Los microARN Como Posibles Biomarcadores en Enfermedades y Toxicología

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Mutation Research 764–765 (2014) 46–57

Contents lists available at ScienceDirect


Mutation Research/Genetic Toxicology and
Environmental Mutagenesis
journal homepage: www.elsevier.com/locate/gentox
Community address: www.elsevier.com/locate/mutres

MicroRNAs as potential biomarkers in diseases and toxicology


Bénazir Siddeek a,b,f , Lilia Inoubli a,b , Nadjem Lakhdari a,b , Paul Bellon Rachel a,b ,
Karma Claire Fussell g , Steffen Schneider g , Claire Mauduit a,b,c,d ,
Mohamed Benahmed a,b,e,∗
a
Inserm, U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 5, Nice, F-06204, France
b
Université de Nice Sophia-Antipolis, UFR Médecine, Nice, F-06000, France
c
Université Lyon 1, UFR Médecine Lyon Sud, Lyon, F-69921, France
d
Hospices Civils de Lyon, Hôpital Lyon Sud, laboratoire d’anatomie et de cytologie pathologiques, Pierre-Bénite, F-69495, France
e
Centre Hospitalier Universitaire de Nice, Pôle Digestif, Gynécologie, Obstetrique, Centre de Reproduction, Nice, F-06202, France
f
BASF Agro, Ecully F-69130, France
g
BASF SE, experimental toxicology and ecology, 67056 Ludwigshafen, Germany

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

Article history: MiRNAs (microRNAs) are single-stranded non-coding RNAs of approximately 21–23 nucleotides in length
Received 7 January 2014 whose main function is to inhibit gene expression by interfering with mRNA processes. MicroRNAs
Received in revised form 20 January 2014 suppress gene expression by affecting mRNA (messenger RNAs) stability, targeting the mRNA for degra-
Accepted 20 January 2014
dation, or both. In this review, we have examined how microRNA expression could be altered following
Available online 30 January 2014
exposure to chemicals and how they could represent appropriate tissue and more interestingly circu-
lating biomarkers. Among the key questions before using the microRNA for evaluation of risk toxicity, it
Keywords:
remains still to clarify how they could be causally involved in the adverse effects and how stable their
MicroRNAs
Chemicals
changes are.
Circulating biomarkers © 2014 Published by Elsevier B.V.

1. Introduction In animals, siRNA typically binds perfectly to its mRNA target, a


perfect match to the sequence, whereas miRNA can inhibit the
Small RNAs of 20–30 nucleotides continue to be discovered, translation of many different mRNA sequences because its pair-
including some that are specific to plants or animal lineages (for ing is imperfect. MiRNAs suppress gene expression by affecting
reviews, see [1,2]). Various small RNAs with distinctive charac- mRNA stability, targeting the mRNA for degradation, or both [4].
teristics have been identified and can be classified into three Near to 1872 miRNAs have been identified in human with the
classes based on their biogenesis mechanisms, the type of RNA potential to regulate the expression of about two-third of human
protein that they are associated with and the nature of their tar- mRNAs and influence almost all genetic pathways. The possibilities
gets: microRNAs (miRNAs), endogenous small interfering RNAs of predictions for miRNA-target recognition have been developed
(endo-siRNAs) and Piwi-interacting RNAs (piRNAs). Endogenous using computer modeling [5]. A growing number of data show that
siRNA are short double-stranded RNAs that contains 21–23 nucleic microRNAs are involved in the expression of genes involved in
acids with a 19-nucleotide duplex region which is able to inhibit xenobiotic exposure [6–8]. Better understanding of the non-coding
the gene expression of specific proteins by a mechanism called RNAs is critical to determine its potential key role in the impact of
RNA interference. The piRNAs are highly enriched in the animal environmental factors and particularly xenobiotic exposure [9] (for
germline only and originate from single-stranded precursor RNAs a review, see [10]).
[3]. MiRNAs are single-stranded non-coding RNAs of approximately Since their discovery, there is a growing interest in miRNAs
21–23 nucleotides in length which main function is to inhibit gene research allowed by the development of efficient and sensitive tools
expression by interfering with messenger RNA (mRNA) processes. for their study. MiRNAs can be extracted from tissues and quanti-
fied easily by RT-qPCR even with low amount of input material [11].
They can also be detected by hybridization (Northern, Panomics,
Nanostring, MicroArrays) ([12,13] or by Next generation sequenc-
∗ Corresponding author at: INSERM (National Institut of Health and Medical
ing [14]. Since a decade, a number of studies are pointing out the
Research), U1065, Team 5, Bâtiment Universitaire Archimed, Centre Méditerranéen
de Médecine Moléculaire (C3M), 151 route Saint-Antoine Ginestière, BP 2 3194,
role of miRNAs in the regulation of key cellular processes and the
06204 Nice Cedex 3, France. Tel.: +33 492 035 630; fax: +33 492 035 640. appearance of pathologies. MicroRNAs (miRNAs) have been impli-
E-mail address: benahmed.m@chu-nice.fr (M. Benahmed). cated in posttranscriptional regulation of many gene expressions

1383-5718/$ – see front matter © 2014 Published by Elsevier B.V.


http://dx.doi.org/10.1016/j.mrgentox.2014.01.010
B. Siddeek et al. / Mutation Research 764–765 (2014) 46–57 47

Table 1
Effects of drugs, natural and chemical compounds on miRNAs expression.

Compound Organ/Cell Species Altered miRNAs References

Cigarette smoke Spermatozoa Human Mir-340, mir-365, 129-3p, 634 [68]


Volatile organic compounds Lung Mouse miR-1187, miR-125a-3p, miR-125b-5p, miR-466c-5p, [100]
(formaldehyde, benzene, toluene, and miR-5105, miR-3472
xylene)
Trichostatin A Primary hepatocyte Rat miR-379, miR-143, miR-122, miR-122, miR-143, miR-379 [101]
Sodium arsenite TK6 cell line miR-222, miR-210, [102]
Arsenic trioxide T24 cell line mir-19a [103]
Cadmium Human Mir-146 [95]
Aluminum-sulfate HN cells Mir-146 [104]
Aluminum-sulfate HN cells miR-9, -128, -125b [105]
Air pollution metal-rich PM Leucocytes Human Mir-222, mir-21 [106]
Cigarette smoke Lung Rat Mir-294, let-7c, miR-34c, -222 [107]
Cigarette smoke Lung Human Mir-218 [108]
4-(methylnitrosamino)-1-(3-pyridyl)-1- Lung Rat Mir-126, Mir-34 [109]
butanone (NNK) (tobacco
carcinogen)
Hexahydro-1,3,5-trinitro-1,3,5-triazine Liver Mouse let-7, miR-15, -16, -26, -181, miR-10b [110]
Hexahydro-1,3,5-trinitro-1,3,5-triazine Neuron Mouse miR-206, -30, -195 [110]
Carbon tetrachloride Hepatocytes Rat miR-298, -370 [111]
Dioxins Hepatocytes Rat Mir-191 [112]
Bisphenol A 3A placental cell Human Mir-146a [113]
Wy-14, 643 Hepatocyte Mouse Let-7c [114]
Estradiol benzoate Testis Rat Mir-29 [66]
Ethanol Neurons Mouse Mir-21, mir-335,mir-153 [115]
Tamoxyfen Hepatocyte Rat Mir-17-92 cluster, mir-106a, mir-34 [116]
Acetaminophen; carbon tetrachloride Hepatocytes Mir-298, mir-370 [111]
Iron – and aluminum – sulfate Neuron Human Mir-9, mir-125b, mir-128 [105]
5-fluorouracil (5-FU) (drug) Colon Human Mir-200b [117]
Residual oil fly ash Heart Rat miR-1 and miR-133, miR-21, miR-24, and miR-29 [118]
Vorinostat, myo-inositol, bexarotene, Lung Mouse [119]
pioglitazone (cigarette smoke)
6-Mercaptopurine (immunosuppressive Placenta Rat miR-195, miR-21, miR-29c and miR-34a, 146bmiR-144 [120]
drug) and miR-451
4-(methylnitrosamino)-1-(3-pyridyl)-1- Lung Rat miR-206 and miR-133b [121]
butanone
Radon Lung BEAS2B cell line Human hsa-miR-483-3p, hsa-miR-494, hsa-miR-2115*, [122]
hsa-miR-33b, hsa-miR-1246, hsa-miR-3202, hsa-miR-18a,
hsa-miR-125b, hsa-miR-17*, and hsa-miR-886-3p
7,12-dimethylbenz(␣)anthracene (DMBA) In the liver, spleen and Mouse miR-34a and miR-155 miR-21 [123]
and N-methyl-N-nitrosourea (MNU) kidneys
Acetaminophen, allyl alcohol, and Liver Rat miR-122 [124]
␣-naphthyl isothiocyanate
2,3,7,8-tetrachlorodibenzo-p-dioxin Thymus Mouse miR-122 and miR-181a miR-23a, miR-18b, miR-31 [125]
(environmental contaminant) miR-182
2,3,7,8-tetrachlorodibenzo-p-dioxin Embryo Zebrafish miR-27e [126]
Doxorubicin Heart Mouse 208b, miR-216b, miR-215, miR-34c and miR-367 [127]
Cyanobacterial hepatotoxin microcystin-LR Whitefish let-7c, miR-9b), (miR-16a, miR-21a, miR-34a) (miR-122) [128]
Ethanol Liver Rat Mir-21 [129]
Printex 90 carbon black nanoparticles Lung Mouse miR-135b [130]
Arsenic Umbilical endothelial Human [128]
cell
Arsenic Liver Mouse [131]
Acetaminophen Liver Mouse 74-5p, 135a*, 466g, 1196, 466f-3p, 877, 342-3p, 195, 375, [132]
29c, 148a, 652
Gentamicin Kidney Mouse Mir-21, 155 [133]
2-Amino-1-methyl-6-phenylimidazo[4,5- Colon Rat Let-7 [134]
b]pyridine (carcinogen from cooked
meat)
Perfluorooctane sulfonic acid Brain Rat miR-466b, -672, and -297 [135]
Cadmium HepG2, liver Human Let-7 [136]
Cisplatin Hela [137]
Folate deprivation and arsenic exposure Lymphocyte Human Mir-222 [102]
Benzo[k]fluoranthene Hepatocyte Human miR-146a, miR-365, let-7f, miR-199b-5p,miR-30c-1* [138]
Thapsigargin, deoxycholic acid Hepatocytes Mouse mir-199a-5p [139]
Fenhexamid and fludioxonil (fungicide) Breast cancer cells Human Mir-21, miR-125b and miR-181a [140]
DDT and BPA Breast cancer cells Human Mir-21 [141]
Nonylphenol Sertoli cells, testis Mouse miR-135a* and miR-199a-5p [142]
Pegylated leptin antagonist Hypothalamus Rat miR-10a, miR-200a, rno-miR-409-5p, miR-125a-3p [143]
Lycopene Liver Rat Mir-21 [144]
Licorice flavonoid Liver Mouse Mir-122 [145]
Quercetin Liver Mouse miR-122 and miR-125b [146]
Ethanol Liver Mouse Mir-217 [147]
Allyl-isothiocyanate Macrophage Mouse Mir-155 [148]
High fat diet Adipocytes Mouse Mir-21 [149]
Conjugated linoleic acid Adipocytes miR-103 mir-107, miR-221, mir-222 [150]
High fat diet Adipocytes Mouse Mir-143 [151]
48 B. Siddeek et al. / Mutation Research 764–765 (2014) 46–57

and control of different processes such as apoptosis, DNA repair,


oxidative stress response, cancer and cellular development. Those
discoveries in miRNAs research open new insights in toxicology and
raise the question of miRNAs regulation in response to exposure to
environmental chemical toxicants or natural compounds such as
alcohol, smoke, stress, hormones, medication, and diets (Table 1).
This would facilitate the bio-monitoring and preventive strategies
for drug development, and clinical pharmacotherapy.
This review describes the current knowledge on the miRNAs
expression, their potential changes in toxicological studies and
circulating miRNA as potential toxicological biomarkers will be
discussed.

2. MiRNA expression (Fig. 1)

The stem-loop structures from which miRNAs are derived


are disseminated throughout the genome, either within intronic
sequences of protein-coding genes, within intronic or exonic
regions of non-coding RNAs, or set between independent tran-
scription units (inter-genic). The location of some of these intronic
miRNAs is evolutionarily conserved and they are similarly coex-
pressed with their host genes in different animals. Here, we review
the recent advances in miRNA biogenesis in animals. Particularly,
we focus on the roles of miRNAs in vertebrate physiology. The
majority of intronic miRNAs are transcribed from the same pro-
moter as the host gene. However, approximately 35% of intronic
miRNAs have upstream regulatory elements consistent with pro-
moter function [15]. Pri-miRNAs are mostly transcribed by RNA
polymerase II (Pol II) [16], and to a less extent by Pol III [17].
Their promoters also contain transcription start sites, CpG islands,
expression sequence tags, and conserved transcription factor bind-
ing sites, enhancers and silencers [18,19]. MiRNA biogenesis is Fig. 1. MiRNA biogenesis. MicroRNA (miRNA) genes are transcribed by RNA poly-
merase II (Pol II) to generate the primary transcripts (pri-miRNAs). Cleavage of the
based on series of processing steps to convert the primary miRNA
pri-miRNA occurs within the nucleus by Drosha and DGCR8 complex (also known as
(pri-miRNA) transcript into the biologically active, mature miRNA the Microprocessor complex) which interact with helicases p68 and p72, to generate
(Fig. 1), [for a review see [1,20]]. Firstly, the transcribed pri-miRNA a ∼65 nucleotide pre-miRNAs. Pre-miRNA has a ∼2-nt 3 overhang, which is recog-
is cleaved by the RNase III-like enzyme Drosha in the nucleus [21] nized by the nuclear export factor exportin 5. Once export through the nuclear pore
generating a miRNA precursor about 60–70 nt called pre-miRNA. complex into the cytoplasm, the RNase III Dicer supported by the proteins TRBP or
PACT catalyses the second processing step to produce miRNA duplexes. Dicer, TRBP
Cleavage by Drosha requires the co-factor DGCR8 (DiGeorge critical or PACT and Argonaute (AGO 1–4) mediate the processing of pre-miRNA and the
region 8), also known as Pasha [22]. The miRNA biogenesis machin- assembly of the RISC (RNA-induced silencing complex). One strand of the duplex is
ery is also subjected to regulation by feedback loops, as evidenced preferentially retained by the Ago protein as the mature miRNA, whereas the other
for the Drosha–DGCR8 complex [23,24]. Once produced, the pre- strand is degraded. The complex contains an Ago protein, GW182 and mature miRNA
which is required for gene silencing, which act by specifically binding to the 3 -UTR
miRNA is translocated to the cytoplasm through the nuclear pore
regions of target mRNAs, causing translational repression or mRNA degradation.
complex by Exportin-5. In the cytoplasm, the pre-miRNA hairpin
associates with the RNase III-like enzyme Dicer that cleaves it into
a double-stranded miRNA duplex comprised of the mature miRNA
and the miRNA* (also known as passenger strand) [25,26]. The localization of p68 [32]. The nuclear-cytoplasmic exportation can
guide strand functions as a mature miRNA and is incorporated into be regulated by steroids like estrogen and progesterone which
an RNA-induced silencing complex (RISC). This complex contains increase Exportin-5 mRNA expression [31]. The tumor suppres-
an Argonaute (Ago) protein as a primary component that binds to sor, BRCA1 (breast cancer susceptibility gene 1), associates with
the target mRNA and degrades the passenger strand. Mature miRNA Microprocessor complex to accelerate processing of pri-miRNAs
guides RISC to recognize target sequences located in the 3 UTR of specifically associated with cancer [33]. Other proteins such as
mRNAs leading to the inhibition of translation or degradation of Esr1 (also known as estrogen receptor alpha) [34], NF90 and NF45
mRNA [27]. MiRNA biogenesis can be regulated at each step by (nuclear factor 90 and 45) [35] associate with Drosha and inhibit
various mechanisms. At the transcriptional level, a number of fac- also the processing of specific pri-miRNAs such as let-7 family
tors like c-myc, or HMGA1 have been showed to regulate miRNAs members.
transcription, and have been reported in databases such as Trans- Proteins regulating pri-miRNA processing by recognition of the
mir [28]. Further steps, like the nuclear processing of miRNAs by stem-loop sequence or structure like hnRNP-A1 (heterogeneous
Drosha can be also modulated by the transforming Growth Factor nuclear ribonucleoprotein A1), KSRP (KH-type splicing regulatory
␤ (TGF␤) signaling pathway, the Smads [29]. The c-Myc oncogenic protein) have been also been described. Lin28 (abnormal cell lin-
transcription factor also transactivates drosha mRNA expression eage factor 28) is an interesting protein involved in regulating
thus upregulating the Drosha protein level in addition to the tran- multiple aspects of miRNA biogenesis. This protein can inhibit pri-
scriptional regulatation of miRNAs [30]. Tumor suppressor p53 let-7 processing [36,37] by sequestering pri-let-7 miRNAs in the
promotes some pri-miRNA processing via interaction with p68 [31]. nucleoli away from the Microprocessor. Interestingly, hnRNP-A1
Furthermore, it has been shown that the p38 MAPK-MK2 signaling protein can also interact with pri-let-7a, but in this case, it nega-
pathway promotes miRNA biogenesis by facilitating the nuclear tively regulates the processing [38].
B. Siddeek et al. / Mutation Research 764–765 (2014) 46–57 49

The terminal loop region is a determinant of microRNA biogene- Concerning AhR, its activation in TH17 cells regulates miR-132/212
sis. This region is crucial for both positive (for example, hnRNP A1) cluster and induces the cells differentiation [51]. Among nuclear
and negative (for example, Lin28) regulators to modulate miRNA receptors that are regulated by miRNAs, Esr1 was reported as a
levels and thereby gene regulation, reviewed in [39]. Finally, miRNA target of miR-206 [52], miR-221, miR-222 [53], and miR-22 [54].
stability and degradation are dependent on different mechanisms Conversely, miRNAs can also be regulated by nuclear receptors
that give to the majority of mature miRNAs, a half-life in the order of at the transcriptional level and during their maturation [55].
several hours to several days [40,41]. This suggests that a cis-acting Some authors pointed out the potential role of these miRNAs in
element in the sequence of mature miRNA regulates specificity anti-estrogen therapies. In the mouse testis, miR-184 was shown
of degradation of miRNAs. Studies in several model systems have to target nuclear receptor corepressor 2 (Ncor2) and by this way,
identified the molecular mechanisms of miRNA-regulated decay. regulates mammalian spermatogenesis [56].
The animal miRNAs generally lack a protective 2 -O methyl group Considering the wide actions of miRNAs on the expression
at their 3 terminus and display template-independent nucleotide of genes in key processes involved in detoxification, chemical
addition, mostly adenylation or uridylation that may regulate metabolic activation, and in the activity nuclear receptor, miRNAs
miRNA stability [42]. studies open new insights in the mode of action of drugs, hormones
The XRN (5 –3 exoribonuclease) family of enzymes and or endocrine disrupting chemicals.
hPNPaseold-35 (human polynucleotide phosphorylase protein) In this way of elucidating miRNAs function, the understand-
play various roles in miRNA stability. ing of cell-type specificity of miRNA expression is an important
In the context of the present brief review aiming at identifying step. However, very few miRNAs are exclusively found in indi-
the role of miRNA expression in toxicology, it will be of major inter- vidual tissues or cells. The miRNA expression varies from highly
est to in the future to identify how drugs/chemicals may affect this specific to ubiquitous and, for conserved miRNAs, is comparable
miRNA biosynthesis and degradation pathways. between rodents and human [57]. One example of tissue specificity
was found in the lung where miR-195 and miR-200c were specif-
ically expressed in the rat lung [58]. In few research areas such as
3. MiRNA in toxicology (Table 1) hepato-toxicity or cardiology, specific miRNAs have been identified
as markers of specific tissue injury. Indeed, miR-208 is described as
Recently, the identification of miRNAs targets highlighted the a myomiR since it is found at high levels in cardiac tissue. MiR-208
roles of miRNA in the posttranscriptional regulation of cytochrome expression is deregulated in various cardiovascular diseases. More-
P450 (CYP) and nuclear receptors and considered their poten- over, its expression in the cardiac tissue is described as a sensitive
tial relevance and application for toxicological studies. P450s are biomarker for cardiac injury in human [59], and inhibition studies
important enzymes that catalyze the metabolism of xenobiotics concluded that it might be an interesting therapeutic target [60].
including drugs, environmental chemicals, and carcinogens. The More often, this is a miRNAs expression profile that is linked to a
transcriptional regulation of CYPs by nuclear receptors has been specific pathology. For instance, Shapiro et al. have recently exam-
well studied and the central role of miRNAs in the regulation of ined the role of miRNAs in renal Ischemia reperfusion injury using
CYPs and nuclear receptors has been evidenced [43]. For example, expression profiling. They found that samples that underwent renal
CYP1A1/1A2 is regulated by miR-142-3p and miR-200a, CYP2C19 injury can be distinguished from controls based on the alterations
by miR-34a, CYP2D6 by let-7b and CYP2E1 by miR-10a and let- in nine miRNAs [61].
7g [44]. In addition, miRNAs can modulate the action of nuclear Interestingly, assessment of Doxorubicin-induced cardiomy-
receptors. Nuclear receptors are ligand-activated transcription fac- opathy versus anthracyclins in rats indicated that miRNAs
tors that regulate the expression of target genes by binding to expression profile could give indications about the specific tissue
their promoters. The expression of cytochrome P450 is regulated response in a dose and a time dependant manner [62]. In this tissue,
by receptors such as pregnane X receptor (PXR), the aryl hydro- they represent early toxicity indicators, since their modifications
carbon receptor (AhR), and the constitutive androstane receptor are detected earlier than changes in gene expression. Thus, miRNAs
(CAR). Those receptors are targeted by a number of miRNA and can help to better understand chronic/acute toxicity mechanism,
represent a way of P450 regulation. Indeed, mir-148a, which is since they may be regulated earlier than genomic toxicological
abundantly expressed in the liver, targets the human PXR [45]. markers. Similarly, MCF7 breast cancer cells showed a dose- and
It was reported that miR-375 with complex regulatory effects on time-dependent modification in the miRNA expression levels after
AhR mRNA could mediate in part the action of pollutants such a treatment with the chemotherapeutic drug 5-flourouracil. Eleven
as ambient particulate matter and diesel exhaust particles in the miRNAs (let-7g, miR-10b, miR-15a, miR-16, miR-21, miR-27a, miR-
induction of asthma [45]. Also, the activation of AhR by Tranilast, 365, miR-374b, miR-483-5p, miR-574-3p and miR-575) previously
an Anti-allergy Drug was shown to facilitate cell reprogramming identified in the microarray to be differentially expressed after
in a miR-302-dependent way [46]. Shizu et al. suggested that treatment were selected to analyze their responsiveness to dif-
phenobarbital-mediated down-regulation of miR-122 is an early ferent doses of 5-FU. Time-response data was also generated for
and important event in the AMPK-dependent CAR activation and miR-10b, miR-21, miR-483-5p, miR-574-3p and miR-575 follow-
transactivation of its target genes [47]. Thus, microRNAs appear ing 12, 24, 36, 48, 60 and 72 h treatment. It was concluded that
as key players in the regulation of genes involved in absorption, miRNAs play an important regulatory role in 5-FU induced cyto-
distribution, metabolism, and excretion. toxicity and fit in perfectly in the intricate network of 5-FU activity
Peroxisome-proliferator-activated receptors (PPARs) are [63].
ligand-activated nuclear receptors that exert in the liver a tran- Furthermore, Koufaris et al. [64] examined the microRNA profile
scriptional activity. PPAR␣ was reported to be specifically regulated in the liver of rats exposed to genotoxic (2-acetylaminofluorene)
by miR-21 and miR-27b, in the liver [48]. Additionally, miR-10b and epigenetic (phenobarbital, diethylhexylphthalate, methapyri-
was shown to target PPAR␣ and induce steatosis in hepatocytes lene HCL, monuron, and chlorendic acid) chemical hepatocar-
[49]. Indirectly, PPAR␥ activity appears to be under the control cinogens, as well as to non-hepatocarcinogenic treatments (ben-
of miR-132. Inversely, miRNA expression profiling indicated that zophenone, and diethylthiourea). All hepatocarcinogens affects the
activated PPAR␣ is a major regulator of hepatic miRNA expres- expression of liver mRNAs, while the hepatic microRNA profiles
sion and revealed that let-7C signaling cascade is critical for are associated with the mode of action of the chemical treatments
PPAR␣ agonist-induced liver proliferation and tumorigenesis [50]. and correspond to chemical carcinogenicity. The three nuclear
50 B. Siddeek et al. / Mutation Research 764–765 (2014) 46–57

receptor-activating chemicals (phenobarbital, benzophenone, and antigens specific of their parental cells [76,77]. Microparticles dis-
diethylhexylphthalate) are characterized by the induction of the play a broad spectrum of bioactive substances and receptors on
miR-200a/200b/429. In humans, a miRNAs-Environmental associ- their surface and harbor a concentrated set of cytokines, signaling
ation study conducted by Xudong Wu identified 1842 concurrent proteins, mRNAs, and microRNAs. During apoptosis, cells release
interactions between 407 miRNAs and 497 environmental chem- larger microparticles or apoptotic bodies that also transport spe-
icals, and thus highlighted distinct miRNAs regulated genes in cific miRNAs. Lipoproteins, particularly high-density lipoprotein
response to different environmental chemicals [65]. Since differ- can also transport endogenous miRNAs and deliver them to recipi-
ent drugs induce various miRNAs profiles, fitting with their cellular ent cells with possible functional targeting capabilities. This cellular
mode of action, the analysis of the miRNAs profile could potentially export is regulated by neutral sphingomyelinase and is dependent
help to categorize a potentially toxic compound. on scavenger receptor class B type I [71]. Most of the extracel-
MiRNAs are not only involved in the effects induced by an acute lular miRNAs in blood plasma and cell culture are independent
exposure, but also in chronic toxicity. In the rat testis, we recently of exosomes and are bound to Argonaute 2 protein (Ago2), a
showed that a neonatal exposure to estradiol benzoate induces part of RNA-induced silencing complex [78]. Ago2 is the main
germ cells death in adulthood [66]. This phenomenon is linked functional component of cytoplasmic miRNA ribonucleoprotein
to increased levels of mir-29 which targets DNA methyl trans- complex (miRNP). Extracellular miRNAs are in the most part by-
ferases and the anti-apoptotic protein MCL-1. The inhibition of products of dead cells that remain in extracellular space due to the
these proteins leads to the over expression of retrotransposons, high stability of the Ago2 protein and Ago2-miRNA complex. It was
to a stimulation of apoptosis and in term to the germ cell death also found that extracellular circulating miRNAs could be bound to
[66]. However, the mechanisms of imprinting and the existence other Ago proteins like Ago1.
of sensitive windows of exposure leading to irreversible miRNAs These observations raised the question as to whether circulating
changes remain poorly understood and represent an interesting miRNAs may play a role, at least as potential biomarkers, in health
area for further studies. and disease. Indeed, circulating miRNAs are abundant in blood
Besides the assessment of drugs on miRNAs expression at the and their levels in serum are stable, reproducible, and consistent
individual level, a few studies showed that such alteration could among individuals of the same species in contrast to mRNA [79,80],
also be observed in the next generations. For instance, the offspring which allow them to be potential non-invasive biomarkers. Several
of pregnant rats submitted to high fat diet presents, at the adult extracellular miRNAs have now revealed specific miRNA profiles
age, alterations in hepatic expression of insulin like growth factor-2 in health and disease allowing them to represent a consistent
and key miRNAs: miR-709, miR-122, miR-192, miR-194, miR-26a, signature in health and some differentiating diseases [74,81,82].
let-7a, let7b, let-7c, miR-494 and miR-483* [67]. Another exam- For example, several tumor-specific miRNA aberrations in serum
ple is represented by the exposure to cigarette smoke. Two of the were used as circulating biomarkers for distinguishing cancer and
constituents of cigarette smoke, benzo[a]pyrene and nicotine, have cancer-free diseases (Table 2). Increasing number of reports indi-
been shown to induce harmful phenotypes that can be transmitted cate that the serum miRNA expression profile can be used as a novel
to future generations. Indeed, in the multigenerational toxicity of serum-based biomarkers potentially offering more sensitive and
cigarette smoke, miRNAs expression alterations (mir-340, mir-365, specific tests than those currently available for early diagnosis of
mir-129-3p) in the spermatozoa from human smokers and that the cancer and other diseases. Those new approaches could improve
altered microRNAs may be at play in pathways for maintenance present clinical management, including the classification of can-
of healthy sperm and normal embryo development by control- cer, the prognosis estimation and the prediction of the therapeutic
ling cell death/apoptosis [68]. Those studies address the potential efficiency [69,79,83]. A number of circulating miRNAs were used as
transgenerational effects of exposure to chemicals and additional potential biomarkers of metabolic disorders, autoimmune, inflam-
experiments should be realized to validate those effects. Indeed, matory diseases (Table 3).
this field is still a matter of debate and remains under discussion. Interestingly, the question related to miRNA as biomarkers in
health and diseases has been now extended to miRNAs as biomark-
ers in toxicology studies (Table 4). This question related to the
4. miRNA as circulating biomarkers role of miRNA expression and epigenetic in chemical safety was
the topic of at least three recent held workshops by ILSI-HESI, US
The majority of miRNAs are intra-cellular and, as such, miRNAs National Academy of Sciences and ECETOC [84–88]. Indeed, that
are the critical mediators in the stress cellular response, disease and the levels of specific circulating miRNA species may also be used to
environmental stimuli [69]. Less is known, though, about extracel- detect and monitor the pathological development associated with
lular action of miRNAs. Stable circulating miRNAs were described chemicals or drug-induced tissue injuries is now suggested by sev-
for the first time in 2008, from the plasma of patients with lym- eral investigators [43,89–92]. Using a mouse model, the level a set
phoma [70]. To date, a significant number of miRNAs have been of plasma miRNAs has been associated with hepato-cellular injuries
observed outside of cells, in various body fluids. Number of miRNA induced by acetaminophen overdose. More specifically mir-122
carriers have been also described such as ribonucleoprotein com- and mir-192, are both enriched in the liver tissue and exhibit dose-
plexes, exosomes, apoptosis bodies, membrane-derived vesicles and exposure duration-dependent changes in the plasma that are
and high-density lipoproteins (Fig. 2) [71]. Exosomes are mem- parallel to the serum amino-transferase levels and the histopathol-
brane vesicles of 40–100 nm in diameter corresponding to the ogy of liver degeneration [91]. MiR-103 was also reported as an
internal vesicle of an endosomal compartment. It is initially formed appropriate biomarker among the circulating miRNAs identified
by the inward budding plasma membrane into multi-vesicular bod- in rats with acetaminophen-induced hepato-toxicity [43]. In the
ies within endosomes. Exosomes and their miRNA content are same way, a study in human and mouse model suggested that cir-
released into the extracellular compartment on the fusion of endo- culating miR-122 can be used as a potential novel predictive and
somes with the plasma membrane [72–74]. Exosomes are released reliable blood marker for viral-, alcohol-, and chemical-induced
when endosomally-derived multi-vesicular bodies fuse with the liver injury [90]. Environmental factors in daily life such as cigarette
plasma membrane [75]. Apoptotic bodies and microparticles are smoking can also affect plasma miRNA profiles. Altered miRNA
larger vesicles than exosomes (100–1000 nm in diameter). They expression has been reported in the smoking-related diseases [93]
represent a heterogeneous population of vesicles that are released and a study performed on smokers and non-smokers revealed that
by budding and blobbing of the plasma membrane and express plasma miRNA profiles clearly discriminate between smokers and
Table 2
Circulating miRNAs associated to cancers.

Disease miRNA Correlation Ref Disease miRNA Correlation Ref

Colorectal cancer miR-92 Differentiation of CRC from gastric [152] Gastric cancer mir-125b, Discrimination of gastric cancer from [153]
(CRC) cancer, inflammatory bowel disease, mir-199a,mir-100, healthy controls
and healthy controls let-7 g, mir-433,
mir-214
mir-21, mir-133b Discrimination of prostate cancer from [154] mir-10b, mir-21, [155]
healthy controls mir-223, mir-338,
let-7a, mir-30a-5p,
mir-126
mir-133 [156] Pancreatic cancer mir-452, mir-105, Discrimination of pancreatic cancer [157]
mir-127, mir-518-2, from healthy controls

B. Siddeek et al. / Mutation Research 764–765 (2014) 46–57


mir-187, mir-30a-3p,
miR-196a-2
mir-221 [158] mir-18a [159]
mir-141 [160] Glioblastoma mir-21 Discrimination of glioblastoma from [161]
healthy controls

Prostate cancer miR-141 Discrimination of prostate cancer from [162] mir-128 [67]
healthy controls
mir-96 [163] Acute leukemia miR-92a decreased Differentiation between leukemia and [164]
(ratio healthy controls
miR-638/miR-92a
decreased)
mir-375, mir-141 [154] CRC and advanced miR-29a and miR-92a Discrimination of CRC and also [165]
adenome increased advanced adenoma from healthy
controls
mir-15, mir-195, [166] Diffuse large B-cell miR-155, miR-210, Differentiation of DLBCL from healthy [70]
mir-26a, let-7i lymphoma (DLBCL) miR-21 increased controls and association of miR-21
with relapse-free survival in DLBCL
Set of 15 miRNAs [167] Non-small cell lung miR- 128b, miR-152, Differentiation of NSCLC from healthy [79]
(including for cancer (NSCLC) miR-125b, miR-205, controls
instance miR-16) miR-27a, miR- 146a,
miR-222, miR-23a,
miR-24, miR-150

Breast cancer Mir-195, let7a Discrimination of breast cancer from [168] Lung cancer mir-155, let-7a Discrimination of lung cancer from [169]
healthy controls; correlation between healthy controls
miRNA levels and lymph node status
mir-145 Discrimination of breast cancer from [170] let-7a, mir-221, [171]
healthy controls mir-137, mir-372,
mir-182
mir-10b, mir-34a, [172] mir-221 and mir-222 [173]
mir-155
92a, 21 [174]

51
52 B. Siddeek et al. / Mutation Research 764–765 (2014) 46–57

Fig. 2. Mechanisms of miRNA release from cells into peripheral blood circulation and transport to target cells.
MP: microparticles; HDL: high-density lipoprotein; Ago2: Argonaute 2

Table 3
Circulating miRNAs associated to metabolic disorders, auto-immune and inflammatory diseases.

Physiopathological status miRNAs Correlation References

Pregnancy mir-527, miR-520d-5p and miR-526a Discrimination of pregnant women from non-pregnant women [175]
Non-small-cell lung carcinoma let-7f, miR-20b and miR-30e-3p Discrimination between NSCLC and healthy control [176]
(NSCLC)
Rheumatoid arthritis miR-132 decreased Distinction between RA or OA [177]
(RA)/osteoarthritis (OA)
mir-16 and miR-146a Correlation with tender joint counts and 28-joint Disease
Activity Score

Coronary artery disease (CAD) miR-126, miR-17, miR-92a, miR-155, miR-145 Distinction between CAD and healthy controls [178]
miR-133a and miR-208a

Chron disease miR-149 [179]


Diabetes miR-144 Increased [180]
Alzheimer’s disease miR-137, 181c, 9, 29a/b Reduced [181]
Duchenne muscular dystrophy miR-1, 133a, 206 Increased [182]

non-smokers. This observation suggested that repeated cigarette miRNA-222 and miRNA-21 was significantly upregulated in post-
smoking substantially alters the plasma miRNA profile [94]. Expo- exposure samples [95].
sure to metal-rich particulate matter has been reported to modify Circulating miRNAs may also represent good markers for
the expression of candidate miRNAs in peripheral blood leukocytes hypertension-induced heart failure and for the response to thera-
of workers at an electric-furnace steel plant. Indeed, expression of peutic treatment. Indeed, in a rat model, miRNAs arrays on plasma

Table 4
MiRNA expression in seminal plasma from infertile men.

Diseases miRNAs Correlation References

Oligoastheno-zoospermia miR-141, miR-29a, miR-429, Upregulated in spermatozoa samples from oligoasthenozoospermic [183]
miR200a patients compared with those from normozoospermic
miR- 34b, miR-19a, miR-16, Downregulated in supernatant sperm samples from
miR-122 oligoasthenozoospermic patients compared with those from
normozoospermic

Astheno-zoospermia miR-30a, miR-26a, miR-200a, Upregulated in supernatant sperm samples from asthenozoospermic
miR-141, miR-29a, miR-24 patients compared with those from normozoospermic
miR-1973, miR-34b, miR-122 Downregulated in supernatant sperm samples from
asthenozoospermic patients compared with those from
normozoospermic

Infertility miR-574-5p, miR-297, Upregulated in the semen of infertile males with semen abnormalities [184]
miR-122, miR-1275, miR-373,
miR-185 and miR-193b
miR-100, miR-512-3p, miR-16, Downregulated in the semen of infertile males with semen
miR-19b, miR-23b and abnormalities
miR-26a

Non obstructive miR-19b and let-7a Upregulated in supernatant sperm from idiopathic infertile males with [185]
azoospermia non obstructive azoospermia compared with fertile controls
B. Siddeek et al. / Mutation Research 764–765 (2014) 46–57 53

highlighted modifications in miR-16, miR-20b, miR-93, miR-106b, interesting biomarkers, the possibility that miRNAs measurement
miR-223, and miR-423-5p. A time course study showed that those could be included in toxicological studies is open. While the field of
changes are correlated to disease progression [96]. epigenetic (including miRNA but also methylation and chromatin
In kidney diseases, a number of studies have highlighted the sen- remodeling) is a rapidly growing field in both understanding of
sitivity of miRNAs detection in plasma and urine (for review [97]). these mechanisms in health and diseases as well as in terms of
In injury induced by renal ischemia reperfusion or streptozotocin availability of tools, some key questions remain still to be clarified:
induced diabetes in mice, miR-10a and miR-30d concentrations (i) are miRNA expression changes causes or consequences of the
in plasma and urine are positively correlated with the degree of adverse effects? (ii) Are they stable and irreversible? (86).
kidney injury [98]. In humans, urinary and plasma miR-21 are asso-
ciated with severe acute kidney injury [99].
Together, there are now many studies establishing a potential Conflict of interest
link between the level of circulating miRNA and clinical diagnosis
or prognosis in many pathologies or injuries. The use of circulating None.
miRNAs in body fluids as potential toxicological biomarkers, and
the link between miRNA-related pharmaco-genomics and adverse
References
drug reactions will be a matter of future investigations [43].
[1] V.N. Kim, J. Han, M.C. Siomi, Biogenesis of small RNAs in animals, Nat. Rev.
Mol. Cell Biol. 10 (2009) 126–139.
5. Conclusions [2] M. Ghildiyal, P.D. Zamore, Small silencing RNAs: an expanding universe, Nat.
Rev. Genet. 10 (2009) 94–108.
[3] C.D. Malone, G.J. Hannon, Small RNAs as guardians of the genome, Cell 136
MiRNAs are highly conserved between human and animal mod- (2009) 656–668.
els and can regulate biological pathways by repressing target [4] J.C. Mathers, G. Strathdee, C.L. Relton, Induction of epigenetic alterations by
dietary and other environmental factors, Adv. Genet. 71 (2010) 3–39.
proteins. Chemicals can affect those pathways by inducing their [5] B.P. Lewis, C.B. Burge, D.P. Bartel, Conserved seed pairing, often flanked by
down or up-regulation. Also, miRNA profiling in response to toxic adenosines, indicates that thousands of human genes are microRNA targets,
compounds can provide toxicant-specific profiles in specific organs. Cell 120 (2005) 15–20.
[6] M. Szyf, The dynamic epigenome and its implications in toxicology, Toxicol.
Given their specificity to tested compounds, the time and dose Sci. 100 (2007) 7–23.
dependency of their modifications, the sensitivity and simplicity [7] Y. Saito, P.A. Jones, Epigenetic activation of tumor suppressor microRNAs in
of their detection in various tissues, miRNAs appear like promis- human cancer cells, Cell Cycle 5 (2006) 2220–2222.
[8] L. Meunier, B. Siddeek, A. Vega, N. Lakhdari, L. Inoubli, R.P. Bellon, G. Lemaire,
ing tools in toxicological studies. Interestingly, circulating miRNAs
C. Mauduit, M. Benahmed, Perinatal programming of adult rat germ cell death
could be accessible through non-invasive protocols and stable. Cir- after exposure to xenoestrogens: role of microRNA miR-29 family in the
culating miRNAs thus represent ideal candidates in toxicological down-regulation of DNA methyltransferases and Mcl-1, Endocrinology 153
studies, and may be used as biomarkers of chemical exposure for (2012) 1936–1947.
[9] M.J. LeBaron, R.J. Rasoulpour, J. Klapacz, R.G. Ellis-Hutchings, H.M. Hollnagel,
safety assessment. B.B. Gollapudi, Epigenetics and chemical safety assessment, Mutat. Res. 705
However, with regard to the involvement of miRNA expres- (2010) 83–95.
sion in the effects of exposure to chemicals, there are at least two [10] M.R. Fabian, N. Sonenberg, The mechanics of miRNA-mediated gene silencing:
a look under the hood of miRISC, Nat. Struct. Mol. Biol. 19 (2012) 586–593.
main observations that should be taken into account. Firstly, while [11] S.D. Fiedler, M.Z. Carletti, L.K. Christenson, Quantitative RT-PCR methods for
there are a growing number of reports showing altered miRNAs mature microRNA expression analysis, Methods Mol. Biol. 630 (2010) 49–64.
profile following exposure to drugs/chemicals, the mechanisms [12] E. Varallyay, J. Burgyan, Z. Havelda, MicroRNA detection by northern blotting
using locked nucleic acid probes, Nat. Protoc. 3 (2008) 190–196.
leading to the pathological phenotype remain poorly understood. [13] S. Ambs, R.L. Prueitt, M. Yi, R.S. Hudson, T.M. Howe, F. Petrocca, T.A. Wallace,
One of the major limitations of numerous studies analyzing the C.G. Liu, S. Volinia, G.A. Calin, H.G. Yfantis, R.M. Stephens, C.M. Croce, Genomic
miRNAs expression changes in experimental models after exposure profiling of microRNA and messenger RNA reveals deregulated microRNA
expression in prostate cancer, Cancer Res. 68 (2008) 6162–6170.
to chemicals is that these changes, generally, are viewed associ- [14] Z. Williams, I.Z. Ben-Dov, R. Elias, A. Mihailovic, M. Brown, Z. Rosenwaks,
ated rather than causal to the adverse apical endpoints. One of the T. Tuschl, Comprehensive profiling of circulating microRNA via small RNA
appropriate approaches to establish a causal link between miRNA sequencing of cDNA libraries reveals biomarker potential and limitations,
Proc. Natl. Acad. Sci. U.S.A. 110 (2013) 4255–4260.
expression and the adverse effects could be the following; iden-
[15] A.M. Monteys, R.M. Spengler, J. Wan, L. Tecedor, K.A. Lennox, Y. Xing, B.L.
tify first the adverse effect and then try to delineate and dissect Davidson, Structure and activity of putative intronic miRNA promoters, RNA
the supporting miRNA expression changes. Recently, by using such 16 (2013) 495–505.
an approach, alterations in mir-29 family members that induced [16] Y. Lee, M. Kim, J. Han, K.H. Yeom, S. Lee, S.H. Baek, V.N. Kim, MicroRNA genes
are transcribed by RNA polymerase II, EMBO J. 23 (2004) 4051–4060.
adult germ cell death process leading to infertility were identified [17] G.M. Borchert, W. Lanier, B.L. Davidson, RNA polymerase III transcribes human
following neonatal exposure to the xenoestrogen estradiol ben- microRNAs, Nat. Struct. Mol. Biol. 13 (2006) 1097–1101.
zoate [66]. Secondly, a number of studies have highlighted the dose [18] Y.S. Lee, A. Dutta, MicroRNAs in cancer, Annu. Rev. Pathol. 4 (2009) 199–227.
[19] F. Ozsolak, L.L. Poling, Z. Wang, H. Liu, X.S. Liu, R.G. Roeder, X. Zhang, J.S. Song,
and time dependency of miRNAs profile after exposure to chemi- D.E. Fisher, Chromatin structure analyses identify miRNA promoters, Genes
cal compounds. Furthermore, studies have showed that exposure Dev. 22 (2008) 3172–3183.
can lead to long term changes, and possibly irreversible. The irre- [20] V. Libri, P. Miesen, R.P. van Rij, A.H. Buck, Regulation of microRNA biogene-
sis and turnover by animals and their viruses, Cell. Mol. Life Sci. 70 (2013)
versibility of miRNA expression changes is likely occurring when 3525–3544.
the exposure takes place during critical developmental periods i.e. [21] Y. Lee, C. Ahn, J. Han, H. Choi, J. Kim, J. Yim, J. Lee, P. Provost, O. Radmark, S.
the in utero and perinatal periods that are specifically sensitive to Kim, V.N. Kim, The nuclear RNase III Drosha initiates microRNA processing,
Nature 425 (2003) 415–419.
chemicals. Those observations are to be viewed in the context of [22] R.I. Gregory, K.P. Yan, G. Amuthan, T. Chendrimada, B. Doratotaj, N. Cooch, R.
the concept of Developmental Origin of Health and Diseases (also Shiekhattar, The Microprocessor complex mediates the genesis of microRNAs,
termed the Barker’s hypothesis). In this context, low-dose effects Nature 432 (2004) 235–240.
[23] R. Triboulet, H.M. Chang, R.J. Lapierre, R.I. Gregory, Post-transcriptional con-
have received considerable attention from the scientific and regula-
trol of DGCR8 expression by the microprocessor, RNA 15 (2009) 1005–1011.
tory communities. Analysis of miRNAs seems to be an appropriate [24] S. Kadener, J. Rodriguez, K.C. Abruzzi, Y.L. Khodor, K. Sugino, M.T. Marr 2nd,
approach in risk assessment at low doses, and long term effects S. Nelson, M. Rosbash, Genome-wide identification of targets of the drosha-
induced by EDCs. Based on the increasing amounts of accumu- pasha/DGCR8 complex, RNA 15 (2009) 537–545.
[25] E. Bernstein, A.A. Caudy, S.M. Hammond, G.J. Hannon, Role for a bidentate
lating data on miRNA expression alterations following exposure ribonuclease in the initiation step of RNA interference, Nature 409 (2001)
to chemicals and the fact that circulating miRNAs appear as very 363–366.
54 B. Siddeek et al. / Mutation Research 764–765 (2014) 46–57

[26] G. Hutvagner, J. McLachlan, A.E. Pasquinelli, E. Balint, T. Tuschl, P.D. Zamore, downregulated in estrogen receptor alpha-positive human breast cancer cell
A cellular function for the RNA-interference enzyme Dicer in the maturation lines and clinical samples, FEBS J. 277 (2010) 1684–1694.
of the let-7 small temporal RNA, Science 293 (2001) 834–838. [55] Z. Yang, L. Wang, Regulation of microRNA expression and function by nuclear
[27] D.P. Bartel, MicroRNAs: target recognition and regulatory functions, Cell 136 receptor signaling, Cell Biosci. 1 (2011) 31.
(2009) 215–233. [56] J. Wu, J. Bao, L. Wang, Y. Hu, C. Xu, MicroRNA-184 downregulates nuclear
[28] J. Wang, M. Lu, C. Qiu, Q. Cui, TransmiR: a transcription factor-microRNA receptor corepressor 2 in mouse spermatogenesis, BMC Dev. Biol. 11 (2011)
regulation database, Nucleic Acids Res. 38 (2010) D119–D122. 64.
[29] B.N. Davis-Dusenbery, A. Hata, Smad-mediated miRNA processing: a critical [57] P. Landgraf, M. Rusu, R. Sheridan, A. Sewer, N. Iovino, A. Aravin, S. Pfef-
role for a conserved RNA sequence, RNA Biol. 8 (2011) 71–76. fer, A. Rice, A.O. Kamphorst, M. Landthaler, C. Lin, N.D. Socci, L. Hermida,
[30] X. Wang, X. Zhao, P. Gao, M. Wu, c-Myc modulates microRNA processing via V. Fulci, S. Chiaretti, R. Foa, J. Schliwka, U. Fuchs, A. Novosel, R.U. Muller,
the transcriptional regulation of Drosha, Sci. Rep. 3 (2013) 1942. B. Schermer, U. Bissels, J. Inman, Q. Phan, M. Chien, D.B. Weir, R. Choksi,
[31] H.I. Suzuki, K. Yamagata, K. Sugimoto, T. Iwamoto, S. Kato, K. Miyazono, Mod- G. De Vita, D. Frezzetti, H.I. Trompeter, V. Hornung, G. Teng, G. Hartmann,
ulation of microRNA processing by p53, Nature 460 (2009) 529–533. M. Palkovits, R. Di Lauro, P. Wernet, G. Macino, C.E. Rogler, J.W. Nagle, J. Ju,
[32] S. Hong, H. Noh, H. Chen, R. Padia, Z.K. Pan, S.B. Su, Q. Jing, H.F. Ding, S. Huang, F.N. Papavasiliou, T. Benzing, P. Lichter, W. Tam, M.J. Brownstein, A. Bosio, A.
Signaling by p38 MAPK stimulates nuclear localization of the microprocessor Borkhardt, J.J. Russo, C. Sander, M. Zavolan, T. Tuschl, A mammalian microRNA
component p68 for processing of selected primary microRNAs, Sci. Signal. 6 expression atlas based on small RNA library sequencing, Cell 129 (2007)
(2013) ra16. 1401–1414.
[33] S. Kawai, A. Amano, BRCA1 regulates microRNA biogenesis via the DROSHA [58] Y. Wang, T. Weng, D. Gou, Z. Chen, N.R. Chintagari, L. Liu, Identification of rat
microprocessor complex, J. Cell Biol. 197 (2012) 201–208. lung-specific microRNAs by micoRNA microarray: valuable discoveries for
[34] K. Yamagata, S. Fujiyama, S. Ito, T. Ueda, T. Murata, M. Naitou, K. Takeyama, the facilitation of lung research, BMC Genomics 8 (2007) 29.
Y. Minami, B.W. O’Malley, S. Kato, Maturation of microRNA is hormonally [59] M. Satoh, Y. Minami, Y. Takahashi, T. Tabuchi, M. Nakamura, Expression of
regulated by a nuclear receptor, Mol. Cell 36 (2009) 340–347. microRNA-208 is associated with adverse clinical outcomes in human dilated
[35] S. Sakamoto, K. Aoki, T. Higuchi, H. Todaka, K. Morisawa, N. Tamaki, E. Hatano, cardiomyopathy, J. Card. Fail. 16 (2010) 404–410.
A. Fukushima, T. Taniguchi, Y. Agata, The NF90-NF45 complex functions as a [60] C.E. Grueter, E. van Rooij, B.A. Johnson, S.M. DeLeon, L.B. Sutherland, X. Qi, L.
negative regulator in the microRNA processing pathway, Mol. Cell. Biol. 29 Gautron, J.K. Elmquist, R. Bassel-Duby, E.N. Olson, A cardiac microRNA gov-
(2009) 3754–3769. erns systemic energy homeostasis by regulation of MED13, Cell 149 (2012)
[36] M.A. Newman, J.M. Thomson, S.M. Hammond, Lin-28 interaction with the Let- 671–683.
7 precursor loop mediates regulated microRNA processing, RNA 14 (2008) [61] M.D. Shapiro, J. Bagley, J. Latz, J.G. Godwin, X. Ge, S.G. Tullius, J. Iacomini,
1539–1549. MicroRNA expression data reveals a signature of kidney damage following
[37] E. Piskounova, C. Polytarchou, J.E. Thornton, R.J. LaPierre, C. Pothoulakis, J.P. ischemia reperfusion injury, PLoS One 6 (2011) e23011.
Hagan, D. Iliopoulos, R.I. Gregory, Lin28A and Lin28B inhibit let-7 microRNA [62] C. Vacchi-Suzzi, Y. Bauer, B.R. Berridge, S. Bongiovanni, K. Gerrish, H.K.
biogenesis by distinct mechanisms, Cell 147 (2011) 1066–1079. Hamadeh, M. Letzkus, J. Lyon, J. Moggs, R.S. Paules, F. Pognan, F. Staedtler,
[38] G. Michlewski, J.F. Caceres, Antagonistic role of hnRNP A1 and KSRP in the M.P. Vidgeon-Hart, O. Grenet, P. Couttet, Perturbation of microRNAs in rat
regulation of let-7a biogenesis, Nat. Struct. Mol. Biol. 17 (2010) 1011–1018. heart during chronic doxorubicin treatment, PLoS One 7 (2012) e40395.
[39] N.R. Choudhury, G. Michlewski, Terminal loop-mediated control of microRNA [63] M.Y. Shah, X. Pan, L.N. Fix, M.A. Farwell, B. Zhang, 5-Fluorouracil drug alters
biogenesis, Biochem. Soc. Trans. 40 (2012) 789–793. the microRNA expression profiles in MCF-7 breast cancer cells, J. Cell. Physiol.
[40] S. Bail, M. Swerdel, H. Liu, X. Jiao, L.A. Goff, R.P. Hart, M. Kiledjian, Differential 226 (2011) 1868–1878.
regulation of microRNA stability, RNA 16 (2006) 1032–1039. [64] C. Koufaris, J. Wright, R.A. Currie, N.J. Gooderham, Hepatic microRNA pro-
[41] M.P. Gantier, C.E. McCoy, I. Rusinova, D. Saulep, D. Wang, D. Xu, A.T. Irving, M.A. files offer predictive and mechanistic insights after exposure to genotoxic
Behlke, P.J. Hertzog, F. Mackay, B.R. Williams, Analysis of microRNA turnover and epigenetic hepatocarcinogens, Toxicol. Sci. 128 (2012) 532–543.
in mammalian cells following Dicer1 ablation, Nucleic Acids Res. 39 (2011) [65] X. Wu, Y. Song, Preferential regulation of miRNA targets by environmental
5692–5703. chemicals in the human genome, BMC Genomics 12 (2011) 244.
[42] J.O. Westholm, E. Ladewig, K. Okamura, N. Robine, E.C. Lai, Common and dis- [66] L. Meunier, B. Siddeek, A. Vega, N. Lakhdari, L. Inoubli, R.P. Bellon, G. Lemaire, C.
tinct patterns of terminal modifications to mirtrons and canonical microRNAs, Mauduit, M. Benahmed, Perinatal programming of adult rat germ cell death
RNA 18 (2012) 177–192. after exposure to xenoestrogens: role of microRNA miR -29 family in the
[43] T. Yokoi, M. Nakajima, microRNAs as mediators of drug toxicity, Annu. Rev. down-regulation of DNA methyltransferases and Mcl-1, Endocrinology 153
Pharmacol. Toxicol. 53 (2013) 377–400. (2012) 1936–1947.
[44] J.K. Rieger, K. Klein, S. Winter, U.M. Zanger, Expression variability of absorp- [67] Y. Zhang, T. Chao, R. Li, W. Liu, Y. Chen, X. Yan, Y. Gong, B. Yin, B. Qiang, J.
tion, distribution, metabolism, excretion-related microRNAs in human liver: Zhao, J. Yuan, X. Peng, MicroRNA-128 inhibits glioma cells proliferation by
influence of nongenetic factors and association with gene expression, Drug targeting transcription factor E2F3a, J. Mol. Med. (Berl.) 87 (2009) 43–51.
Metab. Dispos. 41 (2013) 1752–1762. [68] E.L. Marczylo, A.A. Amoako, J.C. Konje, T.W. Gant, T.H. Marczylo, Smoking
[45] S. Takagi, M. Nakajima, T. Mohri, T. Yokoi, Post-transcriptional regulation induces differential miRNA expression in human spermatozoa: a potential
of human pregnane X receptor by micro-RNA affects the expression of transgenerational epigenetic concern? Epigenetics 7 (2012) 432–439.
cytochrome P450 3A4, J. Biol. Chem. 283 (2008) 9674–9680. [69] J.T. Mendell, E.N. Olson, MicroRNAs in stress signaling and human disease,
[46] W. Hu, J. Zhao, G. Pei, Activation of aryl hydrocarbon receptor (AhR) by Cell 148 (2012) 1172–1187.
Tranilast, an anti-allergy drug, promotes miR-302 expression and cell repro- [70] C.H. Lawrie, S. Gal, H.M. Dunlop, B. Pushkaran, A.P. Liggins, K. Pulford, A.H.
gramming, J. Biol. Chem. 288 (2013) 22972–22984. Banham, F. Pezzella, J. Boultwood, J.S. Wainscoat, C.S. Hatton, A.L. Harris,
[47] R. Shizu, S. Benoki, Y. Numakura, S. Kodama, M. Miyata, Y. Yamazoe, Detection of elevated levels of tumour-associated microRNAs in serum of
K. Yoshinari, Xenobiotic-induced hepatocyte proliferation associated with patients with diffuse large B-cell lymphoma, Br. J. Haematol. 141 (2008)
constitutive active/androstane receptor (CAR) or peroxisome proliferator- 672–675.
activated receptor alpha (PPARalpha) is enhanced by pregnane X receptor [71] K.C. Vickers, B.T. Palmisano, B.M. Shoucri, R.D. Shamburek, A.T. Remaley,
(PXR) activation in mice, PLoS One 8 (2013) e61802. MicroRNAs are transported in plasma and delivered to recipient cells by
[48] K. Kida, M. Nakajima, T. Mohri, Y. Oda, S. Takagi, T. Fukami, T. Yokoi, PPARalpha high-density lipoproteins, Nat. Cell. Biol. 13 (2011) 423–433.
is regulated by miR-21 and miR-27b in human liver, Pharm. Res. 28 (2011) [72] C. Thery, L. Zitvogel, S. Amigorena, Exosomes: composition, biogenesis and
2467–2476. function, Nat. Rev. Immunol. 2 (2002) 569–579.
[49] L. Zheng, G.C. Lv, J. Sheng, Y.D. Yang, Effect of miRNA-10b in regulating cellular [73] W. Stoorvogel, M.J. Kleijmeer, H.J. Geuze, G. Raposo, The biogenesis and func-
steatosis level by targeting PPAR-alpha expression, a novel mechanism for the tions of exosomes, Traffic 3 (2002) 321–330.
pathogenesis of NAFLD, J. Gastroenterol. Hepatol. 25 (2010) 156–163. [74] R.A. Boon, K.C. Vickers, Intercellular transport of microRNAs, Arterioscler.
[50] Y.M. Shah, K. Morimura, Q. Yang, T. Tanabe, M. Takagi, F.J. Gonzalez, Perox- Thromb. Vasc. Biol. 33 (2013) 186–192.
isome proliferator-activated receptor alpha regulates a microRNA-mediated [75] A. Allegra, A. Alonci, S. Campo, G. Penna, A. Petrungaro, D. Gerace, C. Musolino,
signaling cascade responsible for hepatocellular proliferation, Mol. Cell. Biol. Circulating microRNAs: new biomarkers in diagnosis, prognosis and treat-
27 (2007) 4238–4247. ment of cancer (review), Int. J. Oncol. 41 (2012) 1897–1912.
[51] T. Nakahama, H. Hanieh, N.T. Nguyen, I. Chinen, B. Ripley, D. Millrine, S. [76] L. Xu, B.F. Yang, J. Ai, MicroRNA transport: a new way in cell communication,
Lee, K.K. Nyati, P.K. Dubey, K. Chowdhury, Y. Kawahara, T. Kishimoto, Aryl J. Cell. Physiol. 228 (2013) 1713–1719.
hydrocarbon receptor-mediated induction of the microRNA-132/212 cluster [77] S.F. Mause, C. Weber, Microparticles: protagonists of a novel communica-
promotes interleukin-17-producing T-helper cell differentiation, Proc. Natl. tion network for intercellular information exchange, Circ. Res. 107 (2010)
Acad. Sci. U.S.A. 110 (2013) 11964–11969. 1047–1057.
[52] B.D. Adams, H. Furneaux, B.A. White, The micro-ribonucleic acid (miRNA) [78] A. Turchinovich, L. Weiz, A. Langheinz, B. Burwinkel, Characterization of extra-
miR-206 targets the human estrogen receptor-alpha (ERalpha) and represses cellular circulating microRNA, Nucleic Acids Res. 39 (2011) 7223–7233.
ERalpha messenger RNA and protein expression in breast cancer cell lines, [79] X. Chen, Y. Ba, L. Ma, X. Cai, Y. Yin, K. Wang, J. Guo, Y. Zhang, J. Chen, X. Guo,
Mol. Endocrinol. 21 (2007) 1132–1147. Q. Li, X. Li, W. Wang, J. Wang, X. Jiang, Y. Xiang, C. Xu, P. Zheng, J. Zhang, R. Li,
[53] J.J. Zhao, J. Lin, H. Yang, W. Kong, L. He, X. Ma, D. Coppola, J.Q. Cheng, MicroRNA- H. Zhang, X. Shang, T. Gong, G. Ning, K. Zen, C.Y. Zhang, Characterization of
221/222 negatively regulates estrogen receptor alpha and is associated with microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and
tamoxifen resistance in breast cancer, J. Biol. Chem. 283 (2008) 31079–31086. other diseases, Cell Res. 18 (2008) 997–1006.
[54] J. Xiong, D. Yu, N. Wei, H. Fu, T. Cai, Y. Huang, C. Wu, X. Zheng, Q. Du, [80] M.P. Hunter, N. Ismail, X. Zhang, B.D. Aguda, E.J. Lee, L. Yu, T. Xiao, J. Schafer,
D. Lin, Z. Liang, An estrogen receptor alpha suppressor, microRNA-22, is M.L. Lee, T.D. Schmittgen, S.P. Nana-Sinkam, D. Jarjoura, C.B. Marsh, Detection
B. Siddeek et al. / Mutation Research 764–765 (2014) 46–57 55

of microRNA expression in human peripheral blood microvesicles, PLoS One [107] A. Izzotti, G.A. Calin, P. Arrigo, V.E. Steele, C.M. Croce, S. De Flora, Downregula-
3 (2008) e3694. tion of microRNA expression in the lungs of rats exposed to cigarette smoke,
[81] N. Scholer, C. Langer, F. Kuchenbauer, Circulating microRNAs as biomarkers FASEB J. 23 (2009) 806–812.
– true Blood? Genome Med. 3 (2011) 72. [108] F. Schembri, S. Sridhar, C. Perdomo, A.M. Gustafson, X. Zhang, A. Ergun, J. Lu, G.
[82] A. Keller, P. Leidinger, A. Bauer, A. Elsharawy, J. Haas, C. Backes, A. Wendschlag, Liu, X. Zhang, J. Bowers, C. Vaziri, K. Ott, K. Sensinger, J.J. Collins, J.S. Brody, R.
N. Giese, C. Tjaden, K. Ott, J. Werner, T. Hackert, K. Ruprecht, H. Huwer, J. Hue- Getts, M.E. Lenburg, A. Spira, MicroRNAs as modulators of smoking-induced
bers, G. Jacobs, P. Rosenstiel, H. Dommisch, A. Schaefer, J. Muller-Quernheim, gene expression changes in human airway epithelium, Proc. Natl. Acad. Sci.
B. Wullich, B. Keck, N. Graf, J. Reichrath, B. Vogel, A. Nebel, S.U. Jager, P. U.S.A. 106 (2009) 2319–2324.
Staehler, I. Amarantos, V. Boisguerin, C. Staehler, M. Beier, M. Scheffler, M.W. [109] S. Kalscheuer, X. Zhang, Y. Zeng, P. Upadhyaya, Differential expression of
Buchler, J. Wischhusen, S.F. Haeusler, J. Dietl, S. Hofmann, H.P. Lenhof, S. microRNAs in early-stage neoplastic transformation in the lungs of F344 rats
Schreiber, H.A. Katus, W. Rottbauer, B. Meder, J.D. Hoheisel, A. Franke, E. chronically treated with the tobacco carcinogen 4-(methylnitrosamino)-1-
Meese, Toward the blood-borne miRNome of human diseases, Nat. Methods (3-pyridyl)-1-butanone, Carcinogenesis 29 (2008) 2394–2399.
8 (2011) 841–843. [110] B. Zhang, X. Pan, RDX induces aberrant expression of microRNAs in mouse
[83] J.A. Weber, D.H. Baxter, S. Zhang, D.Y. Huang, K.H. Huang, M.J. Lee, D.J. Galas, brain and liver, Environ. Health Perspect. 117 (2009) 231–240.
K. Wang, The microRNA spectrum in 12 body fluids, Clin. Chem. 56 (2010) [111] T. Fukushima, Y. Hamada, H. Yamada, I. Horii, Changes of micro-RNA expres-
1733–1741. sion in rat liver treated by acetaminophen or carbon tetrachloride–regulating
[84] E. Buckley, Epigenetic effects of chemicals not ready for regulatory “prime- role of micro-RNA for RNA expression, J. Toxicol. Sci. 32 (2007) 401–409.
time”, Pestic. Toxic Chem. News 37 (2009), http://www.agra-net.com/aius/ [112] E. Elyakim, E. Sitbon, A. Faerman, S. Tabak, E. Montia, L. Belanis, A. Dov, E.G.
home.jsp?pagetitle=aiusfp&pubId=ag100 Marcusson, C.F. Bennett, A. Chajut, D. Cohen, N. Yerushalmi, hsa-miR-191 is a
[85] J.I. Goodman, K.A. Augustine, M.L. Cunnningham, D. Dixon, Y.P. Dragan, J.G. candidate oncogene target for hepatocellular carcinoma therapy, Cancer Res.
Falls, R.J. Rasoulpour, R.C. Sills, R.D. Storer, D.C. Wolf, S.D. Pettit, What do we 70 (2010) 8077–8087.
need to know prior to thinking about incorporating an epigenetic evaluation [113] M. Avissar-Whiting, K.R. Veiga, K.M. Uhl, M.A. Maccani, L.A. Gagne, E.L. Moen,
into safety assessments? Toxicol. Sci. 116 (2010) 375–381. C.J. Marsit, A. Bisphenol, exposure leads to specific microRNA alterations in
[86] B. Hileman, Chemicals can turn genes on and off, Environ. Health News 3 placental cells, Reprod. Toxicol. 29 (2010) 401–406.
(August) (2009). [114] J.S. Lee, D. Semela, J. Iredale, V.H. Shah, Sinusoidal remodeling and angiogen-
[87] ECETOC WR 23 - Epigenetics and Chemical Safety Rome, 2011. esis: a new function for the liver-specific pericyte? Hepatology (Baltimore,
[88] C.C. Priestley, M. Anderton, A.T. Doherty, P. Duffy, H.R. Mellor, H. Powell, R. Md.) 45 (2007) 817–825.
Roberts, Epigenetics – relevance to drug safety science, Toxicol. Res. 1 (2012) [115] P. Sathyan, H.B. Golden, R.C. Miranda, Competing interactions between micro-
23–31. RNAs determine neural progenitor survival and proliferation after ethanol
[89] L. Hou, X. Zhang, D. Wang, A. Baccarelli, Environmental chemical exposures exposure: evidence from an ex vivo model of the fetal cerebral cortical neu-
and human epigenetics, Int. J. Epidemiol. 41 (2012) 79–105. roepithelium, J. Neurosci. 27 (2007) 8546–8557.
[90] Y. Zhang, Y. Jia, R. Zheng, Y. Guo, Y. Wang, H. Guo, M. Fei, S. Sun, Plasma [116] I.P. Pogribny, V.P. Tryndyak, A. Boyko, R. Rodriguez-Juarez, F.A. Beland, O.
microRNA-122 as a biomarker for viral-, alcohol-, and chemical-related hep- Kovalchuk, Induction of microRNAome deregulation in rat liver by long-term
atic diseases, Clin. Chem. 56 (2010) 1830–1838. tamoxifen exposure, Mutat. Res. 619 (2007) 30–37.
[91] K. Wang, S. Zhang, B. Marzolf, P. Troisch, A. Brightman, Z. Hu, L.E. Hood, D.J. [117] L. Rossi, E. Bonmassar, I. Faraoni, Modification of miR gene expression pattern
Galas, Circulating microRNAs, potential biomarkers for drug-induced liver in human colon cancer cells following exposure to 5-fluorouracil in vitro,
injury, Proc. Natl. Acad. Sci. U.S.A. 106 (2009) 4402–4407. Pharmacol. Res. 56 (2007) 248–253.
[92] T. Yokoi, M. Nakajima, Toxicological implications of modulation of gene [118] A.K. Farraj, M.S. Hazari, N. Haykal-Coates, C. Lamb, D.W. Winsett, Y. Ge, A.D.
expression by microRNAs, Toxicol. Sci. 123 (2011) 1–14. Ledbetter, A.P. Carll, M. Bruno, A. Ghio, D.L. Costa, ST depression, arrhythmia,
[93] A. Banerjee, K. Luettich, MicroRNAs as potential biomarkers of smoking- vagal dominance, and reduced cardiac micro-RNA in particulate-exposed rats,
related diseases, Biomark Med. 6 (2012) 671–684. Am. J. Resp. Cell Mol. Biol. 44 (2011) 185–196.
[94] K. Takahashi, S. Yokota, N. Tatsumi, T. Fukami, T. Yokoi, M. Nakajima, Cigarette [119] A. Izzotti, R. Balansky, F. D’Agostini, M. Longobardi, C. Cartiglia, S. La Maes-
smoking substantially alters plasma microRNA profiles in healthy subjects, tra, R.T. Micale, A. Camoirano, G. Ganchev, M. Iltcheva, V.E. Steele, S. De
Toxicol. Appl. Pharmacol. 272 (2013) 154–160. Flora, Relationships between pulmonary micro-RNA and proteome profiles,
[95] V. Bollati, B. Marinelli, P. Apostoli, M. Bonzini, F. Nordio, M. Hoxha, V. Pego- systemic cytogenetic damage and lung tumors in cigarette smoke-exposed
raro, V. Motta, L. Tarantini, L. Cantone, J. Schwartz, P.A. Bertazzi, A. Baccarelli, mice treated with chemopreventive agents, Carcinogenesis 34 (2013)
Exposure to metal-rich particulate matter modifies the expression of candi- 2322–2329.
date microRNAs in peripheral blood leukocytes, Environ. Health Perspect. 118 [120] K. Taki, T. Fukushima, R. Ise, I. Horii, T. Yoshida, Microarray analysis of 6-
(2010) 763–768. mercaptopurine-induced-toxicity-related genes and microRNAs in the rat
[96] B.A. Dickinson, H.M. Semus, R.L. Montgomery, C. Stack, P.A. Latimer, placenta, J. Toxicol. Sci. 38 (2013) 159–167.
S.M. Lewton, J.M. Lynch, T.G. Hullinger, A.G. Seto, E. van Rooij, Plasma [121] J. Wu, T. Yang, X. Li, Q. Yang, R. Liu, J. Huang, Y. Li, C. Yang, Y. Jiang, Alter-
microRNAs serve as biomarkers of therapeutic efficacy and disease pro- ation of serum miR-206 and miR-133b is associated with lung carcinogenesis
gression in hypertension-induced heart failure, Eur. J. Heart Fail. 15 (2010) induced by 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone, Toxicol. Appl.
650–659. Pharmacol. 267 (2013) 238–246.
[97] J.M. Lorenzen, T. Thum, Circulating and urinary microRNAs in kidney disease, [122] F.M. Cui, J.X. Li, Q. Chen, H.B. Du, S.Y. Zhang, J.H. Nie, J.P. Cao, P.K. Zhou, T.K.
Clin. J. Am. Soc. Nephrol. 7 (2012) 1528–1533. Hei, J. Tong, Radon-induced alterations in micro-RNA expression profiles in
[98] N. Wang, Y. Zhou, L. Jiang, D. Li, J. Yang, C.Y. Zhang, K. Zen, Urinary microRNA- transformed BEAS2B cells, J. Toxicol. Environ. Health 76 (2013) 107–119.
10a and microRNA-30d serve as novel, sensitive and specific biomarkers for [123] K. Juhasz, K. Gombos, M. Szirmai, K. Gocze, V. Wolher, P. Revesz, I. Magda,
kidney injury, PloS One 7 (2012) e51140. A. Sebestyen, A. Nemeth, I. Ember, Very early effect of DMBA and MNU on
[99] J. Du, X. Cao, L. Zou, Y. Chen, J. Guo, Z. Chen, S. Hu, Z. Zheng, MicroRNA-21 microRNA expression, In vivo 27 (2013) 113–117.
and risk of severe acute kidney injury and poor outcomes after adult cardiac [124] S. Starckx, A. Batheja, G.R. Verheyen, S.D. Jonghe, K. Steemans, B.V. Dijck, M.
surgery, PloS One 8 (2013) e63390. Singer, N. Bogdan, J. Snoeys, P. Vinken, J.C. Sasaki, J.V. Gompel, P. Guzzie-
[100] F. Wang, C. Li, W. Liu, Y. Jin, Effect of exposure to volatile organic compounds Peck, A. Lampo, L. Lammens, Evaluation of miR-122 and Other Biomarkers in
(VOCs) on airway inflammatory response in mice, J. Toxicol. Sci. 37 (2012) Distinct Acute Liver Injury in Rats, Toxicol. Pathol. 41 (2013) 795–804.
739–748. [125] N.P. Singh, U.P. Singh, H. Guan, P. Nagarkatti, M. Nagarkatti, Prenatal exposure
[101] J. Bolleyn, J. Fraczek, M. Vinken, D. Lizarraga, S. Gaj, J.H. van Delft, to TCDD triggers significant modulation of microRNA expression profile in the
V. Rogiers, T. Vanhaecke, Effect of Trichostatin A on miRNA expres- thymus that affects consequent gene expression, PLoS One 7 (2012) e45054.
sion in cultures of primary rat hepatocytes, Toxicol. In Vitro 25 (2011) [126] M.J. Jenny, N. Aluru, M.E. Hahn, Effects of short-term exposure to 2,3,7,8-
1173–1182. tetrachlorodibenzo-p-dioxin on microRNA expression in zebrafish embryos,
[102] C.J. Marsit, K. Eddy, K.T. Kelsey, MicroRNA responses to cellular stress, Cancer Toxicol. Appl. Pharmacol. 264 (2012) 262–273.
Res. 66 (2006) 10843–10848. [127] C. Vacchi-Suzzi, Y. Bauer, B.R. Berridge, S. Bongiovanni, K. Gerrish, H.K.
[103] Y. Cao, S.L. Yu, Y. Wang, G.Y. Guo, Q. Ding, R.H. An, MicroRNA-dependent Hamadeh, M. Letzkus, J. Lyon, J. Moggs, R.S. Paules, F. Pognan, F. Staedtler,
regulation of PTEN after arsenic trioxide treatment in bladder cancer cell line M.P. Vidgeon-Hart, O. Grenet, P. Couttet, Perturbation of micro RNAs in rat
T24, Tumour Biol. 32 (2011) 179–188. heart during chronic doxorubicin treatment, PLoS One 7 (2012) e40395.
[104] A.I. Pogue, Y.Y. Li, J.G. Cui, Y. Zhao, T.P. Kruck, M.E. Percy, M.A. Tarr, W.J. Lukiw, [128] P. Brzuzan, M. Wozny, L. Wolinska, A. Piasecka, Expression profiling in vivo
Characterization of an NF-kappaB-regulated, miRNA-146a-mediated down- demonstrates rapid changes in liver microRNA levels of whitefish (Coregonus
regulation of complement factor H (CFH) in metal-sulfate-stressed human lavaretus) following microcystin-LR exposure, Aquat. Toxicol. 122–123 (2012)
brain cells, J. Inorg. Biochem. 103 (2009) 1591–1595. 188–196.
[105] W.J. Lukiw, A.I. Pogue, Induction of specific micro RNA (miRNA) species by [129] R.P. Dippold, R. Vadigepalli, G.E. Gonye, J.B. Hoek, Chronic ethanol feeding
ROS-generating metal sulfates in primary human brain cells, J. Inorg. Biochem. enhances miR-21 induction during liver regeneration while inhibiting prolif-
101 (2007) 1265–1269. eration in rats, Am. J. Physiol. 303 (2012) G733–G743.
[106] V. Bollati, B. Marinelli, P. Apostoli, M. Bonzini, F. Nordio, M. Hoxha, V. Pego- [130] J.A. Bourdon, A.T. Saber, S. Halappanavar, P.A. Jackson, D. Wu, K.S. Hougaard,
raro, V. Motta, L. Tarantini, L. Cantone, J. Schwartz, P.A. Bertazzi, A. Baccarelli, N.R. Jacobsen, A. Williams, U. Vogel, H. Wallin, C.L. Yauk, Carbon black
Exposure to metal-rich particulate matter modifies the expression of can- nanoparticle intratracheal installation results in large and sustained changes
didate microRNAs in peripheral blood leukocytes, Environ. Health. Perspect. in the expression of miR-135b in mouse lung, Environ. Mol. Mutagen. 53
118 (2010) 763–768. (2012) 462–468.
56 B. Siddeek et al. / Mutation Research 764–765 (2014) 46–57

[131] J.C. States, A.V. Singh, T.B. Knudsen, E.C. Rouchka, N.O. Ngalame, G.E. Arteel, Real-time PCR of 13 mature microRNAs differentially expressed in colorectal
Y. Piao, M.S. Ko, Prenatal arsenic exposure alters gene expression in the adult cancer and non-tumoral tissues, Mol. Cancer 5 (2006) 29.
liver to a proinflammatory state contributing to accelerated atherosclerosis, [157] M. Bloomston, W.L. Frankel, F. Petrocca, S. Volinia, H. Alder, J.P. Hagan, C.G. Liu,
PLoS One 7 (2012) e38713. D. Bhatt, C. Taccioli, C.M. Croce, MicroRNA expression patterns to differentiate
[132] J. Ward, S. Bala, J. Petrasek, G. Szabo, Plasma microRNA profiles distinguish pancreatic adenocarcinoma from normal pancreas and chronic pancreatitis,
lethal injury in acetaminophen toxicity: a research study, World J. Gastroen- JAMA 297 (2007) 1901–1908.
terol. 18 (2012) 2798–2804. [158] X.X. Pu, G.L. Huang, H.Q. Guo, C.C. Guo, H. Li, S. Ye, S. Ling, L. Jiang,
[133] J. Saikumar, D. Hoffmann, T.M. Kim, V.R. Gonzalez, Q. Zhang, P.L. Goering, Y. Tian, T.Y. Lin, Circulating miR-221 directly amplified from plasma is
R.P. Brown, V. Bijol, P.J. Park, S.S. Waikar, V.S. Vaidya, Expression, circulation, a potential diagnostic and prognostic marker of colorectal cancer and
and excretion profile of microRNA-21, -155, and -18a following acute kidney is correlated with p53 expression, J. Gastroenterol. Hepatol. 25 (2010)
injury, Toxicol. Sci. 129 (2012) 256–267. 1674–1680.
[134] M.A. Parasramka, S. Ali, S. Banerjee, T. Deryavoush, F.H. Sarkar, S. Gupta, [159] R. Morimura, S. Komatsu, D. Ichikawa, H. Takeshita, M. Tsujiura, H. Nagata,
Garcinol sensitizes human pancreatic adenocarcinoma cells to gemcitabine H. Konishi, A. Shiozaki, H. Ikoma, K. Okamoto, T. Ochiai, H. Taniguchi, E.
in association with microRNA signatures, Mol. Nutr. Food Res. 57 (2013) Otsuji, Novel diagnostic value of circulating miR-18a in plasma of patients
235–248. with pancreatic cancer, Br. J. Cancer 105 (2011) 1733–1740.
[135] F. Wang, W. Liu, J. Ma, M. Yu, Y. Jin, J. Dai, Prenatal and neonatal exposure [160] H. Cheng, L. Zhang, D.E. Cogdell, H. Zheng, A.J. Schetter, M. Nykter, C.C. Har-
to perfluorooctane sulfonic acid results in changes in miRNA expression pro- ris, K. Chen, S.R. Hamilton, W. Zhang, Circulating plasma MiR-141 is a novel
files and synapse associated proteins in developing rat brains, Environ. Sci. biomarker for metastatic colon cancer and predicts poor prognosis, PLoS One
Technol. 46 (2012) 6822–6829. 6 (2011) e17745.
[136] M. Fabbri, C. Urani, M.G. Sacco, C. Procaccianti, L. Gribaldo, Whole genome [161] G. Gabriely, T. Wurdinger, S. Kesari, C.C. Esau, J. Burchard, P.S. Linsley, A.M.
analysis and microRNAs regulation in HepG2 cells exposed to cadmium, Altex Krichevsky, MicroRNA 21 promotes glioma invasion by targeting matrix met-
29 (2012) 173–182. alloproteinase regulators, Mol. Cell. Biol. 28 (2008) 5369–5380.
[137] G. Zhang, L. Sun, X. Lu, Z. Chen, P.J. Duerksen-Hughes, H. Hu, X. Zhu, J. Yang, [162] P.S. Mitchell, R.K. Parkin, E.M. Kroh, B.R. Fritz, S.K. Wyman, E.L. Pogosova-
Cisplatin treatment leads to changes in nuclear protein and microRNA expres- Agadjanyan, A. Peterson, J. Noteboom, K.C. O’Briant, A. Allen, D.W. Lin, N.
sion, Mutat. Res. 746 (2012) 66–77. Urban, C.W. Drescher, B.S. Knudsen, D.L. Stirewalt, R. Gentleman, R.L. Ves-
[138] M.K. Song, M. Song, H.S. Choi, Y.J. Kim, Y.K. Park, J.C. Ryu, Identification of sella, P.S. Nelson, D.B. Martin, M. Tewari, Circulating microRNAs as stable
molecular signatures predicting the carcinogenicity of polycyclic aromatic blood-based markers for cancer detection, Proc. Natl. Acad. Sci. U.S.A. 105
hydrocarbons (PAHs), Toxicol. Lett. 212 (2012) 18–28. (2008) 10513–10518.
[139] B.H. Dai, L. Geng, Y. Wang, C.J. Sui, F. Xie, R.X. Shen, W.F. Shen, J.M. Yang, [163] A. Schaefer, M. Jung, H.J. Mollenkopf, I. Wagner, C. Stephan, F. Jentzmik, K.
microRNA-199a-5p protects hepatocytes from bile acid-induced sustained Miller, M. Lein, G. Kristiansen, K. Jung, Diagnostic and prognostic implica-
endoplasmic reticulum stress, Cell Death Dis. 4 (2013) e604. tions of microRNA profiling in prostate carcinoma, Int. J. Cancer 126 (2010)
[140] Y. Teng, T.T. Manavalan, C. Hu, S. Medjakovic, A. Jungbauer, C.M. Klinge, 1166–1176.
Endocrine disruptors fludioxonil and fenhexamid stimulate miR-21 expres- [164] M. Tanaka, K. Oikawa, M. Takanashi, M. Kudo, J. Ohyashiki, K. Ohyashiki, M.
sion in breast cancer cells, Toxicol. Sci. 131 (2013) 71–83. Kuroda, Down-regulation of miR-92 in human plasma is a novel marker for
[141] S.L. Tilghman, M.R. Bratton, H.C. Segar, E.C. Martin, L.V. Rhodes, M. Li, J.A. acute leukemia patients, PLoS One 4 (2009) e5532.
McLachlan, T.E. Wiese, K.P. Nephew, M.E. Burow, Endocrine disruptor regu- [165] Z. Huang, D. Huang, S. Ni, Z. Peng, W. Sheng, X. Du, Plasma microRNAs are
lation of microRNA expression in breast carcinoma cells, PLoS One 7 (2012) promising novel biomarkers for early detection of colorectal cancer, Int. J.
e32754. Cancer 127 (2010) 118–126.
[142] J.S. Choi, J.H. Oh, H.J. Park, M.S. Choi, S.M. Park, S.J. Kang, M.J. Oh, S.J. Kim, S.Y. [166] R. Mahn, L.C. Heukamp, S. Rogenhofer, A. von Ruecker, S.C. Muller, J. Ellinger,
Hwang, S. Yoon, miRNA regulation of cytotoxic effects in mouse Sertoli cells Circulating microRNAs (miRNA) in serum of patients with prostate cancer,
exposed to nonylphenol, Reprod. Biol. Endocrinol. 9 (2011) 126. Urology 77 (2011), 1265 e1269-1216.
[143] C. Benoit, H. Ould-Hamouda, D. Crepin, A. Gertler, L. Amar, M. Taouis, Early [167] M.J. Lodes, M. Caraballo, D. Suciu, S. Munro, A. Kumar, B. Anderson, Detection
leptin blockade predisposes fat-fed rats to overweight and modifies hypotha- of cancer with serum miRNAs on an oligonucleotide microarray, PLoS One 4
lamic microRNAs, J. Endocrinol. 218 (2013) 35–47. (2009) e6229.
[144] J. Ahn, H. Lee, C.H. Jung, T. Ha, Lycopene inhibits hepatic steatosis via [168] H.M. Heneghan, N. Miller, A.J. Lowery, K.J. Sweeney, M.J. Kerin, MicroRNAs as
microRNA-21-induced downregulation of fatty acid-binding protein 7 in mice novel biomarkers for breast cancer, J. Oncol. 2009 (2009) 950201.
fed a high-fat diet, Mol. Nutr. Food Res. 56 (2012) 1665–1674. [169] N. Yanaihara, N. Caplen, E. Bowman, M. Seike, K. Kumamoto, M. Yi, R.M.
[145] Y.M. Yang, S.Y. Seo, T.H. Kim, S.G. Kim, Decrease of microRNA-122 causes Stephens, A. Okamoto, J. Yokota, T. Tanaka, G.A. Calin, C.G. Liu, C.M. Croce,
hepatic insulin resistance by inducing protein tyrosine phosphatase 1B, which C.C. Harris, Unique microRNA molecular profiles in lung cancer diagnosis and
is reversed by licorice flavonoid, Hepatology 56 (2012) 2209–2220. prognosis, Cancer Cell 9 (2006) 189–198.
[146] C. Boesch-Saadatmandi, A.E. Wagner, S. Wolffram, G. Rimbach, Effect of [170] R. Spizzo, M.S. Nicoloso, L. Lupini, Y. Lu, J. Fogarty, S. Rossi, B. Zagatti, M.
quercetin on inflammatory gene expression in mice liver in vivo - role of Fabbri, A. Veronese, X. Liu, R. Davuluri, C.M. Croce, G. Mills, M. Negrini, G.A.
redox factor 1, miRNA-122 and miRNA-125b, Pharmacol. Res. 65 (2012) Calin, miR-145 participates with TP53 in a death-promoting regulatory loop
523–530. and targets estrogen receptor-alpha in human breast cancer cells, Cell Death
[147] H. Yin, M. Hu, R. Zhang, Z. Shen, L. Flatow, M. You, MicroRNA-217 promotes Differ. 17 (2010) 246–254.
ethanol-induced fat accumulation in hepatocytes by down-regulating SIRT1, [171] S.L. Yu, H.Y. Chen, G.C. Chang, C.Y. Chen, H.W. Chen, S. Singh, C.L. Cheng, C.J.
J. Biol. Chem. 287 (2012) 9817–9826. Yu, Y.C. Lee, H.S. Chen, T.J. Su, C.C. Chiang, H.N. Li, Q.S. Hong, H.Y. Su, C.C.
[148] A.E. Wagner, C. Boesch-Saadatmandi, J. Dose, G. Schultheiss, G. Rimbach, Anti- Chen, W.J. Chen, C.C. Liu, W.K. Chan, K.C. Li, J.J. Chen, P.C. Yang, MicroRNA
inflammatory potential of allyl-isothiocyanate–role of Nrf2, NF-(kappa) B and signature predicts survival and relapse in lung cancer, Cancer Cell 13 (2008)
microRNA-155, J. Cell. Mol. Med. 16 (2012) 836–843. 48–57.
[149] Y.J. Kim, S.H. Hwang, H.H. Cho, K.K. Shin, Y.C. Bae, J.S. Jung, MicroRNA 21 [172] C. Roth, B. Rack, V. Muller, W. Janni, K. Pantel, H. Schwarzenbach, Circulating
regulates the proliferation of human adipose tissue-derived mesenchymal microRNAs as blood-based markers for patients with primary and metastatic
stem cells and high-fat diet-induced obesity alters microRNA 21 expression breast cancer, Breast Cancer Res. 12 (2010) R90.
in white adipose tissues, J. Cell. Physiol. 227 (2012) 183–193. [173] M. Garofalo, G. Di Leva, G. Romano, G. Nuovo, S.S. Suh, A. Ngankeu, C. Taccioli,
[150] P. Parra, F. Serra, A. Palou, Expression of adipose microRNAs is sensitive to F. Pichiorri, H. Alder, P. Secchiero, P. Gasparini, A. Gonelli, S. Costinean, M.
dietary conjugated linoleic acid treatment in mice, PLoS One 5 (2010) e13005. Acunzo, G. Condorelli, C.M. Croce, miR-221&222 regulate TRAIL resistance and
[151] R. Takanabe, K. Ono, Y. Abe, T. Takaya, T. Horie, H. Wada, T. Kita, N. Satoh, A. enhance tumorigenicity through PTEN and TIMP3 downregulation, Cancer
Shimatsu, K. Hasegawa, Up-regulated expression of microRNA-143 in associa- Cell 16 (2009) 498–509.
tion with obesity in adipose tissue of mice fed high-fat diet, Biochem. Biophys. [174] H. Si, X. Sun, Y. Chen, Y. Cao, S. Chen, H. Wang, C. Hu, Circulating microRNA-
Res. Commun. 376 (2008) 728–732. 92a and microRNA-21 as novel minimally invasive biomarkers for primary
[152] E.K. Ng, W.W. Chong, H. Jin, E.K. Lam, V.Y. Shin, J. Yu, T.C. Poon, S.S. Ng, J.J. breast cancer, J. Cancer Res. Clin. Oncol. 139 (2013) 223–229.
Sung, Differential expression of microRNAs in plasma of patients with col- [175] S. Gilad, E. Meiri, Y. Yogev, S. Benjamin, D. Lebanony, N. Yerushalmi, H. Ben-
orectal cancer: a potential marker for colorectal cancer screening, Gut 58 jamin, M. Kushnir, H. Cholakh, N. Melamed, Z. Bentwich, M. Hod, Y. Goren, A.
(2009) 1375–1381. Chajut, Serum microRNAs are promising novel biomarkers, PLoS One 3 (2008)
[153] T. Ueda, S. Volinia, H. Okumura, M. Shimizu, C. Taccioli, S. Rossi, H. Alder, C.G. e3148.
Liu, N. Oue, W. Yasui, K. Yoshida, H. Sasaki, S. Nomura, Y. Seto, M. Kaminishi, [176] J. Silva, V. Garcia, A. Zaballos, M. Provencio, L. Lombardia, L. Almonacid, J.M.
G.A. Calin, C.M. Croce, Relation between microRNA expression and progres- Garcia, G. Dominguez, C. Pena, R. Diaz, M. Herrera, A. Varela, F. Bonilla,
sion and prognosis of gastric cancer: a microRNA expression analysis, Lancet Vesicle-related microRNAs in plasma of nonsmall cell lung cancer patients
Oncol. 11 (2010) 136–146. and correlation with survival, Eur. Respir. J. 37 (2011) 617–623.
[154] J.C. Brase, M. Johannes, T. Schlomm, M. Falth, A. Haese, T. Steuber, T. Beiss- [177] K. Murata, H. Yoshitomi, S. Tanida, M. Ishikawa, K. Nishitani, H. Ito, T.
barth, R. Kuner, H. Sultmann, Circulating miRNAs are correlated with tumor Nakamura, Plasma and synovial fluid microRNAs as potential biomarkers of
progression in prostate cancer, Int. J. Cancer 128 (2011) 608–616. rheumatoid arthritis and osteoarthritis, Arthritis Res. Ther. 12 (2010) R86.
[155] X. Li, Y. Zhang, J. Ding, K. Wu, D. Fan, Survival prediction of gastric cancer by [178] S. Fichtlscherer, S. De Rosa, H. Fox, T. Schwietz, A. Fischer, C. Liebetrau, M.
a seven-microRNA signature, Gut 59 (2010) 579–585. Weber, C.W. Hamm, T. Roxe, M. Muller-Ardogan, A. Bonauer, A.M. Zeiher, S.
[156] E. Bandres, E. Cubedo, X. Agirre, R. Malumbres, R. Zarate, N. Ramirez, A. Dimmeler, Circulating microRNAs in patients with coronary artery disease,
Abajo, A. Navarro, I. Moreno, M. Monzo, J. Garcia-Foncillas, Identification by Circ. Res. 107 (2010) 677–684.
B. Siddeek et al. / Mutation Research 764–765 (2014) 46–57 57

[179] A.M. Zahm, M. Thayu, N.J. Hand, A. Horner, M.B. Leonard, J.R. Friedman, of muscular dystrophy animal models: promising novel blood-based markers
Circulating microRNA is a biomarker of pediatric Crohn disease, J. Pediatr. for muscular dystrophy, PLoS One 6 (2011) e18388.
Gastroenterol. Nutr. 53 (2011) 26–33. [183] M. Abu-Halima, M. Hammadeh, J. Schmitt, P. Leidinger, A. Keller, E. Meese,
[180] D.S. Karolina, A. Armugam, S. Tavintharan, M.T. Wong, S.C. Lim, C.F. Sum, K. C. Backes, Altered microRNA expression profiles of human spermatozoa in
Jeyaseelan, MicroRNA 144 impairs insulin signaling by inhibiting the expres- patients with different spermatogenic impairments, Fertil. Steril. 99 (2013)
sion of insulin receptor substrate 1 in type 2 diabetes mellitus, PLoS One 6 1249–1255, e1216.
(2011) e22839. [184] T. Liu, W. Cheng, Y. Gao, H. Wang, Z. Liu, Microarray analysis of microRNA
[181] J.P. Cogswell, J. Ward, I.A. Taylor, M. Waters, Y. Shi, B. Cannon, K. Kelnar, J. expression patterns in the semen of infertile men with semen abnormalities,
Kemppainen, D. Brown, C. Chen, R.K. Prinjha, J.C. Richardson, A.M. Saunders, Mol. Med. Rep. 6 (2012) 535–542.
A.D. Roses, C.A. Richards, Identification of miRNA changes in Alzheimer’s [185] W. Wu, Z. Hu, Y. Qin, J. Dong, J. Dai, C. Lu, W. Zhang, H. Shen, Y. Xia, X. Wang,
disease brain and CSF yields putative biomarkers and insights into disease Seminal plasma microRNAs: potential biomarkers for spermatogenesis sta-
pathways, J. Alzheimers Dis. 14 (2008) 27–41. tus, Mol. Hum. Reprod. 18 (2012) 489–497.
[182] H. Mizuno, A. Nakamura, Y. Aoki, N. Ito, S. Kishi, K. Yamamoto, M. Sekiguchi,
S. Takeda, K. Hashido, Identification of muscle-specific microRNAs in serum

You might also like