Tink Ov 2018

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Environmental Pollution 235 (2018) 429e434

Contents lists available at ScienceDirect

Environmental Pollution
journal homepage: www.elsevier.com/locate/envpol

Gut as a target for cadmium toxicity*


Alexey A. Tinkov a, b, c, *, Viktor A. Gritsenko c, Margarita G. Skalnaya b,
Sergey V. Cherkasov c, Jan Aaseth d, e, Anatoly V. Skalny a, b, f
a
Yaroslavl State University, Sovetskaya St., 14, Yaroslavl 150000, Russia
b
Peoples' Friendship University of Russia (RUDN University), Miklukho-Maklay St., 10/2, Moscow 117198, Russia
c
Institute of Cellular and Intracellular Symbiosis, Russian Academy of Sciences, Orenburg, 460008, Russia
d
Innlandet Hospital Trust, 2226 Kongsvinger, Norway
e
Inland Norway University of Applied Sciences, Terningen Arena, 2411 Elverum, Norway
f
Orenburg State University, Pobedy Ave., 13, Orenburg 460018, Russia

a r t i c l e i n f o a b s t r a c t

Article history: The primary objective of the present study was to review the impact of Cd exposure on gut microbiota
Received 12 November 2017 and intestinal physiology, as well as to estimate whether gut may be considered as the target for Cd
Received in revised form toxicity. The review is based on literature search in available databases. The existing data demonstrate
26 December 2017
that the impact of Cd on gut physiology is two-sided. First, Cd exposure induces a significant alteration of
Accepted 28 December 2017
bacterial populations and their relative abundance in gut (increased Bacteroidetes-to-Firmicutes ratio),
accompanied by increased lipopolysaccharide (LPS) production, reflecting changed metabolic activity of
the intestinal microbiome. Second, in intestinal wall Cd exposure induces inflammatory response and cell
Keywords:
Cadmium
damage including disruption of tight junctions, ultimately leading to increased gut permeability.
Gut microbiota Together with increased LPS production, impaired barrier function causes endotoxinemia and systemic
Lipopolysaccharide inflammation. Hypothetically, Cd-induced increase gut permeability may also result in increased bac-
Bacteroides terial translocation. On the one hand, bacteriolysis may be associated with aggravation of endotoxemia.
Firmicutes At the same time, together with Cd-induced impairment of macrophage inflammatory response,
increased bacterial translocation may result in increased susceptibility to infections. Such a supposition is
generally in agreement with the finding of higher susceptibility of Cd-exposed mice to infections. The
changed microbiome metabolic activity and LPS-induced systemic inflammation may have a significant
impact on target organs. The efficiency of probiotics in at least partial prevention of the local (intestinal)
and systemic toxic effects of cadmium confirms the role of altered gut physiology in Cd toxicity.
Therefore, probiotic treatment may be considered as the one of the strategies for prevention of Cd
toxicity in parallel with chelation, antioxidant, and anti-inflammatory therapy.
© 2018 Elsevier Ltd. All rights reserved.

1. Introduction exposure is also found to be high in China (Zhang et al., 2016). At


the same time, the highest Cd content in rice was detected in
Cadmium is an environmental pollutant possessing serious Bangladesh and Sri-Lanka (Meharg et al., 2013). Although some
health hazards (Satarug et al., 2011). In particular, Cd pollution is studies demonstrate a significant reduction in Cd exposure (USA),
widespread through Europe with the highest levels in sediments further reduction is required (Tellez-Plaza et al., 2012a,b).
and stream water being observed in Germany and Slovakia, High social costs of Cd exposure are related to its role in various
whereas topsoil Cd levels were maximal in Greece, Italy, France, diseases. In particular, it has been demonstrated that Cd is associ-
Austria, Ireland (Pan et al., 2010). The mean weekly intake of Cd in ated with cancer (Adams et al., 2011; Hartwig, 2013), diabetes
European adults accounts for 2.5 mg/kg (Nawrot et al., 2010). Cd mellitus (Tinkov et al., 2017), cardiovascular diseases (Tellez-Plaza
et al., 2012a,b), chronic kidney disease (Ferraro et al., 2010), oste-
oporosis (Kazantzis, 2004), liver disease (Hyder et al., 2013), neu-
*
This paper has been recommended for acceptance by David Carpenter. rodegeneration (Min and Min, 2016), adverse neurodevelopmental
* Corresponding author. Yaroslavl State University, Sovetskaya St., 14, Yaroslavl outcome (Wang et al., 2016), and other diseases (Satarug et al.,
150000, Russia.
2011). It has been estimated that high Cd exposure significantly
E-mail address: tinkov.a.a@gmail.com (A.A. Tinkov).

https://doi.org/10.1016/j.envpol.2017.12.114
0269-7491/© 2018 Elsevier Ltd. All rights reserved.
430 A.A. Tinkov et al. / Environmental Pollution 235 (2018) 429e434

increases mortality by 17% (Nawrot et al., 2010). observed NF-kB activation and proinflammatory cytokine secretion
Traditionally, the variety of adverse health effects attributed to was at least partially dependent on Cd-induced alteration of gut
Cd exposure has been explained by the general mechanisms of microbiome, as germ-free mice were less prone to proin-
metal toxicity. In particular, it has been reported that Cd is capable flammatory response to Cd exposure (Kim et al., 2015). The
of induction of oxidative stress (Liu et al., 2009), inflammation observed changes in the gut persisted for a long time after elimi-
(Olszowski et al., 2012), endoplasmic reticulum stress (Biagioli nation of the agent. It is also notable, that transfer of gut micro-
et al., 2008), genomic instability (Rani et al., 2014), and essential biome to Cd-unexposed mice resulted in similar inflammatory and
metal dyshomeostasis (Moulis, 2010). Certain effects of the metal allergic responses, whereas no changes were detected in germ-free
are associated with its effect as an endocrine disruptor (Takiguchi mice (Kim et al., 2017). These findings are generally in agreement
and Yoshihara, 2006). However, further investigations of mecha- with the earlier observation of increased intestinal permeability to
nisms of Cd toxicity is highly required for assessment and pre- macromolecules in Cd-exposed mice (28 days) (Kim et al., 2014).
vention of its harmful effects. Liu et al. (2014) demonstrated that administration of the increasing
Gut microbiota is known to play a significant role in a number of doses of Cd significantly reduced intestinal bacterial populations as
physiological functions (Tremaroli and Ba €ckhed, 2012), including well as decreased the Firmicutes to Bacteroidetes ratio starting
regulating immunity (Round and Mazmanian, 2009) and mineral from 3 week of exposure. The number of Bifidobacteria also
metabolism (Skrypnik and Suliburska, 2017). Qualitative, quanti- decreased starting from the 1 week of Cd exposure, whereas the
tative, or functional (metabolic) disturbances of the gut microbiota significant dose-dependent reduction of lactobacilli population was
may mediate the development numerous diseases (Marchesi et al., detected only after 3 weeks of exposure. These changes were also
2015; Scott et al., 2015). Microbiota has been considered as a target accompanied by increased production of TNFa and alteration of
for environmental pollutants including persistent organic pollut- genes involved in the bacterial metabolism of short-chain fatty
ants, antibiotics, pesticides, as well as heavy metals (Jin et al., 2017). acids (SCFAs). Correspondingly, incubation of the cultured fecal
However, existing data and reviews on the possible role of gut microbiota with Cd in vitro resulted in a significant decrease in cell
dysbiosis in mediating Cd toxicity are insufficient. viability (Liu et al., 2014). Oppositely, another study demonstrated a
Therefore, the primary objective of the present study was to significant decrease in Bacteroides abundance in response to Cd
review the impact of Cd exposure on gut microbiota and intestinal treatment for 8 weeks. At the same time, Cd exposure was shown to
physiology as well as estimate whether gut may be considered as reduce SCFAs concentration in the fecal pellet (Li et al., 2016).
the target for Cd toxicity. In parallel with altered intestinal biodiversity under chronic Cd
exposure, short term Cd treatment also affects gut microbiome
1.1. Cd and gut microbiota physiology. In particular, it has been demonstrated that Cd expo-
sure induces a significant shift in bacterial proteome starting from
Gut microbiota is considered to be an important target medi- 15 min of exposure. Based on these findings the authors have
ating toxic effects of heavy metals including cadmium (Jin et al., proposed the characteristic changes of protein expression in gut
2017). The role of gut microbiota as a protective factor of Cd microbiota may be related to three temporal response patterns to
toxicity was indicated by Breton et al. (2013), who observed a sig- Cd exposure: resistance, adaptation, sustained tolerance (Lacerda
nificant increase in blood, liver, kidney, and spleen Cd content, as et al., 2007). In an earlier in vitro study Wang and Crowly also
well as MT1 and MT2 expression in duodenum and colon in germ- demonstrated Cd-induced alteration of global gene expression. Of
free mice treated with Cd (Breton et al., 2013a). Certain studies have note, in line with the impact on energy metabolism, cell cycle,
demonstrated the impact of chronic (and subchronic) and short- transport and binding protein genes, Cd exposure for 0, 5, 15, and
term Cd exposure on gut microbiome physiology in laboratory 25 min resulted in up-regulation of genes that may be related to
animals of different age. biosynthesis/transport of lipopolysaccharides (Wang and Crowley,
Cd exposed (42 days) adult rats were characterized by a signif- 2005). These data are of great importance, being indicative of the
icant reduction in anaerobic, and also aerobic, and lactic bacteria in nearly immediate effects of Cd on gut microbiota.
the gut (Jafarpour et al., 2015). Moreover, Cd induced specific Early life low-dose Cd exposure also significantly affected the
changes in gut microbiota on family and genus levels. An earlier quantity and biodiversity of the gut microbiome. In particular, at 8-
study demonstrated a significant dose-dependent reduction in week age Cd-exposed male (but not female) mice were character-
B. cereus, Lactobacillus spp, Clostridium spp, E. coli intestinal pop- ized by a significant increase in Bacteroides and a concomitant
ulations in mice exposed to Cd in drinking water for 45 days (Fazeli decrease in Firmicutes populations. At the genus levels, the most
et al., 2011). It has been also shown that Cd exposure for 8 weeks significant decrease was detected in Bifidobacterium and Prevotella
resulted in a significant dose-dependent increase in the relative populations. It is also notable that the population of Cd-
abundance of Actinobacteria in caecum of mice, whereas the accumulating Sphingomonas was significantly higher in Cd-
changes in Bacteroidetes and Firmicutes were not significant. At the exposed animals. Interestingly, the observed changes in gut
family level, caecal flora was characterized by a dose-dependent microbiome were associated with increased adiposity in Cd-treated
increase in the relative number of Coriobacteriaceae, Lactobacilla- mice. This consequence of Cd-induced alteration of the microbiome
ceae, whereas the percent of Lachnospiraceae decreased. The was confirmed by the observed increase in adipose tissue mass
changes in fecal bacteria in Cd-exposed mice were less pronounced accumulation after transfer of fecal flora from Cd-exposed to Cd-
(Breton et al., 2013). Zhai et al. (2017) have also demonstrated a unexposed mice. Moreover, pretreatment of mice with Gram-
significant increase in relative abundance of Alistipes and Odor- negative targeting antibiotics prevented Cd-induced increase in
ibacter genera, whereas Mollicutes and unclassified Ruminococca- adiposity (Ba et al., 2017). Another study demonstrated a significant
ceae were characterized by a significant Cd-induced reduction in decrease in caecal Firmicutes in response to Cd exposure in 5-week-
adult male C57black/6 mice after the same period of metal expo- old C57BL/6 male mice, whereas Bacteroidetes and g-proteobacteria
sure (Zhai et al., 2017). It is notable that Cd-induced alterations in were unaffected. These changes were associated with alteration of
microbiota are more pronounced in the first 4 weeks of exposure as energy homeostasis genes (fatty acids synthesis and transport,
compared to the latter 4 weeks. Kim et al. (2015) also demonstrated triglyceride synthesis) in liver that may be related to LPS-induced
that oral Cd treatment reduces bacterial diversity of the gut as well up-regulation of proinflammatory gene expression (Zhang et al.,
as alters Firmicutes to Bacteroidetes ratio. It is also notable that the 2015).
A.A. Tinkov et al. / Environmental Pollution 235 (2018) 429e434 431

Therefore, the majority of studies demonstrate a significant in- compared to control animals, as well as Cd and LPS-exposed only
crease in Bacteroidetes-to-Firmicutes ratio due to increased Bacter- (Srinivasan et al., 2011). In addition, Cd and LPS exposure may have
oidetes abundance and/or reduced Firmicutes populations. The potentiating effect on Cd-induced displacement of essential metals
existing contradictions on the effect of Cd exposure on particular (Cu, Zn) and NO production (Satarug et al., 2000). It is also notable
bacterial populations to Cd may be associated with different that high Cd exposure as assessed by blood Cd levels significantly
exposure regimens (dose, duration, period of life) or particular Cd improved the association between LPS levels and the prevalence of
species. metabolic syndrome (Han et al., 2015).
Other studies have indicated that Cd may interfere with normal
1.2. Gut permeability, inflammation and endotoxemia LPS signaling in cells. In particular, Cd perturbed inflammatory
response to LPS in macrophages via inhibition of NF-kB pathway,
It has been demonstrated that Cd exposure significantly affects thus reducing resistance to infectious agents (Cox et al., 2016), is in
intestinal epithelial cells viability and affects paracellular perme- agreement with the earlier study by Koropatnick and Zalups (1997).
ability through disruption of tight junction (Duizer et al., 1999). It is proposed that Cd-induced oxidative stress may at least partially
Later studies indicated that Cd exposure induced an irregular dis- mediate dysregulation of reactivity in murine macrophages (Jin
tribution of tight junction proteins in HT-29 cell monolayers, as et al., 2016). Moreover, it has been demonstrated that Cd expo-
well as significantly reduced mRNA expression of ZO-1, ZO-2, sure dose-dependently decreased proinflammatory cytokine
occludin, and claudin-1 in jejunum and colon of Cd-exposed mice. expression in colon of mice with dextran sodium sulfate or trini-
These changes were associated with increased gut permeability trobenzene sulfonic acid -induced colitis (Breton et al., 2016).
resulting in elevated blood LPS levels (Zhai et al., 2016). In a culture Generally, the existing studies demonstrate that in parallel with
of Caco-2 cells Cd exposure resulted in a significant alteration of increased LPS levels and endotoxemia due to Cd-induced increase
tight junction permeability associated with intercellular junction in gut permeability, Cd may also interfere with LPS signaling.
damage and increased expression of caspase 3, heat shock protein, However, the interaction between LPS and Cd may have both
and oxidative stress-related genes (Rusanov et al., 2015). These data antagonistic and synergistic effects depending on the dose and
are in agreement with the earlier findings of Boveri et al. (2004), target processes.
who detected a significant increase in paracellular permeability in
Caco-2 layer, being accompanied by a significant elevation of HSP70 1.3. Probiotics as a therapeutic strategy in Cd exposure
levels and the presence of certain necrotic lesions (Boveri et al.,
2004). It has been also demonstrated that Cd exposure resulted In agreement with the primary role of gut dysbiosis in Cd
in a significant dose-dependent increase in serum LPS levels in toxicity, multiple studies have demonstrated that probiotics may
mice, being accompanied by induction of LPS target genes (Lbp, have a significant protective effect. It has been demonstrated that
lipopolysaccharide-binding protein; TLR4, toll-like receptor 4; MD- the use of diets enriched with L. plantarum, B. coagulans, and inulin
2, myeloid differentiation factor 2; CD14) and pro-inflammatory prevented Cd-induced toxicity to liver and kidneys. In particular,
genes including IL-1b, TNFa, and IL-6 in liver (Zhang et al., 2015). probiotic-rich diet significantly reduced Cd accumulation in liver
It has been suggested that an increased synthesis of the protein and kidney, as reflected by decreased serum values of ALT, AST,
zonulin, a prehaptoglobin (Fasano, 2011) mediates the changes of creatinine and carbamide as compared to untreated Cd-exposed
tight junctions and accompanying disruptions in gut dysbiosis rats (Jafarpour et al., 2017). The changes were accompanied by
(Sturgeon and Fasano, 2016. improvement of the number of intestinal bacteria, especially lactic
Prolonged Cd exposure resulted in altered intestinal histology, bacteria, in Cd-exposed animals treated with the probiotics
being characterized by short and thick villi with certain fusion and (Jafarpour et al., 2015). Correspondingly, the prebiotic Lactobacillus
necrotic areas. Altered gut morphology was also associated with plantarum prevented significantly decreased Cd-induced inflam-
increased TNFa, IL-1b, IFN-y, and IL-17 levels in intestine, being matory response in Cd-exposed colonic (HT-29) cells, reducing
accompanied by reduced lactobacilli count. It is notable that Cd- TNFa, IL-1b, IL-6, and IL-8 expression. Moreover, L. plantarum
induced changes were also detected in mesenteric lymph nodes. treatment alleviated tight junction alteration, gut permeability and
Cd exposure induced proliferation resulting in increased lymph endotoxemia in Cd-exposed rats (Zhai et al., 2016). It has been also
node cellularity. Moreover, the increase in lymph node IFN-y, IL-17, demonstrated that the use of L. plantarum diminished Cd-induced
and IL-10 at both protein and mRNA levels was detected in damage to liver and kidney and increased renal antioxidant
response to Cd treatment (Ninkov et al., 2015). Later studies by the response. These findings allowed the authors to propose that the
authors have confirmed these findings and demonstrated strain- use of probiotics may be a more effective tool in treatment of Cd
specific changes, being more profound in Dark Agouti rats as intoxication than antioxidant therapy (Zhai et al., 2013). It is also
compared to Albino Oxford strain (Ninkov et al., 2016). In addition, notable that intraperitoneal administration of L. plantarum to Cd-
CdCl2 treatment resulted in a marked villi damage and infiltration exposed animals did not reduce Cd accumulation in the organs,
of inflammatory cells into the lamina propria of the proximal in- although increasing antioxidant activity and protecting tissues
testine of the mice, being associated with increased macrophage from damage (Zhai et al., 2014). Administration of a probiotic
inflammatory protein-2 (MIP-2) mRNA expression (Zhao et al., containing L. rhamnosus, L. acidophilus and B. longum reversed Cd-
2006). The observed proinflammatory response in intestine may induced depression of lactobacilli and bifidobacilli populations,
be at least partially mediated by Cd-induced activation of NF-kB via being accompanied by a significant reduction in Cd genotoxicity, as
I-kBa degradation as observed in a Caco-2 cell model (Hyun et al., assessed by DNA damage in hepatocytes (Jama et al., 2012).
2007). However, a histological study of gastrointestinal tract in The protective effect of probiotics under Cd-exposure may be
Cd-treated animals did not reveal any significant lesions in intes- related to direct binding of metal ions in the intestine (Ibrahim
tine (Nai et al., 2015). et al., 2006) thus reducing their bioaccessibility (Kumar et al.,
Certain studies have demonstrated the interactive effects be- 2017). It has been demonstrated that the increase in fecal lacto-
tween Cd and LPS toxicity. In particular, it has been demonstrated bacilli content significantly correlates with fecal Cd levels and the
that Cd and LPS coexposure in rats resulted in a more significant decrease in blood metal concentration (Djurasevic et al., 2017). In
liver damage via oxidative (lipid peroxidation) and nitrosative an in vitro model, preincubation of Cd(NO3)2 with Lactobacillus kefir
(nitrate, peroxynitrite) stress and inflammatory response (IL-1b) as significantly reduced its toxicity in HepG2 colony (Gerbino et al.,
432 A.A. Tinkov et al. / Environmental Pollution 235 (2018) 429e434

Fig. 1. The proposed targets and effects of Cd toxicity in the gut.

2014). microbiota metabolism is associated with non-alcoholic fatty liver


The scheme of the protective action of probiotics in Cd toxicity disease and alcoholic steatohepatitis, inflammatory bowel diseases
was proposed by Zhai et al. (2016) who demonstrated that Cd- (Goldsmith and Sartor, 2014), atherosclerosis (Koeth et al., 2013),
induced intestinal inflammation and tight junction disruption airway allergy, altered hemopoiesis (Trompette et al., 2014). In turn,
may be the key targets for probiotics (Zhai et al., 2016). The pro- LPS is involved in numerous inflammatory diseases including
tective action of probiotics may be improved by supplementation obesity and diabetes mellitus (Cani et al., 2007; We˛gielska and
with zinc (Sturniolo et al., 2002). Suliburska, 2016), neurodegenerative disorders (Qin et al., 2007)
including Parkinson's disease (Dutta et al., 2008), cardiovascular
diseases (Neves et al., 2013) including acute coronary syndromes
1.4. Summary of the toxic effects of Cd exposure on gut physiology (Troseid, 2017), liver dysfunction (Fukui, 2015), kidney failure
(McIntyre et al., 2011). Taking into account the significant role of Cd
The existing data demonstrate that the impact of Cd on gut exposure in pathogenesis of these diseases, it is possible that
physiology is two-sided. In gut microbiota Cd exposure induces a altered gut physiology may at least partially mediate the toxic ef-
significant alteration of bacterial populations and their relative fects of Cd. In addition, the efficiency of probiotics in at least partial
abundance (increased Bacteroidetes-to-Firmicutes ratio, etc.), prevention of the local (intestinal) and systemic toxic effects of
resulting in increased LPS production. Moreover, starting from first cadmium confirms the role of altered gut physiology in Cd toxicity.
minutes of exposure Cd affects metabolic activity of the intestinal Therefore, probiotic treatment may be considered as the one of the
microbiome. In intestinal wall Cd exposure induces inflammatory strategies for prevention of Cd toxicity in parallel with chelation,
response and cell damage. The one of the most important aspects is antioxidant, and anti-inflammatory therapy. However, further
disruption of tight junctions leading to increased gut permeability studies are strongly required to clarify the role of Cd exposure on
to macromolecules. Together with increased LPS production, gut microbiota, its biodiversity, symbiotic interactions, and meta-
impaired barrier function causes endotoxemia and systemic bolic activity, as well as on the intestinal wall in general and in-
inflammation. Moreover, Cd-induced alteration of gut lining may testinal epithelium in particular.
itself be a factor of microbiota impairment, whereas direct effect of
Cd on intestinal microbiome may in turn mediate its effects on
Acknowledgements
intestinal wall.
Hypothetically, Cd-induced increase gut permeability may also
This paper was financially supported by the Ministry of Educa-
result in increased bacterial translocation into the tissues as
tion and Science of the Russian Federation on the program to
observed in a number of studies (Condette et al., 2014; Earley et al.,
improve the competitiveness of Peoples' Friendship University of
2015). On the one hand, bacteriolysis may be associated with
Russia (RUDN University) among the world's leading research and
aggravation of endotoxemia (Ginsburg, 2004). At the same time,
education centers in 2016e2020.
together with Cd-induced impairment of macrophage inflamma-
tory response, increased bacterial translocation may result in
increased susceptibility to infections. It is also notable that Cd References
exposure decreases bacteriophage lytic activity (Dorozhko and Adams, S.V., Passarelli, M.N., Newcomb, P.A., 2011. Cadmium exposure and cancer
Rotshild, 1985), that may also decrease the rate of infectious mortality in the third national health and nutrition examination survey cohort.
resistance. Such a supposition is generally in agreement with the Occup. Environ. Med. https://doi.org/10.1136/oemed-2011-100111 oemed-2011.
Ba, Q., Li, M., Chen, P., Huang, C., Duan, X., Lu, L., Li, J., Chu, R., Xie, D., Song, H., Wu, Y.,
finding of higher susceptibility of Cd-exposed mice to infections
Ying, H., Jia, X., Wang, H., 2017. Sex-dependent effects of cadmium exposure in
(Simonyte et al., 2003; Rager et al., 2014). early life on gut microbiota and fat accumulation in mice. Environ. Health
Therefore, both impaired microbiome metabolic activity and Perspect. 125 (3), 437. https://doi.org/10.1289/EHP360.
LPS-induced systemic inflammation may have a significant impact Biagioli, M., Pifferi, S., Ragghianti, M., Bucci, S., Rizzuto, R., Pinton, P., 2008. Endo-
plasmic reticulum stress and alteration in calcium homeostasis are involved in
on target organs (Fig. 1). cadmium-induced apoptosis. Cell Calcium 43 (2), 184e195. https://doi.org/
In particular, it has been demonstrated that impaired gut 10.1016/j.ceca.2007.05.003C.
A.A. Tinkov et al. / Environmental Pollution 235 (2018) 429e434 433

Boveri, M., Pazos, P., Gennari, A., Casado, J., Hartung, T., Prieto, P., 2004. Comparison Ibrahim, F., Halttunen, T., Tahvonen, R., Salminen, S., 2006. Probiotic bacteria as
of the sensitivity of different toxicological endpoints in Caco-2 cells after cad- potential detoxification tools: assessing their heavy metal binding isotherms.
mium chloride treatment. Arch. Toxicol. 78 (4), 201e206. https://doi.org/ Can. J. Microbiol. 52 (9), 877e885. https://doi.org/10.1139/w06-043.
10.1007/s00204-003-0532-1. Jafarpour, D., Shekarforoush, S.S., Ghaisari, H.R., Nazifi, S., Sajedianfard, J., 2015.
Breton, J., Daniel, C., Dewulf, J., Pothion, S., Froux, N., Sauty, M., Thomas, P., Pot, B., Impact of synbiotic diets including inulin, Bacillus coagulans and Lactobacillus
Foligne , B., 2013a. Gut microbiota limits heavy metals burden caused by chronic plantarum on intestinal microbiota of rat exposed to cadmium and mercury.
oral exposure. Toxicol. Lett. 222 (2), 132e138. https://doi.org/10.1016/ Vet. Sci. Dev. 5 (2) https://doi.org/10.4081/vsd.2015.6061.
j.toxlet.2013.07.021. Jafarpour, D., Shekarforoush, S.S., Ghaisari, H.R., Nazifi, S., Sajedianfard, J.,
Breton, J., Daniel, C., Vignal, C., Body-Malapel, M., Garat, A., Ple , C., Foligne , B., 2016. Eskandari, M.H., 2017. Protective effects of synbiotic diets of Bacillus coagulans,
Does oral exposure to cadmium and lead mediate susceptibility to colitis? The Lactobacillus plantarum and inulin against acute cadmium toxicity in rats. BMC
dark-and-bright sides of heavy metals in gut ecology. Sci. Rep.-UK 6. https:// Complem. Alternative Med. 17(1) (291) https://doi.org/10.1186/s12906-017-
doi.org/10.1038/srep19200. 1803-3.
Breton, J., Massart, S., Vandamme, P., De Brandt, E., Pot, B., Foligne , B., 2013. Eco- Jama, A.M., Miti 
c-Culafi 
c, D., Kolarevi c, S., Ðurasevi
c, S.F., Knezevi
c-Vuk cevi
c, J.,
toxicology inside the gut: impact of heavy metals on the mouse microbiome. 2012. Protective effect of probiotic bacteria against cadmium-induced geno-
BMC Pharmacol. Toxico. 14 (1), 62. https://doi.org/10.1186/2050-6511-14-62. toxicity in rat hepatocytes in vivo and in vitro. Arch. Biol. Sci. 64 (3), 1197e1206.
Cani, P.D., Amar, J., Iglesias, M.A., Poggi, M., Knauf, C., Bastelica, D., Neyrink, A.M., https://doi.org/10.2298/ABS1203197J.
Fava, F., Tuohy, K.M., Chabo, C., Waget, A., Delme e, E., Cousin, B., Sulpice, T., Jin, Y., Liu, L., Zhang, S., He, R., Wu, Y., Chen, G., Fu, Z., 2016. Cadmium exposure to
Chamontin, B., Ferrie res, J., Tanti, J.F., Gibson, G.R., Casteilla, L., Delzenne, N.M., murine macrophages decreases their inflammatory responses and increases
Alessi, M.C., Burcelin, R., 2007. Metabolic endotoxemia initiates obesity and their oxidative stress. Chemosphere 144, 168e175. https://doi.org/10.1016/
insulin resistance. Diabetes 56 (7), 1761e1772. https://doi.org/10.2337/db06- j.chemosphere.2015.08.084.
1491. Jin, Y., Wu, S., Zeng, Z., Fu, Z., 2017. Effects of environmental pollutants on gut
Condette, C.J., Khorsi-Cauet, H., Morlie re, P., Zabijak, L., Reygner, J., Bach, V., Gay- microbiota. Environ. Pollut. 222, 1e9. https://doi.org/10.1016/
Que heillard, J., 2014. Increased gut permeability and bacterial translocation j.envpol.2016.11.045.
after chronic chlorpyrifos exposure in rats. PLos One 9 (7), e102217. https:// Kazantzis, G., 2004. Cadmium, osteoporosis and calcium metabolism. Biometals 17
doi.org/10.1371/journal.pone.0102217. (5), 493e498. https://doi.org/10.1023/B: BIOM.0000045727.76054.f3.
Cox, J.N., Rahman, M.A., Bao, S., Liu, M., Wheeler, S.E., Knoell, D.L., 2016. Cadmium Kim, E., Jee, J., Steiner, H., Cormet-Boyaka, E., Boyaka, P., 2014. Chronic exposure to
attenuates the macrophage response to LPS through inhibition of the NF-kB cadmium alters gut immune homeostasis and innate immunity (MUC8P. 810).
pathway. Am. J. Physiol.-Lung C. 311 (4), L754eL765. https://doi.org/10.1152/ J. Immunol. 192 (1 Supplement), 198.11.
ajplung.00022.2016. Kim, E., Lembert, M.M., Opiyo, S.O., Ahmer, B.M., Cormet-Boyaka, E., Boyaka, P.N.,
Djurasevic, S., Jama, A., Jasnic, N., Vujovic, P., Jovanovic, M., Mitic-Culafic, D., Kne- 2017. Differential role of oxidative stress pathways and microbiota in the
zevic-Vukcevic, J., Cakic-Milosevic, M., Ilijevic, K., Djordjevic, J., 2017. The pro- development of allergen specific IgE following chronic ingestion of low doses of
tective effects of probiotic bacteria on cadmium toxicity in rats. J. Med. Food 20 cadmium. J. Immunol. 198 (1 Supplement), 194.5.
(2), 189e196. https://doi.org/10.1089/jmf.2016.0090. Kim, E., Xu, X., Steiner, H., Ahmer, B., Cormet-Boyaka, E., Boyaka, P., 2015. Chronic
Dorozhko, O.V., Rotshild, E.V., 1985. Trace elements in the life of pathogenic mi- ingestion of low doses of cadmium alters the gut microbiome and immune
croorganisms and some other ones. Usp. Sovrem. Biol. 99 (2), 313e319 [in Russ]. homeostasis to enhance allergic sensitization (MUC9P. 743). J. Immunol. 194 (1
Duizer, E., Gilde, A.J., Versantvoort, C.H., Groten, J.P., 1999. Effects of cadmium Supplement), 205.7.
chloride on the paracellular barrier function of intestinal epithelial cell lines. Koeth, R.A., Wang, Z., Levison, B.S., Buffa, J.A., Org, E., Sheehy, B.T., Britt, E.B., Fu, X.,
Toxicol. Appl. Pharmacol. 155 (2), 117e126. https://doi.org/10.1006/ Wu, Y., Li, L., Smith, J.D., DiDonato, J.A., Chen, J., Li, H., Wu, G.D., Lewis, J.D.,
taap.1998.8589. Warrier, M., Brown, J.M., Krauss, R.M., Wilson Tang, W.H., Bushman, F.D.,
Dutta, G., Zhang, P., Liu, B., 2008. The lipopolysaccharide Parkinson's disease animal Lusis, A.J., Hazen, S.L., 2013. Intestinal microbiota metabolism of L-carnitine, a
model: mechanistic studies and drug discovery. Fund. Clin. Pharmacol. 22 (5), nutrient in red meat, promotes atherosclerosis. Nat. Med. 19 (5), 576e585.
453e464. https://doi.org/10.1111/j.1472-8206.2008.00616.x. https://doi.org/10.1038/nm.3145.
Earley, Z.M., Akhtar, S., Green, S.J., Naqib, A., Khan, O., Cannon, A.R., Hammer, A.M., Koropatnick, J., Zalups, R.K., 1997. Effect of non-toxic mercury, zinc or cadmium
Morris, N.L., Li, X., Eberhardt, J.M., Gamelli, R.M., Kennedy, R.H., Choudhry, M.A., pretreatment on the capacity of human monocytes to undergo
2015. Burn injury alters the intestinal microbiome and increases gut perme- lipopolysaccharide-induced activation. Br. J. Pharmacol. 120 (5), 797e806.
ability and bacterial translocation. PLos One 10 (7), e0129996. https://doi.org/ https://doi.org/10.1038/sj.bjp.0700975.
10.1371/journal.pone.0129996. Kumar, N., Kumar, V., Panwar, R., Ram, C., 2017. Efficacy of indigenous probiotic
Fasano, A., 2011. Zonulin and its regulation of intestinal barrier function: the bio- Lactobacillus strains to reduce cadmium bioaccessibility-an in vitro digestion
logical door to inflammation, autoimmunity, and cancer. Physiol. Rev. 91 (1), model. Environ. Sci. Pollut. Res. 24 (2), 1241e1250. https://doi.org/10.1007/
151e175. https://doi.org/10.1152/physrev.00003.2008. s11356-016-7779-6.
Fazeli, M., Hassanzadeh, P., Alaei, S., 2011. Cadmium chloride exhibits a profound Lacerda, C.M., Choe, L.H., Reardon, K.F., 2007. Metaproteomic analysis of a bacterial
toxic effect on bacterial microflora of the mice gastrointestinal tract. Hum. Exp. community response to cadmium exposure. J. Proteome Res. 6 (3), 1145e1152.
Toxicol. 30 (2), 152e159. https://doi.org/10.1177/0960327110369821. https://doi.org/10.1021/pr060477v.
Ferraro, P.M., Costanzi, S., Naticchia, A., Sturniolo, A., Gambaro, G., 2010. Low level Li, Y., Liu, K., Shen, J., Liu, Y., 2016. Wheat bran intake can attenuate chronic cad-
exposure to cadmium increases the risk of chronic kidney disease: analysis of mium toxicity in mice gut microbiota. Food Funct. 7 (8), 3524e3530. https://
the NHANES 1999-2006. BMC Publ. Health 10 (1), 304. https://doi.org/10.1186/ doi.org/10.1039/C6FO00233A.
1471-2458-10-304. Liu, J., Qu, W., Kadiiska, M.B., 2009. Role of oxidative stress in cadmium toxicity and
Fukui, H., 2015. Gut-liver axis in liver cirrhosis: how to manage leaky gut and carcinogenesis. Toxicol. Appl. Pharmacol. 238 (3), 209e214. https://doi.org/
endotoxemia. World J. Hepatology 7 (3), 425. https://doi.org/10.4254/ 10.1016/j.taap.2009.01.029.
wjh.v7.i3.425. Liu, Y., Li, Y., Liu, K., Shen, J., 2014. Exposing to cadmium stress cause profound toxic
Gerbino, E., Carasi, P., Tymczyszyn, E.E., Go  mez-Zavaglia, A., 2014. Removal of effect on microbiota of the mice intestinal tract. PLos One 9 (2), e85323. https://
cadmium by Lactobacillus kefir as a protective tool against toxicity. J. Dairy Res. doi.org/10.1371/journal.pone.0085323.
81 (3), 280e287. https://doi.org/10.1017/S0022029914000314. Marchesi, J.R., Adams, D.H., Fava, F., Hermes, G.D., Hirschfield, G.M., Hold, G.,
Ginsburg, I., 2004. Bactericidal cationic peptides can also function as bacteriolysis- Quraishi, M.N., Kinross, J., Smidt, H., Tuohy, T.M., Thomas, L.V., Zoetendal, E.G.,
inducing agents mimicking beta-lactam antibiotics?; it is enigmatic why this Hart, A., 2015. The gut microbiota and host health: a new clinical frontier. Gut
concept is consistently disregarded. Med. Hypotheses 62 (3), 367e374. https:// 2015 (0), 1e10. https://doi.org/10.1136/gutjnl-2015-309990.
doi.org/10.1016/j.mehy.2003.11.017. McIntyre, C.W., Harrison, L.E., Eldehni, M.T., Jefferies, H.J., Szeto, C.C., John, S.G.,
Goldsmith, J.R., Sartor, R.B., 2014. The role of diet on intestinal microbiota meta- Sigrist, M.K., Burton, J.O., Hothi, D., Korsheed, S., Owen, P.J., Lai, K.B., Li, P.K.T.,
bolism: downstream impacts on host immune function and health, and ther- 2011. Circulating endotoxemia: a novel factor in systemic inflammation and
apeutic implications. J. Gastroenterol. 49 (5), 785e798. https://doi.org/10.1007/ cardiovascular disease in chronic kidney disease. Clin. J. Am. Soc. Nephrol. 6 (1),
s00535-015-1043-6. 133e141. https://doi.org/10.2215/CJN.04610510.
Han, S.J., Ha, K.H., Jeon, J.Y., Kim, H.J., Lee, K.W., Kim, D.J., 2015. Impact of cadmium Meharg, A.A., Norton, G., Deacon, C., Williams, P., Adomako, E.E., Price, A., Zhu, Y.,
exposure on the association between lipopolysaccharide and metabolic syn- Li, G., Zhao, F.J., McGrath, S., Villada, A., Sommella, A., De Silva, P.M.C.S.,
drome. Int. J. Environ. Res. Publ. Health 12 (9), 11396e11409. https://doi.org/ Brammer, H., Dasgupta, T., Islam, R.L., 2013. Variation in rice cadmium related to
10.3390/ijerph120911396. human exposure. Environ. Sci. Technol. 47 (11), 5613e5618. https://doi.org/
Hartwig, A., 2013. Cadmium and Cancer. In Cadmium: from Toxicity to Essentiality. 10.1021/es400521h.
Springer Netherlands, pp. 491e507. https://doi.org/10.1007/978-94-007-5179- Min, J.Y., Min, K.B., 2016. Blood cadmium levels and Alzheimer's disease mortality
8_15. risk in older US adults. Environ. Health-Glob. 15(1) (69) https://doi.org/10.1186/
Hyder, O., Chung, M., Cosgrove, D., Herman, J.M., Li, Z., Firoozmand, A., Gurakar, A., s12940-016-0155-7.
Koteish, A., Pawlik, T.M., 2013. Cadmium exposure and liver disease among US Moulis, J.M., 2010. Cellular mechanisms of cadmium toxicity related to the ho-
adults. J. Gastrointest. Surg. 17 (7), 1265e1273. https://doi.org/10.1007/s11605- meostasis of essential metals. Biometals 23 (5), 877e896. https://doi.org/
013-2210-9. 10.1007/s10534-010-9336-y.
Hyun, J.S., Satsu, H., Shimizu, M., 2007. Cadmium induces Interleukin-8 production Nai, G.A., Gonçalves Filho, M.A., Estrella, M.P.S., Teixeira, L.D.S., 2015. Study of the
via NF-kB activation in the human intestinal epithelial cell, Caco-2. Cytokine 37 influence of the ph of water in the initiation of digestive tract injury in cad-
(1), 26e34. https://doi.org/10.1016/j.cyto.2007.02.011. mium poisoning in rats. Toxicology Reports 2, 1033e1038. https://doi.org/
434 A.A. Tinkov et al. / Environmental Pollution 235 (2018) 429e434

10.1016/j.toxrep.2015.07.012. Takiguchi, M., Yoshihara, S.I., 2006. New aspects of cadmium as endocrine disruptor.
Nawrot, T.S., Staessen, J.A., Roels, H.A., Munters, E., Cuypers, A., Richart, T., Environmental sciences: an international journal of environmental physiology
Ruttens, A., Smeets, K., Clijsters, H., Vangronsveld, J., 2010. Cadmium exposure and toxicology 13 (2), 107e116.
in the population: from health risks to strategies of prevention. Biometals 23 Tellez-Plaza, M., Navas-Acien, A., Caldwell, K.L., Menke, A., Muntner, P., Guallar, E.,
(5), 769e782. https://doi.org/10.1007/s10534-010-9343-z. 2012a. Reduction in cadmium exposure in the United States population,
Neves, A.L., Coelho, J., Couto, L., Leite-Moreira, A., Roncon-Albuquerque, R., 2013. 1988e2008: the contribution of declining smoking rates. Environ. Health Per-
Metabolic endotoxemia: a molecular link between obesity and cardiovascular spect. 120 (2), 204. https://doi.org/10.1289/ehp.1104020.
risk. J. Mol. Endocrinol. 51 (2), R51eR64. https://doi.org/10.1530/JME-13-0079. Tellez-Plaza, M., Navas-Acien, A., Menke, A., Crainiceanu, C.M., Pastor-Barriuso, R.,
Ninkov, M., Aleksandrov, A.P., Demenesku, J., Mirkov, I., Mileusnic, D., Petrovic, A., Guallar, E., 2012b. Cadmium exposure and all-cause and cardiovascular mor-
Grigorov, I., Zolotarevski, L., Tolinacki, M., Kataranovski, D., Brceski, I., tality in the US general population. Environ. Health Perspect. 120 (7), 1017.
Kataranovski, M., 2015. Toxicity of oral cadmium intake: impact on gut im- https://doi.org/10.1289/ehp.1104352.
munity. Toxicol. Lett. 237 (2), 89e99. https://doi.org/10.1016/ Tinkov, A.A., Filippini, T., Ajsuvakova, O.P., Aaseth, J., Gluhcheva, Y.G., Ivanova, J.M.,
j.toxlet.2015.06.002. Bjørklund, G., Skalnaya, M.G., Gatiatulina, E.R., Popova, E.V., Nemereshina, O.N.,
Ninkov, M., Aleksandrov, A.P., Mirkov, I., Demenesku, J., Mileusnic, D., Stojanov, S.J., Vinceti, M., Skalny, A.V., 2017. The role of cadmium in obesity and diabetes. Sci.
Golic, N., Tolinacki, M., Zolotarevski, L., Kataranovski, D., Brceski, I., Total Environ. 601, 741e755. https://doi.org/10.1016/j.scitotenv.2017.05.224.
Kataranovski, M., 2016. Strain differences in toxicity of oral cadmium intake in Tremaroli, V., Ba €ckhed, F., 2012. Functional interactions between the gut microbiota
rats. Food Chem. Toxicol. 96, 11e23. https://doi.org/10.1016/j.fct.2016.07.021. and host metabolism. Nature 489 (7415), 242e249. https://doi.org/10.1038/
Olszowski, T., Baranowska-Bosiacka, I., Gutowska, I., Chlubek, D., 2012. Pro-in- nature11552.
flammatory properties of cadmium. Acta Biochim. Pol. 59 (4), 475e482. Trompette, A., Gollwitzer, E.S., Yadava, K., Sichelstiel, A.K., Sprenger, N., Ngom-
Pan, J., Plant, J.A., Voulvoulis, N., Oates, C.J., Ihlenfeld, C., 2010. Cadmium levels in Bru, C., Blanchard, C., Junt, T., Nicod, L.P., Harris, N.L., Marsland, B.J., 2014. Gut
Europe: implications for human health. Environ. Geochem. Health 32 (1), 1e12. microbiota metabolism of dietary fiber influences allergic airway disease and
https://doi.org/10.1007/s10653-009-9273-2. hematopoiesis. Nat. Med. 20 (2), 159e166. https://doi.org/10.1038/nm.3444.
Qin, L., Wu, X., Block, M.L., Liu, Y., Breese, G.R., Hong, J.S., Knapp, D.J., Crews, F.T., Trøseid, M., 2017. Gut microbiota and acute coronary syndromes: ready for use in
2007. Systemic LPS causes chronic neuroinflammation and progressive neuro- the emergency room? Eur. Heart J. 38 (11), 825e827. https://doi.org/10.1093/
degeneration. Glia 55 (5), 453e462. https://doi.org/10.1002/glia.20467. eurheartj/ehx005.
Rager, J.E., Yosim, A., Fry, R.C., 2014. Prenatal exposure to arsenic and cadmium Wang, A., Crowley, D.E., 2005. Global gene expression responses to cadmium
impacts infectious disease-related genes within the glucocorticoid receptor toxicity in Escherichia coli. J. Bacteriol. 187 (9), 3259e3266. https://doi.org/
signal transduction pathway. Int. J. Mol. Sci. 15 (12), 22374e22391. https:// 10.1128/JB.187.9.3259-3266.2005.
doi.org/10.3390/ijms151222374. Wang, Y., Chen, L., Gao, Y., Zhang, Y., Wang, C., Zhou, Y., Hu, Y., Shi, R., Tian, Y., 2016.
Rani, A., Kumar, A., Lal, A., Pant, M., 2014. Cellular mechanisms of cadmium-induced Effects of prenatal exposure to cadmium on neurodevelopment of infants in
toxicity: a review. Int. J. Environ. Health Res. 24 (4), 378e399. https://doi.org/ Shandong, China. Environ. Pollut. 211, 67e73. https://doi.org/10.1016/
10.1080/09603123.2013.835032. j.envpol.2015.12.038.
Round, J.L., Mazmanian, S.K., 2009. The gut microbiota shapes intestinal immune We˛ gielska, I., Suliburska, J., 2016. The role of intestinal microbiota in the patho-
responses during health and disease. Nat. Rev. Immunol. 9 (5), 313e323. genesis of metabolic diseases. Acta Sci. Pol. Technol. Aliment. 15 (2), 201e211.
https://doi.org/10.1038/nri2515. https://doi.org/10.17306/J.AFS.2016.2.20.
Rusanov, A.L., Smirnova, A.V., Poromov, A.A., Fomicheva, K.A., Luzgina, N.G., Zhai, Q., Li, T., Yu, L., Xiao, Y., Feng, S., Wu, J., Zhao, J., Zhang, H., Chen, W., 2017.
Majouga, A.G., 2015. Effects of cadmium chloride on the functional state of Effects of subchronic oral toxic metal exposure on the intestinal microbiota of
human intestinal cells. Toxicol. Vitro 29 (5), 1006e1011. https://doi.org/10.1016/ mice. Sci. Bull. https://doi.org/10.1016/j.scib.2017.01.031.
j.tiv.2015.03.018. Zhai, Q., Tian, F., Zhao, J., Zhang, H., Narbad, A., Chen, W., 2016. Oral administration
Satarug, S., Baker, J.R., Reilly, P.E., Esumi, H., Moore, M.R., 2000. Evidence for a of probiotics inhibits absorption of the heavy metal cadmium by protecting the
synergistic interaction between cadmium and endotoxin toxicity and for nitric intestinal barrier. Appl. Environ. Microbiol. 82 (14), 4429e4440. https://doi.org/
oxide and cadmium displacement of metals in the kidney. NATO Sci. S. A. Lif. Si. 10.1128/AEM.00695-16.
4 (4), 431e440. https://doi.org/10.1006/niox.2000.0295. Zhai, Q., Wang, G., Zhao, J., Liu, X., Narbad, A., Chen, Y.Q., Zhang, H., Tian, F., Chen, W.,
Satarug, S., Garrett, S.H., Sens, M.A., Sens, D.A., 2011. Cadmium, environmental 2014. Protective effects of Lactobacillus plantarum CCFM8610 against chronic
exposure, and health outcomes. Cienc. Saude Coletiva 16 (5), 2587e2602. cadmium toxicity in mice: intestinal sequestration is not the only route of
https://doi.org/10.1590/S1413-81232011000500029. protection. Appl. Environ. Microbiol. 80 (13), 4063e4071. https://doi.org/
Scott, K.P., Jean-Michel, A., Midtvedt, T., van Hemert, S., 2015. Manipulating the gut 10.1128/AEM.00762-14, 2014 Jul.
microbiota to maintain health and treat disease. Microb. Ecol. Health Dis. 26 (1), Zhai, Q., Wang, G., Zhao, J., Liu, X., Tian, F., Zhang, H., Chen, W., 2013. Protective
25877. effects of Lactobacillus plantarum CCFM8610 against acute cadmium toxicity in

Simonyte, S., Cerka sin, G., Plan  niene,
ciu _ R., Naginiene,
_ R., Ryselis, S., Ivanov, L., 2003. mice. Appl. Environ. Microbiol. 79 (5), 1508e1515. https://doi.org/10.1128/
Influence of cadmium and zinc on the mice resistance to Listeria mono- AEM.03417-12.
cytogenes infection. Medicina 39 (8), 767e772. Zhang, S., Jin, Y., Zeng, Z., Liu, Z., Fu, Z., 2015. Subchronic exposure of mice to cad-
Skrypnik, K., Suliburska, J., 2017. Association between the gut microbiota and mium perturbs their hepatic energy metabolism and gut microbiome. Chem.
mineral metabolism. J. Sci. Food Agric. https://doi.org/10.1002/jsfa.8724. Res. Toxicol. 28 (10), 2000e2009. https://doi.org/10.1021/
Srinivasan, P., Li, Y.H., Hsu, D.Z., Su, S.B., Liu, M.Y., 2011. Ostensibly ineffectual doses acs.chemrestox.5b00237.
of cadmium and lipopolysaccharide causes liver damage in rats. Hum. Exp. Zhang, W., Zheng, J., Han, X., Yao, D., Han, J., 2016. Comparison of dietary cadmium
Toxicol. 30 (7), 624e635. https://doi.org/10.1177/0960327110376553. exposure among the general population from two cadmium-polluted regions in
Sturgeon, C., Fasano, A., 2016. Zonulin, a regulator of epithelial and endothelial China. J. Pollut. Eff. Cont. 4(167) (2) https://doi.org/10.4172/2375-4397.1000167.
barrier functions, and its involvement in chronic inflammatory diseases. Tissue Zhao, Z., Hyun, J.S., Satsu, H., Kakuta, S., Shimizu, M., 2006. Oral exposure to cad-
Barriers 4 (4), e1251384. https://doi.org/10.1080/21688370.2016.1251384. mium chloride triggers an acute inflammatory response in the intestines of
Sturniolo, G.C., Fries, W., Mazzon, E., Di Leo, V., Barollo, M., D'inca, R., 2002. Effect of mice, initiated by the over-expression of tissue macrophage inflammatory
zinc supplementation on intestinal permeability in experimental colitis. J. Lab. protein-2 mRNA. Toxicol. Lett. 164 (2), 144e154. https://doi.org/10.1016/
Clin. Med. 139 (5), 311e315. https://doi.org/10.1067/mlc.2002.123624. j.toxlet.2005.12.004.

You might also like