You are on page 1of 17

Materials Research Express

TOPICAL REVIEW

Green synthesis of zinc nanoparticles through plant extracts:


establishing a novel era in cancer theranostics
To cite this article: Safia Hameed et al 2019 Mater. Res. Express 6 102005

View the article online for updates and enhancements.

This content was downloaded from IP address 130.241.16.16 on 23/10/2019 at 20:21


Mater. Res. Express 6 (2019) 102005 https://doi.org/10.1088/2053-1591/ab40df

TOPICAL REVIEW

Green synthesis of zinc nanoparticles through plant extracts:


establishing a novel era in cancer theranostics
RECEIVED
23 May 2019
REVISED
10 August 2019
ACCEPTED FOR PUBLICATION
Safia Hameed1 , Javed Iqbal2 , Muhammad Ali1, Ali Talha Khalil3 , Banzeer Ahsan Abbasi2,
3 September 2019 Muhammad Numan1 and Zabta Khan Shinwari1,3
1
PUBLISHED Department of Biotechnology, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
18 September 2019 2
Department of Plant Sciences, Quaid-i-Azam University, Islamabad, Pakistan
3
Department of Eastern Medicine and Surgery, Qarshi University, Lahore, Pakistan
E-mail: safiahameed77@gmail.com

Keywords: zinc oxide nanoparticles, phyto-synthesis, cancer theranostics, green synthesis

Abstract
Cancer represents one of the leading causes of mortalities worldwide. Recent progress in
nanotechnology has opened a new avenue for research in cancer theranostics. Among the different
metal oxide nanoparticles, the semiconductor zinc oxide nanoparticles have gained significant
importance. Following to tremendous potential of biosynthesized metal nanoparticles as anticancer
agents, there is a dire need to review the current studies available on phytogenic zinc oxide
nanoparticles. We aim to bridge the knowledge vacuum and further elaborate the mechanistic as well
as futuristic insights from biosynthesis to application of the phytogenic zinc oxide nanoparticles. The
present manuscript is a systemic review of the research articles published in recent years by searching
through different online web repositories. Furthermore, in this review, we have discussed about the
recent milestones and possible hurdles for the therapies based on phytogenic zinc oxide nanoparticles.

Cancer: a worldwide threat to existence

Cancer is a dreadful disease which develops by a series of genetic events at the molecular level ultimately
malfunction the regulation of cell division causing uncontrolled division eventually causing cancer. The genetic
instability is further driven by various risk factors leading to DNA mutations followed by cancer initiation and
progression [1]. Cancer is a compound genetic disease comprised of structural and expression abnormalities of
both non-coding and coding genes. From almost 30 years, the source of tumorigenesis has assumed to be in the
alteration of tumor suppressor gene and protein coding oncogenes [2]. Being the main health care problem,
cancer has equally affected the developing and underdeveloped world. Some of the well-established risk factors
associated with cancer are aging overweight, smoking, alcoholic, physical inactivity etc [3]. In 2015, reported 8.8
million people died from cancer, largely in middle and low income countries. In developed regions cancer is the
principal cause of mortalities while in the developing regions it is the second major cause [4]. As of 2017, ∼13%
of the cancers prevailed in adults of age 20 years while the incidence was less prevalent in mature [5]. Current
data analysis shows that, two among 5 people are at risk of developing cancer in their lifespan at a point.
Chemotherapy, radiation therapy, immunotherapy, hormonal therapies are some of the existing modalities for
the treatment of cancer. The selection depends on the stage of disease, position, status of cancer and physical
status of the patient. Many of experimental cancer treatments are also in progress [6]. Many aforementioned
treatment are accompanied with severe side effects. Chemotherapy-concerned harmfulness can occur terribly
after administration, within few hours or days, or even chronically, from weeks to years [7]. Various kinds of
toxicities are related to the administration of chemotherapy. For example, Doxorubicin can lead to
myelotoxicity, cardiotoxicity and renal toxicity. Cutaneous and bladder toxicities are related with the use of
Bleomycin. 5-fluorouracil is another chemotherapeutic agent which is linked to cardio and myelotoxicity.
Therefore, there is a continuous surge for the development of novel treatment strategies with enhanced
theranostic potential which is characterized by features like enhanced efficiency and minimum side effects [8].

© 2019 IOP Publishing Ltd


Mater. Res. Express 6 (2019) 102005 S Hameed et al

Emerging interface of phyto-nano-oncology

Over the past few years, there has been increasing concern in the nanotechnology-based interventions or
treatment modalities with reference to cancer. Nanotechnology is a dynamic blend of interdisciplinary research
through the core principles of chemistry, biology, physics, medicine and engineering and have tremendous
capacity for the treatment of cancer [9, 10]. The interface of medicinal plants, nanotechnology and oncology
proposed as ‘phytonanocology’ is delivering exciting horizons for the treatment of cancer. Medicinal plants are
not only a valuable resource of therapeutic chemical entities which can have anticancer potential but can also be
used to fabricate metal or metal oxide NPs by an ecofriendly and green route.
Phytogenic metal oxide nanoparticles have interesting physical, optical and biological properties, and
therefore widely researched for therapeutic applications. Such exceptional properties are due to their small size
and surface area to volume ratio making them ideal for theranostic applications. Such characteristic structures
and morphologies are usually distinct from their macroscale counterparts. Metal oxide nanoparticles such as
zinc oxide can also be synthesized using different physical and chemical means. Hitherto, being effective, they
are accompanied by disadvantages like cost, energy and generating toxic wastes. On the contrary, the phytogenic
methods are safe, easy and environment friendly. Other biological resources such as bacteria, fungi, algae can
also be used during biosynthesis of metallic nanoparticles, however, the use of plants are preferred as they are
devoid of any extensive processing methods like using antibiotics, or culture conditions which also makes
phytosynthesis an economical process. Microorganisms usually require large incubation periods to allow metals
reduction and also downstream processing make them costly. Use of microorganisms can also raise biosafety
concerns and therefore plants are the perfect candidates for the synthesis of nanoparticles. Additional
advantages of phytomediated synthesis of nanoparticles are enhanced stability, rapid synthesis and cost-
effectiveness. In addition, NPs of several shapes and size can be produced using plant material [11, 12].
The two alternate approaches for the production of metallic NPs are the ‘bottom-up’ method and the ‘top-
down’ method. The bottom-up, means the building of structure atom by atom, molecule by molecule and
cluster by cluster [13]. In this method, primarily the nanostructured key elements (NPs) are produced by using
biological or chemical procedure(s) for production which is assembled into the final material [14]. A distinctive
benefit of the bottom-up method is the improved probability of getting metallic NPs with more homogeneous
chemical composition (s) and relatively lesser defects. In the top-down method, an appropriate beginning
material is squeezed in magnitude using physical and mechanical resources [15]. The main deficiency of top
down method is surface structure imperfection [16, 17]. Figure 1 shows chemical, biological and physical
approaches for the synthesis of zinc oxide nanoparticles.
Such flaws in the surface arrangement can have a substantial effect on surface chemistry and physical
properties of the metallic NPs because of high aspect ratio. Research for development of nanotechnology based
treatment options against cancer is continuously rising since early 2000’s [18]. The speed at which
pharmaceutical companies have made partnerships to use registered nanoparticle technologies has been
significantly quicker [19].

Zinc oxide
In past few years, growing number of available articles displays abundant attention of scientists and engineers for
the significance of metal nanoparticles like gold NPs, silver NPs, cupper NPs and zinc nanoparticles etc, due to
their very small size (nm), the surface plasmon behavior and physicochemical properties [20]. Metal NPs are
being broadly used in different sectors, cancer treatment, drug delivery, wastewater treatment, biosensors, DNA
analysis, antibacterial agents and catalysts [21]. Among the numerous metal oxides, zinc oxides (due to their
fascinating properties) are considered ideal for various biological applications. Zinc oxide is an n-type
semiconductor metal oxide which has been exploited for various applications because of its tunable and
multifunctional spintronic, photonic and morphological features. The ZnO is characterized by a great excitation
energy (60 meV) and wide direct band gape (3.37 eV). Zinc oxide has strong pyroelectric and piezoelectric
characteristics. ZnO is biocompatible, bio-safe with exceptional ability like structure-dependent properties,
thermal and electrical transport properties, which may change with respect to particle size, morphology, shape,
aspect ratio and orientation [22, 23]. Owing to its interesting properties, ZnO has successfully been used in
numerous devices such as metal-insulator semiconductor diodes, miniaturized semiconductor lasers, optically
transparent electrodes, nanogenerators and ultraviolet photo detectors, surface acoustic devices and gas
sensors). In addition, ZnO is among the 5 compounds of zinc that are listed under the category of ‘generally
recognized as safe’. Because of their biocompatible nature. It has also been studied for their potential biomedical
applications such as sunscreens, lotions and cosmetics. They can been used for drug delivery and bio-imaging.
Extensive research work has been carried out on ZnO nanoparticles to establish them as a potential
antimicrobial and anticancer agent. To date, mostly physical and chemical ways are used as a method of

2
Mater. Res. Express 6 (2019) 102005 S Hameed et al

Figure 1. Illustration of chemical, biological and physical approaches for the synthesis of zinc oxide nanoparticles.

synthesis for ZnO NPs. The chemical methods for synthesis that are mostly employed for nano-scaled ZnO
synthesis include hydrothermal, spray pyrolysis, sol-gel, sonochemical, solvothermal and electrodeposition. The
physical methods usually employed for the synthesis include thermal evaporation, pulsed laser deposition,
chemical laser deposition and molecular beam epitaxy etc [24].
The chemical resources of production comprise compounds of chemical nature to reduce Zn++ to ZnONPs;
however, the chemical methods are often objectionable because of their minimum biocompatibility [25].
Similarly, it was reported that the surface of nanoparticles contain certain noxious chemicals and therefore could
not be used for biomedical applications [26].

Mechanism of phytogenic synthesis of zinc oxide NPs


Recently, phytogenic formation of zinc oxide nanoparticles have gained tremendous interest as the method itself
is environmentally benign, easy and complete reliance is on phytochemical entities like phenols, flavonoids,
alkaloids etc [27]. Different plants have been successfully demonstrated in the biosynthesis of ZnO NPs (table 1).
Plant mediated synthesis of ZnO NPs in different sizes and shapes have been successfully established using
different parts of the plants. A typical procedure is based on the addition of precursor salts like zinc acetate, zinc
nitrate to the plant extracts. The bioactive components in the plants such as phenols, alkaloids, tannins, flavones,
proteins have the tendency to reduce metal atoms/ions. The aromatic OH groups are generally proposed to
adhere to the Zn++ ions leading to a stable complex. After annealing, the complex decomposes give rise to ZnO
nanoparticles. Highly crystalline zinc oxide nanoparticles can be obtained by annealing them at high
temperatures [28]. Figure 2 indicates plant-mediated procedures for the biogenic synthesis of zinc oxide NPs

Optimization conditions
Tailored ZnO NPs in terms of size and shape can be produced by varying and optimizing the reaction conditions
like temperature, concentration, duration, pH etc. The reaction parameters have significant role in optimizing
size, yield and stability of the biogenic ZnONPs. For the synthesis different parameters can be optimized, like
concentration of zinc salt, amount of plant material used, pH and temperature at which the reaction is carried
out and the time of incubation for the reaction [29, 30].
Different pH has been reported but the pH range from 6–7 has been reported to be the best for the synthesis
of ZnONPs at 100 °C. Formation of ZnO NPs by means of Vitex negundo plant solution using ZnNO3.6H2O as
starter was defined according to Ambika et al The formation of ZnONPs presented a noticeable and sharp
absorption band on 375 nm. This is because of the point that transition happens from valence toward band of
conduction and at same time the blue shift has also noted with respect to absorption of large amount of ZnO

3
Mater. Res. Express 6 (2019) 102005
Table 1. List of studies elaborating plant extracts for green formation of zinc NPs as a diagnostics and therapeutics agents.

Characterizations Optimal variables Reference

Cancer ZnO Incubation


Reference Plant Part used cell line IC50 value Size (nm) Shape Techniques used (mM) pH Temp (°C) time (hour)

(35) Saccharum officinarum Juice MCF-7 16.7±0.5 μg ml−1 19.4±2.3 nm Spherical FESEM, HRTEM, XRD, UV 1 Neutral 65 4h
visible
(36) Abutilon indicum Leave Extract Calu-6 9.34 ± 0.4 35.2±2.3 nm Spherical FTIR, XRD, TEM, SEM 1 Neutral 60 2–3 h
(37) Rosa indica L petals extract HT-29 9.54 ± 1.2 47 nm Spherical NTA, XRD, SEM, FTIR 1 Neutral Room temp 5–6 h
(38) Eclipta alba Whole plant A43 16.49 ± 1.6 30±2 Spherical XRD, SEM, TEM, FTIR 3 Neutral 100 2–3 h
(39) Tectona grandis (L.) Leaf extract MC3t3-E1 9.79 ± 0.8 27±4 Spherical SEM, TEM, FTIR, XRD, UV 2 7.8 60 3h
visible
(40) Laurus nobilis Leave Extract A549 11.31 ± 0.9 47.27 nm Hexagonal FTIR, XRD, TEM, SEM, EDX 1 Neutral 75 4h
(41) Sargassum muticum Whole plant CaOV-3 10.8±0.3 μg ml−1 10.2±1.5 nm Hexagonal XRD, TEM, SEM 1 neutral 70 2–3 h
(42) Withania Somnifera Leave Extract WEHI-3 12.41 ± 1.6 51.34 nm Hexagonal XRD, TEM, SEM, UV visi- 1 5.4 60 2–3 h
4

cells ble, FTIR


(43) Micrococca mercurialis Leaf and stem WEHI-3 15.34 ± 1.3 53 nm Spherical XRD, SEM, TEM, UV visible 1 6.7 65 2–3 h
extracts cells
(44) Sargassum muticum Leaves WEHI-3 2.25 ± 0.4 22.5±3.5 nm Spherical FTIR, XRD, UV visible, TEM 2 6.4 70 3–4 h
cells
(45) Anabaena variabilis Phycobili MCF-7 16.49 ± 1.6 42 nm±3 Spherical SEM, XRD Raman, FTIR 1 6.8 Room temp 5–6 h
pigment
(46) Cannabis sativa Leaves A549 18.32 ±1.3 40±1.5 nm Hexagonal XRD, TEM, SEM, UV visible 1 6.6 60 3h
(47) Atropa belladonna Leaves MCF-7 12 ±0.9 34±3.2 nm Hexagonal XRD, TEM, Raman 1 Neutral 80 2h
spectroscopy, SEM
(48) Eclipta prostrate Leave Extract Hep-G2 14.15 ± 0.7 29±1.3 nm Spherical SEM-EDX, UV visible, SPR, 1 Neutral 60 3–4 h
(49) Borassus flabellifer fruit extract HT-29 9 ±0.125 55 nm Spherical XRD, TEM, SEM, UV visible, 1 5.8 60 2–3 h
SPR, EDS
(50) Tabernaemontana Leaves MCF-7 30.65 μg ml−1 36±5 nm Spherical XRD, TEM, Raman 1 5.6 60 3
divaricate spectroscopy
(51) Eclipta alba Seed extract HT-29 16.41 ± 1.6 43±3 nm Spherical XRD, SEM, TEM 1 Neutral Room temp 24 h
(52) Coptidis rhizoma Whole plant RAW 264.7 8.47 ± 0.5 2.90–25.20 nm Spherical TGA, TEM, SEM, UV 1 7.1 100 2h
(53) Holorheena Seed extract HepG2 16.27 ± 0.7 13±2 nm Spherical Raman spectroscopy XRD, 2 Neutral 60 2–3 h
antidysentrica TEM, SEM, UV visi-

S Hameed et al
ble, SPR
(54) Tabernaemontana Leaf extract MCF-7 30.65 36±5 Spherical XRD, SEM, Raman 1 Neutral 60 4h
divaricate
(55) Catharanthus roseus Leaf extract MCF-7 21.54 ± 1.9 57 nm spherical TEM, FTIR, XRD 1 Neutral 75 2–3 h
Mater. Res. Express 6 (2019) 102005
Table 1. (Continued.)
Characterizations Optimal variables Reference

Cancer ZnO Incubation


Reference Plant Part used cell line IC50 value Size (nm) Shape Techniques used (mM) pH Temp (°C) time (hour)

(56) Terminalia chebula Seed extract MDAMB- 13.72 ± 1.3 42 nm±3 Spherical XRD, SEM, Raman, XRD, 1 6.7 60 2–3 h
5

231 SEM, Raman


(57) Tabernaemontana Leaf extract MCF-7 30.65 36±5 Spherical Raman spectroscopy XRD, 3 Neutral 80 3–4 h
TEM, SEM, UV visi-
ble, SPR
(58) Saraca asoca Flower extract WEHI-3 13.23 ± 0.8 7–19 nm roughly XRD, TEM, SEM, UV visi- 3 6.5 60 2h
cells spherical ble, EDS
(59) Calotropis gigantea leaves extract Calu-6 11.61 ± 0.9 30–35 nm Spherical XRD, SEM 1 Neutral 80 3h
(60) Borassus flabellifer Leaf extract MCF-7 0.125 55 nm Spherical PL, XRD, SEM, FTIR 1 6.5 75 3h
(61) Calotropis procera leaves extract A549 15.25 ± 1.6 5–40 nm Spherical PL, XRD, SEM, TEM 1 6.6 room temp 4h
(62) Coptidis rhizoma Leaf extract SKBR3 19.38 ± 1.5 20–40 nm Spherical XRD, SEM, TEM 1 Neutral 60 3h
(63) Embelia ribes Root extract MCF-7 9.62 ± 1.9 130 to 150 nm Spherical SEM-EDX, UV visible, 1 6.5 60 2h
SPR, XRD

S Hameed et al
Mater. Res. Express 6 (2019) 102005 S Hameed et al

Figure 2. Plant-mediated procedures for biogenic synthesis of Zinc oxide NPs.

which is normally seems at 400 nm. All these factors recognized hexagonal and spherical shape and approximate
size of the zinc oxide nanoparticles [31].
In a comparative study of the effect of temperature on the shapes of biogenic zinc oxide NPs, different
calcination temperatures (200, 400, 600 °C) were provided to the sample for two hours. In case of precursor of
zinc nitrate, irregular and triangular shaped NPs were produced before and after calcination, while, in case of
precursor ZnSO4, nanoparticles before calcinations were observed to be rod-shaped and grain shaped structures
(modifications of flower type). There are minor changes on 200 °C, where both rod and flakes were observed.
But the shapes of all NPs were altered to flakes or petals type at 400 °C. Though, there are so many differences at
600 °C [32].

Phytogenic ZnO NPs as a candidate for cancer treatment

Though ZnO NPs have been utilized in cosmetic industry since several years, they have newly been discovered as
a candidate for cancer therapies. ZnO NPs are now being extensively studied and well documented in different
research papers. Phytogenic ZnO NPs have been investigated against various cell lines such as PC3 (human
prostate cancer), COLO 205 (colon carcinoma), MCF7 (breast cancer of humans), cell line of jurkat (human
T-cell lymphoma), AGS (human gastric carcinoma), SiHa (cervical cancer), HEPG2 (human liver cancer), HCT-
116, HCT-15 (human colon adenocarcinoma), Caco-2 (intestinal adenocarcinoma), Hep-G2 (liver
carcinomaHek-293 (kidney cancer), H1299, A549 (lung cancer), HeLa (cervical cancer), PA1 (ovarian cancer),
B16 (cell line of mouse melanoma), HL-60 ( the human promyelocytic leukemia cells), VCaP (prostate cancer)
and A431 (epidermoid carcinoma) [33, 34]. Different medicinal plants used for the synthesis of zinc oxide
nanoparticles and their efficacy against different cancer cell lines. Table 1 comprehensively explains the
anticancer results of ZnO NPs synthesis and optimized conditions for biogenic synthesis from studies done in
the previous years.

Mechanism of action of biogenic ZnO NPs


Multiple pathways leading to ROS generation causing oxidative stress is mostly described as a powerful
mechanism of cytotoxic action of biogenic zinc oxide towards malignant cells [35, 36]. Some other mechanism

6
Mater. Res. Express 6 (2019) 102005 S Hameed et al

like increase in the intracellular concentration of Zn++ ions and physical interaction of ZnO NPs with the
cellular membranes are also considered for the potential cytotoxicity towards cancer [37]. Free radicals
produced by zinc oxide nanoparticles would intermingle with macromolecules comprising membrane lipids,
proteins, enzymes and even DNA that might be modified, denatured and oxidized which lastly cause destruction
and dysfunction of various cellular organelles. Here, we discuss various proposed mechanisms for ROS
production and subsequent oxidative stress produced by zinc oxide nanoparticles to have improved knowledge
of their mechanism of toxicity.

Mechanistic perspectives for ROS generation


Different mechanisms have been established for the generation of ROS which are;

(i) Electron hole pairs activated by light or UV responsible for radical formation.
(ii) Electronically active configurations and formation of functional groups on the surface of nanoparticles
because of irregular defects and crystal planes.
(iii) On the surface of nanoparticles active redox cycling owing to transition metal based NPs [38, 39].

Band gap and shrinkage size of biogenic zinc oxide NPs


Semiconductors have a void region known as band gap between the valence band and the conduction band. The
movement of electrons from the valence band to the conduction band leaves holes or unoccupied states in the
valence band [40]. The electron can react with hydroxyl ion and oxygen respectively in aqueous environments
[41] to produce extremely reactive free radical compounds comprising of hydroxyl radical (from holes),
superoxide anion radical (from electrons). Furthermore, shrinkage in NPs size produces structural flaws
accountable for the existence of groups that is improved interstitial Zn ions and oxygen vacant places (in crystal
when the oxygen atom is detached from its exact place the intrinsic defect occur). Moreover, smaller NPs size is
the reason for the disruption of well-structured electronic configurations, so as give rise to altered electronic
properties on the particle surface [42]. Hence, maximum number of electron acceptor and donor active sites can
produce due to crystal defect which can transfer to surface of NPs and contribute in formation of reactive oxygen
species (ROS). Hence, dissimilarity among nanoparticles could describe their discrepancy in their cytotoxic
effects. For a while, silicon and zinc oxides nanoparticles with different shape and size have varied toxic
responses according to their surface properties and composition. Zinc oxide has been revealed to be extra active
chemically than SiO2 and produce greater O−2 production causing in ROS [43]. Hence, surface and chemical
characteristics of zinc oxide nanoparticles lead to impulsive ROS formation at their surface. Photo activated zinc
oxide nanoparticles observed to act like an oxidizing agent with anti-tumor and anti-bacterial activities. Park
et al (2011) recommended that zinc oxide nanomaterial cytotoxicity is associated with ROS and ROS generation
is associated to their production method as well as physiochemical features [44]. Simplified procedure of ROS
formation by zinc oxide nanoparticles are shown in figure 3.
The contact of NPs with cellular compounds like mitochondria can also lead to the production of ROS [45].
The other proposed method used in oxidative injury including metabolization of Zinc oxide nanoparticles with
P450 cytochrome which can be used as a source of ROS as showed in case of other NPs [46].

Correlation among the dissolution of biogenic zinc oxide and oxidative stress (possible cytotoxicity)
Here are few researches which attribute to the cytotoxicity of biogenic Zinc oxide nanoparticles to particle
dissolution and consequent discharge of Zn2+ ions that ultimately causes ROS associated injury and damages
[47, 48]. On the other hand, there are numerous articles which attribute zinc oxide nanoparticles cytotoxicity to
its dissolution; few studies have associated it to systematic pattern like oxidative damage. Though, the results of
some studies indicated interrelationship among dissolution of the zinc oxide nanoparticles and results in
oxidative stress. Biogenic ZnONPs create intrinsically a minor amount of ROS, while the intracellular ROS is
chiefly created after the uptake of zinc oxide nanoparticles or the dissolved Zn2+ that move in into the cell [49].
The other experimental results showed that production of ROS was considerable procedure of cytotoxicity due
to the discharge of zinc ions from zinc oxide nanoparticles, because of their instable nature in the lysosome
acidic compartment which also increases rate of ROS formation. Zhu et al (2012) recommended that the
contrast of zinc oxide nanoparticles and Zn2+ prompted embryonic cytotoxicity by producing reactive oxygen
species or moderate cellular response [50]. Another study determined that zinc oxide nanoparticles cellular
toxicity arose from Zn+2 ions produce from zinc oxide nanoparticles in vivo and in vitro [51]. Moreover,
upsurges in Zn++ intracellular amount observed to interconnect with increased quantity of ROS and ultimate
death of cell. Furthermore, these all changes inside of cell Zn2+ homeostasis induce great stimulatory influence

7
Mater. Res. Express 6 (2019) 102005 S Hameed et al

Figure 3. Various mechanisms of ROS generation by Zinc oxide nanoparticles, (a) Light or UV activation lead to electron hole pair and
free radical generation and (b) show Electron donor/accepter active groups.

on multi conductance calcium channels in the interior of mitochondria membrane leading to production of
ROS in organelles and cell apoptosis and oxidative damage [52]. Other experiments indicated that release of
transition metal cations by engineered NPs is responsible for the induction of OS [53]. The research study stated
that cellular toxicity of zinc oxide nanoparticles perhaps straight forwardly associated to zinc oxide dissolution
via connections at consecutive nano bio interfaces which can prompt a chain of injurious cell related
consequences like ROS generation [54]. Certain studies presented that suspension of ZnONPs and surplus
discharge of Zn2+ leading to distraction of the redox state of mitochondria. Extracted mitochondria
experimentations have revealed that interference of Zn2+ with cytochrome bc1 and that of the complex of α
ketoglutarate dehydrogenase inside the ETC (Electron Transport Chain) hang-up cell mediated respiration
[16, 55]. Diminished mitochondrial function due to improved cell inside Zn2+ produced higher ROS inside the
cell leading to necrotic or apoptotic cell death. Some experiments recommended that surplus free Zn2+ rising
from dissolved zinc oxide nanoparticles inside mitochondria interrupts zinc to protein connections, leading to
distracting of redox state of mitochondria that increases the level of intracellular ROS causing cell death and cell
membrane damage [56].

DNA damage
DNA damage is thought to be a promising means supposed to be accountable for the lethal effects of applied
nanomaterial through oxidative stress [57, 58]. Hydroxyl radical is the principal species accountable for DNA
damaging. Various nanoparticles from their surfaces discharge transition metals ions which have capability to
transform metabolic products of cellular oxygen, like super oxide ion and H2O2 to OH radicals [59]. Reactive
oxygen species (ROS) interact with DNA, causing damage to nucleotide bases and backbone of DNA. Here are
various kinds of ROS prompted DNA impairment such as single and double stranded DNA breaks and base
modifications (e.g. creation of 8 hydroxy deoxyguanosine adducts) and cross links DNA, all of which if
unrepaired possess the capability to start and stimulate carcinogenesis [60]. Toyokuni et al (1998) described that
extremely responsive molecules are ROS that could easily react with cellular macromolecules comprising of
DNA, proteins, lipids and interrupt the homeostasis of intracellular milieu [61]. Zinc oxide nanoparticles are
considered to have the potential of genotoxicity in human epidermal cells though at fewer amounts which
perhaps produce via oxidative stress and lipid peroxidation [62, 63]. Malondialdehyde (MDA) is a core product
of lipid peroxidation which is carcinogenic and mutagenic. It has been stated to act with DNA nucleotides to

8
Mater. Res. Express 6 (2019) 102005 S Hameed et al

form adducts to that of deoxyguanosine, deoxyadenocine and deoxycytocine. Yang et al (2008) inspected
oxidative stress and genotoxicity of zinc oxide nanoparticles tested on fibroblast cells of the embryo of primary
mouse. Their outcomes indicate that oxidative impairment and cytotoxicity of zinc oxide nanoparticles were far
higher compare to other nonmetal nanoparticles [64]. In promise through this research, other study established
that zinc oxide nanoparticles prompts developmental toxicity, DNA damage and oxidative stress on embryo of
Zebra fish, relatively because of the dissolved Zn2+ [65]. A research established that zinc oxide nanoparticles
produce oxidative damage of DNA and cytotoxicity in bronchoalveolar carcinoma derived cells of humans
(A549) in time and dose dependent [66]. Moreover, NPs can damage DNA via indirect mechanisms where they
do not physically react with DNA molecule, but with other cellular proteins such as cell division proteins [67].
The other probable mechanism through which zinc nanoparticles induced ROS and produces DNA injury for
particular signaling pathways activation subsequent in the release of proinflammatory cytokines. Numerous
research studies showed contact of human endothelial cells to diverse nanoparticles cause inflammatory
response which is dependent on NPs concentration and composition [68, 69]. In the research of a group the
production of ROS is an obvious candidate pathway for NPs induced inflammation however, more
investigations are needed to exactly establish a connection between ROS generation and inflammatory responses
Inflammatory factors observed to be induce in point mutation form; DNA adducts formation, DNA damage and
chromosomal partitions. Likewise, these factors can stimulate aberrant methylation patterns and avoid DNA
repair and leading to altered gene expression profile [70]. In a research under changed irradiation environments
using the chromosome aberration test inspected the zinc oxide nanoparticles genotoxicity in cells of Chinese
hamster ovary (CHO) [71]. They observed that ZnO produced a concentration-related increase in chromosome
aberrations in the dark. Chromosome aberrations induced by ZnO NPs showed to enhance in the presence of
UV light. Zinc oxide nanoparticle is genetically toxic in somatic cells (lymphocytes, keratinocytes and
hepatocytes) and germ cells [72]. Conclusions of a novel study exhibited oxidative lesions of DNA in cultures of
epithelial cells of human lungs (A549) after disclosure to zinc nanoparticles [73]. Contact epithelial cells of
human lungs (L-132) to zinc oxide nanoparticles noted to produce DNA fragmentation illustrating cell death in
form of apoptosis. Moreover, damaging effect of DNA via zinc oxide on L02 (human hepatocyte cells) and
HEK293 (embryonic human kidney cells) were observed due to lipid peroxidation [74]. The substantial increase
in the Fpg sites was noticed inside people’s livers who were treated with zinc oxide nanoparticles representing
oxidative damage of DNA. Substantial raises may produce in tail length, percentage of DNA in tail, keratinocytes
and moment of Olive tail after PMEF cells were reacted with [75]. Completely, it could be specified that certain
nanoparticles due to their smaller size have potential to enter inside the nucleus and intermingle with DNA [53].
Whereas few other nanoparticles like zinc oxide nanoparticles might also display an indirect influence on DNA
via their ability of ROS generation [76]. Damaging of DNA can interrupt functions of normal cells which might
lead to cell death or cancer.

Role of nanoparticles in gene expression


However former studies exhibited that these NPs can induce DNA injury and oxidative stress. Though, it could
not supply inclusive evidence at the molecular level to direct by NPs induced stress that how regulation of certain
unnatural effect and antioxidant enzyme activities are affected. Hence, in order to observe the effects of ROS
generation with more details, the necessary genes associated with antioxidant DNA repair system and
antioxidant enzymes, necessity to be assessed. Calculation of oxidative destruction triggered via zinc oxide
nanoparticles in the embryo larval stages of zebra fish was identified through down regulating of Bcl-2, Gstp2, up
regulation of Ucp2 and Nqo1 gene expression [77]. Other research data specified that exposure of BEAS-2B cells
to sub toxic amount of zinc oxide NPs prompts expression of PRDX3 (peroxiredoxin 3), BNIP3 (BCL2/
adenovirus E1B 19 kDa intermingling protein 3), TRXND1 (thioredoxin reductase 1) and PRNP (prion protein)
genes which are accountable in apoptosis and OS [16]. Xia et al (2006) stated that zinc oxide nanoparticles could
prompt enriched NQO1 and the Nrf2, mRNA expression in BEAS2B cells of RAW 264.7 [78]. Another research
study showed that zinc oxide nanoparticles exposed to L-132 cells existing upregulation of the gene such as MT
(metallothionein). One of the vigorous biomarker in toxicity is MT produced via metals which enables metal
detoxification and provide defense from the free radicals [79].

Intracellular enzymes and ZnO NPs


Oxidative cytotoxicity can be summarized as damaging effects due to ROS cytotoxic characteristics which in the
organism could inhibit or activate compound arrays of antioxidant enzymes. Hence, numerous intracellular
enzymes were measured in various articles as oxidative stress biomarkers to gain further information. In primary
mouse embryo fibroblasts (PMEF) cells, the intracellular levels of SOD (superoxide dismutase) activity displayed
a dose related reduction after 24 h exposure to zinc oxide NPs. By decreasing catalase and SOD levels zinc oxide
nanoparticles could prompt OS in epidermal cell line of human (A431). Zhao et al (2013) stated the relationship

9
Mater. Res. Express 6 (2019) 102005 S Hameed et al

among zinc oxide nanoparticles induced OS and in embryo level zebra fish the antioxidant enzymes properties
[53, 80]. They revealed that by exposure to nano zinc oxide of 100 mg l−1 responsible for the extreme generation
of ROS and substantially inhibit the activity of SOD. As concentration of ROS formed by zinc oxide
nanoparticles was surpassed antioxidant capacity of cells, antioxidant system could not remove it and their
outcomes as well accepted that nano ZnO had inhibitory consequences on the activity of catalase,
recommending that H2O2 produced by SOD caused intracellular accumulation of the ROS as it was not
completely removed by catalase. Their results also shown that GPx (glutathione peroxidase) activity in ZnONPs
plus corresponding soluble Zn2+ treated groups enlarged with comparison of control, demonstrating its
improved capability to scavenge lipid hydroperoxides and H2O2 [81]. Research study study recommended that
exposure of A549 cells with zinc oxide nano rods triggered intracellular generation of reactive oxygen species
leading to generation of lipid peroxidation (LPO), catalase, SOD and proposing that OS might be the main
method of zinc oxide nano rods harmfulness in A549 cells. Exhaustion of the SOD level was also noted on 24 h
contact of the kidney cells of human embryo to zinc oxide nanoparticles [82].

Hypercalcemia and nanoparticles


Inside of the cell amount of Ca2+ has key effects on signal transduction, metabolism of cells, and expression of
genes. Ca2+ amount is firmly controlled whereas its upsurges in are related with energetic and metabolic
discrepancy, different disease states which could be harmful to the cell, cellular dysfunction. Numerous
researches established that nanoparticles raise intracellular Ca2+ and ROS levels. A research establish that
carbon black ultrafine nanoparticles brought open up of Ca2+ channels of plasma membrane through a
mechanism comprising of ROS [83]. Huang et al (2010) revealed that Zinc oxide nanoparticles prompt the ROS
intracellular formation and Ca2+ amount bronchial epithelial cells (BEAS-2B) of humans. They recommended
that zinc oxide may be firstly interacting with membrane of cytoplasm because of the lipid peroxidation
beginning damage of cell membrane integrity resulting calcium influx via membrane channels [84]. Zinc oxide
nanoparticles treatment has been reported to prompt significant and continued Ca2+ concentration raised in
RAW 264.7 cell line and BEAS-2B. Zinc oxide nanoparticles induced calcium boost probably produced
cytotoxicity by various methods. A research stated that raise in calcium amount intracellular triggers calmodulin
reliant signaling pathways which are critical for cell toxicity and cytoskeletal dysfunctions which eventually cause
cell death [85]. Main OS response is the intracellular calcium discharge that can lead to trigger mitochondrial
permeability transition pore (PTP), causes degeneracy of mitochondrial membrane capability and discharge of
proapoptotic factors and ultimately cell death [86]. Furthermore, the cell toxicity effects of zinc oxide
nanoparticles were stated to be related with the discharge of Zn2+ from nanoparticles and consequently upsurge
of intracellular Ca2+ level which is additional crucial signal pathway thoroughly correlated to cell necrosis and
apoptosis. Additionally, studies using methods other than NPs indicated that hypercalcemia and OS have
mutual effect on each other. Research findings revealed that the increase in intracellular Ca2+ amount can
trigger enzymes (e.g., hydrolytic enzymes, proteases, dehydrogenases in the citric cycle) that produce RNS/ROS
[66]. In contrast, excess production of PNS/ROS can oxidatively deactivate thiol dependent Ca2+ pump, which
ultimately aggravates hypercalcemia. Both ROS/RNS and hypercalcemia cause intensive prime metabolic
disorders leading to ATP exhaustion and finally death of cells (apoptosis and necrosis) [87].

Nanoparticles and cellular inflammation


In demand to retain the redox equilibrium at an intermediary level of OS, pro inflammatory pathways are
triggered. Hence, effects of peroxidase of certain nanoparticles lead to the activation of cytokine cascade and
signaling pathways that contribute to wide range of cellular responses. Numerous metal oxide nanoparticles like
Zn, Cd, Si, and Fe apply their toxic effects through ROS dependent nuclear factor-5ØßB (NF-5ØßB) triggering
[88]. The NF-5ØßB family plays a significant role in regulation of immune and inflammatory responses, cell
proliferation, and apoptosis. In NPs associated lung damage, activation of NF-5ØßB moderates production of
proinflammatory cytokines (IL-2, TNF-5ØüÞ, IL-6, and IL-8) by lung epithelial cells and macrophages.
Ultrafine zinc oxide nanoparticles range the alveoli and create symptomatic responses and pulmonary
inflammation in the lung via increase in the proinflammatory cytokines TNF-α, IL-6 and IL-8. Previous studies
shows that ROS generation by zinc oxide nanoparticles affects amplified early growth responce-1 (Egr-1)
expression through extracellular signal regulating kinase (ERK) leading to induction of transcriptional
activations of TNF-α expression and cause inflammation of cell. Furthermore, further numerous in vivo and
in vitro studies of pulmonary inhalation have shown that ZnONPs exposure prompt the production of
numerous proinflammatory cytokines, like TNF-α, IL-12 and IFN-γ. In addition substantial reduction in cell
viability, notable changes occur morphologically, apoptosis stimulation through ROS generation and IL- 8
discharge, after treating with zinc oxide nanoparticles exposure proposing that OS was the main mechanism for
the cell injury [89]. Zinc oxide nanoparticles induce the production of proinflammatory cytokines at
concentrations less than those causing considerable cell death, suggesting that zinc oxide nanoparticles could

10
Mater. Res. Express 6 (2019) 102005 S Hameed et al

improve killing of tumor cells via the construction of TNF-α when used in appropriate amount. Exposure of
RAW 264.7 and BEAS-2B cells to zinc oxide nanoparticles could effectively trigger cascade of Jun kinase (JNK)
which happened in parallel with enlarged TNF- α and IL-8 formation respectively [88].

Lipid peroxidation and NPs


The lipid peroxidation of unsaturated fatty acids existing in phospholipids of membrane is the most authentic
lethal effects of oxygen based free radicals. One of the end products of lipid peroxidation is MDA is known as
marker of lipid peroxidation and related to the amount of ROS generated. MDA content in the zebra fish
exposed to high concentrations of nano-ZnO increased significantly compared to the control group [68].
Consequences of this research design were in accord to conclude the work of other researchers. Findings of the
other trials shown that zinc oxide nanoparticles treatment at 50 and 100 μg ml−1 increases the inside amount of
MDA in a quantity reliant manner in the PMEF cell. Creation of hyper peroxide as another remarkable marker
of oxidation of lipids by ROS was also specified by the improvement of lactate dehydrogenase (LDH) discharge
after exposing of the cells like A431 to zinc oxide nanoparticles [90]. In HepG2 cells enhance in lipid peroxides
on exposure to zinc oxide nanoparticles was also detected which shows an additional marker of OS. Lipid
peroxidation can furthermore give rise to extra free radicals and harmed biomolecules [91]. Particle associated
reactive oxygen species generation and consequent lipid peroxidation of cellular membrane were also planned to
clarify zinc oxide nanoparticles phytotoxicity to regress and yeast [92].

NPs induced oxidation and mitochondrial dysfunction


Mitochondria are experienced target organelle for cytotoxic damage. By favoring aerobic ATP production, they
play significant role in retaining functions of cells. Besides providing cellular energy, they are also involve in
other processes like cellular differentiation, signal transduction apoptosis, cell growth and cell proliferation [91].
Mitochondrion is considered as one of the main cell targets for nanoparticles induced OS and the intrinsic
mitochondrial apoptotic pathway and play significant role in metal oxide nanoparticles prompted cell death.
Numerous metal oxide nanoparticles produce ROS associated cell death through dysfunction of mitochondria.
When nanoparticles reach inside mitochondria, they can generate mitochondrial ROS through depolarization
mitochondrial membrane and impaired electron transport chain. Nanoparticles could catalyze the formation of
superoxide anion (O2) either by speeding the rate of electron transfer to molecular oxygen or by blocking
mitochondrial electron transport chain. Assessment of the power of the membrane of mitochondria (Δψ) by
Xia et al (2006) showed that zinc nanoparticles could efficiently enhance the cells percentage with a lesser Δψm
while CeO2 and TiO2 did not disturb the Δψm [81].

NPs-induced apoptosis
Numerous research works have recognized the oxidative stress as simple pathway for zinc oxide nanoparticles
related apoptosis. A study presented zinc oxide nanoparticles shows an increase of cellular ROS in a quantity
dependent method followed by JNK phosphorylation representing that JNK signaling pathway was responsible
in apoptosis in response to zinc oxide nanoparticles [93]. Furthermore, in liver cells of treated mice a substantial
amount of cells experiencing apoptosis were found in contrast to the control group proposing that during the
in vivo exposure the oxidative stress may produce apoptosis. A research exhibited that sub-acute oral exposure to
zinc oxide NPs in mice leads to a storage of NPs in the liver triggering DNA damage and oxidative stress
mediated apoptosis. Another study was intended to examine the cytotoxicity, apoptosis and oxidative stress
caused by zinc oxide nanoparticles in skin melanoma cells of humans. Results showed that zinc oxide
nanoparticle was responsible for induction of apoptosis and oxidative stress. Data of the other experiment
demonstrated that zinc oxide nano rod initiated apoptosis in alveolar adenocarcinoma (A549) cells of humans
via oxidative stress and ROS by regulating p53, survivin, bax/bcl-2 and caspase pathways [94].

Drug delivery of ZnO nano-therapeutics

ZnO quantum dots: a clever drug delivery nano carrier


In several cases, chemotherapeutic drugs become progressively lethal to the healthy cells. To progress and
advance this problem of chemotherapeutic drugs toxicity to healthy cells, directed drug supply and quick
exposure of cancer cells both need to be extensively examined [95]. Tumor directed drug transport systems
usually combine a tumor identifying moiety like folate receptors which are greatly exposed on the tumor cells
along with the drug loaded vesicle. But, current anticancer chemotherapies frequently demonstrate adverse
effects because of failure to distinguish among normal and cancerous cells. Drug Delivery Systems (DDS) based
on nanotechnology display excessive ability in the treatment of cancer and used to convey anti-cancer drugs to

11
Mater. Res. Express 6 (2019) 102005 S Hameed et al

Figure 4. Schematic diagram of the DOX delivery from the ZnO nanoparticles. [101] (Adapted from Satinder et al 2016 with
permission).

the target tissues. In view of the intracellular parts such as lysosomes and endosomes and extracellular slightly
acidic atmosphere inside the solid tumor tissues [40].

NPs designing
NPs were designed in such a way to move in the cells by endocytic pathway of cells and combine with lysosomes
proliferation [96], thus a pH reactive DDS can be a perfect selection intended for the treatment of cancer in
which NPs will be released in a specific pH conditions. Luckily, ZnO nanoparticles may display excessive
strength in physiological condition that is pH of 7.4, but quickly soluble at pH 5 to 6.
ZnONPs protect the holes of mesoporous silica nanoparticles (MSNs) and were used as cappers and once
these DDS come across with acids, the ZnO nanoparticles decayed to discharge doxorubicin molecules of DOX
from that of MSNs. Though, this kind of DDS has trouble in breakdown so it may not fully discharge drugs [97].
Direct loading of therapeutic drugs on to ZnO nanoparticles has also been reported. This drug delivery
mechanism is shown in figure 4. Even though the stabilized nanoparticle shown less toxicity and good
biocompability, later ZnO decay, Zn2+ ions are toxic for cells [98]. In the past few years, due to their low toxicity,
ZnO quantum dots have been established as multifunctional nano carriers for drugs. For the delivery of
doxorubicin (DOX) the ‘ZnO-chitosan-folate’ system can be used as a nano carrier which manifest therapeutic
action by chemical as well as physical interaction with cancer cells. Various features like particle size, surface
properties, pH response, degradation etc are considered before loading a drug to the carrier. Usually, drugs
loaded nanoparticle electrostatic interactions; physical adsorption and p–p stacking while drug release is
accomplished by breaking of these interactions. Electrostatic interaction among ZnO QDs and DOX become
weakens because of the existence of FA (folic acid) inside the ZnO-QD-chitosan folate carrier, responsible for the
release of DOX. In other study, Zinc Oxide QDs also have been considered for pH reactive intracellular transport
of DOX [99]. Here, acidic conditions produce rapid dissolution of ZnO QDs releasing DOX and Zn2+,
consequential in the cytosols killing the cancer cells. Latest work [100] points towards the application of water
dispersed Zinc Oxide quantum dots with durable stability of fluorescence in design of novel drug release carriers.

Zinc oxide nanoparticles: photodynamic and cancer therapy


Photodynamic therapy (PDT) is a developing and encouraging substitute for non-invasive treatment of cancer
[102]. By uptake of photosensitizers into cancer cells, irradiation having light of appropriate dosage and
wavelength can generate ROS reactive oxygen species which can induce necrosis or death of cells. ZnO
nanoparticles can induce ROS that of hydrogen peroxide, hydroxyl radicals and superoxide in aqueous solutions
by absorption of UV illumination, proving it good candidates for PDT. ZnO NPs together with grapheme under
visible light irradiation can be used for targeting photodynamic therapy (PDT). Through imide linkage, the Folic
acid, a tumor targeting agent was conjugated on graphene oxide [103]. The combination of Zinc Oxide with
GO–FA prompted an amazing enhancement in targeting of tumor, which has been validated by the cellular

12
Mater. Res. Express 6 (2019) 102005 S Hameed et al

uptake examine. Because of the communication among ZnO and graphene, [104–106] great electrical
conductivity of the graphene, inhibition of aggregation, hybrid of GO–FA and Zinc Oxide photodynamic
property is considerably increase [107]. This was observed that below visible light irradiation the photodynamic
property of the non-cytotoxic GO–FA–ZnO is facilitated by the reactive oxygen species (ROS) production.
Resulting, the ROS production, GO–FA–ZnO produced a substantial reduction in viability of cell, catalase and
glutathione peroxidase, superoxide dismutase activity, mitochondrial membrane potential, besides the rise in
malonodialdehyde formation. Furthermore, GO–FA–ZnO by elevating the caspase-3 activity, prompted
apoptotic death. Therefore, GO–FA–ZnO can act as an encouraging approach in PDT for cancer cure and a
novel tumor targeting photosensitizer [108].

Conclusion and future perspective

Nanosciences has delivered significant breakthroughs in medicine, especially for cancer. Zinc oxide
nanoparticles have diverse applications like drug delivery, tumor imaging and targeting etc which provides
effective clinical platforms for theranostic perspectives. Additional features of ZnO nanoparticles are their
compatibility, easy synthesis and tunable features makes them an ideal candidate [109]. The compatibility is
further enhanced through green chemistry based biosynthesis, however to get in to the real applications of
biogenic ZnO nanoparticles, their toxicity studies needs to be extensively elaborated. Mechanistic studies needs
to be undertaken so as to find the functional groups and exact phytochemicals involved in the stabilization and
capping of the biogenic ZnO. Phytosynthesis of nanoparticles is still in infancy. Although initial research
indicates promising findings in nanomedicine but their toxicity and biocompatibility aspects are still not clear.

Conflicts of interest

There are no conflicts to declare.

ORCID iDs

Safia Hameed https://orcid.org/0000-0002-5052-7172


Javed Iqbal https://orcid.org/0000-0003-3036-4481
Ali Talha Khalil https://orcid.org/0000-0002-9848-8723

References
[1] Ahmedin J D V M, Freddie B and Melissa M C 2011 Global Cancer Statistics 90 784–99
[2] Iqbal J, Abbasi B A, Mahmood T, Kanwal S, Ali B, Shah S A and Khalil A T 2017 Plant-derived anticancer agents: a green anticancer
approach Asian Pacific Journal of Tropical Biomedicine 7 1129–50
[3] Iqbal J, Abbasi B A, Batool R, Mahmood T, Ali B, Khalil A T and Ahmad R 2018 Potential phytocompounds for developing breast
cancer therapeutics: nature’s healing touch Eur. J. Pharmacol. 827 125–48
[4] George A C 2006 MicroRNA signatures in human cancers Nature 6 857–66
[5] Hoeijmakers H J J 2001 Genome maintenance mechanisms for preventing cancer Nature 411 366–74
[6] Abbasi B A, Iqbal J, Mahmood T, Khalil A T, Ali B, Kanwal S and Ahmad R 2018 Role of dietary phytochemicals in modulation of
miRNA expression: natural swords combating breast cancer Asian Pacific Journal of Tropical Medicine 11 501–9
[7] Mieog J S D and van D H 2007 Neo adjuvant chemotherapy for operable breast cancer British Journal of Surgery 94 1189–200
[8] Chow A Y 2010 Cell cycle control by oncogenes and tumor suppressors: driving the transformation of normal cells into cancerous
cells Nano Biotechnology 8 317–25
[9] Suriamoorthy P, Zhang X and Hao G 2010 Folic acid-CdTe quantum dot conjugates and their applications for cancer cell targeting
Cancer Nanotechnol. 1 7743–62
[10] Azizi S, Ahmad M B, Namvar F and Mohamad R 2014 Green biosynthesis and characterization of zinc oxide nanoparticles using
brown marine macroalga Sargassum muticum aqueous extract Mater. Lett. 116 275–7
[11] Iqbal J, Abbasi B A, Mahmood T, Kanwal S, Ahmad R and Ashraf M 2019 Plant-extract mediated green approach for the synthesis of
ZnONPs: Characterization and evaluation of cytotoxic, antimicrobial and antioxidant potentials J. Mol. Struct. 1189 315–27
[12] Abbasi B A, Iqbal J, Mahmood T, Ahmad R, Kanwal S and Afridi S 2019 Plant-mediated synthesis of nickel oxide nanoparticles (NiO)
via Geranium wallichianum: characterization and different biological applications Mater. Res. Express 2019 6 0850a7
[13] Mukherjee P, Roy M and Mandal B 2008 Green synthesis of highly stabilized nanocrystalline silver particles by a non-pathogenic and
agriculturally important fungus T. asperellum Nanotechnology 19 75103–10
[14] Shankar S, Ahmad A and Sastry M 2003 Geranium leaf assisted biosynthesis of silver nanoparticles Biotechnol Prog 2003 19 1627–31
[15] Shankar S, Rai A and Ankamwar B 2004 Biological synthesis of triangular gold nanoprisms Nat. Mater. 482–8
[16] Huang C C et al 2010 Oxidative stress, calcium homeostasis, and altered gene expression in human lung epithelial cells exposed to
ZnO nanoparticles Toxicol. in Vitro 24 45–55
[17] Armendariz V, Herrera I and Peralta V J R 2004 Size controlled gold nanoparticle formation by Avena sativa biomass: use of plants in
nanobiotechnology J Nanoparticle Res 6 377–82

13
Mater. Res. Express 6 (2019) 102005 S Hameed et al

[18] Gautama A and Van V 2013 Synthesis of nanoparticles, their biocompatibility, and toxicity behavior for biomedical applications
J. Mater. Chem. 1 5186–200
[19] Nagarajan S and Kuppusamy K A 2013 Extracellular synthesis of zinc oxide nanoparticle using seaweeds of gulf of Mannar, India
Journal of Nanobiotechnology 11 39–45
[20] Raveendran P, Fu J and Wallen S L 2003 Completely ‘green’ synthesis and stabilization of metal nanoparticles JACS 125 13940–1
[21] Iqbal J, Abbasi B A, Mahmood T, Hameed S, Munir A and Kanwal S 2019 Green synthesis and characterizations of Nickel oxide
nanoparticles using leaf extract of Rhamnus virgata and their potential biological applications Appl. Organomet. Chem. 23 49–50
[22] Rajeshkumar S, Malarkodi C and Paulkumar K M 2014 Algae mediated green fabrication of silver nanoparticles and examination of
its antifungal activity against clinical pathogens Int. J. Met 2014 674–89
[23] Namver F, Chisti Y and Banerjee U C 2016 Synthesis of metallic nanoparticles using plant extracts Biotechnol. Adv. 31 346–56
[24] Shakeel A and Annu S A C 2017 A review on biogenic synthesis of ZnO nanoparticles using plant extracts and microbes A Prospect
Towards Green Chemistry 166 272–84
[25] Farideh N J W 2015 Zinc oxide nanoparticles for selective destruction of tumor cells and potential for drug delivery applications
Expert Opin. Drug Deliv. 7 1063–77
[26] Hanley C 2010 Preferential killing of cancer cells and activated human T cells using ZnO nanoparticles Nanotechnology 19 295103
[27] Sidra S, Manikandan M and Dhlamini M S 2015 Green synthesis of ZnO nanoparticles via Agathosma betulina natural extract Mater.
Lett. 161 124–7
[28] Thema F T, Manikandan M and Dhlamini M S 2015 Green synthesis of ZnO nanoparticles via Agathosma betulina natural extract
Mater. Lett. 161 124–7
[29] Ambika S and Sundrarajan M 2015 Biology antibacterial behaviour of Vitex negundo extract assisted ZnO nanoparticles against
pathogenic bacteria J. Photochem. Photobiol. B 146 52–7
[30] Rich I N, Worthington D, Garden O A and Musk P 2000 Apoptosis of leukemic cells accompanies reduction in intracellular pH after
targeted inhibition of the Na(+)/H(+) exchanger Blood. 95 1427–34
[31] Prashanth G K, Prashanth P A and Manoj G 2017 In vitro antibacterial and anticancer studies of ZnO nanoparticles prepared by sugar
fueled combustion synthesis Advanced Materials Letters 8 24–9
[32] Prashanth G K, Prashanth P A and Nagabhushana B M 2017 Comparison of anticancer activity of biocompatible ZnO nanoparticles
prepared by solution combustion synthesis using aqueous leaf extracts of Abutilon indicum, Melia azedarach and Indigofera tinctoria
as biofuels Artificial Cells, Nanomedicine, and Biotechnology 46 968–79
[33] Rasmussen J W, Martinez E and Louka P 2010 Zinc oxide nanoparticles for selective destruction of tumor cells and potential for drug
delivery applications Expert Opinion on Drug Delivery 7 1063–77
[34] Namver F et al 2016 Green synthesis, characterization, and anticancer activity of hyaluronan/zinc oxide nanocomposites Dovepress 9
4549–59
[35] Mukherjee S et al 2014 Potential theranostics application of bio-synthesized silver nanoparticles (4-in-1 system) Theranostics 4 316–35
[36] Firdhouse M J and Lalitha P 2013 Biosynthesis of silver nanoparticles using the extract of Alternanthera sessilis—antiproliferative
effect against prostate cancer cells Cancer Nanotechnol. 4 137–43
[37] Yang Z and Xie C 2006 Zn2+release from zinc and zinc oxide particles in simulated uterine solution Colloids Surf B Biointerfaces 10
5418–21
[38] Premanathan M, Karthikeyan K and Jeyasubramanian K 2011 Selective toxicity of ZnO nanoparticles toward Gram-positive bacteria
and cancer cells by apoptosis through lipid peroxidation Nanomedicine. 7 184–92
[39] Asha Rani P, Low K M G and Hande M P 2008 Cytotoxicity and genotoxicity of silver nanoparticles in human cells ACS Nano. 3
279–90
[40] Rasmussen J W, Martinez E and Louka P 2010 Zinc oxide nanoparticles for selective destruction of tumor cells and potential for drug
delivery applications Expert Opin Drug Deliv. 7 1063–77
[41] Talalay P, Dinkova-Kostova A T and Holtzclaw W D 2003 Importance of phase 2 gene regulation in protection against electrophile
and reactive oxygen toxicity and carcinogenesis Adv. Enzyme Regul. 43 121–34
[42] Min K-S 2007 [Physiological significance of metallothionein in oxidative stress Yakugaku Zasshi. 84 695–702
[43] Donaldson K et al 2002 The pulmonary toxicology of ultrafine particles J. Aerosol Med. 100 213–20
[44] Park S J et al 2011 Comparing the toxic mechanism of synthesized zinc oxide nanomaterials by physicochemical characterization and
reactive oxygen species properties Toxicol. Lett. 80 197–203
[45] Griffitt R J et al 2007 Exposure to copper nanoparticles causes gill injury and acute lethality in zebrafish (Danio rerio) Environ. Sci.
Technol. 24 8178–86
[46] Bishop G M, Dringen R and Robinson S 2007 Zinc stimulates the production of toxic reactive oxygen species (ROS) and inhibits
glutathione reductase in astrocytes Free Radic. Biol. Med. 117 1222–30
[47] Singh N et al 2009 NanoGenotoxicology: the DNA damaging potential of engineered nanomaterials Biomaterials 1 118–120
[48] Zhao X et al 2013 Acute ZnO nanoparticles exposure induces developmental toxicity, oxidative stress and DNA damage in embryo-
larval zebrafish Aquat. Toxicol. 2 49–59
[49] Horie M et al 2009 Protein adsorption of ultrafine metal oxide and its influence on cytotoxicity toward cultured cells Chem. Res.
Toxicol. 22 543–53
[50] Zhu J, Liao L and Bian X J 2012 pH-Controlled delivery of doxorubicin to cancer cells, based on small mesoporous carbon
nanospheres Small. 1 118–120
[51] Stone V et al 2000 Increased calcium influx in a monocytic cell line on exposure to ultrafine carbon black Eur. Respir. J. 15 297–303
[52] Moller W et al 2005 Ultrafine particles cause cytoskeletal dysfunctions in macrophages: role of intracellular calcium Part. Fibre
Toxicol. 2 7
[53] Boonstra J and Post J A 2004 Molecular events associated with reactive oxygen species and cell cycle progression in mammalian cells
Gene. 337 1–13
[54] Wang L et al 2005 Essential role of p53 in silica-induced apoptosis Am. J. Physiol. Lung Cell Mol. Physiol. 1 L488–96
[55] Yin Y et al 2012 Cytotoxic effects of ZnO hierarchical architectures on RSC96 Schwann cells Nanoscale Res. Lett. 7 1–8
[56] Ma H, Williams P L and Diamond S A 2013 Ecotoxicity of manufactured ZnO nanoparticles–a review. A review Environ. Pollut. 172
76–85
[57] Alarifi S, Ali D and Alkahtani S 2013 Induction of oxidative stress, DNA damage, and apoptosis in a malignant human skin melanoma
cell line after exposure to zinc oxide nanoparticles Int. J. Nanomed. 41 10347–56
[58] Franklin N M, Rogers N J and Apte S C 2007 Comparative toxicity of nanoparticulate ZnO, bulk ZnO and ZnCl2 to a freshwater
microalga (Pseudokirchneriella subcapitata): the importance of particle solubility Environ. Sci. Technol. 6 208–14

14
Mater. Res. Express 6 (2019) 102005 S Hameed et al

[59] Moreau J W, Weber P K and Martin M C 2007 Extracellular proteins limit the dispersal of biogenic nanoparticles Science 7 1063–77
[60] Nel K W, Zhao W and Li N 2009 Air-pollutant chemicals and oxidized lipids exhibit genome-wide synergistic effects on endothelial
cells Genome Biol. 30 R149
[61] Toyokuni A M, Kristal B S and Effron M S 2008 Zn2+inhibits α-ketoglutarate-stimulated mitochondrial respiration and the isolated
α-ketoglutarate dehydrogenase complex J. Biol. Chem. 133 178–181
[62] Manke A, Wang L and Rojanasakul Y 2013 Mechanisms of nanoparticle-induced oxidative stress and toxicity BioMed Res 213 1–15
[63] Yuan Q, Hein S and Misra R D K 2010 New generation of chitosan-encapsulated ZnO quantum dots loaded with drug: synthesis,
characterization and in vitro drug delivery response Acta Biomater. 6 5342–65
[64] Hubbard A K, Timblin C R and Shukla A 2002 Activation of NF-κB-dependent gene expression by silica in lungs of luciferase reporter
mice Am. J. Physiol. Lung Cell Mol. Physiol. 282 L968–75
[65] Syama S, Sreekanth P and Varma H 2014 Zinc oxide nanoparticles induced oxidative stress in mouse bone marrow mesenchymal
stem cells Toxicol. Mech. Methods. 24 644–53
[66] Yu K N, Yoon T J and Minai T A 2013 Zinc oxide nanoparticle induced autophagic cell death and mitochondrial damage via reactive
oxygen species generation Toxicol. in Vitro 27 1187–95
[67] Zabirnyk O, Yezhelyev M and Seleverstov O 2007 Nanoparticles as a novel class of autophagy activators Autophagy. 3 278–81
[68] Sharma V, Anderson D and Dhawan A 2012 Zinc oxide nanoparticles induce oxidative DNA damage and ROS-triggered
mitochondria mediated apoptosis in human liver cells (HepG2) Apoptosis. 17 852–70
[69] Frazzini V, Rockabrand E and Mocchegiani E 2006 Oxidative stress and brain aging: is zinc the link? Biogerontology. 7 307–14
[70] Yang D, Wells S M and Wilham J 2008 Endothelial response to stress from exogenous Zn2+ resembles that of NO-mediated
nitrosative stress, and is protected by MT-1 overexpression Am. J. Physiol. Cell Physiol. 291 C555–68
[71] Dufour E K, Kumaravel T, Nohynek G J, Kirkland D and Toutain H 2006 Clastogenicity, photo clastogenicity or pseudophoto-
clastogenicity: genotoxic effects of zinc oxide in the dark, in pre-irradiated or simultaneously irradiated Chinese hamster ovary cells
Mutat Res. 607 215–24
[72] Lin H, Williams P L and Diamond S A 2009 Ecotoxicity of manufactured ZnO nanoparticles–a review A review. Environ. Pollut. 172
76–85
[73] Sharma V, Singh P, Pandey A K and Dhawan A 2012 Induction of oxidative stress, DNA damage and apoptosis in mouse liver after
sub-acute oral exposure to zinc oxide nanoparticles Mutat Res. 745 84–91
[74] Sahu D, Kannan G, Vijayaraghavan R, Anand T and Khanum F 2013 Nanosized zinc oxide induces toxicity in human lung cells ISRN
toxicology 2013 1–8
[75] Knaapen A M, Borm P J and Albrecht C 2004 Inhaled particles and lung cancer. Part A: mechanisms Int. J. Cancer. 109 799–809
[76] Yu L, Lu Y and Man N 2009 rare earth oxide nanocrystals induce autophagy in HeLa cells Small. 5 2784–7
[77] Dufour X, Wang J and Zhang X 2006 The impact of ZnO nanoparticle aggregates on the embryonic development of zebrafish (Danio
rerio) Nanotechnology 20 195103
[78] Xia T, Kovochich M and Brant J 2006 Comparison of the abilities of ambient and manufactured nanoparticles to induce cellular
toxicity according to an oxidative stress paradigm Nano Lett. 8 1794–807
[79] Chen Y, Yang L and Feng C 2005 Nano neodymium oxide induces massive vacuolization and autophagic cell death in non-small cell
lung cancer NCI-H460 cells Biochem. Biophys. Res. 337 52–60
[80] Karlsson K, Lin Q and Sun H 2008 Cytotoxic effects of ZnO hierarchical architectures on RSC96 Schwann cells Nanoscale Res. Lett. 7
1–8
[81] Lanone S and Boczkowski J 2006 Biomedical applications and potential health risks of nanomaterials: molecular mechanisms Curr.
Mol. Med. 6 651–63
[82] Murray A R, Kisin E R and Tkach A V 2012 Factoring-in agglomeration of carbon nanotubes and nanofibers for better prediction of
their toxicity versus asbestos Part. Fibre Toxicol. 9 1–19
[83] Moller W, Brown D M, Kreyling L and Stone V 2005 Ultrafine particles cause cytoskeletal dysfunctions in macrophages: role of
intracellular calcium Part. Fibre Toxicol. 2 7
[84] Huang C C, Aronstam R S, Chen D R and Huang Y W 2010 Oxidative stress, calcium homeostasis, and altered gene expression in
human lung epithelial cells exposed to ZnO nanoparticles Toxicol. in Vitro 8 45–55
[85] Zhao X, Wang S and Wu Y 2013 Acute ZnO nanoparticles exposure induces developmental toxicity, oxidative stress and DNA damage
in embryo-larval zebrafish Aquat. Toxicol. 136 49–59
[86] Hsiao I L and Huang Y J 2011 Effects of various physicochemical characteristics on the toxicities of ZnO and TiO2 nanoparticles
toward human lung epithelial cells Sci. Total Environ. 409 1219–28
[87] Ahamed T, Kovochich M and Brant J 2011 Comparison of the abilities of ambient and manufactured nanoparticles to induce cellular
toxicity according to an oxidative stress paradigm Nano Lett. 6 1794–807
[88] Gojova A, Guo B and Kota R S 2007 Induction of inflammation in vascular endothelial cells by metal oxide nanoparticles: effect of
particle composition Environ. Health Perspect. Environmental health Perspectives 115 403–9
[89] Peters K, Unger R E and Kirkpatrick C J 2004 Effects of nano-scaled particles on endothelial cell function in vitro: studies on viability,
proliferation and inflammation J. Mater. Sci., Mater. Med. 213 1–15
[90] Gopalan R C, Osman I F and Amani A 2009 The effect of zinc oxide and titanium dioxide nanoparticles in the Comet assay with UVA
photoactivation of human sperm and lymphocytes Nanotoxicology. 6 33–9
[91] Harma V, Anderson D and Dhawan A 2012 Zinc oxide nanoparticles induce oxidative DNA damage and ROS-triggered mitochondria
mediated apoptosis in human liver cells (HepG2) Apoptosis. 17 852–70
[92] Sahu D, Kannan G and Vijayaraghavan R 2013 Nanosized zinc oxide induces toxicity in human lung cells ISRN toxicology 2013 1–8
[93] Singh N, Manshian B and Jenkins G J 2009 NanoGenotoxicology: the DNA damaging potential of engineered nanomaterials
Biomaterials 30 3891–914
[94] Wang V, Rockabrand E and Mocchegiani E 2013 Oxidative stress and brain aging: is zinc the link? Biogerontology. 7 307–14
[95] Wiseman D A, Wells S M and Wilham J 2006 Endothelial response to stress from exogenous Zn2+ resembles that of NO-mediated
nitrosative stress, and is protected by MT-1 overexpression Am. J. Physiol. Cell Physiol. 291 C555–68
[96] Moghimi S M and Rajabi S A R 2000 Adv.megnatic resonance imaging Drug Deliver. Rev. 1 129
[97] Jaracz S, Chen J and Kuznetsova L V 2005 Recent advances in tumor-targeting anticancer drug conjugates Med. Chem. 13 5043
[98] Weitman S D, Lark R H and Coney L R 1992 Distribution of the folate receptor GP38 in normal and malignant cell lines and tissues
Cancer Res. 52 3396
[99] Ke C J, Su T Y and Chen H L 2011 Smart multifunctional hollow microspheres for the quick release of drugs in intracellular lysosomal
compartments Angew. Chem. Int. Ed. 50 4011–26

15
Mater. Res. Express 6 (2019) 102005 S Hameed et al

[100] Jin E, Zhang B and Sun X R 2013 Acid-active cell-penetrating peptides for in vivo tumor-targeted drug delivery J. Am. Chem. Soc.
135 933
[101] Satinder P K, Malhotra k and Mandal T K 2016 Biomedical applications of zinc oxide nanomaterials in cancer treatment: a review
SCIREA Journal of Chemistry 2013 1–8
[102] Pearce T R, Shroff K and Kokkoli E 2012 Peptide targeted lipid nanoparticles for anticancer drug delivery Adv. Mater. 24 5372–85
[103] Lin Y S, Hurley K R and Haynes C L 2012 Critical considerations in the biomedical use of mesoporous silica nanoparticles J. Phys.
Chem. Lett. 3 364 87
[104] Zhang Z Y, Xu Y D and Ma Y Y 2013 Biodegradable ZnO@polymer core–shell nanocarriers: pH-triggered release of doxorubicin
in vitro Angew. Chem. Int. Ed. 52 1242–63
[105] Rasmussen J W, Martinez E and Louka P 2010 Zinc oxide nanoparticles for selective destruction of tumor cells and potential for drug
delivery applications Expert Opin. Drug Delivery. 7 1831–45
[106] Brown S B, Brown E A and Walker I 2004 The present and future role of photodynamic therapy in cancer treatment Lancet Oncol. 5
5120–42
[107] Zhang H, Chen B and Jiang H 2011 A strategy for ZnO nanorod mediated multi-mode cancer treatment Biomaterials 32 2567–81
[108] Hu Z, Zhao S and Wang Y 2013 Folic acid-conjugated graphene–ZnO nanohybrid for targeting photodynamic therapy under visible
light irradiation J. Mater. Chem. 1 2238–54
[109] Ovais M, Ali T K and Raza A 2016 Green synthesis of silver nanoparticles via plant extracts: beginning a new era in cancer theranostics
Nanomedicine 12 3157–3177

16

You might also like