Mecanismos Moleculares de La Toxicidad de La Acroleína-Relevancia A La Enfermedad Humana

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

TOXICOLOGICAL SCIENCES, 143(2), 2015, 242–255

doi: 10.1093/toxsci/kfu233
REVIEW

Molecular Mechanisms of Acrolein Toxicity: Relevance


to Human Disease

Downloaded from https://academic.oup.com/toxsci/article/143/2/242/1676672 by guest on 10 November 2022


Akshata Moghe*, Smita Ghare†, Bryan Lamoreau†, Mohammad Mohammad†,
Shirish Barve*,†, Craig McClain*,†,‡, and Swati Joshi-Barve*,†,1
*Department of Pharmacology and Toxicology, †Department of Medicine and ‡Robley Rex VAMC, Louisville,
Kentucky 40202
1
To whom correspondence should be addressed at Department of Medicine, University of Louisville, 505 S. Hancock Street, Room 505 CTRB, Louisville,
Kentucky 40202. Fax: 502-852-8927. E-mail: s0josh01@louisville.edu.

ABSTRACT
Acrolein, a highly reactive unsaturated aldehyde, is a ubiquitous environmental pollutant and its potential as a serious
environmental health threat is beginning to be recognized. Humans are exposed to acrolein per oral (food and water),
respiratory (cigarette smoke, automobile exhaust, and biocide use) and dermal routes, in addition to endogenous
generation (metabolism and lipid peroxidation). Acrolein has been suggested to play a role in several disease states
including spinal cord injury, multiple sclerosis, Alzheimer’s disease, cardiovascular disease, diabetes mellitus, and neuro-,
hepato-, and nephro-toxicity. On the cellular level, acrolein exposure has diverse toxic effects, including DNA and protein
adduction, oxidative stress, mitochondrial disruption, membrane damage, endoplasmic reticulum stress, and immune
dysfunction. This review addresses our current understanding of each pathogenic mechanism of acrolein toxicity, with
emphasis on the known and anticipated contribution to clinical disease, and potential therapies.

Key words: antioxidants; apoptosis; DNA adducts; inflammation; exposure; environmental; oxidative injury

Acrolein (CAS No 107-02-8) is listed by the Department of propagated. Our analysis suggests that such a common unifying
Homeland Security (DHS), Agency for Toxic Substances and pathway might not exist or is not currently discernable, placing
Disease Registry (ATSDR), and Environmental Protection even more onus on the need to further investigate acrolein tox-
Agency (EPA) as a high priority toxic chemical (Toxicological icity in various disease conditions. This comprehensive review
Profile For Acrolein, 2007), with recommendations on acrolein starts with succinct introductory comments on the generation,
exposure levels in the environment, food, and water. Acrolein is sources, and metabolism of acrolein, followed by a detailed dis-
an industrial chemical and a biocide, and over 500 million cussion on the various mechanisms of acrolein toxicity, which
pounds of acrolein are produced annually in the United States include protein adduction, oxidative stress, mitochondrial dys-
(Toxicological Profile For Acrolein, 2007). The past decade has function, DNA adduction, endoplasmic reticulum (ER) stress, in-
witnessed a dramatic increase in research regarding acrolein, flammation and immune alterations, structural and membrane
and this review attempts to assimilate this growing literature effects, and dysregulated signal transduction.
with a focus on the molecular mechanisms of acrolein toxicity
pertaining to human disease. Although some early literature
has been cited, this review is focused on more recent literature.
ACROLEIN SOURCES AND METABOLISM
The theme of this review is that there are multiple mechanisms Sources. Acrolein is a common dietary and environmental pollu-
involved in acrolein toxicity. In our extensive appraisal of the tant and is also generated by cellular metabolism (Figure 1). It is
current literature, our goal was not only to characterize these an a,b-unsaturated aldehyde released during the combustion of
various mechanisms, but also to identify an overarching path- petroleum fuels, biodiesel, plastic, paper and wood, and is a
way through which acrolein toxicity may be initiated and major component of tobacco smoke; such exposures are directly

C The Author 2015. Published by Oxford University Press on behalf of the Society of Toxicology.
V
All rights reserved. For Permissions, please e-mail: journals.permissions@oup.com

242
MOGHE ET AL. | 243

SOURCES

Endogenous Dietary Exogenous

Lipid Peroxidation Fried food Automobile Exhaust


Anti-Cancer Drugs Alcoholic beverages Cigarette Smoke
Polyamines Charred Meat Industrial Waste
Threonine Forest Fires

ACROLEIN

Downloaded from https://academic.oup.com/toxsci/article/143/2/242/1676672 by guest on 10 November 2022


GSH-Acrolein

Protein and Acrylic acids


HPMA OPMA CEMA
DNA Adducts Glyceraldehyde
Malonic acid
METABOLITES
FIG. 1. The sources and metabolic fate of acrolein. GSH: glutathione; CEMA: 2-carboxyethylmercapturic acid; HPMA: 3-hydroxypropylmercapturic acid; OPMA: S-(3-oxo-
propyl)-N-acetylcysteine.

relevant in dermal and respiratory toxicity. Firefighters, acrolein or may be mediated by glutathione-S-transferases; these acro-
industry workers, persons inhabiting densely polluted cities, lein-GSH conjugates may be metabolized by enzymatic cleavage
and cigarette and hookah smokers (Kassem et al., 2014) face sub- of c-glutamic acid and glycine residues. Further metabolism via
stantial exposures (reviewed in Cahill (2014); Stevens and Maier aldehyde dehydrogenase results in 2-carboxyethylmercapturic
(2008)). Acrolein has been identified as a health hazard com- acid (CEMA), or catalyzed by aldo-keto reductase results in S-(3-
monly found at significant levels in most homes and schools in hydroxypropyl)-N-acetylcysteine (3-HPMA), the main urinary
the United States (Logue et al., 2012). Acrolein can be generated metabolite of acrolein. Acrylic acid and glycidaldehyde are alter-
from overheated vegetable and animal fats; hence, cooked, nate acrolein by-products, should oxidation and epoxidation pre-
fried, and charred foods are notable sources of acrolein, as are cede its conjugation with GSH; glycidaldehyde may represent a
beer, wine, rum, and breads; the best current estimate for human health risk as it has carcinogenic properties in mice and
Tolerable Daily Intake (TDI) for acrolein is 7.5 mg/kg body weight rats when applied dermally. Acrolein may be also converted to
(reviewed in Abraham et al. (2011)). Moreover, acrolein exposure malonic acid by aldehyde dehydrogenase. Finally, acrolein may
from consumption of foods may be considerably underesti- form irreversible adducts with cellular nucleophiles, a metabolic
mated (Watzek et al., 2012). Exposure to reactive unsaturated reaction that may be the basis of several of its toxic effects
aldehydes like acrolein by dietary consumption is estimated at (Figure 1) (reviewed in Stevens and Maier (2008)).
nearly 5 mg/kg of food (Wang et al., 2008). The gastrointestinal
(GI) system is likely affected the most by ingested acrolein. MECHANISMS OF TOXICITY
Notably, acrolein can also be generated endogenously during
cellular metabolism by (1) degradation of threonine by neutro- An interesting feature of acrolein is the diversity with which it
phil-derived myeloperoxidase; (2) amine oxidase-mediated exerts its toxic effects in different tissues and organs. Both dis-
catabolism of polyamines such as spermine and spermidine; (3) ease- and organ-specific, as well as shared mechanisms have
metabolism of cancer drugs such as cyclophosphamide; and (4) distinct value in the search for therapeutic targets; the former
lipid peroxidation (LPO) of polyunsaturated fatty acids (PUFAs) enables specific, targeted therapy, whereas the latter facilitates
(Stevens and Maier, 2008; Uchida et al., 1998a,b). The levels of the broad management of acrolein toxicity. In the following sec-
acrolein formed by metabolism are difficult to quantify, and tions, the various direct mechanisms of acrolein toxicity such
may reach very high levels in certain cellular microenviron- as protein and DNA adduction, and indirect mechanisms
ments; hence, a determination of adverse effects is challenging. including induction of oxidative, mitochondrial, and ER stress,
Importantly, both acute and chronic acrolein exposures are are critically discussed with possible connection to affected
described to have significant adverse effects (Centers for organs and pathological conditions. Wherever known or tested,
Disease and Prevention, 2013). the effectiveness of acrolein scavengers and other compounds
in mitigating acrolein toxicity has also been described (Table 1).
Metabolism. Acrolein is highly water soluble and can rapidly enter
bodily tissues. It is known to rapidly form conjugates with cellular Protein Adduction
glutathione (GSH), cysteine, N-acetylcysteine (NAC), and/or thio- Being a strong electrophile, acrolein shows high reactivity with
redoxin. Acrolein-GSH conjugation can occur without a catalyst cellular nucleophiles such as proteins, DNA and RNA. Acrolein
244 | TOXICOLOGICAL SCIENCES, 2015, Vol. 143, No. 2

TABLE 1. Acrolein Toxicity and Protective Agents

Mechanisms of Acrolein Toxicity Protective Agents

Protein adduction Hydralazine


Oxidative stress NAC, Hydralazine, MESNA, Lipoic acid, Hydroxytyrosol, Vitamin E, Diallyl disulfide, Rapamycin,
L-Cysteine, Penicillamine, Aminoguanidine, Hydroxylamine, Sodium Borohydride, Sodium
Bisulfite, Ascorbic acid, Trolox
Mitochondrial dysfunction NAC, Polyethylene glycol-catalase, SOD mimetic-MnTBAP, Pifithrin- p53 inhibitor
Inflammation and Immune Alterations NAC, MESNA, U0126- AKT inhibitor, FR180204 - ERK1/2 inhibitor, Tetramethylthiourea
ER stress NAC, Caspase inhibitor, Phenylbutyric acid, JNK inhibitor
Structural and membrane effects NAC, Hydralazine
Deregulated signal transduction NAC, Rottlerin- PKC-d inhibitor, JNK inhibitor, Simvastatin, Rosiglitazone, Sildenafil, NBHA
(N-benzylhydroxylamine), SIS3- SMAD3 inhibitor

Downloaded from https://academic.oup.com/toxsci/article/143/2/242/1676672 by guest on 10 November 2022


readily targets the sulfhydryl group of cysteine, the imidazole dehydrogenase (LDH) activity was reduced by cigarette smoke
group of histidine and the amino group of lysine, to form mainly (34%), and more so by 10 mM acrolein (61%) via introducing a car-
Michael addition-type adducts or Schiff base cross-links bonyl group into the protein and causing dysfunction; this
(reviewed by Pizzimenti et al. (2013); Zarkovic et al. (2013)). These established acrolein as the main ingredient in cigarette smoke
susceptible amino acid residues have major physiological responsible for salivary LDH activity diminution (Avezov et al.,
importance and are involved in cellular processes including 2014a).
enzyme catalysis, redox signaling, reactive oxygen species A pathological role for acrolein in disease development/
(ROS) sensing, and cellular buffering; hence, adduction by acro- progression has been proposed based on the detection of higher
lein may lead to significant alterations in protein function. Also, levels of acrolein adducts in the sera of patients with various
acrolein can interfere with its own metabolism by inhibiting diseases, including diabetic nephropathy; renal disease;
two enzymes that metabolize acrolein-GSH conjugates, Sjogren’s syndrome autoimmune disorder (Brooks, 2013); cere-
namely, alcohol dehydrogenase and aldehyde dehydrogenase. bral stoke/infarction (Igarashi and Kashiwagi, 2011); colon carci-
Furthermore, acrolein may augment the effects of other known nogenesis; and diabetes (reviewed by Aldini et al. (2011)).
targets via irreversible adduction and inactivation of arylamine A recent study using sera from patients with diabetic nephrop-
N-acetyltransferases, a major family of xenobiotic-metabolizing athy described the utility of “aldehyde capture”, a procedure
enzymes involved in metabolizing many drugs and pollutants used to calculate the protein carbonyl stress derived solely from
(Bui et al., 2013). LPO, suggesting future promise in the monitoring of renal dis-
Acrolein protein adducts are assessed primarily by antibody- ease and treatment (Medina-Navarro et al., 2011). The potential
based techniques, high-performance liquid chromatography utility of plasma levels of protein-conjugated acrolein (along
(HPLC), and mass spectrometry, and such adducts have been with amyloid b40/42 (Ab40/42)) to detect and differentiate
demonstrated in vitro and in vivo in several cell types and tissues between mild cognitive impairment and Alzheimer’s disease
(Aldini et al., 2011). Alterations in protein function caused by was recently demonstrated (Mizoi et al., 2014). Thus, substantial
acrolein adduction have been demonstrated in several studies, evidence suggests that acrolein toxicity in major organ systems
albeit primarily in vitro. In 1998, Uchida et al. were the first to involves the common mechanism of formation of adducts with
show that an acrolein-lysine adduct (FDP-Lys) was involved in proteins that play critical functional roles in cellular processes.
the oxidation of human low-density lipoprotein (LDL); they also However, the question of causality versus association still
demonstrated the presence of specific antibodies against this remains, and the direct/indirect mechanisms by which the
adduct in vivo (Uchida et al., 1998b). Prabhu’s group showed that acrolein adducts may lead to injury remain to be determined.
direct exposure to acrolein (as low as 0.01 mM) induced myofila-
ment dysfunction in vitro and in vivo (1 mg/kg/day intravenous) Oxidative Stress
in mice (Ismahil et al., 2011), with accumulation of acrolein-pro- Aerobic metabolism and pathogen defense mechanisms pro-
tein adducts and protein carbonyls within the myocardium. duce ROS which can damage lipids, proteins, carbohydrates,
Burcham et al. showed in mouse hepatocytes that exposure to and nucleic acids. To counter oxidative damage, the human
allyl alcohol and its metabolite acrolein led to protein carbony- body is equipped with antioxidant compounds and antioxidant
lation, which correlated with hepatocyte death. They later enzymes. However, extensive ROS production can overwhelm
showed that the acrolein scavenger hydralazine effectively these defense mechanisms, and results in oxidative stress
trapped acrolein adducts in vivo to reduce allyl alcohol- (reviewed by Birben et al. (2012)). Acrolein is considered both a
mediated hepatotoxicity in mice, and that mobilization of the product and an initiator of LPO (Adams and Klaidman, 1993;
chaperone protein HSP90 was protective against acrolein- Uchida et al., 1998a) and is thus a perpetrator of oxidative stress.
induced carbonylation (Burcham et al., 2012). Acrolein was Both in vitro and in vivo studies have shown that acrolein can
suggested to play a role in liver injury in a murine model of itself cause oxidative damage, leading to membrane disruption,
early alcoholic liver disease and two novel in vivo protein targets DNA and mitochondrial damage and can exacerbate apoptosis.
for acrolein were identified: neutral alpha-glucosidase AB and Acrolein exposure (0.1–5 mM) in the human retinal epithelial
nesprin-1 (Galligan et al., 2012). Treatment of recombinant rat cells (ARPE19) significantly increased oxidant levels by decreas-
apoE with a 10-fold molar excess of acrolein resulted in an ing GSH, anti-oxidant enzymes (SOD and GSH-peroxidase), and
impairment in cholesterol efflux abilities, and a significant total and nuclear levels of the antioxidant regulator-Nrf2, coin-
decrease in lipid-binding and LDLr- and heparin-binding capa- cident with reduced mitochondrial function, suggesting a role
bilities, and overall stability of the protein (Tran et al., 2014). for acrolein in retinal damage and age-related macular degener-
Recently, it was demonstrated that salivary lactate ation. Notably, lipoic acid was protective (Jia et al., 2007), as was
MOGHE ET AL. | 245

hydroxytyrosol (antioxidant polyphenol in olives) via induction damage, and toxicity in cell and cell-free models; starting
of phase II detoxifying enzymes and stimulation of mitochon- with the most effective: reducing agents > thiol-containing
drial biogenesis (Zhu et al., 2010), and a-tocopherol (Vitamin E) compounds > carbonyl scavengers/amines > antioxidants/ROS
by upregulating Nrf2 (Feng et al., 2010). scavengers (MacAllister et al., 2013).
With the abundance of PUFAs available for LPO and the lack Like many other inducers of oxidative stress, acrolein is
of enzymes that metabolize reactive aldehydes, neurons and known to activate cellular antioxidant systems, most likely
the central nervous system are particularly vulnerable to oxida- as an adaptive response. These effects are seen primarily at
tive stress and acrolein toxicity (Hamann and Shi, 2009). low sublethal acrolein concentrations, and may be due to
Acrolein-induced neuronal damage was demonstrated in pigs “hormesis” described for other toxins, wherein hormesis-stimu-
and rats, suggesting that it may be a potential risk factor for spi- lating compounds initiate an adaptive response that confers
nal cord injury (SCI) (Due et al., 2014; Shi et al., 2011; Zheng et al., protection against harmful doses of the same or similar agents.
2013). The neuroprotective role of hydralazine was shown in the One such example is the acrolein-induced activation of
attenuation of acrolein-mediated damage in SCI (Park et al., Nrf2 in vitro in endothelial cells leading to induction of heme-
oxygenase-1, the enzyme that plays a major role in the cellular

Downloaded from https://academic.oup.com/toxsci/article/143/2/242/1676672 by guest on 10 November 2022


2014). Plasma levels of protein-conjugated acrolein (along with
acrolein producing enzymes—spermine oxidase and acetylpoly- antioxidative response (Wu et al., 2006); and in vivo in the lungs
amine oxidase) were shown to be good biomarkers for human of mice exposed to 5 ppm of acrolein vapor for 6 h/day for 14–17
stroke. Interestingly, an inverse correlation was seen between days (Zhang and Forman, 2008). The effects of acrolein on two
stroke and 3-HPMA the urinary GSH-metabolite of acrolein; the major redox regulators (thioredoxin and thioredoxin reductase)
authors proposed that the decrease in 3-HPMA reflects reduced have been recently discussed (Myers et al., 2011). In the event
GSH caused by increased acrolein generation at the region of that induction of cellular antioxidant systems fails or is subpar,
brain infarct (Yoshida et al., 2012). acrolein may induce pathological oxidative mechanisms that
Acrolein-induced oxidative stress is associated with severe contribute to disease development.
toxicity in the renal system due to the use of anticancer agents
such as cyclophosphamide and ifosfamide that get Mitochondrial Dysfunction
metabolized to acrolein; the acrolein toxicity is prevented by Mitochondria play a dynamic role in human cells, controlling
Mesna (2-mercaptoethane sulfonate) which binds with and oxidative phosphorylation to produce ATP, aiding in fatty acid
clears acrolein. A major contributor to acrolein toxicity in such breakdown and steroid synthesis, and playing an integral role
cases appears to be the activation of intracellular ROS and nitric in calcium signaling and apoptosis. Acrolein has been impli-
oxide production, either directly or via NF-jB and AP-1 (Korkmaz cated as a mitochondrial toxin affecting several of these func-
et al., 2007). A recent report showed that pretreatment with diallyl tions, most notably apoptosis, and whether apoptosis or
disulfide protected against cyclophosphamide-induced oxidative necrosis ensues after acrolein exposure appears to be related to
damage, histopathological lesions, and apoptosis; notably, the dose and cell type. The majority of these studies are in vitro, and
accumulation of acrolein-adducts in the bladder was reduced the lack of much-needed in vivo data raises questions about
showing that the protection was by detoxifying acrolein (Kim their relevance; nonetheless, some key studies are discussed
et al., 2014). A recent review identifies acrolein-lysine adducts as a below.
promising urinary biomarker of oxidative stress and oxidative Although the mitochondrial death pathway leading to apop-
damage (Il’yasova et al., 2012). Acrolein-induced oxidative dam- tosis typically involves the activation of caspases, acrolein-
age is shown to have an indirect impact on cardiovascular health induced apoptosis was shown to be both caspase-dependent in
via apoptosis of adult mice cardiomyocytes in vitro (Wang et al., human neuroblastoma cells (Dong et al., 2013) and in A549 lung
2011); and via myocardial oxidative stress, increased apoptosis, cells (Roy et al., 2010), and caspase-independent in CHO cells
and myocyte hypertrophy and dilated cardiomyopathy in vivo (Tanel and Averill-Bates, 2005). On the other hand, acrolein may
after long-term (48-day) oral exposure of mice to 1 mg/kg acrolein inhibit caspases and ultimately inhibit apoptosis in some cells,
(Ismahil et al., 2011). Also, acrolein-induced oxidation of apolipo- such as murine proB lymphocytes (Kern and Kehrer, 2002) and
protein A-I (apoA-I, the major high density lipoprotein) impaired in B lymphoblastoid SKW6.4 cells via direct alkylation of cas-
cellular cholesterol efflux (Shao, 2012). The exposure of oral pase active sites (Hristova et al., 2007).
tissues to environmental hazards is immense, especially in Tanel et al. showed that NAC prevented apoptotic events in
smokers, and acrolein was shown to reduce GSH, and increase CHO cells exposed to low doses of acrolein (4 fmol/cell, 30 min),
oxidative stress and protein carbonylation in oral keratinocytes including Bad and Bcl-2 translocations, depolarization of the
in vitro (Avezov et al., 2014b). Studies in chronic obstructive mitochondrial membrane potential, procaspase-9 processing,
pulmonary disease (COPD) have shown that cigarette smoke caspase-9, 7, 8 activation, and poly (ADP-ribose) polymerase
depletes total GSH levels in bronchial cells (van der Toorn et al., cleavage (Tanel and Averill-Bates, 2007b); however, apoptosis
2007) and human gingival fibroblasts (Colombo et al., 2012); in was not prevented by the Fas receptor antagonist KP7-6 (Tanel
both cell types, mass spectrometric analyses demonstrated that and Averill-Bates, 2007a). Roy demonstrated in A549 human
the GSH was depleted by binding to acrolein. Rapamycin was lung cells that low-dose acute acrolein exposures (3–27 mM,
demonstrated to inhibit acrolein-induced apoptosis in male germ 30–60 min) could be overcome by an adaptive response involv-
cells by alleviating acrolein-induced ROS, and reducing ROS- ing activation of antiapoptosis survival factors AKT and cIAP1/2,
driven mitochondrial dysfunction and apoptosis by increasing despite triggering of early apoptotic changes in Bax and cyto-
ratio of Bcl2/Bax and protecting mitochondrial membranes (He chrome c release; the survival proteins did not protect against
et al., 2014). Our work has shown that acrolein-induced oxidative higher acrolein exposures (10–50 mM) induced apoptosis (Roy
stress was accompanied by mitochondrial dysfunction and ER et al., 2009). Most adverse changes were inhibited by catalase,
stress in liver cells; acrolein toxicity was attenuated by the anti- the antioxidant polyethylene glycol-catalase, and the SOD mim-
oxidant and GSH-prodrug NAC (Mohammad et al., 2012). A recent etic-MnTBAP, reiterating the initiating role of acrolein-induced
article ranked the overall effectiveness of known protective oxidative stress. Apoptosis in A549 lung cells was also reversible
agents in suppressing acrolein-induced ROS formation, protein with pifithrin-a, an inhibitor of p53 (Roy et al., 2010). Acrolein is
246 | TOXICOLOGICAL SCIENCES, 2015, Vol. 143, No. 2

known to induce mitochondrial dysfunction leading to neuronal inhibition of repair enzymes, likely by acrolein-adduction lead-
death in HT22 mouse hippocampal cells, and lithium was ing to increased proteasome-dependent degradation (Wang
shown to protect against acrolein-induced neurotoxicity by et al., 2012) or slower repair activity (Choudhury et al., 2013).
enhancement of GSH, and attenuation of ROS and mitochon- Additionally, acrolein inhibited DNA methylase by 30%–50%
drial dysfunction caused by acrolein (Huang et al., 2014). (Cox et al., 1988), and inhibited histone acetylation and compro-
Acrolein also impacts the ability of the mitochondria to take mised chromatin assembly suggesting that acrolein exposure
part in cellular respiration. In rat liver mitochondria acrolein may also cause epigenetic alterations in gene expression (Chen
dose-dependently inhibited select components of the respira- et al., 2013). However, in vivo evidence of mutagenicity has not
tory machinery, including complexes I and II, pyruvate dehy- been demonstrated thus far.
drogenase, and a-ketoglutarate dehydrogenase, over a broad Acrolein-induced DNA damage and inhibition of DNA repair
range of acrolein exposures from 10 mM (relevant to human are likely to promote cancer development; however, the carci-
exposures) to 1000 mM (highly unlikely) (Sun et al., 2006). nogenicity of acrolein is controversial (reviewed in Abraham
Mitochondrial dysfunction due to inhibition of key mitochon- et al. (2011)). Intraperitoneal administration of acrolein (75 and
drial enzymes (pyruvate dehydrogenase and a-ketoglutarate 150 nmol per animal) increased DNA adducts but not tumor

Downloaded from https://academic.oup.com/toxsci/article/143/2/242/1676672 by guest on 10 November 2022


dehydrogenase) has been observed in Alzheimer’s disease brain incidences in male or female B6C3F1 neonatal mice, suggesting
tissue; and lipoic acid, a cofactor for these enzymes, can be that DNA adduction may not parallel cancer development (Von
covalently bound and sequestered by acrolein, thereby decreas- Tungeln et al., 2002). In contrast, a case has been made for the
ing enzyme activity. In keeping with this, supplementation with carcinogenicity of acrolein based on higher acrolein DNA
lipoic acid was shown to decrease oxidative stress and mito- adducts levels in lung (Feng et al., 2006), liver (Kawai et al., 2003),
chondrial enzyme loss, partly by scavenging of acrolein and bladder cancers (Lee et al., 2014). In an early study, acrolein
(Maczurek et al., 2008). The presence of increased protein- was postulated to promote urinary bladder cancer since intra-
carbonyl adducts in this study emphasizes that particular mito- peritoneal and intravesicular administration of acrolein demon-
chondrial enzymes may be more susceptible to covalent strated abnormal cell proliferation and hyperplasia of the
modification, thereby acting as a wrench in the cogwheels of bladder epithelium and the urothelium (Cohen et al., 1992).
electron transport. Moreover, in vivo inhalation exposure of rodents to acrolein
resulted in inflammation, hyperplasia, and squamous metapla-
DNA Adduction sia of the respiratory airway epithelium (Dorman et al., 2008). In
DNA damage can be induced by exposure to chemicals, ROS and epidemiological studies, an increased lung cancer incidence
reactive metabolites such as acrolein; DNA damage has been was reported for nonsmoking Chinese women cooking in a tra-
associated with mutations and carcinogenesis. Acrolein can ditional wok style. The women exhibited significantly higher
produce interchain cross-links of double-stranded DNA as well levels of the mercapturic acid metabolites of acrolein, crotonal-
as DNA-protein cross-links. Acrolein readily reacts with deoxy- dehyde, and benzene, suggesting a link between lung cancer
guanosine (dG) producing two exocyclic DNA adducts, a- and and volatile toxicants (such as acrolein) produced by high tem-
c-hydroxy-1, N2-propano-20 -deoxyguanosine (a-HOPdG and perature wok cooking/frying (Wang et al., 2013).
c-HOPdG), which are mutagenic to varying degrees (Tang et al., A novel and contradictory hypothesis was recently proposed
2011). Additionally, acrolein adducts of 20 -deoxyadenosine suggesting that acrolein may, in fact, be part of a physiological
(Pawlowicz et al., 2006a,b) and 20 -deoxycytidine DNA bases and anticancer system that controls the growth/proliferation of can-
thymidine (Pawlowicz and Kronberg, 2008) have been reported. cer cells (Alarcon, 2012). This hypothesis is based on properties
Acrolein-DNA adducts have been characterized in vitro, and of acrolein such as GSH depletion and direct cytotoxicity, and
have also been detected in vivo in several different animal and the fact that acrolein is a metabolite of anticancer compounds,
human tissues and cells (Tang et al., 2011; Voulgaridou et al., spermine and cyclophosphamide. The hypothesis is interesting
2011). but speculative; and needs further investigation. Detailed stud-
Acrolein adduction of DNA, if not repaired efficiently, has ies on the type and magnitude of DNA damage and mutations
the potential to cause critical gene mutations, suggesting that arising from endogenous and/or exogenous acrolein exposure,
acrolein may be mutagenic and may contribute to the process and the biological consequences are essential to understand the
of carcinogenesis. Mutagenicity studies of acrolein have yielded true role of acrolein in cancer (Figure 2).
inconsistent results, and both cell type and sequence context
appear to play a role (reviewed in Liu et al. (2010)). A lack of Inflammation and Immune Alterations
mutagenicity by acrolein was demonstrated in various shuttle Adverse health effects of acrolein are anticipated to arise from
vector-based systems (Kim et al., 2007); in contrast, Demir’s its impact on innate and adaptive immune responses.
group demonstrated dose-dependent in vitro mutagenic effects Inflammation is an essential protective immune response for
of acrolein (10 and 25 mM) using the mouse lymphoma and the combating infections; however, uncontrolled or chronic inflam-
Drosophila wing spot mutagenicity assay (Demir et al., 2013). mation is a major cause of tissue injury. Interestingly, reports of
Furthermore, CpG methylation, at CpG-rich hot spots that are acrolein-induced alterations in inflammatory signaling and
prone to acrolein-DNA adduction, increased the acrolein- gene expression, particularly via the NF-jB pathway, are diverse
induced G-to-T and G-to-A mutation frequency to levels found and often conflicting, implying a significant multilevel com-
in the CpG sites in the p53 gene, a known mutational hot spot in plexity influenced by cell type, duration of acrolein exposure,
tobacco smoke-associated lung cancer (Wang et al., 2013). In the and environmental context (reviewed by Yadav and Ramana
mitochondria, the natural absence of nucleotide excision repair (2013)).
enzymes permits accumulation of acrolein adducts in mito- Acrolein can potentially diminish defenses against bacterial
chondrial DNA, leading to transversion mutations which are and viral infections by inhibiting macrophage responses/func-
thought to contribute to neurodegenerative disorders tion via suppression of NF-jB (Horton et al., 1999; Kehrer and
(Kasiviswanathan et al., 2013). Interestingly, acrolein not only Biswal, 2000; Kirkham et al., 2004; Lambert et al., 2005), or by
induced DNA damage but also inhibited DNA repair by direct alkylation of signaling proteins (Hristova et al., 2012;
MOGHE ET AL. | 247

Acrolein

Downloaded from https://academic.oup.com/toxsci/article/143/2/242/1676672 by guest on 10 November 2022


Carcinogenesis Altered Gene Expression

Disease State

FIG. 2. Effects of acrolein on DNA.

Kasahara et al., 2008; Valacchi et al., 2005), or by suppressing M1 effectively suppressed by melatonin (Kim et al., 2012). Similar
proinflammatory reactions and favoring anti-inflammatory M2 proinflammatory effects along with increased susceptibility to
responses, consistent with observations in smokers (Hristova influenza-A virus was seen in vivo wherein mice exposed to
et al., 2012). Acrolein is shown to suppress TNF-a-induced IL-8, a acrolein for 4 days exhibited an accelerated inflammatory
proinflammatory neutrophil chemoattractant, in both primary response with higher macrophages and neutrophils, and greater
and immortalized human bronchial epithelial cells (Valacchi IL-1a, IL-1b, IL-6, TNF, IFN-c, KC, and MCP-1 in the bronchial
et al., 2005). Immunosuppressive effects of acrolein exposure fluid; the effects were suppressed by the carbonyl scavenger
were also seen in human T cells, in which acrolein directly alky- bisulphite (Ong et al., 2012). In vitro acrolein exposure of nasal
lated Cys-61 and Arg-307 of NF-jB1 (p50), significantly reducing epithelial cells was inflammatory and cytotoxic (Comer et al.,
binding to the promoters of IL-2, IL-10, TNF-a, GMCSF, and IFNc, 2014). Also, intranasal acrolein exposure of mice caused both
with a notable exception being IL-8 (Lambert et al., 2007). pulmonary inflammation and death of lung epithelial cells,
In contrast to these immunosuppressive effects, acrolein is with increased activated macrophages in the lungs and
demonstrated to activate NF-jB and induce proinflammatory increased ROS formation via induction of NF-jB signaling; these
mediators. In rat lung epithelial cells, acute acrolein exposure effects were decreased by NAC (Sun et al., 2014).
(20 mM, 6 h) induced the NF-jB dependent marker of inflamma- Recent data indicate that acrolein may contribute to the
tion COX-2 via calcium release and subsequent proteolytic deg- highly atherogenic transformation of macrophages into foam
radation of IjBa. This was reversed with GW5074, a Ras/Raf-1/ cells via modification of LDL (Watanabe et al., 2013). In keeping
ERK pathway inhibitor (Sarkar and Hayes, 2007). Acrolein with this, acrolein-lysine adducts in LDL are detected in athero-
induced an inflammatory response and increased mucin gene sclerotic lesions in humans. Acrolein also increased macro-
expression in human middle ear epithelial cells supporting the phages expression of the atherogenic factors 5-lipoxygenase,
hypothesis that acrolein may be a risk factor for otitis media leukotriene B4 and matrix metalloproteinase (Kim et al., 2010;
(Song et al., 2013). IL-8 upregulation is associated with many O’Toole et al., 2009). Other proinflammatory effects of acrolein
inflammatory disorders, and IL-8 production due to acrolein include mast cell degranulation which can increase inflamma-
exposure has been reported in vitro in multiple cell types, tory injury. Environmentally, relevant exposures of acrolein
including normal human bronchial smooth muscle cells and in (1 ppm) promoted the generation of ROS and initiated
airway epithelial cells (Moretto et al., 2012), alveolar macro- degranulation of the airway mast cells (RBL-2H3); these effects
phages, cultured normal human lung fibroblasts, and in small were significantly reduced by antioxidants NAC and tetrame-
airway epithelial cells which was prevented by antioxidants thylthiourea (Hochman et al., 2014). These studies suggest a sig-
NAC and MESNA, and by U0126 and FR180204, inhibitors of Akt nificant role for acrolein in chronic inflammatory disorders via
and ERK1/2 respectively, suggesting their mechanistic involve- dysregulation of inflammatory mediators both locally and sys-
ment in IL-8 induction (Moretto et al., 2009). Finkelstein showed temically; however, such conclusions may be premature since
acrolein-induced IL-8 production by isolated neutrophils which in-vivo studies are limited. Although acrolein appears to have
was accompanied by inhibition of apoptosis via suppressed cas- contradictory pro- and anti-inflammatory effects, they may not
pase activation, extrapolating the potential for prolonged and be incongruous. It is likely that the magnitude and duration of
increased inflammation within the lung (Finkelstein et al., 2001). acrolein exposure may critically determine outcomes, wherein
Acrolein-induced IL-8 in human pulmonary fibroblasts was acute high-dose acrolein exposure might suppress innate
248 | TOXICOLOGICAL SCIENCES, 2015, Vol. 143, No. 2

Acrolein

Acute High Chronic Low


Exposure Dose Exposure Dose

Suppression of Immune Enhancement of Inflammatory


Responses Responses
NFkB, TNFα, IL-10, IFNγ, IL-2, NFkB, IL-8, COX-2, IL-1β, IL-6,TNF,IFN γ,
GMCSF KC, MCP-1, 5-lipoxygenase, LTB4, MMP

Downloaded from https://academic.oup.com/toxsci/article/143/2/242/1676672 by guest on 10 November 2022


Susceptibility to Infections Inflammation & Tissue Injury

FIG. 3. Effects of acrolein on immune responses.

immunity and inflammatory responses thereby increasing sus- smoke-induced ER stress and activation of PERK, and ATF4
ceptibility to infections, whereas chronic low-dose exposures (Hengstermann and Muller, 2008). In vivo acute (a single intra-
may increase inflammation leading to tissue injury (Figure 3). peritoneal injection of acrolein 12 mmol/kg) and chronic (three
weekly intraperitoneal injections of acrolein at 6, 12, or 24 mmol/
ER Stress kg) systemic acrolein exposure in rats caused ER stress and
The ER is the site of synthesis and folding of proteins, and the upregulation of ATF4, CHOP and GADD34, and VEGF, leading to
accumulation of misfolded proteins can trigger ER stress and emphysematous lung tissue remodeling and airspace enlarge-
the unfolded protein response (UPR). ER stress is thought to con- ment; caspase-3 activation and apoptosis were concurrently
tribute to several pathological conditions that may also be asso- observed in the septal cells in acrolein-treated rat lungs.
ciated with increased acrolein, such as fatty liver disease Moreover, accumulated acrolein-protein adducts were observed
(Vacaru et al., 2014), Alzheimer’s disease (Li et al., 2014), and in the lungs of smokers with COPD, indicating that acrolein may
inflammatory and cardiovascular diseases (Zhou and Tabas, contribute to lung disorders like emphysema and COPD
2013). Based on its proclivity to form protein adducts, acrolein is (Kitaguchi et al., 2012). Recent reports have demonstrated ER
highly likely to trigger ER stress. Indeed, several recent studies stress associated with cigarette smoke and the development of
demonstrate acrolein-induced ER stress and UPR. The effects of lung disorders such as emphysema (Gan et al., 2011) and COPD
acrolein (2–25 mM) on ER stress and endothelial cell activation (Ribeiro and O’Neal, 2012). Thus, acrolein-induced ER stress may
were studied in human umbilical vein endothelial cells be a relevant mechanism of toxicity in various disorders.
(Haberzettl et al., 2009), wherein both the alarm and the adapta-
tion pathways of UPR were triggered, and inflammatory genes Structural and Membrane Effects
TNF-a, IL-6, and IL-8 were upregulated, demonstrating the Myelin is a critical structural component of the nervous system
proinflammatory and atherogenic properties of acrolein. composed of lipids and proteins including myelin basic protein
Notably, the acrolein effects were prevented by the chemical (MBP), and demyelination is a hallmark of neurodegenerative
chaperone (phenylbutyric acid), suggesting its usefulness in diseases, including multiple sclerosis (MS) and SCI. Increased
treating pathologies associated with acrolein toxicity. The same acrolein levels are seen in the spinal cord in MS and SCI animal
group also demonstrated that the elimination of LPO products models, with acrolein levels correlating to neuronal membrane
(eg acrolein) by aldose reductase diminishes ER stress and damage, loss of conduction, and even degeneration (Leung et al.,
decreases ischemia-reperfusion injury (Keith et al., 2009). Our 2011; Shi et al., 2011). The acrolein scavenger hydralazine less-
recent work demonstrates that acrolein-induced cytotoxicity in ened myelin damage and improved behavioral outcomes
hepatocytes involves ER stress, in combination with mitochon- (Leunget al., 2011); reversed SCI-induced damage by alleviating
drial disruption and oxidative stress, resulting in cell death superoxide production, preventing GSH depletion (Hamann
(Mohammad et al., 2012) Acrolein-induced cell death was only et al., 2008); and effectively reducing acrolein levels, tissue dam-
partially attenuated by NAC, phenyl-butyric acid, and caspase age, motor deficits, and neuropathic pain (Park et al., 2014).
or JNK inhibitors individually, indicating that combination Acrolein exposure in vitro also elicited a concentration-
therapies may be more effective against acrolein-induced dependent modification and aggregation of neurofilament-L
toxicity. Interestingly, acrolein caused ER stress but failed to (NF-L) which may contribute to neurodegeneration. The acro-
upregulate the protective ER-chaperones, GRP78 and GRP94. lein concentrations used (100–500 mM) correspond to concentra-
Acrolein is demonstrated to modify and reduce function of tions of 1.0–5.2 nmol/mg protein, comparable to levels observed
another chaperone protein disulfide isomerase; this may lead to in Alzheimer’s disease, and thus are within the expected range
the enhancement and perpetuation of ER stress (Carbone et al., of SCI and MS. These effects of acrolein were blocked by NAC
2005). Activation of protective versus injurious ER responses to and GSH (Jeong and Kang, 2008).
acrolein exposure may be dependent on dose and time, as Acrolein exposures as low as 1 lM for 4 h resulted in
shown in A549 lung cells (Tanel et al., 2014). increased permeability of cell membranes to ethidium bromide
In vitro acrolein exposure (<10 mM) of Swiss 3T3 cells in ex vivo spinal cords; as the dose and duration increased,
was demonstrated to be primarily responsible for cigarette larger molecules such as horseradish peroxidase and LDH were
MOGHE ET AL. | 249

able to pass through (Luo and Shi, 2004). Exposure to acrolein lung fibroblasts exposed to subcytotoxic concentrations of
in vivo and in vitro was associated with reduced presynaptic neu- acrolein (10–100 mM) resulted in the production of a potent
rotransmitter release; this effect involved inhibition of key - proangiogenic factor, vascular endothelial growth factor (VEGF);
proteins that regulate membrane-vesicle fusion, such as the effects were prevented by p38 inhibitor and NAC (Volpi et al.,
N-ethylmaleimide-sensitive fusion protein (NSF) and synapto- 2011). Activation and membrane translocation of another pro-
somal-associated protein of 25 kDa (SNAP-25). Acrolein, result- tein kinase, PKC-d, by allyl alcohol and its primary metabolite
ing from acrylamide, also inhibited presynaptic membrane acrolein (up to 125 mM), resulted in cytotoxicity in hepatocytes,
neurotransmitter uptake and vesicular storage in vivo and which was blocked by rottlerin, a selective PKC-d inhibitor
in vitro (LoPachin et al., 2004, 2006); proteomic analyses showed (Maddox et al., 2003). Our work showed that acrolein exposure of
that these dysfunctions were associated with the formation of cultured hepatocytes led to a sustained activation of ERK1/2,
acrolein adducts with the dopamine transporter and v-ATPase JNK, and p38, which was associated with ER and mitochondrial
(Lopachin et al., 2007). stress and apoptosis; the cytotoxic effects of acrolein were
Acrolein exposure resulted in several structural changes decreased by JNK inhibitor, suggesting that kinase activation
including induced F-actin microfilament aggregation, and mar- may be linked to cell death and liver injury (Mohammad et al.,

Downloaded from https://academic.oup.com/toxsci/article/143/2/242/1676672 by guest on 10 November 2022


ginalization and regionalization in Sertoli cells, which are 2012). Via p38 activation, acrolein also caused upregulation of
important in spermatogenesis; such effects may underlie the the muscle-specific E3 ligases atrogin-1 and MuRF1, suggesting
testicular toxicity of the anticancer drug cyclophosphamide a possible role in degradation of myosin heavy chain, myotube
which is metabolized into acrolein (Liu et al., 2012). Another catabolism, and muscle wasting (Rom et al., 2013). In contrast,
membrane effect of acrolein was the stimulation of erythrocyte acrolein was shown to suppress phosphorylation of JNK and
cell membrane scrambling, a typical feature of suicidal death or activation of c-Jun; moreover, JNK was a direct target for alkyla-
eryptosis. Membrane phospholipid scrambling was accompa- tion by acrolein at key cysteine residues in the active site
nied by the generation of ceramide, cell shrinkage, and (Hristova et al., 2012).
apoptosis (Ahmed et al., 2013). Airborne acrolein was shown to Acrolein-induced deregulated signaling is known to induce
induce hyperphosphorylation and damage of keratin-8 and mucin production and inflammation (major contributors to
ubiquitination of intermediate filament in bronchiolar lung cell lung disorders) via many upstream signals, including (1) MAPKs
monolayers (Burcham et al., 2014). Numerous studies have and epidermal growth factor receptor (Deshmukh et al., 2005);
shown in vitro and in vivo toxicity of acrolein associated with (2) matrix metalloproteinase-9 and the Ras/extracellular signal-
structural (and functional) effects in the cardiovascular system, regulated kinase pathway, which was attenuated by simvasta-
including increased susceptibility to vasospasm (Conklin et al., tin (Chen et al., 2010); (3) Peroxisome proliferator-activated
2006), increased heart rate and blood pressure (Perez et al., receptor-gamma (PPAR-gamma), which was reduced by rosigli-
2013), increased atherosclerotic lesions (Srivastava et al., 2011), tazone (Liu et al., 2009); and (4) Phosphodiesterase 5 that selec-
and decreased endothelial cell migration (O’Toole et al., 2014); tively degrades cyclic guanosine 30 ,50 -monophosphate (cGMP),
however, the exact toxic mechanisms remain unclear. which was prevented by sildenafil (Wang et al., 2009). Acrolein
has also been implicated in the demise of retinal pigment epi-
Deregulated Signal Transduction thelium (ARPE-19) cells, with overproduction of the proangio-
Protein kinases and phosphatases are critical components of genic cytokines, TGFb2 and VEGF that are important in macular
intracellular signaling that regulate cellular function, and acro- degeneration and in diabetic retinopathy. These effects were
lein is reported to inactivate as well as activate them in a con- potentiated by high glucose and were attenuated by NBHA
textual manner, depending on dose and cell type. Acrolein, at a (N-benzylhydroxylamine, a hydroxylamine that sequesters
fairly high concentrations (100–1000 mM), was shown to be a aldehydes) and by SIS3, a specific inhibitor of SMAD3 that blocks
potent, time-dependent inactivator of protein tyrosine phos- TGFb2 signaling (Grigsby et al., 2012; Vidro-Kotchan et al., 2011).
phatase-1B via covalent modification of an active site cysteine Recently, a novel mechanism of acrolein cytotoxicity involving
(Seiner et al., 2007); and phosphatase PP2A was also inhibited by the acrolein-conjugation and inactivation of glyceraldehyde-
acrolein, with concurrent apoptosis (Fathi et al., 2000). On the 3-phosphate dehydrogenase, an enzyme with many diverse
other hand, a lower acrolein exposure (25 mM) in human hepato- functions, was described in vitro (Martyniuk et al., 2011) and in
cytes led to a modest  2-fold increase in both serine and tyro- mouse mammary carcinoma FM3A cells (Nakamura et al., 2013).
sine phosphatase activity and reduced interferon-a antiviral Thus, acrolein-induced changes in cellular signaling and gene
activity, which may contribute to therapy resistance in hepatitis expression may alter function and physiology, and potentially
C virus infection (Joshi-Barve et al., 2009). Acrolein exposure contribute to the disease process in multiple organ systems.
(10–30 mM) in breast cancer cells altered the thioredoxin-
dependent redox system and caused a G2-M cell cycle arrest,
but did not inhibit phosphatase-CDC25 which controls this cell
CONCLUSIONS AND FUTURE DIRECTON
cycle checkpoint (Beillerot et al., 2012). In this review, we focused on the myriad effects mediated by
Several in vitro studies described below have demonstrated acrolein with an emphasis on the molecular mechanisms of
acrolein-induced alterations in cellular mitogen activated pro- toxicity that provide evidence to suggest that acrolein plays a
tein kinases (MAPKs) which are likely to contribute to other critical role in disease pathogenesis (Figure 4). Acrolein operates
mechanisms of toxicity; however, few findings are validated via disruption of cell organelles such as mitochondria, ER, and
in vivo. Acrolein-induced MAPK activation and cytotoxicity were membranes. In addition, it increases oxidative stress and apop-
observed in cultured neurons along with the activation of c-jun tosis, alters proinflammatory mediators, and changes cellular
cAMP response element binding protein, CREB (Pugazhenthi signal transduction leading to a variety of defects that
et al., 2006), and in cultured rat vascular smooth muscle cells; may cause or predispose to disease in various organs.
however, in the latter study, MAPK inhibitors failed to block cell Moreover, acrolein may have cooperative effects with other
death, suggesting divergent mechanisms (Ranganna et al., 2002). toxic insults. Many of the studies described here were done
Activation of p38 in human airway smooth muscle cells and in vitro or in cultured cells, rather than in vivo; hence, despite
250 | TOXICOLOGICAL SCIENCES, 2015, Vol. 143, No. 2

MECHANISMS OF TOXICITY AFFECTED ORGANS

DNA
Adduction Brain

Mitochondrial Eye
Dysfunction

Skin
Inflammation
Lung

Oxidative Heart
ACROLEIN Stress
Liver

Downloaded from https://academic.oup.com/toxsci/article/143/2/242/1676672 by guest on 10 November 2022


Protein Kidney
Adduction

Gut

Membrane Reproductive
Disruption system
Bladder

Endoplasmic
Reticulum Stress

FIG. 4. Toxicity mechanisms and organ systems affected by acrolein (images courtesy Google).

strong links to several diseases, the causal role of acrolein in Institutes of Health (NIH); and U.S. Department of Veterans
disease pathogenesis, though highly likely, can only be sur- Affairs. Grant numbers are: R01AA018869-02 (CJM),
mised. A limitation of this review stems from the fact that levels T35ES014559-06 (CJM), P01AA017103-01 (CJM), K01 ES017105-
of acrolein are difficult to quantify, particularly in vivo, while the 01A1 (S.J.-B.) and (BX000350 (CJM).
in vitro literature describes acrolein concentrations in various
units that are problematic to interconvert and challenging to
ACKNOWLEDGMENTS
compare.
Several unanswered questions remain regarding acrolein The authors would like to acknowledge Ms Marion McClain
toxicology. Detailed studies are necessary to determine not only for proof-reading the article. Nothing to disclose.
the temporal sequence of molecular events, but also the inter-
dependence, cross-talk, and the relative contributions of differ-
ent mechanisms of acrolein toxicity to the pathogenesis/
REFERENCES
progression of disease. The development of small and large Abraham, K., Andres, S., Palavinskas, R., Berg, K., Appel, K. E.,
mammalian models of exogenous and endogenous acrolein and Lampen, A. (2011). Toxicology and risk assessment of
exposures, both acute and chronic, will promote detailed mecha- acrolein in food. Mol. Nutr. Food Res. 55, 1277–1290.
nistic investigations and toxicity studies. Research, particularly in Adams, J. D., Jr. and Klaidman, L. K. (1993). Acrolein-induced oxy-
humans, has been limited by availability of techniques to quan- gen radical formation. Free Radic. Biol. Med. 15, 187–193.
tify acrolein and acrolein-adducts in body fluids and tissues. Ahmed, M. S., Langer, H., Abed, M., Voelkl, J., and Lang, F. (2013).
Hence, the development of sensitive, high-throughput and quan- The uremic toxin acrolein promotes suicidal erythrocyte
titative techniques will facilitate large population-based human death. Kidney Blood Press. Res. 37, 158–167.
studies. Acrolein scavengers with proven effectiveness in small Alarcon, R. A. (2012). Anticancer system created by acrolein and
in vitro/in vivo studies may provide effective and safe means to hydroxyl radical generated in enzymatic oxidation of sper-
mitigate acrolein toxicity and associated disorders; however, their mine and other biochemical reactions. Med. Hypotheses 79,
bioavailability, effective concentrations, and delivery to the
522–530.
affected areas would require extensive investigations. A true
Aldini, G., Orioli, M., and Carini, M. (2011). Protein modification
determination of causality and the contribution of acrolein need
by acrolein: relevance to pathological conditions and inhibi-
to be established by detailed in vivo studies in animal models and
tion by aldehyde sequestering agents. Mol. Nutr. Food Res. 55,
in human populations. Moreover, it will be important to examine
1301–1319.
the impact of age, sex, lifestyle, and genetic variation on the
Avezov, K., Reznick, A. Z., and Aizenbud, D. (2014a). LDH enzyme
susceptibility to acrolein exposures in correlation with disease
activity in human saliva: the effect of exposure to cigarette
states.
smoke and its different components. Arch. Oral Biol. 59,
142–148.
Avezov, K., Reznick, A. Z., and Aizenbud, D. (2014b). Oxidative
FUNDING
damage in keratinocytes exposed to cigarette smoke and
This work was supported by National Institute on Alcohol aldehydes. Toxicol. in vitro 28, 485–491.
Abuse and Alcoholism (NIAAA) and National Institute of Beillerot, A., Battaglia, E., Aline, B., and Bagrel, D. (2012).
Environmental Health Sciences (NIEHS) at the National Protection of CDC25 phosphatases against oxidative stress in
MOGHE ET AL. | 251

breast cancer cells: evaluation of the implication of the thio- Cox, R., Goorha, S., and Irving, C. C. (1988). Inhibition of DNA
redoxin system. Free Radic. Res. 46, 674–689. methylase activity by acrolein. Carcinogenesis 9, 463–465.
Birben, E., Sahiner, U. M., Sackesen, C., Erzurum, S., and Kalayci, Demir, E., Turna, F., Kaya, B., Creus, A., and Marcos, R. (2013).
O. (2012). Oxidative stress and antioxidant defense. World Mutagenic/recombinogenic effects of four lipid peroxi-
Allergy Organ. J. 5, 9–19. dation products in Drosophila. Food Chem. Toxicol. 53,
Brooks, W. H. (2013). Increased polyamines alter chromatin and 221–227.
stabilize autoantigens in autoimmune diseases. Front. Deshmukh, H. S., Case, L. M., Wesselkamper, S. C., Borchers, M.
Immunol. 4, 91. T., Martin, L. D., Shertzer, H. G., Nadel, J. A., and Leikauf, G. D.
Bui, L. C., Manaa, A., Xu, X., Duval, R., Busi, F., Dupret, J. M., (2005). Metalloproteinases mediate mucin 5AC expression by
Rodrigues Lima, F., and Dairou, J. (2013). Acrolein, an alpha, epidermal growth factor receptor activation. Am. J. Respir.
beta-unsaturated aldehyde, irreversibly inhibits the acetyla- Crit. Care Med. 171, 305–314.
tion of aromatic amine xenobiotics by human Nat1. Drug Dong, L., Zhou, S., Yang, X., Chen, Q., He, Y., and Huang, W.
Metab. Dispos. 41, 1300-1305. (2013). Magnolol protects against oxidative stress-

Downloaded from https://academic.oup.com/toxsci/article/143/2/242/1676672 by guest on 10 November 2022


Burcham, P. C., Raso, A., and Henry, P. J. (2014). Airborne acrolein mediated neural cell damage by modulating mitochondrial
induces keratin-8 (Ser-73) hyperphosphorylation and inter- dysfunction and PI3K/Akt signaling. J. Mol. Neurosci. 50,
mediate filament ubiquitination in bronchiolar lung cell 469–481.
monolayers. Toxicology 319, 44–52. Dorman, D. C., Struve, M. F., Wong, B. A., Marshall, M. W., Gross,
Burcham, P. C., Raso, A., and Kaminskas, L. M. (2012). Chaperone E. A., and Willson, G. A. (2008). Respiratory tract responses in
heat shock protein 90 mobilization and hydralazine cytopro- male rats following subchronic acrolein inhalation. Inhal.
tection against acrolein-induced carbonyl stress. Mol. Toxicol. 20, 205–216.
Pharmacol. 82, 876–886. Due, M. R., Park, J., Zheng, L., Walls, M., Allette, Y. M., White, F.
Cahill, T. M. (2014). Ambient acrolein concentrations in coastal, A., and Shi, R. (2014). Acrolein involvement in sensory and
remote, and urban regions in California. Environ. Sci. Technol. behavioral hypersensitivity following spinal cord injury in
48, 8507–8513. the rat. J. Neurochem. 128, 776–786.
Carbone, D. L., Doorn, J. A., Kiebler, Z., and Petersen, D. R. (2005). Fathi, A. R., Krautheim, A., Kaap, S., Eger, K., and Steinfelder, H. J.
Cysteine modification by lipid peroxidation products in- (2000). Michael adducts of ascorbic acid as inhibitors of pro-
hibits protein disulfide isomerase. Chem. Res. Toxicol. 18, tein phosphatase 2A and inducers of apoptosis. Bioorg. Med.
1324–1331. Chem. Lett. 10, 1605–1608.
Centers for Disease, C., and Prevention (2013). Notes from the Feng, Z., Hu, W., Hu, Y., and Tang, M. S. (2006). Acrolein is a major
field: acute pesticide-related illness resulting from occupa- cigarette-related lung cancer agent: Preferential binding at
tional exposure to acrolein - washington and california, p53 mutational hotspots and inhibition of DNA repair. Proc.
1993-2009. MMWR 62, 313–314. Natl. Acad. Sci. U.S.A. 103, 15404–15409.
Chen, Y. J., Chen, P., Wang, H. X., Wang, T., Chen, L., Wang, X., Feng, Z., Liu, Z., Li, X., Jia, H., Sun, L., Tian, C., Jia, L., and Liu, J.
Sun, B. B., Liu, D. S., Xu, D., An, J., and Wen, F. Q. (2010). (2010). Alpha-tocopherol is an effective Phase II enzyme in-
Simvastatin attenuates acrolein-induced mucin production ducer: protective effects on acrolein-induced oxidative stress
in rats: involvement of the Ras/extracellular signal-regulated and mitochondrial dysfunction in human retinal pigment
kinase pathway. Int. Immunopharmacol. 10, 685–693. epithelial cells. J. Nutr. Biochem. 21, 1222–1231.
Chen, D., Fang, L., Li, H., Tang, M. S., and Jin, C. (2013). Cigarette Finkelstein, E. I., Nardini, M., and Van der Vliet, A. (2001).
smoke component acrolein modulates chromatin assem- Inhibition of neutrophil apoptosis by acrolein: a mechanism
bly by inhibiting histone acetylation. J. Biol. Chem. 288, of tobacco-related lung disease?. Am. J. Physiol. Lung C 281,
21678–21687. L732–L739.
Choudhury, S., Dyba, M., Pan, J., Roy, R., and Chung, F. L. (2013). Galligan, J. J., Smathers, R. L., Fritz, K. S., Epperson, L. E., Hunter,
Repair kinetics of acrolein- and (E)-4-hydroxy-2-nonenal- L. E., and Petersen, D. R. (2012). Protein carbonylation in a
derived DNA adducts in human colon cell extracts. Mutat. murine model for early alcoholic liver disease. Chem. Res.
Res. 751–752, 15–23. Toxicol. 25, 1012–1021.
Cohen, S. M., Garland, E. M., St John, M., Okamura, T., and Smith, Gan, G., Hu, R., Dai, A., Tan, S., Ouyang, Q., Fu, D., and Jiang, D.
R. A. (1992). Acrolein initiates rat urinary bladder carcinogen- (2011). The role of endoplasmic reticulum stress in emphy-
esis. Cancer Res. 52, 3577–3581. sema results from cigarette smoke exposure. Cell. Physiol.
Colombo, G., Dalle-Donne, I., Orioli, M., Giustarini, D., Rossi, R., Biochem. 28, 725–732.
Clerici, M., Regazzoni, L., Aldini, G., Milzani, A., Butterfield, D. Grigsby, J., Betts, B., Vidro-Kotchan, E., Culbert, R., and Tsin, A.
A., and Gagliano, N. (2012). Oxidative damage in human gin- (2012). A possible role of acrolein in diabetic retinopathy: in-
gival fibroblasts exposed to cigarette smoke. Free Radic. Biol. volvement of a VEGF/TGFbeta signaling pathway of the reti-
Med. 52, 1584–1596. nal pigment epithelium in hyperglycemia. Curr. Eye Res. 37,
Comer, D. M., Elborn, J. S., and Ennis, M. (2014). Inflammatory 1045–1053.
and cytotoxic effects of acrolein, nicotine, acetylaldehyde Haberzettl, P., Vladykovskaya, E., Srivastava, S., and Bhatnagar,
and cigarette smoke extract on human nasal epithelial cells. A. (2009). Role of endoplasmic reticulum stress in acrolein-
BMC Pulm. Med. 14, 32. induced endothelial activation. Toxicol. Appl. Pharmacol. 234,
Conklin, D. J., Bhatnagar, A., Cowley, H. R., Johnson, G. H., 14–24.
Wiechmann, R. J., Sayre, L. M., Trent, M. B., and Boor, P. J. Hamann, K., Nehrt, G., Ouyang, H., Duerstock, B., and Shi, R.
(2006). Acrolein generation stimulates hypercontraction in (2008). Hydralazine inhibits compression and acrolein-
isolated human blood vessels. Toxicol. Appl. Pharmacol. 217, mediated injuries in ex vivo spinal cord. J. Neurochem. 104,
277–288. 708–718.
252 | TOXICOLOGICAL SCIENCES, 2015, Vol. 143, No. 2

Hamann, K. and Shi, R. (2009). Acrolein scavenging: a potential adducts by human mitochondrial DNA polymerase gamma.
novel mechanism of attenuating oxidative stress following J. Biol. Chem. 288, 14247–14255.
spinal cord injury. J. Neurochem. 111, 1348–1356. Kassem, N. O., Daffa, R. M., Liles, S., Jackson, S. R., Kassem, N. O.,
He, X., Song, W., Liu, C., Chen, S., and Hua, J. (2014). Rapamycin Younis, M. A., Mehta, S., Chen, M., Jacob, P., 3rd, Carmella, S.
inhibits acrolein-induced apoptosis by alleviating ROS- G., et al. (2014). Children’s exposure to secondhand and third-
driven mitochondrial dysfunction in male germ cells. Cell hand smoke carcinogens and toxicants in homes of hookah
Prolif. 47, 161–171. smokers. Nicotine Tobacco Res. 16, 961–975.
Hengstermann, A. and Muller, T. (2008). Endoplasmic reticulum Kawai, Y., Furuhata, A., Toyokuni, S., Aratani, Y., and Uchida, K.
stress induced by aqueous extracts of cigarette smoke in 3T3 (2003). Formation of acrolein-derived 2’-deoxyadenosine
cells activates the unfolded-protein-response-dependent adduct in an iron-induced carcinogenesis model. J. Biol.
PERK pathway of cell survival. Free Radic. Biol. Med. 44, Chem. 278, 50346–50354.
1097–1107. Kehrer, J. P., and Biswal, S. S. (2000). The molecular effects of
Hochman, D. J., Collaco, C. R., and Brooks, E. G. (2014). Acrolein acrolein. Toxicol. Sci. 57, 6–15.

Downloaded from https://academic.oup.com/toxsci/article/143/2/242/1676672 by guest on 10 November 2022


induction of oxidative stress and degranulation in mast cells. Keith, R. J., Haberzettl, P., Vladykovskaya, E., Hill, B. G.,
Environ. Toxicol. 29, 908–915. Kaiserova, K., Srivastava, S., Barski, O., and Bhatnagar, A.
Horton, N. D., Biswal, S. S., Corrigan, L. L., Bratta, J., and Kehrer, J. (2009). Aldose reductase decreases endoplasmic reticulum
P. (1999). Acrolein causes inhibitor kappaB-independent de- stress in ischemic hearts. Chem. Biol. Int. 178, 242–249.
creases in nuclear factor kappaB activation in human lung Kern, J. C. and Kehrer, J. P. (2002). Acrolein-induced cell death: a
adenocarcinoma (A549) cells. J. Biol. Chem. 274, 9200–9206. caspase-influenced decision between apoptosis and oncosis/
Hristova, M., Heuvelmans, S., and van der Vliet, A. (2007). GSH- necrosis. Chem. Biol. Int. 139, 79–95.
dependent regulation of Fas-mediated caspase-8 activation Kim, S. H., Lee, I. C., Baek, H. S., Shin, I. S., Moon, C., Bae, C. S.,
by acrolein. Febs Lett. 581, 361–367. Kim, S. H., Kim, J. C., and Kim, H. C. (2014). Mechanism for the
Hristova, M., Spiess, P. C., Kasahara, D. I., Randall, M. J., Deng, B., protective effect of diallyl disulfide against cyclophos-
and van der Vliet, A. (2012). The tobacco smoke component, phamide acute urotoxicity in rats. Food Chem. Toxicol. 64,
acrolein, suppresses innate macrophage responses by direct 110–118.
alkylation of c-Jun N-terminal kinase. Am. J. Respir. Cell Mol. Kim, G. D., Lee, S. E., Kim, T. H., Jin, Y. H., Park, Y. S., and Park, C.
Biol. 46, 23–33. S. (2012). Melatonin suppresses acrolein-induced IL-8 pro-
Huang, Y., Qin, J., Chen, M., Chao, X., Chen, Z., Ramassamy, C., Pi, duction in human pulmonary fibroblasts. J. Pineal Res. 52,
R., and Jin, M. (2014). Lithium prevents acrolein-induced neu- 356–364.
rotoxicity in HT22 mouse hippocampal cells. Neurochem. Res. Kim, C. E., Lee, S. J., Seo, K. W., Park, H. M., Yun, J. W., Bae, J. U., Bae,
39, 677–684. S. S., and Kim, C. D. (2010). Acrolein increases 5-lipoxygenase
Igarashi, K. and Kashiwagi, K. (2011). Protein-conjugated acrolein expression in murine macrophages through activation of ERK
as a biochemical marker of brain infarction. Mol. Nutr. Food pathway. Toxicol. Appl. Pharmacol. 245, 76–82.
Res. 55, 1332–1341. Kim, S. I., Pfeifer, G. P., and Besaratinia, A. (2007). Lack of mutage-
Il’yasova, D., Scarbrough, P., and Spasojevic, I. (2012). Urinary nicity of acrolein-induced DNA adducts in mouse and
biomarkers of oxidative status. Clin. Chim. Acta 413, human cells. Cancer Res. 67, 11640–11647.
1446–1453. Kirkham, P. A., Spooner, G., Rahman, I., and Rossi, A. G. (2004).
Ismahil, M. A., Hamid, T., Haberzettl, P., Gu, Y., Chandrasekar, B., Macrophage phagocytosis of apoptotic neutrophils is com-
Srivastava, S., Bhatnagar, A., and Prabhu, S. D. (2011). Chronic promised by matrix proteins modified by cigarette smoke
oral exposure to the aldehyde pollutant acrolein induces di- and lipid peroxidation products. Biochem. Biophys. Res.
lated cardiomyopathy. Am. J. Physiol. Heart Circ. Physiol. 301, Commun. 318, 32–37.
H2050–H2060. Kitaguchi, Y., Taraseviciene-Stewart, L., Hanaoka, M., Natarajan,
Jeong, M. S. and Kang, J. H. (2008). Acrolein, the toxic endogenous R., Kraskauskas, D., and Voelkel, N. F. (2012). Acrolein indu-
aldehyde, induces neurofilament-L aggregation. BMB Rep. 41, ces endoplasmic reticulum stress and causes airspace
635–639. enlargement. PloS One 7, e38038.
Jia, L., Liu, Z., Sun, L., Miller, S. S., Ames, B. N., Cotman, C. W., and Korkmaz, A., Topal, T., and Oter, S. (2007). Pathophysiological
Liu, J. (2007). Acrolein, a toxicant in cigarette smoke, causes aspects of cyclophosphamide and ifosfamide induced hem-
oxidative damage and mitochondrial dysfunction in RPE orrhagic cystitis; implication of reactive oxygen and nitrogen
cells: protection by (R)-alpha-lipoic acid. Invest. Ophthalmol. species as well as PARP activation. Cell Biol. Toxicol. 23,
Vis. Sci. 48, 339–348. 303–312.
Joshi-Barve, S., Amancherla, K., Patil, M., Bhatnagar, A., Lambert, C., Li, J., Jonscher, K., Yang, T. C., Reigan, P., Quintana,
Mathews, S., Gobejishvili, L., Cave, M., McClain, C., and Barve, M., Harvey, J., and Freed, B. M. (2007). Acrolein inhibits cyto-
S. (2009). Acrolein, a ubiquitous pollutant and lipid hydroper- kine gene expression by alkylating cysteine and arginine
oxide product, inhibits antiviral activity of interferon-alpha: residues in the NF-kappaB1 DNA binding domain. J. Biol.
relevance to hepatitis C. Free Radic. Biol. Med. 47, 47–54. Chem. 282, 19666–19675.
Kasahara, D. I., Poynter, M. E., Othman, Z., Hemenway, D., and Lambert, C., McCue, J., Portas, M., Ouyang, Y., Li, J., Rosano, T. G.,
van der Vliet, A. (2008). Acrolein inhalation suppresses lipo- Lazis, A., and Freed, B. M. (2005). Acrolein in cigarette smoke
polysaccharide-induced inflammatory cytokine production inhibits T-cell responses. J. Allergy Clin. Immunol. 116,
but does not affect acute airways neutrophilia. J. Immunol. 916–922.
181, 736–745. Lee, H. W., Wang, H. T., Weng, M. W., Hu, Y., Chen, W. S., Chou,
Kasiviswanathan, R., Minko, I. G., Lloyd, R. S., and Copeland, W. D., Liu, Y., Donin, N., Huang, W. C., Lepor, H., et al. (2014).
C. (2013). Translesion synthesis past acrolein-derived DNA Acrolein- and 4-aminobiphenyl-DNA adducts in human
MOGHE ET AL. | 253

bladder mucosa and tumor tissue and their mutagenicity in Moretto, N., Bertolini, S., Iadicicco, C., Marchini, G., Kaur, M.,
human urothelial cells. Oncotarget 5, 3526–3540. Volpi, G., Patacchini, R., Singh, D., and Facchinetti, F. (2012).
Leung, G., Sun, W., Zheng, L., Brookes, S., Tully, M., and Shi, R. Cigarette smoke and its component acrolein augment IL-8/
(2011). Anti-acrolein treatment improves behavioral out- CXCL8 mRNA stability via p38 MAPK/MK2 signaling in hu-
come and alleviates myelin damage in experimental man pulmonary cells. Am. J. Physiol. Lung Cell. Mol. Physiol.
autoimmune encephalomyelitis mouse. Neuroscience 173, 303, L929–L938.
150–155. Moretto, N., Facchinetti, F., Southworth, T., Civelli, M., Singh, D.,
Li, J. Q., Yu, J. T., Jiang, T., and Tan, L. (2014). Endoplasmic reticu- and Patacchini, R. (2009). Alpha, beta-unsaturated aldehydes
lum dysfunction in Alzheimer’s Disease. Mol. Neurobiol. contained in cigarette smoke elicit IL-8 release in pulmonary
doi:10.1007/s12035-014-8695-8. cells through mitogen-activated protein kinases. Am. J.
Liu, D. S., Liu, W. J., Chen, L., Ou, X. M., Wang, T., Feng, Y. L., Physiol. Lung Cell. Mol. Physiol. 296, L839–L848.
Zhang, S. F., Xu, D., Chen, Y. J., and Wen, F. Q. (2009). Myers, C. R., Myers, J. M., Kufahl, T. D., Forbes, R., and
Rosiglitazone, a peroxisome proliferator-activated receptor- Szadkowski, A. (2011). The effects of acrolein on the thiore-

Downloaded from https://academic.oup.com/toxsci/article/143/2/242/1676672 by guest on 10 November 2022


gamma agonist, attenuates acrolein-induced airway mucus doxin system: implications for redox-sensitive signaling.
hypersecretion in rats. Toxicology 260, 112–119. Mol. Nutr. Food Res. 55, 1361–1374.
Liu, F., Li, X. L., Lin, T., He, D. W., Wei, G. H., Liu, J. H., and Li, L. S. Nakamura, M., Tomitori, H., Suzuki, T., Sakamoto, A., Terui, Y.,
(2012). The cyclophosphamide metabolite, acrolein, induces Saiki, R., Dohmae, N., Igarashi, K., and Kashiwagi, K. (2013).
cytoskeletal changes and oxidative stress in Sertoli cells. Mol. Inactivation of GAPDH as one mechanism of acrolein toxic-
Biol. Rep. 39, 493–500. ity. Biochem. Biophys. Res. Commun. 430, 1265–1271.
Liu, X. Y., Zhu, M. X., and Xie, J. P. (2010). Mutagenicity of acrolein O’Toole, T. E., Abplanalp, W., Li, X., Cooper, N., Conklin, D. J.,
and acrolein-induced DNA adducts. Toxicol. Mech. Methods 20, Haberzettl, P., and Bhatnagar, A. (2014). Acrolein decreases
36–44. endothelial cell migration and insulin sensitivity through
Logue, J. M., Price, P. N., Sherman, M. H., and Singer, B. C. induction of Let-7a. Toxicol. Sci. 140, 271–282.
(2012). A method to estimate the chronic health impact of O’Toole, T. E., Zheng, Y. T., Hellmann, J., Conklin, D. J., Barski, O.,
air pollutants in U.S. residences. Environ. Health Persp. 120, and Bhatnagar, A. (2009). Acrolein activates matrix metallo-
216–222. proteinases by increasing reactive oxygen species in macro-
Lopachin, R. M., Barber, D. S., Geohagen, B. C., Gavin, T., He, D., phages. Toxicol. Appl. Pharmacol. 236, 194–201.
and Das, S. (2007). Structure-toxicity analysis of type-2 Ong, F. H., Henry, P. J., and Burcham, P. C. (2012). Prior exposure
alkenes: in vitro neurotoxicity. Toxicol. Sci. 95, 136–146. to acrolein accelerates pulmonary inflammation in influenza
Luo, J. and Shi, R. (2004). Acrolein induces axolemmal disruption, A-infected mice. Toxicol. Lett. 212, 241–251.
oxidative stress, and mitochondrial impairment in spinal Park, J., Zheng, L., Marquis, A., Walls, M., Duerstock, B., Pond, A.,
cord tissue. Neurochem. Int. 44, 475–486. Vega-Alvarez, S., Wang, H., Ouyang, Z., and Shi, R. (2014).
MacAllister, S. L., Martin-Brisac, N., Lau, V., Yang, K., and Neuroprotective role of hydralazine in rat spinal cord injury-
O’Brien, P. J. (2013). Acrolein and chloroacetaldehyde: an ex- attenuation of acrolein-mediated damage. J. Neurochem. 129,
amination of the cell and cell-free biomarkers of toxicity. 339–349.
Chem. Biol. Int. 202, 259–266. Pawlowicz, A. J. and Kronberg, L. (2008). Characterization of
Maczurek, A., Hager, K., Kenklies, M., Sharman, M., Martins, R., adducts formed in reactions of acrolein with thymidine and
Engel, J., Carlson, D. A., and Munch, G. (2008). Lipoic acid as calf thymus DNA. Chem. Biodivers. 5, 177–188.
an anti-inflammatory and neuroprotective treatment for Pawlowicz, A. J., Munter, T., Klika, K. D., and Kronberg, L. (2006a).
Alzheimer’s disease. Adv. Drug Deliv. Rev. 60, 1463–1470. Reaction of acrolein with 2’-deoxyadenosine and 9-
Maddox, J. F., Roth, R. A., and Ganey, P. E. (2003). Allyl alcohol ac- ethyladenine–formation of cyclic adducts. Bioorg. Chem. 34, 39–48.
tivation of protein kinase C delta leads to cytotoxicity of rat Pawlowicz, A. J., Munter, T., Zhao, Y., and Kronberg, L. (2006b).
hepatocytes. Chem. Res. Toxicol. 16, 609–615. Formation of acrolein adducts with 2’-deoxyadenosine in
Martyniuk, C. J., Fang, B., Koomen, J. M., Gavin, T., Zhang, L., calf thymus DNA. Chem. Res. Toxicol. 19, 571–576.
Barber, D. S., and Lopachin, R. M. (2011). Molecular mecha- Perez, C. M., Ledbetter, A. D., Hazari, M. S., Haykal-Coates, N.,
nism of glyceraldehyde-3-phosphate dehydrogenase inacti- Carll, A. P., Winsett, D. W., Costa, D. L., and Farraj, A. K.
vation by alpha, beta-unsaturated carbonyl derivatives. (2013). Hypoxia stress test reveals exaggerated cardiovascu-
Chem. Res. Toxicol. 24, 2302–2311. lar effects in hypertensive rats after exposure to the air
Medina-Navarro, R., Nieto-Aguilar, R., and Alvares-Aguilar, C. pollutant acrolein. Toxicol. Sci. 132, 467–477.
(2011). Protein conjugated with aldehydes derived from lipid Pizzimenti, S., Ciamporcero, E., Daga, M., Pettazzoni, P., Arcaro,
peroxidation as an independent parameter of the carbonyl A., Cetrangolo, G., Minelli, R., Dianzani, C., Lepore, A.,
stress in the kidney damage. Lipids Health Dis. 10, 201. Gentile, F., and Barrera, G. (2013). Interaction of aldehydes
Mizoi, M., Yoshida, M., Saiki, R., Waragai, M., Uemura, K., Akatsu, derived from lipid peroxidation and membrane proteins.
H., Kashiwagi, K., and Igarashi, K. (2014). Distinction between Front. Physiol. 4, 242.
mild cognitive impairment and Alzheimer’s disease by CSF Pugazhenthi, S., Phansalkar, K., Audesirk, G., West, A., and
amyloid beta40 and beta42, and protein-conjugated acrolein. Cabell, L. (2006). Differential regulation of c-jun and CREB
Clin. Chim. Acta 430, 150–155. by acrolein and 4-hydroxynonenal. Free Radic. Biol. Med. 40,
Mohammad, M. K., Avila, D., Zhang, J., Barve, S., Arteel, G., 21–34.
McClain, C., and Joshi-Barve, S. (2012). Acrolein cytotoxicity Ranganna, K., Yousefipour, Z., Nasif, R., Yatsu, F. M., Milton, S. G.,
in hepatocytes involves endoplasmic reticulum stress, mito- and Hayes, B. E. (2002). Acrolein activates mitogen-activated
chondrial dysfunction and oxidative stress. Toxicol. Appl. protein kinase signal transduction pathways in rat vascular
Pharmacol. 265, 73–82. smooth muscle cells. Mol. Cell. Biochem. 24, 83–98.
254 | TOXICOLOGICAL SCIENCES, 2015, Vol. 143, No. 2

Ribeiro, C. M. and O’Neal, W. K. (2012). Endoplasmic reticulum Toxicological Profile For ACROLEIN. (2007). Available at:
stress in chronic obstructive lung diseases. Curr. Mol. Med. 12, http://www.atsdr.cdc.gov/ToxProfiles/tp124.pdf. Accessed
872–882. November 10, 2014.
Rom, O., Kaisari, S., Aizenbud, D., and Reznick, A. Z. (2013). The Tran, T. N., Kosaraju, M. G., Tamamizu-Kato, S., Akintunde, O.,
effects of acetaldehyde and acrolein on muscle catabolism in Zheng, Y., Bielicki, J. K., Pinkerton, K., Uchida, K., Lee, Y. Y.,
C2 myotubes. Free Radic. Biol. Med. 65C, 190–200. and Narayanaswami, V. (2014). Acrolein modification
Roy, J., Pallepati, P., Bettaieb, A., and Averill-Bates, D. A. (2010). impairs key functional features of rat apolipoprotein
Acrolein induces apoptosis through the death receptor path- E: identification of modified sites by mass spectrometry.
way in A549 lung cells: role of p53. Can. J. Physiol. Pharmacol. Biochemistry 53, 361–375.
88, 353–368. Uchida, K., Kanematsu, M., Morimitsu, Y., Osawa, T., Noguchi,
Roy, J., Pallepati, P., Bettaieb, A., Tanel, A., and Averill-Bates, D. N., and Niki, E. (1998a). Acrolein is a product of lipid peroxida-
A. (2009). Acrolein induces a cellular stress response and trig- tion reaction. Formation of free acrolein and its conjugate
gers mitochondrial apoptosis in A549 cells. Chem. Biol. Int. with lysine residues in oxidized low density lipoproteins.

Downloaded from https://academic.oup.com/toxsci/article/143/2/242/1676672 by guest on 10 November 2022


181, 154–167. J. Biol. Chem. 273, 16058–16066.
Sarkar, P. and Hayes, B. E. (2007). Induction of COX-2 by acrolein Uchida, K., Kanematsu, M., Sakai, K., Matsuda, T., Hattori, N.,
in rat lung epithelial cells. Mol. Cell. Biochem. 301, 191–199. Mizuno, Y., Suzuki, D., Miyata, T., Noguchi, N., Niki, E., and
Seiner, D. R., LaButti, J. N., and Gates, K. S. (2007). Kinetics and Osawa, T. (1998b). Protein-bound acrolein: potential markers
mechanism of protein tyrosine phosphatase 1B inactivation for oxidative stress. Proc. Natl. Acad. Sci. U.S.A. 95, 4882–4887.
by acrolein. Chem. Res. Toxicol. 20, 1315–1320. Vacaru, A. M., Di Narzo, A. F., Howarth, D. L., Tsedensodnom, O.,
Shao, B. (2012). Site-specific oxidation of apolipoprotein A-I im- Imrie, D., Cinaroglu, A., Amin, S., Hao, K., and Sadler, K. C.
pairs cholesterol export by ABCA1, a key cardioprotective (2014). Molecularly defined unfolded protein response sub-
function of HDL. Biochim. Biophys. Acta 1821, 490–501. classes have distinct correlations with fatty liver disease in
Shi, Y., Sun, W., McBride, J. J., Cheng, J. X., and Shi, R. (2011). zebrafish. Dis. Model. Mech. 7, 823–835.
Acrolein induces myelin damage in mammalian spinal cord. Valacchi, G., Pagnin, E., Phung, A., Nardini, M., Schock, B. C., Cross,
J. Neurochem. 117, 554–564. C. E., and van der Vliet, A. (2005). Inhibition of NFkappaB acti-
Song, J. J., Lee, J. D., Lee, B. D., Chae, S. W., and Park, M. K. (2013). vation and IL-8 expression in human bronchial epithelial cells
Effect of acrolein, a hazardous air pollutant in smoke, on hu- by acrolein. Antioxid. Redox Signal. 7, 25–31.
man middle ear epithelial cells. Int. J. Pediatr. Otorhinolaryngol. van der Toorn, M., Smit-de Vries, M. P., Slebos, D. J., de Bruin, H.
77, 1659–1664. G., Abello, N., van Oosterhout, A. J., Bischoff, R., and
Srivastava, S., Sithu, S. D., Vladykovskaya, E., Haberzettl, P., Kauffman, H. F. (2007). Cigarette smoke irreversibly modifies
Hoetker, D. J., Siddiqui, M. A., Conklin, D. J., D’Souza, S. E., and glutathione in airway epithelial cells. Am. J. Physiol. Lung Cell.
Bhatnagar, A. (2011). Oral exposure to acrolein exacerbates Mol. Physiol. 293, L1156–L1162.
atherosclerosis in apoE-null mice. Atherosclerosis 215, Vidro-Kotchan, E., Yendluri, B. B., Le-Thai, T., and Tsin, A. (2011).
301–308. NBHA reduces acrolein-induced changes in ARPE-19 cells:
Stevens, J. F. and Maier, C. S. (2008). Acrolein: sources, metabo- possible involvement of TGFbeta. Curr. Eye Res. 36, 370–378.
lism, and biomolecular interactions relevant to human Volpi, G., Facchinetti, F., Moretto, N., Civelli, M., and Patacchini,
health and disease. Mol. Nutr. Food Res. 52, 7–25. R. (2011). Cigarette smoke and alpha, beta-unsaturated alde-
Sun, L., Luo, C., Long, J., Wei, D., and Liu, J. (2006). Acrolein is a hydes elicit VEGF release through the p38 MAPK pathway in
mitochondrial toxin: effects on respiratory function and human airway smooth muscle cells and lung fibroblasts.
enzyme activities in isolated rat liver mitochondria. Br. J. Pharmacol. 163, 649–661.
Mitochondrion 6, 136–142. Von Tungeln, L. S., Yi, P., Bucci, T. J., Samokyszyn, V. M., Chou, M.
Sun, Y., Ito, S., Nishio, N., Tanaka, Y., Chen, N., and Isobe, K. I. W., Kadlubar, F. F., and Fu, P. P. (2002). Tumorigenicity of
(2014). Acrolein induced both pulmonary inflammation chloral hydrate, trichloroacetic acid, trichloroethanol,
and the death of lung epithelial cells. Toxicol. Lett. 229, malondialdehyde, 4-hydroxy-2-nonenal, crotonaldehyde,
384–392. and acrolein in the B6C3F(1) neonatal mouse. Cancer Lett. 185,
Tanel, A. and Averill-Bates, D. A. (2005). The aldehyde acrolein 13–19.
induces apoptosis via activation of the mitochondrial path- Voulgaridou, G. P., Anestopoulos, I., Franco, R., Panayiotidis, M.
way. Biochim. Biophys. Acta 1743, 255–267. I., and Pappa, A. (2011). DNA damage induced by endoge-
Tanel, A. and Averill-Bates, D. A. (2007a). Activation of the death nous aldehydes: current state of knowledge. Mutat. Res. 711,
receptor pathway of apoptosis by the aldehyde acrolein. Free 13–27.
Radic. Bio. Med. 42, 798–810. Wang, G. W., Guo, Y., Vondriska, T. M., Zhang, J., Zhang, S., Tsai,
Tanel, A. and Averill-Bates, D. A. (2007b). Inhibition of acrolein- L. L., Zong, N. C., Bolli, R., Bhatnagar, A., and Prabhu, S. D.
induced apoptosis by the antioxidant N-acetylcysteine. (2008). Acrolein consumption exacerbates myocardial ische-
J. Pharmacol. Exp. Ther. 321, 73–83. mic injury and blocks nitric oxide-induced PKCepsilon
Tanel, A., Pallepati, P., Bettaieb, A., Morin, P., and Averill-Bates, signaling and cardioprotection. J. Mol. Cell. Cardiol. 44,
D. A. (2014). Acrolein activates cell survival and apoptotic 1016–1022.
death responses involving the endoplasmic reticulum in Wang, H. T., Hu, Y., Tong, D., Huang, J., Gu, L., Wu, X. R., Chung,
A549 lung cells. Biochim. Biophys. Acta 1843, 827–835. F. L., Li, G. M., and Tang, M. S. (2012). Effect of carcinogenic
Tang, M. S., Wang, H. T., Hu, Y., Chen, W. S., Akao, M., Feng, Z., acrolein on DNA repair and mutagenic susceptibility. J. Biol.
and Hu, W. (2011). Acrolein induced DNA damage, mutage- Chem. 287, 12379–12386.
nicity and effect on DNA repair. Mol. Nutr. Food Res. 55, Wang, T., Liu, Y., Chen, L., Wang, X., Hu, X. R., Feng, Y. L., Liu, D.
1291–1300. S., Xu, D., Duan, Y. P., Lin, J., et al. (2009). Effect of sildenafil on
MOGHE ET AL. | 255

acrolein-induced airway inflammation and mucus produc- Yadav, U. C. and Ramana, K. V. (2013). Regulation of NF-kappaB-
tion in rats. Eur. Respir. J. 33, 1122–1132. induced inflammatory signaling by lipid peroxidation-
Wang, L., Sun, Y., Asahi, M., and Otsu, K. (2011). Acrolein, an derived aldehydes. Oxid. Med. Cell. Longev. 2013, 690545.
environmental toxin, induces cardiomyocyte apoptosis via Yoshida, M., Mikami, T., Higashi, K., Saiki, R., Mizoi, M., Fukuda,
elevated intracellular calcium and free radicals. Cell Biochem. K., Nakamura, T., Ishii, I., Nishimura, K., Toida, T., et al.
Biophys. 61, 131–136. (2012). Inverse correlation between stroke and urinary
Wang, H. T., Weng, M. W., Chen, W. C., Yobin, M., Pan, J., Chung, 3-hydroxypropyl mercapturic acid, an acrolein-glutathione
F. L., Wu, X. R., Rom, W., and Tang, M. S. (2013). Effect of CpG metabolite. Clin. Chim. Acta 413, 753–759.
methylation at different sequence context on acrolein- and Zarkovic, N., Cipak, A., Jaganjac, M., Borovic, S., and Zarkovic, K.
BPDE-DNA binding and mutagenesis. Carcinogenesis 34, (2013). Pathophysiological relevance of aldehydic protein
220–227. modifications. J. Proteomics 92, 239–247.
Watanabe, K., Nakazato, Y., Saiki, R., Igarashi, K., Kitada, M., and Zhang, H. and Forman, H. J. (2008). Acrolein induces heme oxy-
Ishii, I. (2013). Acrolein-conjugated low-density lipoprotein genase-1 through PKC-delta and PI3K in human bronchial

Downloaded from https://academic.oup.com/toxsci/article/143/2/242/1676672 by guest on 10 November 2022


induces macrophage foam cell formation. Atherosclerosis 227, epithelial cells. Am. J. Respir. Cell Mol. Biol. 38, 483–490.
51–57. Zheng, L., Park, J., Walls, M., Tully, M., Jannasch, A., Cooper, B.,
Watzek, N., Scherbl, D., Feld, J., Berger, F., Doroshyenko, O., Fuhr, and Shi, R. (2013). Determination of urine 3-HPMA, a stable
U., Tomalik-Scharte, D., Baum, M., Eisenbrand, G., and acrolein metabolite in a rat model of spinal cord injury.
Richling, E. (2012). Profiling of mercapturic acids of acrolein J. Neurotrauma 30, 1334–1341.
and acrylamide in human urine after consumption of potato Zhou, A. X. and Tabas, I. (2013). The UPR in atherosclerosis.
crisps. Mol. Nutr. Food Res. 56, 1825–1837. Semin. Immunopathol. 35, 321–332.
Wu, C. C., Hsieh, C. W., Lai, P. H., Lin, J. B., Liu, Y. C., and Wung, B. Zhu, L., Liu, Z., Feng, Z., Hao, J., Shen, W., Li, X., Sun, L., Sharman, E.,
S. (2006). Upregulation of endothelial heme oxygenase-1 ex- Wang, Y., Wertz, K., et al. (2010). Hydroxytyrosol protects against
pression through the activation of the JNK pathway by suble- oxidative damage by simultaneous activation of mitochondrial
thal concentrations of acrolein. Toxicol. Appl. Pharmacol. 214, biogenesis and phase II detoxifying enzyme systems in retinal
244–252. pigment epithelial cells. J. Nutr. Biochem. 21, 1089–1098.

You might also like