ArticlePDF Available

Core Gut Microbiota of Shrimp Function as a Regulator to Maintain Immune Homeostasis in Response to WSSV Infection

Authors:

Abstract and Figures

Abundant gut microorganisms constitute a complex microecosystem with the intestinal environment of the host, which plays a critical role in the adjustment of various physiological states of the organism. Sequencing and mass spectrometry data collected from intestinal samples of shrimp after virus infection helped to investigate the special function of the microecosystem and suggested that the gut microbiota has a functional potential in maintaining immune homeostasis of the host under environmental challenge.
Content may be subject to copyright.
Core Gut Microbiota of Shrimp Function as a Regulator to
Maintain Immune Homeostasis in Response to WSSV Infection
Siyuan Zhang,
a
Xumei Sun
a
a
School of Marine Science, Ningbo University, Ningbo, Peoples Republic of China
ABSTRACT The gut microbiota is an integral part of the host and has a functional
potential in host physiology. Numerous scientic efforts have opened new horizons in
gut microbiota research and enhanced the understanding of host-microbe interactions
in vertebrates. However, evidence on the association between the gut microbiota and
immunity in invertebrates, especially in shrimp, which is an important aquatic animal, is
limited. Herein, we conducted a comprehensive analysis based on 16S rRNA gene
sequencingandliquidchromatography-coupledmassspectrometry(LC-MS)toinvesti-
gate the correlation between them. Comparing the gut microbiota among the four dif-
ferent species of shrimp, we found huge variations and determined a core gut micro-
biota composed of 55 microbes. The environmental challenge of white spot syndrome
virus (WSSV) infection led to changes in core microbial structures, but the alteration of
coremicrobiotaamongdifferentshrimpfollowedthesametrendandshowedimmune-
related function in the prediction of its metabolic potential. In metabolomic analysis,
nine signicantly upregulated metabolites found after viral infection indicated that they
have antiviral potential. Moreover, we found a tight correlation between them and
almost half of the core microbiota. These data demonstrate that these metabolites are
responsible for maintaining the immune homeostasis of the host and prove the function
of the gut microbiota and the related metabolome in antiviral immunity of shrimp.
IMPORTANCE Abundant gut microorganisms constitute a complex microecosystem
with the intestinal environment of the host, which plays a critical role in the adjust-
ment of various physiological states of the organism. Sequencing and mass spec-
trometry data collected from intestinal samples of shrimp after virus infection helped
to investigate the special function of the microecosystem and suggested that the
gut microbiota has a functional potential in maintaining immune homeostasis of the
host under environmental challenge.
KEYWORDS gut microbiota, metabolome, immune response, WSSV, shrimp
The gut microbiota is a complex microbial ecosystem with important roles in health
and development of organisms (13). Some deterministic and stochastic processes
are thought to shape the gut microbiota. These processes are generally driven by envi-
ronmental and biological factors. Factors such as host immune system, pH in the gut,
and dietary composition are considered to be the dominant factors in shaping the gut
microbiota, processes also termed environmental selection (4, 5). Biological factors, for
example, interspecies interactions (competitive, mutualistic, and some synergistic
interactions) may further affect the composition of the microbiota (68). Under the syn-
ergistic effect of these processes, the gut microbiota in different habitats can be di-
vided into core microbes and habitat-specic microbes (911). The gut microbiota pro-
foundly regulates homeostasis mechanisms by assisting the establishment of the
intestinal epithelial barrier and maintenance of immune homeostasis in hosts (12, 13).
In the human body, previous studies indicated intense communication between the
gut microbiota and intestinal epithelial cells and immune cells shaped specic immune
Editor Daniel R. Perez, University of Georgia
Copyright © 2022 Zhang and Sun. This is an
open-access article distributed under the terms
of the Creative Commons Attribution 4.0
International license.
Address correspondence to Xumei Sun,
sunxumei@nbu.edu.cn.
The authors declare no conict of interest.
Received 3 December 2021
Accepted 22 March 2022
Published 12 April 2022
March/April 2022 Volume 10 Issue 2 10.1128/spectrum.02465-21 1
RESEARCH ARTICLE
responses to antigens, balancing tolerance and effector immune functions (14). For the
regulation of immunity by the gut microbiota, the wide range of secondary metabo-
lites produced by the commensal gut microbiota were proven to be crucial for host
physiology and host immunity regulation (15). To date, numerous studies of the gut
microbiota and its functional potential have been conducted on vertebrates, and infor-
mation concerning invertebrates is limited, especially in aquatic invertebrates.
Shrimp are one of the most important animals in aquatic aquaculture (16).
Although the farming industry of shrimp has increased considerably in recent years,
with the expansion of farming, threats by various environmental challenges restrict the
sustainable development of the industry worldwide, such as pathogen infection (17).
White spot syndrome virus (WSSV) is a typical pathogen of shrimp and with a wide
range of hosts (18). Infection with WSSV causes white spot syndrome of shrimp and
leads to 100% mortality within 7 to 10 days (19). As reported, the gut microbiota plays
a key role in regulating the immune system of the host (14). Multiple pathways such as
carbohydrate metabolism and cofactor/vitamin biosynthesis that the gut microbiota
participates in can promote host metabolism and anti-infection and anti-inammation
processes and regulate autoimmune reactions (20). Secondary metabolites produced
by the gut microbiota include short-chain fatty acids (SCFAs), polyamines, the aryl
hydrocarbon receptor (AHR), and so on (21) and can interact with host cells through
the intestinal epithelia, thus inuencing immune responses and disease risk (22). A few
studies have found that the gut microbiota is related to growth and development in
shrimp, and gut microbiota dysbiosis is responsible for shrimp white feces syndrome
(16, 23, 24). Several studies have reported the impact of WSSV infection on the intesti-
nal microbiota in Litopenaeus vannamei. However, few studies have assessed the gut
microbiota associated with host immunity responses to WSSV infection.
Here, WSSV infection was used as an environmental challenge for four different spe-
cies of shrimp (Marsupenaeus japonicus,Litopenaeus vannamei,Macrobrachium rose-
nbergii, and Procambarus clarkii). By using 16S rRNA gene sequencing and liquid chro-
matography-coupled mass spectrometry (LC-MS), the gut microbiota and metabolome
before and after virus infection were identied. We determined a group of microbes
that perform immunomodulatory functions in the gut of different species of shrimp
and assist the host in maintaining environmental adaption. These valuable ndings
greatly enhanced our understanding of the functions of the gut microbiota in main-
taining host tness under environmental challenge and provide a new strategy for the
prevention and treatment of viral infection in shrimp.
RESULTS
Gut microbiota of different shrimp. In light of exploring the shrimp gut microbiota
as a whole, four common shrimp in freshwater and seawater were used in this study:
Macrobrachium rosenbergii,Procambarus clarkia,Marsupenaeus japonicus,andLitopenaeus
vannamei. Intact intestinal tracts were sampled, and total DNA was extracted followed by
16S rRNA gene amplicon sequencing (n= 3). In total, the sequencing of the shrimp gut
microbiota yielded 383,028 reads, resulting in 1,135 operational taxonomic units (OTUs;
GenBank accession number PRJNA780955). OTUs were classied into 24 phyla and 488
genera. The composition of the microbial communities across the gut revealed signicant
discrepancies between different species of shrimp (Fig. 1A andB).Thedominantbacteria
in gut of Macrobrachium rosenbergii and Procambarus clarkii were Tenericutes and
Firmicutes, respectively. The dominant bacteria of the other two shrimp were both
Proteobacteria (Fig.1A).Amongthefourshrimpspecies,Procambarus clarkii showed signif-
icantly higher species richness (paired Wilcoxon ttest, P,0.05; Fig. 1C) and diversity
(Shannon index, P,0.01), while Macrobrachium rosenbergii and Litopenaeus vannamei
showed signicantly lower richness and diversity (P,0.05; Fig. 1C). The diversity and com-
position of gut microbes may be related to the habitant-intestinal environment of shrimp
and suggests that the gut serves as a strong environmental lter, enabling the establish-
ment of distinct microbial communities in the different shrimp.
Potential Immune Function of Microbiota in Shrimp Gut Microbiology Spectrum
March/April 2022 Volume 10 Issue 2 10.1128/spectrum.02465-21 2
A core microbiota persists across different species of shrimp. To identify the
core gut microbiota among four species of shrimp, the niche breadth of individual
microbes was characterized at the taxonomic level of the genus rst, which could reect
the adaptation of species to the environment. In general, the larger the niche breadth of
a species, the less specialized it is, that is, the more likely it is to be a generalized species.
The niche breadth of each microbe was calculated by using the Levins measure, as pre-
viously described (8, 25), and the results revealed that only a few of the microbes inhab-
ited a wide range of environments along the different shrimp, and most of them had a
narrow range of occupancy across different environments (Fig. 2A), suggesting that
microbes with high niche breadth had greater odds of being core microbes. To address
this speculation, we used a Venn diagram to analyze the core microbes that exist in ev-
ery environment and found 55 microbes that fell into this denition (Fig. 2B). Notably,
when we examined the correspondence between Venn diagram and niche breadth mea-
surement, we found some inconformity. The niche breadth of some core microbes was
lower, such as Alistipes,Rikenella,Erysipelotrichaceae_uncultured, and so on (Fig. 2C),
which was due to their uneven distribution in different environments. The abundance of
FIG 1 Microbial diversity in the gut among different species of shrimp. (A) The relative abundance of the microbial communities at the genus level found
in the guts (Macrobrachium rosenbergii,Procambarus clarkii,Marsupenaeus japonicus, and Litopenaeus vannamei) of different shrimp that are from seawater
or freshwater. (B) Principal-coordinate analysis of samples based on the weighted UniFrac method. (C) The
a
-diversity comparison between different
species of shrimp (n= 3). Signicant differences are indicated by asterisks (*,P,0.05; **,P,0.01).
Potential Immune Function of Microbiota in Shrimp Gut Microbiology Spectrum
March/April 2022 Volume 10 Issue 2 10.1128/spectrum.02465-21 3
FIG 2 Fifty-ve microbes were found as core microbes that persist across the guts of all shrimp species. (A) Niche breadth of the overall microbial
communities calculated using Levins measure. The larger the niche breadth, the more generalized the microbe. (B) Venn diagram showing the unique and
shared bacterial genera in the gut between four shrimp species. At the genus level, 55 microbes that were shared among all habitats. (C) The niche
breadth of each microbe, in descending order. The listed microbes are a part of microbes that are found in the guts of all shrimp (core microbe). (D) Heat
map showing the abundance of each core microbe across different shrimp guts at the genus level. The shade of color indicates the abundance of
microbes. (E) Proportion of core microbes and specic microbes in the guts of different shrimp species.
Potential Immune Function of Microbiota in Shrimp Gut Microbiology Spectrum
March/April 2022 Volume 10 Issue 2 10.1128/spectrum.02465-21 4
these bacteria could reach up to 9% in Procambarus clarkii but less than 1% in the intes-
tines of the other three shrimp species (Fig. 2D). The situation showed that it was the dif-
ferent species of shrimp that shapes various gut microbial communities. Even so, some
microbes will not disappear due to this inuence, and they were consistently present in
the guts of shrimp, although their abundance varied greatly. In this term, 55 such
microbes were identied as core microbes in the gut of shrimp (Fig. 2D), and others
were identied as specic microbes. Although these microbes contributed almost 90%
(except for core microbe in Macrobrachium rosenbergii that was due to differences in the
dominant microbes) of the total relative abundance, they comprised less than 10% of
the overall richness (Fig. 2E). The distribution of these microbes clearly varied in guts of
different species.
Abundance of core microbes in the shrimp gut is changed by WSSV infection. It
is recognized that the gut microbiota is related to pathogen infection in shrimp, but
detailed study of the relationship between core microbiota and pathogen infection
and their causal roles have not been clearly elucidated. Therefore, WSSV, which is a
prevalent viral pathogen and able to cause extremely high mortality in shrimp, was
used to explore the issue in this study. Based on the determined core microbiota in the
shrimp gut, we analyzed the inuence of WSSV infection on core microbial abundance.
We found that the abundance of core gut microbes in Procambarus clarkii had the
greatest disturbance after virus infection, with nearly half of the core microbial abun-
dance signicantly changed (P,0.01; Fig. 3). Additionally, a Sankey diagram showed
some differences in the abundance change of core gut microbes between shrimp from
seawater and shrimp from freshwater after WSSV infection (Fig. 3A). The most obvious
distinction was in the abundance of Vibrio and Photobacterium. After virus infection,
the abundance of Vibrio in the guts of seawater shrimp decreased signicantly, and
the abundance of Photobacterium increased signicantly, while the changes in the gut
of freshwater shrimp were opposite (Fig. 3A). A similar condition was seen in other
microorganisms, such as Citrobacter,Lactobacillus,Bacteroides, and so on. The abun-
dance changes induced by WSSV infection were consistent in some core microorgan-
isms, such as Achromobacter,Chryseobacterium, and Flavobacterium (Fig. 3B). Although
the changes were signicant, their abundance in the guts of shrimp were relatively
low, so the response to virus infection may be small (Fig. 3B). Notably, the taxonomic
composition of the microbial community in shrimp after virus infection showed distinct
successional trajectories (Fig. 3C), with all microbial communities of the gut developing
toward the rst and fourth quadrants of the principal-coordinate analysis (PCoA), sug-
gesting that the responses of gut microorganisms among different shrimp to viral
infection may eventually follow the same trend.
Virus infection disrupted the initial metabolism of the shrimp gut. As reported,
the gut microbiota plays a major role in amino acid metabolism, lipid metabolism, pro-
tein digestion, and the fermentation of complex carbohydrates into short-chain fatty
acids (SCFAs) that are important for the health of organisms (26, 27). Hence, the micro-
bial composition obtained by 16S rRNA gene sequencing was used to predict the Kyoto
Encyclopedia of Genes and Genomes (KEGG) metabolic pathways that are involved, and
the differences between different samples and groups were analyzed. Based on explor-
ing the proportions of each KEGG metabolic pathway (level 2), we found some discrep-
ancies between infected and uninfected shrimp. These discrepancies had some com-
monalities in the changes of each species of shrimp after treatment with WSSV (Fig. 4).
The proportions of 12 pathways were altered in the guts of the four shrimp after virus
infection, including biosynthesis of other secondary metabolites, carbohydrate metabo-
lism, cell growth and death, the endocrine system, energy metabolism, metabolism of
terpenoids and polyketides, the immune system, infectious diseases, lipid metabolism,
membrane transport, and metabolism of cofactors and vitamins and signal transduction
(Fig. 4). Some of these pathways have been previously reported to be related to host me-
tabolism and anti-infection and anti-inammation processes (20). These results reect
metabolic changes that may be caused by alterations in the microbiome of the shrimp
Potential Immune Function of Microbiota in Shrimp Gut Microbiology Spectrum
March/April 2022 Volume 10 Issue 2 10.1128/spectrum.02465-21 5
gut after viral infection, indicating that the response of the core microbiota to viral stim-
ulation in different shrimp intestines was consistent.
Alterations in the gut metabolome by microbiota function in maintaining
shrimp immune homeostasis. By altering the structure of the gut microbiota, infection
with WSSV may alter the function of the community. This change in function is reected
in the gut metabolome, which includes both host- and microbial-derived metabolites. To
determine which metabolites changed in the guts of shrimp after viral infection and par-
ticipate in the anti-infection response, the gut metabolome of shrimp was explored using
mass spectrometry platforms with an untargeted approach. Shrimp intestinal contents
from different time points after infection with WSSV were used to characterize the
changes of the gut metabolome, and the top 50 metabolites with maximum fold change
and signicant changes were selected for display (Fig. 5A). Most metabolites were seen
with signicant decreases after WSSV infection, some of them varying by more than
50-fold, such as 4-dimethylallyl-L-tryptophan, bactoprenyl diphosphate, gymnodi-
mine,andsoon(Fig.5A).Themostsignicant increase (with 5-fold to 11-fold) was
seen in nine metabolites, including toluene-cis-dihydrodiol, eicosapentaenoic acid,
FIG 3 The abundance of core gut microorganisms changed after shrimp were infected with WSSV. (A) Sankey charts of three time points from the core
microbial community of the shrimp gut after WSSV infection. The taxonomy level, including phylum and genus, is displayed. The top 10 most abundant
genera and the relative changes over time are visible. (B) Balloon plot showing the signicant changes of core gut microbial abundance across different
shrimp species after WSSV infection. The size of the circles represents the change of bacterial abundance, while the color indicates increase or decline
(n=3;*,P,0.05 and **,P,0.01). (C) Principal-coordinate analysis of samples after WSSV infection.
Potential Immune Function of Microbiota in Shrimp Gut Microbiology Spectrum
March/April 2022 Volume 10 Issue 2 10.1128/spectrum.02465-21 6
(1)-carvone, myxalamid S, N-nitrosodimethylamine, oplophorus luciferin, traumatic
acid, 4-hydroxyretinoic acid, and 6-hydroxy-3,7-dimethyloctanoate, suggesting that
they have functional potential in anti-infection of WSSV in the shrimp gut (Fig. 5A).
Metabolites that change signicantly mostly belonged to 18 KEGG metabolic path-
ways compared with the initial states, such as secondary bile acid biosynthesis, lino-
leic acid metabolism, biosynthesis of unsaturated fatty acids, necroptosis, and the
AMP-activated protein kinase (AMPK) signaling pathway that are correlated with the
immunity and health of organisms, consistent with what the sequencing predicted
(Fig. 4 and 5B). Additionally, a correlation analysis conducted with the Spearman
algorithm found that the alteration of the gut metabolome was tightly correlated
with the changes of the core gut microbiota (Fig. 5A). For example, nine signicantly up-
regulated metabolites after infection strongly positively correlate with half of the core
microbes of the gut, while those signicantly downregulated metabolites have a strong
positive correlation with the rest of the microbes (Fig. 5A). These results showed that de-
spite that the decrease in the large number of metabolites and the enriched KEGG path-
way that they belonged to resulted in a reduction in shrimp immunity and health after
WSSV infection, the increase of some antiviral metabolites under the inuence of gut
microbes may assist host adaptation to environmental challenge.
DISCUSSION
In hosts, multiple factors shape the diversity of the gut microbiome, such as the de-
velopmental cycle, dietary differences, intestinal pH, and geographical location of the
host (28). The microorganisms that are prevalent among different hosts regardless of
these factors are dened as core microbes (8). A functional core microbiota was pro-
vided by abundant bacterial taxa. To date, identications of core gut microbiota typi-
cally focus on vertebrates, and information about those of invertebrates is very limited,
especially in aquatic invertebrates. Thus, shrimp, an essential invertebrate in global
aquatic aquaculture (29) were used to explore the core microbiota in our study. Based
on 16S rRNA gene sequencing, the gut microbial composition of four different species
of shrimp was determined, and we found that the gut environment of different shrimp
is a strong habitat lter for the microbial community, resulting in the high diversity of
the gut microbial community among different species of shrimp. By calculating the
niche breadth of microorganisms and analyzing their presence or not in different guts,
a core gut microbiota containing 55 genera was identied. They were present in the guts
FIG 4 Scatter plot showing the alterations of KEGG metabolic pathways among four species of shrimp after virus infection. Purple, green, red, and blue
represent Marsupenaeus japonicus,Litopenaeus vannamei,Macrobrachium rosenbergii, and Procambarus clarkia, respectively.
Potential Immune Function of Microbiota in Shrimp Gut Microbiology Spectrum
March/April 2022 Volume 10 Issue 2 10.1128/spectrum.02465-21 7
of each shrimp species regardless of the environment changes, even though their abun-
dance varies (inhomogeneity) from high to low. Therefore, our study provides a character-
ization of the core microbiota among different species of shrimp for the rst time, and we
speculate that they perform a vital role in host response to environmental stimuli.
Recent large-scale studies have provided insights into gut microbial structure and
functional potential (30). As reported, the gut microbiota profoundly inuences host
tness, and it is able to inuence various host physiological processes by regulating
multiple processes, including nutrient absorption, immune function, oxidative stress,
inammation, and anabolic balance, when the host is stimulated by the environment
(31, 32). The gut microbiota performs its function on some different landscapes in the
host, including metabolic, protective, structural, and neurological functions. For
instance, some gut microbial species are the main producers of short-chain fatty acids
FIG 5 Changes in the intestinal metabolome of Marsupenaeus japonicus after WSSV infection. (A) Gut metabolites with signicant changes after treatment
with WSSV and their correlation with gut core microbiota. The Z-score = (x2
m
)/
s
, where xrepresents a specic score,
m
represents the mean value, and
s
represents the standard deviation. Red boxes indicate positive correlations, while the blue boxes show negative correlations. (B) Enrichment of
metabolites in KEGG pathways in response to WSSV infection. The color of the dot represents the Pvalue, and the bluer the dot is, the more signicant
the enrichment is. The size of the dot represents rich number.
Potential Immune Function of Microbiota in Shrimp Gut Microbiology Spectrum
March/April 2022 Volume 10 Issue 2 10.1128/spectrum.02465-21 8
(SCFAs; formate, acetate, propionate, butyrate, valerate, isovalerate, and hexanoate) and
function in biodegradation of various undigested organics, bile salt, choline, polyphe-
nols, and so on (3335). Activities of the gut microbiota have an inuential role in modu-
lating the gut-brain axis and in maintaining gut homeostasis in the human body via pro-
duction, expression, and turnover of these metabolites (3638). Additionally, activation
of goblet cells to secrete mucin is associated with fucose, a product that is cleaved from
glycans by gut microbes (39). In shrimp, biolm formation by gut bacteria was found to
be related to regulating homeostasis in invertebrates (40). Shrimp white feces syndrome
was also reported to be related to intestinal microbiota dysbiosis (16). Wangsstudyhad
shown that the growth and developmental of shrimp is accompanied by alterations in
the gut microbiota, and some pivotal microbes are crucial in the growth of shrimp (24).
However, little is known about the response of the gut microbiota to environmental
changes, such as viral infection, and its effects on the host. Herein, we found that the
core gut microbiota in different species of shrimp adjusts and develops toward the same
trend when the host is exposed to virus infection. Such an alteration ultimately leads to
downregulation of a large number of metabolites in the gut when the shrimp was
infected with viruses, and these metabolites are involved in multiple immune-related
pathways. However, the upregulation of nine metabolites and the closely correlated
core microbes suggests that they have antiviral function potential and play an important
role in host resistance to viral stimulation. This reects an adaptive adjustment of gut
microbes to the environmental challenges of shrimp, which may be manifested as a
coadaptation of the host and gut microbes under environmental challenges.
Conclusions. In this study, WSSV infection was used as an environmental challenge
for four different species of shrimp (Marsupenaeus japonicus,Litopenaeus vannamei,
Macrobrachium rosenbergii, and Procambarus clarkii). By using 16S rRNA gene sequenc-
ing and liquid chromatography-coupled mass spectrometry (LC-MS), the gut micro-
biota and metabolome before and after virus infection were identied. Our ndings
provided the rst attempt to compare the gut microbiota among the four different
species of shrimp (Marsupenaeus japonicus,Litopenaeus vannamei,Macrobrachium
rosenbergii, and Procambarus clarkii), found huge variations, and determined a core gut
microbiota composed of 55 microbes. The environmental challenge of WSSV infection
led to changes in core microbial structures, but the alteration of core microbiota
among different shrimp species followed the same trend and showed immune-related
function in the prediction of its metabolic potential. In metabolomic analysis, nine sig-
nicantly upregulated metabolites that were found after viral infection suggested that
they have antiviral potential. Moreover, the tight correlation between them and almost
half of the core microbiota demonstrated that they were responsible for maintaining
immune homeostasis. These valuable ndings greatly enhanced our understanding of
the gut microbiota in maintaining host tness under environmental challenge and pro-
vide a new strategy for prevention and treatment of viral infection in shrimp.
MATERIALS AND METHODS
Shrimp culture and WSSV infection. Shrimp were cultured as previously described (18), and three
from each group were randomly selected for PCR detection of WSSV with specic primers (59-
TTGGTTTCATGCCCGAGATT-39and 59-CCTTGGTCAGCCCCTTGA-39) to ensure that the shrimp used for
experiments were WSSV free. WSSV-free shrimp were infected with WSSV (10
5
copies/mL) by injection
(100
m
L of WSSV inoculum/shrimp) into the lateral area of the fourth abdominal segment. The WSSV-
infected shrimp were collected for later experiments at different times after infection.
Sample collection and DNA extraction. Three shrimp were randomly selected from each group for
aseptically collecting intestines after sterilizing the surface of shrimp with 70% ethanol. The intestine
was dissected using sterile instruments, and microbial DNA was isolated from gut samples using the
bacterial genome DNA extraction kit (Generay, China) following the manufacturers protocols.
Sequencing and data analysis of microbial 16S rRNA. Amplicon sequencing covering the V4-V5
regions of bacterial 16S rRNA gene was performed by Mingke Biotechnology Co., Ltd. (Hangzhou, China), using
universal bacterial primers 515F (59-GTGCCAGCMGCCGCGG-39) and 907R (59-CCGTCAATTCMTTTRAGTTT-39).
Sequencing was performed using an Illumina PE250 (Illumina, USA), and the barcoded library was constructed
using an Illumina TruSeq DNA library kit (Illumina, USA) (41). Sequencing data were uploaded to NCBI
(GenBank accession number PRJNA780955).
The paired-end reads were overlapped to assemble the sequences using the Flash program. After
Potential Immune Function of Microbiota in Shrimp Gut Microbiology Spectrum
March/April 2022 Volume 10 Issue 2 10.1128/spectrum.02465-21 9
removal of low-quality fragments, spacers, primers, and the sequences shorter than 50 bp, the remaining
sequences were denoised and screened for chimeric sequences with the pre.cluster command and
chimera.uchime command in Mothur. The candidate sequences were classied into operational taxo-
nomic units (OTUs) by 97% sequence similarity using the Usearch program.
Principal-coordinates analysis. Beta diversity was evaluated by principal-coordinates analysis (PCoA)
plots based on unweighted UniFrac metrics using the vegan of package R (version 3.4.4; https://www.r
-project.org/). The potential principal components that affect the difference of sample community compo-
sition were found out through dimension reduction based on Euclidean distance and other distances.
Alpha diversity analysis. Alpha diversity includes a series of statistical analysis indices to estimate
the species abundance and diversity of the environmental community. Community richness was calcu-
lated using the following indices: Chao (http://www.mothur.org/wiki/Chao) and Ace (http://www
.mothur.org/wiki/Ace). The indices used to calculate community diversity were Shannon (http://www
.mothur.org/wiki/Shannon) and Simpson (http://www.mothur.org/wiki/Simpson).
The calculation of niche breadth. The niche breadth was calculated as the formula described. The
core bacteria inhabit a wide range of environments along the different samples, such as environmental
types, and specic bacteria have a narrower range of occupancy across these different environments. B
i
represents niche breadth (1 #B
i
#n), nrepresents the number of habitats, and irepresents the microbial
genus. P
ij
is the ratio of genus iin the nth habitat to the total number of this genus in all habitats (25).
Bi¼1=X
n
j¼1
Pij
ðÞ
2
Pij ¼nij=Pi1
Functional prediction by PICRUSt2. The PICRUSt2 (phylogenetic investigation of communities by
reconstruction of observed states, v2.1.0-b) pipeline was used to predict functional potentials of the gut
microbiota. Functional proles were predicted using the script picrust2_pipeline.py, generating a table
of KEGG orthologs (KOs). KEGG Mapper was used to reconstruct KEGG reference categories (KEGG level
1) and modules (KEGG level 2) according to the KO annotations.
Metabolome analysis of shrimp intestinal contents based on LC-MS. Three shrimp were randomly
selected from each group, and their intestines were dissected. Intestines (100 mg) were diluted with 1 mL
of a mixture of methanol-acetonitrile-water (2:2:1 [vol/vol]), followed by centrifugation at 13,000 rpm for
15 min at 4°C. Subsequently, 100
m
L of the supernatant was harvested for liquid chromatography-coupled
mass spectrometry (LC-MS) analysis. The metabolic proles were performed by Mingke Biotechnology Co.,
Ltd. (Hangzhou, China), on an Agilent 1290 Innity LC system and Accurate-Mass QTOF/MS-6545 (Agilent
Technologies, USA). For chromatographic separation, a C
18
(2.1 mm 100 mm) reversed-phase column
(Thermo Scientic, USA) preheated at 35°C was used. A prepared sample of 1
m
L was injected and main-
tained at 35°C for analysis. The gradient conditions for elution were 95% acetonitrile for 2 min, 95 to 90%
from 2 to 3 min of linear gradient, 90 to 30% from 3 to 9 min, 10% from 10 to 12 min, 10 to 95% from 12
to 12.1 min, and 95% from 12.1 to 14 min. The mobile phase for negative ion mode (ES2) and positive ion
mode (ES1) was composed of water with 0.04% formic acid as solvent A and acetonitrile with 0.04% for-
mic acid as solvent B, and the ow rate was at 0.3 mL/min.
Metabolome data processing. The original data were converted into m/zformat by ProteoWizard,
and peak detection, alignment, and retention time correction were carried out by the XCMS program.
The SVRmethod was used to correct the peak positions, and the peaks with a loss rate of .50% in
each group were ltered. After correcting the screened peaks, the metabolite identication was
obtained by searching the Metlin metabolite database. Statistical analysis was performed by the R
program.
KEGG annotation of differential metabolites. Different metabolites interact with each other in
organisms and form different pathways. The KEGG database was used to annotate the metabolic path-
ways of the differential metabolites involved. KEGG pathway enrichment was conducted according to
the results of differential metabolites. Metabolites with signicant alterations between groups were
dened as differential metabolites and were obtained at a variable inuence on projection (VIP) of .1.5,
with a Pvalue of ,0.05 (ttest statistics) based on the peak intensities.
Data availability. The data we obtained from next-generation sequencing were uploaded to the
NCBI database under GenBank accession number PRJNA780955.
ACKNOWLEDGMENTS
This work was supported by National Natural Science Foundation of China (32102832)
and China Postdoctoral Science Foundation (2021M701795).
REFERENCES
1. Lynch SV, Pedersen O. 2016. The human intestinal microbiome in health
and disease. N Engl J Med 375:23692379. https://doi.org/10.1056/
NEJMra1600266.
2. Adelman MW, Woodworth MH, Langelier C, Busch LM, Kempker JA, Kraft
CS, Martin GS. 2020. The gut microbiomes role in the development, main-
tenance, and outcomes of sepsis. Crit Care 24:278. https://doi.org/10
.1186/s13054-020-02989-1.
3. Bäckhed F, Roswall J, Peng Y, Feng Q, Jia H, Kovatcheva-Datchary P, Li Y,
XiaY,XieH,ZhongH,KhanMT,ZhangJ,LiJ,XiaoL,Al-AamaJ,ZhangD,
Lee YS, Kotowska D, Colding C, Tremaroli V, Yin Y, Bergman S, Xu X,
Madsen L, Kristiansen K, Dahlgren J, Wang J, Jun W. 2015. Dynamics and
stabilization of the human gut microbiome during the rst year of life.
Cell Host Microbe 17:690703. https://doi.org/10.1016/j.chom.2015.04
.004.
Potential Immune Function of Microbiota in Shrimp Gut Microbiology Spectrum
March/April 2022 Volume 10 Issue 2 10.1128/spectrum.02465-21 10
4. Donaldson GP, Lee SM, Mazmanian SK. 2016. Gut biogeography of the
bacterial microbiota. Nat Rev Microbiol 14:2032. https://doi.org/10.1038/
nrmicro3552.
5. Shaani Y, Zehavi T, Eyal S, Miron J, Mizrahi I. 2018. Microbiome niche mod-
ication drives diurnal rumen community assembly, overpowering indi-
vidual variability and diet effects. ISME J 12:24462457. https://doi.org/10
.1038/s41396-018-0203-0.
6. Pacheco AR, Moel M, Segrè D. 2019. Costless metabolic secretions as driv-
ers of interspecies interactions in microbial ecosystems. Nat Commun 10:
103. https://doi.org/10.1038/s41467-018-07946-9.
7. Coyte KZ, Schluter J, Foster KR. 2015. The ecology of the microbiome: net-
works, competition, and stability. Science 350:663666. https://doi.org/10
.1126/science.aad2602.
8. Kokou F, Sasson G, Friedman J, Eyal S, Ovadia O, Harpaz S, Cnaani A,
Mizrahi I. 2019. Core gut microbial communities are maintained by bene-
cial interactions and strain variability in sh. Nat Microbiol 4:24562465.
https://doi.org/10.1038/s41564-019-0560-0.
9. Astudillo-García C, Bell JJ, Webster NS, Glasl B, Jompa J, Montoya JM,
Taylor MW. 2017. Evaluating the core microbiota in complex commun-
ities: a systematic investigation. Environ Microbiol 19:14501462. https://
doi.org/10.1111/1462-2920.13647.
10. Shabat SK, Sasson G, Doron-Faigenboim A, Durman T, Yaacoby S, Berg
Miller ME, White BA, Shterzer N, Mizrahi I. 2016. Specic microbiome-de-
pendent mechanisms underlie the energy harvest efciency of rumi-
nants. ISME J 10:29582972. https://doi.org/10.1038/ismej.2016.62.
11. Shade A, Handelsman J. 2012. Beyond the Venn diagram: the hunt for a
core microbiome. Environ Microbiol 14:412. https://doi.org/10.1111/j
.1462-2920.2011.02585.x.
12. Fung TC, Olson CA, Hsiao EY. 2017. Interactions between the microbiota,
immune and nervous systems in health and disease. Nat Neurosci 20:
145155. https://doi.org/10.1038/nn.4476.
13. Vaarala O. 2012. Is the origin of type 1 diabetes in the gut? Immunol Cell
Biol 90:271276. https://doi.org/10.1038/icb.2011.115.
14. Takiishi T, Fenero CIM, Câmara NOS. 2017. Intestinal barrier and gut
microbiota: shaping our immune responses throughout life. Tissue Bar-
riers 5:e1373208. https://doi.org/10.1080/21688370.2017.1373208.
15. Sommer F, Bäckhed F. 2013. The gut microbiotamasters of host devel-
opment and physiology. Nat Rev Microbiol 11:227238. https://doi.org/
10.1038/nrmicro2974.
16. Huang Z, Zeng S, Xiong J, Hou D, Zhou R, Xing C, Wei D, Deng X, Yu L,
Wang H, Deng Z, Weng S, Kriengkrai S, Ning D, Zhou J, He J. 2020. Micro-
ecological Kochs postulates reveal that intestinal microbiota dysbiosis
contributes to shrimp white feces syndrome. Microbiome 8:32. https://
doi.org/10.1186/s40168-020-00802-3.
17. Li C, Wang S, He J. 2019. The two NF-
k
B pathways regulating bacterial
and WSSV infection of shrimp. Front Immunol 10:1785. https://doi.org/10
.3389/mmu.2019.01785.
18. Zhang S, Xin F, Zhang X. 2021. The compound packaged in virions is the
key to trigger host glycolysis machinery for virus life cycle in the cyto-
plasm. iScience 24:101915. https://doi.org/10.1016/j.isci.2020.101915.
19. Lo CF, Hsu HC, Tsai MF, Ho CH, Peng SE, Kou GH, Lightner DV. 1999. Spe-
cic genomic DNA fragment analysis of different geographical clinical
samples of shrimp white spot syndrome virus. Dis Aquat Org 35:175185.
https://doi.org/10.3354/dao035175.
20. Vatanen T, Kostic AD, d'Hennezel E, Siljander H, Franzosa EA, Yassour M,
Kolde R, Vlamakis H, Arthur TD, Hämäläinen AM, Peet A, Tillmann V, Uibo
R, Mokurov S, Dorshakova N, Ilonen J, Virtanen SM, Szabo SJ, Porter JA,
Lähdesmäki H, Huttenhower C, Gevers D, Cullen TW, Knip M, DIABIM-
MUNE Study Group, Xavier RJ. 2016. Variation in microbiome LPS immu-
nogenicity contributes to autoimmunity in humans. Cell 165:842853.
https://doi.org/10.1016/j.cell.2016.04.007.
21. Adak A, Khan MR. 2019. An insight into gut microbiota and its functionalities.
Cell Mol Life Sci 76:473493. https://doi.org/10.1007/s00018-018-2943-4.
22. Cummings JH, Pomare EW, Branch WJ, Naylor CP, Macfarlane GT. 1987.
Short chain fatty acids in human large intestine, portal, hepatic and ve-
nous blood. Gut 28:12211227. https://doi.org/10.1136/gut.28.10.1221.
23. Guri M, Durand L, Cueff-Gauchard V, Zbinden M, Crassous P, Shillito B,
Cambon-Bonavita MA. 2012. Acquisition of epibiotic bacteria along the
life cycle of the hydrothermal shrimp Rimicaris exoculata. ISME J 6:
597609. https://doi.org/10.1038/ismej.2011.133.
24. Wang Y, Wang K, Huang L, Dong P, Wang S, Chen H, Lu Z, Hou D, Zhang D.
2020. Fine-scale succession patterns and assembly mechanisms of bacterial
community of Litopenaeus vannamei larvae across the developmental
cycle. Microbiome 8:106. https://doi.org/10.1186/s40168-020-00879-w.
25. Levins R. 1968. Evolution in changing environments: some theoretical
explorations. Princeton University Press, Princeton, NJ.
26. Dai ZL, Wu G, Zhu WY. 2011. Amino acid metabolism in intestinal bacteria:
links between gut ecology and host health. Front Biosci (Landmark Ed)
16:17681786. https://doi.org/10.2741/3820.
27. Swann JR, Want EJ, Geier FM, Spagou K, Wilson ID, Sidaway JE, Nicholson
JK, Holmes E. 2011. Systemic gut microbial modulation of bile acid me-
tabolism in host tissue compartments. Proc Natl Acad Sci U S A 108:
45234530. https://doi.org/10.1073/pnas.1006734107.
28. Kim S, Jazwinski SM. 2018. The gut microbiota and healthy aging: a mini-
review. Gerontology 64:513520. https://doi.org/10.1159/000490615.
29. Cao L, Naylor R, Henriksson P, Leadbitter D, Metian M, Troell M, Zhang W.
2015. Chinas aquaculture and the worlds wild sheries. Science 347:
133135. https://doi.org/10.1126/science.1260149.
30. Rosshart SP, Vassallo BG, Angeletti D, Hutchinson DS, Morgan AP, Takeda
K, Hickman HD, McCulloch JA, Badger JH, Ajami NJ, Trinchieri G, Pardo-
Manuel de Villena F, Yewdell JW, Rehermann B. 2017. Wild mouse gut
microbiota promotes host tness and improves disease resistance. Cell
171:10151028. https://doi.org/10.1016/j.cell.2017.09.016.
31. Ticinesi A, Nouvenne A, Cerundolo N, Catania P, Prati B, Tana C, Meschi T.
2019. Gut microbiota, muscle mass and function in aging: a focus on
physical frailty and sarcopenia. Nutrients 11:1633. https://doi.org/10
.3390/nu11071633.
32. Groussin M, Mazel F, Alm EJ. 2020. Co-evolution and co-speciation of
host-gut bacteria systems. Cell Host Microbe 28:1222. https://doi.org/10
.1016/j.chom.2020.06.013.
33. Louis P, Flint HJ. 2009. Diversity, metabolism and microbial ecology of bu-
tyrate-producing bacteria from the human large intestine. FEMS Micro-
biol Lett 294:18. https://doi.org/10.1111/j.1574-6968.2009.01514.x.
34. Portune KJ, Beaumont M, Davila AM, Tome D, Blachier F, Sanz Y. 2016.
Gut microbiota role in dietary protein metabolism and health-related out-
comes: the two sides of the coin. Trends Food Sci Technol 57:213232.
https://doi.org/10.1016/j.tifs.2016.08.011.
35. Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, Pettersson
S. 2012. Host-gut microbiota metabolic interactions. Science 336:
12621267. https://doi.org/10.1126/science.1223813.
36. Romano KA, Vivas EI, Amador-Noguez D, Rey FE. 2015. Intestinal micro-
biota composition modulates choline bioavailability from diet and accu-
mulation of the proatherogenic metabolite trimethylamine-N-oxide.
mBio 6:e02481. https://doi.org/10.1128/mBio.02481-14.
37. Most J, Penders J, Lucchesi M, Goossens GH, Blaak EE. 2017. Gut micro-
biota composition in relation to the metabolic response to 12-week com-
bined polyphenol supplementation in overweight men and women. Eur J
Clin Nutr 71:10401045. https://doi.org/10.1038/ejcn.2017.89.
38. Sarkar A, Lehto SM, Harty S, Dinan TG, Cryan JF, Burnet PWJ. 2016. Psycho-
biotics and the manipulation of bacteria-gut-brain signals. Trends Neuro-
sci 39:763781. https://doi.org/10.1016/j.tins.2016.09.002.
39. Xu J, Bjursell MK, Himrod J, Deng S, Carmichael LK, Chiang HC, Hooper LV,
Gordon JI. 2003. A genomic view of the human-Bacteroides thetaiotaomi-
cron symbiosis. Science 299:20742076. https://doi.org/10.1126/science
.1080029.
40. Zhang YX, Zhang ML, Wang XW. 2021. C-Type lectin maintains the home-
ostasis of intestinal microbiota and mediates biolm formation by intesti-
nal bacteria in shrimp. J Immunol 206:11401150. https://doi.org/10
.4049/jimmunol.2000116.
41. Sun X, Yang J, Zheng M, Zhang X. 2021. Articial construction of the bio-
coenosis of deep-sea ecosystem via seeping methane. Environ Microbiol
23:11861198. https://doi.org/10.1111/1462-2920.15347.
Potential Immune Function of Microbiota in Shrimp Gut Microbiology Spectrum
March/April 2022 Volume 10 Issue 2 10.1128/spectrum.02465-21 11
... But commensals can also directly impact bacteria (either true pathogen or opportunistic ones) through interference with their entry and installation by direct competition or production of antimicrobial compounds. The cnidarian Hydra vulgaris is the aquatic invertebrate model for which the beneficial effect of the microbiota has been largely addressed and a substantial number of works clearly shows that the mucosal innate immunity is shaped by host-microbiota interactions (Schröder and Bosch 2016 been shown in different shrimp species where a higher diversity of metabolites with potential beneficial properties are produced by half of the core microbiota and likely contributes to their immune homeostasis (Zhang and Sun 2022). Otherwise, functional redundancy has been described in the more diverse microbial community of the yellow abalone compared to the blue abalone and this correlates with a lower prevalence of the Withering Syndrome disease . ...
... In order to prevent these infectious diseases, several approaches based on the use of micro- Several studies also have shown that a higher microbiota diversity is correlated with the health status of the host in both crustaceans and molluscs Holt et al. 2020;Zhang and Sun 2022). From this perspective, Fallet and colleagues decided to use the microbiota to educate the immune system of C. gigas. ...
Thesis
Full-text available
Recently, the frequency and severity of marine diseases have increased in association with global changes, andmollusks of economic interest are particularly concerned. A striking example of a devastating disease is the Pacific Oyster Mortality Syndrome(POMS) caused by the Ostreid Herpesvirus-1 μVar (OsHV-1 μVar) that emerged in 2008 and which heavily impacts Crassostrea gigasproduction worldwide by affecting juvenile oysters. Adult oysters are also affected by infectious diseases, especially those caused by thebacterial pathogen Vibrio aestuarianus.In my PhD, I investigated the possibility of using the beneficial effect of the oyster microbiota to fightagainst these infectious diseases. It has been reported that some bacterial strains are preferentially associated with oysters with better survivalcapacity to the POMS disease. In addition, previous work has shown that it is possible to educate the oyster immune system through an exposureto healthy microbiota during their larval development. This experiment had been achieved by using oyster donors of microbiota. This microbialeducation had the advantage of using all the potential of the whole microorganism diversity, but the transferred community were not fullycontrolled and could be hazardous. This made this whole microbiota exposure not applicable in aquaculture. In this sense, I decided to focus mywork on controlled microbial environment and investigated their potential beneficial effect in oyster health.I generated a collection of bacterialspecies from naturally disease-resistant C. gigas collected in the field, and I characterized their effects on oysters. I investigated two possiblebeneficial strategies: (1) I performed a microbial education by adding multi-strain bacterial mixes during larval development and investigated thelong-term effect of this exposure at juvenile and adult stages when oysters were challenged with OsHV-1 and V. aestuarianus. (2) I identifiedantimicrobial-producing bacterial strains and investigated their short-term effect by adding them to juvenile and adult oysters just before pathogenchallenges. Overall, I showed that both strategies can help fighting against oyster infectious diseases but strongly relies on oyster genetic.Thesefindings open new avenues for the development of microbiota-targeted prophylactic approaches to mitigate diseases in oyster farming.
... Enterococcus, the domain bacteria in the gut of C. medinalis, was the most enriched bacteria with no particular association with different treatment groups. The abundance of the domain bacteria was important to gut microbiota homeostasis [49]. Wolbachia was also the core gut bacteria of C. medinalis and more highly enriched at 24 hpi and 72 hpi in the mock-infected group but at 48 hpi in the CnmeGV-infected group. ...
Article
Full-text available
The composition of microbiota in the digestive tract gut is essential for insect physiology, homeostasis, and pathogen infection. Little is known about the interactions between microbiota load and oral infection with baculoviruses. CnmeGV is an obligative baculovirus to Cnaphalocrocis medinalis. We investigated the impact of CnmeGV infection on the structure of intestinal microbes of C. medinalis during the initial infection stage. The results revealed that the gut microbiota profiles were dynamically driven by pathogen infection of CnmeGV. The numbers of all the OTU counts were relatively higher at the early and later stages, while the microbial diversity significantly increased early but dropped sharply following the infection. The compositional abundance of domain bacteria Firmicutes developed substantially higher. The significantly enriched and depleted species can be divided into four groups at the species level. Fifteen of these species were ultimately predicted as the biomarkers of CnmeGV infection. CnmeGV infection induces significant enrichment of alterations in functional genes related to metabolism and the immune system, encompassing processes such as carbohydrate, amino acid, cofactor, and vitamin metabolism. Finally, the study may provide an in-depth analysis of the relationship between host microbiota, baculovirus infection, and pest control of C. medinalis.
... Since core ASVs were already present in the eggs and might have been kept throughout the whole life cycle, we can hypothesize that the larvae and maybe even the breeders have selected particular bacterial communities harboring specific functions to be beneficial for the animal's health and welfare. Indeed, members of the core microbiota might act as probiotics or be involved in immune homeostasis [66,67]. The putative functions and ecological activities of the core microbiota assigned with FRAPROTAX displayed different abundance profiles according to the larval stage ( Figure S4), while the correlogram indicated specific putative functions among the core microbiota according to the larval stage (Figure 7). ...
Article
Full-text available
During their entire lifecycle, mariculture animals are farmed in water that contains various microorganisms with which they are in close associations. Microbial exchanges between the animals and their surrounding water can occur. However, little is known about the interactions between shrimp larvae and water, and more especially, about larval bacterial selection and microbiota modulation across ontogeny. To address this gap, using HiSeq sequencing targeting the V4 region of the 16S rRNA molecule, we investigated the active prokaryotic diversity and structure of healthy Penaeus stylirostris larvae and seawater. Comparisons between different larval stages revealed evidence of stage-specific microbiotas and biomarkers, a core microbiota common to all stages, and shared taxa between successive stages, suggesting vertical transmission of bacterial taxa. Comparisons between stage-specific microbiotas and core microbiotas with water storages highlighted that many taxa associated with the larvae were originally present in the natural seawater, underlining horizontal transmission of bacteria from water to larvae. As some of these lineages became active at specific larval stages, we suggest that larvae were able to modulate their microbiota. This study provides insight into larvae-microbiota interactions at the larval stage scale.
... In different shrimp species, a greater diversity of metabolites with potential beneficial properties is produced by half of the core microbiota likely contributing to their immune homeostasis. 76 Additionally, functional redundancy has been described in the more diverse microbial community of the yellow abalone compared to the blue abalone correlating with a lower prevalence of the Withering Syndrome disease. 78,79 However, it's essential to note that while the mentioned studies are mostly correlative, only two of them have demonstrated a direct causality between healthy or diseased microbiota and health status. ...
Article
Full-text available
The holobiont theory expands the notion of individual multicellular organisms as a community composed of a host and all its associated microorganisms. This concept has been extensively studied in the field of aquaculture, where increasing evidence has highlighted the importance of the host associated microorganisms in species fitness. Here, we focus our review on mollusc and crustacean species in which micro-biota dysbiosis has recently been described in the context of various diseases, resulting in significant economic losses. Influencing the holobiont structure through the use of probiotics is a potential strategy that could improve the fitness or the robustness of cultivated species. We discuss here the possibility of developing micro-biome targeted prophylactic approaches by promoting (1) methods to identify host microbial community that fosters good health status and (2) early life microbial education to favour long-term resistance to stress or disease. This review aims to inform the aquaculture industry about potential strategies in rearing practices to mitigate diseases and economic losses.
... The microbiome analysis in WSSV-infected fresh water shrimp revealed the increase in relative abundance of Vibrio (Zhang and Sun 2022). Further, WSSV condition significantly increased the relative abundance of Propionigenium, Photobacterium, and Arcobacter and decreased the relative abundance of Flavobacterium and Candidatus bacilloplasma among the gut microbiota of L. vannamei. ...
Article
Full-text available
Aquaculture plays a vital role in meeting global food demand, and shrimp farming is a major contributor to this industry. Despite the immense economic importance of farmed shrimp, disease outbreaks continue to pose a significant challenge. This was attributed to fluctuations in various biotic and abiotic factors such as temperature, salinity, nutrition absorption, and water quality, which favor the abundance of invaders or opportunistic pathogens. Furthermore, the alterations in the gut microbiota, particularly decreased levels of commensals and increased prevalence of pathogens were reported during disease outbreaks. Shrimp physiology and immune function are intimately intertwined with the gut microbiome, leading to a dynamic interplay where the host shapes the microbial community through selection, while the microbes in turn influence the host’s metabolism and immune responses. Although various literatures report the change in gut microbial diversity and the immune responses generated by the host against various pathogens, a crucial knowledge gap exists regarding the intricate communication between these two systems. This review bridges this gap by deciphering the intricate interplay between host immunity and gut microbes, elucidating on the mechanisms underlying successful shrimp disease establishment. Thus, this review could offer possibilities for generating ideas to carry out effective aquaculture practices.
... The gut bacterial community is a microecosystem containing a diverse range of microorganisms, which play an important role in growth and development, nutrient metabolism and host immunity [20]. In the present study, the dominant bacterial species of A. japonicus exhibiting differential growth pattern were relatively similar, mainly concentrated in Bacteroidetes and Proteobacteria. ...
Preprint
Full-text available
Background Gut microbiome and their metabolites are increasingly recognized for their pivotal role in regulating the health and growth of the host. The mechanism by which the host-gut microbiome co-metabolism affects the growth rate of sea cucumber (Apostichopus japonicus) remains unclear. In this study, the intestinal wall tissues of fast-growing (FG) and slow-growing (SG) A. japonicus were collected from the full-sib family for 16S amplicon sequencing, metagenomics, metabolomics, and transcriptomics analyses. Results The results showed a marked variation in the gut bacterial community of A. japonicus with different growth rates. Compared with SG group, FG A. japonicus had a higher abundance of Bacteroidetes. Many of these bacteria were associated with the degradation of polysaccharides, which could potentially affect growth performance. The abundance of Proteobacteria, however, was found to be higher in SG A. japonicus, thereby increasing the potential risk of disease. The metabolome results were further supported by the predicated KEGG functions of the gut microbiomes, lots of metabolites related to the metabolism and biosynthesis of fructose 6-phosphate, arachidonic acid, oleic acid, and dihydroxyacetone phosphate were upregulated in the FG A. japonicus. Furthermore, the genes (IGF1, HK, PK, PCK1) were significantly upregulated in the FG A. japonicus, which were mainly enriched in the metabolic pathways of lipids, amino acids, and carbohydrates. Conclusions Gut bacterial community via their unique metabolic pathways, which in turn affected the metabolic phenotypes of the organism and eventually led to differences in growth performance of A. japonicus. These findings might help understand the molecular mechanisms of growth gap in sea cucumber, thereby increasing the mariculture production.
... However, the stomach microbiome of L. vannamei from two different ponds in the same study varied significantly suggesting the influence of culture conditions on the gut microbiome (Imaizumi et al. 2022). Zhang and Sun (2022) reported significant variations in the core gut microbiota among the four different species of shrimp. Challenge with WSSV resulted in alterations in core microbial structures; however, variation of core microbiota followed the same trend among different species and displayed immune-related function. ...
Chapter
Farmed shrimp are exposed to environmental conditions that are distinctly different from their natural habitat. Bacteria that colonize the shrimp surface and the intestine vary depending on the culture conditions as well as the health status of shrimp. From disease perspective, the gut microbiome of shrimp has been of intense focus in the recent times owing to its potential application in disease prediction and management. Gut being the focal point of various diseases in shrimp of known and unknown etiologies, microbiome studies using powerful next-generation sequencing (NGS) technologies can unravel the microbiome dynamics of shrimp gut under different pond conditions and the life stages of shrimp, and help understand their influence on the growth, physiology, immunity, and health status of shrimp. Disturbances in gut microbiome (dysbiosis), which predispose the shrimp, have been noticed in conditions such as early mortality syndrome (EMS)/acute hepatopancreatic necrosis disease (AHPND), slow/retarded growth, white feces syndrome (WFS), etc. As the composition of gut microbiome changes with the infection status of shrimp, identification of specific microbiota associated with a specific disease can serve as a useful diagnostic biomarker to predict the onset of disease. This can eventually help in disease management and decision-making. However, a multitude of factors in shrimp aquaculture system influence the gut microbiome of shrimp and, therefore, healthy gut microbiome can vary depending on the culture system and the environmental conditions. Nevertheless, microbiome studies can provide deeper insights into the etiology of various diseases and help to delineate the interactions between various factors, both biotic and abiotic, that determine the health of farmed shrimp.
Chapter
After the ban on the use of antibiotics in aquaculture farming, efforts are being made to find out other alternative methods which can replace antibiotics and augment aquaculture production safely without compromising the quality of the yield. Some of the alternative methods devised are the use of bacteriophage and biofloc technology and also the microbiome in the form of probiotics. But there is not much information regarding the use of gut microbiome species of fish and shellfish for the development of aquaculture farming/industry. There is a lot of potential for development in this particular area, provided the knowledge pertaining to the functional aspects and the ingredient molecules that are present in these microbiome species is available. Therefore, extensive efforts are needed to develop R and D programs on the metagenomics aspects of the gut microbiome species. In order to promote the future prospects of the bio industry, there is an urgent need for intensive research to be undertaken on fish and shellfish microbiomes, particularly in the areas of genomics and metagenomics. The Functionality aspects of the gut microbiome are an important issue for future investigation and therefore, in-depth knowledge of functional metagenomics is very much essential. Metagenomics has emerged in the last decade as a promising technology that attempts to analyze the complex genomes contained within a microbial niche.
Article
Full-text available
Viruses depend on the host metabolic machinery to complete their life cycle in the host cytoplasm. However, the key viral factors initiating the host machinery after the virus enters the cytoplasm remain unclear. Here, we found that compounds packaged in the virions of white spot syndrome virus, such as palmitic amide, could trigger the viral life cycle in the host cytoplasm. Palmitic amide promoted virus infection by enhancing host glycolysis by binding to triosephosphate isomerase to enhance its enzymatic activity. The glycolysis enhancement resulted in lactate accumulation, thereby promoting hypoxia-inducible factor 1 (HIF-1) expression. HIF-1 upregulation further enhanced glycolysis, which in turn promoted virus infection. Therefore, our study presented novel insight into the initiation of the virus life cycle in host cells.
Article
Full-text available
Mammalian gut microbiomes profoundly influence host fitness, but the processes that drive the evolution of host-microbiome systems are poorly understood. Recent studies suggest that mammals and their individual gut symbionts can have parallel evolutionary histories, as represented by their congruent phylogenies. These “co-phylogenetic” patterns are signatures of ancient co-speciation events and illustrate the cohesiveness of the mammalian host-gut microbiome entity over evolutionary times. Theory predicts that co-speciation between mammals and their gut symbionts could result from their co-evolution. However, there is only limited evidence of such co-evolution. Here, we propose a model that explains cophylogenetic patterns without relying on co-evolution. Specifically, we suggest that individual gut bacteria are likely to diverge in patterns recapitulating host phylogeny when hosts undergo allopatric speciation, limiting inter-host bacterial dispersal and genomic recombination. We provide evidence that the model is empirically grounded and propose a series of observational and experimental approaches to test its validity.
Article
Full-text available
Background: Microbiome assembly in early life may have a long-term impact on host health. Larval nursery is a crucial period that determines the success in culture of Litopenaeus vannamei, the most productive shrimp species in world aquaculture industry. However, the succession patterns and assembly mechanisms of larval shrimp bacterial community still lack characterization at a fine temporal scale. Here, using a high-frequency sampling strategy and 16S rRNA gene amplicon sequencing, we investigated dynamics of larval shrimp bacterial community and its relationship with bacterioplankton in the rearing water across the whole developmental cycle in a realistic aquaculture practice. Results: Alpha-diversity of larval shrimp bacteria showed a U-shaped pattern across the developmental cycle with the stages zoea and mysis as the valley. Correspondingly, the compositions of dominant bacterial taxa at the stages nauplius and early postlarvae were more complex than other stages. Remarkably, Rhodobacteraceae maintained the overwhelming dominance after the mouth opening of larvae (zoea I~early postlarvae). The taxonomic and phylogenetic compositions of larval bacterial community both showed stage-dependent patterns with higher rate of taxonomic turnover, suggesting that taxonomic turnover was mainly driven by temporal switching among closely related taxa (such as Rhodobacteraceae taxa). The assembly of larval bacteria was overall governed by neutral processes (dispersal among individuals and ecological drift) at all the stages, but bacterioplankton also had certain contribution during three sub-stages of zoea, when larval and water bacterial communities were most associated. Furthermore, the positive host selection for Rhodobacteraceae taxa from the rearing water during the zoea stage and its persistent dominance and large predicted contribution to metabolic potentials of organic matters at post-mouth opening stages suggest a crucial role of this family in larval microbiome and thus a potential source of probiotic candidates for shrimp larval nursery. Conclusions: Our results reveal pronounced succession patterns and dynamic assembly processes of larval shrimp bacterial communities during the developmental cycle, highlighting the importance of the mouth opening stage from the perspective of microbial ecology. We also suggest the possibility and potential timing in microbial management of the rearing water for achieving the beneficial larval microbiota in the nursery practice.
Article
Full-text available
The gut microbiome regulates a number of homeostatic mechanisms in the healthy host including immune function and gut barrier protection. Loss of normal gut microbial structure and function has been associated with diseases as diverse as Clostridioides difficile infection, asthma, and epilepsy. Recent evidence has also demonstrated a link between the gut microbiome and sepsis. In this review, we focus on three key areas of the interaction between the gut microbiome and sepsis. First, prior to sepsis onset, gut microbiome alteration increases sepsis susceptibility through several mechanisms, including (a) allowing for expansion of pathogenic intestinal bacteria, (b) priming the immune system for a robust pro-inflammatory response, and (c) decreasing production of beneficial microbial products such as short-chain fatty acids. Second, once sepsis is established, gut microbiome disruption worsens and increases susceptibility to end-organ dysfunction. Third, there is limited evidence that microbiome-based therapeutics, including probiotics and selective digestive decontamination, may decrease sepsis risk and improve sepsis outcomes in select patient populations, but concerns about safety have limited uptake. Case reports of a different microbiome-based therapy, fecal microbiota transplantation, have shown correlation with gut microbial structure restoration and decreased inflammatory response, but these results require further validation. While much of the evidence linking the gut microbiome and sepsis has been established in pre-clinical studies, clinical evidence is lacking in many areas. To address this, we outline a potential research agenda for further investigating the interaction between the gut microbiome and sepsis.
Article
Full-text available
Background: Recently, increasing evidence supports that some complex diseases are not attributed to a given pathogen, but dysbiosis in the host intestinal microbiota (IM). The full intestinal ecosystem alterations, rather than a single pathogen, are associated with white feces syndrome (WFS), a globally severe non-infectious shrimp disease, while no experimental evidence to explore the causality. Herein, we conducted comprehensive metagenomic and metabolomic analysis, and intestinal microbiota transplantation (IMT) to investigate the causal relationship between IM dysbiosis and WFS. Results: Compared to the Control shrimp, we found dramatically decreased microbial richness and diversity in WFS shrimp. Ten genera, such as Vibrio, Candidatus Bacilloplasma, Photobacterium, and Aeromonas, were overrepresented in WFS, whereas 11 genera, including Shewanella, Chitinibacter, and Rhodobacter were enriched in control. The divergent changes in these populations might contribute the observation that a decline of pathways conferring lipoic acid metabolism and mineral absorption in WFS. Meanwhile, some sorts of metabolites, especially lipids and organic acids, were found to be related to the IM alteration in WFS. Integrated with multiomics and IMT, we demonstrated that significant alterations in the community composition, functional potentials, and metabolites of IM were closely linked to shrimp WFS. The distinguished metabolites which were attributed to the IM dysbiosis were validated by feed-supplementary challenge. Both homogenous selection and heterogeneous selection process were less pronounced in WFS microbial community assembly. Notably, IMT shrimp from WFS donors eventually developed WFS clinical signs, while the dysbiotic IM can be recharacterized in recipient shrimp. Conclusions: Collectively, our findings offer solid evidence of the causality between IM dysbiosis and shrimp WFS, which exemplify the 'microecological Koch's postulates' (an intestinal microbiota dysbiosis, a disease) in disease etiology, and inspire our cogitation on etiology from an ecological perspective. Video abstract.
Article
Intestinal microbiota are closely related to host physiology. Over the long course of evolution and interaction, both commensal bacteria and their host have evolved multiple strategies to adapt to each other. However, in invertebrates, the regulatory mechanism of intestinal microbiota homeostasis is largely unknown. In the current study, a digestive tract-specific C-type lectin, designated as CTL33, was identified because of its abundance and response to bacteria in the intestine of kuruma shrimp (Marsupenaeus japonicus). Silencing of CTL33 expression led directly to intestinal dysbiosis, tissue damage, and shrimp death. CTL33 could facilitate biofilm formation by the intestinal bacteria. This function originated from its unique architecture, with a lectin domain responsible for bacteria recognition and a coiled coil region that mediated CTL33 dimerization and cross-linked the bacteria into a biofilm-like complex. By mediating the formation of a biofilm, CTL33 promoted the establishment of intestinal bacteria in intestine and maintained the homeostasis of the microbiota. Thus, to our knowledge, we demonstrated a new mechanism of C-type lectin-mediated biofilm formation by intestinal bacteria, providing new insights into intestinal homeostasis regulation in invertebrates.
Article
Deep-sea ecosystems, such as cold seeps and hydrothermal vents, have high biomass, even though they are located in the benthic zone, where no sunlight is present to provide energy for organism proliferation. Based on the coexistence of the reduced gases and chemoautotrophic microbes, it is inferred that the energy from the reduced gases supports the biocoenosis of deep-sea ecosystems. However, there is no direct evidence to support this deduction. Here, we developed and placed a biocoenosis generator, a device that continuously seeped methane, on the 1000-m deep-sea floor of the South China Sea to artificially construct a deep-sea ecosystem biocoenosis. The results showed that microorganisms, including bacteria and archaea, appeared in the biocoenosis generator first, followed by jellyfish and Gammaridea arthropods, indicating that a biocoenosis had been successfully constructed in the deep sea. Anaerobic methane-oxidizing archaea, which shared characteristics with the archaea of natural deep-sea cold seeps, acted as the first electron acceptors of the emitted methane; then, the energy in the electrons was transferred to downstream symbiotic archaea and bacteria and finally to animals. Nitrate-reducing bacteria served as partners to complete anaerobic oxidation of methane process. Further analysis revealed that viruses coexisted with these organisms during the origin of the deep-sea biocoenosis. Therefore, our study mimics a natural deep-sea ecosystem and provides the direct evidence to show that the chemical energy of reduced organic molecules, such as methane, supports the biocoenosis of deep-sea ecosystems.