You are on page 1of 19

Fish and Shellfish Immunology 114 (2021) 263–281

Contents lists available at ScienceDirect

Fish and Shellfish Immunology


journal homepage: www.elsevier.com/locate/fsi

Overview of the latest developments in the role of probiotics, prebiotics and


synbiotics in shrimp aquaculture
Usman Dawood Butt a, Na Lin b, Najeeb Akhter c, Tooba Siddiqui d, Sihui Li a, Bin Wu a, *
a
Ocean College, Zhejiang University, Zhoushan, 316021, China
b
Lishui Hospital of Traditional Chinese Medicine, Lishui, 323000, China
c
Centre of Excellence in Marine Biology, University of Karachi, Karachi, 75270, Pakistan
d
Institute of Marine Science, University of Karachi, Karachi, 75270, Pakistan

A R T I C L E I N F O A B S T R A C T

Keywords: With the growing world population, the demand for food has increased, leading to excessive and intensive
Probiotics breeding and cultivation of fisheries, simultaneously exacerbating the risk of disease. Recently, shrimp producers
Prebiotics have faced major losses of stocks due to the prevalence of periodical diseases and inappropriate use of antibiotics
Synbiotics
for disease prevention and treatment, leading to bacterial resistance in shrimp, along with imposing health
Shrimp aquaculture
Litopenaeus vannamei
hazards on human consumers. Strict regulations have been placed to ban or reduce the use of prophylactic
antibiotics to lessen their detrimental effects on aquatic life. Dietary and water supplements have been used as
substitutes, among which probiotics, prebiotics, and synbiotics have been the most beneficial for controlling or
treating bacterial, viral, and parasitic diseases in shrimp. The present analysis addresses the issues and current
progress in the administration of pro-, pre-, and synbiotics as disease controlling agents in the field of shrimp
farming. Furthermore, the benefits of pro-, pre-, and synbiotics and their mechanism of action have been
identified such as; strengthening of immune responses, growth of antibacterial agents, alteration in gut micro­
flora, competition for nutrients and binding sites, and enzymes related activities. Overall, this study aims to
depict the antagonistic action of these supplements against a variety of pathogens and their mode of action to
counter diseases and benefit shrimp species.

1. Introduction human food consumption, and it is expected to reach 109 mt by 2030. A


rise of 32% is estimated, reaching to 26 mt over 2018 [3]. The increase
It is believed that aquaculture initially started in China and has now in shrimp aquaculture practices in the last few decades is due to the
become one of the fastest-growing sectors of the animal-based food rising demand and shrimp’s nutritional quality (see Table 1). According
supply in the world [1]. Due to the increased demand for food from to the latest survey by the Boston Consulting Group (BCG) in 2019,
marine and freshwater resources, aquaculture has emerged as an alter­ global production of shrimp has tripled in less than two decades, and
native source of nourishment and nutrition. Its usage in raising marine cultivated shrimp is listed as one of the fastest-growing foods in the
and freshwater crustaceans in a culturally and artificially controlled world [4]. It is estimated that global farmed shrimp production reached
environment, has not only helped to meet the demand for consumption 4.455 mt in 2019, rising to about 17% compared to 2018 [5]. However,
but also to compensate for the scarce resources. According to the United the latest pandemic, Coronavirus (COVID-19) has hindered global
Nations Food and Agricultural Organization (FAO), most of the development and prospects, which has led to a decreased demand for
economically and commercially important fish (including shrimp) shrimp, shortage of labour, and suspension of trade between countries;
stocks have been destroyed or overfished and as a result, are either increasing the likelihood that the economy will struggle in the near
extinct or on the verge of collapse [2]. In contrast, the global production future [5].
of aquaculture reached around 82.1 mt in 2018, accounting for 46% of Since aquatic species interact closely with the surrounding envi­
the world’s overall fish (including shrimp) production and 52% of ronment, aquaculture microbiomes have a direct impact on their health

* Corresponding author.
E-mail addresses: usman_butt@zju.edu.cn (U.D. Butt), 854954086@qq.com (N. Lin), na_memon@marinebiology.edu.pk (N. Akhter), toobasiddiqui202@gmail.
com (T. Siddiqui), 21934038@zju.edu.cn (S. Li), wubin@zju.edu.cn (B. Wu).

https://doi.org/10.1016/j.fsi.2021.05.003
Received 17 January 2021; Received in revised form 4 April 2021; Accepted 3 May 2021
Available online 8 May 2021
1050-4648/© 2021 Elsevier Ltd. All rights reserved.
U.D. Butt et al. Fish and Shellfish Immunology 114 (2021) 263–281

Table 1 the action of cortical sensing in pathogens [36]. At present, there is


Nutritional value of shrimp [213]. evidence that probiotics can not only enhance the absorption of nutri­
NUTRITIONAL VALUE ents and increase stress resistance but also enhance the fertility of the
host species [37]. According to Llewellyn et al. [38] probiotics can also
Amount Per 100g Serving
Total Calories 118 .8 (15.3 From Fat) be added to the culture system to improve water quality and to monitor
DV % * infectious diseases caused by bacteria (Fig. 1). For example, a variety of
Total Fat 1.6g 3% bacteria, phages, microalgae, and yeast are conventionally used in
Saturated Fat 0.5g 3% aquaculture or feed supplements. Probiotics can improve immunity and
Cholesterol 210.6 mg 70%
Sodium 947.2 mg 29%
survival rate, produce digestive enzymes and reduce disease occurrence
Potassium 170.6 mg 5% (Fig. 1) [39,40]. There are several prototypes commercially available in
Total Carbohydrates 1.5g 1% mono or multi-stains [41]. However, the development phase of com­
Proteins 22.4g mercial probiotic-based feed requires rigorous experiments, formation
Vitamin A 6%
of suitable observatory tools, and controlled manufacturing [25,42].
Calcium 9.1%
Iron 1.8%
1.2. Prebiotics
* DV = Daily Value.
* Daily Values % is Based on 2000 Calorie Diet.
The concept of prebiotics was first introduced by Gibson and
Roberfroid who defined it as “an indigestible fibre that can enhance the
and development. Therefore, investigating the microbial fauna of growth and activity of health-promoting bacteria in the intestine and
aquatic species and their surroundings is important [6]. As shrimp are beneficially affect the host” [43]. Similarly, Bindels et al. [44] described
closely interdependent on their environment, any changes can lead to prebiotics as “non-digestible food ingredients that are broken down into
various types of viral and bacterial diseases. Therefore centralized, simpler substances, enhancing the growth and activity of preferred mi­
intensive, and rigorous management is needed to conserve shrimp croorganisms found in the gastrointestinal tract and benefiting the
stocks. Due to their proven beneficial effect, the use of probiotics, pre­ health of the host”. Guerreiro et al. [45] presented them as “useful food
biotics, and synbiotics in aquaculture has now become a popular prac­ ingredients that have numerous favourable effects on the host”.
tice [7–17]. Recently, various evaluations of professional pro-, pre-, and Recently, the application of antibiotics in aquaculture has been
synbiotics have been carried out in shrimp aquaculture, namely for the widely criticized, due to the growth of antibiotic-resistant strains, the
activation of innate, humoral, and cell-mediated immunity, as dietary emergence of antibiotic residues in the host, changes in the microbial
and nutritional supplements, environmental capacity builder agents, population of the gut and the culture system, and reduced immunity
growth promoters, and prophylactic against infectious diseases. These [46–49]. Probiotics may not survive or grow during the production,
supplements also directly induce artificial immune responses, including preserving and pelleting process of the probiotic-based feed. The feed, if
activating phagocytes, stimulating neutrophils, activating alternative prepared under inappropriate and adverse conditions may reduce its
regeneration systems, and enhancing lysosomal activity. efficiency (Fig. 1) [50]. Thus, prebiotics have been recently adopted as
However, some studies have raised concerns about the continuous an alternative to antibiotics and probiotics. Several analyses and com­
and inappropriate use of pro-, pre-, and synbiotics, as they may lead to ments have been published emphasizing the beneficial effect of pre­
changes in the bacterial composition of the culture system and the biotics in aquaculture [45,51–54]. Inulin, fructooligosaccharides (FOS),
naturally protected flora of the cultured species. This increases the host short-chain fructooligosaccharides (scFOS), mannan oligosaccharides
animal’s vulnerability to opportunistic pathogens and reduces the feed (MOS), galactooligosaccharides (GOS), xylooligosaccharides (XOS),
conversion rate due to the disruption of the GIT microflora [13]. Thus, arabinoxylooligosaccharides (AXOS), isomaltooligosaccharides (IMO),
developing an experimental test for identifying and checking strain and GroBiotic-A, which are usually derived from plants and fibres, are
mechanisms, a clear understanding of the professional pro-, pre-, and commonly recognized prebiotics in shrimp farming [55–57]. Some
synbiotic-based products and studies about the efficiency of these carbohydrates are also classified as prebiotics on the degree of their
products to weigh their strengths or weaknesses is vital. By far, an polymerization into monosaccharides, oligosaccharides, and poly­
insufficient amount of studies has been conducted to examine the saccharides [58].
effectiveness of pro, pre, and synbiotics in shrimp farming, rendering the Prebiotics possess the ability to be metabolized by beneficial bacte­
existing data inadequate for analysis. Thus, this review emphasizes the ria, Lactobacillus and Bifidobacterium, producing short-chain fatty acids
importance of identifying the benefits and limitations of such in­ (SCFAs) and lactate [58]. SCFAs can be absorbed through the intestinal
terventions. It also briefly summarizes the current evidence on the main tract to act as a source of energy while lactate acts as a stimulant for
uses of pro-, pre-, and synbiotic in shrimp aquaculture. It describes and gluconeogenesis [59]. The production of SCFAs helps to reduce the pH
focuses on the most significant characteristics that have been proven and of the colon, which prevents the proliferation of pathogens and enhances
disclosed to date. the growth of Bifidobacterium and other lactic acid-type bacterial species
[17]. Similarly, the administration of prebiotics promotes the growth of
1.1. Probiotics beneficial bacteria and reduces the number of pathogenic bacteria by
competing with glycoconjugates on the epithelial lining, resulting in
Probiotic is a Greek term, where pro means benefit, and bios means mucus production, increased SCFAs and cytokines [50,60]. They posi­
life [18]. It is a bacteria-based product in which essential bacteria is used tively affect the growth parameters of the host, such as final weight,
to promote health, prevent multiple diseases and pathogenic growth in weight gain, daily weight gain, specific growth rate, dietary conversion
aquaculture. The use of non-pathogenic bacteria can also improve di­ ratio, feed efficiency ratio, and protein efficiency ratio [61–64].
etary safety and environmental performance, which can control allergies A diet supplemented with prebiotics can also increase digestive
and improve the immune system by altering host-related microorgan­ enzyme activity (Fig. 1) [62,65]. Increased levels of digestive enzymes
isms [19–21]. Several gram-positive and gram-negative bacterial can­ associated with weight gain and effective feed conversion efficiency
didates such as Bacillus species [22–28], Vibrio species [29–31], lactic were reported [62], but consistent improvements were not observed as
acid bacteria [32], and some other bacterial species [13,33–35] have some studies also reported ineffective behaviour of prebiotics [54,65,
been used as probiotics in shrimp aquaculture in recent times. 66]. Prebiotics can further enhance immunity [61,66] by activating the
Probiotics are mainly focused on energy production, immune non-specific immune system and promoting beneficial bacterial health
response, and the production of inhibitory compounds which impairs [67]. The beneficial microbiota provides protection against harmful

264
U.D. Butt et al. Fish and Shellfish Immunology 114 (2021) 263–281

Fig. 1. Constraints and potentials of probiotics, prebiotics and synbiotics in shrimp aquaculture.

bacteria present in the colon, and improves the host’s immunity, and various shrimp species [15,80–82]. Their effectiveness in shrimp aqua­
also provides a clear balance between the gastrointestinal and immune culture is further demonstrated by increased stimulation of host species’
system’s microbiota [68]. Prebiotic candidates must fulfill the following growth and survival [83]. This occurs via the secretion of pancreatic
criteria by both in vivo and in vitro experiments to be considered as po­ enzymes which leads to improvement in enzymatic digestion, modula­
tential prebiotics: (i) they must be resistant to gastric/abdominal acidity tion in intestinal microflora, reduced rate of mortality by changing the
(ii) initiate various enzymatic processes and effects (iii) enhance potential immune responses, and antagonism to pathogenic species
gastrointestinal digestion (iv) support microbial fermentation (v) stim­ (Fig. 1) [82,84–86].
ulate the activity of desirable microbes that can confer health benefits
[44,69]. However, not all prebiotics have these specifications [50]. 2. Modes of application

1.3. Synbiotics Probiotics can be classified into several types depending on their
application;
The term synbiotic is a combination of two Greek words "συν" and
"βíoς", meaning “joint or together” and “life” respectively [70]. The term
synbiotics denotes a dietary supplement that includes both prebiotics
and probiotics in a synergistic relationship. A nutrient that helps pro­
mote the growth and survival of probiotics in the gastrointestinal tract of
the host species by altering the opportunistic bacterial community [71,
72]. The choice of prebiotics for the formulation of synbiotic is based on
their ability to indirectly promote the growth and survival of beneficial
bacteria within their target range. Probiotics are chosen based on their
specific beneficial effects on the host. However, the choice of prebiotics,
as per its synergistic effects, is based on their ability to enhance the
growth of probiotic bacteria as its primary function, while the choice of
probiotics as per its synergistic effects is based on their overall benefits
on the host correspondingly [70]. Most recent studies have focused and
emphasized solely on the synergistic effects of synbiotics [73–75].
The composition of synbiotics depends mainly on the mechanism of
action of prebiotics in the host species [76]. However, prebiotic with
low-degree-of-polymerization is better suited for the development of
synbiotics than the ones with higher-degree-of-polymerization [77], as
they hydrolyze the grown beneficial strains and produce primary and
secondary metabolites that may have beneficial effects on the host [76].
Additionally, the administration of synbiotics has been proven to be
more effective than the use of probiotics alone [78], as better survival of
probiotics has been observed when used in combination with prebiotics
and vice versa. Prebiotics, however, provide better resistance to signif­
icant changes in oxygen, temperature, and pH levels [79]. The following
studies have reported synbiotic effectiveness as a positive alternative to Fig. 2. Common viral diseases in shrimp.

265
U.D. Butt et al. Fish and Shellfish Immunology 114 (2021) 263–281

a) Probiotics that are collected from the gut and can be orally admin­ d) Raw addition of prebiotic-rich Artemia nauplii increased shrimp
istered in combination with feed to improve the beneficial microflora larval and post-larval survival [92].
of the intestine [87].
b) Those that are administered as water additives and can grow in the Similarly, synbiotics also have several forms depending on their
water medium by absorbing all the ingestible food thus, resulting in applications;
starving of pathogenic bacteria due to malnutrition [88].
c) Which can be administered through micro-encapsulation, and have a a) The incorporation of synbiotics in the diet results in better growth
direct and positive impact on water quality, physical parameters, and [93], strengthened immune responses, and resistance against disease
shrimp health [26]. [74,94].
d) In the form of living cells [30]. b) Bio-encapsulation of Artemia species with synbiotics not only pro­
e) Those that can be administered via probiotic-rich Artemia or micro­ mote growth but also provide optimal size and nutrients to growing
algae and can promote growth and survival during the nursing phase larvae of shrimp [80].
[28]. c) Micro-encapsulation of synbiotics can support the survival and
growth of probiotics bacteria [95] and promote the growth of in­
However, prebiotics are a popular set of alternative disease man­ testinal microflora [96].
agement strategies that promote non-specific immune responses. There
are several varieties of prebiotics, depending on their application. For 3. Mechanism of action
example.
Shrimp are vulnerable to various viral and bacterial epizootics [97].
a) Supplementing an aquatic diet with prebiotics can increase glucose Several infections and diseases have dominated shrimp farming in
uptake [89], and bioavailability of trace elements [90]. recent years (Fig. 2) [98–100]. The periodical damage may result in
b) Prebiotics when used as a water additive for a specific time period, widespread mortality and huge economic losses. It is estimated that
stimulate immune responses and enhance resistance against diseases shrimp disease outbreaks result in global losses of approximately 3
[91]. billion US dollars [101]. The development of new techniques and
c) Bio-encapsulation of Artemia species with prebiotics provided methods was inevitable. Thus, pro-, pre-, and synbiotics were produced
appropriate size for consumption, high nutritional value, and pro­ and effectively used in shrimp aquaculture.
moted larval growth [80]. Some mechanisms have been proposed as the mode of action of

Fig. 3. Expected pond condition with and without probiotic treatment.

266
U.D. Butt et al. Fish and Shellfish Immunology 114 (2021) 263–281

probiotics in recent years but the pathways of their activities in aqua­ Several Bacillus probiotic species are known for enhancing the host’s
culture are not well known. According to some hypotheses, stimulation innate and adaptive immunity by immunostimulatory effects and stim­
or regulation of immunity, enhancement of growth and tolerance, ulation of beneficial gut microflora [124,134]. Bacillus subtilis, Bacillus
antagonism against pathogens, and alteration in gut microflora are the licheniformis, Bacillus coagulans, Bacillus pumilus, Bacillus amyloliquefa­
key mechanisms by which probiotics exert their effects. Several other ciens, Bacillus velezensis and Bacillus megaterium are the most used Ba­
mechanisms of probiotics are also observed during in vitro and in vivo cillus species in shrimp cultures [27,32,135]. The combination of Bacillus
experiments such as production of inhibitory compounds, competition and Vibrio spp. Enhanced the resistance of Litopenaeus vannamei to Vibrio
for adhesion sites, Iron (Fe) and nutrients (as well as nutrient source harveyi and WSSV [136]. Chiu et al. [127] recorded an increase in
themselves), increased enzymatic activities (such as contribution to transcription of peroxinectin (PE) and prophenoloxidase (proPO) mRNA
digestion), bioremediation of accumulated ammonia, nitrite and other as immune profiling factors in white shrimp supplemented with Lacto­
organic material from the culture system and pond bottom through bacillus plantarum. Besides, an increase in activities of superoxide dis­
bacterial mediation (Fig. 3), and antiviral activities [23,26,87,102]. mutase (SOD), phenoloxidase (PO), respiratory burst, and the removal
Recently, numerous articles and reviews on the use of prebiotics in efficiency of Vibrio alginolyticus has also been observed.
shrimp farming have been published [103–106]. Prebiotics’ mechanism These beneficial microbes can further enhance the immunity of
of action observed in various shrimp species includes increased sus­ shrimp significantly by both in vivo and in vitro; for instance, 58%
tainability against disease [107,108], stimulation and modulation of average increase in the survival rate of shrimp Penaeus orientalis (Post
immunity [80,109,110], enhancement, modulation, and alteration of Larvae) in the pond, while 21% on average in control and remarkable
intestinal microbiota [109,111,112], improvement in growth and sur­ reduction in hazardous pathogens in shrimp ponds has been observed as
vival of host species [113–115], antioxidant activity [61,115] and compared to the control [137,138]. Rengpipat et al. [139] mentioned
beneficial changes in enzyme activity [114,115]. Moreover, the use of the use of three different approaches for the welfare of shrimp; a) by
prebiotics enhances the activity of amylase and hydrolysis of carbohy­ using disease-resistant shrimp b) by vaccinating or immunostimulation
drates in shrimps [116]. It facilitates and promotes the growth of and, c) by using probiotic strains for immunity improvement and
beneficial bacteria such as Bifidobacterium and Lactobacillus species exclusion of pathogenic bacteria. However, vaccination or immunosti­
[117]. mulation might not result in effective protection against disease-causing
During the past decades, the role of pro-, and prebiotics in shrimp bacterium [140] due to uncertain immune responses in shrimp [141].
aquaculture have been studied separately, while the concept of syn­ The probiotic treatment has been proven to be broadly effective in
biotics emerged recently. They flourished after their first use in 2009 disease resistance by improving serological immunity and competitive
[118]. Synbiotics have demonstrated several mechanisms of action since exclusion in the shrimp’s gut. The modulation of humoral immune re­
their application on shrimps. According to Boonanuntanasarn et al. sponses and expression of immune-related genes in probiotic-treated
[119], synbiotics can provide greater yields and better sustainability in shrimps by stimulation of innate immunity was observed [40]. Dietary
shrimp aquaculture. Litopenaeus vannamei when administered with supplementation of selected probiotics is also reported to enhance
probiotic Bacillus OJ and isomalto-oligosaccharide as a feed additive, cellular and humoral parameters [142], especially Bacillus species re­
resulted in significant effects on gastrointestinal microbes and antago­ flected as disease-resistant [135]. The use of Bacillus S11 strain
nism against White Spot Syndrome Virus (WSSV) [120]. Synbiotics strengthens the immunity of tiger shrimp [139]. Serum immune stipu­
improved the efficiency of growth-enhancing probiotic bacteria in lations such as lysozyme, peroxidase, superoxide dismutase (SOD),
cultured shrimp [121]. They can also proliferate immune parameters protease and antiprotease, catalase (CAT), and myeloperoxidase (MPO)
such as lysozyme activity, production of hemocytes, respiratory burst have shown vigorous antibacterial activity against infectious microbes
activity, phenoloxidase activity, superoxide dismutase activity, and in­ [133], and increased immune response in the host species [143]. As
crease competitiveness against bacterial and viral infections [122]. compared to serum, few studies emphasized probiotic’s role as mucosal
Moreover, better digestive enzyme activities in shrimp gut have been immunomodulator [144]. Mucus plays an important role in protection
observed which resulted in improved growth [123]. Recently, the syn­ against infectious agents [145] and probiotics interact with
biotics and probiotics in combination with plant products or yeast/β-­ mucosa-associated lymphoid tissues to help strengthen the defence
glucan are used in aquaculture and demonstrated potential benefits on against infections [68]. Several Bacillus species are also identified as
gut microbiota and gut morphology, and mucosal immune responses mucosal immunomodulators [144,146].
[124]. The ability of species to resist certain bacterial and viral loads de­
pends on their antagonism capacity triggered by genetic makeup as well
3.1. Immune modulation and disease resistance as certain immune parameters [147]. Shrimps normally possess
non-specific immune genes which exert defensive responses against
Recently, several studies have illustrated the key functions and ef­ pathogenic strains [148] such as recognition, phagocytosis, melaniza­
ficacy of probiotics as immunostimulants (see Table 2) [13,51,125,126]. tion, cytotoxicity, and cell to cell communication in hemocytes [149].
The major mechanism of probiotics as an immunostimulant is to stim­ Several articles reported the efficiency of prebiotics as immunostimu­
ulate phagocytosis, an inflammatory response before antibody produc­ lator/modulator in aquaculture (see Table 3) [53,54,150]. A diet sup­
tion and antibacterial activity in the host [127,128]. In particular, plemented with Bio-Mos® and β-1,3-D-glucan, promoted growth and
probiotics can effectively modulate the growth of anti-inflammatory survival of Penaeus latisulcatus and increased the efficiency of immune
cytokines such as IL-10 and Transforming Growth Factor b (TGF-b) parameters [151]. The use of xylooligosaccharides in shrimp, promotes
and pro-inflammatory cytokines such as interleukin-1 (IL-1), IL-6, IL-12, growth factors [152], the feed conversion ratio (FCR), protection against
and tumour necrosis factor-a (TNF-a) in several life forms [129]. Like­ disease and potential immune responses [153]. The immunostimulatory
wise, Kim and Austin [130] reported the impact of probiotic isolates function of prebiotics is to interact with pattern recognition (PR) mol­
from rainbow trout on cytokine mRNA expression in head kidney leu­ ecules such as β-glucan and dectin-1 receptors present on macrophages
cocytes and gut cells. These gut leukocyte isolates demonstrated and to activate signalling molecules such as NF-kB, which enhances the
phagocytosis and secretion of bactericidal oxygen free radicals [131]. ability of immune cells [154,155]. The saccharides may also interact
The same isolates from the head kidney when used as a feed supplement with the Microbe-Associated Molecular Patterns (MAMPs) such as tei­
demonstrated enhanced immune function, such as phagocytosis by choic acid, peptidoglycan, glycosylated proteins, and bacterial capsule
neutrophils and macrophages [132], as enhancement of host immunity polysaccharides to stimulate the immune response [156]. Growth
is one important benefit of probiotics when implied as dietary additives stimulation of anaerobic bacteria specifically Bifidobacterium, preven­
[133]. tion of opportunistic/pathogenic adhesion, reduction in pH of the colon

267
U.D. Butt et al. Fish and Shellfish Immunology 114 (2021) 263–281

Table 2
Probiotics evaluated for shrimp aquaculture applications.
Probiotic Strains Source Targeted Mode of Application Concentration and Responses References
Species Duration

a. Gram-Positive Bacteria
Arthrobacter XE-7 Isolated from the L. vannamei Feed Additive 1010 CFU g− 1
feed for 63 Modulates the intestinal [214]
culture water of (Pacific white days microflora, enhanced the
Pacific white shrimp) immune parameters,
Shrimp. significantly decreased
mortalities to
V. parahaemolyticus
Arthrobacter MCCB 104 Isolated from P. monodon Water Additive 106 CFU mL − 1
No Significant activity against [34]
seawater hatchery (Larvae) V. harveyi, enhanced SR as well
as functioned as probiotics
through immunostimulatory
and digestive enzyme-
supporting modes of action.
Probiotic B. thuringiensis Isolated from White Shrimp Water Additive (As a 109 CFU mL − 1 daily3–4 Effectively improves GR, SR, [26]
A BUU 001 P. monodon intestine (Post Larvae) freeze-dried Bacillus or nauplii per shrimp beneficial bacteria, water
B. megaterium microencapsulated 4 times daily quality parameters, especially
BUU 002 Bacillus)Fed with pH, ammonia and nitrite as
B. polymyxa probiotic enriched compared to controls.
BUU 003 Artemia
B. licheniformis
BUU 004
B. subtilis BUU
005
Probiotic B. subtilis BUU
B 006
B. polymyxa
BUU 007
B. megaterium
BUU 009
B. circulans BUU
010
B. pumilus BUU
012
Bacillus pumilus Obtained from the P. japonicas Added as alive cells Starting with 104 to 1010 94.9% Larval survival increase [30]
Bacillus Genetic (Larvae) cells g− 1 daily for 10 after 7 days
Stock Center days
(Columbus, Ohio)
(http://Bacillus.bi
osci.ohio-state.edu)
Bacillus PC465 Isolated from the Litopenaeus Feed Additive 107 CFU g − 1 Higher biomass of microbes as [22]
gut of vannamei 10% of body weight 3 compare to 109 CFU g − 1. DEA
Fenneropenaeus times daily for 30 days such as amylase, protease, and
chinensis lipase, in the mid-gut
significantly increased.
Protects against viral
infections.
109 CFU g − 1 Higher GR as compare to 107
10% of body weight 3 CFU g − 1. Improve SR,
times daily for 30 days digestion, nutrient absorption.
DEA such as amylase, protease,
and lipase, in the mid-gut
significantly increased. Protect
against viral infections.
B. subtilis Not Mentioned White Shrimp Included in diet Not Mentioned Increased SOD, GPX, LYZ & PO [23]
enzymes. Increased SR and GR.
(A) B. subtilisand (B) Not Mentioned White Shrimp Feed Additive 1.2 × 104 CFU/g (1 g/ (A) Very good GR, higher FCR [27]
B. megaterium (Both strains Kg) values, 100% SR and
administered separately) 3 times daily for 4 weeks haemolymph metabolites
positively affected.
(B) Good FCR values, 96.67%
SR and improved culture
oxygen concentration.
B. subtilis Isolated from White Shrimp Feed Additive 105 and 108 CFU g− 1
Improve GR, DEA, resistance to [102]
fermented pickles (juvenile) 8 weeks V. harveyi and upregulation of
immune gene expression.
Commercial Probiotic Belgium, www.inve. Litopenaeus Water Additive 2.5 × 104 CFU mL− 1 for Increased survival and growth [28]
NVE Sanolife® MIC, INVE be vannamei with microalgae Nauplius4-5 to Zoea2 and of zoea and mysis phases.
(B. subtilis, B. licheniformis, (Larvae & probiotic enriched 5.0 × 104 CFU mL− 1 for
B. pumilus) Post Larvae) Artemia Zoea3 to PL10 (in water
daily)
10.0 × 104 CFU mL− 1
(with microalgae daily)
(continued on next page)

268
U.D. Butt et al. Fish and Shellfish Immunology 114 (2021) 263–281

Table 2 (continued )
Probiotic Strains Source Targeted Mode of Application Concentration and Responses References
Species Duration

20.0 × 104 CFU mL− 1


(via Artemia)
Bacillus DMS Series Supplied by Penaeids Water Additive 1 x 104 to 1 x 105 mL− 1
Reduce Vibrio and luminous [215]
Biomanagement for 160 days Vibrio in pond, low Vibrio
Systems, Darwin, numbers in sediments and no
Australia luminous Vibrio occurred.
Improved production.
Bacillus tequilensis YC5-2 Isolated from the Whiteleg Water Additive 1 × 105 CFU mL− 1
daily Improved survival and [25]
B. endophyticus C2-2 and guts of healthy wild shrimp showed remarkable
YC3-b adult shrimp (Larvae) antagonistic activity against
B. subtilis YC2-a Vibrio campbelli, V. vulnificus,
V. parahaemolyticus, and
V. alginolyticus.
Bacillus S11 (As wet or Isolated from black Penaeids Feed Additive ~1010 CFU g− 1 Lack of any success other than [216]
lyophilized cells, or as tiger shrimp 3 times daily for 100 better survival against
saline suspensions) habitats days V. harveyi.
Antagonistic Bacillus spp. Indigenously P. monodon Lab Experiment Average zone of Reduce the pathogenic effect of [24]
isolated Bacillus spp. (Juvenile) inhibition against virulent luminous V. harveyi.
luminescent V. harveyi
strains was 10 mm.
Bacillus S11 Isolated from black Black Tiger Feed Supplement ~1010 CFU g− 1 mixed Significantly larger growth and [217]
tiger shrimp Shrimp (P. with 3 kg of feed daily higher survival and
habitats monodon) for 100 days controlled V. harveyi
infections.
B. subtilis BT23 Isolated from Black Tiger Water Additive After 6 days of treatment Reduced growth of V. harveyi [218]
shrimp culture Shrimp (P. at 106–108 CFU mL− 1 at and
ponds monodon) 28◦ shrimp infected with reduced mortality.
103–104 CFU mL− 1 of
V. harveyi for 1 h then
again treated with the
same amount of BT23 for
1h
B. subtilis UTM 126 Isolated from Litopenaeus Feed Supplement 105 CFU g− 1 Reduced mortality against [172]
shrimp culture vannamei 28 days pathogenic V. harveyi.
ponds (White
shrimp)
B. subtilis E20 Isolated from Litopenaeus Water Additive 109 CFU L− 1 once every Improved larval SR, [219]
human health food vannamei 3 days during 14 days of development, stress resistance,
‘Natto’ (Larvae) a breeding experiment and immune status.
Bacillus OJ + IMO Isolated from the Litopenaeus Feed Additive 108 CFU/g of feed + Immunity improved, intestinal [120]
Pacific white shrimp vannamei 0.2% IMO microbes altered and
(Litopenaeus (White 28 days resistance to WSSV increased.
vannamei) shrimp)
Bacillus P64 Isolated from the L. vannamei By Inoculation 107 bacteria mL− 1
Showed inhibitory and [220]
hepatopancreas of (White 24 h immunomodulatory effect.
healthy wild shrimp shrimp)
Enterococcus faecium MC13 Isolated from Penaeus Feed Additive 200 mg/100 PL in feed Protects against V. harveyi and [35]
brackish water fish monodon Water Additive twice a day V. parahaemolyticus.
and shrimp intestine 107 CFU mL− 1 at the rate
of 200 μl/100 PL in
water twice a day
Streptococcus phocae P180 Feed Additive 200 mg/100 PL in feed Growth and protection against
Water Additive twice a day V. harveyi infection increased.
107 CFU mL− 1 at the rate
of 200 μl/100 PL in
water twice a day
Streptomyces spp. Isolated from P. monodon Feed Supplement 2.5, 5.0, 7.5, or 10.0 g Changes in physical [221]
sediment samples per kg feed twice a day parameters observed.
collected from on day 1, 10 and 20 Improved growth, survival and
Vellar estuary India disease resistance. Decreased
pathogenic Vibriosis
Streptomyces spp. Isolated from Black Tiger Feed Supplement Streptomyces cells at 1% Enhanced GR and survival [33]
marine sediment Shrimp (Post 15 days in 3 treatments against V. harveyi.
samples of shrimp Larvae)
farms at
Queensland,
Australia
Bacillus coagulans NJ105 Isolated from Penaeus Water Additive 1 × 107 CFU mL− 1
daily Beneficial in terms of [32]
shrimps and vannamei for 35 days increasing GR and enhancing
common carp ponds immune responses, such as PO
activity, SOD activity, POD
activity and antibacterial
activity.
b. Gram-Negative Bacteria
Pseudomonas 1-2 Shrimp [222]
(continued on next page)

269
U.D. Butt et al. Fish and Shellfish Immunology 114 (2021) 263–281

Table 2 (continued )
Probiotic Strains Source Targeted Mode of Application Concentration and Responses References
Species Duration

Isolated from Lab ExperimentWater In well and disc diffusion Showed good inhibition in
estuarine water Additive assay diameter of the pathogenic Vibrio damsela,
sample collected zone of inhibition was 17 Vibrio harveyi, Vibrio fluvialis,
from Mangalore, and 9 mm respectively. Vibrio parahaemolyticus and
India In cross-streak method, Vibrio vulnificus.
inhibition was averaging Nonpathogenic as a water
41.4 mm in all five additive.
pathogens106 CFU mL − 1
Pseudomonas MCCB 102 and Isolated from Penaeus Water Additive 106 CFU mL − 1 Proved to be resistant, [34]
MCCB 103 seawater hatchery monodon antagonistic and inhibitory.
Higher survival against
V. harveyi MCCB 111 was
observed.
Rhodopseudomonas palustris Isolated from Penaeus Water Additive 1 × 107 CFU mL− 1
daily Beneficial in terms of [32]
GH642 shrimps and vannamei for 35 days increasing GR and enhancing
common carp ponds immune responses, such as PO
activity, SOD activity, POD
activity and antibacterial
activity.
Synechocystis MCCB 114 and Isolated from Penaeus Feed Additive Daily for 20 days Exhibited antagonistic activity, [223]
115 surface seawater of monodon (Post higher survival against
Vypeen Island Larvae) V. harveyi, a substantial
reduction or total absence of
Vibrio spp.. Cultures were
found to contain high levels of
protein.
V. fluvialis Isolated from Penaeus Added as alive cells Starting with 104 to 1010 94.9% Larval survival increase [30]
shrimp cultures japonicus cells g− 1 daily for 10 after 7 days.
days
Vibrio fluvialis PM 17 Isolated from the Tiger Shrimp By inoculation in rearing 103 CFU mL− 1 every 3 No effectiveness observed. [29]
Pseudomonas sp. PM 11 gut of farm-reared (Penaeus water days and 7 days intervals
(Both strains administered sub-adult shrimp monodon) for 45 days
separately)
Vibrio gazogenes NCIMB 2250 Isolated from the GI Pacific white Injection 3 x 107 or 3 x 105 Higher mortality observed. [31]
tract of marine shrimp bacteria per shrimp
invertebrates mL− 1
Dragon Feeds 3 x 107 live bacteria Reduced levels of Vibrios in the
Supreme™ + Chitin (V. gazogenes) per gram fore and hindgut. Improved
Powder (Sigma-Aldrich) of feed health & welfare.
+ Live V. gazogenes
culture

Abbreviations: CFU: colony-forming unit, DEA: digestive enzyme activity, FCR: feed conversion rate, GR: growth rate, GI: gastrointestinal, IMO: Iso­
maltooligosaccharide, LYZ: Lysozyme, PO: Phenoloxidase, POD: peroxidase, SOD: superoxide dismutase, SR: survival rate, WSSV: white spot syndrome virus.

by producing SCFAs, suppressing the space for toxin receptors, compe­ Various studies also presented prebiotics in shrimp aquaculture as
tition for nutrition, and up-regulation of immune responses against in­ anti-disease agents (see Table 3). According to Sun et al. [108] the
fections are the immunomodulatory functions of prebiotics [17,157]. sustainability of Pacific white shrimp against Vibrio alginolyticus
White shrimp larvae (Litopenaeus vannamei) when fed with MOS, enhanced by the up-regulation of some defensive parameters such as
enhanced immunity by increasing total hemocyte count, phenoloxidase albumin, superoxide dismutase, lysozyme and phenoloxidase activity.
and respiratory burst activity [80]. This increase can be noticed due to Dietary supplementation of MOS can stimulate the bactericidal activity
recognition of foreign particles through pattern recognition receptors, in plasma and enhance the resistance against Vibrio harveyi [161]. Sac­
intracellular signalling cascade and nodular aggregations [158]. Innate charides are similar to the specific receptors of bacteria, toxic molecules
immunity of shrimp was also enhanced by triggering of immune-related and viruses, so they prevent the adhesion of pathogens by acting as
gene expressions [107] such as lipopolysaccharide, β-1,3-glucan-bind­ competing receptors in intestinal epithelial cells and reduce the preva­
ing protein (LGBT) [159], serine protein and peroxinectin. Diet sup­ lence of specific viral species [160]. Prebiotics are also reported to
plemented with commercial prebiotics also stimulated the expressions of stimulate shrimp’s hemocyte activity to eliminate certain viruses and
immune-related genes in white shrimp such as prophenoloxidase, increase the degree of resistance by activating phagocytosis and
penaeidin, crustin and lysozyme activity in shrimp’s gut [109,110]. expression of antimicrobial peptides [107]. Moreover, microbial surface
Non-specific immune responses such as alkaline phosphate and acid antigens such as peptidoglycans, lipopolysaccharides (LPS), and β-1,
phosphate in serum also improved immune responses [114]. On some 3-glucans stimulate lysis in hemocytes which induces various humoral
occasions, the outcome of prebiotic administration may be defence factors including agglutinins, clotting proteins, lysosomal hy­
dose-dependent; for instance, appropriate doses of β-glucan can accel­ drolytic enzymes and antimicrobial glucan peptides [162,163]. Sabry
erate immune-related genes, expression of Toll-Like Receptors (TLRs), Neto and Nunes [164] observed that the dietary supplementation of β-1,
myostatin and heat shock protein 70 [115]. Higher concentration of 3/1, 6-glucan enhanced resistance genes (R-Genes) of shrimp without
MOS along with β-glucan supplemented with the diet resulted in cell altering their total hemocyte count (THC) and reduced mortality against
damage, suppressed immunity and reduced resistance against in­ viruses; however, the reduction in THC may lead to stress in crustaceans
fections. However, when used in appropriate quantities, it resulted in [165]. These hemocytes are also known to play a vital role in the acti­
increased immunity, humoral responses and respiratory burst activity vation of proPO cascade, cytotoxicity, and haemolymph clotting
[160]. mechanism [149,166].

270
U.D. Butt et al. Fish and Shellfish Immunology 114 (2021) 263–281

Table 3
Prebiotics evaluated for shrimp aquaculture applications.
Prebiotic Source Targeted Species Mode of Concentration and Responses References
Strains Application Duration

a) Oligosaccharides
1
FOS Sucrose, 1–3 fructose, Fish Litopenaeus Diet 1.0, 2.0, 4.0 g kg− feed Significant improvement in SGR, SOD, AKP, [114]
meal, Soybean meal, Fish vannamei Additive for 60 days ACP, [lipase and amylase activities (0.4%)],
oil, Soybean oil, and microbial diversity. GOT, GPT dropped
Cholesterol, Lecithin, with increasing supplementation of FOS
Starch diets.
1
ScFOS Barley & wheat Litopenaeus Feed 0, 0.4, 0.8, 1.2, 1.6 g kg− Remarkable Iincrease in the growth rate, feed [112]
vannamei Additive feed for 8 weeks intake and decreased FCR.
(Juvenile) ScFOS affected the gut microbial community.
0.17g
ScFOS Not Mentioned White Shrimp Feed 0.25, 0.5, 0.75, 1, 2, 4 & No effect on WG, FC, Survival. [111]
(L. vannamei) Additive 8 g kg− 1 feed for 6 weeks ScFOS affected the gut microbial community.
(75.4 ± 0.8g)
IMO Prepared from cornstarch Pacific White Feed 0.2% IMO in feed for 28 No significant effect on microbial population, [120]
Shrimp Additive days immune responses & resistance to WSSV.
(L. vannamei)
1
MOS Cell wall of yeast Tiger Shrimp Feed 0, 1.5, 3, 4.5 g kg− feed Increased growth, FC (3g kg− 1) and survival. [200]
(Penaeus Additives for 48 days No harmful effect on hepatopancreas tissue.
semisulcatus) Reduced protein content with increasing
MOS.
1
MOS Not Mentioned Pacific White Water 12mgL− Supplementation of MOS increased THC, PO, [80]
Shrimp (Larvae) Additive RB and Ssurvival of shrimp larvae.
(M3 – PL12)
MOS & Inulin MOS: Cell wall of yeast Pacific White Diet 2, 5, 4 & 10 mg/g for 4 Significant improvement in the TLC 1, 2, 3, [16]
Inulin: Not Mentioned Shrimp Additive weeks proPO, STAT, crustin, anti-
L. vannamei lipopolysaccharide factor (5 mg/g inulin and
MOS). Decreased mortality against WSSV and
Vibrio alginolyticus challenges.
1
XOS 35% XOS, 65% Defatted L. vannamei Feed 0, 1, 2, 4, 6 g kg− feed Final body weight and WGR not affected. [108]
rice bran (Juvenile) Additive for 60 days SR, VL & A were higher in diet supplemented
with XOS. Provide resistance against Vibrio
alginolyticus.
β-1,4 Coconut meal enzymatic Kuruma Shrimp Diet 0%, 0.02% & 2% MNB Stimulate PO, PA, SR, immune response, [107]
mannobiose hydrolysate (Marsupenaeus Additive for 12 days increase resistance against Vibrio
japonicus) parahaemolyticus.
b) Polysaccharides
Inulin Not Mentioned Fenneropenaeus Water (A) No significant changes in TL, CL, weight/CL [92]
indicus Additive 150 mgL− 1 Selco & SGR of larvae and post larvae.
Larvae (M1- M3) enriched Artemia nauplii, No significant differences between the
3–4 nauplii per larvae 4 survivals of individuals from M1 – M3.
times a day Survival of PL was remarkably higher in the
(B) control group & the group fed with inulin-
60 mgL− 1 Inulin enriched Artemia.
enriched Artemia nauplii,
3–4 nauplii per larvae 4
times a day
(C)
150 mgL− 1 Selco + 60
mgL− 1 Inulin
Post larvae (PL1 – (A)
PL8) 50 mgL− 1 Selco enriched
Artemia nauplii, 3–4
nauplii per larvae 5–6
times a tay
(B)
50 mgL− 1 Inulin
enriched Artemia nauplii,
3–4 nauplii per larvae
5–6 times a day
(C)
50 mgL− 1 Selco + 50
mgL− 1 Inulin
Inulin Blue agave (Agave White Leg Shrimp Feed 0, 1.25, 2.5, 5.0, 10 g Enhanced growth, remarkably increased the [160]
tequilana) (L. vannamei) Additive kg− 1 of feed for 62 days PO.
(1.1 ± 0.08g) Decreased the prevalence of WSSV, Survive
1
(1.09 ± 0.07g) 2.5 g kg− of feed for 73 rate has not increased.
days
c) Fungal Polysaccharides
β –glucan S. cerevisiae P. monodon (Post- Water 0.25, 0.5, 1.0, and 2.0 Significant improvement in WG, PO. Reduced [91]
larvae) Additive mg mL− 1 for 43 days mortality rate and resistance against
V. vulnificus.
1
β − 1, 3 – S. commune P. monodon (Post- Feed 2 g kg− feed for 11 days Significantly improve DR, SR and resistance [224]
glucan larvae) Additive against V. harveyi.
(continued on next page)

271
U.D. Butt et al. Fish and Shellfish Immunology 114 (2021) 263–281

Table 3 (continued )
Prebiotic Source Targeted Species Mode of Concentration and Responses References
Strains Application Duration

β − 1, 3 – S. commune P. monodon Feed 0.2–10 g kg− 1 feed for 20 Significant increase in resistance against [225]
glucan Additive days V. damsela
β − 1, 3 – S. commune P. monodon Feed 2 g kg− 1 feed for 20 days Showed enhancement in sustainability [226]
glucan (Juveniles) Additive against disease and reduced the rate of
− 1
P. monodon (Post- 2 g kg feed for 15 days mortality.
larvae)
β − 1, 3 – S. commune P. monodon Feed 2.0 g kg− 1 feed for 40 Stimulate SR, haemocyte PA, cell adhesion, [227]
glucan (Brooders) Additive days superoxide anion production.
β – glucan S. cerevisiae P. monodon Feed 2 g kg− 1 feed for 3 days Stimulate PO, THC, survival and resistance [228]
Additive against V. harveyi.
− 1
β – glucan S. cerevisiae P. monodon Feed 1–1.3 g kg feed for 75 Significantly increased WG, SGR, survival, [229]
Additive days DR against V. alginolyticus.
β – glucan Cell wall of marine yeast F. indicus (Post- Feed 0.05, 0.1, 0.2, 0.3,0.4 g/ Improved THC, PO, NBT, survival and [230]
larvae) Additive 100g Feed for 21 days resistance to WSSV.
β − 1, 3 – S. cerevisiae L. vannamei Feed 2 g kg− 1 feed for 40 days Stimulate DGC, PO, DGG, TAG, Hc. [231]
glucan (Juveniles) Additive
β − 1, 3 – S. commune L. vannamei Feed 0, 0.2%, 1% in feed for 7 Stimulate PO, SOD, PA, superoxide anion [232]
glucan Additive days production. No significant effect on proPO,
Hc, transglutaminase activity.
1
β – glucan S. cerevisiae L. vannamei Feed 2 g kg− feed for 42 days Stimulate SGR, THC, SOD, PO, superoxide [113]
(Juveniles) Additive anion production.
β – glucan S. cerevisiae L. vannamei Feed 1 or 2 g kg− 1 feed for 35 Significantly increase THC, PO, SOD, RB, [233]
(Juveniles) Additive days disease resistance and resistance to WSSV.
β-1, 3/1, 6- S. cerevisiae L. vannamei Feed 1.0 mg kg− 1 feed for 70 Improved SR. No effect on THC, PO and total [164]
glucan (Juveniles) Additive days protein. Resist against infectious myonecrosis
virus.
β-1, 3/1, 6- G. lucidum L. vannamei Feed 0, 1, 2, and 4 g/kg for 30 Stimulate PO, SOD, superoxide anion [234]
glucan Additive days production.
β – glucan Not Mentioned White Shrimp Diet 0.5 g kg− 1 for 90 days Stimulated the level of lipopolysaccharide [159]
Larvae Additive and β-1, 3- glucan binding protein.
No significant effect on peroxinectin, LYZ,
and THC.
β – glucan Extracted from yeast cell L. vannamei Diet 0, 0.01%, 0.02%, 0.04% Significant increase in WG, SOD, GP, SR, [115]
wall Additive dry diets for 8 weeks protease, amylase, glutathione (0.02% &
0.04%), mRNA expression of TLR, myostatin,
heat shock protein 70. Decreased mRNA
expression of tumour necrosis factor- α,
richness and diversity of IMC (0.04).
MOS & Cell wall of yeast L. vannamei Feed 0, 0.2%, 0.4%, 0.8% for 100% mortality when challenged with [235]
β-glucan (Juvenile) Additive 45 days Vibriosis (0.8%). Significant increase in THC,
RB, PO (0.4% & 0.2%). Immune responses
suppressed (0.8%).
d) Commercial Prebiotics
Immunogen Cell wall of yeast Pacific White Feed 0, 0.5, 1 & 1.5 g kg-1 for No remarkable effect on WG, FCR, SGR, [110]
(β-glucan & Shrimp Additive 60 days Amylase activity.
MOS) L. vannamei (Post Stimulate protease activity (1.5 g kg− 1),
larvae) lipase activity (1 & 1.5 g kg− 1), proPO,
crustin, g-type lysozyme (1.5 g kg− 1),
expression of penaeidin gene (1g kg− 1).
Previda 100% hemicellulose (83% Pacific White Feed 0, 0.2, 0.5, 1.0 & 1.6 g Increased HPC, hyaline cell counts, [109]
sugar, 5% ash, 12% Shrimp Additive kg− 1 feed for 35 days haemolymph glucose, haemolymph protein
hemicellulose extract) L. vannamei (1.6 g kg− 1).
Modification of microbial communities in the
shrimp gut. WG, SR, FCR were not affected by
supplementation of Previda in the diet.

Abbreviations: A: area, ACP: acid phosphate, AKP: alkaline phosphate, CL: carapace length, DGC: daily growth coefficient, DGG: digestive gland glycogen, DR: disease
resistance, FCR: feed conversion rate, FOS: fructooligosaccharide, GOT: glutamic-oxaloacetic transaminase, GPT: glutamic-pyruvic transaminase, GP: growth per­
formance, HPC: haemocyte phagocytic capacity, Hc: hemocyanin, IMO: isomaltooligosaccharide, LYZ: lysozyme, MOS: mannan oligosaccharide, NBT: nitroblue
tetrazolium activity, ProPO: prophenoloxidase activity, PO: phenoloxidase activity, PA: phagocytic activity, PL: post larvae, RB: respiratory burst, SR: survival rate,
SGR: specific growth rate, SOD: superoxide dismutase, STAT: signal transducer and activator of transcription, ScFOS: short-chain fructooligosaccharide, TL: total
length, THC: total haemocyte count, TLR: toll-like receptor, VL: villi length, WG: weight gain, WGR: weight gain rate, WSSV: white spot syndrome virus, XOS:
xylooligosaccharide.

Several studies have stressed the use of synbiotics as immunomod­ amino acids in activation of T lymphocytes, B lymphocytes, natural
ulators (see Table 4). Synbiotics enhance immunity by facilitating the killer cells, macrophages, as well as lymphocyte proliferation, and the
growth of beneficial bacterial strains colonized on mucus lining and production of antibodies, cytokines, and other cytotoxic substances
prevent the pathogenic growth by competing for substrates and adhe­ [167,168]. They activate non-specific immune responses by enhancing
sion sites [84]. The host’s mucosal immune system serves as the first line respiratory burst, phenoloxidase, nitric oxide synthetase, and serum
of defence as it is exposed to the outside environment, while intestinal lysozyme activity [82]. Similarly, the dietary administration of
epithelium acts as a natural barrier for the digestive tract by increasing beta-glucan along with probiotic strains (B. subtilis and Pediococcus
antagonism against pathogens (see Table 4) [84]. Synbiotic metabolites acidilactici) enhanced the immunity of shrimp by stimulating superoxide
also act as immunity boosters in aquaculture, for example, the role of dismutase activity and also induced the gene expressions of serine

272
U.D. Butt et al. Fish and Shellfish Immunology 114 (2021) 263–281

Table 4
Synbiotics evaluated for shrimp aquaculture applications.
Probiotic Candidate Prebiotic Targeted Mode of Concentration and Duration Responses References
Candidate Species Application

B. licheniformis IMO Penaeus Feed Additive (A) Stimulated TBC, THC, PO, RB, LYZ, NOS, [82]
+ japonicus 108 CFU g− 1 B. licheniformis & SOD. Decreased Vibrio counts, cumulative
B. subtilis 0.2% IMO (T1) for 56 days mortality.
(B) Positive effects on bacterial flora &
108 CFU g− 1 B. subtilis & 0.2% activation of non-specific immunity.
IMO (T2) for 56 days
(C)
108 CFU g− 1 B. licheniformis +
B. subtilis & 0.2% IMO (T3) for 56
days
Bacillus sp. Mixture of White Shrimp Feed Additive 0, 1.5, 3 g kg− 1 or Powder of 109 Significantly increase WG, SGR, body [201]
Herbs L. vannamei CFU g− 1 (Bacillus) + 0, 1, 2 g lipid (1 g kg− 1), stimulated amylase and
(1.91 ± 0.03g) kg− 1 (Mixture of medical herbs) protease activities.
for 10 weeks
B. subtilis β – glucans White Feed Additive (1) Diet β-glucan + B. subtilis induced [159]
+ Shrimp Larvae 5 × 107 CFU kg− 1 (β-glucan + B. expression of serine protease and PO
Pediococcus (156 ± 39 mg) subtlis) for 90 days genes.
acidilactici (2) Diet β-glucan + P. acidilactici improved
0, 5 × 107 CFU kg− 1 (β-glucan + SOD & PO.
P. acidilactici) for 90 days
B. subtilis β – glucans L. vannamei Feed Additive 5 × 107 CFU kg− 1 (Probiotics) Remarkable increase in GP, BPC (β-glucan [119]
+ (PL 5) + & β-glucan + P. acidilactici), haemolymph
Pediococcus 0.35 g kg− 1 (β-glucans) for 90 glucose level, osmolarity (β-glu), the
acidilactici days height of intestinal villi (β-glucan +
P. acidilactici), intestinal LAB (β-glu +
B. subtilis). Decreased Vibrio species. No
remarkable changes in HLTG, cholesterol,
protein, haemolymph urea nitrogen or
chloride.
Bacillus OJ IMO White Shrimp Feed Additive 0, 108, 1010 CFU g− 1 (Bacillus) Significantly improved immune response, [120]
L. vannamei + AKP, PA, PO, RB, SR, disease resistance.
(1.75g) 0.2% IMO for 28 days Decreased TBC, Vibrio counts.
Mixture of Bacteria Inulin White Shrimp Feed Additive 0, 105 CFU g− 1 (Probiotics) Stimulated THC, ACP in haemocyte lysate [202]
(2LAB + 2Bacillus) L. vannamei + supernatant. Increased GP, SR, SGR,
(1.4 ± 0.31g) 0, 0.4, 0.8 g kg− 1 (Inulin) for 60 enzymatic activity of several enzymes in
days plasma.
Decreased prevalence of WSSV.
Pediococcus parvulus Mixture of White Shrimp Feed Additive 0, 2.1, 4.2, 8.4 mg kg− 1 Significantly increased PO, protein [236]
+ Echinacea L. vannamei (Probiotics) concentration of haemocyte lysate
Candida parapsilosis purpurea + (3.6 ± 0.58g) + supernatant.
(yeast) Uncaria 0, 1, 2, 4, g kg− 1 (Prebiotics) for Decreased prevalence of WSSV.
tomentosa 21 days
Pseudoalteromonas MOS Pacific White Through bio- 106 CFU mL− 1 (P. piscicida 1UB) Stimulated THC, PO, RB, SR, expression [80]
piscicida 1UB Shrimp encapsulation of + of immune-related genes, increased
(Larvae) Artemia 12 mg L− 1 (IMO) for 13 days disease resistance.
M3 – PL12
Lactobacillus GOS White Shrimp Feed Additive 108 CFU kg− 1 (L. plantarum 7–40) Improved colonization of Lactobacillus [86]
plantarum L. vannamei + plantarum and reduced prevalence of
0.4% GOS for 60 days Vibrio harveyi and Photobacterium
damselae in the intestines.
Lactobacillus GOS White Shrimp Feed Additive 108 CFU kg− 1 (L. plantarum 7–40) Significantly improved WG, LAB, [85]
plantarum L. vannamei + protease, leu-aminopeptidase and β
(Post-larvae) 0.4% GOS for 60 days -galactosidase activity. Reduced Vibrio
counts in the intestine, DEA.
Vibrio alginolyticus 1.115% inulin, Pacific White Feed Additive Diet P1 (daily), Diet P2 (twice a Significant increase in GP, enhanced SR, [15]
SKT-bR 1.015% FOS, Shrimp week), Diet P3 (once a week) for PO, RB in shrimp fed with diet (P1), THC.
1.488% GOS L. vannamei 30 days
(PL 15)
1
Bacillus sp. NP5 Ipomoea batatas White Shrimp Encapsulated diet 0.5%, 1%, 2% g kg− for 30 days Improved GP, FCR, THC, PO, differential [96]
L (sweet potato) L. vannamei haemocyte count, intestine bacteria.
Bacillus sp. NP5 Rf Ipomoea batatas White Shrimp Mirco- Group A (Once a week), Group B Improved GR & SR, stimulated immune [14]
L (sweet potato) L. vannamei encapsulated diet (Twice a week), Group C (Daily) responses in treatment C as compared to
for 40 days treatment A and B. Decreased FCR in
treatment C. Overall, Improvement in
intestinal bacterial count.
Probiotic SKT-b Ipomoea batatas White Shrimp Feed Additive 1% Probiotic +2% Prebiotic for Stimulated THC, PA activity, and PO [103]
L (sweet potato) L. vannamei 14 days activity.
Pseudoalteromonas MOS Pacific white Through Enriched 10 g/L (Pseudoalteromonas Promoted SGR & SR, stimulated THC, PO [81]
piscicida 1Ub shrimp larvae Artemia piscicida 1Ub) + & RB. It increased bacterial population
12 mg L− 1 (MOS) for 13 days and antagonism against disease.
Bacillus NP5 RfR MOS Pacific White Mirco- 0.5%, 1%, 2% for 30 days Stimulated THC, RB, and ProPO. [83]
Shrimp encapsulated Significant increase in growth & survival.
L. vannamei synbiotic Decreased FCR as compared to the control
group.

273
U.D. Butt et al. Fish and Shellfish Immunology 114 (2021) 263–281

Abbreviations: ACP: acid phosphate activity, AKP: alkaline phosphate activity, BPC: body protein content, CFU: colony-forming unit, DEA: digestive enzyme activity,
FCR: feed conversion rate, GP: growth performance, GOS: galactooligosaccharides, HLTG: haemolymph triglyceride, IMO: isomaltooligosaccharide, LYZ: lysozyme,
LAB: lactic acid bacteria, MOS: mannan oligosaccharide, NOS: nitric oxide synthetase, PO: phenoloxidase, PA: phagocytic activity, RB: respiratory burst, SR: survival
rate, SGR: specific growth rate, THC: total haemocyte count, TBC: total bacterial count, WG: weight gain, WSSV: white spot syndrome virus.

protease and phenoloxidase [159]. Synergistic effects of synbiotics may from the infected GI tract [175] and prevents the disease occurrence by
also enhance immune parameters of shrimp such as phagocytosis, res­ interfering with the disease cycle. Several gram-positive, gram-negative,
piratory burst activity and alkaline phosphate activity and reduce Vibrio and lactate-producing probiotics have superposed other bacteria in
counts [120]. These effects further protect shrimp against WSSV and adhesion capacity [40,190,191]. Probiotic Bacillus can replace Vibrio
Vibrio harveyi [96,120]. Several other scholars also observed increased through competitive exclusion for nutrients and adhesion sites [40],
immunity to various infections due to increased hemocyte count, phe­ thereby becoming a prepotent component of intestinal microflora (see
noloxidase and respiratory burst activity [15,81,96] which resulted in Table 2) [192]. Furthermore, in vitro and in vivo experiments suggested
better protection against pathogens [14,103]. that some potential strains are able to attach to the mucus by dislocating
pathogenic bacteria [190] to compete for essential nutrients and space.
3.2. Competitiveness against pathogens The adhering mechanism of probiotics can be summarized in the
following steps; (a) it starts with attraction (b) binds to surface-secreting
Probiotics are well known for their antagonism against pathogens in gel and (c) ends with attachment to animal tissue cells [132]. However,
the host species as well as in the culture system (see Table 2). Several constant exposure to inadequate feeding ratios may lead to a reduction
mechanisms are defined by which probiotic bacteria can induce bacte­ in probiotic’s efficiency as well as intermittent production of organic
rial antagonism such as production of siderophore substances, but more matter and nutrients (Fig. 4). Vine et al. [175] proposed the idea of
importantly, antimicrobial agents (antibiotics, antimicrobial peptides) careful determination of dose-effect relationship to avoid such
[169]. The application of probiotics can further induce a competitive conditions.
exclusion process, preventing pathogens from developing vital resis­
tance genes. This ecological process can be intentionally altered to 3.3. Alteration in gut microflora
modify the microbial composition of pond water and soil. Numerous
Bacillus species having the ability to produce opportunistic antibiotics The complex polymicrobial ecology of the alimentary canal of
and metabolites in response to pathogenic microbes are indicated in the shrimp is an interface between the external environment and the body
literature [170,171]. Sieved supernatant of B. subtilis UTM 126 has and has an important influence on health and disease. The intestine
shown significant inhibition against three different strains of Vibrio performs multiple functions such as supporting digestion and absorption
harveyi. However, inhibitory activity against V. alginolyticus or of feed, maintaining osmotic balance, regulating endocrine and immu­
V. parahaemolyticus (the isolates from deceased shrimp) was insignifi­ nity. The alimentary microbiota of adult penaeid shrimp (Penaeus chi­
cant. The observed diameter of the zone of inhibition of the nensis) may serve as an additional source of food, vitamins, necessary
above-mentioned supernatant on the culture dish during in vitro was amino acids and enhance microbial activity in the gastrointestinal tract
about 10–15 mm. Overall, B. subtilis UTM 126 resulted in the suppres­ [174]. This microbiota can be favoured by many factors, for example,
sion of pathogenic expansion both in vivo and in vitro [172]. Further­ increased population, density and types of microbes and the complex
more, probiotics are cheap, non-pathogenic and mostly non-toxic interactions between them [37].
sources of antibiotics, having the ability to synthesize a variety of me­ It has become a common practice to alter shrimp’s gut microbiota by
tabolites with antibacterial function, thus are worthwhile for commer­ adding probiotics in feed or culture systems to improve digestion,
cial production. They also have been experimentally used to control growth, and survival. Some microorganisms participate in the digestion
microbial pathogenicity in fish [170,173]. processes by producing extracellular enzymes, such as proteases and
lipases, and can provide necessary growth factors [175]. Similarly,
3.4. Production of inhibitory compounds probiotics can consume carbohydrates from the intestine for self-growth
and produce various digestive enzymes such as amylase, protease,
The production of inhibitory biological compounds such as antibi­ lipase, increase growth rate, and pre-digestibility of secondary com­
otics, antibacterial substances, siderophores, bacteriolytic enzymes, pounds [176]. Ziaei-Nejad et al. [177] investigated Bacillus spp. For its
proteases and protease inhibitor, lactic acid and other organic com­ effects on Indian white shrimp (Fenneropenaeus indicus) at different
pounds like bacteriocins, hydrogen peroxide [181] and butyric acid stages of its life cycle, and noted a significant increase in growth per­
[182] are widely studied and well-documented functioning mechanism formance as compared to control. Additionally, treated ponds
of probiotics [183]. Probiotic microbes possess the potential to inhibit or
even eliminate some pathogenic bacteria. However, in vitro inhibition
may not be observed during in vivo experiments, due to incessantly
changing physicochemical environmental factors [184].
Several Bacillus spp. are known to compete with pathogenic micro­
organisms and produce antibiotic-inhibiting compounds [128,185]. The
production of bacteriocins [186], inhibition of virulence gene expres­
sion [187], and lytic enzymes such as β-1,3-glucanase that inhibit and
lyse the cell wall of the pathogens, protease, chitinase, and cellulose
have been widely observed [188]. Other antibacterial compounds such
as organic acid and hydrogen peroxide may also have an inhibitory ef­
fect due to the residual activity after catalase and acid treatment [189].

3.5. Competition for nutrients and adhesion sites

The potential probiotic bacterium is usually able to colonize and


adhere to the intestinal mucosa as it prevents the adhesion of pathogens Fig. 4. Possible scenarios of organic matter and nutrient production in pond
through inhibition of etiological agents. It also eradicates pathogens water, related bacterial growth and its effects.

274
U.D. Butt et al. Fish and Shellfish Immunology 114 (2021) 263–281

manifested increased activity of amylase, protease, and lipase with survival by reducing the prevalence of WSSV [202]. Synbiotics also
evidently higher digestibility of essential nutrients. promote shrimp growth by increasing the efficiency of active feed intake
Several studies reported the potential mechanism by which pre­ [121]. Improved growth of treated species may be correlated with
biotics alter the intestinal microflora in aquaculture [51,69,112], increased bacterial counts and improved intestinal morphology [203];
including Pacific white shrimp Litopenaeus vannamei [112,114]. Pre­ for instance, synbiotic application promoted growth by altering physi­
biotics regulate local cytokine and antibodies to increase intestinal ological properties of GIT and morphology of epithelium [15].
SCFAs production and improve the binding capability of SCFAs to
G-protein coupled receptors on leukocytes and carbohydrate receptors 3.7. Enzymatic activities
on intestinal epithelial [178]. Supplementation of Previda (a commer­
cially produced prebiotic) modulated intestinal bacterial community It is speculated that the application of probiotics in aquaculture can
and stimulated immunity of shrimp [109]. Prebiotic administration help attain higher enzymatic activities and enhance feed utilization and
further stimulated nutrient absorption and improved homeostasis sta­ digestive capacity by modulating extracellular and antioxidant enzymes
bility [179]. However, improper doses may result in proliferation of (see Table 2) [180]. Glutathione peroxidase (GPx), superoxide dismut­
intestinal microbiota. ase (SOD) and catalase (CAT) are commonly found antioxidant enzymes
Synbiotic administration can enhance, modulate and alter gut in aquatic species [204]. SOD helps decompose harmful oxygen mole­
microbiota (see Table 4). Gastrointestinal bacteria can produce physi­ cules (O−2 ) into H2O2 [205] while CAT catalyzes the dismutation of H2O2
ologically active substances such as enzymes, amino acids, and vitamins into H2O and O2 [206]. Antioxidant enzymes are also known to coun­
through nutrient decomposition, which can provide increased utiliza­ teract the damage caused by reactive oxygen species (ROS) [207] thus
tion and digestion of feed [94]. The probiotic component of synbiotics protect the host against oxidative stress [173]. It is reported that the
can produce molecules such as bacteriocin to prohibit the growth of secretion of digestive enzymes is another characteristic of Bacillus spe­
pathogens on the intestinal wall and enhance the mucosal barrier [14, cies [208]. Usually, Bacillus species can modulate almost all major
180] whereas, the prebiotic component of synbiotics, serves as a sub­ digestive enzymes such as protease, amylase, trypsin, and lipase [9,
strate for probiotics to grow [119]. The presence of sulfate-reducing and 209]. In particular, B. coagulans have been reported to enhance digestive
nitrifying probiotic bacteria in the colon of Litopenaeus vannamei may enzyme activity of freshwater prawn [209]. Similarly, Bacillus PC465
increase resistance against ambient sulfide, ammonia or nitrite [86, improved feed absorption of Litopenaeus vannamei by increasing diges­
121]. Synbiotics can further reduce mortality, regulate intestinal flora tive enzyme activity [22].
and enhance the immune status of shrimp against Vibrio alginolyticus Administration of prebiotics has also been reported to make en­
[82,96,120]. hancements in enzymatic activities (see Table 3). Dietary supplemen­
tation of beta-glucan as prebiotic may result in significant enhancement
3.6. Stimulation of growth and survival of protease and amylase activities, increased nutrients and feed assim­
ilation in the host, alteration in Bacillus and Geobacillus microbial com­
Literature has proven that both dietary supplementation and oral munities and a reduction in lipase levels through hypolipidemic activity
administration of probiotics enhanced the growth of the target species [210]. The combination of β-glucan (prebiotic) and Bacillus (probiotic)
by providing the necessary nutrients and improved feed utilization and can enhance digestive enzymes as Bacillus triggers extracellular enzymes
digestibility by increasing digestive enzymes (see Table 2) [78,193]. in the shrimp’s colon, such as protease and amylase [211]. Commer­
Several strains of Bacillus as probiotics have been tested in shrimp cially available prebiotic immunogen may also increase digestive en­
aquaculture and proved to be effective (see Table 2). Bacillus generally zymes in Pacific white shrimp Litopenaeus vannamei [110]. However,
facilitates nutrient assimilation by using a variety of nutrients for their excessive dosage of prebiotics may result in reduced enzymatic activities
own growth and simultaneously releasing necessary digestive enzymes such as glutamic-pyruvic transaminase (GPT) and glutamic-oxaloacetic
for the host, resulting in higher growth and survival [194]. Litopenaeus transaminase (GOT) [114].
vannamei and Fenneropenaeus indicus administered with Bacillus at the Synbiotics are also known to produce necessary digestive enzymes
rate of 50 g kg− 1 of feed resulted in increased growth [195]. which improve digestion of the host and provide better FCR (see
B. megaterium BM1 at a concentration of 106 cells g− 1 diet of L. vannamei Table 4). Yu et al. [201] observed stimulation in the level of digestive
during in vivo study resulted in better feed utilization and growth [196]. enzymes by synbiotic supplementation. The use of probiotics for syn­
However, the application of microencapsulated and freeze-dried Bacillus biotic preparation, resulted in biosynthesis and secretion of digestive
has shown no significant effect on shrimp larvae but increased the enzymes in shrimp’s gut [212]. Synbiotics stimulate the level of intes­
growth and survival of Post Larvae (PL) remarkably [177]. This uncer­ tinal proteases and leu-aminopeptidase, which play an important role in
tainty may be associated with shrimp’s exposure time to probiotics. protein digestibility [121]. Synbiotic supplementation may also increase
Giant tiger shrimp (Penaeus monodon) supplemented with Bacillus spp. enzymes in plasma such as N-acetyl-β-glucosaminidase, β-galactosidase
DDKRC1, induced exceptional growth, better protein ratio and lower and naphthol-AS-BI-phosphohydrolase, which is mainly due to probiotic
FCR [197]. Similar effects were observed in Litopenaeus vannamei (white strains [202]. However, the effects of prebiotics as a synbiotic compo­
shrimp) [198]. nent requires further investigations to understand their effects on
A wide variety of prebiotics are administered as feed supplements to digestive enzyme activities.
attain better growth activity in treated shrimp [199]. Live feed has also
been reported to promote the growth of shrimp larvae, specifically 4. Conclusion and future outlook
Artemia enriched feed due to its high nutritional value and digestibility
[80]; for instance, bioencapsulation of Artemia species with MOS, Shrimp aquaculture has been dominated by various pathogenic and
improved survival of Litopenaeus vannamei while enhanced growth and viral diseases over the past few decades. The use of antibiotics, to
FCR when supplemented with diet [200]. Prebiotic application may also counter these diseases, backfired as it equally threatened the health of
enhance the survival rate by stimulating several immune parameters the host and the consuming bodies. Besides resisting the pathogens
such as phagocytic, bactericidal and phenoloxidase activities [107,115]. inappropriate antibiotic application led to bacterial resistance in shrimp
Synbiotics are proved to have beneficial effects on growth parame­ and human consumers. Considering the gradual spike in the global de­
ters by increasing fat decomposition [201]. Dietary supplementation of mand and preference for healthy and hygienic shrimp, there is a need for
B. subtilis and Pediococcus acidilactici, along with beta-glucan have natural alternatives such as pro-, pre-, and synbiotics as dietary sup­
improved growth performance [119]. Administration of a mixture of plements to improve competitive exclusion of pathogens from the sys­
bacteria (2LAB + 2Bacillus) and inulin in white shrimp improved its tem and to enhance the immune parameters of shrimp without affecting

275
U.D. Butt et al. Fish and Shellfish Immunology 114 (2021) 263–281

its health. However, to achieve optimal protection, a clear understand­ [12] F.K.A. Kuebutornye, E.D. Abarike, Y. Lu, A review on the application of Bacillus as
probiotics in aquaculture,, Fish Shellfish Immunol. 87 (2019) 820–828, https://
ing of these immunostimulants is necessary. As per the experiments
doi.org/10.1016/j.fsi.2019.02.010.
conducted in recent decades, pro-, pre-, and synbiotics could be [13] A.S. Ninawe, J. Selvin, Probiotics in shrimp aquaculture: avenues and challen,
considered a better alternative, as compared to antibiotics and similar Crit. Rev. Microbiol. 35 (2009) 43–66, https://doi.org/10.1080/
products, for achieving protection while maintaining environmental 10408410802667202.
[14] W. Munaeni, M. Yuhana, W. Widanarni, Effect of micro-encapsulated synbiotic at
stability thereby increasing the shrimp yields. The experiments further different frequencies for luminous Vibriosis control in white shrimp (Litopenaeus
confirmed that the addition of these supplements in shrimp feed can vannamei), Microbiol. Indones 8 (2014) 73–80, https://doi.org/10.5454/
significantly decrease the occurrence of disease and increase enzymatic mi.8.2.5.
[15] A. Oktaviana, Widanarni, M. Yuhana, the use of synbiotics to prevent IMNV and
activities, feed consumption, growth and survival of cultured shrimp. Vibrio harveyi co-infection in Litopenaeus vannamei, HAYATI J, Biosci 21 (2014)
Few studies also raised some concerns regarding the use of these sup­ 127–134, https://doi.org/10.4308/hjb.21.3.127.
plements in inappropriate quantities which can lead to excessive [16] Y. Li, H. Liu, X. Dai, J. Li, F. Ding, Effects of dietary inulin and mannan
oligosaccharide on immune related genes expression and disease resistance of
nutrient production and disturbed microbial balance. In conclusion, a Pacific white shrimp, Litopenaeus vannamei, Fish Shellfish Immunol. 76 (2018)
methodical and comprehensive understanding of the utilities of pro-, 78–92, https://doi.org/10.1016/j.fsi.2018.02.034.
pre-, and synbiotics in the field of shrimp farming can be achieved [17] S.I. Mussatto, I.M. Mancilha, Non-digestible oligosaccharides: a review,
Carbohydr, Polym 68 (2007) 587–597, https://doi.org/10.1016/j.
through profound knowledge of the genetic makeup and transcriptomic carbpol.2006.12.011.
and proteomic profiling of these products. [18] J. Schrezenmeir, M. De Vrese, Probiotics, prebiotics, and synbiotics - approaching
Furthermore, other alternatives, such as paraprobiotics (non-bio­ a definition, Am, J. Clin. Nutr. 73 (2001) 361s–364s, https://doi.org/10.1093/
ajcn/73.2.361s.
logical counterparts of the probiotic organisms), algae and plant extracts
[19] M.A.O. Dawood, N.M. Eweedah, E.M. Moustafa, M.G. Shahin, Synbiotic effects of
with prebiotic properties are highly recommended, as the use of natural Aspergillus oryzae and β-glucan on growth and oxidative and immune responses of
products can yoke comparable advantageous effects and may reduce the Nile tilapia, Oreochromis niloticus, Probiotics Antimicrob, Proteins 12 (2020)
production cost. In summary, the present review reveals the higher 172–183, https://doi.org/10.1007/s12602-018-9513-9.
[20] A. Llewellyn, A. Foey, Probiotic modulation of innate cell pathogen sensing and
effectiveness of pro-, pre-, and synbiotics; however, immediate diagnosis signaling events, Nutrients 9 (2017), https://doi.org/10.3390/nu9101156.
of the molecular pathways regulating the mechanisms of the pro-, pre-, [21] H. Van Doan, S.H. Hoseinifar, W. Tapingkae, M. Seel-audom, S. Jaturasitha, M.A.
and synbiotics in shrimp metabolism is obligatory. O. Dawood, S. Wongmaneeprateep, T.T.N. Thu, M.Á. Esteban, Boosted growth
performance, mucosal and serum immunity, and disease resistance Nile tilapia
(Oreochromis niloticus) fingerlings using corncob-derived xylooligosaccharide and
Lactobacillus plantarum CR1T5, Probiotics Antimicrob. Proteins 12 (2020)
Declaration of competing interest 400–411, https://doi.org/10.1007/s12602-019-09554-5.
[22] P.C. Chai, X.L. Song, G.F. Chen, H. Xu, J. Huang, Dietary supplementation of
The authors declare that they have no competing financial interests probiotic Bacillus PC465 isolated from the gut of Fenneropenaeus chinensis
improves the health status and resistance of Litopenaeus vannamei against white
or personal relationships that seem to affect the work done in this paper.
spot syndrome virus, Fish Shellfish Immunol. 54 (2016) 602–611, https://doi.
org/10.1016/j.fsi.2016.05.011.
Acknowledgements [23] H. Chen, J. Ullah, J. Jia, Progress in Bacillus subtilis spore surface display
technology towards environment, vaccine development, and biocatalysis, J. Mol.
Microbiol. Biotechnol. 27 (2017) 159–167, https://doi.org/10.1159/000475177.
This work was supported by the science fundation of Lishui Science [24] K. Janarthanam, M.R. George, K.R. John, M.J.P. Jeyaseelan, In vitro and in vivo
and Technology Bureau (2017ZDYF17) and the National Key Research biocontrol of Vibrio harveyi using indigenous bacterium, Bacillus SPP, Indian J.
Geo-Marine Sci. 41 (2012) 83–89.
and Development (R&D) Program of China (No. 2017YFE0103102).
[25] I.E. Luis-Villaseñor, M.E. Macías-Rodríguez, B. Gómez-Gil, F. Ascencio-Valle, Á.I.
Campa-Córdova, Beneficial effects of four Bacillus strains on the larval cultivation
References of Litopenaeus vannamei, Aquaculture 321 (2011) 136–144, https://doi.org/
10.1016/j.aquaculture.2011.08.036.
[26] S. Nimrat, S. Suksawat, T. Boonthai, V. Vuthiphandchai, Potential Bacillus
[1] T. Nakajima, M.J. Hudson, J. Uchiyama, K. Makibayashi, J. Zhang, Common carp
probiotics enhance bacterial numbers, water quality and growth during early
aquaculture in Neolithic China dates back 8,000 years, Nat, Ecol. Evol. 3 (2019)
development of white shrimp (Litopenaeus vannamei), Vet. Microbiol. 159 (2012)
1415–1418, https://doi.org/10.1038/s41559-019-0974-3.
443–450, https://doi.org/10.1016/j.vetmic.2012.04.029.
[2] FAO, FAO Fisheries & Aquaculture, The State of World Fisheries and Aquaculture
[27] J. Olmos, L. Ochoa, J. Paniagua-Michel, R. Contreras, Functional feed assessment
(SOFIA), 2002, 1, http://www.fao.org/fishery/sofia/en.
on Litopenaeus vannamei using 100% fish meal replacement by soybean meal, high
[3] Fao, The State of World Fisheries and Aquaculture 2020, In brief. Sustainability in
levels of complex carbohydrates and Bacillus probiotic strains, Mar. Drugs 9
action., FAO, Rome, 2020, https://doi.org/10.4060/ca9231en.
(2011) 1119–1132, https://doi.org/10.3390/md9061119.
[4] H. Rubel, W. Woods, D. Pérez, A. Meyer, Z. Felde, S. Zielcke, C. Lidy, A strategic
[28] E.F. Silva, M.A. Soares, N.F. Calazans, J.L. Vogeley, B.C. do Valle, R. Soares,
approach to sustainable shrimp production in Thailand the case for improved
S. Peixoto, Effect of probiotic (Bacillus spp.) addition during larvae and postlarvae
economics and sustainability Carolin Lanfer 2, A strategic approach to sustainable
culture of the white shrimp Litopenaeus vannamei, Aquacult. Res. 44 (2012)
shrimp production in Thailand (2019) 2–56. https://media-publications.bcg.co
13–21, https://doi.org/10.1111/j.1365-2109.2011.03001.x.
m/BCG-A-Strategic-Approach-to-Sustainable-Shrimp-Production-Thailand-July
[29] S.V. Alavandi, K.K. Vijayan, T.C. Santiago, M. Poornima, K.P. Jithendran, S.A. Ali,
-2019.pdf.
J.J.S. Rajan, Evaluation of Pseudomonas sp. PM 11 and Vibrio fluvialis PM 17 on
[5] FAO, Globefish highlights, Issue, with Annual 2019 Statistics – A Quarterly
immune indices of tiger shrimp, Penaeus monodon, Fish Shellfish Immunol. 17
Update on World Seafood Markets. Globefish Highlights No. 2–2020, vol. 2020,
(2004) 115–120, https://doi.org/10.1016/j.fsi.2003.11.007.
FAO, Rome, April 2020, https://doi.org/10.4060/ca9528en.
[30] N.A. El-Sersy, F.A. AbdelRazek, S.M. Taha, Evaluation of various probiotic
[6] C.-Y. Chen, P.-C. Chen, F.C.-H. Weng, G.T.-W. Shaw, D. Wang, Habitat and
bacteria for the survival of Penaeus japonicus larvae, Fresenius Environ. Bull. 15
indigenous gut microbes contribute to the plasticity of gut microbiome in oriental
(2006) 1506–1511.
river prawn during rapid environmental change, PloS One 12 (2017), https://doi.
[31] J. Thompson, S. Gregory, S. Plummer, R.J. Shields, A.F. Rowley, An in vitro and
org/10.1371/journal.pone.0181427 e0181427.
in vivo assessment of the potential of Vibrio spp. as probiotics for the Pacific white
[7] C.L. Ayisi, A. Apraku, G. Afriyie, A Review of probiotics, prebiotics, and
shrimp, Litopenaeus vannamei, J. Appl. Microbiol. 109 (2010) 1177–1187, https://
synbiotics in crab: present research, problems, and future perspective,,
doi.org/10.1111/j.1365-2672.2010.04743.x.
J. Shellfish Res. 36 (2017) 799–806, https://doi.org/10.2983/035.036.0329.
[32] Y. Wang, Q. Gu, Effect of probiotics on white shrimp (Penaeus vannamei) growth
[8] H. Cordero, Use of probiotic bacteria against bacterial and viral infections in
performance and immune response, Mar. Biol. Res. 6 (2010) 327–332, https://
shellfish and fish aquaculture, in: M. ÁEsteban (Ed.), IntechOpen, Rijeka vol. 8,
doi.org/10.1080/17451000903300893.
2014, https://doi.org/10.5772/57198. Ch.
[33] S. Das, L.R. Ward, C. Burke, Screening of marine Streptomyces spp. for potential
[9] M.A.O. Dawood, S. Koshio, M. Ishikawa, M. El-Sabagh, M.A. Esteban, A.
use as probiotics in aquaculture, Aquaculture 305 (2010) 32–41, https://doi.org/
I. Zaineldin, Probiotics as an environment-friendly approach to enhance red sea
10.1016/j.aquaculture.2010.04.001.
bream, Pagrus major growth, immune response and oxidative status, Fish Shellfish
[34] S.S. Pai, A. Anas, N.S. Jayaprakash, P. Priyaja, B. Sreelakshmi, R. Preetha,
Immunol. 57 (2016) 170–178, https://doi.org/10.1016/j.fsi.2016.08.038.
R. Philip, A. Mohandas, I.S.B. Singh, Penaeus monodon larvae can be protected
[10] S.H. Hoseinifar, Y.Z. Sun, A. Wang, Z. Zhou, Probiotics as means of diseases
from Vibrio harveyi infection by pre-emptive treatment of a rearing system with
control in aquaculture, a review of current knowledge and future perspectives,
antagonistic or non-antagonistic bacterial probiotics, Aquacult. Res. 41 (2009)
Front. Microbiol. 9 (2018), https://doi.org/10.3389/fmicb.2018.02429.
847–860, https://doi.org/10.1111/j.1365-2109.2009.02362.x.
[11] S.H. Hoseinifar, H. Van Doan, M. Dadar, E. Ringø, R. Harikrishnan, Feed
[35] S.M. Swain, C. Singh, V. Arul, Inhibitory activity of probiotics Streptococcus
additives, gut microbiota, and health in finfish aquaculture, Microb. Communities
phocae PI80 and Enterococcus faecium MC13 against Vibriosis in shrimp Penaeus
Aquac, Ecosyst (2019) 121–142, https://doi.org/10.1007/978-3-030-16190-3_6.

276
U.D. Butt et al. Fish and Shellfish Immunology 114 (2021) 263–281

monodon, World J. Microbiol. Biotechnol. 25 (2009) 697–703, https://doi.org/ [60] S.H. Hoseinifar, A. Ahmadi, M. Raeisi, S.M. Hoseini, M. Khalili, N. Behnampour,
10.1007/s11274-008-9939-4. Comparative study on immunomodulatory and growth enhancing effects of three
[36] M.A.O. Dawood, S. Koshio, M. Ishikawa, M. El-Sabagh, S. Yokoyama, W.L. Wang, prebiotics (galactooligosaccharide, fructooligosaccharide and inulin) in common
Z. Yukun, A. Olivier, Physiological response, blood chemistry profile and mucus carp (Cyprinus carpio), Aquacult. Res. 48 (2017) 3298–3307, https://doi.org/
secretion of red sea bream (Pagrus major) fed diets supplemented with 10.1111/are.13156.
Lactobacillus rhamnosus under low salinity stress, Fish Physiol, Biochem 43 (2017) [61] Y. Chen, X. Zhu, Y. Yang, D. Han, J. Jin, S. Xie, Effect of dietary chitosan on
179–192, https://doi.org/10.1007/s10695-016-0277-4. growth performance, haematology, immune response, intestine morphology,
[37] P. Martínez Cruz, A.L. Ibáñez, O.A. Monroy Hermosillo, H.C. Ramírez Saad, Use intestine microbiota and disease resistance in gibel carp (Carassius auratus gibelio),
of probiotics in aquaculture, ISRN Microbiol. (2012) 1–13, https://doi.org/ Aquacult. Nutr. 20 (2014) 532–546, https://doi.org/10.1111/anu.12106.
10.5402/2012/916845, 2012. [62] H. Kühlwein, D.L. Merrifield, M.D. Rawling, A.D. Foey, S.J. Davies, Effects of
[38] M.S. Llewellyn, S. Boutin, S.H. Hoseinifar, N. Derome, Teleost microbiomes: the dietary β-(1,3)(1,6)-D-glucan supplementation on growth performance, intestinal
state of the art in their characterization, manipulation and importance in morphology and haemato-immunological profile of mirror carp (Cyprinus carpio
aquaculture and fisheries, Front, Microbiol. 5 (2014), https://doi.org/10.3389/ L.), J. Anim. Physiol. Anim. Nutr. 98 (2014) 279–289, https://doi.org/10.1111/
fmicb.2014.00207, 1–1. jpn.12078.
[39] C.E. Boyd, L. Massaut, Risks associated with the use of chemicals in pond [63] G. Ebrahimi, H. Ouraji, M.K. Khalesi, M. Sudagar, A. Barari, M. Zarei
aquaculture, Aquacult. Eng. 20 (1999) 113–132, https://doi.org/10.1016/S0144- Dangesaraki, K.H. Jani Khalili, Effects of a prebiotic, Immunogen ®, on feed
8609(99)00010-2. utilization, body composition, immunity and resistance to Aeromonas hydrophila
[40] L. Verschuere, G. Rombaut, P. Sorgeloos, W. Verstraete, Probiotic bacteria as infection in the common carp Cyprinus carpio (Linnaeus) fingerlings, J. Anim.
biological control agents in aquaculture, Microbiol. Mol. Biol. Rev. 64 (2000) Physiol. Anim. Nutr. 96 (2012) 591–599, https://doi.org/10.1111/j.1439-
655–671, https://doi.org/10.1128/mmbr.64.4.655-671.2000. 0396.2011.01182.x.
[41] H. Van Doan, S.H. Hoseinifar, M.A.O. Dawood, C. Chitmanat, K. Tayyamath, [64] R.Z. Reza Akrami, Hossein Chitsaz, Ali Hezarjaribi, Effect of dietary mannan
Effects of Cordyceps militaris spent mushroom substrate and Lactobacillus oligosaccharide (MOS) on growth performance and immune response of gibel
plantarum on mucosal, serum immunology and growth performance of Nile tilapia carp juveniles (Carassius auratus gibelio), J. Vet. Adv. 2 (2012) 507–513. htt
(Oreochromis niloticus), Fish Shellfish Immunol. 70 (2017) 87–94, https://doi.org/ p://www.scopemed.org/?mno=26971.
10.1016/j.fsi.2017.09.002. [65] H. Eshaghzadeh, S.H. Hoseinifar, H. Vahabzadeh, E. Ringø, The effects of dietary
[42] I.E. Luis-Villaseñor, T. Castellanos-Cervantes, B. Gomez-Gil, Á.E. Carrillo-García, inulin on growth performances, survival and digestive enzyme activities of
Á.I. Campa-Córdova, F. Ascencio, Probiotics in the intestinal tract of juvenile common carp (Cyprinus carpio) fry, Aquac, Nutrition 21 (2015) 242–247, https://
whiteleg shrimp Litopenaeus vannamei: modulation of the bacterial community, doi.org/10.1111/anu.12155.
World J. Microbiol. Biotechnol. 29 (2013) 257–265, https://doi.org/10.1007/ [66] R. Dobšíková, J. Blahová, I. Mikulíková, H. Modrá, E. Prášková, Z. Svobodová,
s11274-012-1177-0. M. Škorič, J. Jarkovský, A.-K. Siwicki, The effect of oyster mushroom β-1.3/1.6-D-
[43] G.R. Gibson, M.B. Roberfroid, Dietary modulation of the human colonic glucan and oxytetracycline antibiotic on biometrical, haematological,
microbiota: introducing the concept of prebiotics, J. Nutr. 125 (1995) biochemical, and immunological indices, and histopathological changes in
1401–1412, https://doi.org/10.1093/jn/125.6.1401. common carp (Cyprinus carpio L.),, Fish Shellfish Immunol. 35 (2013) 1813–1823,
[44] L.B. Bindels, N.M. Delzenne, P.D. Cani, J. Walter, Towards a more comprehensive https://doi.org/10.1016/j.fsi.2013.09.006.
concept for prebiotics, Nat. Rev. Gastroenterol. Hepatol. 12 (2015) 303–310, [67] S.K. Song, B.R. Beck, D. Kim, J. Park, J. Kim, H.D. Kim, E. Ringø, Prebiotics as
https://doi.org/10.1038/nrgastro.2015.47. immunostimulants in aquaculture: a review,, Fish Shellfish Immunol. 40 (2014)
[45] I. Guerreiro, A. Oliva-Teles, P. Enes, Prebiotics as functional ingredients: focus on 40–48, https://doi.org/10.1016/j.fsi.2014.06.016.
Mediterranean fish aquaculture, Rev. Aquacult. 10 (2018) 800–832, https://doi. [68] G.D. Gómez, J.L. Balcázar, A review on the interactions between gut microbiota
org/10.1111/raq.12201. and innate immunity of fish: Table 1, FEMS Immunol. Med, Microbiol. 52 (2008)
[46] F.C. Cabello, Antibióticos y acuicultura en Chile: consecuencias para la salud 145–154, https://doi.org/10.1111/j.1574-695X.2007.00343.x.
humana y animal, Rev. Med. Chile 132 (2004) 1001–1006, https://doi.org/ [69] M. De Vrese, J. Schrezenmeir, Probiotics, Prebiotics, and Synbiotics, in: Adv.
10.4067/S0034-98872004000800014. Biochem. Eng. Biotechnol, 111th ed., Springer, Berlin, Heidelberg, 2008,
[47] A. Sapkota, A.R. Sapkota, M. Kucharski, J. Burke, S. McKenzie, P. Walker, pp. 1–66, https://doi.org/10.1007/10_2008_097.
R. Lawrence, Aquaculture practices and potential human health risks: current [70] S. Kolida, G.R. Gibson, Synbiotics in health and disease, Annu. Rev. Food Sci.
knowledge and future priorities, Environ. Int. 34 (2008) 1215–1226, https://doi. Technol 2 (2011) 373–393, https://doi.org/10.1146/annurev-food-022510-
org/10.1016/j.envint.2008.04.009. 133739.
[48] V.J. Smith, J.H. Brown, C. Hauton, Immunostimulation in crustaceans: does it [71] Sweta V. Chauhan, Mehul R. Chorawala, Probiotics, prebiotics and synbiotics, Int.
really protect against infection? Fish Shellfish Immunol. 15 (2003) 71–90, J. Pharm. Sci. Res. 3 (2012) 711–726, https://doi.org/10.13040/IJPSR.0975-
https://doi.org/10.1016/S1050-4648(02)00140-7. 8232.3(3).711-26.
[49] H. Sørum, Antimicrobial drug resistance in fish pathogens, in: antimicrob. Resist. [72] K. Mondal, S. Haque, S. Das, K. Mondal Associate Professor, C. Kausik Mondal
Bact. Anim. Orig, American Society of Microbiology (2006) 213–238, https://doi. Associate Professor, A review on application of probiotic, prebiotic and synbiotic
org/10.1128/9781555817534.ch13. for sustainable development of aquaculture,, J. Entomol. Zool. Stud 5 (2017)
[50] H.L. Lauzon, A. Dimitroglou, D.L. Merrifield, E. Ringø, S.J. Davies, Probiotics and 422–429. https://www.researchgate.net/publication/335222699.
prebiotics: concepts, definitions and history, in: Aquac. Nutr, John Wiley & Sons, [73] R. Akrami, M. Nasri-Tajan, A. Jahedi, M. Jahedi, M. Razeghi Mansour, S.
Ltd, Chichester, UK, 2014, pp. 169–184, https://doi.org/10.1002/ A. Jafarpour, Effects of dietary synbiotic on growth, survival, Lactobacillus
9781118897263.ch7. bacterial count, blood indices and immunity of beluga (Huso huso Linnaeus, 1754)
[51] N. Akhter, B. Wu, A.M. Memon, M. Mohsin, Probiotics and prebiotics associated juvenile, Aquacult. Nutr. 21 (2015) 952–959, https://doi.org/10.1111/
with aquaculture: a review,, Fish Shellfish Immunol. 45 (2015) 733–741, https:// anu.12219.
doi.org/10.1016/j.fsi.2015.05.038. [74] S. Lin, S. Mao, Y. Guan, L. Luo, L. Luo, Y. Pan, Effects of dietary chitosan
[52] B. Ali, A. El-Feky, Enhancing growth performance and feed utilization using oligosaccharides and Bacillus coagulans on the growth, innate immunity and
prebiotics in commercial diets of Nile tilapia (Oreochromis niloticus) fingerlings, resistance of koi (Cyprinus carpio koi), Aquaculture. 342–343 (2012) 36–41,
Egypt. J. Nutr. Feed 22 (2019) 219–225, https://doi.org/10.21608/ https://doi.org/10.1016/j.aquaculture.2012.02.009.
ejnf.2019.76355. [75] C.-N. Zhang, X.-F. Li, W.-N. Xu, D.-D. Zhang, K.-L. Lu, L.-N. Wang, H.-Y. Tian, W.-
[53] A. Nawaz, A. Bakhsh javaid, S. Irshad, S.H. Hoseinifar, H. Xiong, The B. Liu, Combined effects of dietary fructooligosaccharide and Bacillus licheniformis
functionality of prebiotics as immunostimulant: evidences from trials on on growth performance, body composition, intestinal enzymes activities and gut
terrestrial and aquatic animals,, Fish Shellfish Immunol. 76 (2018) 272–278, histology of triangular bream (Megalobrama terminalis), Aquacult. Nutr. 21 (2015)
https://doi.org/10.1016/j.fsi.2018.03.004. 755–766, https://doi.org/10.1111/anu.12200.
[54] S.H. Hoseinifar, M.Á. Esteban, A. Cuesta, Y.-Z. Sun, Prebiotics and fish immune [76] G. Mazzola, I. Aloisio, B. Biavati, D. Di Gioia, Development of a synbiotic product
response: a review of current knowledge and future perspectives,, Rev. Fish. Sci. for newborns and infants, LWT - Food Sci. Technol. (Lebensmittel-Wissenschaft
Aquac 23 (2015) 315–328, https://doi.org/10.1080/23308249.2015.1052365. -Technol.) 64 (2015) 727–734, https://doi.org/10.1016/j.lwt.2015.06.033.
[55] A. Dimitroglou, S. Davies, J. Sweetman, The effect of dietary mannan [77] J. Grimoud, H. Durand, C. Courtin, P. Monsan, F. Ouarné, V. Theodorou,
oligosaccharides on the intestinal histology of rainbow trout (Oncorhynchus C. Roques, In vitro screening of probiotic lactic acid bacteria and prebiotic
mykiss), Comp. Biochem. Physiol, Part A Mol. Integr. Physiol 150 (2008) S63, glucooligosaccharides to select effective synbiotics, Anaerobe 16 (2010)
https://doi.org/10.1016/j.cbpa.2008.04.069. 493–500, https://doi.org/10.1016/j.anaerobe.2010.07.005.
[56] J.S. Sweetman, A. Dimitroglou, S. Davies, Torrecillas, Nutrient uptake: gut [78] D.L. Merrifield, A. Dimitroglou, A. Foey, S.J. Davies, R.T.M. Baker, J. Bøgwald,
morphology a key to efficient nutrition, Int. Aquafeed 26 (2008) 26–30. M. Castex, E. Ringø, The current status and future focus of probiotic and prebiotic
[57] E. Yilmaz, M.A. Genc, E. Genc, Effects of dietary mannan oligosaccharides on applications for salmonids,, Aquaculture 302 (2010) 1–18, https://doi.org/
growth, body composition, and intestine and liver histology of rainbow trout, 10.1016/j.aquaculture.2010.02.007.
Oncorhynchus mykiss, Isr. J. Aquac. Bamidgeh 59 (2007) 182–188. [79] B.S. Sekhon, S. Jairath, Prebiotics, probiotics and synbiotics: an overview,
[58] E. Ringø, R.E. Olsen, T.Ø. Gifstad, R.A. Dalmo, H. Amlund, G.-I. Hemre, A. J. Pharm. Educ. Res. 1 (2010) 13.
M. Bakke, Prebiotics in aquaculture: a review, Aquacult. Nutr. 16 (2010) [80] H. Hamsah, W. Widanarni, A. Alimuddin, M. Yuhana, M.Z. Junior,
117–136, https://doi.org/10.1111/j.1365-2095.2009.00731.x. D. Hidayatullah, Immune response and resistance of Pacific white shrimp larvae
[59] G. Burr, D. Gatlin, S. Ricke, Microbial ecology of the gastrointestinal tract of fish administered probiotic, prebiotic, and synbiotic through the bio-encapsulation of
and the potential application of prebiotics and probiotics in finfish aquaculture, Artemia sp, Aquacult. Int. 27 (2019) 567–580, https://doi.org/10.1007/s10499-
J. World Aquacult. Soc. 36 (2005) 425–436, https://doi.org/10.1111/j.1749- 019-00346-w.
7345.2005.tb00390.x.

277
U.D. Butt et al. Fish and Shellfish Immunology 114 (2021) 263–281

[81] D.E. Ramadhani, W. Widanarni, S. Sukenda, Microencapsulation of probiotics and [103] I.I. Arisa, W. Widanarni, M. Yuhana, Z.A. Muchlisin, A.A. Abdullah, The
its applications with prebiotic in Pacific white shrimp larvae through Artemia sp, application of probiotics, prebiotics and synbiotics to enhance the immune
J. Akuakultur Indones 18 (2019) 130–140, https://doi.org/10.19027/ responses of vannamei shrimp (Litopenaeus vannamei) to Vibrio harveyi infection,,
jai.18.2.130-140. AACL Bioflux 8 (2015) 772–778.
[82] Q. Zhang, B. Tan, K. Mai, W. Zhang, H. Ma, Q. Ai, X. Wang, Z. Liufu, Dietary [104] W. Widanarni, A. Taufik, M. Yuhana, J. Ekasari, Dietary mannan oligosaccharides
administration of Bacillus (B. licheniformis and B. subtilis) and positively affect the growth, digestive enzyme activity, immunity and resistance
isomaltooligosaccharide influences the intestinal microflora, immunological against Vibrio harveyi of Pacific white shrimp (Litopenaeus vannamei) larvae, Turk.
parameters and resistance against Vibrio alginolyticus in shrimp, Penaeus J. Fish. Aquat. Sci. 19 (2018) 271–278.
japonicus (Decapoda: Penaeidae), Aquac. Res. 42 (2011) 943–952, https://doi. [105] M.T.D. Tei, S. Aslamyah, S. Sriwulan, The utilization of sweet potatoes as
org/10.1111/j.1365-2109.2010.02677.x. prebiotics on the performance of Lactobacillus sp. in the vanamei shrimp digestion
[83] D. Febrianti, M. Yuhana, Dietary Synbiotic microcapsule influence the immune (Litopenaeus vannamei), Int, J. Environ. Agric. Biotechnol 4 (2019) 1087–1091,
responses, growth performance and microbial populations to white spot https://doi.org/10.22161/ijeab.4429.
syndrome virus in Pacific white shrimp (Litopenaeus vannamei), J. Fish. Aquat. Sci. [106] M. Yazici, Y. Mazlum, Prebiotic applications in cultured crayfish and shrimps.,
11 (2016) 28–42, https://doi.org/10.3923/jfas.2016.28.42. KSU tarım ve doğa dergisi-KSU J, Agric. Nat 22 (2019) 153–163, https://doi.org/
[84] T.-G. Huynh, Y.-L. Shiu, T.-P. Nguyen, Q.-P. Truong, J.-C. Chen, C.-H. Liu, Current 10.18016/ksutarimdoga.vi.471559.
applications, selection, and possible mechanisms of actions of synbiotics in [107] G.E. Elshopakey, E.F. Risha, O.A. Abdalla, Y. Okamura, V.D. Hanh, M. Ibuki,
improving the growth and health status in aquaculture: a review,, Fish Shellfish R. Sudhakaran, T. Itami, Enhancement of immune response and resistance against
Immunol. 64 (2017) 367–382, https://doi.org/10.1016/j.fsi.2017.03.035. Vibrio parahaemolyticus in kuruma shrimp (Marsupenaeus japonicus) by dietary
[85] T.-G. Huynh, C.-C. Chi, T.-P. Nguyen, T.-T.-T.H. Tran, A.-C. Cheng, C.-H. Liu, supplementation of β-1,4-mannobiose, Fish Shellfish Immunol. 74 (2018) 26–34,
Effects of synbiotic containing Lactobacillus plantarum 7-40 and https://doi.org/10.1016/j.fsi.2017.12.036.
galactooligosaccharide on the growth performance of white shrimp, Litopenaeus [108] Y. Sun, G. Wang, K. Peng, Y. Huang, J. Cao, W. Huang, B. Chen, J. Hu, Effects of
vannamei, Aquacult. Res. 49 (2018) 2416–2428, https://doi.org/10.1111/ dietary xylooligosaccharides on growth performance, immunity and Vibrio
are.13701. alginolyticus resistance of juvenile Litopenaeus vannamei, Aquacult. Res. 50 (2019)
[86] T.-G. Huynh, S.-Y. Hu, C.-S. Chiu, Q.-P. Truong, C.-H. Liu, Bacterial population in 358–365, https://doi.org/10.1111/are.13911.
intestines of white shrimp, Litopenaeus vannamei fed a synbiotic containing [109] J.D. Anuta, A. Buentello, S. Patnaik, M.E. Hume, A. Mustafa, D.M. Gatlin, A.
Lactobacillus plantarum and galactooligosaccharide, Aquacult. Res. 50 (2019) L. Lawrence, Effects of dietary supplementation of a commercial prebiotic Previda
807–817, https://doi.org/10.1111/are.13951. ® on survival, growth, immune responses and gut microbiota of Pacific white
[87] E. Li, C. Xu, X. Wang, S. Wang, Q. Zhao, M. Zhang, J.G. Qin, L. Chen, Gut shrimp, Litopenaeus vannamei, Aquacult. Nutr. 22 (2016) 410–418, https://doi.
microbiota and its modulation for healthy farming of Pacific white shrimp org/10.1111/anu.12257.
Litopenaeus vannamei, Rev. Fish. Sci. Aquac 26 (2018) 381–399, https://doi.org/ [110] H.K. Miandare, A.T. Mirghaed, M. Hosseini, N. Mazloumi, A. Zargar, S. Nazari,
10.1080/23308249.2018.1440530. Dietary Immunogen® modulated digestive enzyme activity and immune gene
[88] P. V Nageswara, D.E. Babu, Probiotics as an alternative therapy to minimize or expression in Litopenaeus vannamei post larvae, Fish Shellfish Immunol. 70 (2017)
avoid antibiotics use in aquaculture, Fish. Chimes 26 (2006) 112–114. 621–627, https://doi.org/10.1016/j.fsi.2017.09.048.
[89] G. Breves, Effects of oligosaccharides on functional parameters of the intestinal [111] P. Li, G.S. Burr, D.M. Gatlin, M.E. Hume, S. Patnaik, F.L. Castille, A.L. Lawrence,
tract of growing pigs, Dtsch. Tierarztl. Wochenschr 108 (2001) 246–248. Dietary supplementation of short-chain fructooligosaccharides influences
[90] A. Bongers, E.G.H.M. van den Heuvel, Prebiotics and the bioavailability of gastrointestinal microbiota composition and immunity characteristics of Pacific
minerals and trace elements, Food Rev. Int. 19 (2003) 397–422, https://doi.org/ white shrimp, Litopenaeus vannamei, cultured in a recirculating system, J. Nutr.
10.1081/FRI-120025482. 137 (2007) 2763–2768, https://doi.org/10.1093/jn/137.12.2763.
[91] H.H. Sung, G.H. Kou, Y.L. Song, Vibriosis resistance induced by glucan treatment [112] Z. Zhou, Z. Ding, L.V. Huiyuan, Effects of dietary short-chain
in tiger shrimp (Penaeus monodon), Fish Pathol. 29 (1994) 11–17, https://doi.org/ fructooligosaccharides on intestinal microflora, survival, and growth performance
10.3147/jsfp.29.11. of juvenile white shrimp, Litopenaeus vannamei, J. World Aquacult. Soc. 38 (2007)
[92] S.H. Hoseinifar, P. Zare, D.L. Merrifield, The effects of inulin on growth factors 296–301, https://doi.org/10.1111/j.1749-7345.2007.00099.x.
and survival of the Indian white shrimp larvae and postlarvae (Fenneropenaeus [113] N. Bai, W. Zhang, K. Mai, X. Wang, W. Xu, H. Ma, Effects of discontinuous
indicus),, Aquacult. Res. 41 (2010) e348–e352, https://doi.org/10.1111/j.1365- administration of β-glucan and glycyrrhizin on the growth and immunity of white
2109.2010.02485.x. shrimp Litopenaeus vannamei, Aquaculture 306 (2010) 218–224, https://doi.org/
[93] Z. Geraylou, C. Souffreau, E. Rurangwa, L. De Meester, C.M. Courtin, J. 10.1016/j.aquaculture.2010.06.017.
A. Delcour, J. Buyse, F. Ollevier, Effects of dietary arabinoxylan-oligosaccharides [114] X. Hu, H.L. Yang, Y.Y. Yan, C.X. Zhang, J. dan Ye, K. Le Lu, L.H. Hu, J.J. Zhang,
(AXOS) and endogenous probiotics on the growth performance, non-specific L. Ruan, Y.Z. Sun, Effects of fructooligosaccharide on growth, immunity and
immunity and gut microbiota of juvenile Siberian sturgeon (Acipenser baerii), Fish intestinal microbiota of shrimp (Litopenaeus vannamei) fed diets with fish meal
Shellfish Immunol. 35 (2013) 766–775, https://doi.org/10.1016/j. partially replaced by soybean meal, Aquacult. Nutr. 25 (2019) 194–204, https://
fsi.2013.06.014. doi.org/10.1111/anu.12843.
[94] Q. Ai, H. Xu, K. Mai, W. Xu, J. Wang, W. Zhang, Effects of dietary [115] H. Li, C. Xu, L. Zhou, Y. Dong, Y. Su, X. Wang, J.G. Qin, L. Chen, E. Li, Beneficial
supplementation of Bacillus subtilis and fructooligosaccharide on growth effects of dietary β-glucan on growth and health status of Pacific white shrimp
performance, survival, non-specific immune response and disease resistance of Litopenaeus vannamei at low salinity, Fish Shellfish Immunol. 91 (2019) 315–324,
juvenile large yellow croaker, Larimichthys crocea, Aquaculture 317 (2011) https://doi.org/10.1016/j.fsi.2019.05.052.
155–161, https://doi.org/10.1016/j.aquaculture.2011.04.036. [116] Y. Mustafa, Aplikasi Prebiotik Berbeda pada Pakan Terhadap Kinerja Bakteri
[95] A.K. Anal, H. Singh, Recent advances in microencapsulation of probiotics for Lactobacillus Sp. dalam Saluran Pencernaan Udang Vaname (Litopenaeus
industrial applications and targeted delivery, Trends Food Sci. Technol. 18 (2007) Vannamei), Tesis, Program Pascasarjana Universitas Hasanuddin (2017).
240–251, https://doi.org/10.1016/j.tifs.2007.01.004. [117] R. Suri, P. Berta, S. Oktora, Studi tentang penggunaan pakan komersil yang
[96] A. Zubaidah, M. Yuhana, Widanarni, Encapsulated synbiotic dietary dicampur dengan bakteri Bacillus coagulans terhadap performa Litopenaeus
supplementation at different dosages to prevent Vibriosis in white shrimp, vannamei, e-Jurnal Rekayasa dan Teknologi Budidaya Perairan 7 (1) (2018)
Litopenaeus vannamei, HAYATI J, Biosci 22 (2015) 163–168, https://doi.org/ 751–762, https://doi.org/10.23960/jrtbp.v7i1.p751-762.
10.1016/j.hjb.2015.10.007. [118] S.J. Rodriguez-Estrada U, S. Satoh, Y. Haga, H. Fushimi, Effects of single and
[97] W. Purivirojkul, S. Khidprasert, Diseases and parasites on cultured fairy shrimps, combined supplementation of Enterococcus faecalis, mannan oligosaccharide and
Branchinella thailandensis and Streptocephaluss irindhornae, in: Proceeding polyhydroxybutyrate acid on growth performance and immune response of
Asian Pacific Aquac, 2009, p. 444. Kuala Lumpur, Malaysia, 2009. rainbow trout Oncorhynchus mykiss, Aquac. Sci. 57 (2009) 609–617, https://doi.
[98] J.C. Vergel, L.D. Cabawatan, V.A. Madrona, A.F. Rosario, J.B. Sta, Ana, M.V. Tare, org/10.11233/aquaculturesci.57.609.
M.B. Maningas, detection of taura syndrome virus (TSV) in Litopenaeus vannamei [119] S. Boonanuntanasarn, U. Wongsasak, T. Pitaksong, S. Chaijamrus, Effects of
in the Philippines, Philipp. J. Fish 26 (2019) 8–14, https://doi.org/10.31398/ dietary supplementation with β-glucan and synbiotics on growth, haemolymph
tpjf/25.2.2018-0003. chemistry, and intestinal microbiota and morphology in the Pacific white shrimp,
[99] K.F.J. Tang, L.F. Aranguren, P. Piamsomboon, J.E. Han, I.Y. Maskaykina, M. Aquacult. Nutr. 22 (2016) 837–845, https://doi.org/10.1111/anu.12302.
M. Schmidt, Detection of the microsporidian Enterocytozoon hepatopenaei (EHP) [120] J. Li, B. Tan, K. Mai, Dietary probiotic Bacillus OJ and isomaltooligosaccharides
and Taura syndrome virus in Penaeus vannamei cultured in Venezuela, influence the intestine microbial populations, immune responses and resistance to
Aquaculture 480 (2017) 17–21, https://doi.org/10.1016/j. white spot syndrome virus in shrimp (Litopenaeus vannamei), Aquaculture 291
aquaculture.2017.07.043. (2009) 35–40, https://doi.org/10.1016/j.aquaculture.2009.03.005.
[100] Q. Ren, X. Huang, Y. Cui, J. Sun, W. Wang, X. Zhang, Two white spot syndrome [121] T.-G. Huynh, A.-C. Cheng, C.-C. Chi, K.-H. Chiu, C.-H. Liu, A synbiotic improves
virus microRNAs target the dorsal gene to promote virus infection in the immunity of white shrimp, Litopenaeus vannamei: metabolomic analysis reveal
Marsupenaeus japonicus Shrimp, J. Virol. 91 (2017), https://doi.org/10.1128/ compelling evidence,, Fish Shellfish Immunol. 79 (2018) 284–293, https://doi.
JVI.02261-16. org/10.1016/j.fsi.2018.05.031.
[101] C.G. Lundin, Globabl Attempts to Address Shrimp Disease, The World Bank, 1995. [122] D. Nurhayati, W.M. Yuhana, Dietary synbiotic influence on the growth
[102] H. Zokaeifar, J.L. Balcázar, C.R. Saad, M.S. Kamarudin, K. Sijam, A. Arshad, performances and immune responses to co-infection with infectious Myonecrosis
N. Nejat, Effects of Bacillus subtilis on the growth performance, digestive enzymes, virus and Vibrio harveyi in Litopenaeus vannamei, J. Fish. Aquat. Sci. 10 (2015)
immune gene expression and disease resistance of white shrimp, Litopenaeus 13–23, https://doi.org/10.3923/jfas.2015.13.23.
vannamei, Fish Shellfish Immunol. 33 (2012) 683–689, https://doi.org/10.1016/ [123] W. Widanarni, M. Yuhana, A. Muhammad, Bacillus NP5 improves growth
j.fsi.2012.05.027. performance and resistance against infectious myonecrosis virus in white shrimp
(Litopenaeus vannamei)(Bacillus NP5 meningkatkan pertumbuhan dan ketahanan

278
U.D. Butt et al. Fish and Shellfish Immunology 114 (2021) 263–281

terhadap infeksi virus myonecrosis pada udang putih (L. vannamei), ILMU kelaut, glucan binding protein (LGBP) from the white shrimp Litopenaeus vannamei, Fish
Indones. J. Mar. Sci. 19 (2014) 211–218. Shellfish Immunol. 18 (2005) 297–310, https://doi.org/10.1016/j.
[124] E. Ringø, S.K. Song, Application of dietary supplements (synbiotics and probiotics fsi.2004.08.002.
in combination with plant products and β-glucans) in aquaculture, Aquacult. [150] E.D. Abarike, F.K.A. Kuebutornye, J. Jian, J. Tang, Y. Lu, J. Cai, Influences of
Nutr. 22 (2016) 4–24, https://doi.org/10.1111/anu.12349. immunostimulants on phagocytes in cultured fish: a mini review, Rev. Aquacult.
[125] M.A.O. Dawood, S. Koshio, M.M. Abdel-Daim, H. Van Doan, Probiotic application 11 (2019) 1219–1227, https://doi.org/10.1111/raq.12288.
for sustainable aquaculture, Rev. Aquacult. 11 (2019) 907–924, https://doi.org/ [151] N. Van Hai, R. Fotedar, Comparison of the effects of the prebiotics (Bio-Mos® and
10.1111/raq.12272. β-1,3-D-glucan) and the customised probiotics (Pseudomonas synxantha and
[126] M.A.O. Dawood, S. Koshio, M.Á. Esteban, Beneficial roles of feed additives as P. aeruginosa) on the culture of juvenile western king prawns (Penaeus latisulcatus
immunostimulants in aquaculture: a review, Rev. Aquacult. 10 (2018) 950–974, Kishinouye, 1896), Aquaculture 289 (2009) 310–316, https://doi.org/10.1016/j.
https://doi.org/10.1111/raq.12209. aquaculture.2009.02.001.
[127] C.H. Chiu, Y.K. Guu, C.H. Liu, T.M. Pan, W. Cheng, Immune responses and gene [152] G.X. Wang, Y. Zhou, W.Q. Huang, Y.H. Huang, X.H. Liu, S.Z. Dong, Effects of
expression in white shrimp, Litopenaeus vannamei, induced by Lactobacillus xylooligosaccharide on growth, body composition and non-specific immunity in
plantarum, Fish Shellfish Immunol. 23 (2007) 364–377, https://doi.org/10.1016/ Litopenaeus vannamei, Freshw. Fish. 5 (2010) 54–58. http://en.cnki.com.cn/Artic
j.fsi.2006.11.010. le_en/CJFDTotal-DSYY201005011.htm.
[128] S. Mohapatra, T. Chakraborty, V. Kumar, G. Deboeck, K.N. Mohanta, Aquaculture [153] M. Yousefian, M.S. Amiri, A review of the use of prebiotic in aquaculture for fish
and stress management: a review of probiotic intervention,, J. Anim. Physiol. and shrimp, African J, Biotechnol. 8 (2009) 7313–7318, https://doi.org/
Anim. Nutr. 97 (2013) 405–430, https://doi.org/10.1111/j.1439- 10.5897/AJB2009.000-9566.
0396.2012.01301.x. [154] G.D. Brown, P.R. Taylor, D.M. Reid, J.A. Willment, D.L. Williams, L. Martinez-
[129] L.E.M. Niers, H.M. Timmerman, G.T. Rijkers, G.M. Van Bleek, N.O.P. Van Uden, Pomares, S.Y.C. Wong, S. Gordon, Dectin-1 is a major β-glucan receptor on
E.F. Knol, M.L. Kapsenberg, J.L.L. Kimpen, M.O. Hoekstra, Identification of strong macrophages, J. Exp. Med. 196 (2002) 407–412, https://doi.org/10.1084/
interleukin-10 inducing lactic acid bacteria which down-regulate T helper type 2 jem.20020470.
cytokines, Clin. Exp. Allergy 35 (2005) 1481–1489, https://doi.org/10.1111/ [155] M. Yadav, J.S. Schorey, The β-glucan receptor dectin-1 functions together with
j.1365-2222.2005.02375.x. TLR2 to mediate macrophage activation by mycobacteria,, Blood 108 (2006)
[130] D.H. Kim, B. Austin, Cytokine expression in leucocytes and gut cells of rainbow 3168–3175, https://doi.org/10.1182/blood-2006-05-024406.
trout, Oncorhynchus mykiss Walbaum, induced by probiotics, Vet. Immunol. [156] P.A. Bron, P. van Baarlen, M. Kleerebezem, Emerging molecular insights into the
Immunopathol. 114 (2006) 297–304, https://doi.org/10.1016/j. interaction between probiotics and the host intestinal mucosa, Nat. Rev.
vetimm.2006.08.015. Microbiol. 10 (2012) 66–78, https://doi.org/10.1038/nrmicro2690.
[131] P. Clerton, D. Troutaud, P. Deschaux, The chemiluminescence response of [157] B.L.E. Trujillo, L. Rivera, E. Hardy, E.M. Llumiquinga, F. Garrido, Natural
leucocytes isolated from the gut of rainbow trout (Oncorhynchus mykiss),, Fish strategies to improve growth and health of farmed shrimp examining the use of
Shellfish Immunol. 8 (1998) 73–76, https://doi.org/10.1006/fsim.1997.0114. prebiotics, probiotics and, Glob. Aquac. Advocate. (2018). https://www.aquacult
[132] J.L. Balcázar, I. de Blas, I. Ruiz-Zarzuela, D. Vendrell, A.C. Calvo, I. Márquez, urealliance.org/advocate/natural-strategies-growth-health-farmed-shrimp/.
O. Gironés, J.L. Muzquiz, Changes in intestinal microbiota and humoral immune (Accessed 3 July 2020).
response following probiotic administration in brown trout (Salmo trutta), Br. J. [158] J. Rodríguez, G. Le Moullac, State of the art of immunological tools and health
Nutr. 97 (2007) 522–527, https://doi.org/10.1017/S0007114507432986. control of penaeid shrimp, Aquaculture 191 (2000) 109–119, https://doi.org/
[133] S.K. Nayak, Probiotics and immunity: a fish perspective,, Fish Shellfish Immunol. 10.1016/S0044-8486(00)00421-X.
29 (2010) 2–14, https://doi.org/10.1016/j.fsi.2010.02.017. [159] U. Wongsasak, S. Chaijamrus, S. Kumkhong, S. Boonanuntanasarn, Effects of
[134] M. Suva, V. Sureja, D. Kheni, Novel insight on probiotic Bacillus subtilis: dietary supplementation with β-glucan and synbiotics on immune gene expression
mechanism of action and clinical applications, J. Curr. Res. Sci. Med, 3 136 and immune parameters under ammonia stress in Pacific white shrimp,
(2017), https://doi.org/10.4103/jcrsm.jcrsm_47_17. Aquaculture 436 (2015) 179–187, https://doi.org/10.1016/j.
[135] Y. Yi, Z. Zhang, F. Zhao, H. Liu, L. Yu, J. Zha, G. Wang, Probiotic potential of aquaculture.2014.10.028.
Bacillus velezensis JW: antimicrobial activity against fish pathogenic bacteria and [160] A. Luna-González, J.C. Almaraz-Salas, J.A. Fierro-Coronado, M. del C. Flores-
immune enhancement effects on Carassius auratus, Fish Shellfish Immunol. 78 Miranda, H.A. González-Ocampo, V. Peraza-Gómez, The prebiotic inulin increases
(2018) 322–330, https://doi.org/10.1016/j.fsi.2018.04.055. the phenoloxidase activity and reduces the prevalence of WSSV in whiteleg
[136] J.L. Balcázar, Evaluation of Probiotic Bacterial Strains in Litopenaeus Vannamei, shrimp (Litopenaeus vannamei) cultured under laboratory conditions, Aquaculture,
Ecuador., Guayaquil, 2003. 362–363 (2012) 28–32, https://doi.org/10.1016/j.aquaculture.2012.07.022.
[137] M. Zhang, J. Shi, Application of photosynthetic bacteria in aquatic rearing., [161] W. Rungrassamee, Y. Kingcha, Y. Srimarut, S. Maibunkaew, N. Karoonuthaisiri,
Chinese J. Appl. Environ. Biol. S1. http://en.cnki.com.cn/Article_en/CJFDTota W. Visessanguan, Mannooligosaccharides from copra meal improves survival of
l-YYHS1999S1053.htm, 1999. the Pacific white shrimp (Litopenaeus vannamei) after exposure to Vibrio harveyi,
[138] J. Mao, G. Zhou, B. Chen, X. Du, The effects of compound microorganisms on Aquaculture 434 (2014) 403–410, https://doi.org/10.1016/j.
improving water quality. J,, Aquaculture 27 (2006) 25–27. aquaculture.2014.08.032.
[139] S. Rengpipat, S. Rukpratanporn, S. Piyatiratitivorakul, P. Menasaveta, Immunity [162] D. Destoumieux, M. Muñoz, C. Cosseau, J. Rodriguez, P. Bulet, M. Comps,
enhancement in black tiger shrimp (Penaeus monodon) by a probiont bacterium E. Bachère, Penaeidins, antimicrobial peptides with chitin-binding activity, are
(Bacillus S11), Aquaculture 191 (2000) 271–288, https://doi.org/10.1016/ produced and stored in shrimp granulocytes and released after microbial
S0044-8486(00)00440-3. challenge, J. Cell Sci. 113 (2000) 461–469.
[140] H.H. Sung, Y.L. Yang, Y.L. Song, Enhancement of microbicidal activity in the tiger [163] T. Muta, S. Iwanaga, The role of hemolymph coagulation in innate immunity,
shrimp Penaeus monodon via immunostimulation, J. Crustac Biol. 16 (1996) 278, Curr. Opin, Immunol. 8 (1996) 41–47, https://doi.org/10.1016/S0952-7915(96)
https://doi.org/10.2307/1548883. 80103-8.
[141] E. Bachère, Shrimp immunity and disease control, Aquaculture 191 (2000) 3–11, [164] H. Sabry Neto, A.J.P. Nunes, Performance and immunological resistance of
https://doi.org/10.1016/S0044-8486(00)00413-0. Litopenaeus vannamei fed a β-1,3/1,6-glucan-supplemented diet after per os
[142] F.-J. Gatesoupe, Updating the importance of lactic acid bacteria in fish farming: challenge with the Infectious Myonecrosis Virus (IMNV), Rev. Bras. Zootec. 44
natural occurrence and probiotic treatments, J. Mol. Microbiol. Biotechnol. 14 (2015) 165–173, https://doi.org/10.1590/S1806-92902015000500001.
(2008) 107–114, https://doi.org/10.1159/000106089. [165] W. Cheng, J.C. Chen, Effects of intrinsic and extrinsic factors on the haemocyte
[143] P.R. Rauta, B. Nayak, S. Das, Immune system and immune responses in fish and profile of the prawn, Macrobrachium rosenbergii, Fish Shellfish Immunol. 11
their role in comparative immunity study: a model for higher organisms,, (2001) 53–63, https://doi.org/10.1006/fsim.2000.0293.
Immunol. Lett. 148 (2012) 23–33, https://doi.org/10.1016/j.imlet.2012.08.003. [166] L. Cerenius, B.L. Lee, K. Söderhäll, The proPO-system: pros and cons for its role in
[144] N. Sheikhzadeh, A. Karimi Pashaki, K. Nofouzi, M. Heidarieh, H. Tayefi- invertebrate immunity,, Trends Immunol. 29 (2008) 263–271, https://doi.org/
Nasrabadi, Effects of dietary Ergosan on cutaneous mucosal immune response in 10.1016/j.it.2008.02.009.
rainbow trout (Oncorhynchus mykiss), Fish Shellfish Immunol. 32 (2012) 407–410, [167] P. Li, Y.-L. Yin, D. Li, S. Woo Kim, G. Wu, Amino acids and immune function, Br.
https://doi.org/10.1016/j.fsi.2011.11.028. J. Nutr. 98 (2007) 237–252, https://doi.org/10.1017/S000711450769936X.
[145] T.N. McNeilly, S.W. Naylor, A. Mahajan, M.C. Mitchell, S. McAteer, D. Deane, D. [168] M. Machado, R. Azeredo, P. Díaz-Rosales, A. Afonso, H. Peres, A. Oliva-Teles,
G.E. Smith, J.C. Low, D.L. Gally, J.F. Huntley, Escherichia coli O157:H7 B. Costas, Dietary tryptophan and methionine as modulators of European seabass
colonization in cattle following systemic and mucosal immunization with purified (Dicentrarchus labrax) immune status and inflammatory response, Fish Shellfish
H7 flagellin, Infect. Immun. 76 (2008) 2594–2602, https://doi.org/10.1128/ Immunol. 42 (2015) 353–362, https://doi.org/10.1016/j.fsi.2014.11.024.
IAI.01452-07. [169] H. Sugita, Y. Hirose, N. Matsuo, Y. Deguchi, Production of the antibacterial
[146] T. Sangma, D. Kamilya, Dietary Bacillus subtilis FPTB13 and chitin, single or substance by Bacillus sp. strain NM 12, an intestinal bacterium of Japanese coastal
combined, modulate systemic and cutaneous mucosal immunity and resistance of fish, Aquaculture 165 (1998) 269–280, https://doi.org/10.1016/S0044-8486(98)
catla, Catla catla (Hamilton) against edwardsiellosis, Comp. Immunol. Microbiol. 00267-1.
Infect. Dis. 43 (2015) 8–15, https://doi.org/10.1016/j.cimid.2015.09.003. [170] M. Amin, Z. Rakhisi, A. Zarei Ahmady, Isolation and identification of Bacillus
[147] D.M. Gatlin, A.M. Peredo, Prebiotics and probiotics : definitions and applications, species from soil and evaluation of their antibacterial properties, Avicenna J. Clin.
South. Reg. Aquauclture Cent 4711 (2012) 1–8. Microbiol. Infect 2 (2015), https://doi.org/10.17795/ajcmi-23233.
[148] L. Cerenius, K. Soderhall, The prophenoloxidase-activating system in [171] M. Mutaz Al-Ajlani, S. Hasnain, Bacteria exhibiting antimicrobial activities;
invertebrates,, Immunol. Rev. 198 (2004) 116–126, https://doi.org/10.1111/ Screening for antibiotics and the associated genetic studies, Open Conf. Proc. J. 1
j.0105-2896.2004.00116.x. (2010) 230–238, https://doi.org/10.2174/2210289201001010230.
[149] W. Cheng, C.-H. Liu, C.-H. Tsai, J.-C. Chen, Molecular cloning and [172] J.L. Balcázar, T. Rojas-Luna, Inhibitory activity of probiotic Bacillus subtilis UTM
characterisation of a pattern recognition molecule, lipopolysaccharide- and β-1,3- 126 against Vibrio species confers protection against Vibriosis in juvenile shrimp

279
U.D. Butt et al. Fish and Shellfish Immunology 114 (2021) 263–281

(Litopenaeus vannamei), Curr. Microbiol. 55 (2007) 409–412, https://doi.org/ [197] D. De, R. Ananda Raja, T.K. Ghoshal, S. Mukherjee, K.K. Vijayan, Evaluation of
10.1007/s00284-007-9000-0. growth, feed utilization efficiency and immune parameters in tiger shrimp
[173] E.D. Abarike, J. Cai, Y. Lu, H. Yu, L. Chen, J. Jian, J. Tang, L. Jun, F.K. (Penaeus monodon) fed diets supplemented with or diet fermented with gut
A. Kuebutornye, Effects of a commercial probiotic BS containing Bacillus subtilis bacterium Bacillus sp. DDKRC1. isolated from gut of Asian seabass (Lates
and Bacillus licheniformis on growth, immune response and disease resistance in calcarifer), Aquacult. Res. 49 (2018) 2147–2155, https://doi.org/10.1111/
Nile tilapia, Oreochromis niloticus, Fish Shellfish Immunol. 82 (2018) 229–238, are.13669.
https://doi.org/10.1016/j.fsi.2018.08.037. [198] N. Sadat Hoseini Madani, T.J. Adorian, H. Ghafari Farsani, S.H. Hoseinifar, The
[174] X. Wang, H. Li, X. Zhang, Y. Li, W. Ji, H. Xu, Microbial flora in the digestive tract effects of dietary probiotic Bacilli (Bacillus subtilis and Bacillus licheniformis) on
of adult penaeid shrimp (Penaeus chinensis), J. Ocean Univ. Qingdao 30 (2000) growth performance, feed efficiency, body composition and immune parameters
493–498, abstract/CBA/338145. of whiteleg shrimp (Litopenaeus vannamei) postlarvae,, Aquacult. Res. 49 (2018)
[175] N.G. Vine, W.D. Leukes, H. Kaiser, Probiotics in marine larviculture, FEMS 1926–1933, https://doi.org/10.1111/are.13648.
Microbiol. Rev. 30 (2006) 404–427, https://doi.org/10.1111/j.1574- [199] S. Das, K. Mondal Associate Professor, S. Haque, C. Kausik Mondal Associate
6976.2006.00017.x. Professor, K. Mondal, A review on application of probiotic, prebiotic and
[176] E.R. EL-Haroun, A.M.A.-S. Goda, M.A. Kabir Chowdhury, Effect of dietary synbiotic for sustainable development of aquaculture, ~ 422 ~ J. Entomol. Zool,
probiotic Biogen - supplementation as a growth promoter on growth performance For. Stud. 5 (2017) 422–429.
and feed utilization of Nile tilapia Oreochromis niloticus (L.), Aquacult. Res. 37 [200] M.A. Genc, M. Aktas, E. Genc, E. Yilmaz, Effects of dietary mannan
(2006) 1473–1480, https://doi.org/10.1111/j.1365-2109.2006.01584.x. oligosaccharide on growth, body composition and hepatopancreas histology of
[177] S. Ziaei-Nejad, M.H. Rezaei, G.A. Takami, D.L. Lovett, A.-R. Mirvaghefi, Penaeus semisulcatus (de Haan 1844), Aquacult. Nutr. 13 (2007) 156–161, https://
M. Shakouri, The effect of Bacillus spp. bacteria used as probiotics on digestive doi.org/10.1111/j.1365-2095.2007.00469.x.
enzyme activity, survival and growth in the Indian white shrimp Fenneropenaeus [201] M.-C. Yu, Z.-J. Li, H.-Z. Lin, G.-L. Wen, S. Ma, Effects of dietary medicinal herbs
indicus,, Aquaculture 252 (2006) 516–524, https://doi.org/10.1016/j. and Bacillus on survival, growth, body composition, and digestive enzyme activity
aquaculture.2005.07.021. of the white shrimp Litopenaeus vannamei, Aquacult. Int. 17 (2009) 377–384,
[178] S. Seifert, B. Watzl, Inulin, Oligofructose: review of experimental data on immune https://doi.org/10.1007/s10499-008-9209-3.
modulation, J. Nutr. 137 (2007) 2563S–2567S, https://doi.org/10.1093/jn/ [202] B.O. Partida-Arangure, A. Luna-González, J.A. Fierro-Coronado, M. del Carmen
137.11.2563S. Flores-Miranda, H.A. González-Ocampo, Effect of inulin and probiotic bacteria on
[179] L.V. Hooper, A.J. Macpherson, Immune adaptations that maintain homeostasis growth, survival, immune response, and prevalence of White Spot Syndrome
with the intestinal microbiota, Nat. Rev. Immunol. 10 (2010) 159–169, https:// Virus (WSSV) in Litopenaeus vannamei cultured under laboratory conditions, Afr.
doi.org/10.1038/nri2710. J. Biotechnol. 12 (2013), https://doi.org/10.5897/AJB12.1569.
[180] R.C. Jose Meseguer, Current knowledge in synbiotic use for fish aquaculture: a [203] E. Amenyogbe, G. Chen, Z. Wang, J. Huang, B. Huang, H. Li, The exploitation of
review, J. Aquac. Res. Dev. s1 (2011) 1–7, https://doi.org/10.4172/2155-9546. probiotics, prebiotics and synbiotics in aquaculture: present study, limitations and
S1-008. future directions. : a review, Aquac, Bar Int. 28 (2020) 1017–1041, https://doi.
[181] Y.K. Lee, C.Y. Lim, W.L. Teng, A.C. Ouwehand, E.M. Tuomola, S. Salminen, org/10.1007/s10499-020-00509-0.
Quantitative approach in the study of adhesion of lactic acid bacteria to intestinal [204] R.T. Di Giulio, C. Habig, E.P. Gallagher, Effects of Black Rock Harbor sediments
cells and their competition with Enterobacteria, Appl. Environ. Microbiol. 66 on indices of biotransformation, oxidative stress, and DNA integrity in channel
(2000) 3692–3697, https://doi.org/10.1128/AEM.66.9.3692-3697.2000. catfish, Aquat. Toxicol 26 (1993) 1–22, https://doi.org/10.1016/0166-445X(93)
[182] X. Pan, T. Wu, L. Zhang, Z. Song, H. Tang, Z. Zhao, In vitro evaluation on 90002-I.
adherence and antimicrobial properties of a candidate probiotic Clostridium [205] J. Li, Y. Xu, L. Jin, X. Li, Effects of a probiotic mixture (Bacillus subtilis YB-1 and
butyricum CB2 for farmed fish, J. Appl. Microbiol. 105 (2008) 1623–1629, Bacillus cereus YB-2) on disease resistance and non-specific immunity of sea
https://doi.org/10.1111/j.1365-2672.2008.03885.x. cucumber, Apostichopus japonicus (Selenka), Aquac. Res. 46 (2015) 3008–3019,
[183] A. Kesarcodi-Watson, H. Kaspar, M.J. Lategan, L. Gibson, Probiotics in https://doi.org/10.1111/are.12453.
aquaculture: the need, principles and mechanisms of action and screening [206] L. Wang, C. Ge, J. Wang, J. Dai, P. Zhang, Y. Li, Effects of different combinations
processes,, Aquaculture 274 (2008) 1–14, https://doi.org/10.1016/j. of Bacillus on immunity and antioxidant activities in common carp, Aquacult. Int.
aquaculture.2007.11.019. 25 (2017) 2091–2099, https://doi.org/10.1007/s10499-017-0175-5.
[184] P. Pandiyan, D. Balaraman, R. Thirunavukkarasu, E.G.J. George, [207] I. Messaoudi, S. Barhoumi, K. Saïd, A. Kerken, Study on the sensitivity to
K. Subaramaniyan, S. Manikkam, B. Sadayappan, Probiotics in aquaculture, drug cadmium of marine fish Salaria basilisca (Pisces: blennidae), J. Environ. Sci. 21
invent, Today Off. 5 (2013) 55–59, https://doi.org/10.1016/j.dit.2013.03.003. (2009) 1620–1624, https://doi.org/10.1016/S1001-0742(08)62464-X.
[185] I.V. Pinchuk, P. Bressollier, B. Verneuil, B. Fenet, I.B. Sorokulova, F. Mégraud, M. [208] N. Van Hai, Research findings from the use of probiotics in tilapia aquaculture: a
C. Urdaci, In vitro anti-Helicobacter pylori activity of the probiotic strain Bacillus review,, Fish Shellfish Immunol. 45 (2015) 592–597, https://doi.org/10.1016/j.
subtilis 3 is due to secretion of antibiotics, Antimicrob. Agents Chemother. 45 fsi.2015.05.026.
(2001) 3156–3161, https://doi.org/10.1128/AAC.45.11.3156-3161.2001. [209] A. Gupta, G. Verma, P. Gupta, Growth performance, feed utilization, digestive
[186] T. Stein, Bacillus subtilis antibiotics: structures, syntheses and specific functions, enzyme activity, innate immunity and protection against Vibrio harveyi of
Mol. Microbiol. 56 (2005) 845–857, https://doi.org/10.1111/j.1365- freshwater prawn, Macrobrachium rosenbergii fed diets supplemented with
2958.2005.04587.x. Bacillus coagulans, Aquacult. Int. 24 (2016) 1379–1392, https://doi.org/
[187] H.K. Miandare, P. Yarahmadi, M. Abbasian, Immune related transcriptional 10.1007/s10499-016-9996-x.
responses and performance of Litopenaeus vannamei post-larvae fed on dietary [210] X. Zhou, W. Lin, L. Tong, X. Liu, K. Zhong, L. Liu, L. Wang, S. Zhou,
probiotic PrimaLac®, Fish Shellfish Immunol. 55 (2016) 671–678, https://doi. Hypolipidaemic effects of oat flakes and β -glucans derived from four Chinese
org/10.1016/j.fsi.2016.06.053. naked oat (Avena nuda) cultivars in Wistar-Lewis rats, J. Sci. Food Agric. 96
[188] M.C. Urdaci, I. Pinchuk, Antimicrobial Activity of Bacillus Probiotics - Bacterial (2016) 644–649, https://doi.org/10.1002/jsfa.7135.
Spore Formers : Probiotics and Emerging Applications, 2004. [211] D. Abdollahi-Arpanahi, E. Soltani, H. Jafaryan, M. Soltani, M. Naderi-Samani, A.
[189] K. Kongnum, T. Hongpattarakere, Effect of Lactobacillus plantarum isolated from I. Campa-Córdova, Retracted: efficacy of two commercial and indigenous
digestive tract of wild shrimp on growth and survival of white shrimp (Litopenaeus probiotics, Bacillus subtilis and Bacillus licheniformis on growth performance,
vannamei) challenged with Vibrio harveyi, Fish Shellfish Immunol. 32 (2012) immuno-physiology and resistance response of juvenile white shrimp (Litopenaeus
170–177, https://doi.org/10.1016/j.fsi.2011.11.008. vannamei), Aquaculture 496 (2018) 43–49, https://doi.org/10.1016/j.
[190] M. Lara-Flores, G. Aguirre-Guzman, The use of probiotic in fish and shrimp aquaculture.2018.06.082.
aquaculture, A review, Probiotics Prod. Eval. Uses Anim. Feed. Res. Signpost, [212] C.-H. Liu, C.-S. Chiu, P.-L. Ho, S.-W. Wang, Improvement in the growth
Kerala (2009) 75–89. performance of white shrimp, Litopenaeus vannamei, by a protease-producing
[191] J.A. Vázquez, M.P. González, M.A. Murado, Effects of lactic acid bacteria cultures probiotic, Bacillus subtilis E20, from natto, J. Appl. Microbiol. 107 (2009)
on pathogenic microbiota from fish, Aquaculture 245 (2005) 149–161, https:// 1031–1041, https://doi.org/10.1111/j.1365-2672.2009.04284.x.
doi.org/10.1016/j.aquaculture.2004.12.008. [213] U.S. Food and Drug Administration, Nutrition information for cooked seafood
[192] P. Utiswannakul, S. Sangchai, S. Rengpipat, Enhanced growth of black tiger (Purchased Raw), (2018). https://www.fda.gov/food/food-labeling-nutrition/n
shrimp Penaeus Monodon by dietary supplementation with Bacillus (BP11) as a utrition-information-cooked-seafood-purcahsed-raw. (Accessed 17 June 2020).
probiotic, J. Aquac. Res. Dev. 2 (Special Issue) (2011) 1–9, https://doi.org/ [214] J. Li, B. Tan, K. Mai, Q. Ai, W. Zhang, Z. Liufu, W. Xu, Immune responses and
10.4172/2155-9546.S1-006. resistance against Vibrio parahaemolyticus induced by probiotic bacterium
[193] R.M. Reda, K.M. Selim, Evaluation of Bacillus amyloliquefaciens on the growth Arthrobacter XE-7 in Pacific white shrimp, Litopenaeus vannamei, J. World
performance, intestinal morphology, hematology and body composition of Nile Aquacult. Soc. 39 (2008) 477–489, https://doi.org/10.1111/j.1749-
tilapia, Oreochromis niloticus, Aquacult. Int. 23 (2015) 203–217, https://doi.org/ 7345.2008.00188.x.
10.1007/s10499-014-9809-z. [215] D.J. Moriarty, Control of luminous Vibrio species in penaeid aquaculture ponds,
[194] M. Lara-flores, The use of probiotic in aquaculture : an overview,, Int. Res. J. Aquaculture 164 (1998) 351–358, https://doi.org/10.1016/S0044-8486(98)
Microbiol. 2 (2011) 471–478. http://www.interesjournals.org/IRJM. 00199-9.
[195] G. Cooke, J. Behan, M. Costello, Newly identified vitamin K-producing bacteria [216] S. Rengpipat, W. Phianphak, S. Piyatiratitivorakul, P. Menasveta, Effects of a
isolated from the neonatal faecal flora, Microb. Ecol. Health Dis. 18 (2006) probiotic bacterium on black tiger shrimp Penaeus monodon survival and growth,
133–138, https://doi.org/10.1080/08910600601048894. Aquaculture 167 (1998) 301–313, https://doi.org/10.1016/S0044-8486(98)
[196] A. Yuniarti, D.A. Guntoro, A.M. Hariati, Response of indigenous Bacillus 00305-6.
megaterium supplementation on the growth of Litopenaeus vannamei ( Boone ), a [217] S. Rengpipat, A. Tunyanun, A. Fast, S. Piyatiratitivorakul, P. Menasveta,
new target species for shrimp culture in East Java of Indonesia, J Basic Appl Sci Enhanced growth and resistance to Vibrio challenge in pond-reared black tiger
Res. 3 (2013) 747–754.

280
U.D. Butt et al. Fish and Shellfish Immunology 114 (2021) 263–281

shrimp Penaeus monodon fed a Bacillus probiotic, Dis. Aquat. Org. 55 (2003) shrimp feed, World J. Microbiol. Biotechnol. 18 (2002) 527–539, https://doi.org/
169–173, https://doi.org/10.3354/dao055169. 10.1023/A:1016322227535.
[218] B. Vaseeharan, P. Ramasamy, Control of pathogenic Vibrio spp. by Bacillus subtilis [229] N. Felix, M.J.P. Jeyaseelan, C.J.W. Kirubakaran, Growth improvement and
BT23, a possible probiotic treatment for black tiger shrimp Penaeus monodon, Lett. enhanced disease resistance against Vibrio alginolyticus using beta -glucan as a
Appl. Microbiol. 36 (2003) 83–87, https://doi.org/10.1046/j.1472- dietary supplement for Penaeus monodon (Fabricius), Indian J. Fish. 55 (2008)
765X.2003.01255.x. 247–250.
[219] K.F. Liu, C.H. Chiu, Y.L. Shiu, W. Cheng, C.H. Liu, Effects of the probiotic, Bacillus [230] T.P. Sajeevan, R. Philip, I.S. Bright Singh, Dose/frequency: a critical factor in the
subtilis E20, on the survival, development, stress tolerance, and immune status of administration of glucan as immunostimulant to Indian white shrimp
white shrimp, Litopenaeus vannamei larvae, Fish Shellfish Immunol 28 (2010) Fenneropenaeus indicus,, Aquaculture 287 (2009) 248–252, https://doi.org/
837–844, https://doi.org/10.1016/j.fsi.2010.01.012. 10.1016/j.aquaculture.2008.10.045.
[220] M. Gullian, F. Thompson, J. Rodriguez, Selection of probiotic bacteria and study [231] N. López, G. Cuzon, G. Gaxiola, G. Taboada, M. Valenzuela, C. Pascual,
of their immunostimulatory effect in Penaeus vannamei, Aquaculture 233 (2004) A. Sánchez, C. Rosas, Physiological, nutritional, and immunological role of
1–14, https://doi.org/10.1016/j.aquaculture.2003.09.013. dietary β 1-3 glucan and ascorbic acid 2-monophosphate in Litopenaeus vannamei
[221] S. Das, P.S. Lyla, S. Ajmal Khan, Application of Streptomyces as a probiotic in the juveniles, Aquaculture 224 (2003) 223–243, https://doi.org/10.1016/S0044-
laboratory culture of Penaeus monodon (Fabricius), Isr. J. Aquac. Bamidgeh 58 8486(03)00214-X.
(2006) 198–204. [232] Y.-C. Wang, P.-S. Chang, H.-Y. Chen, Differential time-series expression of
[222] R. Chythanya, I. Karunasagar, I. Karunasagar, Inhibition of shrimp pathogenic immune-related genes of Pacific white shrimp Litopenaeus vannamei in response to
vibrios by a marine Pseudomonas I-2 strain, Aquaculture 208 (2002) 1–10, dietary inclusion of β-1,3-glucan, Fish Shellfish Immunol. 24 (2008) 113–121,
https://doi.org/10.1016/S0044-8486(01)00714-1. https://doi.org/10.1016/j.fsi.2007.09.008.
[223] R. Preetha, N.S. Jayaprakash, I.S. Bright Singh, Synechocystis MCCB 114 and 115 [233] N. Bai, M. Gu, W. Zhang, W. Xu, K. Mai, Effects of β-glucan derivatives on the
as putative probionts for Penaeus monodon post-larvae, Dis. Aquat. Org. 74 (2007) immunity of white shrimp Litopenaeus vannamei and its resistance against white
243–247, https://doi.org/10.3354/dao074243. spot syndrome virus infection, Aquaculture, 426–427 (2014) 66–73, https://doi.
[224] M.S. Su, K.F. Liu, C.F. Chang, I.C. Liao, Enhancement of grass prawn Penaeus org/10.1016/j.aquaculture.2014.01.019.
monodon postlarvae viability by beta-1, 3-glucan from Schizophyllum commune, [234] Y.-S. Wu, S.-Y. Liau, C.-T. Huang, F.-H. Nan, Beta 1,3/1,6-glucan and vitamin C
J. Taiwan Fish. Res. 3 (1995) 125–132. immunostimulate the non-specific immune response of white shrimp (Litopenaeus
[225] I.C. Liao, M.-S. Su, C.-F. Chang, B.-Y. Her, T. Kojima, Enhancement of the vannamei), Fish Shellfish Immunol. 57 (2016) 269–277, https://doi.org/10.1016/
resistance of grass prawn Penaeus monodon against Vibrio damsela infection by j.fsi.2016.08.046.
beta-1, 3-glucan, J. Fish. Soc. Taiwan 23 (1996) 109–116. [235] N.S. Solidum, R.C. Sanares, K.G.S. Andrino-Felarca, V.L. Corre Jr., Immune
[226] C. Chang, M. Su, H. Chen, C. Lo, G. Kou, I. Liao, Effect of dietary β-1,3-glucan on responses and resistance to Vibriosis of juvenile Pacific whiteleg shrimp Penaeus
resistance to white spot syndrome virus (WSSV) in postlarval and juvenile Penaeus Vannamei fed with high dose mannan oligosaccharide and β-glucan, AACL Bioflux
monodon, Dis. Aquat. Org. 36 (1999) 163–168, https://doi.org/10.3354/ 9 (2016) 239–249.
dao036163. [236] V. Peraza-Gómez, A. Luna-González, J.M. González-Prieto, A. Fierro-Coronado, H.
[227] C.-F. Chang, H.-Y. Chen, M.-S. Su, I.-C. Liao, Immunomodulation by dietary β-1, A. González-Ocampo, Protective effect of microbial immunostimulants and
3-glucan in the brooders of the black tiger shrimp Penaeus monodon, Fish Shellfish antiviral plants against WSSV in Litopenaeus vannamei cultured under laboratory
Immunol. 10 (2000) 505–514, https://doi.org/10.1006/fsim.2000.0266. conditions, Aquaculture, 420–421 (2014) 160–164, https://doi.org/10.1016/j.
[228] P. Thanardkit, P. Khunrae, M. Suphantharika, C. Verduyn, Glucan from spent aquaculture.2013.10.044.
brewer’s yeast: preparation, analysis and use as a potential immunostimulant in

281

You might also like