Вы находитесь на странице: 1из 59

Accepted Manuscript

Recent progress in biomedical applications of chitosan and its


nanocomposites in aquaculture: A review

Fatma Ahmed, Faiza M. Soliman, Mohamed A. Adly, Hamdy


A.M. Soliman, Mansour El-Matbouli, Mona Saleh

PII: S0034-5288(19)30163-8
DOI: https://doi.org/10.1016/j.rvsc.2019.08.005
Reference: YRVSC 3850
To appear in: Research in Veterinary Science
Received date: 15 February 2019
Revised date: 3 July 2019
Accepted date: 4 August 2019

Please cite this article as: F. Ahmed, F.M. Soliman, M.A. Adly, et al., Recent progress
in biomedical applications of chitosan and its nanocomposites in aquaculture: A review,
Research in Veterinary Science, https://doi.org/10.1016/j.rvsc.2019.08.005

This is a PDF file of an unedited manuscript that has been accepted for publication. As
a service to our customers we are providing this early version of the manuscript. The
manuscript will undergo copyediting, typesetting, and review of the resulting proof before
it is published in its final form. Please note that during the production process errors may
be discovered which could affect the content, and all legal disclaimers that apply to the
journal pertain.
ACCEPTED MANUSCRIPT

Recent progress in biomedical applications of chitosan and its nanocomposites in

aquaculture: A review

Authors: Fatma Ahmed1,2 , Faiza M. Soliman2 , Mohamed A. Adly2 , Hamdy A.M.

Soliman2 , Mansour El-Matbouli1 , Mona Saleh*1

1- Clinical Division of Fish Medicine; University of Veterinary Medicine, Vienna,

Austria.

PT
2- Department of Zoology, Faculty of Science, Sohag University, Sohag, Egypt.

RI
*Corresponding Author: Dr. Mona Saleh Clinical Division of Fish Medicine, University

SC
of Veterinary Medicine Veterinärplatz 1, 1210 Vienna, Austria.

Tel: 0043-1-250774708
NU
Fax: 0043-1-250775192

E-mail: mona.sale h@vetme duni.ac.at


MA

Abstract word count: 173

Manuscript word count: 3986


ED

Number of references: 169

Number of figures: 4
T
EP

Tables: 5

The authors declare that they don’t have any commercial associations, current and within
C

the past five years, that might pose a potential, perceived or real conflict of interest.
AC

Abstract

Chitosan nanoparticles (CSNPs) are the nanostructures of chitosan biopolymer which

is derived from chitin polysaccharide, the main component of crustacean shells. Chitosan is a

biocompatible, nontoxic and biodegradable polymer soluble in acidic solutions and easily
ACCEPTED MANUSCRIPT

excreted from kidneys. It is widely used in medical and pharmaceutical applications

including artificial matrices for tissue engineering, drug transport, targeted drug delivery and

protein or gene delivery. The antimicrobial activities of chitosan and CSNPS against different

bacterial, fungal and viral pathogens made them valuable for several biological applications

including food preservation purposes. In addition, they have immunomodulatory effects on

fish and crustaceans providing direct positive impact on aquaculture and fish farming

PT
industry. Sustained release of some bioactive ingredients such as hormones, vitamins,

RI
nutrients and antioxidants has improved the biological activities of fish. Furthermore, CSNPs

SC
have recently been employed to diagnose fish diseases. In this review, we present the medical

and biological applications of chitosan and CSNPs on aquatics to provide an update on recent
NU
advances and the potential for further advanced applications for aquaculture in the future.
MA

Keywords

Chitosan nanoparticles, Biomedical applications, Antimicrobial agents, Immunomodulation,


ED

Fish and seafood preservation


T

Table of contents:
EP

1. Introduction
C

2. Preparation and characterization of CSNPs


AC

3. Biomedical applications of chitosan and nano-chitosan complexes on aquatics

3.1 Immunomodulation

3.2 Delivery

3.3 Sustained release of bioactive ingredients

3.4 Potential biomedical fish disease diagnosis

3.5 Fish and seafood preservation

Conclusions
ACCEPTED MANUSCRIPT

References

1. Introduction

Chitosan is a natural polysaccharide prepared by alkaline N-de-acetylation of chitin (Zhao

et al., 2011) that is a water insoluble linear polysaccharide consists of repeated N-acetyl-D-

PT
glucosamine units which are linked by β-(1→4) glycosidic bonds (Gomes et al., 2017). It is a

biocompatible, nontoxic and biodegradable polymer soluble in acidic aqueous solutions (Ji et

RI
al., 2015), thus it is easily excreted from the kidney. Therefore, chitosan and its derivatives

SC
are widely used in medical and pharmaceutical applications (Pan et al., 2002) (Kim et al.,

2005). Solubility of chitosan depends mainly on two items; the average degree of acetylation
NU
and the distribution of acetyl groups along the polymer chains (Brugnerotto et al., 2001).
MA

Owing to the mucoadhesive properties of chitosan, it could be used as a vehicle improving

penetration of large molecules across mucosal surfaces (Xu and Du, 2003). However, its

cytotoxicity was found to be variable in freshwater and salt water media; the lower the
ED

salinity, the higher the cytotoxicity (Apetroaei et al., 2018). Therefore, we can expect
T

advanced applications of chitosan on marine aquacultures.


EP

Nanoparticles are small solid particles with a size range of 1–1000 nm providing bigger
C

surface areas for the attachment of bioactive ingredients (Jia et al., 2003) and a shorter
AC

diffusion path for immobilization (Kim and Grate, 2003) (Kim et al., 2005) and slow

sustainable drug release (Lanka and Mittapally, 2016) (Gomes et al., 2017). Recently,

nanoparticles have been reported as novel antimicrobial agents against several fish pathogens

(Shaalan et al., 2016). Gold, silver and zinc oxide nanoparticles were reported by our group

to combat bacterial, fungal and parasitic fish diseases (Saleh et al., 2016) (Shaalan et al.,

2017) and our ongoing research includes the application of chitosan nanoparticles as

antimicrobial and immunostimulatory agent against deleterious fish pathogens. Several


ACCEPTED MANUSCRIPT

applications in drug and vaccine delivery using CSNPs were summarized previously in

review articles (Agnihotri et al., 2004) (Elgadir et al., 2015). In this review, we aimed to

highlight the potential and significance of using chitosan and CSNPs in various applications

on aquatics encouraging more researchers to further explore the variable characteristics of

CSNPs for more benefits to the aquaculture.

PT
2. Preparation and characterization of CSNPs

RI
Chitosan could be prepared via enzymatic or chemical method. The enzymatic methods

are controlled processes form a well-defined chitosan (Younes and Rinaudo, 2015) however

SC
the chemical methods require lower coasts and then are extensively used in the preparation of
NU
chitosan for the commercial applications (Ji et al., 2015). A survey on the common

techniques used for the preparation of CSNPs from chitosan biopolymer was reported by
MA

Grenha (2012) including, ionic gelation (Qi et al., 2004), emulsion (Kataoka et al., 2000),

self-assembling (Liu et al., 2005), reverse micellar (Banerjee et al., 2002) and ultrafine
ED

milling (Zhang et al., 2012). Ionic gelation (simple ionotropic gelation) is the most widely

used method because it is a simple chemical reaction that needs few organic solvents, could
T
EP

be proceeded at room temperature and allows safe and successful encapsulation of sensitive

and fragile molecules such as vaccines and proteins (Zhao et al., 2011). Formation of CSNPs
C

starts spontaneously by ionic gelation mechanism via dropwise addition of the gelating
AC

solution, contains anionic molecules, into chitosan solution, contains positive amino groups

(NH2 +) (Huang and Lapitsky, 2012) and the process could be controlled by changing the pH

to change the charge density of both solutions. The gelating agent, tripolyphosphate (TPP)

has often been used for CSNPs preparation because it is nontoxic, multivalent, negatively

charged and able to form gels through ionic interactions under continuous stirring (Rampino

et al., 2013). Factors influencing the formation of CSNPs, such as pH, concentrations and
ACCEPTED MANUSCRIPT

ratios of components, and the method of mixing were previously reported by Nasti et al.

(2009). The Mw of chitosan affects the size of the formed nanoparticles; low Mw generates

smaller nanoparticles with tunable size, morphology, and delivery rate (Xu and Du, 2003).

The higher the Mw, the larger the nanoparticles size, and the lower Mw, the smaller the

nanoparticles size formed (Alishahi et al., 2011b). It is worth mentioning that nanoparticles

formed by the ionic gelation are characterized by limited physical and chemical stability

PT
when stored for an extended period due to particles aggregation and/or fusion. Larger

RI
particles may form because single small particles tend to aggregate and fuse together

SC
generating a larger entity (Rampino et al., 2013). Therefore, the nanoparticles solutions

should be prepared or gently mixed prior to use (Alishahi et al., 2011b).


NU
Physicochemical properties of the nanoparticles prepared from biopolymers, including

chitosan, have been assessed using different approaches. Size and particles morphology have
MA

been evaluated by scanning electron microscopy (SEM) and transmission electron

microscopy (TEM) as shown in figure 1 (Parida et al., 2013). In addition, morphology and
ED

distribution of the nanoparticles in solutions has also been studied by tapping mode atomic

force microscopy after being air dried (Ferosekhan et al., 2014). The size of nanoparticles be
T
EP

accurately measured by Fourier transform infrared (FTIR) spectra. The size distribution range

(Zeta Potential) and the average particle size (mean size) can be assessed by laser doppler
C

electrophoresis and dynamic light scattering, respectively using Zeta Sizer (Dubey et al.,
AC

2016).

3. Biomedical applications of chitosan and nano-chitosan complexes on aquatics

The NH2 + and hydroxyl (OH-) groups provide chitosan with many special

characteristics, making it suitable for chemical reactions and applicable in many areas

(Zhao et al., 2011). In addition, chitosan is characterized by its safety and significant

biological and chemical properties such as biodegradability, biocompatibility, bioactivity


ACCEPTED MANUSCRIPT

and polycationicity (Muzzarelli, 2010) (Kean and Thanou, 2010) (Plant and LaPatra,

2011). Chitosan and its nanostructures have several antimicrobial properties against the

antibiotic resistant Gram negative and Gram positive bacteria in addition to fungi (Ma et

al., 2016) (Ma et al., 2017) (Table 1). This potential antimicrobial activity needs further

investigations against major bacterial and fungal fish pathogens. Additionally, functional

studies are required to elucidate their mechanisms of action. Owing to their antimicrobial

PT
activities, chitosan nanocomposites are very important in several applications in the

RI
textile industry, for water disinfection of fish farms, in food packaging and medicine

SC
(Yang et al., 2010) (Hajipour et al., 2012) (Gokulalakshmi et al., 2015). CSNPs can bind

with particles that have negative charges on their surfaces (Wisdom et al., 2018) and this
NU
make them applicable for wide range of purposes. Bioadhesive CSNPs have been also

used as carriers, encapsulators or immobilizers for several bioactive ingredients as


MA

catechins from green tea (Dudhani and Kosaraju, 2010) (Tang et al., 2013), chlorogenic

acid from fruits and vegetables (Llaiyaraja et al., 2014) and the food preservative
ED

“Thyme” essential oil (Ghahfarokhi et al., 2016). Fish collagen/chitosan films have been

used for controlled localized drug delivery and chemotherapy (Chen et al., 2015). The
T
EP

reported biological and medical applications of chitosan-based complexes on fish and

crustaceans are summarized in this review article to highlight their benefits on the health
C

of aquatic organisms. Undoubtedly, the applications related to the immunomodulation


AC

and antimicrobial activity gained the highest importance since they have high impact on

improving the aquaculture production.

3.1. Immunomodulation

Diets supplemented with chitosan improved growth, survival and meat quality of several

freshwater and seawater fish species including, Rainbow trout (Oncorhynchus mykiss)
ACCEPTED MANUSCRIPT

(Meshkini et al., 2012), olive flounder (Paralichthys olivaceus) (Cha et al., 2008), koi

(Cyprinus carpio koi) (Lin et al., 2011), kelp grouper (Epinephelus bruneus) (Harikrishnan et

al., 2012a) (Harikrishnan et al., 2012b), turbot (Scophthalmus maximus L.) (Cui et al., 2013),

gibel carp (Carassius auratus gibelio) (Chen et al., 2014), mrigal carp (Cirrhina mrigala)

(Shanthi Mari et al., 2014), Asian seabass (Lates calcarifer) (Ranjan et al., 2014) and sea bass

(Dicentrarchus labrax) (Zaki et al., 2015). Chitosan as well as CSNPs supplemented diets

PT
enhanced the survival, growth and meat quality of Nile tilapia (Oreochromis nilotica) (Wang

RI
and Li, 2011) (Abd El-Naby et al., 2019) (Abdel-Tawwab et al., 2019) and the African catfish

SC
(Clarius gariepinus) fingerlings (Imefon Udo, 2018). However, CSNPs are more effective

than chitosan in improving growth performance and food quality status. Dietary chitosan and
NU
chitin supplemented diets can demonstrate immunomodulatory effects (Plant and LaPatra,

2011) and improve the resistance of several fish species against infections and stresses (Ji et
MA

al., 2015) by enhancing the innate immune response (Vahedi and Ghodratizadeh, 2011). For

the oral immunization against infections, the black tiger shrimp (Penaeus monodon) was
ED

vaccinated against white spot syndrome virus (WSSV) by the oral feeding on DNA vaccine

encapsulated with CSNPs (Rajeshkumar et al., 2009). Additionally, the crayfish


T
EP

(Procambarus clarkii) was protected against WSSV infection via the dietary CSNPs with

significantly higher survival rate (Sun et al., 2016). The reported studies on the
C

immunomodulation of fish against infections and stresses are summarized in table 2.


AC

However, fish immunomodulation effects of CSNPs need further studies to improve our

knowledge on how and to which extent they can enhance fish health.

3.2. Delivery

Since they have been used for longer release, CSNPs are promising carriers for drug oral

administration (Zhao et al., 2014). CSNPs have the ability to penetrate deeply into tissues
ACCEPTED MANUSCRIPT

through fine capillaries allowing efficient delivery into body organs (Vimal et al., 2014).

Chitosan and CSNPs are bioactive ingredient carriers, encapsulators and immobilizers used in

oral delivery of drugs, vitamins, nutrients, genes (DNA) and vaccines into fish

gastrointestinal tract. CSNPs enhance the mucoadhesive potential improving intestinal

absorption and oral bioavailability. Therefore, they can be incorporated with further natural

ingredients and antioxidants to enhance fish immunity (Ferosekhan et al., 2014). FTIR

PT
spectroscopy can be used to confirm cross linking of loaded bioactive ingredients (Bhat et al.,

RI
2018).

CSNPs are used as stabilizers for bio-molecules and dietary supplements as RNA and α-

SC
tocopherol with limited bioavailability, due to unsuitable pH and intestinal cell repulsion, and
NU
short half life in blood which significantly improve growth, immunity and disease resistance

of fish (Ferosekhan et al., 2014).


MA

In vitro, Zein/chitosan complexes were used for food supplementation and considered

promising for delivery of hydrophobic molecules (Luo and Wang, 2014) (Luo et al., 2011).
ED

Currently, gene (DNA) delivery is considered the most effective strategy for fish vaccination

(Mandal, 2010). Delivery systems improve the solubility, bioavailability and sustained
T
EP

release of hydrophobic drugs and protect therapeutic agents from degradation, thereby,

facilitating cell and tissue targeting (Shi et al., 2010). CSNPs can be used as gene vectors for
C

the rapidly degrading molecules such as DNA, plasmids (pDNA), siRNA (Vimal et al.,
AC

2014). DNA immunization of fish is usually applied either by direct intramuscular injection

(Kumar et al., 2007) or by immersion (Fernandez-Alonso et al., 2001). However, oral

vaccination is the ideal way for fish immunization as it can effectively induce the mucosal

immunity of the gastrointestinal tract avoiding the exhausting injection of large numbers of

fish in aquacultures (Ramos et al., 2005) (Tian et al., 2008). Because chitosan is resistant to

degradation, it can protect the activity of encapsulated compounds from destruction by the
ACCEPTED MANUSCRIPT

critical conditions inside gastrointestinal tract enhancing their absorption (Aranaz et al.,

2009). Therefore, CSNPs are one of the most widely used drug and gene delivery vehicles in

aquaculture (Ji et al., 2015). Immune genes expression depends on CSNPs/plasmid ratio, pH

and the combination of deacetylation degree (DD) and M W of chitosan (Lavertu et al., 2006).

Oral plasmid DNA (pDNA) delivery into gastrointestinal tract of fish can be achieved via

encapsulation with chitosan or its micro or nanoparticles. OMPs of bacteria act as antigenic

PT
sites with high immunogenic properties on the outer surfaces of bacterial cell membranes

RI
(Dubey et al., 2016). Antigen vaccines constructed using recombinant OMPs of several

SC
pathogens have been used for fish vaccination via intramuscular injection (Kumar et al.,

2007). Currently, CSNPs provide oral vaccines administration system that enables higher
NU
tissue availability enhancing fish immunity. Figure 2 showing size distribution intensity and

TEM of CSNPs encapsulated with Edwardsiella tarda OmpA for the oral vaccination of
MA

fringed-Lipped peninsula carp (Dubey et al., 2016). Moreover, the combination between

microspheres of chitosan and other biodegradable composites has been used as antigen
ED

carriers for both parenteral and oral immunization of fish. Chitosan and gantrez has been

effective in incorporating the surface antigens of P. dicentrarchi for the stimulation of innate
T
EP

immune response against scuticociliatosis (León-Rodríguez et al., 2013). Reports on the

delivery of bio-molecules and vaccines into fish tissues via encapsulation with chitosan, its
C

microspheres and nanocomplexes are summarized in table 3.


AC

3.3. Sustained release of bioactive ingredients

Nano-encapsulation is one of the most promising strategies for controlled release of

bioactive compounds at the right time and the right site with efficient cell absorption

(Ezhilarasi et al., 2013). CSNPs have specific physical properties (size, surface charge, or

loading capacity), which allow controlled delivery, improve targeting and stimulation of the

immune system (Ji et al., 2015) and provide protection against gastrointestinal conditions
ACCEPTED MANUSCRIPT

(Luo et al., 2011). In fish, CSNPs have been reported in the controlled release of some

bioactive components including vitamins (Alishahi et al., 2011a) and proteins such as

enzymes and hormones (Wisdom et al., 2018). Figure 3 shows particle size distribution of

leutinizing hormone releasing hormone (LHRH) encapsulated CSNPs and the effect of their

injection on the ovarian histology (Rather et al., 2013).

CSNPs can persist inside cells up to two weeks and are considered as good carriers

PT
for sustained release of various biological substances. Moreover, they can diffuse into the

RI
cytoplasm and nucleus, therefore they are able to pass from mother to daughter cells through

SC
several mitotic cycles (Malatesta et al., 2015). They are enriching additives for freshwater

and marine organisms enhancing their immunity (Sun et al., 2016) (Abd El-Naby et al.,
NU
2019). When encapsulated with antioxidants, they can penetrate intestinal epithelium

providing controlled oral delivery of bioactive ingredients (Rojas et al., 2015). The
MA

functionality, the thermal stability at 45°C and the shelf life of vitamin C has been enhanced

by encapsulation with CSNPs (Rojas et al., 2015) in vitro as well as in vivo (Alishahi et al.,
ED

2011a). In vitro, the stability and control release of vitamin C has been improved through

encapsulation with double layer coating from Zein/CS nanoparticle complex (Dong and
T
EP

Wang, 2016). Similarly, the chemical stability of vitamin D3 was enhanced when loaded on

zein nanoparticles coated with carboxymethyl chitosan (Luo et al., 2012). Additionally,
C

significant increase in the total antioxidant capacity of vitamin C has been seen in zebrafish
AC

liver cell line when loaded on CSNPs without affecting cell viability (Jiménez-Fernández et

al., 2014). In vivo, vitamin C has been control released from CSNPs into the gastrointestinal

tract and serum of some fish species inducing their nonspecific immunity. In addition,

releasing of vitamin C from CSNPs was pH dependent and its shelf life increased when

compared with the non encapsulated form (Alishahi et al., 2011b).


ACCEPTED MANUSCRIPT

CSNPs are sufficiently stable to carry and protect proteins (Katas et al., 2013). The

higher Mw and degree of deacetylation (DD) of chitosan, the more the encapsulation

efficiency (EE) and the slower the protein release rate obtained. However, increasing in

loading capacity of proteins decreases the EE and accelerates its release rate from

nanocomplexes (Xu and Du, 2003). EE of bovine serum albumin (BSA) as a protein model

loaded on CSNPs reached up to 80% provided continued release for up to 1 week (Calvo et

PT
al., 1997). Recently, BSA/siRNA was controlled released for up to 48 h in vitro when loaded

RI
on CSNPs (Katas et al., 2013). Additionally, CSNPs improved mucoadhesive properties and

SC
permeability when adsorbed in vitro onto the excised carp intestinal mucosa (Liu et al.,

2012b) without toxicity or pathological damage in carp tissues (Liu et al., 2012a) (Liu et al.,
NU
2013).

Oral administration of protein derivatives including, hormones and enzymes in diets is


MA

limited because of its thermal nature, pH specificity and intestinal damage hazard.

Conjugation and encapsulation with CSNPs provides oral controlled delivery systems to
ED

overcome the problem of their short life in blood and enhance the efficacy and safety of their

oral administration. Figure 4 shows particle size distribution of trypsin enzyme encapsulated
T
EP

with CSNPs and the effect of their oral administration on the intestinal histology (Kumari et

al., 2013). Recently, nanoconjugation with chitosan was reported for the oral administration
C

of various gonadal enzymes avoiding multiple injections of fish (Wisdom et al., 2018).
AC

Studies using chitosan and CSNPs for vitamin and protein delivery into fish are presented in

table 4.

3.4. Potential biomedical fish disease diagnosis (fish pathogen detection)

Fernandes et al. (2015) have used chitosan-based nanoparticles in the formation of a

highly sensitive electrochemical genosensor for the detection of fish pathogenic Aeromonas

spp. in spiked tap water. This genosensor is a promising potential biomedical tool for fish
ACCEPTED MANUSCRIPT

diseases diagnosis. It has been constructed from multi-walled carbon nanotubes chitosan–

bismuth complex (MWCNT–Chi–Bi) and lead sulphide nanoparticles. Thiol-modified

oligonucleotides have been used as fixing probe DNA. Lead sulphide NPs have been capped

via amide bond with 5′- (NH2) oligonucleotides and used as signalizing probe DNA (sz-

DNA). The two probes and the DNA of Aeromonas sequence are forming a hybrid complex.

Lead nanoparticles released from sz-DNA have been electrodeposited on the surface of glass

PT
carbon electrode provided with the MWCNT–Chi–Bi complex for improvement of lead

RI
deposition and electrical signal transmission. Finally, the differential pulse voltammetry is

SC
used in detecting the hybridization signal.

3.5. Fish and Seafood preservation


NU
In spite of the reported toxicity of CSNPs on zebrafish embryos (Hu et al., 2011)
MA

(Wang et al., 2016), there is a good number of investigations considering the inorganic

biodegradable nanoparticles, including CSNPs safe. For seawater aquaculture, chitosan,


ED

mostly from marine sources, was reported as the most promising stabilizer for seafood

products that are highly susceptible to deterioration because of their high water activity,
T
EP

neutral pH and their high contents of autolytic enzymes, polyunsaturated fatty acids, and free

amino acids (Alishahi and Aïder, 2012). Several in vitro and in vivo studies have considered
C

chitosan safe, nontoxic (Kean and Thanou, 2010) and effective antibacterial agent for food
AC

preservation (Islam et al., 2011) and fish additive (Aref et al., 2018) improving their quality

(Gomes et al., 2017). CSNPs are non genotoxic and can enhance food protection by reducing

the permeation of gases, minimizing odor loss and increasing mechanical strength and

thermal stability (de Lima et al., 2010). Moreover, CSNPs have been incorporated in

biosensors for detection of xanthine in fish meat as an indicator for meat freshness (Devi et

al., 2012) (Devi et al., 2013). Owing to its antioxidative and antimicrobial properties,
ACCEPTED MANUSCRIPT

chitosan and its nanocomplexes as N-stearoyl O-butylglyceryl/chitosan have been used for

encapsulation and sustained release of fish oil for protection against oxidative degradation by

decreasing heat transfer and increasing the thermal stability of the encapsulated fish oil

(Chatterjee and Judeh, 2015). The quality of the unsaturated fatty acids (ω-3) content

obtained from carp viscera has been preserved against oxidative degradation when

encapsulated with CSNPs (Esquerdo et al., 2015).

PT
Nanocapsules constructed from the encapsulation of the liquid smoke of coconut shell

RI
have been obtained by the condensation of coconut wood smoke under limited oxygen

SC
conditions, with chitosan and maltodextrin and used for tuna fish preservation at ambient

temperature (Saloko et al., 2014). During refrigeration of fish meat at 4°C, chitosan
NU
composites have been used for the inhibition of the bacterial growth and reduction of the
MA

formed volatile bases and oxidation products (Mohan et al., 2012). Additionally, CSNPs have

been used in bio-nanocomposite technology applications for fabrication and strengthening


ED

edible films of food packaging and preservative coatings to increase shelf life and decrease

the deterioration of fish meat during refrigerating and freezing storage (De Moura et al.,
T

2008). Chitosan coatings reduce the off-flavour compounds and improve flavour retention of
EP

chilled fish (Yu et al., 2018). Combination of chitosan with natural preservatives such as
C

plant extracts, essential oils or antioxidants significantly improves the preservation quality
AC

(Zarei et al., 2015) (Qiu et al., 2014) (Li et al., 2012) (T. Li et al., 2013).

Recently, the physicochemical properties of the food edible films constructed from

fish gelatine (FG) fillers have been enhanced via incorporation with chitosan (Hosseini et al.,

2013). Strong, applicable, eco-friendly and bio-nanocomposite films have been produced via

reinforcing the FG matrix by incorporation with CSNPs (Hosseini et al., 2015) (Hosseini et

al., 2016). These films are safe to eat, hence, can be used in manufacturing the packaging

materials of food and helps in decreasing the environmental pollution resulting from plastic
ACCEPTED MANUSCRIPT

materials usually used for food packaging (Feng et al., 2016). FG/chitosan coating and film

can control the bacterial contamination, provide good quality characteristics and extend the

meat shelf life, however, the coating is better than the film in reducing the lipid oxidation of

fish fillets during chilled storage (Nowzari et al., 2013). Additionally, FG/chitosan edible

films with the plasticizer hydrophobic D-limonene have provided excellent mechanical

resistance against light and water penetration and enhanced the antibacterial properties of the

PT
films (Yao et al., 2017). Further, combination with natural preservatives significantly

RI
enhances the preservative characters of FG/chitosan-based edibles (Gómez-Estaca et al.,

SC
2010). During freezing, the aqueous chitosan solution has been used to form coating for

frozen storage of fish meat (Fan et al., 2009). Supplying the edible coatings with chitosan and
NU
CSNPs introduces effective barriers for protection of fish meat against deterioration for long

frozen storage periods of fish fingers (Abdou et al., 2012). It provides considerable
MA

improvements in chemical, microbial, sensory and textural properties even at -18°C frozen

storage (Berizi et al., 2018). The applications for fish meat preservation during chilled and
ED

frozen storage are presented in table 5.

On crustaceans, chitosan has been used as an antimicrobial agent to reduce the microbial
T
EP

loads on the surface of fresh shrimp and for preservation of shrimp meat (Alfaro et al., 2018).

Chitosan combined with shrimp protein/lipid concentrate has been used for coating and
C

improving shrimp quality during cold storage (Arancibia et al., 2015). Chitosan coatings
AC

incorporated with some bioactive factors such as oregano and thyme essential oil preserved

ready-to-eat peeled shrimp and controlled the growth of microorganisms during refrigeration

storage (Carrión-Granda et al., 2016). In addition, chitosan coating combined with

pomegranate peel extract significantly lowered the melanosis and increased the quality of

Pacific white shrimp (Litopenaeus vannamei) during iced storage up to 10 days (Yuan et al.,

2016). Moreover, gelatin coating supplemented with chitosan improved the quality of shrimp
ACCEPTED MANUSCRIPT

under refrigerated storage (Farajzadeh et al., 2016). Recently, a novel packaging film had

been produced from chitosan coatings containing the essential oil of (Ziziphora

clinopodioides), pomegranate peel extract and cellulose nanoparticles, either separately or in

combination. High antibacterial activity had been observed during refrigerating storage of

peeled shrimp for up to 11 days (Mohebi and Shahbazi, 2017). More recently,

chitosan/carvacrol coating has been used for active packaging and extending the shelf life of

PT
L. vannamei during iced storage for 10 days (Wang et al., 2018). CSNPs have been used as

RI
glazing material for controlling lipid oxidation and reducing aerobic counts, yeasts and molds

SC
during 30 days frozen storage of shrimp at -21°C (Solval et al., 2014). Chitosan applied by

vacuum tumbling was helpful in the preservation of shrimp meat quality frozen at -20°C
NU
(Chouljenko et al., 2016). Similarly, vacuum tumbling with chitosan and CSNPs reduced the

aerobic plate counts and lipid oxidation besides maintaining the desired physicochemical
MA

properties of shrimp during frozen storage at -20°C (Chouljenko et al., 2017).


ED

Conclusions

There is a potential need for the development of novel encapsulation or immobilization


T
EP

carriers of several bioactive ingredients for several applications in aquaculture. This review

highlights the reported applications of chitosan and its nanostructure complexes on fish and
C

crustaceans. CSNPs have many favorable biological properties such as safety,


AC

biocompatibility, biodegradability and antibacterial ability that make them promising

candidates for several applications in fish medicine. They are promising carriers for

biological ingredients including DNA vaccines, dietary supplements and nutrients such as

RNA nucleotides, antioxidants like vitamin C and even proteins including hormones and

enzymes. Dietary CSNPs exhibit bioactive immune-inducing activity and antimicrobial

properties on fish and crustaceans. However, additional studies are required particularly in
ACCEPTED MANUSCRIPT

the field of fish disease diagnosis and the oral delivery of DNA vaccines, siRNA, and

bioactive ingredients including vitamins, proteins and nutrients aiming at inducing of fish

immunity for the protection against infections.

Acknowledgement

We acknowledge support for a PhD scholarship offered for Fatma El Zahraa Ahmed by

PT
mission sectors in the ministry of higher education, Egypt.

RI
References

SC
Aathi, K., Ramasubramanian, V., Uthayakumar, V., Munirasu, S., 2013. Effect of
NU
chitosan supplemented diet on survival, growth, hematological, biochemical and

immunological responses of Indian major carp Labeo Rohita. Int. Res. J. Pharm.
MA

4, 141–147. https://doi.org/10.7897/2230-8407.04529

Abd El-Naby, F.S., Naiel, M.A.E., Al-Sagheer, A.A., Negm, S.S., 2019. Dietary chitosan
ED

nanoparticles enhance the growth, production performance, and immunity in


T

Oreochromis niloticus. Aquaculture 501, 82–89.


EP

https://doi.org/10.1016/j.aquaculture.2018.11.014

Abdel-Tawwab, M., Razek, N.A., Abdel-Rahman, A.M., 2019. Immunostimulatory effect


C
AC

of dietary chitosan nanoparticles on the performance of Nile tilapia, Oreochromis

niloticus (L.). Fish Shellfish Immunol. 88, 254–258.

https://doi.org/10.1016/j.fsi.2019.02.063

Abdou, E.S., Osheba, A, Sorour, M., 2012. Effect of chitosan and chitosan-nanoparticles

as active coating on microbiological characteristics of fish fingers. Int. J. Appl.

Sci. Technol. 2, 158–169.


ACCEPTED MANUSCRIPT

Abu-Elala, N.M., Mohamed, S.H., Zaki, M.M., Eissa, A.E., 2015. Assessment of the

immune-modulatory and antimicrobial effects of dietary chitosan on Nile tilapia

(Oreochrmis niloticus) with special emphasis to its bio-remediating impacts. Fish

Shellfish Immunol. 46, 678–685. https://doi.org/10.1016/j.fsi.2015.08.004

Agnihotri, S.A., Mallikarjuna, N.N., Aminabhavi, T.M., 2004. Recent advances on

chitosan-based micro- and nanoparticles in drug delivery. J. Control. Release 100, 5–

PT
28.

RI
Alboghbeish, H., Khodanazary, A., 2018. The comparison of quality characteristics of

SC
refrigerated Carangoides coeruleopinnatus fillets with chitosan and nanochitosan
NU
coating 19, 957–967.

Alfaro, L., Chotiko, A., Chouljenko, A., Janes, M., King, J.M., Sathivel, S., 2018.
MA

Development of water-soluble chitosan powder and its antimicrobial effect against

inoculated Listeria innocua NRRL B-33016 on shrimp. Food Control 85, 453–
ED

458. https://doi.org/10.1016/j.foodcont.2017.10.023
T

Alishahi, A., Aïder, M., 2012. Applications of chitosan in the seafood industry and
EP

aquaculture: A review. Food Bioprocess Technol. 5, 817–830.

https://doi.org/10.1007/s11947-011-0664-x
C
AC

Alishahi, A., Mirvaghefi, A., Tehrani, M.R., Farahmand, H., Koshio, S., Dorkoosh, F.A.,

Elsabee, M.Z., 2011a. Chitosan nanoparticle to carry vitamin C through the

gastrointestinal tract and induce the non-specific immunity system of rainbow

trout (Oncorhynchus mykiss). Carbohydr. Polym. 86, 142–146.

https://doi.org/10.1016/j.carbpol.2011.04.028

Alishahi, A., Mirvaghefi, A., Tehrani, M.R., Farahmand, H., Shojaosadati, S.A.,

Dorkoosh, F.A., Elsabee, M.Z., 2011b. Shelf life and delivery enhancement of
ACCEPTED MANUSCRIPT

vitamin C using chitosan nanoparticles. Food Chem. 126, 935–940.

https://doi.org/10.1016/j.foodchem.2010.11.086

Alishahi, M., Esmaeili Rad, A., Zarei, M., Ghorbanpour, M., 2014. Effect of dietary

chitosan on immune response and disease resistance in Cyprinus carpio. Iran. J.

Vet. Med. Eff. 8, 125–133.

PT
Apetroaei, M.R., Pădureţu, C., Rău, I., Schroder, V., 2018. New-chitosan characterization

and its bioassay in different salinity solutions using Artemia salina as bio tester.

RI
Chem. Pap. 72, 1853–1860. https://doi.org/10.1007/s11696-018-0440-8

SC
Aranaz, I., Mengibar, M., Harris, R., Panos, I., Miralles, B., Acosta, N., Galed, G., Heras,
NU
A., 2009. Functional characterization of chitin and chitosan. Curr. Chem. Biol. 3,

203–230. https://doi.org/10.2174/187231309788166415
MA

Arancibia, M.Y., López-Caballero, M.E., Gómez-Guillén, M.C., Montero, P., 2015.

Chitosan coatings enriched with active shrimp waste for shrimp preservation.
ED

Food Control 54, 259–266. https://doi.org/10.1016/j.foodcont.2015.02.004


T

Aref, S., Morsy, N., Habiba, R.A., Zayat, F., 2018. Effect of transglutaminase enzyme,
EP

chitosan and rosemary extract on some quality characteristics of ready to eat fish

fingers made from catfish (Clarias gariepinus) during frozen storage 8, 716–731.
C
AC

Avadi, M.R., Sadeghi, A.M.M., Tahzibi, A., Bayati, K., Pouladzadeh, M., Zohuriaan-

Mehr, M.J., Rafiee-Tehrani, M., 2004. Diethylmethyl chitosan as an antimicrobial

agent: Synthesis, characterization and antibacterial effects. Eur. Polym. J. 40,

1355–1361. https://doi.org/10.1016/j.eurpolymj.2004.02.015

Banerjee, T., Mitra, S., Kumar Singh, A., Kumar Sharma, R., Maitra, A., 2002.

Preparation, characterization and biodistribution of ultrafine chitosan

nanoparticles. Int. J. Pharm. 243, 93–105. https://doi.org/10.1016/S0378-


ACCEPTED MANUSCRIPT

5173(02)00267-3

Behera, T., Swain, P., 2014. Antigen encapsulated alginate-coated chitosan microspheres

stimulate both innate and adaptive immune responses in fish through oral

immunization. Aquac. Int. 22, 673–688. https://doi.org/10.1007/s10499-013-

9696-8

PT
Behera, T., Swain, P., 2013. Alginate-chitosan-PLGA composite microspheres induce

both innate and adaptive immune response through parenteral immunization in

RI
fish. Fish Shellfish Immunol. 35, 785–791.

SC
https://doi.org/10.1016/j.fsi.2013.06.012 NU
Berizi, E., Hosseinzadeh, S., Shekarforoush, S.S., Barbieri, G., 2018. Microbial,

chemical, textural and sensory properties of coated rainbow trout by chitosan


MA

combined with pomegranate peel extract during frozen storage. Int. J. Biol.

Macromol. 106, 1004–1013. https://doi.org/10.1016/j.ijbiomac.2017.08.099


ED

Bhat, I.A., Nazir, M.I., Ahmad, I., Pathakota, G.B., Chanu, T.I., Goswami, M., Sundaray,

J.K., Sharma, R., 2018. Fabrication and characterization of chitosan conjugated


T
EP

eurycomanone nanoparticles: In vivo evaluation of the biodistribution and toxicity

in fish. Int. J. Biol. Macromol. 112, 1093–1103.


C

https://doi.org/10.1016/j.ijbiomac.2018.02.067
AC

Brugnerotto, J., Desbrières, J., Heux, L., Mazeau, K., Rinaudo, M., 2001. Overview on

structural characterization of chitosan molecules in relation with their behavior in

solution. Macromol. Symp. 168, 1–20. https://doi.org/10.1002/1521-

3900(200103)168:1<1::AID-MASY1>3.0.CO;2-W

Calvo, P., Remuñán-López, C., Vila-Jato, J.L., Alonso, M.J., 1997. Novel hydrophilic

chitosan-polyethylene oxide nanoparticles as protein carriers. J. Appl. Polym. Sci.


ACCEPTED MANUSCRIPT

63, 125–132. https://doi.org/10.1002/(SICI)1097-4628(19970103)63:1<125::AID-

APP13>3.0.CO;2-4

Carrión-Granda, X., Fernández-Pan, I., Jaime, I., Rovira, J., Maté, J.I., 2016.

Improvement of the microbiological quality of ready-to-eat peeled shrimps

(Penaeus vannamei) by the use of chitosan coatings. Int. J. Food Microbiol. 232,

PT
144–149. https://doi.org/10.1016/j.ijfoodmicro.2016.05.029

Cha, S.H., Lee, J.S., Song, C.B., Lee, K.J., Jeon, Y.J., 2008. Effects of chitosan-coated

RI
diet on improving water quality and innate immunity in the olive flounder,

SC
Paralichthys olivaceus. Aquaculture 278, 110–118.

https://doi.org/10.1016/j.aquaculture.2008.01.025
NU
Chatterjee, S., Judeh, Z.M.A., 2015. Encapsulation of fish oil with N-stearoyl O-
MA

butylglyceryl chitosan using membrane and ultrasonic emulsification processes.

Carbohydr. Polym. 123, 432–442. https://doi.org/10.1016/j.carbpol.2015.01.072


ED

Chen, M.-M., Huang, Y.-Q., Cao, H., Liu, Y., Guo, H., Chen, L.S., Wang, J.-H., Zhang,

Q.-Q., 2015. Collagen/chitosan film containing biotinylated glycol chitosan


T
EP

nanoparticles for localized drug delivery. Colloids Surfaces B Biointerfaces 128,

339–346. https://doi.org/10.1016/j.colsurfb.2015.02.024
C
AC

Chen, Y., Zhu, X., Yang, Y., Han, D., Jin, J., Xie, S., 2014. Effect of dietary chitosan on

growth performance, haematology, immune response, intestine morphology,

intestine microbiota and disease resistance in gibel carp (Carassius auratus

gibelio). Aquac. Nutr. 20, 532–546. https://doi.org/10.1111/anu.12106

Chouljenko, A., Chotiko, A., Bonilla, F., Moncada, M., Reyes, V., Sathivel, S., 2017.

Effects of vacuum tumbling with chitosan nanoparticles on the quality

characteristics of cryogenically frozen shrimp. LWT - Food Sci. Technol. 75,


ACCEPTED MANUSCRIPT

114–123. https://doi.org/10.1016/j.lwt.2016.08.029

Chouljenko, A., Chotiko, A., Reyes, V., Alfaro, L., Liu, C., Dzandu, B., Sathivel, S.,

2016. Application of water-soluble chitosan to shrimp for quality retention. LWT

- Food Sci. Technol. 74, 571–579. https://doi.org/10.1016/j.lwt.2016.08.024

Chowdappa, P., Gowda, S., Chethana, C.S., Madhura, S., 2014. Antifungal activity of

PT
chitosan-silver nanoparticle composite against Colletotrichum gloeosporioides

associated with mango anthracnose. African J. Microbiol. Res. 8, 1803–1812.

RI
https://doi.org/10.5897/AJMR2013.6584

SC
Chung, Y., Su, Y., Chen, C., Jia, G., Wang, H., Wu, J.C.G., Lin, J., 2004. Relationship
NU
between antibacterial activity of chitosan and surface characteristics of cell wall.

Acta Pharmacol Sin 932–936.


MA

Cui, L., Xu, W., Ai, Q., Wang, D., Mai, K., 2013. Effects of dietary chitosan

oligosaccharide complex with rare earth on growth performance and innate


ED

immune response of turbot, Scophthalmus maximus L. Aquac. Res. 44, 683–690.

https://doi.org/10.1111/j.1365-2109.2011.03072.x
T
EP

de Lima, R., Feitosa, L., Pereira, A. do E.S., De Moura, M.R., Aouada, F.A., Mattoso,

L.H.C., Fraceto, L.F., 2010. Evaluation of the genotoxicity of chitosan


C
AC

nanoparticles for use in food packaging films. J. Food Sci. 75, 89–96.

https://doi.org/10.1111/j.1750-3841.2010.01682.x

De Moura, M.R., Avena-Bustillos, R.J., McHugh, T.H., Krochta, J.M., Mattoso, L.H.C.,

2008. Properties of novel hydroxypropyl methylcellulose films containing

chitosan nanoparticles. J. Food Sci. 73. https://doi.org/10.1111/j.1750-

3841.2008.00872.x

de Paz, L.E.C., Resin, A., Howard, K.A., Sutherland, D.S., Wejse, P.L., 2011.
ACCEPTED MANUSCRIPT

Antimicrobial Effect of chitosan nanoparticles on streptococcus mutans biofilms.

Appl. Environ. Microbiol. 77, 3892–3895. https://doi.org/10.1128/AEM.02941-10

Devi, R., Yadav, S., Pundir, C.S., 2012. Amperometric determination of xanthine in fish

meat by zinc oxide nanoparticle/chitosan/multiwalled carbon nanotube/polyaniline

composite film bound xanthine oxidase. Analyst.

PT
Devi, R., Yadav, Sandeep, Nehra, R., Yadav, Sujata, Pundir, C.S., 2013. Electrochemical

biosensor based on gold coated iron nanoparticles/chitosan composite bound

RI
xanthine oxidase for detection of xanthine in fish meat. J. Food Eng. 115, 207–

SC
214. https://doi.org/10.1016/j.jfoodeng.2012.10.014
NU
Divya, K., Vijayan, S., George, T.K., Jisha, M.S., 2017. Antimicrobial properties of

chitosan nanoparticles: Mode of action and factors affecting activity. Fibers


MA

Polym. 18, 221–230. https://doi.org/10.1007/s12221-017-6690-1

Dong, F., Wang, Y., 2016. Encapsulation of vitamin C by a double- layer zein /chitosan
ED

structure with improved. Int. J. Nanomedicine Nanosurgery 2, 1–7.


T

Du, W.L., Xu, Y.L., Xu, Z.R., Fan, C.L., 2008. Preparation, characterization and
EP

antibacterial properties against E. coli K88 of chitosan nanoparticle loaded copper

ions. Nanotechnology 19. https://doi.org/10.1088/0957-4484/19/8/085707


C
AC

Du, W.L., Niu, S.S., Xu, Y.L., Xu, Z.R., Fan, C.L., 2009. Antibacterial activity of

chitosan tripolyphosphate nanoparticles loaded with various metal ions.

Carbohydr. Polym. 75, 385–389. https://doi.org/10.1016/j.carbpol.2008.07.039

Dubey, S., Avadhani, K., Mutalik, S., Sivadasan, S., Maiti, B., Girisha, S., Venugopal,

M., Mutoloki, S., Evensen, Ø., Karunasagar, I., Munang′andu, H., 2016.

Edwardsiella tarda OmpA encapsulated in chitosan nanoparticles shows superior

protection over inactivated whole cell vaccine in orally vaccinated fringed-lipped


ACCEPTED MANUSCRIPT

peninsula carp (Labeo fimbriatus). Vaccines 4, 40.

https://doi.org/10.3390/vaccines4040040

Dudhani, A.R., Kosaraju, S.L., 2010. Bioadhesive chitosan nanoparticles: Preparation and

characterization. Carbohydr. Polym. 81, 243–251.

https://doi.org/10.1016/j.carbpol.2010.02.026

PT
Elgadir, M.A., Uddin, M.S., Ferdosh, S., Adam, A., Chowdhury, A.J.K., Sarker, M.Z.I.,

2015. Impact of chitosan composites and chitosan nanoparticle composites on

RI
various drug delivery systems: A review. J. Food Drug Anal. 23, 619–629.

SC
https://doi.org/10.1016/j.jfda.2014.10.008
NU
Esquerdo, V.M., Dotto, G.L., Pinto, L.A.A., 2015. Preparation of nanoemulsions

containing unsaturated fatty acid concentrate-chitosan capsules. J. Colloid


MA

Interface Sci. 445, 137–142. https://doi.org/10.1016/j.jcis.2014.12.094

Ezhilarasi, P.N., Karthik, P., Chhanwal, N., Anandharamakrishnan, C., 2013.


ED

Nanoencapsulation techniques for food bioactive components: A review. Food

Bioprocess Technol. 6, 628–647. https://doi.org/10.1007/s11947-012-0944-0


T
EP

Fan, W., Sun, J., Chen, Y., Qiu, J., Zhang, Y., Chi, Y., 2009. Effects of chitosan coating

on quality and shelf life of silver carp during frozen storage. Food Chem. 115, 66–
C
AC

70. https://doi.org/10.1016/j.foodchem.2008.11.060

Farajzadeh, F., Motamedzadegan, A., Shahidi, S.A., Hamzeh, S., 2016. The effect of

chitosan-gelatin coating on the quality of shrimp (Litopenaeus vannamei) under

refrigerated condition. Food Control 67, 163–170.

https://doi.org/10.1016/j.foodcont.2016.02.040

Feng, X., Bansal, N., Yang, H., 2016. Fish gelatin combined with chitosan coating

inhibits myofibril degradation of golden pomfret (Trachinotus blochii) fillet


ACCEPTED MANUSCRIPT

during cold storage. Food Chem. 200, 283–292.

https://doi.org/10.1016/j.foodchem.2016.01.030

Fernandes, A.M., Abdalhai, M.H., Ji, J., Xi, B.W., Xie, J., Sun, J., Noeline, R., Lee, B.H.,

Sun, X., 2015. Development of highly sensitive electrochemical genosensor based

on multiwalled carbon nanotubes-chitosan-bismuth and lead sulfide nanoparticles

for the detection of pathogenic Aeromonas. Biosens. Bioelectron. 63, 399–406.

PT
https://doi.org/10.1016/j.bios.2014.07.054

RI
Fernandez-Alonso, M., Rocha, A., Coll, J.M., 2001. DNA vaccination by immersion and

SC
ultrasound to trout viral haemorrhagic septicaemia virus. Vaccine 19, 3067–3075.

https://doi.org/10.1016/S0264-410X(01)00046-9
NU
Fernández-Díaz, C., Coste, O., Malta, E. jan, 2017. Polymer chitosan nanoparticles
MA

functionalized with Ulva ohnoi extracts boost in vitro ulvan immunostimulant

effect in Solea senegalensis macrophages. Algal Res. 26, 135–142.


ED

https://doi.org/10.1016/j.algal.2017.07.008

Ferosekhan, S., Gupta, S., Singh, A.R., Rather, M.A., Kumari, R., Kothari, D.C., Pal,
T
EP

A.K., Jadhao, S.B., 2014. RNA-Loaded chitosan nanoparticles for enhanced

growth, immunostimulation and disease resistance in fish. Curr. Nanosci. 10, 453–
C

464. https://doi.org/10.2174/1573413710666140115220300
AC

Ghahfarokhi, G.M., Barzegar, M., Sahari, M.A., Azizi, M.H., 2016. Enhancement of

thermal stability and antioxidant activity of thyme essential oil by encapsulation in

chitosan nanoparticles. J. Agric. Sci. Technol. 18, 1781–1792.

Gokulalakshmi, E., Vanitha, M.C., Ramalingam, K., Srikumaran, N., 2015. Extraction

and characterization of chitosan nanoparticles from fish waste and its applications

on waste water treatment of fish food processing industry. J Biotechnol Biomater


ACCEPTED MANUSCRIPT

5, 341.

Gomes, L.P., Paschoalin, V.M.F., Del Aguila, E.M., 2017. Chitosan nanoparticles:

Production, physicochemical characteristics and nutraceutical applications. Rev.

Virtual Quim. 9, 387–409. https://doi.org/10.21577/1984-6835.20170022

Gómez-Estaca, J., López de Lacey, A., López-Caballero, M.E., Gómez-Guillén, M.C.,

PT
Montero, P., 2010. Biodegradable gelatin-chitosan films incorporated with

essential oils as antimicrobial agents for fish preservation. Food Microbiol. 27,

RI
889–896. https://doi.org/10.1016/j.fm.2010.05.012

SC
Grenha, A., 2012. Chitosan nanoparticles: A survey of preparation methods. J. Drug
NU
Target. 20, 291–300. https://doi.org/10.3109/1061186X.2011.654121

Guohua, H., Wei, L., Hailin, F., Jian, L., Gao Yuanyuan, 2016. Effects of chitosan
MA

combined with nisin treatment on storage quality of large yellow croaker

(Pseudosciaena crocea). Food Chem. 203, 276–282.


ED

https://doi.org/10.1016/j.foodchem.2016.01.122
T

Hajipour, M.J., Fromm, K.M., Akbar Ashkarran, A., Jimenez de Aberasturi, D.,
EP

Larramendi, I.R. de, Rojo, T., Serpooshan, V., Parak, W.J., Mahmoudi, M., 2012.

Antibacterial properties of nanoparticles. Trends Biotechnol. 30, 499–511.


C
AC

https://doi.org/10.1016/j.tibtech.2012.06.004

Harikrishnan, R., Kim, J.S., Balasundaram, C., Heo, M.S., 2012a. Dietary

supplementation with chitin and chitosan on haematology and innate immune

response in Epinephelus bruneus against Philasterides dicentrarchi. Exp.

Parasitol. 131, 116–124. https://doi.org/10.1016/j.exppara.2012.03.020

Harikrishnan, R., Kim, J.S., Balasundaram, C., Heo, M.S., 2012b. Immunomodulatory
ACCEPTED MANUSCRIPT

effects of chitin and chitosan enriched diets in Epinephelus bruneus against Vibrio

alginolyticus infection. Aquaculture 326–329, 46–52.

https://doi.org/10.1016/j.aquaculture.2011.11.034

Hebeish, A., Ramadan, M.A., Montaser, A.S., 2015. In vitro and in vivo antibacterial

potential of chitosan-g-acrylonitrile silver nanocomposite against a pathogenic

PT
bacterium. Int.J.Curr.Microbiol.App.Sci 4, 5–19.

Hosseini, S.F., Rezaei, M., Zandi, M., Farahmandghavi, F., 2016. Preparation and

RI
characterization of chitosan nanoparticles-loaded fish gelatin-based edible films. J.

SC
Food Process Eng. 39, 521–530. https://doi.org/10.1111/jfpe.12246
NU
Hosseini, S.F., Rezaei, M., Zandi, M., Farahmandghavi, F., 2015. Fabrication of bio-

nanocomposite films based on fish gelatin reinforced with chitosan nanoparticles.


MA

Food Hydrocoll. 44, 172–182. https://doi.org/10.1016/j.foodhyd.2014.09.004

Hosseini, S.F., Rezaei, M., Zandi, M., Farhid Farahmand Ghavi, 2013. Preparation and
ED

functional properties of fish gelatin-chitosan blend edible films. Food Chem. 136,

1490–1495. https://doi.org/10.1016/j.foodchem.2012.09.081
T
EP

Hu, Y.-L., Qi, W., Han, F., Shao, J.-Z., Gao, J.-Q., 2011. Toxicity evaluation of

biodegradable chitosan nanoparticles using a zebrafish embryo model. Int. J.


C
AC

Nanomedicine 6, 3351–9. https://doi.org/10.2147/IJN.S25853

Huang, Y., Lapitsky, Y., 2012. Salt-assisted mechanistic analysis of

chitosan/tripolyphosphate micro- and nanogel formation. Biomacromolecules 13,

3868–3876. https://doi.org/10.1021/bm3014236

Imefon Udo, U., 2018. Effects of chitosan and chitosan nanoparticles on water quality,

growth performance, survival rate and meat quality of the African catfish, Clarias

gariepinus. Nanoscience 1, 12–25. https://doi.org/10.31058/j.nano.2018.11002


ACCEPTED MANUSCRIPT

Ing, L.Y., Zin, N.M., Sarwar, A., Katas, H., 2012. Antifungal activity of chitosan

nanoparticles and correlation with their physical properties. Int. J. Biomater. 2012,

1–9. https://doi.org/10.1155/2012/632698

Islam, M.M., Masum, S.M., Mahbub, K.R., 2011. In vitro antibacterial activity of shrimp

chitosan against Salmonela paratyphi and Staphylococcus aureus. J. Bangladesh

PT
Chem. Soc. 24, 185–190. https://doi.org/10.3329/jbcs.v24i2.9707

Jeon, S.J., Oh, M., Yeo, W.-S., Galvao, K.N., Jeong, K.C., 2014. Underlying mechanism

RI
of antimicrobial activity of chitosan microparticles and implications for the

SC
treatment of infectious diseases. PLoS One 9, e92723.

doi:10.1371/journal.pone.009272. https://doi.org/10.1371/journal.pone.0092723
NU
Ji, J., Torrealba, D., Ruyra, À., Roher, N., 2015. Nanodelivery systems as new tools for
MA

immunostimulant or vaccine administration: Targeting the fish immune system.

Biology (Basel). 4, 664–696. https://doi.org/10.3390/biology4040664


ED

Jia, H., Zhu, G., Wang, P., 2003. Catalytic behaviors of enzymes attached to

nanoparticles: the effect of particle mobility. Biotechnol. Bioeng. 84, 406–414.


T
EP

https://doi.org/10.1002/bit.10781

Jiménez-Fernández, E., Ruyra, A., Roher, N., Zuasti, E., Infante, C., Fernández-Díaz, C.,
C
AC

2014. Nanoparticles as a novel delivery system for vitamin C administration in

aquaculture. Aquaculture 432, 426–433.

https://doi.org/10.1016/j.aquaculture.2014.03.006

Kakaei, S., Shahbazi, Y., 2016. Effect of chitosan-gelatin film incorporated with ethanolic

red grape seed extract and Ziziphora clinopodioides essential oil on survival of

Listeria monocytogenes and chemical, microbial and sensory properties of minced

trout fillet. LWT - Food Sci. Technol. 72, 432–438.


ACCEPTED MANUSCRIPT

https://doi.org/10.1016/j.lwt.2016.05.021

Kataoka, K., Matsumoto, T., Yokoyama, M., Okano, T., Sakurai, Y., Fukushima, S.,

Okamoto, K., Kwon, G.S., 2000. Doxorubicin-loaded poly (ethylene glycol)-poly

(β-benzyl-L-aspartate) copolymer micelles: Their pharmaceutical characteristics

and biological significance. J. Control. Release 64, 143–153.

PT
https://doi.org/10.1016/S0168-3659(99)00133-9

Katas, H., Raja, M.A.G., Lam, K.L., 2013. Development of chitosan nanoparticles as a

RI
stable drug delivery system for protein/siRNA. Int. J. Biomater. 2013, 1–9.

SC
https://doi.org/10.1155/2013/146320 NU
Kean, T., Thanou, M., 2010. Biodegradation, biodistribution and toxicity of chitosan.

Adv. Drug Deliv. Rev. 62, 3–11. https://doi.org/10.1016/j.addr.2009.09.004


MA

Kim, B.C., Nair, S., Kim, J., Kwak, J.H., Grate, J.W., Kim, S.H., Gu, M.B., 2005.

Preparation of biocatalytic nanofibres with high activity and stability via enzyme
ED

aggregate coating on polymer nanofibres. Nanotechnology 16, 382–388.


T

Kim, J., Grate, J.W., 2003. Single-enzyme nanoparticles armored by a nanometer-scale


EP

organic/inorganic network. Nano Lett. 3, 1219–1222.

https://doi.org/10.1021/nl034404b
C
AC

Kumar, S.R., Parameswaran, V., Ahmed, V.P.I., Musthaq, S.S., Hameed, A.S.S., 2007.

Protective efficiency of DNA vaccination in Asian seabass (Lates calcarifer)

against Vibrio anguillarum. Fish Shellfish Immunol. 23, 316–326.

https://doi.org/10.1016/j.fsi.2006.11.005

Kumari, R., Gupta, S., Singh, A.R., Ferosekhan, S., Kothari, D.C., Pal, A.K., Jadhao,

S.B., 2013. Chitosan nanoencapsulated exogenous trypsin biomimics zymogen-

like enzyme in fish gastrointestinal tract. PLoS One 8, 1–12.


ACCEPTED MANUSCRIPT

https://doi.org/10.1371/journal.pone.0074743

Lanka, D., Mittapally, V.K., 2016. Preparation and applications of chitosan nanoparticles:

A brief review. Res. Rev. J. Mater. Sci. 04, 34–38. https://doi.org/10.4172/2321-

6212.1000r001

Lavertu, M., Méthot, S., Tran-Khanh, N., Buschmann, M.D., 2006. High efficiency gene

PT
transfer using chitosan/DNA nanoparticles with specific combinations of

molecular weight and degree of deacetylation. Biomaterials 27, 4815–4824.

RI
https://doi.org/10.1016/j.biomaterials.2006.04.029

SC
León-Rodríguez, L., Luzardo-álvarez, A., Blanco-Méndez, J., Lamas, J., Leiro, J., 2013.
NU
Biodegradable microparticles covalently linked to surface antigens of the

scuticociliate parasite P. dicentrarchi promote innate immune responses in vitro.


MA

Fish Shellfish Immunol. 34, 236–243. https://doi.org/10.1016/j.fsi.2012.10.029

León-Rodríguez, L., Luzardo-álvarez, A., Blanco-Méndez, J., Lamas, J., Leiro, J., 2012.
ED

A vaccine based on biodegradable microspheres induces protective immunity


T

against scuticociliatosis without producing side effects in turbot. Fish Shellfish


EP

Immunol. 33, 21–27. https://doi.org/10.1016/j.fsi.2012.03.028


C

Li, L., Lin, S.L., Deng, L., Liu, Z.G., 2013. Potential use of chitosan nanoparticles for
AC

oral delivery of DNA vaccine in black seabream Acanthopagrus schlegelii

Bleeker to protect from Vibrio parahaemolyticus. J. Fish Dis. 36, 987–995.

https://doi.org/10.1111/jfd.12032

Li, R., Guo, Z., 2010. Synthesis, characterization and antifungal properties of N, O- (acyl)

-N- (trimethyl) chitosan chloride. e-Polymers 1–6.

Li, T., Hu, W., Li, J., Zhang, X., Zhu, J., Li, X., 2012. Coating effects of tea polyphenol
ACCEPTED MANUSCRIPT

and rosemary extract combined with chitosan on the storage quality of large

yellow croaker (Pseudosciaena crocea). Food Control 25, 101–106.

https://doi.org/10.1016/j.foodcont.2011.10.029

Li, T., Li, J., Hu, W., Li, X., 2013. Quality enhancement in refrigerated red drum

(Sciaenops ocellatus) fillets using chitosan coatings containing natural

preservatives. Food Chem. 138, 821–826.

PT
https://doi.org/10.1016/j.foodchem.2012.11.092

RI
Lin, S., Pan, Y., Luo, Lin, Luo, Li, 2011. Effects of dietary β-1,3-glucan, chitosan or

SC
raffinose on the growth, innate immunity and resistance of koi (Cyprinus carpio

koi). Fish Shellfish Immunol. 31, 788–794.


NU
https://doi.org/10.1016/j.fsi.2011.07.013
MA

Liu, C.G., Chen, X.G., Park, H.J., 2005. Self-assembled nanoparticles based on linoleic-

acid modified chitosan: Stability and adsorption of trypsin. Carbohydr. Polym. 62,
ED

293–298. https://doi.org/10.1016/j.carbpol.2005.08.010

Liu, N., Chen, X.-G., Park, H., Liu, C.-G., Liu, C.-S., Meng, X.-H., Yu, L.-J., 2006.
T
EP

Effect of MW and concentration of chitosan on antibacterial activity of

Escherichia coli. Carbohydr. Polym. 64, 60–65.


C

https://doi.org/10.1016/j.carbpol.2005.10.028
AC

Liu, X.F., Guan, Y.L., Yang, D.Z., Li, Z., Yao, K. De, 2001. Antibacterial action of

chitosan and carboxymethylated. J. Appl. Polym. Sci. 79, 1324–1335.

Liu, Y., Cheng, X.J., Dang, Q.F., Ma, F.K., Chen, X.G., Park, H.J., Kim, B.K., 2012a.

Preparation and evaluation of oleoyl-carboxymethy-chitosan (OCMCS)

nanoparticles as oral protein carriers. J. Mater. Sci. Mater. Med. 23, 375–384.

https://doi.org/10.1007/s10856-011-4470-9
ACCEPTED MANUSCRIPT

Liu, Y., Di Zang, H., Kong, M., Ma, F.K., Dang, Q.F., Cheng, X.J., Ji, Q.X., Chen, X.G.,

2012b. In vitro evaluation of mucoadhesion and permeation enhancement of

polymeric amphiphilic nanoparticles. Carbohydr. Polym. 89, 453–460.

https://doi.org/10.1016/j.carbpol.2012.03.028

Liu, Y., Kong, M., Feng, C., Yang, K.K., Li, Y., Su, J., Cheng, X.J., Park, H.J., Chen,

X.G., 2013. Biocompatibility, cellular uptake and biodistribution of the polymeric

PT
amphiphilic nanoparticles as oral drug carriers. Colloids Surfaces B Biointerfaces

RI
103, 345–353. https://doi.org/10.1016/j.colsurfb.2012.11.012

SC
Liu, Y., Wang, F.Q., Shah, Z., Cheng, X.J., Kong, M., Feng, C., Chen, X.G., 2016. Nano-

polyplex based on oleoyl-carboxymethy-chitosan (OCMCS) and hyaluronic acid


NU
for oral gene vaccine delivery. Colloids Surfaces B Biointerfaces 145, 492–501.
MA

https://doi.org/10.1016/j.colsurfb.2016.05.035

Llaiyaraja, N., Devi, A., Khanum, F., 2014. Chlorogenic acid loaded chitosan
ED

nanoparticles with sustained release property, retained antioxidant activity and

enhanced bioavailability. Asian J. Pharm. Sci. 10, 203–211.


T
EP

https://doi.org/10.1016/j.ajps.2014.09.005

Luo, Y., Teng, Z., Wang, Q., 2012. Development of zein nanoparticles coated with
C

carboxymethyl chitosan for encapsulation and controlled release of vitamin D3. J.


AC

Agric. Food Chem. 60, 836–843. https://doi.org/10.1021/jf204194z

Luo, Y., Wang, Q., 2014. Zein-based micro- and nano-particles for drug and nutrient

delivery: A review. J. Appl. Polym. Sci. 131, 40696.

https://doi.org/10.1002/app.40696

Luo, Y., Zhang, B., Whent, M., Yu, L.L., Wang, Q., 2011. Preparation and

characterization of zein/chitosan complex for encapsulation of α-tocopherol, and


ACCEPTED MANUSCRIPT

its in vitro controlled release study. Colloids Surfaces B Biointerfaces 85, 145–

152. https://doi.org/10.1016/j.colsurfb.2011.02.020

Ma, Z., Garrido-maestu, A., Jeong, K.C., 2017. Application, mode of action, and in vivo

activity of chitosan and its micro- and nanoparticles as antimicrobial agents: A

review. Carbohydr. Polym. 176, 257–265.

PT
https://doi.org/10.1016/j.carbpol.2017.08.082

Ma, Z., Kim, D., Adesogan, A.T., Ko, S., Galvao, K., Jeong, K.C., 2016. Chitosan

RI
microparticles exert broad-spectrum antimicrobial activity against antibiotic-

SC
resistant micro-organisms without increasing resistance. ACS Appl. Mater.

Interfaces 8, 10700–10709. https://doi.org/10.1021/acsami.6b00894


NU
Malatesta, M., Grecchi, S., Chiesa, E., Cisterna, B., Costanzo, M., Zancanaro, C., 2015.
MA

Internalized chitosan nanoparticles persist for long time in cultured cells. Eur. J.

Histochem. 59, 17–21. https://doi.org/10.4081/ejh.2015.2492


ED

Mandal, A., 2010. Modification of a DNA vaccine for oral acministration in fish for

aquaculture by using non-microbial nanoparticles.


T
EP

Mehrpak, M., Banaee, M., Haghi, B.N., Noori, A.N., 2015. Protective effects of vitamin

C and chitosan against cadmium-induced oxidative stress in the liver of common


C
AC

carp (Cyprinus carpio). Iran. J. Toxicol. 9, 1360–1367.

Meshkini, S., Tafy, A.A., Tukmechi, A., Farhang-Pajuh, F., 2012. Effects of chitosan on

hematological parameters and stress resistance in rainbow trout (Oncorhynchus

mykiss). Vet Res Forum 3, 49–54.

Mohan, C.O., Ravishankar, C.N., Lalitha, K. V., Srinivasa Gopal, T.K., 2012. Effect of

chitosan edible coating on the quality of double filleted Indian oil sardine

(Sardinella longiceps) during chilled storage. Food Hydrocoll. 26, 167–174.


ACCEPTED MANUSCRIPT

https://doi.org/10.1016/j.foodhyd.2011.05.005

Mohebi, E., Shahbazi, Y., 2017. Application of chitosan and gelatin based active

packaging films for peeled shrimp preservation: A novel functional wrapping

design. LWT - Food Sci. Technol. 76, 108–116.

https://doi.org/10.1016/j.lwt.2016.10.062

PT
Mustafa, S.A., Alfaragi, J.K., Aref, Z., 2014. The influence of chitosan on immune status

and survival rate of Cyprinus carpio L . challenged with Aeromonas hydrophila.

RI
Vet. Med. Sci. 5, 93–104.

SC
Muzzarelli, R.A.A., 2010. Chitins and chitosans as immunoadjuvants and non-allergenic
NU
drug carriers. Mar. Drugs 8, 292–312. https://doi.org/10.3390/md8020292

Najafabad, M.K., Imanpoor, M.R., Taghizadeh, V., Alireza Alishahi, 2016. Effect of
MA

dietary chitosan on growth performance, hematological parameters, intestinal

histology and stress resistance of Caspian kutum (Rutilus frisii kutum Kamenskii,
ED

1901) fingerlings. Fish Physiol. Biochem. 42, 1063–1071.

https://doi.org/10.1007/s10695-016-0197-3
T
EP

Nasti, A., Zaki, N.M., De Leonardis, P., Ungphaiboon, S., Sansongsak, P., Rimoli, M.G.,

Tirelli, N., 2009. Chitosan/TPP and chitosan/TPP-hyaluronic acid nanoparticles:


C
AC

Systematic optimisation of the preparative process and preliminary biological

evaluation. Pharm. Res. 26, 1918–1930. https://doi.org/10.1007/s11095-009-

9908-0

Nikapitiya, C., Dananjaya, S.H.S., De Silva, B.C.J., Heo, G.J., Oh, C., De Zoysa, M., Lee,

J., 2018. Chitosan nanoparticles: A positive immune response modulator as

display in zebrafish larvae against Aeromonas hydrophila infection. Fish Shellfish

Immunol. 76, 240–246. https://doi.org/10.1016/j.fsi.2018.03.010


ACCEPTED MANUSCRIPT

No, K.H., Park, N.Y., Lee, S.H., Meyers, S.P., 2002. Antibacterial activity of chitosans

and chitosan oligomers with different molecular weights. Int. J. Food Microbiol.

74, 65–72.

Nowzari, F., Shábanpour, B., Ojagh, S.M., 2013. Comparison of chitosan-gelatin

composite and bilayer coating and film effect on the quality of refrigerated

rainbow trout. Food Chem. 141, 1667–1672.

PT
https://doi.org/10.1016/j.foodchem.2013.03.022

RI
Pan, Y., Li, Y.J., Zhao, H.Y., Zheng, J.M., Xu, H., Wei, G., Hao, J.S., Cui, F. De, 2002.

SC
Bioadhesive polysaccharide in protein delivery system: Chitosan nanoparticles

improve the intestinal absorption of insulin in vivo. Int. J. Pharm. 249, 139–147.
NU
https://doi.org/10.1016/S0378-5173(02)00486-6
MA

Parida, U.K., Rout, N., Bindhani, B.K., 2013. In vitro properties of chitosan nanoparticles

induce apoptosis in human lymphoma SUDHL-4 cell line. Adv. Biosci.


ED

Biotechnol. 4, 1118–1127. https://doi.org/10.4236/abb.2013.412148

Plant, K.P., LaPatra, S.E., 2011. Advances in fish vaccine delivery. Dev. Comp.
T
EP

Immunol. 35, 1256–1262. https://doi.org/10.1016/j.dci.2011.03.007


C

Qi, L., Xu, Z., Jiang, X., Hu, C., Zou, X., 2004. Preparation and antibacterial activity of
AC

chitosan nanoparticles. Carbohydr. Res. 339, 2693–2700.

https://doi.org/10.1016/j.carres.2004.09.007

Qiu, X., Chen, S., Liu, G., Yang, Q., 2014. Quality enhancement in the Japanese sea bass

(Lateolabrax japonicas) fillets stored at 4°C by chitosan coating incorporated with

citric acid or licorice extract. Food Chem. 162, 156–160.

https://doi.org/10.1016/j.foodchem.2014.04.037
ACCEPTED MANUSCRIPT

Rajesh Kumar, S., Ishaq Ahmed, V.P., Parameswaran, V., Sudhakaran, R., Sarath Babu,

V., Sahul Hameed, A.S., 2008. Potential use of chitosan nanoparticles for oral

delivery of DNA vaccine in Asian sea bass (Lates calcarifer) to protect from

Vibrio (Listonella) anguillarum. Fish Shellfish Immunol. 25, 47–56.

https://doi.org/10.1016/j.fsi.2007.12.004

Rajeshkumar, S., Venkatesan, C., Sarathi, M., Sarathbabu, V., Thomas, J., Anver Basha,

PT
K., Sahul Hameed, A.S., 2009. Oral delivery of DNA construct using chitosan

RI
nanoparticles to protect the shrimp from white spot syndrome virus (WSSV). Fish

SC
Shellfish Immunol. 26, 429–437. https://doi.org/10.1016/j.fsi.2009.01.003

Ramezani, Z., Zarei, M., Raminnejad, N., 2015. Comparing the effectiveness of chitosan
NU
and nanochitosan coatings on the quality of refrigerated silver carp fillets. Food
MA

Control 51, 43–48. https://doi.org/10.1016/j.foodcont.2014.11.015

Ramos, E.A., Relucio, J.L. V., Torres-Villanueva, C.A.T., 2005. Gene expression in
ED

tilapia following oral delivery of chitosan-encapsulated plasmid DNA

incorporated into fish feeds. Mar. Biotechnol. 7, 89–94.


T
EP

https://doi.org/10.1007/s10126-004-3018-0

Rampino, A.., Borgogna, M.., Blasi, P.., Bellich, B.., Cesàro, A.., 2013. Chitosan
C

nanoparticles: Preparation, size evolution and stability. Int. J. Pharm. 455, 219–
AC

228. https://doi.org/10.1016/j.ijpharm.2013.07.034

Ranjan, R., Prasad, K.P., Vani, T., Kumar, R., 2014. Effect of dietary chitosan on

haematology, innate immunity and disease resistance of Asian seabass Lates

calcarifer (Bloch). Aquac. Res. 45, 983–993. https://doi.org/10.1111/are.12050

Rather, M.A., Sharma, R., Gupta, S., Ferosekhan, S., Ramya, V.L., Jadhao, S.B., 2013.

Chitosan-nanoconjugated hormone nanoparticles for sustained surge of


ACCEPTED MANUSCRIPT

gonadotropins and enhanced reproductive output in female fish. PLoS One 8, 1–

10. https://doi.org/10.1371/journal.pone.0057094

Rivas-Aravena, A., Fuentes, Y., Cartagena, J., Brito, T., Poggio, V., La Torre, J.,

Mendoza, H., Gonzalez-Nilo, F., Sandino, A.M., Spencer, E., 2015. Development

of a nanoparticle-based oral vaccine for Atlantic salmon against ISAV using an

alphavirus replicon as adjuvant. Fish Shellfish Immunol. 45, 157–166.

PT
https://doi.org/10.1016/j.fsi.2015.03.033

RI
Rojas, J., Ciro, Y., Zapata, S., 2015. Chitosan as a potential microencapsulation carrier

SC
for ascorbic acid stabilization in heterodisperse systems. Int. J. Pharm. Pharm. Sci.

7, 69–72.
NU
Sáez, M.I., Vizcaíno, A.J., Alarcón, F.J., Martínez, T.F., 2018. Feed pellets containing
MA

chitosan nanoparticles as plasmid DNA oral delivery system for fish: In vivo

assessment in gilthead sea bream (Sparus aurata) juveniles. Fish Shellfish


ED

Immunol. 80, 458–466. https://doi.org/10.1016/j.fsi.2018.05.055

Saleh, M., Kumar, G., Abdel-Baki, A.A., Al-Quraishy, S., El-Matbouli, M., 2016. In vitro
T
EP

antimicrosporidial activity of gold nanoparticles against Heterosporis saurida.

BMC Vet. Res. 12. https://doi.org/10.1186/S12917-016-0668-X


C
AC

Saloko, S., Darmadji, P., Setiaji, B., Pranoto, Y., 2014. Antioxidative and antimicrobial

activities of liquid smoke nanocapsules using chitosan and maltodextrin and its

application on tuna fish preservation. Food Biosci. 7, 71–79.

https://doi.org/10.1016/j.fbio.2014.05.008

Samarakoon, K.W., Cha, S.H., Lee, J.H., Jeon, Y.J., 2013. The growth, innate immunity

and protection against H2 O 2 -induced oxidative damage of a chitosan-coated diet in

the olive flounder paralichthys olivaceus. Fish. Aquat. Sci. 16, 149–158.
ACCEPTED MANUSCRIPT

https://doi.org/10.5657/FAS.2013.0149

Shaalan, M., Saleh, M., El-Mahdy, M., El-Matbouli, M., 2016. Recent progress in

applications of nanoparticles in fish medicine: A review. Nanomedicine:

Nanotechnology, Biology, and Medicine.

https://doi.org/10.1016/j.nano.2015.11.005

PT
Shaalan, M.I., El-Mahdy, M.M., Theiner, S., El-Matbouli, M., Saleh, M., 2017. In vitro

assessment of the antimicrobial activity of silver and zinc oxide nanoparticles

RI
against fish pathogens. Acta Vet. Scand. 59, 1–11. https://doi.org/10.1186/s13028-

SC
017-0317-9 NU
Shanthi Mari, L.S., Jagruthi, C., Anbazahan, S.M., Yogeshwari, G., Thirumurugan, R.,

Arockiaraj, J., Mariappan, P., Balasundaram, C., Harikrishnan, R., 2014.


MA

Protective effect of chitin and chitosan enriched diets on immunity and disease

resistance in Cirrhina mrigala against Aphanomyces invadans. Fish Shellfish


ED

Immunol. 39, 378–385. https://doi.org/10.1016/j.fsi.2014.05.027


T

Sharifinasab, Z., Banaee, M., Mohiseni, M., Noori, A., 2016. Vitamin C and chitosan
EP

alleviate toxic effects of paraquat on some biochemical parameters in hepatocytes

of common carp. Iran. J. Toxicol. 10, 31–40.


C
AC

Shi, J., Votruba, A.R., Farokhzad, O.C., Langer, R., 2010. Nanotechnology in drug

delivery and tissue engineering: From discovery to applications. Nano Lett. 10,

3223–3230. https://doi.org/10.1021/nl102184c

Šimůnek, J., Brandysová, V., Koppová, I., Šimůnek, J., 2012. The antimicrobial action of

chitosan, low molar mass chitosan, and chitooligosaccharides on human colonic

bacteria. Folia Microbiol. (Praha). 57, 341–345. https://doi.org/10.1007/s12223-

012-0138-1
ACCEPTED MANUSCRIPT

Soares, N.M., Mendes, T.S., Vicente, A.A., 2013. Effect of chitosan-based solutions

applied as edible coatings and water glazing on frozen salmon preservation - A

pilot-scale study. J. Food Eng. 119, 316–323.

https://doi.org/10.1016/j.jfoodeng.2013.05.018

Solval, K.M., Espinoza Rodezno, L.A., Moncada, M., Bankston, J.D., Sathivel, S., 2014.

Evaluation of chitosan nanoparticles as a glazing material for cryogenically frozen

PT
shrimp. LWT - Food Sci. Technol. 57, 172–180.

RI
https://doi.org/10.1016/j.lwt.2013.12.033

SC
Sun, B., Quan, H., Zhu, F., 2016. Dietary chitosan nanoparticles protect crayfish

Procambarus clarkii against white spot syndrome virus (WSSV) infection. Fish
NU
Shellfish Immunol. 54, 241–246. https://doi.org/10.1016/j.fsi.2016.04.009
MA

Tang, D.W., Yu, S.H., Ho, Y.C., Huang, B.Q., Tsai, G.J., Hsieh, H.Y., Sung, H.W., Mi,

F.L., 2013. Characterization of tea catechins-loaded nanoparticles prepared from


ED

chitosan and an edible polypeptide. Food Hydrocoll. 30, 33–41.

https://doi.org/10.1016/j.foodhyd.2012.04.014
T
EP

Tian, J., Yu, J., Sun, X., 2008. Chitosan microspheres as candidate plasmid vaccine

carrier for oral immunisation of Japanese flounder (Paralichthys olivaceus). Vet.


C

Immunol. Immunopathol. 126, 220–229.


AC

https://doi.org/10.1016/j.vetimm.2008.07.002

Vahedi, G., Ghodratizadeh, S., 2011. Effect of chitin supplemented diet on innate

immune response of rainbow trout. World J. fish Mar. Sci. 3, 509–513.

Valero, Y., Awad, E., Buonocore, F., Arizcun, M., Esteban, M.Á., Meseguer, J., Chaves-

Pozo, E., Cuesta, A., 2016. An oral chitosan DNA vaccine against nodavirus

improves transcription of cell-mediated cytotoxicity and interferon genes in the


ACCEPTED MANUSCRIPT

European sea bass juveniles gut and survival upon infection. Dev. Comp.

Immunol. 65, 64–72. https://doi.org/10.1016/j.dci.2016.06.021

Vimal, S., Abdul Majeed, S., Nambi, K.S.N., Madan, N., Farook, M.A., Venkatesan, C.,

Taju, G., Venu, S., Subburaj, R., Thirunavukkarasu, A.R., Sahul Hameed, A.S.,

2014. Delivery of DNA vaccine using chitosan-tripolyphosphate (CS/TPP)

nanoparticles in Asian sea bass, Lates calcarifer (Bloch, 1790) for protection

PT
against nodavirus infection. Aquaculture 420–421, 240–246.

RI
https://doi.org/10.1016/j.aquaculture.2013.11.017

SC
Vimal, S., Taju, G., Nambi, K.S.N., Abdul Majeed, S., Sarath Babu, V., Ravi, M., Sahul

Hameed, A.S., 2012. Synthesis and characterization of CS/TPP nanoparticles for


NU
oral delivery of gene in fish. Aquaculture 358–359, 14–22.
MA

https://doi.org/10.1016/j.aquaculture.2012.06.012

Wang, Q., Lei, J., Ma, J., Yuan, G., Sun, H., 2018. Effect of chitosan-carvacrol coating on
ED

the quality of Pacific white shrimp during iced storage as affected by caprylic

acid. Int. J. Biol. Macromol. 106, 123–129.


T
EP

https://doi.org/10.1016/j.ijbiomac.2017.07.180

Wang, Y., Li, J., 2011. Effects of chitosan nanoparticles on survival, growth and meat
C

quality of tilapia, Oreochromis nilotica. Nanotoxicology 5, 425–431.


AC

https://doi.org/10.3109/17435390.2010.530354

Wang, Y., Zhou, J., Liu, L., Huang, C., Zhou, D., Fu, L., 2016. Characterization and

toxicology evaluation of chitosan nanoparticles on the embryonic development of

zebrafish, Danio rerio. Carbohydr. Polym. 141, 204–210.

https://doi.org/10.1016/j.carbpol.2016.01.012

Wisdom, K.S., Bhat, I.A., Kumar, P., Pathan, M.K., Chanu, T.I., Walke, P., Sharma, R.,
ACCEPTED MANUSCRIPT

2018. Fabrication of chitosan nanoparticles loaded with aromatase inhibitors for

the advancement of gonadal development in Clarias magur (Hamilton, 1822).

Aquaculture 497, 125–133. https://doi.org/10.1016/j.aquaculture.2018.07.049

Xu, Y., Du, Y., 2003. Effect of molecular structure of chitosan on protein delivery

properties of chitosan nanoparticles. Int. J. Pharm. 250, 215–226.

PT
https://doi.org/10.1016/S0378-5173 (02)00548-3

Yang, H.C., Wang, W.H., Huang, K.S., Hon, M.H., 2010. Preparation and application of

RI
nanochitosan to finishing treatment with anti-microbial and anti-shrinking

SC
properties. Carbohydr. Polym. 79, 176–179.

https://doi.org/10.1016/j.carbpol.2009.07.045
NU
Yao, Y., Ding, D., Shao, H., Peng, Q., Huang, Y., 2017. Antibacterial activity and
MA

physical properties of fish gelatin-chitosan edible films supplemented with D-

Limonene. Int. J. Polym. Sci. 2017, 1–9. https://doi.org/10.1155/2017/1837171


ED

Yildirim-Aksoy, M., Beck, B.H., 2017. Antimicrobial activity of chitosan and a chitosan

oligomer against bacterial pathogens of warmwater fish. J. Appl. Microbiol. 122,


T
EP

1570--1578. https://doi.org/10.1111/jam.13460

Younes, I., Rinaudo, M., 2015. Chitin and chitosan preparation from marine sources.
C
AC

Structure, properties and applications. Mar. Drugs 13, 1133–1174.

https://doi.org/10.3390/md13031133

Yu, D., Xu, Y., Regenstein, J.M., Xia, W., Yang, F., Jiang, Q., Wang, B., 2018. The

effects of edible chitosan-based coatings on flavor quality of raw grass carp

(Ctenopharyngodon idellus) fillets during refrigerated storage. Food Chem. 242,

412–420. https://doi.org/10.1016/j.foodchem.2017.09.037

Yuan, G., Lv, H., Tang, W., Zhang, X., Sun, H., 2016. Effect of chitosan coating
ACCEPTED MANUSCRIPT

combined with pomegranate peel extract on the quality of Pacific white shrimp

during iced storage. Food Control 59, 818–823.

https://doi.org/10.1016/j.foodcont.2015.07.011

Zaki, M.A., Salem, M.E., Gaber, M.M., Nour, A.M., 2015. Effect of chitosan

supplemented diet on survival, growth, feed utilization, body composition &

histology of sea bass (Dicentrarchus labrax). World J. Eng. Technol. 3, 38–47.

PT
https://doi.org/10.4236/wjet.2015.34C005

RI
Zarei, M., Ramezani, Z., Ein-Tavasoly, S., Chadorbaf, M., 2015. Coating effects of

SC
orange and pomegranate peel extracts combined with chitosan nanoparticles on

the quality of refrigerated silver carp fillets. J. Food Process. Preserv. 39, 2180–
NU
2187. https://doi.org/10.1111/jfpp.12462
MA

Zhang, W., Zhang, J., Jiang, Q., Xia, W., 2012. Physicochemical and structural

characteristics of chitosan nanopowders prepared by ultrafine milling. Carbohydr.


ED

Polym. 87, 309–313. https://doi.org/10.1016/j.carbpol.2011.07.057

Zhao, L., Shi, L., Zhang, Z., Chen, J., Shi, D., Yang, J., Tang, Z., 2011. Preparation and
T
EP

application of chitosan nanoparticles and nanofibers 28, 353–362.

Zhao, L., Su, C., Zhu, B., Jia, Y., 2014. Development and optimization of insulin-chitosan
C
AC

nanoparticles. Trop. J. Pharm. Res. 13, 3–8. https://doi.org/10.4314/tjpr.v13i1.1

Figures and table legends

Figure 1. (A) SEM and (B) TEM of chitosan nanoparticles (Parida et al., 2013).

Figure 2. (A) Size distribution intensity and (B) TEM of freeze-dried CSNPs, encapsulating

OmpA of E. tarda (Dubey et al., 2016).

Figure 3. (Rather et al., 2013)


ACCEPTED MANUSCRIPT

I. (A) Particle size distributions and (B) TEM of chitosan-LHRH and chitosan-gold-LHRH

nanoparticles.

II. Ovarian histology of Cyprinus carpio:

A] Control: 1. Ovigerous lamellae with immature oocyte; 2. Mature oocyte; 3. Maturing

oocyte; 4. Atretic oocyte.

B] After injection with bare LHRH: 1. Mature/ripe oocyte; 2. Partially spent oocyte; 3.

PT
Cortical alveoli stage oocyte; 4. Spent follicle; 5. Nest of immature oocytes.

RI
C] After injection with CS/ LHRH NPs: 1. Lamellae containing nests of Oocytes 2. Spent

SC
follicle 3. Spent oocytes 4. Immature oocytes in ovigerous lamellae

D] After injection with CS–gold/LHRH NPs: 1. Mature oocytes 2. Spent oocytes 3. Immature
NU
oocytes 4. Discharged follicle.

Figure 4. (Kumari et al., 2013)


MA

a) Particle size distribution and three-dimensional images of CSNPs: A] 0.4% free

CSNPs, B&D] 0.4% CSNPs/0.01% trypsin and C&E] 0.4% CSNPs/0.02% trypsin.
ED

b) Intestinal histology (H&E, 40X) after feeding of Cyprinus carpio with:

(A): Control fish fed: Regular villi with continuous basement membrane lined the
T
EP

intestinal mucosa.

(B): 0.4% free CSNPs: 1. Villi with mildly swollen apical surfaces.
C

(C1 & C2) 0.02% bare trypsin: 2. Broadened villi, marked foamy cells with lipid
AC

vacuoles. 3. Atrophied submucosal layer and muscularis.

(D) 0.4% CSNPs/0.01% trypsin: 4. longer villi with healthy apical surfaces. 5.

Improved morphological appearance of villi with continuous basement membrane. 7.

Distributed goblet cells along the villi for mucin production indicating better gastro-

intestinal health.
ACCEPTED MANUSCRIPT

(E) 0.4% CSNPs/0.02% trypsin: 6. Less crypt depth. 7. Villi containing goblet cells

and are similar to control group.

Table 1. Collection of the reported In vitro studies on the antimicrobial (antibacterial and

antifungal) activities of chitosan and its nanocomposites.

Table 2. Immunomodulatory effects of chitosan-based complexes on several fish species

against infections and stresses.

PT
Table 3. Published studies on bio-molecules and vaccine delivery via loading on chitosan, its

RI
microspheres and nanocomplexes.

SC
Table 4. Published studies on control and sustain release of bioactive ingredients via loading

on chitosan nanocomposites.
NU
Table 5. The reported applications in fish meat preservation during chilled and frozen

storage.
MA
T ED
C EP
AC
ACCEPTED MANUSCRIPT

Table 1

microorganism Reagent Reference

Gram negative bacteria Gram positive bacteria Fungi

Salmonella paratyphi, Staphylococcus aureus ___________ Chitosan (Islam et al.,

Escherchia coli 2011)

Human colonic anaerobic, nonpathogenic bacteria ___________ Chitosan (Šimůnek et

al., 2012)

PT
Escherchia coli __________________ ____________ N-diethylmethyl (Avadi et al.,

chitosan 2004)

RI
SC
__________________________________ filamentous fungi & N,O-(chloracetyl)-N- (Li and Guo,

yeasts (trimethyl) chitosan 2010)


NU
chloride

Klebsiella pneumonia, Staphylococcus aureus ______ CSNPs (Divya et al.,


MA

Escherchia coli, 2017)

Pseudomonas aeruginosa

________________________________ Yeast Chitosan & trimethyl (Ing et al.,


ED

(Candida albicans) CSNPs 2012)


T

& Fungi
EP

(Aspergillus niger)

& Fusarium solani


C

Escherchia coli, Staphylococcus aureus __________ CSNPs & copper-loaded (Qi et al.,
AC

Salmonella choleraesuis, CSNPs 2004) (Du et

Salmonella typhimurium al., 2008) (Du

et al., 2009)

Escherchia coli ______________ ______________ Chitosan, carboxy- (Liu et al.,

methylated chitosan 2001)

______________ ______________ Chitosan (Liu et al.,

2006)

Staphylococcus aureus Candida albicans chitosan-g– (Hebeish et


ACCEPTED MANUSCRIPT

Aspergillus niger acrylonitrile/silver al., 2015)

nanoparticles

__________ ___________________ Colletotrichum Chitosan/silver (Chowdappa

gloeosporioides nanoparticles composite et al., 2014)

Escherchia coli, Pseudomonas Listeria monocytogenes, ______________ Chitosan & chitosan (No et al.,

fluorescens, Salmonella Bacillus megaterium, oligomers 2002)

typhimurium, Vibrio Bacillus cereus,

PT
parahaemolyticus, Staphylococcus aureus,

Lactobacillus plantarum,

RI
Lactobacillus brevis,

SC
Lactobacillus bulgaricus

Escherchia coli, Salmonella Staphylococcus aureus, ______________ Chitosan (Chung et al.,


NU
typhimurium, Pseudomonas Streptococcus faecalis 2004)

aeruginosa
MA

______________ Streptococcus mutans ______________ CSNPs (de Paz et al.,

biofilms 2011)

Aeromonas hydrophila, ______________ ______________ Chitosan (Yildirim-


ED

Edwardsiella ictaluri, Aksoy and

Flavobacterium columnare Beck, 2017)


T
EP

Escherchia coli, Vibrio Streptococcus uberis, ______________ Chitosan microparticles (Jeon et al.,

cholerae, Staphylococcus aureus, 2014)


C

Salmonella enterica, Enterococcus


AC

Klebsiella pneumonia
ACCEPTED MANUSCRIPT

Table 2

Immunomodulati on against infections Immunomodulati on against stresses

Fish species Pathogen Diets Reference Fish species Stress Diets Reference

Kelp grouper Philasterides (Harikrishnan Olive flounder Oxidative (Samarakoon

(Epinephelus dicentrarchi et al., 2012a) (Paralichthys cellular and et al., 2013)

bruneus) olivaceus) DNA damage

induced by
Chitin and Chitosan

PT
H2 O2 .

Chitosan
RI
Kelp grouper Vibrio (Harikrishnan Caspian kutum Environmental (Najafabad

SC
(Epinephelus alginolyticus et al., 2012b) fingerlings stresses et al., 2016)

bruneus) (Rutilus frisii


NU
kutum)

Indian major carp Aeromonas (Aathi et al., Common carp Oxidative stress (Mehrpak et
MA

(Labeo rohita) hydrophila 2013) (Cyprinus and toxicity of al., 2015)

Carpio L.) CdCl2 on liver

Chitosan and Vitamin C


tissues
T ED

Common carp Aeromonas (Alishahi et Common carp Toxicity of (Sharifinasab


Chitosan
EP

(Cyprinus Carpio hydrophila al., 2014) (Cyprinus paraquat on et al., 2016)

L.) (Mustafa et Carpio L.) muscle tissues


C

al., 2014)
AC

Nile tilapia Aeromonas (Abu-Elala et

(Oreochromis hydrophila al., 2015)

nilotica)

Zebrafish larvae Aeromonas (Nikapitiya et

(Danio rerio) hydrophila CSNPs al., 2018)


ACCEPTED MANUSCRIPT

PT
RI
SC
NU
MA
T ED
EP
C
AC
ACCEPTED MANUSCRIPT

Table 3

Bio-molecules delivery

Bio-molecules Carrier Fish species Delivery Reference

The plant extract Indian freshwater catfish Parenteral (Bhat et al.,

(Eurycomanone) (Clarias magur) administration 2018)

(induces the secretion of

PT
CSNPs

testosterone hormone)

The exogenous nucleotide Indian major carp (Ferosekhan

RI
RNA extracted from yeast fingerlings et al., 2014)

SC
(important nutrients) (Labeo rohita)

Oral administration
NU
“Ulvan” polysaccharide CSNPs Solea senegalensis (Fernández-

extracted from Ulva ohnoi Díaz et al.,


MA

macroalgae 2017)

(immunomodulation)
ED

Vaccines delivery

Vaccine Antigen carrier Fish species Delivery Reference


T
EP

The exogenous reporter Nile tilapia (Ramos et al.,

gene, encoding for the (Oreochromis nilotica) 2005)


C

bacterial enzyme (β-


Chitosan
AC

galactosidase)
Oral administration

Nodavirus DNA vaccine European sea bass juveniles (Valero et al.,

(Dicentrarchus labrax) 2016)

The aerolysin antigen of Oleoyl- Common carp (Liu et al.,

Aeromonas hydrophila carboxymethy- (Cyprinus carpio) 2016)

chitosan/hyaluronic
ACCEPTED MANUSCRIPT

acid

pDNA containing major Chitosan Japanese flounder (Tian et al.,

capsid protein gene of microspheres (Paralichthys olivaceus) 2008)

lymphocystis disease virus

The surface antigens of Chitosan/gantrez Turbot Parenteral (León-

PT
Philasterides dicentrarchi microparticles (Scophthalmus maximus) administration Rodríguez et

parasite causing al., 2012)

RI
scuticociliatosis

SC
The OMP antigen of Indian major carp juveniles Parenteral (Behera and
NU
Aeromonas hydrophila (Labeo rohita) administration Swain, 2013)
chitosan composite microspheres

Oral (Behera and


Alginate-coated

MA

administration Swain, 2014)

Model pDNA vaccine Gilthead sea bream juveniles Oral & parenteral (Sáez et al.,
ED

(Sparus aurata) administration 2018)


T
EP

pDNA constructed from the Asian sea bass (Vimal et al.,

RNA2 capsid protein gene (Lates calcarifer) 2014)


C

of nodavirus
AC

Oral administration

pDNA vaccine of Vibrio Asian sea bass (Vimal et al.,


CSNPs

anguillarum Juveniles 2012)

(Lates calcarifer)

pDNA encoding OMP38 of Asian sea bass (Rajesh

Vibrio anguillarum (Lates calcarifer) Kumar et al.,

2008)
ACCEPTED MANUSCRIPT

A combination of OMP K black seabream (Acanthopagrus (L. Li et al.,

gene of Vibrio schlegelii) Bleeker 2013)

parahaemolyticus strain and

the eukaryotic expression

vector against infection.

DNA coding for the replicas Atlantic salmon (Rivas-

PT
of alphavirus of infectious (Salmo salar) Aravena et

salmon anemia virus al., 2015)

RI
SC
The recombinant OMP fringed-lipped peninsula carp (Dubey et al.,

A of Edwardsiella tarda (Labeo fimbriatus) 2016)


NU
MA
ED
T
C EP
AC
ACCEPTED MANUSCRIPT

Table 4

Bioactive ingredients Fish species Delivery Carrier Reference

Vitamins Rainbow trout (Alishahi

(Oncorhynchus mykiss) et al.,

2011a)
Vitamin C

PT
Brachionus plicatilis & the (Jiménez-

post-metamorphic larvae of Fernández

RI
Solea senegalensis et al.,

SC
2014)

Oral delivery

CSNPs
Enzymes The exogenous Indian major carp (Kumari et
NU
proteolyses enzyme (Labeo rohita) al., 2013)

“trypsin”
MA

Aromatase inhibitors Indian freshwater catfish (Wisdom

“letrozole” for the fingerlings et al.,


ED

gonadal development (Clarias magur) 2018)

of fish
T
EP

Hormones Salmon gonadotropic Female grass carp Intramuscular Chitosan & (Rather et

hormone "leutinizing (Ctenopharyngodon idellus) injection chitosan gold al., 2013)


C

hormone releasing nanoconjugates


AC

hormone" (LHRH)
ACCEPTED MANUSCRIPT

Table 5
Fish meat preservation during chilled storage
Fish species Coating/Film Duration/°C Reference

Indian oil sardine Edible chitosan coatings up to 8 days for 1% (Mohan et


(Sardinella longiceps) chitosan and 10 days for al., 2012)
2% chitosan /(4°C)

PT
Large yellow croaker fillets (Li et al.,
(Pseudosciaena crocea) Chitosan combined with tea polyphenol 20 days/(4°C) 2012)

RI
& rosemary & grape seed extract (T. Li et al.,
Red drum fillets extracts 2013)

SC
(Sciaenops ocellatus)

Large yellow croaker Chitosan combined with different 8 days/(4°C) (Guohua et


NU
(Pseudosciaena crocea) concentrations of nisin peptide al., 2016)
MA

Japanese sea bass fillets Chitosan incorporated with citric acid & 12 days/(4°C) (Qiu et al.,
(Lateo labrax japonicas) licorice extract 2014)

Raw grass carp fillets Chitosan combined with glycerol 7 days/(4°C) (Yu et al.,
ED

(Ctenopharyngodon idellus) monolaurate & clove bud essential oil 2018)


T

Rainbow trout fillet FG/chitosan coating & film 16 days/(4±1°C) (Nowzari et


(Oncorhynchus mykiss) al., 2013)
EP

Golden pomfret fillet FG/chitosan up to 17 days/(4°C) (Feng et al.,


C

(Trachinotus blochii) 2016)


AC

Dolphinfish FG/chitosan incorporated with clove 10 days/2°C (Gómez-


(Coryphaena hippurus) essential oil with edible films Estaca et al.,
2010)
Minced trout fillet FG/chitosan films incorporated with (1 11 days /(4 ± 1 °C) (Kakaei and
& 2%) grape seed extract & (1 & 2%) Shahbazi,
Ziziphora clinopodioides essential oil, 2016)
either separately or in combination.
ACCEPTED MANUSCRIPT

Silver carp fillets (Ramezani et


(Hypophthalmicthys molitrix) al., 2015)
Chitosan & CSNPs coating
Carangoides coeruleopinnatus (Alboghbeish

12 days/(4°C)
fillets and
Khodanazary,
2018)

Silver carp fillets CSNPs incorporated with orange & (Zarei et al.,

PT
(Hypophthalmicthys molitrix) pomegranate peel extracts 2015)

RI
Fish meat preservation during frozen storage
Fish species Coating Duration / °C Reference

SC
Silver carp fingers Chitosan & CSNPs up to 6 months/ -18°C (Fan et al.,
(Hypophthalmicthys molitrix) 2009)
NU
Silver carp (Hypophthalmicthys 2% aqueous chitosan solution 30 days/ -3°C (Fan et al.,
molitrix) 2009)
MA

Salmon 0.5% and 0.75% chitosan solutions 14 weeks/ -5°C (Soares et al.,
directly on the surface of frozen fish 2013)
ED

Rainbow trout Chitosan & Pomegranate peel extract up to 6 months/ -18°C (Berizi et al.,
(Oncorhynchus mykiss) 2018)
T
C EP
AC
ACCEPTED MANUSCRIPT

Highlights

Chitosan-based complexes are effective immunomodulators of fish and crustaceans.

Chitosan nanoparticles utilization for controlled drug delivery and sustained release.

Chitosan improves fish meat and oil preservation.

PT
RI
SC
NU
MA
T ED
C EP
AC
Figure 1
Figure 2
Figure 3
Figure 4

Вам также может понравиться