graphene-based antibacterial deezinc oxide composite, Appl. Surf. Sci. 358 (2015) 137e145.
paper, ACS Nano 4 (2010) 4317e4323, http://dx.doi.org/10.1021/nn101097v. [43] S. Cao, C. Chen, J. Zhang, C. Zhang, W. Yu, B. Liang, et al., MnOx quantum dots
[21] S. Liu, M. Hu, T.H. Zeng, R. Wu, R. Jiang, J. Wei, et al., Lateral dimension- decorated reduced graphene oxide/TiO2 nanohybrids for enhanced activity
dependent antibacterial activity of graphene oxide sheets, Langmuir 28 by a UV pre-catalytic microwave method, Appl. Catal. B Environ. 176e177
(2012) 12364e12372, http://dx.doi.org/10.1021/la3023908. (2015) 500e512, http://dx.doi.org/10.1016/j.apcatb.2015.04.041.
[22] S. Gurunathan, J.W. Han, A.A. Dayem, V. Eppakayala, J.-H. Kim, Oxidative [44] S. Chella, P. Kollu, E.V.P.R. Komarala, S. Doshi, M. Saranya, S. Felix, et al.,
stress-mediated antibacterial activity of graphene oxide and reduced gra- Solvothermal synthesis of MnFe2O4-graphene compositedInvestigation of
phene oxide in Pseudomonas aeruginosa, Int. J. Nanomedicine 7 (2012) its adsorption and antimicrobial properties, Appl. Surf. Sci. 327 (2015)
5901e5914, http://dx.doi.org/10.2147/IJN.S37397. 27e36, http://dx.doi.org/10.1016/j.apsusc.2014.11.096.
[23] B.Z. Ristic, M.M. Milenkovic, I.R. Dakic, B.M. Todorovic-Markovic, [45] Y. Ouyang, X. Cai, Q. Shi, L. Liu, D. Wan, S. Tan, et al., Poly-l-lysine-modified
M.S. Milosavljevic, M.D. Budimir, et al., Photodynamic antibacterial effect of reduced graphene oxide stabilizes the copper nanoparticles with higher
graphene quantum dots, Biomaterials 35 (2014) 4428e4435, http:// water-solubility and long-term additively antibacterial activity, Colloids Surf.
dx.doi.org/10.1016/j.biomaterials.2014.02.014. B Biointerfaces 107 (2013) 107e114, http://dx.doi.org/10.1016/
[24] A.F. de Faria, D.S.T. Martinez, S.M.M. Meira, A.C.M. de Moraes, A. Brandelli, j.colsurfb.2013.01.073.
A.G.S. Filho, et al., Anti-adhesion and antibacterial activity of silver nano- [46] X. Wang, N. Zhou, W. Wang, Y. Tang, J. Zhang, J. Shen, The antimicrobial
particles supported on graphene oxide sheets, Colloids Surf. B. Biointerfaces properties of carboxylated graphene oxide decorated with La particles,
113 (2014) 115e124, http://dx.doi.org/10.1016/j.colsurfb.2013.08.006. Carbon N. Y. 52 (2013) 625, http://dx.doi.org/10.1016/j.carbon.2012.10.026.
[25] S. Vijay Kumar, N.M. Huang, H.N. Lim, A.R. Marlinda, I. Harrison, C.H. Chia, [47] T. Li, J. Shen, N. Li, M. Ye, Facile in situ synthesis of hydrophilic RGOeCDeAg
One-step size-controlled synthesis of functional graphene oxide/silver supramolecular hybrid and its enhanced antibacterial properties, Mater. Sci.
nanocomposites at room temperature, Chem. Eng. J. 219 (2013) 217e224, Eng. C 39 (2014) 352e358, http://dx.doi.org/10.1016/j.msec.2014.03.027.
http://dx.doi.org/10.1016/j.cej.2012.09.063. [48] C.-H. Deng, J.-L. Gong, G.-M. Zeng, Y. Jiang, C. Zhang, H.-Y. Liu, et al., Gra-
[26] Z. Zhu, M. Su, L. Ma, L. Ma, D. Liu, Z. Wang, Preparation of graphene oxide- pheneeCdS nanocomposite inactivation performance toward Escherichia
silver nanoparticle nanohybrids with highly antibacterial capability, coli in the presence of humic acid under visible light irradiation, Chem. Eng. J.
Talanta 117 (2013) 449e455, http://dx.doi.org/10.1016/ 284 (2016) 41e53, http://dx.doi.org/10.1016/j.cej.2015.08.106.
j.talanta.2013.09.017. [49] O. Akhavan, E. Ghaderi, Photocatalytic Reduction of Graphene Oxide Nano-
[27] M.R. Das, R.K. Sarma, R. Saikia, V.S. Kale, M.V. Shelke, P. Sengupta, Synthesis sheets on TiO 2 Thin Film for Photoinactivation of Bacteria in Solar Light
of silver nanoparticles in an aqueous suspension of graphene oxide sheets Irradiation, J. Phys. Chem. C 113 (2009) 20214e20220, http://dx.doi.org/
and its antimicrobial activity, Colloids Surf. B. Biointerfaces 83 (2011) 16e22, 10.1021/jp906325q.
http://dx.doi.org/10.1016/j.colsurfb.2010.10.033. [50] M. Mazaheri, O. Akhavan, A. Simchi, Flexible bactericidal graphene oxi-
[28] M.R. Das, R.K. Sarma, S.C. Borah, R. Kumari, R. Saikia, A.B. Deshmukh, et al., deechitosan layers for stem cell proliferation, Appl. Surf. Sci. 301 (2014)
The synthesis of citrate-modified silver nanoparticles in an aqueous sus- 456e462, http://dx.doi.org/10.1016/j.apsusc.2014.02.099.
pension of graphene oxide nanosheets and their antibacterial activity, Col- [51] Y. Liu, M. Park, H.K. Shin, B. Pant, J. Choi, Y.W. Park, et al., Facile preparation
loids Surf. B. Biointerfaces 105 (2013) 128e136, http://dx.doi.org/10.1016/ and characterization of poly(vinyl alcohol)/chitosan/graphene oxide bio-
j.colsurfb.2012.12.033. composite nanofibers, J. Ind. Eng. Chem. 20 (2014) 4415e4420, http://
[29] M.S. Haider, A.C. Badejo, G.N. Shao, S.M. Imran, N. Abbas, Y.G. Chai, et al., dx.doi.org/10.1016/j.jiec.2014.02.009.
Sequential repetitive chemical reduction technique to study size-property [52] R. Major, M. Sanak, A. Mzyk, L. Lipinska, M. Kot, P. Lacki, et al., Graphene
relationships of graphene attached Ag nanoparticle, Solid State Sci. 44 based porous coatings with antibacterial and antithrombogenous func-
(2015) 1e9, http://dx.doi.org/10.1016/j.solidstatesciences.2015.03.024. tiondMaterials and design, Arch. Civ. Mech. Eng. 14 (2014) 540e549, http://
[30] V.H. Nguyen, B.-K. Kim, Y.-L. Jo, J.-J. Shim, Preparation and antibacterial ac- dx.doi.org/10.1016/j.acme.2014.04.012.
tivity of silver nanoparticles-decorated graphene composites, J. Supercrit. [53] H.M. Hegab, A. ElMekawy, T.G. Barclay, A. Michelmore, L. Zou, C.P. Saint, et
Fluids 72 (2012) 28e35, http://dx.doi.org/10.1016/j.supflu.2012.08.005. al., Fine-Tuning the Surface of Forward Osmosis Membranes via Grafting
[31] Q. Bao, D. Zhang, P. Qi, Synthesis and characterization of silver nanoparticle Graphene Oxide: Performance Patterns and Biofouling Propensity, ACS Appl.
and graphene oxide nanosheet composites as a bactericidal agent for water Mater. Interfaces 7 (2015) 18004e18016, http://dx.doi.org/10.1021/
disinfection, J. Colloid Interface Sci. 360 (2011) 463e470, http://dx.doi.org/ acsami.5b04818.
10.1016/j.jcis.2011.05.009. [54] C. Bora, P. Bharali, S. Baglari, S.K. Dolui, B.K. Konwar, Strong and conductive
[32] J. Shen, T. Li, M. Shi, N. Li, M. Ye, Polyelectrolyte-assisted one-step hydro- reduced graphene oxide/polyester resin composite films with improved
thermal synthesis of Ag-reduced graphene oxide composite and its anti- mechanical strength, thermal stability and its antibacterial activity, Compos.
bacterial properties, Mater. Sci. Eng. C 32 (2012) 2042e2047, http:// Sci. Technol. 87 (2013) 1e7, http://dx.doi.org/10.1016/
dx.doi.org/10.1016/j.msec.2012.05.017. j.compscitech.2013.07.025.
[33] X. Cai, M. Lin, S. Tan, W. Mai, Y. Zhang, Z. Liang, et al., The use of [55] L. Duan, Y. Wang, Y. Zhang, J. Liu, Graphene immobilized enzyme/poly-
polyethyleneimine-modified reduced graphene oxide as a substrate for sil- ethersulfone mixed matrix membrane: Enhanced antibacterial, permeable
ver nanoparticles to produce a material with lower cytotoxicity and long- and mechanical properties, Appl. Surf. Sci. 355 (2015) 436e445, http://
term antibacterial activity, Carbon N. Y. 50 (2012) 3407e3415, http:// dx.doi.org/10.1016/j.apsusc.2015.07.127.
dx.doi.org/10.1016/j.carbon.2012.02.002. [56] L. He, L.F. Dume e, C. Feng, L. Velleman, R. Reis, F. She, et al., Promoted water
[34] H. Zhou, Y. Liu, W. Chi, C. Yu, Y. Yu, Preparation and antibacterial properties transport across graphene oxideepoly(amide) thin film composite mem-
of Ag@polydopamine/graphene oxide sheet nanocomposite, Appl. Surf. Sci. branes and their antibacterial activity, Desalination 365 (2015) 126e135,
282 (2013) 181e185, http://dx.doi.org/10.1016/j.apsusc.2013.05.099. http://dx.doi.org/10.1016/j.desal.2015.02.032.
[35] Z. Tai, H. Ma, B. Liu, X. Yan, Q. Xue, Facile synthesis of Ag/GNS-g-PAA [57] Z.-B. Zhang, J.-J. Wu, Y. Su, J. Zhou, Y. Gao, H.-Y. Yu, et al., Layer-by-layer
nanohybrids for antimicrobial applications, Colloids Surf. B Biointerfaces 89 assembly of graphene oxide on polypropylene macroporous membranes via
(2012) 147e151, http://dx.doi.org/10.1016/j.colsurfb.2011.09.006. click chemistry to improve antibacterial and antifouling performance, Appl.
[36] L. Liu, H. Bai, J. Liu, D.D. Sun, Multifunctional graphene oxide-TiO2-Ag Surf. Sci. 332 (2015) 300e307, http://dx.doi.org/10.1016/
nanocomposites for high performance water disinfection and decontami- j.apsusc.2015.01.193.
nation under solar irradiation, J. Hazard. Mater 261 (2013) 214e223, http:// [58] L. Yu, Y. Zhang, B. Zhang, J. Liu, H. Zhang, C. Song, Preparation and charac-
dx.doi.org/10.1016/j.jhazmat.2013.07.034. terization of HPEI-GO/PES ultrafiltration membrane with antifouling and
[37] M.K. Joshi, H.R. Pant, H.J. Kim, J.H. Kim, C.S. Kim, One-pot synthesis of Ag-iron antibacterial properties, J. Memb. Sci. 447 (2013) 452e462, http://dx.doi.org/
oxide/reduced graphene oxide nanocomposite via hydrothermal treatment, 10.1016/j.memsci.2013.07.042.
Colloids Surf. A Physicochem. Eng. Asp. 446 (2014) 102e108, http:// [59] V. Vatanpour, A. Shockravi, H. Zarrabi, Z. Nikjavan, A. Javadi, Fabrication and
dx.doi.org/10.1016/j.colsurfa.2014.01.058. characterization of anti-fouling and anti-bacterial Ag-loaded graphene ox-
[38] T. Kavitha, A.I. Gopalan, K.-P. Lee, S.-Y. Park, Glucose sensing, photocatalytic ide/polyethersulfone mixed matrix membrane, J. Ind. Eng. Chem. 30 (2015)
and antibacterial properties of grapheneeZnO nanoparticle hybrids, Carbon 342e352, http://dx.doi.org/10.1016/j.jiec.2015.06.004.
N. Y. 50 (2012) 2994e3000, http://dx.doi.org/10.1016/j.carbon.2012.02.082. [60] X.-F. Sun, J. Qin, P.-F. Xia, B.-B. Guo, C.-M. Yang, C. Song, et al., Graphene
[39] Y.-N. Chang, X.-M. Ou, G.-M. Zeng, J.-L. Gong, C.-H. Deng, Y. Jiang, et al., oxideesilver nanoparticle membrane for biofouling control and water pu-
Synthesis of magnetic graphene oxideeTiO2 and their antibacterial proper- rification, Chem. Eng. J. 281 (2015) 53e59, http://dx.doi.org/10.1016/
ties under solar irradiation, Appl. Surf. Sci. 343 (2015) 1e10, http:// j.cej.2015.06.059.
H.M. Hegab et al. / Carbon 105 (2016) 362e376 375

[61] E. Mahmoudi, L.Y. Ng, M.M. Ba-Abbad, A.W. Mohammad, Novel nanohybrid J. Parthenios, et al., Deformation of wrinkled graphene, ACS Nano 9 (2015)
polysulfone membrane embedded with silver nanoparticles on graphene 3917e3925, http://dx.doi.org/10.1021/nn507202c.
oxide nanoplates, Chem. Eng. J. 277 (2015) 1e10, http://dx.doi.org/10.1016/ [85] A.E. Nel, L. Ma€dler, D. Velegol, T. Xia, E.M.V. Hoek, P. Somasundaran, et al.,
j.cej.2015.04.107. Understanding biophysicochemical interactions at the nano-bio interface,
[62] A. Jankovi c, S. Erakovi c, M. Vukasinovi c-Sekuli
c, V. Miskovi c-Stankovic, Nat. Mater 8 (2009) 543e557, http://dx.doi.org/10.1038/nmat2442.
S.J. Park, K.Y. Rhee, Graphene-based antibacterial composite coatings elec- [86] C.N.R. Rao, K.S. Subrahmanyam, H.S.S.R. Matte, A. Govindaraj, synthesis,
trodeposited on titanium for biomedical applications, Prog. Org. Coatings 83 functionalization and properties, in: S.K. Pati (Ed.), Graphene its Fascin,
(2015) 1e10, http://dx.doi.org/10.1016/j.porgcoat.2015.01.019. World Scientific, 2011, p. 12.
[63] O. Akhavan, E. Ghaderi, Toxicity of graphene and graphene oxide nanowalls [87] M. Taghioskoui, Trends in graphene research, Mater. Today 12 (2009) 34e37,
against bacteria, ACS Nano 4 (2010) 5731e5736, http://dx.doi.org/10.1021/ http://dx.doi.org/10.1016/S1369-7021(09)70274-3.
nn101390x. [88] A. Terada, A. Yuasa, S. Tsuneda, A. Hirata, A. Katakai, M. Tamada, Elucidation
e, N. Reckinger, C. Didembourg, J.-J. Letesson, M. Sarrazin,
[64] L. Dellieu, E. Laware of dominant effect on initial bacterial adhesion onto polymer surfaces pre-
et al., Do CVD grown graphene films have antibacterial activity on metallic pared by radiation-induced graft polymerization, Colloids Surf. B. Bio-
substrates? Carbon N. Y. 84 (2015) 310e316, http://dx.doi.org/10.1016/ interfaces 43 (2005) 99e107, http://dx.doi.org/10.1016/
j.carbon.2014.12.025. j.colsurfb.2005.03.016.
[65] Y. Wang, P. Zhang, C.F. Liu, C.Z. Huang, A facile and green method to fabricate [89] O. Habimana, A.J.C. Semia ~o, E. Casey, The role of cell-surface interactions in
graphene-based multifunctional hydrogels for miniature-scale water puri- bacterial initial adhesion and consequent biofilm formation on nano-
fication, RSC Adv. 3 (2013) 9240, http://dx.doi.org/10.1039/c3ra22687e. filtration/reverse osmosis membranes, J. Memb. Sci. 454 (2014) 82e96,
[66] X. Wang, Z. Liu, X. Ye, K. Hu, H. Zhong, X. Yuan, et al., A facile one-pot method http://dx.doi.org/10.1016/j.memsci.2013.11.043.
to two kinds of graphene oxide-based hydrogels with broad-spectrum [90] E.P. Ivanova, J. Hasan, H.K. Webb, V.K. Truong, G.S. Watson, J.A. Watson, et al.,
antimicrobial properties, Chem. Eng. J. 260 (2015) 331e337, http:// Natural bactericidal surfaces: mechanical rupture of Pseudomonas aerugi-
dx.doi.org/10.1016/j.cej.2014.08.102. nosa cells by cicada wings, Small 8 (2012) 2489e2494, http://dx.doi.org/
[67] R. Surudzi c, A. Jankovi c, N. Bibi
c, M. Vukasinovic-Sekuli c, A. Peri
c-Gruji
c, 10.1002/smll.201200528.
V. Miskovi c-Stankovic, et al., Physico-chemical and mechanical properties [91] Russell J. Crawford, Elena P. Ivanova, Shannon M. Notley, Bacterial Interac-
and antibacterial activity of silver/poly(vinyl alcohol)/graphene nano- tion with Graphene Particles and Surfaces, in: M. Aliofkhazraei (Ed.), Adv.
composites obtained by electrochemical method, Compos. Part B Eng. 85 Graphene Sci, InTech, 2013, pp. 99e118, http://dx.doi.org/10.5772/51689.
(2015) 102e112. [92] S.A. Hasan, J.L. Rigueur, R.R. Harl, A.J. Krejci, I. Gonzalo-Juan, B.R. Rogers, et
[68] Z. Fan, B. Liu, J. Wang, S. Zhang, Q. Lin, P. Gong, et al., A Novel Wound al., Transferable graphene oxide films with tunable microstructures, ACS
Dressing Based on Ag/Graphene Polymer Hydrogel: Effectively Kill Bacteria Nano 4 (2010) 7367e7372, http://dx.doi.org/10.1021/nn102152x.
and Accelerate Wound Healing, Adv. Funct. Mater 24 (2014) 3933e3943, [93] H.M. Hegab, L. Zou, Graphene oxide-assisted membranes: Fabrication and
http://dx.doi.org/10.1002/adfm.201304202. potential applications in desalination and water purification, J. Memb. Sci.
[69] E.C. Salas, Z. Sun, A. Lüttge, J.M. Tour, Reduction of graphene oxide via bac- 484 (2015) 95e106, http://dx.doi.org/10.1016/j.memsci.2015.03.011.
terial respiration, ACS Nano 4 (2010) 4852e4856, http://dx.doi.org/10.1021/ [94] C.-T. Hsieh, W.-Y. Chen, Water/oil repellency and work of adhesion of liquid
nn101081t. droplets on graphene oxide and graphene surfaces, Surf. Coatings Technol.
[70] Y.-C. Yong, Y.-Y. Yu, X. Zhang, H. Song, Highly active bidirectional electron 205 (2011) 4554e4561, http://dx.doi.org/10.1016/j.surfcoat.2011.03.128.
transfer by a self-assembled electroactive reduced-graphene-oxide- [95] G. Liu, Y. Chen, G. Zhang, S. Yang, Protein resistance of (ethylene oxide)n
hybridized biofilm, Angew. Chem. Int. Ed. Engl. 53 (2014) 4480e4483, monolayers at the air/water interface: effects of packing density and chain
http://dx.doi.org/10.1002/anie.201400463. length, Phys. Chem. Chem. Phys. 9 (2007) 6073e6082, http://dx.doi.org/
[71] G. Wang, F. Qian, C.W. Saltikov, Y. Jiao, Y. Li, Microbial reduction of graphene 10.1039/b711037e.
oxide by Shewanella, Nano Res. 4 (2011) 563e570, http://dx.doi.org/ [96] T.R. Garrett, M. Bhakoo, Z. Zhang, Bacterial adhesion and biofilms on surfaces,
10.1007/s12274-011-0112-2. Prog. Nat. Sci. 18 (2008) 1049e1056, http://dx.doi.org/10.1016/
[72] H. Zhang, X. Yu, D. Guo, B. Qu, M. Zhang, Q. Li, et al., Synthesis of bacteria j.pnsc.2008.04.001.
promoted reduced graphene oxide-nickel sulfide networks for advanced [97] H.M. Hegab, Y. Wimalasiri, M. Ginic-Markovic, L. Zou, Improving the fouling
supercapacitors, ACS Appl. Mater. Interfaces 5 (2013) 7335e7340, http:// resistance of brackish water membranes via surface modification with gra-
dx.doi.org/10.1021/am401680m. phene oxide functionalized chitosan, Desalination 365 (2015) 99e107,
[73] S. Raveendran, N. Chauhan, Y. Nakajima, H. Toshiaki, S. Kurosu, Y. Tanizawa, http://dx.doi.org/10.1016/j.desal.2015.02.029.
et al., Ecofriendly Route for the Synthesis of Highly Conductive Graphene [98] E. Behzadfar, M.H. Abdolrasouli, F. Sharif, H. Nazockdast, Effect of solid
Using Extremophiles for Green Electronics and Bioscience, Part. Part. Syst. loading and aggregate size on the rheological behavior of PDMS/Calcium
Charact. 30 (2013) 573e578, http://dx.doi.org/10.1002/ppsc.201200126. Carbonate suspensions, Braz. J. Chem. Eng. 26 (2009) 713e721, http://
[74] A.G. Nandgaonkar, Q. Wang, K. Fu, W.E. Krause, Q. Wei, R. Gorga, et al., A one- dx.doi.org/10.1590/S0104-66322009000400010.
pot biosynthesis of reduced graphene oxide (RGO)/bacterial cellulose (BC) [99] R.-Q. Song, H. Co €lfen, Mesocrystalseordered nanoparticle superstructures,
nanocomposites, Green Chem. 16 (2014) 3195, http://dx.doi.org/10.1039/ Adv. Mater 22 (2010) 1301e1330, http://dx.doi.org/10.1002/
c4gc00264d. adma.200901365.
[75] O. Akhavan, E. Ghaderi, Escherichia coli bacteria reduce graphene oxide to [100] S. Stankovich, D.A. Dikin, R.D. Piner, K.A. Kohlhaas, A. Kleinhammes, Y. Jia, et
bactericidal graphene in a self-limiting manner, Carbon N. Y. 50 (2012) al., Synthesis of graphene-based nanosheets via chemical reduction of
1853e1860, http://dx.doi.org/10.1016/j.carbon.2011.12.035. exfoliated graphite oxide, Carbon N. Y. 45 (2007) 1558e1565, http://
[76] S. Gurunathan, J.W. Han, V. Eppakayala, J.-H. Kim, Microbial reduction of dx.doi.org/10.1016/j.carbon.2007.02.034.
graphene oxide by Escherichia coli: a green chemistry approach, Colloids [101] J. Wang, Y. Wei, X. Shi, H. Gao, Cellular entry of graphene nanosheets: the
Surf. B. Biointerfaces 102 (2013) 772e777, http://dx.doi.org/10.1016/ role of thickness, oxidation and surface adsorption, RSC Adv. 3 (2013) 15776,
j.colsurfb.2012.09.011. http://dx.doi.org/10.1039/c3ra40392k.
[77] E.C. Salas, Z. Sun, A. Lüttge, J.M. Tour, Reduction of graphene oxide via bac- [102] L.-F. Liu, S.-C. Yu, L.-G. Wu, C.-J. Gao, Study on a novel polyamide-urea
terial respiration, ACS Nano 4 (2010) 4852e4856, http://dx.doi.org/10.1021/ reverse osmosis composite membrane (ICIC-MPD), J. Memb. Sci. 283
nn101081t. (2006) 133e146, http://dx.doi.org/10.1016/j.memsci.2006.06.033.
[78] S. Gurunathan, J.W. Han, V. Eppakayala, J.-H. Kim, Biocompatibility of [103] P.M. Biesheuvel, M. van der Veen, W. Norde, A modified Poisson-Boltzmann
microbially reduced graphene oxide in primary mouse embryonic fibroblast model including charge regulation for the adsorption of ionizable poly-
cells, Colloids Surf. B. Biointerfaces 105 (2013) 58e66, http://dx.doi.org/ electrolytes to charged interfaces, applied to lysozyme adsorption on silica,
10.1016/j.colsurfb.2012.12.036. J. Phys. Chem. B 109 (2005) 4172e4180, http://dx.doi.org/10.1021/
[79] M. Dallavalle, M. Calvaresi, A. Bottoni, M. Melle-Franco, F. Zerbetto, Graphene jp0463823.
can wreak havoc with cell membranes, ACS Appl. Mater. Interfaces 7 (2015) [104] D. Low, A. Hamood, T. Reid, T. Mosley, P. Tran, L. Song, et al., Attachment of
4406e4414, http://dx.doi.org/10.1021/am508938u. selenium to a reverse osmosis membrane to inhibit biofilm formation of
[80] F. Perreault, A.F. de Faria, S. Nejati, M. Elimelech, Antimicrobial Properties of S. aureus, J. Memb. Sci. 378 (2011) 171e178, http://dx.doi.org/10.1016/
Graphene Oxide Nanosheets: Why Size Matters, ACS Nano 9 (2015) j.memsci.2011.04.041.
7226e7236, http://dx.doi.org/10.1021/acsnano.5b02067. [105] D. Saeki, S. Nagao, I. Sawada, Y. Ohmukai, T. Maruyama, H. Matsuyama,
[81] H. Yue, W. Wei, Z. Yue, B. Wang, N. Luo, Y. Gao, et al., The role of the lateral Development of antibacterial polyamide reverse osmosis membrane modi-
dimension of graphene oxide in the regulation of cellular responses, Bio- fied with a covalently immobilized enzyme, J. Memb. Sci. 428 (2013)
materials 33 (2012) 4013e4021, http://dx.doi.org/10.1016/ 403e409, http://dx.doi.org/10.1016/j.memsci.2012.10.038.
j.biomaterials.2012.02.021. [106] H.-L. Yang, J.C.-T. Lin, C. Huang, Application of nanosilver surface modifica-
[82] Y. Li, H. Yuan, A. von dem Bussche, M. Creighton, R.H. Hurt, A.B. Kane, et al., tion to RO membrane and spacer for mitigating biofouling in seawater
Graphene microsheets enter cells through spontaneous membrane pene- desalination, Water Res. 43 (2009) 3777e3786, http://dx.doi.org/10.1016/
tration at edge asperities and corner sites, Proc. Natl. Acad. Sci. U. S. A. 110 j.watres.2009.06.002.
(2013) 12295e12300, http://dx.doi.org/10.1073/pnas.1222276110. [107] X. Wei, Z. Wang, J. Chen, J. Wang, S. Wang, A novel method of surface
[83] S. Gurunathan, J.W. Han, J.-H. Kim, Green chemistry approach for the syn- modification on thin-film-composite reverse osmosis membrane by grafting
thesis of biocompatible graphene, Int. J. Nanomedicine 8 (2013) 2719e2732, hydantoin derivative, J. Memb. Sci. 346 (2010) 152e162, http://dx.doi.org/
http://dx.doi.org/10.2147/IJN.S45174. 10.1016/j.memsci.2009.09.032.
[84] Z. Li, I.A. Kinloch, R.J. Young, K.S. Novoselov, G. Anagnostopoulos, [108] R. Bernstein, S. Belfer, V. Freger, Bacterial attachment to RO membranes
376 H.M. Hegab et al. / Carbon 105 (2016) 362e376

surface-modified by concentration-polarization-enhanced graft polymeri- penetration at edge asperities and corner sites, Proc. Natl. Acad. Sci. U. S. A.
zation, Environ. Sci. Technol. 45 (2011) 5973e5980, http://dx.doi.org/ 110 (2013) 12295e12300, http://dx.doi.org/10.1073/pnas.1222276110.
10.1021/es1043694. [129] J.D. Mangadlao, C.M. Santos, M.J.L. Felipe, A.C.C. de Leon, D.F. Rodrigues,
[109] Y.-C. Chiang, Y. Chang, C.-J. Chuang, R.-C. Ruaan, A facile zwitterionization in R.C. Advincula, On the antibacterial mechanism of graphene oxide (GO)
the interfacial modification of low bio-fouling nanofiltration membranes, Langmuir-Blodgett films, Chem. Commun. (Camb) 51 (2015) 2886e2889,
J. Memb. Sci. 389 (2012) 76e82, http://dx.doi.org/10.1016/ http://dx.doi.org/10.1039/c4cc07836e.
j.memsci.2011.10.017. [130] O. Akhavan, E. Ghaderi, Toxicity of graphene and graphene oxide nanowalls
[110] S.-T. Yang, Y. Chang, H. Wang, G. Liu, S. Chen, Y. Wang, et al., Folding/ag- against bacteria, ACS Nano 4 (2010) 5731e5736, http://dx.doi.org/10.1021/
gregation of graphene oxide and its application in Cu2þ removal, J. Colloid nn101390x.
Interface Sci. 351 (2010) 122e127, http://dx.doi.org/10.1016/ [131] H.Y. Mao, S. Laurent, W. Chen, O. Akhavan, M. Imani, A.A. Ashkarran, et al.,
j.jcis.2010.07.042. Graphene: promises, facts, opportunities, and challenges in nanomedicine,
[111] L.J. Cote, F. Kim, J. Huang, Langmuir-Blodgett assembly of graphite oxide Chem. Rev. 113 (2013) 3407e3424, http://dx.doi.org/10.1021/cr300335p.
single layers, J. Am. Chem. Soc. 131 (2009) 1043e1049, http://dx.doi.org/ [132] Z. Xiu, Q. Zhang, H.L. Puppala, V.L. Colvin, P.J.J. Alvarez, Negligible particle-
10.1021/ja806262m. specific antibacterial activity of silver nanoparticles, Nano Lett. 12 (2012)
[112] A. Bagri, C. Mattevi, M. Acik, Y.J. Chabal, M. Chhowalla, V.B. Shenoy, Struc- 4271e4275, http://dx.doi.org/10.1021/nl301934w.
tural evolution during the reduction of chemically derived graphene oxide, [133] M. Sametband, I. Kalt, A. Gedanken, R. Sarid, Herpes simplex virus type-1
Nat. Chem. 2 (2010) 581e587, http://dx.doi.org/10.1038/nchem.686. attachment inhibition by functionalized graphene oxide, ACS Appl. Mater.
[113] Y. Tu, M. Lv, P. Xiu, T. Huynh, M. Zhang, M. Castelli, et al., Destructive Interfaces 6 (2014) 1228e1235, http://dx.doi.org/10.1021/am405040z.
extraction of phospholipids from Escherichia coli membranes by graphene [134] O. Akhavan, M. Choobtashani, E. Ghaderi, Protein Degradation and RNA
nanosheets, Nat. Nanotechnol. 8 (2013) 594e601, http://dx.doi.org/10.1038/ Efflux of Viruses Photocatalyzed by GrapheneeTungsten Oxide Composite
nnano.2013.125. Under Visible Light Irradiation, J. Phys. Chem. C 116 (2012) 9653e9659,
[114] N.M. Huang, H.N. Lim, S. Radiman, P.S. Khiew, W.S. Chiu, R. Hashim, et al., http://dx.doi.org/10.1021/jp301707m.
Sucrose ester micellar-mediated synthesis of Ag nanoparticles and the [135] X. Hu, L. Mu, J. Wen, Q. Zhou, Covalently synthesized graphene oxide-
antibacterial properties, Colloids Surf. A Physicochem. Eng. Asp. 353 (2010) aptamer nanosheets for efficient visible-light photocatalysis of nucleic
69e76, http://dx.doi.org/10.1016/j.colsurfa.2009.10.023. acids and proteins of viruses, Carbon N. Y. 50 (2012) 2772e2781, http://
[115] H.N. Lim, N.M. Huang, C.H. Loo, Facile preparation of graphene-based chi- dx.doi.org/10.1016/j.carbon.2012.02.038.
tosan films: Enhanced thermal, mechanical and antibacterial properties, [136] S. Kim, S.-R. Ryoo, H.-K. Na, Y.-K. Kim, B.-S. Choi, Y. Lee, et al., Deoxy-
J. Non. Cryst. Solids 358 (2012) 525e530, http://dx.doi.org/10.1016/ ribozyme-loaded nano-graphene oxide for simultaneous sensing and
j.jnoncrysol.2011.11.007. silencing of the hepatitis C virus gene in liver cells, Chem. Commun. (Camb)
[116] W.-P. Xu, L.-C. Zhang, J.-P. Li, Y. Lu, H.-H. Li, Y.-N. Ma, et al., Facile synthesis of 49 (2013) 8241e8243, http://dx.doi.org/10.1039/c3cc43368d.
silver@graphene oxide nanocomposites and their enhanced antibacterial [137] S. Ye, K. Shao, Z. Li, N. Guo, Y. Zuo, Q. Li, et al., Antiviral Activity of Graphene
properties, J. Mater. Chem. 21 (2011) 4593, http://dx.doi.org/10.1039/ Oxide: How Sharp Edged Structure and Charge Matter, ACS Appl. Mater.
c0jm03376f. Interfaces 7 (2015) 21571e21579, http://dx.doi.org/10.1021/
[117] S. Kang, M. Herzberg, D.F. Rodrigues, M. Elimelech, Antibacterial effects of acsami.5b06876.
carbon nanotubes: size does matter!, Langmuir 24 (2008) 6409e6413, [138] S. Gurunathan, J.W. Han, A.A. Dayem, V. Eppakayala, M.-R. Park, D.-N. Kwon,
http://dx.doi.org/10.1021/la800951v. et al., Antibacterial activity of dithiothreitol reduced graphene oxide, J. Ind.
[118] A. Nel, T. Xia, L. M€ adler, N. Li, Toxic potential of materials at the nanolevel, Eng. Chem. 19 (2013) 1280e1288, http://dx.doi.org/10.1016/
Science (80-. ) 311 (2006) 622e627, http://dx.doi.org/10.1126/ j.jiec.2012.12.029.
science.1114397. [139] D. Zhang, X. Liu, X. Wang, Green synthesis of graphene oxide sheets deco-
[119] K.A. Foster, F. Galeffi, F.J. Gerich, D.A. Turner, M. Müller, Optical and phar- rated by silver nanoprisms and their anti-bacterial properties, J. Inorg. Bio-
macological tools to investigate the role of mitochondria during oxidative chem. 105 (2011) 1181e1186, http://dx.doi.org/10.1016/
stress and neurodegeneration, Prog. Neurobiol. 79 (2006) 136e171, http:// j.jinorgbio.2011.05.014.
dx.doi.org/10.1016/j.pneurobio.2006.07.001. [140] B. Pant, P. Pokharel, A.P. Tiwari, P.S. Saud, M. Park, Z.K. Ghouri, et al., Char-
[120] E.-J. Park, J. Choi, Y.-K. Park, K. Park, Oxidative stress induced by cerium oxide acterization and antibacterial properties of aminophenol grafted and Ag NPs
nanoparticles in cultured BEAS-2B cells, Toxicology 245 (2008) 90e100, decorated graphene nanocomposites, Ceram. Int. 41 (2015) 5656e5662,
http://dx.doi.org/10.1016/j.tox.2007.12.022. http://dx.doi.org/10.1016/j.ceramint.2014.12.150.
[121] S. Romero-Vargas Castrillo n, F. Perreault, A.F. de Faria, M. Elimelech, Inter- [141] C. Chen, S. Cao, W. Yu, X. Xie, Q. Liu, Y. Tsang, et al., Adsorption, photo-
action of Graphene Oxide with Bacterial Cell Membranes: Insights from catalytic and sunlight-driven antibacterial activity of Bi2WO6/graphene
Force Spectroscopy, Environ. Sci. Technol. Lett. 2 (2015) 112e117, http:// oxide nanoflakes, Vacuum 116 (2015) 48e53, http://dx.doi.org/10.1016/
dx.doi.org/10.1021/acs.estlett.5b00066. j.vacuum.2015.02.031.
[122] K. Krishnamoorthy, M. Veerapandian, L.-H. Zhang, K. Yun, S.J. Kim, Anti- [142] P. Li, S. Sun, A. Dong, Y. Hao, S. Shi, Z. Sun, et al., Developing of a novel
bacterial Efficiency of Graphene Nanosheets against Pathogenic Bacteria via antibacterial agent by functionalization of graphene oxide with guanidine
Lipid Peroxidation, J. Phys. Chem. C 116 (2012) 17280e17287, http:// polymer with enhanced antibacterial activity, Appl. Surf. Sci. 355 (2015)
dx.doi.org/10.1021/jp3047054. 446e452, http://dx.doi.org/10.1016/j.apsusc.2015.07.148.
[123] X. Wang, P.J. Quinn, Vitamin E and its function in membranes, Prog. Lipid [143] V. Poornima Parvathi, M. Umadevi, R. Bhaviya Raj, Improved waste water
Res. 38 (1999) 309e336. http://www.ncbi.nlm.nih.gov/pubmed/10793887 treatment by bio-synthesized Graphene Sand Composite, J. Environ. Manage
(accessed 10.11.15). 162 (2015) 299e305, http://dx.doi.org/10.1016/j.jenvman.2015.07.055.
[124] O. Akhavan, E. Ghaderi, A. Esfandiar, Wrapping bacteria by graphene nano- [144] M. Haniff Wahid, U.H. Stroeher, E. Eroglu, X. Chen, K. Vimalanathan,
sheets for isolation from environment, reactivation by sonication, and C.L. Raston, et al., Aqueous based synthesis of antimicrobial-decorated gra-
inactivation by near-infrared irradiation, J. Phys. Chem. B 115 (2011) phene, J. Colloid Interface Sci. 443 (2015) 88e96, http://dx.doi.org/10.1016/
6279e6288, http://dx.doi.org/10.1021/jp200686k. j.jcis.2014.11.043.
[125] A.V. Titov, P. Kr al, R. Pearson, Sandwiched grapheneemembrane super- [145] A. Soroush, W. Ma, M. Cyr, M.S. Rahaman, B. Asadishad, N. Tufenkji, Situ
structures, ACS Nano 4 (2010) 229e234, http://dx.doi.org/10.1021/ Silver Decoration on Graphene Oxide-Treated Thin Film Composite Forward
nn9015778. Osmosis Membranes: Biocidal Properties and Regeneration Potential, Envi-
[126] L. Hui, J.-G. Piao, J. Auletta, K. Hu, Y. Zhu, T. Meyer, et al., Availability of the ron. Sci. Technol. Lett. 3 (2016) 13e18, http://dx.doi.org/10.1021/
basal planes of graphene oxide determines whether it is antibacterial, ACS acs.estlett.5b00304.
Appl. Mater. Interfaces 6 (2014) 13183e13190, http://dx.doi.org/10.1021/ [146] F. Perreault, M.E. Tousley, M. Elimelech, Thin-Film Composite Polyamide
am503070z. Membranes Functionalized with Biocidal Graphene Oxide Nanosheets, En-
[127] Y. Tu, M. Lv, P. Xiu, T. Huynh, M. Zhang, M. Castelli, et al., Destructive viron. Sci. Technol. Lett. 1 (2014) 71e76, http://dx.doi.org/10.1021/
extraction of phospholipids from Escherichia coli membranes by graphene ez4001356.
nanosheets, Nat. Nanotechnol. 8 (2013) 594e601, http://dx.doi.org/10.1038/ [147] A. Nourmohammadi, R. Rahighi, O. Akhavan, A. Moshfegh, Graphene oxide
nnano.2013.125. sheets involved in vertically aligned zinc oxide nanowires for visible light
[128] Y. Li, H. Yuan, A. von dem Bussche, M. Creighton, R.H. Hurt, A.B. Kane, et al., photoinactivation of bacteria, J. Alloys Compd. 612 (2014) 380e385, http://
Graphene microsheets enter cells through spontaneous membrane dx.doi.org/10.1016/j.jallcom.2014.05.195.

You might also like