PreprintPDF Available
Preprints and early-stage research may not have been peer reviewed yet.

Figures

Content may be subject to copyright.
Science of the Total Environment 900 (2023) 165809
Available online 26 July 2023
0048-9697/© 2023 Elsevier B.V. All rights reserved.
Bacteria pyruvate metabolism modulates AFB
1
toxicity in
Caenorhabditis elegans
Bowen Tang, Kathy S. Xue, Jia-Sheng Wang, Phillip L. Williams, Lili Tang
*
Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, GA 30602, USA
ARTICLE INFO
Editor: Lidia Minguez Alarcon
Keywords:
Host-microbe-chemical interaction
Aatoxin B
1
C. elegans
Engineered bacteria
Pyruvate metabolic pathway
ABSTRACT
Aatoxin B
1
(AFB
1
), the most potent mycotoxin and Group 1 human carcinogen, continues to pose a signicant
public health burden, particularly in developing countries. Increasing evidence has shown the gut microbiota as a
key mediator of AFB
1
toxicity through multiple interactive host-microbiota activities. In our previous study we
observed that disturbances in bacterial pyruvate metabolism might have a signicant impact on AFB
1
in the host.
To further investigate the impact of the pyruvate pathway on AFB
1
toxicity in C. elegans, we engineered two
bacterial strains (triple-overexpressed and triple-knockout strains with aceB, lpd, and pB). Additionally, we
employed two mutant worm strains (pyk-1 and pdha-1 mutants) known to affect pyruvate metabolism. Our
results revealed that the co-metabolism of pyruvate by the host and bacterial strains synergistically inuences
AFB
1
toxicity. Remarkable, we found that bacterial pyruvate metabolism, rather than that of the host, plays a
pivotal role in modulating AFB
1
toxicity in C. elegans. Our study sheds light on the role of gut microbiota involved
in pyruvate metabolism in inuencing AFB
1
toxicity in C. elegans.
1. Introduction
Aatoxin B
1
(AFB
1
) is the most toxic and hepatocarcinogenic
mycotoxin among the know mycotoxins produced by Aspergillus avus
and A. parasiticus and remains a crucial environmental contamination in
many developing countries (Creppy, 2002). AFB
1
can be metabolically
activated in the liver, which may cause acute hepatic toxicity and im-
mune toxicity, forming DNA adducts and inducing gene mutations, and
has been thereby categorized as Group I human carcinogen (Baan et al.,
2009). AFB
1
is well-documented to be a causative agent of hepatocel-
lular carcinoma (HCC), growth suppression, immune system modula-
tion, and malnutrition (Smoke and Smoking, 2004).
Recent evidence suggested that dietary exposure to AFB
1
can disrupt
the composition and function of gut microbiome, which potentially
inuencing its toxic effects (Chang et al., 2020; Wang et al., 2016; Zhou
et al., 2018). However, the underlying mechanism of the complex re-
lationships among host, gut microbes, and AFB
1
toxicity remains largely
unknown. Further investigation is needed to unravel the complex re-
lationships and shed light on the potential involvement of the gut
microbiome in AFB
1
toxicity.
The nematode Caenorhabditis elegans is an important model organism
in biomedical and toxicological research, mainly due to its well-
characterized genome, short reproductive life cycle, a large number of
offspring, and ease of maintenance (Leung et al., 2008; Stiernagle,
1999). C. elegans is of great convenience for microbiome research
because worms have intestinal cells similar in structure to human in-
testinal cells (McGhee, 2007). Furthermore, the genetic manipulability
of C. elegans allows for precise genetic modications, which can be
complemented by conducting genetic screens and analysing bacterial
mutant collections. The integration of these advantages enables a
comprehensive exploration of microbial-host interactions and the un-
derlying mechanisms involved (Zhang et al., 2017).
In our previous study, we established a 3-way (microbiome-host-
chemical) high-throughput screen (HTS) platform, using C. elegans fed
with E. coli Keio collection on an integrated robotic platform COPAS
Biosort to explore the role of the gut microbiota in modulating AFB
1
toxicity in the context of microbiota-host interaction. We performed 2-
step screenings using 3985 Keio mutants and identied 73 E. coli mu-
tants that modulated C. elegans growth phenotype. Notably, among these
mutants, four genes (aceA, aceB, lpd, and pB) involved in the pyruvate
pathway were identied and subsequently conrmed to be more sensi-
tive to AFB
1
. Our results suggested that disturbances in bacterial
* Corresponding author at: 148 Environmental Health Science Building, 150 Green Street, Athens, GA 30602, USA.
E-mail addresses: bowentang@outlook.com (B. Tang), ksxue@uga.edu (K.S. Xue), jswang@uga.edu (J.-S. Wang), pwilliam@uga.edu (P.L. Williams), ltang@uga.
edu (L. Tang).
Contents lists available at ScienceDirect
Science of the Total Environment
journal homepage: www.elsevier.com/locate/scitotenv
https://doi.org/10.1016/j.scitotenv.2023.165809
Received 28 May 2023; Received in revised form 23 July 2023; Accepted 24 July 2023
Science of the Total Environment 900 (2023) 165809
2
pyruvate metabolism might have a signicant impact on AFB
1
in the
host (Tang et al., 2023).
In this study, we aimed to gain a deeper understand the impact of the
pyruvate pathway on AFB
1
toxicity in C. elegans. To achieve this, we
employed genetic engineering techniques to create two bacterial strains:
one with triple overexpression (aceB, lpd, and pB) and the other with
triple knockout of these genes. Additionally, we utilized two mutant
worm strains, pyk-1 and pdha-1 mutants, which are known to disrupt
pyruvate metabolism. By employing these genetic modications, we
were able to directly examine the impact of specic bacterial alterations
on the interaction between pyruvate pathway and AFB
1
toxicity.
2. Method and materials
2.1. Chemicals
Aatoxin B
1
(AFB
1
), sodium pyruvate and Pyruvate Assay Kit
(MAK071) were purchased from Sigma-Aldrich (St. Louis, MO). Stock
solutions at 1 M were prepared in DMSO. Fresh working solutions at
different concentrations were diluted from the stock using K-medium
(32 mM KCl and 51 mM NaCl) (Williams and Dusenbery, 1990) con-
taining E. coli (wild type or engineered type) as a food source at 1 mg/mL
(Brenner, 1974).
2.2. Nematode and bacterial strains
The nematodes, N2, VC3879 pyk-1 (gk3762), and VC4479 pdha-1
(gk5568), were purchased from the Caenorhabditis Genetics Center
(Minneapolis, MN, USA). All C. elegans strains were maintained at 20 C
on solid nematode growth medium (NGM) plates seeded with OP50
(Brenner, 1974) or engineered bacteria.
Escherichia coli Keio collection, including the parent strain BW25113
and knockout strains aceB, lpd, and pB), as the food source for
C. elegans were purchased from the NBRP at the National Institute of
Genetics, Shizuoka, Japan. Wild-type E. coli strain (BW25113) was
grown in Luria Broth (LB) medium (tryptone 10 g/L, yeast extract 5 g/L,
and NaCl 5 g/L) while E. coli Keio mutants were grown in LB containing
25
μ
g/L kanamycin. All the bacteria were grown for ~18 h at 37 C to
reach OD600 value to 1.01.5 and washed with K-medium 3 times
before being diluted to 1 mg/mL as a food source for nematodes.
2.3. High-throughput screening for growth in C. elegans
High-throughput assay testing for growth was performed using
COPAS Biosort (Boyd et al., 2010b; Tang et al., 2019; Tang et al., 2020).
Isolated C. elegans eggs with bleachsodium hydroxide were hatched in
K-medium overnight to obtain synchronized L1 growth-arrested larvae
(Lewis and Fleming, 1995). A total of 50 age-synchronized L1-stage
worms were sorted and dispensed into each well containing 90
μ
L of test
solutions which contained food source and the test chemical. After in-
cubation for 48 h at 20 C, the plates were loaded onto COPAS Biosort
for growth measurement. Prior to measurement, 10
μ
L formalin (v/v, 10
%) were added to each well to kill and straighten the animals for proper
measurement. The TOF values for each event were recorded as size in-
dicators for each worm.
2.4. Plasmid construction for triple overexpression of targeted genes in
BW25113
All DNA manipulations were performed based on the standard mo-
lecular cloning protocols (Sambrook et al., 1989). E. coli competent cells,
XL1-Blue (Stratagene), were used for plasmid construction. All primers
used in this study were listed in Supplementary Table 1. Gene products
(pB) amplied by PCR were digested with the corresponding enzymes
and ligated into similarly digested medium-copy-number plasmid pCS27
between SalI and MluI to form pCS27-pB. The amplied aceB and lpd
from E. coli were digested with Acc65I and NdeI, NdeI and SalI,
respectively, and then integrated into pCS27-pB to form pCS27-aceB-
lpd-pB. 2 % of the overnight cell culture was inoculated into 50 mL LB
medium in 125 mL asks and incubated for 2 h at 37 C and 280 rpm.
Isopropyl β-D-1-thiogalactopyranoside (IPTG) was added at a nal
concentration of 0.5 mM, and cell cultures were switched to 30 C for
induction. LB medium with kanamycin (25
μ
g/mL) was used for plasmid
propagation and cell inoculation.
2.5. Gene disruption for triple knockout of targeted genes in BW25113
The disruption of aceB, lpd, and pB in E. coli was conducted by P1
phage-based transduction (Thomason et al., 2007). Specically, P1 ly-
sates for target knockouts were prepared from the Keio collection
strains. The transduced cells were screened on an LB-agar plate with
kanamycin and 100 mM sodium citrate. Kanamycin cassette on Keio
mutants was anked by the FLP recombinase site, thus allowing for the
excision of the cassette. The kanamycin cassette on the gene of interest
was removed by electroporation of pCP20, containing FLP recombinase,
into target strains (Datsenko and Wanner BLJPotNAoS., 2000). Trans-
formants were selected for ampicillin resistance at 30 C and then raised
at 42 C to remove the pCP20 plasmid. Colonies were veried by the
inability to grow on LB plates with the kanamycin. Knockout strains
were conrmed by colony PCR.
2.6. Qualication of pyruvate level
To measure the pyruvate level in C. elegans (N2, pyk-1, and pdha-1),
worms were washed with K-medium 3 times and extracted with meth-
anol/water (4:1) using a FastPrep-24 (MP Biomedicals) for ve cycles of
60 s. The tubes were then centrifuged at 3000g for 20 min to separate the
supernatant. The extraction process was repeated twice, and the su-
pernatants were combined for pyruvate analysis. Three types of Bacteria
(BW25113, triple overexpression, and triple knockout) at exponential
growth phase (OD600 =0.5) were collected and centrifuged at 1500g
for 1 min. The supernatant was used for pyruvate measurement. Pyru-
vate levels were estimated using the ELISA kit from Abcam (MA, USA)
according to the manufacturer instruction.
2.7. Statistical analysis
All data were processed with the statistical program R, version 4.0.2.
Bacterial genes which induced statistically signicant changes in worms
in response to AFB
1
in exposed groups (worms fed E. coli Keio mutants)
compared to controls (worms fed the wild-type strain BW25113) were
identied using one-way analysis of variance (ANOVA) with Tukeys t-
test. A p-value of <0.05 was considered statistically signicant. To
model decreasing activity of growth with increasing dose, the Hill
expression Hill=vm*dosex
kmx+dosex was subtracted from V
m
+a
0
. EC
50
(the 50 %
effective concentration of a toxicant) and corresponding 95 % CI (con-
dence intervals) for each cohort were calculated by tting the mean
values of parameters (TOF for growth) to the decreasing Hill equation
(Eq. (1)) (Boyd et al., 2010a). The V
m
+a
0
refers to the worm response
after exposure for the non-treated nematodes and is the maximum value
the expression can have. The value of the expression decreases to the
lower asymptote, a
0
, as chemical concentration increases. The lower
asymptote indicates a maximum possible decrease in the response. The
parameter, Km, is the estimated concentration that decreases the
response to halfway between Vm +a
0
and a
0
, which is the EC
50
. At last,
x is the shape parameter that governs the steepness of the decrease.
Decreasing Hill =a0+Vm
1+(dose
Km )x(1)
B. Tang et al.
Science of the Total Environment 900 (2023) 165809
3
3. Results
3.1. Bacterial pyruvate metabolism plays an important role in modulating
AFB
1
toxicity
In our previous study, we utilized a genetic screen with the Keio
collection and subsequent enrichment analysis, which revealed the
involvement of four bacterial genes (aceA, aceB, lpd, and pB) in
modulating AFB
1
toxicity in C. elegans through the pyruvate biosynthesis
pathway (Fig. 1). To investigate the impact of bacterial pyruvate
metabolism on AFB
1
toxicity in the host, we performed genetic engi-
neering on two bacterial strains derived from the wild-type BW25113:
one strain with triple knockouts of the three genes, and another strain
with triple overexpression. According to the bacterial metabolic
pathway (Fig. 1), aceB, lpd, and pB play crucial roles in regulating the
conversion of pyruvate into either acetyl Co-A or formate. We hypoth-
esis that the triple knockout of aceB, lpd, and pB leads to an increase in
pyruvate production, whereas triple overexpression results in a reduc-
tion in pyruvate levels. This is conrmed by measuring the pyruvate
levels in the bacteria medium (Fig. 2A). Compared to BW25113, the
pyruvate level in the culture medium was 11.6 % higher in triple
knockout bacteria and 18.4 % lower in triple overexpression bacteria.
The wild-type worm N2 was fed the two types of engineered bacteria
(triple knockouts and triple overexpression) as well as wild-type bacteria
(BW25113), and subsequently exposed to AFB
1
at concentrations
ranging from 0.1 to 10
μ
mol/L
,
and their body length was measured
afterward (Fig. 2B). Animals fed the pyruvate-downregulated bacterial
showed reduced sensitivity to AFB
1
with longer body length at each
concentration (p <0.05) compared to those fed the wild-type bacteria.
Conversely, animals fed the pyruvate-upregulated triple-knockout bac-
teria exhibited increased sensitivity to AFB
1
, with shorter body lengths
at each concentration (p <0.05). The calculated EC
50
values for the
three bacteria are showed in Table 1. Triple overexpression bacterial has
higher EC50 (8.05
μ
M), the EC50 of BW25113 and Triple knockout
bacterial was 3.92 and 3.12
μ
M, separately. To assess the potential
impact of the engineered bacterial strains on the response of worms, the
effects of these strains on the growth of C. elegans were also examined in
the absence of AFB
1
(Supplemental Fig. 1), the results indicated no
signicant difference in the body length, compared to wild-type bacte-
rial (p >0.05). These ndings suggest that pyruvate in bacteria
potentially plays a role in modulating host response to AFB
1
.
3.2. Host pyruvate metabolism involved in modulating AFB
1
toxicity
To gain a deeper understanding of pyruvates role in the host, we
conducted further investigations to determine if the hosts pyruvate
metabolism is involved in modulating AFB
1
toxicity. Two mutant worm
strains, pyruvate kinase-decient (pyk-1) worms and pyruvate
dehydrogenase-decient (pdha-1) worms were employed. According to
pyruvate metabolic pathway in C. elegans (Fig. 3), knocking out the
pdha-1 gene resulted in increased pyruvate levels, while knocking out
the pyk-1 gene led to decreased pyruvate levels. As shown in Fig. 4, pyk-1
knockout worms showed 8 % lower (p >0.05) pyruvate level while
pdha-1 worms showed 23.2 % (p <0.01) higher level of pyruvate,
compared to the wild-type N2.
Fig. 4B demonstrates that the pyk-1 mutant worm resulted in reduced
sensitivity to AFB
1
as indicated by body length, in a concentration-
dependent manner with the highest EC
50
value (4.60
μ
M, Table 2),
compared to the wild-type worm and pdha-1 mutant worm (p <0.05).
Conversely, the pdha-1 mutant worm led to increased sensitive to AFB
1
,
in a concentration-dependent manner, with the lowest EC
50
value (2.38
μ
M, Table 2), compared to the wild-type worm and pyk-1 mutant worm
(p <0.05).
3.3. Host-bacteria co-metabolism synergistic modulating AFB
1
toxicity
To further investigate the effect of host-bacterial interaction on AFB
1
toxicity and determine whether that of the host or the microbe, plays a
more important role in modulating host response to AFB
1
, we examined
the worms body length change in response to AFB
1
by using both
engineered bacteria strains and mutant worm strains. As mentioned
above, knocking out pyk-1 worm and triple-overexpression bacterial
strain led to decrease pyruvate level, while the pdha-1 mutant worm and
triple-knockout bacterial strain resulted in increasing production of
pyruvate. As expected, the response to AFB
1
has signicantly difference,
in both a concentration- and bacteria-dependent manner, compared to
those fed with wild-type bacterial (Fig. 5).
When pyruvate level was downregulated in both bacteria (triple-
overexpression) and host (pyk-1 mutant worms), pyk-1 mutant worms
became less sensitive to AFB
1
as indicated by body length, compared to
those fed the wild-type bacteria, even when compared to their untreated
controls, at the lowest concentration. At the highest concentration (10
μ
M), pyk-1 mutant worms fed the triple-overexpressed bacteria were
1.39-fold more resistant to AFB
1
compared to those fed the wild-type
bacteria, suggesting the protective effects of decreased pyruvate level
in triple-overexpressed bacteria. However, the worms growth was
signicantly inhibited fed with triple-knockout bacterial, compared to
those fed the wild-type bacteria. On the other hand, when pyruvate level
was upregulated in both bacteria (triple-knockout) and host (pdha-1
mutant worms) (Fig. 5A), pdha-1 mutant worms exhibited greater size
reduction. At the highest (10
μ
M) concentrations, animals became 1.34-
fold more sensitive to AFB
1
than worms fed the wild-type bacteria,
suggesting the synergistic effects of increased pyruvate level in the
triple-overexpressed bacteria.
For determining whether the host or the bacterial pyruvate plays a
more important role in modulating host response to AFB
1
, we examined
worms body length change in response to AFB
1
using an antagonistic
mechanism by increasing pyruvate metabolism in bacteria while
decreasing that in the host, and vice versa. Fig. 5B showed that when
pyruvate levels were upregulated in bacteria (triple-knockout) but
downregulated in worms (pyk-1 mutants), animals growth was signif-
icantly inhibited, compared to those fed the wild-type and triple-
overexpression bacteria. However, when pyruvate levels were
decreased in bacteria (triple-overexpression) but increased in worms
(pdha-1 mutants), wormsgrowth was signicantly promoted, compared
to those fed the wild-type and triple-knockout bacteria. This indicates
Fig. 1. Disrupted bacterial metabolism based on KEGG enrichment analysis.
PFL (pB), pyruvate formate lyase; lpd, lipoamide dehydrogenase, pyruvate
dehydrogenase E3 monomer; PDH (aceB), pyruvate dehydrogenase complex;
MS (aceB), malate synthase; OAA, oxaloacetate; GOX, glyoxylate.
B. Tang et al.
Science of the Total Environment 900 (2023) 165809
4
that pyruvate metabolism in the bacteria, rather than that in the host,
plays an essential role in modulating AFB
1
toxicity in C. elegans.
3.4. Pure pyruvate supplement modulating AFB
1
toxicity
To further conrm the effects of pyruvate in modulating AFB
1
toxicity, 1 mM pyruvate was supplemented to worm mutants fed
different bacteria, whose growth was then examined after exposure to 1
μ
M AFB
1
. All the animals became more sensitive to AFB
1
when sup-
plemented with pyruvate (Fig. 6). Animals supplemented with pyruvate
showed increased sensitivity to AFB
1
with shorter body length (p <0.01)
compared to those without pyruvate supplement. In wild-type worms
N2, the addition of pyruvate caused a 1.1, 2, and 1.5-fold increase in
sensitivity to AFB
1
when the animals were fed triple-overexpressed,
wild-type, and triple-knockout bacteria, respectively. In pyruvate
kinase-decient worms (pyk-1), pyruvate supplement induced 1.3- and
1.2-fold increased sensitivity to AFB
1
when worms were fed wild-type
and triple-overexpressed bacteria, respectively. In pyruvate
dehydrogenase-decient worms (pdha-1), worms fed triple-knockout
and wild-type bacteria suffered 1.45- and 2.13-fold increased sensi-
tivity to AFB
1
, respectively. To summarize, our nding suggested that
pyruvate plays an important role in modulating AFB
1
toxicity in
C. elegans.
4. Discussion
The gut microbiota plays a signicant role in the toxicity of envi-
ronmental contaminants (Claus et al., 2016). In our previous study, we
established a 3-way high-throughput screening system involving mi-
crobes, worms and chemicals. Through this system, we identied four
bacteria genes (aceA, aceB, lpd, and pB) involved in the pyruvate
metabolism that displayed signicant contribution to induce different
growth rates in response to AFB
1
in C. elegans (Tang et al., 2023).To
further explore the inuence of the pyruvate pathway in modulating
AFB
1
toxicity in C. elegans, we engineered two bacteria strains (triple-
overexpressed and triple-knockout with aceB, lpd, and pB) together
with two mutant worm strains (pyk-1 and pdha-1 mutants). Our ndings
indicate that co-metabolism of pyruvate by the host and bacterial syn-
ergistically modulated AFB
1
toxicity. Specically, and the bacterial py-
ruvate metabolism, rather than that of the host, plays a crucial role in
modulating AFB
1
toxicity in C. elegans.
Pyruvate metabolism is a crucial process in cellular respiration and
plays a central role in energy production and various metabolic path-
ways. Pyruvate, a threecarbon molecule, is derived from glucose
metabolism through glycolysis. Once formed, pyruvate can be further
metabolized through different pathways depending on the cells energy
demands and metabolic conditions (Gray et al., 2014). Studies have
demonstrated that mycotoxins, including AFB
1
, impaired ATP avail-
ability and resulted in decreased pyruvate levels (Baldissera et al., 2018;
Po´
or et al., 2014). However, the mechanisms of pyruvates effects on
AFB
1
toxicity remain largely unknown. Research has shown that certain
metabolic pathways can modulate the toxicity of AFB
1
, including those
involving pyruvate. Our results showed that addition of pyruvate
increased wormssensitivity to AFB
1
by fed with triple- knockout bac-
teria and supplementing pyruvate (1 mM) to worms. This might be
related to oxidative stress and the activity of cytochrome P450. The
Fig. 2. (A) Pyruvate levels in bacteria medium. (B) The effects of different bacteria on growth of wild type C. elegans (N2) in response to AFB
1
. Raw data were
normalized to the TOF value for untreated animals, shown as a percentage of the TOF value for untreated animals. Results are presented as mean of 5 replicates and
error bars represent standard error. *p <0.05, **p <0.01 determined by Students t-test.
Table 1
The estimated EC
50
values and corresponding 95 % condence interval on
growth in wild type C. elegans fed on different bacteria mutants to AFB
1
exposure.
BW25113 Triple knockout Triple overexpression
N2 EC
50
(
μ
M) 3.92 3.12 8.05
95 % CI (2.984.86) (06.83) (1.8814.22)
Fig. 3. Pyruvate metabolism in C. elegans. Modied based on C. elegans meta-
bolism pathway in KEGG. In pyruvate kinase mutant worms (pyk-1), pyruvate
level is decreased while in pyruvate dehydrogenase mutant worms (pdha-1),
pyruvate level is increased.
B. Tang et al.
Science of the Total Environment 900 (2023) 165809
5
pyruvate was found to induce hydrogen peroxide accumulation and
cause excessive ROS production in C. elegans (Mouchiroud et al., 2011),
which further promoted DNA damage and cell death induced by AFB
1
.
Although in one study found that pyruvate supplement at 100 mM in
wormsdiet inducing stress resistance and cellular defence and promote
longevity in worms (Tauffenberger et al., 2019). However, lower con-
centrations of pyruvate (10 mM) did not induce this phenomenon
(Tauffenberger et al., 2019). To avoid the above effects, a pyruvate
supplement at a lower concentration (1 mM) was selected in this study.
In addition, we also found pyruvate supplement alone at such concen-
tration did not cause any different growth rate in C. elegans in the
absence of AFB
1
.
In C. elegans, Pyruvate kinase (PYK) catalyses irreversibly transfer
phosphor from phosphoenolpyruvate (PEP) to ADP to form pyruvate and
ATP. Another important enzyme complex involved in pyruvate meta-
bolism is pyruvate dehydrogenase (PDH), which converts of pyruvate
into citric acid cycle to generate energy (Wang et al., 2002). Our result
Fig. 4. (A) Pyruvate levels in C. elegans. (B) The effects of BW25113 on growth of wild type and mutant C. elegans in response to AFB
1
. Raw data were normalized to
the TOF value for untreated animals, shown as a percentage of the TOF value for untreated animals. Results are presented as mean of 5 replicates and error bars
represent standard error. *p <0.05, **p <0.01 determined by Students t-test.
Table 2
The estimated EC
50
values and corresponding 95 % condence interval on
growth in wild type and mutant C. elegans fed on BW25113 in response to AFB
1
exposure.
N2 pyk-1 pdha-1
BW25113 EC
50
(
μ
M) 3.92 4.60 2.38
95 % CI (2.984.86) (0.958.25) (0.664.10)
Fig. 5. The effects of BW25113 and engineered bacteria on growth of mutant C. elegans (pyk-1 and pdha-1) in response to AFB
1
. (A) pyk-1 and pdha-1 worms treated
with triple overexpression and triple knockout bacteria, respectively. (B) pyk-1 and pdha-1 worms treated with triple knockout and triple overexpression bacteria,
respectively. Raw data were normalized to the TOF value for untreated animals, shown as a percentage of the TOF value for untreated animals. Results are presented
as mean of 5 replicates and error bars represent standard error. *p <0.05, ***p <0.001 determined by Students t-test.
B. Tang et al.
Science of the Total Environment 900 (2023) 165809
6
indicated that pyk-1 and pdha-1 mutant worms, which knocking out pyk
and pdh genes, affected the pyruvate content in C. elegans, consequently
synergistically modulating AFB
1
toxicity in combination with bacterial
pyruvate pathway. Interestingly, despite the pyruvate level in pyk-1
knockout worms didnt show a signicant reduction compared to
wild-type worms. However, the pyk-1 mutant worm still exhibited
reduced sensitivity to AFB
1.
This observation suggests that the decreased
sensitivity may be attributed to the role of PYK itself. Previous studies
have reported that PYK can react with AFB
1
-dialdehyde, which is
derived from the rapid hydrolysis of AFBO to AFB
1
-dihydrodiol in AFB
1
phase II detoxication pathway, thereby leading to increased cytotox-
icity (Hayes et al., 1993; Neal et al., 1981). Furthermore, PYK has
demonstrated efcient bind AFB
1
and signicant enhancement of AFB
1
activation in the nucleus (McLean and Dutton, 1995).
AFB
1
, the most toxic and hepatocarcinogenic mycotoxin known,
continues to pose a signicant public health burden, particularly in
developing countries. (Benkerroum, 2009; Liu and Wu, 2010; Rasheed
et al., 2021). In 2004, it was estimated that ~4.5 billion people in
developing countries were at risk for chronic and uncontrolled AFB
1
exposure (Williams et al., 2004). To address this issue, many countries
have implemented strict regulations to minimize AFB
1
levels in food.
However, these regulations have led to substantial annual losses in the
agricultural industry. In the United States alone, losses due to aatoxins,
including AFB
1
, have been estimated to range from $52.1 million to
$1.68 billion annually (Mitchell et al., 2016). Current strategies for AFB
1
control primarily focus on decontamination by degrading or removing
AFB
1
from crops, as well as prevention by implementing appropriate
management systems to reduce AFB
1
contamination in the eld and
during storage (Benkerroum, 2009). However, these methods are
insufcient when AFB
1
is already absorbed by the human body. In recent
years, probiotic bacteria have emerged as potential detoxication tools
for mycotoxins by inhibiting intestinal absorption, limiting the entry of
toxicants into the systemic circulation, and facilitating their excretion
(Paulina et al., 2021). The results of our study provide signicant in-
sights into microbiome-based intervention strategies for AFB
1
exposure.
By investigating the role of the gut microbiota, specically the bacteria
involved in pyruvate metabolism, in modulating AFB
1
toxicity in
C. elegans, our ndings contribute to the growing body of knowledge on
the potential of microbiota-based approaches to mitigate the adverse
effects of AFB
1
. Understanding the interplay between the gut microbiota
and AFB
1
toxicity can pave the way for the development of novel in-
terventions that target the microbiome and its interactions with
mycotoxins.
The main limitation of this study is the differences in AFB
1
meta-
bolism between C. elegans and mammalian models. AFB
1
requires
metabolic activation by CYP enzymes to form ultimate epoxide
metabolites, which bind to DNA molecules, and resulting in DNA dam-
age. AFB
1
can be activated in mammals by CYP1, CYP2, and CYP3 family
enzymes, however, C. elegans lacks CYP1 family enzymes (Gotoh, 1998;
Leung et al., 2010). This will decrease the sensitivity of C. elegans
response to the toxicity of AFB
1
, though there was a study found that
C. elegans can metabolize AFB
1
into DNA-binding metabolites and that
this activation is CYP dependent (Leung et al., 2010). Differences in
study models, such as variations in species, have been challenges for the
research eld of toxicology and risk assessment, especially for extrap-
olations from lab study ndings to human population risks(Queiros
et al., 2019).
5. Conclusion
Our previous study identied four bacterial genes (aceA, aceB, lpd,
and pB) that related to pyruvate metabolism pathways play a signi-
cant role in modulating AFB
1
toxicity in C. elegans. To further investigate
the impact of the pyruvate pathway on AFB
1
toxicity in C. elegans, we
engineered two bacterial strains (triple-overexpressed and triple-
knockout strains with aceB, lpd, and pB) along with two mutant
worm strains (pyk-1 and pdha-1 mutants). Our ndings demonstrate
that the co-metabolism of pyruvate by both the host and bacterial strains
synergistically inuences AFB
1
toxicity. Notably, bacterial pyruvate
metabolism, rather than that of the host, plays a pivotal role in modu-
lating AFB
1
toxicity in C. elegans. Our study sheds light on the role of gut
microbiota, particularly bacteria involved in pyruvate metabolism, in
inuencing AFB
1
toxicity in C. elegans. These ndings add to the
expanding understanding of the potential of microbiota-based strategies
in alleviating the detrimental impacts of AFB
1
.
Supplementary data to this article can be found online at https://doi.
org/10.1016/j.scitotenv.2023.165809.
Funding sources
This work was supported by the USDA/NIFA under Grant 2022-
67017-36237. N2 worms were provided by the CGC, funded by the NIH
Ofce of Research Infrastructure Programs (P40 OD010440).
CRediT authorship contribution statement
Bowen Tang: Methodology, Investigation, Data curation, Formal
analysis, Writing original draft. Kathy S. Xue: Methodology, Valida-
tion, Resources, Writing review & editing, Visualization. Jia-Sheng
Wang: Validation, Supervision, Writing review & editing. Phillip L.
Williams: Validation, Resources, Writing review & editing. Lili Tang:
Supervision, Project administration, Funding acquisition, Writing
Fig. 6. The effects of AFB
1
(1
μ
M) in C. elegans mutants fed different bacteria strains, with or without pyruvate supplement (1 mM). Mean TOF values were obtained
directly from COPAS Biosort without any normalization. Results are presented as mean of 5 replicates and error bars represent standard error. ***p <0.01
determined by Students t-test.
B. Tang et al.
Science of the Total Environment 900 (2023) 165809
7
review & editing, All authors have read and agreed to the published
version of the manuscript.
Declaration of competing interest
The authors declare that they have no known competing nancial
interests or personal relationships that could have appeared to inuence
the work reported in this paper.
Data availability
Data will be made available on request.
References
Baan, R., Grosse, Y., Straif, K., Secretan, B., El Ghissassi, F., Bouvard, V., et al., 2009.
A review of human carcinogenspart F: chemical agents and related occupations,
10, pp. 11431144.
Baldissera, M.D., Souza, C.F., Zeppenfeld, C.C., Descovi, S., da Silva, A.S.,
BJFP, Baldisserotto, et al., 2018. Changes in the Cerebral Phosphotransfer Network
Impair Energetic Homeostasis in an Aatoxin B 1-contaminated Diet, 44,
pp. 10511059.
Benkerroum, N., 2009. Aatoxins: producing-molds, structure, health issues and
incidence in Southeast Asian and Sub-Saharan African countries. Int. J. Environ. Res.
Public Health 17 (4), 1215.
Boyd, W.A., McBride, S.J., Rice, J.R., Snyder, D.W., Freedman, J.H., 2010a. A high-
throughput method for assessing chemical toxicity using a Caenorhabditis elegans
reproduction assay. Toxicol. Appl. Pharmacol. 245, 153159.
Boyd, W.A., Smith, M.V., Kissling, G.E., Freedman, J.H., 2010b. Medium-and high-
throughput screening of neurotoxicants using C. elegans. Neurotoxicol. Teratol. 32,
6873.
Brenner, S., 1974. The genetics of Caenorhabditis elegans. Genetics 77, 7194.
Chang, J., Wang, T., Wang, P., Yin, Q., Liu, C., Zhu, Q., et al., 2020. Compound probiotics
alleviating aatoxin B1 and zearalenone toxic effects on broiler production
performance and gut microbiota. Ecotoxicol. Environ. Saf. 194, 110420.
Claus, S.P., Guillou, H., Ellero-Simatos, S., 2016. The gut microbiota: a major player in
the toxicity of environmental pollutants? npj Biolms Microbiomes 2, 16003.
Creppy, E.E., 2002. Update of survey, regulation and toxic effects of mycotoxins in
Europe. Toxicol. Lett. 127, 1928.
Datsenko, K.A., Wanner BLJPotNAoS., 2000. One-step Inactivation of Chromosomal
Genes in Escherichia coli K-12 Using PCR Products, 97, pp. 66406645.
Gotoh, O., 1998. Divergent structures of Caenorhabditis elegans cytochrome P450 genes
suggest the frequent loss and gain of introns during the evolution of nematodes. Mol.
Biol. Evol. 15, 14471459.
Gray, L.R., Tompkins, S.C., Taylor, E.B., 2014. Regulation of pyruvate metabolism and
human disease. Cell. Mol. Life Sci. 71, 25772604.
Hayes, J.D., Judah, D.J., Neal GEJCr., 1993. Resistance to Aatoxin B1 is Associated
With the Expression of a Novel Aldo-keto Reductase Which has Catalytic Activity
Towards a Cytotoxic Aldehyde-containing Metabolite of the Toxin, 53,
pp. 38873894.
Leung, M.C., Williams, P.L., Benedetto, A., Au, C., Helmcke, K.J., Aschner, M., et al.,
2008. Caenorhabditis elegans: an emerging model in biomedical and environmental
toxicology. Toxicol. Sci. 106, 528.
Leung, M.C., Goldstone, J.V., Boyd, W.A., Freedman, J.H., Meyer, J.N., 2010.
Caenorhabditis elegans generates biologically relevant levels of genotoxic
metabolites from aatoxin B1 but not benzo[a]pyrene in vivo. Toxicol. Sci. 118,
444453.
Lewis, J.A., Fleming, J.T., 1995. Basic culture methods. In: Methods in Cell Biology, 48.
Elsevier, pp. 329.
Liu, Y., Wu, F., 2010. Global burden of aatoxin-induced hepatocellular carcinoma: a
risk assessment. Environ. Health Perspect. 118 (6), 818824.
McGhee, J.D., 2007. The C. elegans intestine. In: WormBook: The Online Review of C.
Elegans Biology [Internet].
McLean, M., Dutton, M.F.J.P., 1995. Therapeutics. In: Cellular Interactions and
Metabolism of Aatoxin: An Update, 65, pp. 163192.
Mitchell, N.J., Bowers, E., Hurburgh, C., Wu, F., 2016. Potential economic losses to the
US corn industry from aatoxin contamination. Food Addit. Contam. Part A Chem.
Anal. Control Expo. Risk Assess. 33, 540550.
Mouchiroud, L., Molin, L., Kasturi, P., Triba, M.N., Dumas, M.E., Wilson, M.C., et al.,
2011. Pyruvate Imbalance Mediates Metabolic Reprogramming and Mimics Lifespan
Extension by Dietary Restriction in Caenorhabditis Elegans, 10, pp. 3954.
Neal, G., Metcalfe, S., Legg, R., Judah, D., Green, J.J.C., 1981. Mechanism of the
Resistance to Cytotoxicity Which Precedes Aatoxin B 1 Hepatocarcinogenesis, 2,
pp. 457461.
Paulina, S., Edyta, J.-K., Michał, W., Monika, A., Marek, R.L.J.F., 2021. Probiotics as a
biological detoxication tool of food chemical contamination: a review. Food Chem.
Toxicol. 112306.
Po´
or, M., Veres, B., Jakus, P.B., Antus, C., Montsk´
o, G., Zrínyi, Z., et al., 2014. Flavonoid
Diosmetin Increases ATP Levels in Kidney Cells and Relieves ATP Depleting Effect of
Ochratoxin A, 132, pp. 19.
Queiros, L., Pereira, J.L., Goncalves, F.J.M., Pacheco, M., Aschner, M., Pereira, P., 2019.
Caenorhabditis elegans as a tool for environmental risk assessment: emerging and
promising applications for a nobelized worm. Crit. Rev. Toxicol. 49, 411429.
Rasheed, H., Xu, Y., Kimanya, M.E., Pan, X., Li, Z., Zou, X., et al., 2021. Estimating the
Health Burden of Aatoxin Attributable Stunting Among Children in Low Income
Countries of Africa, 11, pp. 111.
Sambrook, J., Fritsch, E.F., Maniatis, T., 1989. Molecular Cloning: A Laboratory Manual.
Cold Spring Harbor Laboratory Press.
Smoke, T., Smoking, I., 2004. IARC Monographs on the Evaluation of Carcinogenic Risks
to Humans, 1. IARC, Lyon, pp. 11452.
Stiernagle TJCe, 1999. Maintenance of C. Elegans, 2, pp. 5167.
Tang, B., Tong, P., Xue, K.S., Williams, P.L., Wang, J.S., Tang, L., 2019. High-throughput
assessment of toxic effects of metal mixtures of cadmium(Cd), lead(Pb), and
manganese(Mn) in nematode Caenorhabditis elegans. Chemosphere 234, 232241.
Tang, B., Williams, P.L., Xue, K.S., Wang, J.-S., Tang, L., 2020. Detoxication
mechanisms of nickel sulfate in nematode Caenorhabditis elegans. Chemosphere
260, 127627.
Tang, B., Xue, K.S., Wang, J.-S., Williams, P.L., Tang, L., 2023. Host-microbiota affects
the toxicity of aatoxin B1 in Caenorhabditis elegans. Food Chem. Toxicol. 176,
113804.
Tauffenberger, A., Fiumelli, H., Almustafa, S., PJJCd, Magistretti, 2019. Lactate and
pyruvate promote oxidative stress resistance through hormetic ROS signaling. Cell
Death Dis. 10, 116.
Thomason, L.C., Costantino, N., DLJCpimb, Court, 2007. E. Coli Genome Manipulation
by P1 Transduction, 79, 1.17. 1-1.17. 8.
Wang, H., Chu, W., Das, S.K., Ren, Q., Hasstedt, S.J., Elbein, S.C.J.D., 2002. Liver
Pyruvate Kinase Polymorphisms are Associated With Type 2 Diabetes in Northern
European Caucasians, 51, pp. 28612865.
Wang, J., Tang, L., Glenn, T.C., Wang, J.S., 2016. Aatoxin B1 induced compositional
changes in gut microbial communities of male F344 rats. Toxicol. Sci. 150, 5463.
Williams, J.H., Phillips, T.D., Jolly, P.E., Stiles, J.K., Jolly, C.M., Aggarwal, D., 2004.
Human aatoxicosis in developing countries: a review of toxicology, exposure,
potential health consequences, and interventions. Am. J. Clin. Nutr. 80, 11061122.
Williams, P.L., Dusenbery, D.B., 1990. Aquatic toxicity testing using the nematode,
Caenorhabditis elegans. Environ. Toxicol. Chem. 9, 12851290.
Zhang, J., Holdorf, A.D., Walhout, A.J.M., 2017. C. elegans and its bacterial diet as a
model for systems-level understanding of hostmicrobiota interactions. Curr. Opin.
Biotechnol. 46, 7480.
Zhou, J., Tang, L., Wang, J., Wang, J.S., 2018. Aatoxin B1 disrupts gut-microbial
metabolisms of short-chain fatty acids, long-chain fatty acids, and bile acids in male
F344 rats. Toxicol. Sci. 164, 453464.
B. Tang et al.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Aflatoxins are a group of potent fungal metabolites produced by Aspergillus and commonly contaminate groundnuts and cereal grains. Aflatoxin B1 (AFB1), the most potent mycotoxin, has been classified as Group 1 human carcinogen because it can be metabolically activated by the cytochrome P450 (CYP450) in the liver to form AFB1-DNA adducts and induce gene mutations. Increasing evidence has shown the gut microbiota as a key mediator of AFB1 toxicity through multiple interactive host-microbiota activities. To identify specific bacterial activity that modulates AFB1 toxicity in Caenorhabditis (C.) elegans, we established a 3-way (microbe-worm-chemical) high-throughput screening system using C. elegans fed E. coli Keio collection on an integrated robotic platform, COPAS Biosort. We performed 2-step screenings using 3,985 Keio mutants and identified 73 E. coli mutants that modulated C. elegans growth phenotype. Four genes (aceA, aceB, lpd, and pflB) involved in the pyruvate pathway were identified from the screening and confirmed to increase the sensitivity of all animals to AFB1. Taking together, our results indicated that disturbances in bacterial pyruvate metabolism might have a significant impact on AFB1 toxicity in the host. Abstract Aflatoxins are a group of potent fungal metabolites produced by Aspergillus and commonly
Article
Full-text available
Nowadays, people are exposed to diverse environmental and chemical pollutants produced by industry and agriculture. Food contaminations such as persistent organic pollutants (POPs), heavy metals, and mycotoxins are a serious concern for global food safety with economic and public health implications especially in the newly industrialized countries (NIC). Mounting evidence indicates that chronic exposure to food contaminants referred to as xenobiotics exert a negative effect on human health such as inflammation, oxidative stress, and intestinal disorders linked with perturbation of the composition and metabolic profile of the gut microflora. Although the physicochemical technologies for food decontamination are utilized in many cases but require adequate conditions which are often not feasible to be met in many industrial sectors. At present, one promising approach to reduce the risk related to the presence of xenobiotics in foodstuffs is a biological detoxification done by probiotic strains and their enzymes. Many studies confirmed that probiotics are an effective, feasible, and inexpensive tool for preventing xenobiotic-induced dysbiosis and alleviating their toxicity. This review aims to summarize the current knowledge of the direct mechanisms by which probiotics can influence the detoxification of xenobiotics. Moreover, probiotic-xenobiotic interactions with the gut microbiota and the host response were also discussed.
Article
Full-text available
Numerous population-based studies have documented high prevalence of aflatoxin associated childhood stunting in low income countries. We provide an estimate of the disease burden of aflatoxin related stunting using data from the four African countries. For this empirical analysis, we obtained blood aflatoxin albumin adduct biomarker based exposure data as measured using ELISA technique and anthropometric measurement data from surveys done over a 12-year period from 2001 to 2012 in four low income countries in Africa. We used these data to calculate population attributable risk (PAR), life time disease burden for children under five by comparing two groups of stunted children using both prevalence and incidence-based approaches. We combined prevalence estimates with a disability weight, measuring childhood stunting and co-occurrence of stunting-underweight to produce years lived with disability. Using a previously reported mortality, years of life lost were estimated. We used probabilistic analysis to model these associations to estimate the disability-adjusted life-years (DALYs), and compared these with those given by the Institute for Health Metrics and Evaluation’s Global Burden of Disease (GBD) 2016 study. The PAR increased from 3 to 36% for aflatoxin-related stunting and 14–50% for co-occurrence of stunting and underweight. Using prevalence-based approach, children with aflatoxin related stunting resulted in 48,965.20 (95% uncertainty interval (UI): 45,868.75–52,207.53) DALYs per 100,000 individuals. Children with co-occurrence of stunting and underweight due to exposure to aflatoxin resulted in 40,703.41 (95% UI: 38,041.57–43,517.89) DALYs per 100,000 individuals. Uncertainty analysis revealed that reducing aflatoxin exposure in high exposure areas upto non-detectable levels could save the stunting DALYs up to 50%. The burden of childhood all causes stunting is greater in countries with higher aflatoxin exposure such as Benin. In high exposure areas, these results might help guide research protocols and prioritisation efforts and focus aflatoxin exposure reduction. HEFCE Global Challenge Research Fund Aflatoxin project.
Article
Full-text available
Nickel is the most prevailing metal allergen with the highest sensitization rate among the “TOP 25” contact allergens and can affect about 15% of the human population. It is an essential trace metal in plants, animals, and humans. However, the environmental levels of nickel are considerably higher than what is needed for human life. Exposure to high levels of nickel can lead to skin allergies, lung fibrosis, and carcinogenesis. Few existing studies have closely examined the toxicity of nickel, let alone investigated the effective detoxification pathways. Here, we developed a high-throughput screening platform to comprehensively evaluate the nickel toxicity in wild-type C. elegans and explore the underlying detoxification mechanisms in transgenic nematodes. We demonstrated that nickel exerted multiple toxic effects on growth, brood size, feeding, and locomotion in C. elegans. Of which, brood size is the most sensitive endpoint. Nickel was found to first bind to phytochelatin (PC) after entering the worms’ body and this PC-Ni complex was further transported by the ABC transporter, CeHMT-1, into the coelomocytes for further detoxification. Our study also demonstrated that the high-throughput screening platform is a promising system for evaluation and investigation of the ecological risks of heavy metals.
Article
Full-text available
This review aims to update the main aspects of aflatoxin production, occurrence and incidence in selected countries, and associated aflatoxicosis outbreaks. Means to reduce aflatoxin incidence in crops were also presented, with an emphasis on the environmentally-friendly technology using atoxigenic strains of Aspergillus flavus. Aflatoxins are unavoidable widespread natural contaminants of foods and feeds with serious impacts on health, agricultural and livestock productivity, and food safety. They are secondary metabolites produced by Aspergillus species distributed on three main sections of the genus (section Flavi, section Ochraceorosei, and section Nidulantes). Poor economic status of a country exacerbates the risk and the extent of crop contamination due to faulty storage conditions that are usually suitable for mold growth and mycotoxin production: temperature of 22 to 29 °C and water activity of 0.90 to 0.99. This situation paralleled the prevalence of high liver cancer and the occasional acute aflatoxicosis episodes that have been associated with these regions. Risk assessment studies revealed that Southeast Asian (SEA) and Sub-Saharan African (SSA) countries remain at high risk and that, apart from the regulatory standards revision to be more restrictive, other actions to prevent or decontaminate crops are to be taken for adequate public health protection. Indeed, a review of publications on the incidence of aflatoxins in selected foods and feeds from countries whose crops are classically known for their highest contamination with aflatoxins, reveals that despite the intensive efforts made to reduce such an incidence, there has been no clear tendency, with the possible exception of South Africa, towards sustained improvements. Nonetheless, a global risk assessment of the new situation regarding crop contamination with aflatoxins by international organizations with the required expertise is suggested to appraise where we stand presently.
Article
Full-text available
L-lactate was long considered a glycolytic by-product but is now being recognized as a signaling molecule involved in cell survival. In this manuscript, we report the role of L-lactate in stress resistance and cell survival mechanisms using neuroblastoma cells (SH-SY5Y) as well as the C. elegans model. We observed that L-lactate promotes cellular defense mechanisms, including Unfolded Protein Response (UPR) and activation of nuclear factor erythroid 2–related factor 2 (NRF2), by promoting a mild Reactive Oxygen Species (ROS) burst. This increase in ROS triggers antioxidant defenses and pro-survival pathways, such as PI3K/AKT and Endoplasmic Reticulum (ER) chaperones. These results contribute to the understanding of the molecular mechanisms involved in beneficial effects of L-lactate, involving mild ROS burst, leading to activation of unfolded protein responses and detoxification mechanisms. We present evidence that this hormetic mechanism induced by L-lactate protects against oxidative stress in vitro and in vivo. This work contributes to the identification of molecular mechanisms, which could serve as targets for future therapeutic approaches for cell protection and aging-related disorders.
Article
Full-text available
Heavy metals, a class of persistent environmental toxicants, are harmful to human health. Cd and Pb are two of the most common toxic heavy metals that have been linked with cancers and malfunction of the nervous system. Notably, contamination of Mn usually coexisted with Cd and Pb in environmental and occupational settings. Studies regularly examined the toxic effects on individual metals; however, potential health and toxic effects of mixtures containing two or more heavy metals are unknown. Here, we investigated toxic effects of Cd, Pb, Mn, and their binary and ternary mixtures in the nematode Caenorhabdities elegans. The toxic outcomes, including effects on growth, reproduction, and feeding, were measured via high-throughput platform analysis. The transgenic strain BY250 with GFP in dopaminergic neurons was used to explore the neurodegenerative effects induced by single metals or their mixtures. The combination index(CI) for mixtures effect was calculated using isobolograms methods. Following the exposure, we found significant toxic effects in C. elegans. For single metals, the toxicity order for growth, reproduction, and feeding were Pb > Cd > Mn. For mixtures, the mixture of Cd + Mn induced a less than addictive effect in C. elegans, whereas the mixtures of Cd + Pb, Pb + Mn, and Cd + Pb + Mn induced greater-than-additive effects. Both single metals and their mixtures induced abnormality in dopaminergic neurons. These results showed combinative toxic and neurodegenerative effects of heavy metal mixtures, and future studies will focus on characterization of concentration-response patterns and identification of potential molecular mechanisms in C. elegans model.
Article
In order to alleviate toxic effects of aflatoxins B1 (AFB1) and zearalenone (ZEA) on broiler production performance and gut microbiota, three kinds of compound probiotics (CP) were selected. The optimal ratios of Bacillus subtilis, Lactobacillus casei and Candida utilis in broiler diets were 7, 5 and 6 log CFU/g for ZEA biodegradation (CP1); 6, 7 and 7 log CFU/g for AFB1 biodegradation (CP2); 7, 6 and 7 log CFU/g for ZEA + AFB1 biodegradation (CP3). A total of 350 1-day-old Ross broilers were randomly divided into 7 groups. Group A was the basal diet, group B-G contained ZEA, AFB1, ZEA + AFB1, ZEA + CP1, AFB1+CP2, ZEA + AFB1+CP3, respectively. The experiment showed that AFB1 or AFB1+ZEA significantly decreased broiler production performance, damaged liver and jejunum, increased mycotoxin residues in broiler body; however, three kinds of compound probiotics additions could alleviate mycotoxin negative effects on the above parameters (p < 0.05). The gut microbiota analysis indicated that AFB1+ZEA increased jejunal microbial richness, but which were decreased to almost the same level as the control group by CP3 addition. CP3 addition significantly increased jejunal Firmicutes and Lactobacillus aviarius abundances. The correlative analysis showed that gut Lactobacillus aviarius abundance was positively correlated with average daily gain (ADG) of broilers (p < 0.05), while AFB1+ZEA addition decreased its relative abundance, indicating that CP3 addition increased broiler growth by increasing Lactobacillus aviarius abundance. AFB1 and ZEA residues in broiler body were negatively correlated with the gut beneficial bacterial abundances (p < 0.01), but positively correlated with the potentially harmful bacterial abundances (p < 0.05), which inferred that CP3 addition could decrease mycotoxin residues through positively regulating gut relative bacterial abundances. In conclusion, compound probiotics could keep gut microbiota stable, degrade mycotoxins, alleviate histological lesions, increase production performance and reduce mycotoxin toxicity for broilers.
Article
Caenorhabditis elegans has been an invaluable model organism in research fields such as developmental biology and neurobiology. Neurotoxicity is one of the subfields greatly profiting from the C. elegans model within biomedical context, while the corresponding potential of the organism applied to environmental studies is relevant but has been largely underexplored. Within the biomedical scope, the implication of metals and organic chemicals with pesticide activity (hereinafter designated as pesticides) in the etiology of several neurodegenerative diseases has been extensively investigated using this nematode as a primary model organism. Additionally, as a well-known experimental model bearing high sensitivity to different contaminants and representing important functional levels in soil and aquatic ecosystems, C. elegans has high potential to be extensively integrated within Environmental Risk Assessment (ERA) routines. In spite of the recognition of some regulatory agencies, this actual step has yet to be made. The purpose of this review is to discuss the major advantages supporting the inclusion of C. elegans in lower tiers of ERA. Special emphasis was given to its sensitivity to metals and pesticides, which is similar to that of other model organisms commonly used in ERA (e.g. Daphnia magna and Eisenia sp.), and to the large array of endpoints that can be tested with the species, both concerning the aquatic and the soil compartments. The inclusion of C. elegans testing may hence represent a relevant advance in ERA, providing ecologically relevant insights toward improvement of the regulatory capacity for establishing appropriate environmental protection benchmarks.
Article
In this study, male F344 rats were orally exposed to aflatoxin B1 (AFB1) at 0, 5, 25, and 75 μg/kg for four weeks. Rat feces were collected from 2-4 weeks following exposure and were assessed for gut-microbiota dependent metabolites. Gut-microbiota related organic acids were quantitated in the feces using 2-nitrophenylhydrazine derivatization coupled HPLC-profiling method which was validated and showed good reliability, accuracy and sensitivity. After 2-week exposure, AFB1 significantly reduced the levels of fecal short chain fatty acids (SCFAs) with an over 70% reduction in the high-dose group (75 μg/kg). Mixed-effects model revealed an inverse correlation between AFB1 dose and fecal levels of SCFAs, but no significant time effect was found. Compared with the control, oral exposure to middle-dose AFB1 (25 μg/kg) resulted in remarkable elevations of fecal cholic acid (2.18-fold), linoleic acid (cis-9,cis-12-18:2) (11.3-fold), pentadecanoic acid (15:0) (3.68-fold), pyruvic acid (4.56-fold) and 3-phenyllactic acid (3.74-fold), but deoxycholic acid level was reduced by 41% in the low-dose group (5 μg/kg). These results demonstrated the disruptions of several important gut-microbiota metabolic pathways, including the synthesis of SCFAs, pyruvic acid related pathways, metabolisms of amino acids, bile acids and long chain fatty acids, which may further affect host digestive efficiency, energy supply, intestinal immunity, production of neurotransmitters, and enterohepatic cross-talk. Our study suggests that the impairment of gut-microbiota dependent metabolism may contribute to pathological mechanisms of AFB1-induced adverse health effects.