Anda di halaman 1dari 17

Perspective

pubs.acs.org/crt

Pyrrolizidine Alkaloids: Potential Role in the Etiology of Cancers,


Pulmonary Hypertension, Congenital Anomalies, and Liver Disease
John A. Edgar,*,† Russell J. Molyneux,‡ and Steven M. Colegate§

CSIRO Food and Nutrition, 11 Julius Avenue, North Ryde, NSW 2113, Australia

Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, 34 Rainbow Drive, Hilo, Hawaii 96720, United States
§
Poisonous Plant Research Laboratory, ARS/USDA, 1150 East 1400 North, Logan, Utah 84341, United States

ABSTRACT: Large outbreaks of acute food-related poisoning,


characterized by hepatic sinusoidal obstruction syndrome, hemor-
rhagic necrosis, and rapid liver failure, occur on a regular basis in
some countries. They are caused by 1,2-dehydropyrrolizidine
alkaloids contaminating locally grown grain. Similar acute poisoning
can also result from deliberate or accidental consumption of 1,2-
dehydropyrrolizidine alkaloid-containing herbal medicines, teas, and
spices. In recent years, it has been confirmed that there is also
significant, low-level dietary exposure to 1,2-dehydropyrrolizidine
alkaloids in many countries due to consumption of common foods
such as honey, milk, eggs, salads, and meat. The level of 1,2-
dehydropyrrolizidine alkaloids in these foods is generally too low
and too intermittent to cause acute toxicity. However, these
alkaloids are genotoxic and can cause slowly developing chronic diseases such as pulmonary arterial hypertension, cancers,
cirrhosis, and congenital anomalies, conditions unlikely to be easily linked with dietary exposure to 1,2-dehydropyrrolizidine
alkaloids, especially if clinicians are unaware that such dietary exposure is occurring. This Perspective provides a comprehensive
review of the acute and chronic toxicity of 1,2-dehydropyrrolizidine alkaloids and their potential to initiate certain chronic
diseases, and suggests some associative considerations or indicators to assist in recognizing specific cases of diseases that may
have resulted from dietary exposure to these hazardous natural substances. If it can be established that low-level dietary exposure
to 1,2-dehydropyrrolizidine alkaloids is a significant cause of some of these costly and debilitating diseases, then this should lead
to initiatives to reduce the level of these alkaloids in the food chain.

■ CONTENTS
1. Introduction 4
Notes
Abbreviations
13
13
References 13
2. Toxicology 6
2.1. Occurrence, Evolution, and Chemistry 6
2.2. Bioactivation and Mechanism of Toxicity 6
1. INTRODUCTION
3. Pathological Effects 7
3.1. Sinusoidal Obstruction Syndrome and Cir- 1,2-Dehydropyrrolizidine ester alkaloids (dehydroPAs) have
rhosis 7 been known for almost a century to be the cause of large and
3.2. Pulmonary Arterial Hypertension (PAH) 8 recurring episodes of acute hepatotoxicity in a number of
3.3. Carcinogenesis 9 countries.1−16 The alkaloids responsible for these outbreaks of
3.4. Teratogenicity 9 poisoning are produced by weeds that often infest grain crops,
4. Possible Indicators of Dehydropyrrolizidine Alka- and the primary cause is consumption of bread made using
loid Involvement 9 dehydroPA-contaminated flour. Typical infestations of dehy-
4.1. Cancer 9 droPA plants are shown in Figure 1.
4.2. Pulmonary Arterial Hypertension 10 Episodes of acute dehydroPA toxicity involving the
4.3. Congenital Anomalies 11 consumption of bread are avoided in many countries by
4.4. Liver Disease 11 more effective control of weeds in crops and by strictly applying
4.5. Exacerbating Factors 11 food safety standards limiting the number of foreign seeds in
5. Selected Indicators Suggesting a Possible Dehy- grain entering the human food chain, including some known to
dropyrrolizidine Alkaloid Etiology 12 contain dehydroPAs. Flour millers also normally screen grain
6. Conclusions and Future Directions 12
Author Information 13 Received: July 31, 2014
Corresponding Author 13 Published: December 6, 2014

© 2014 American Chemical Society 4 DOI: 10.1021/tx500403t


Chem. Res. Toxicol. 2015, 28, 4−20
Chemical Research in Toxicology Perspective

Figure 1. Typical rural scenes showing plants producing dehydroPAs associated with agricultural production: (A) horses grazing a pasture infested
with Senecio madagascariensis (fireweed) in Hawaii, (B) Jacobaea vulgaris (Senecio jacobaea, tansy ragwort) in a wheat crop in Germany, (C) Echium
vulgare (blueweed, blue borage, viper’s bugloss) in a grain crop in Germany, and (D) a pasture infestation of Echium plantagineum (Paterson’s curse,
Salvation Jane) in Australia.

prior to milling to ensure that all foreign seeds and plant contaminated by dehydroPAs, e.g., up to 2000−4000 μg of
fragments are removed. However, if dehydroPA-containing dehydroPAs/kg in some supermarket honeys.28,29,40 Levels of
plants were present in the crop, then dust containing dehydroPAs up to 5647 μg/kg have recently been reported in
dehydroPAs is not removed by screening and, consequently, teas purchased in retail markets in Germany.21 The levels of
some dehydroPAs remain associated with the grain.17−20 It is dehydroPAs found in these foods are generally too low to cause
normal practice in countries undertaking large-scale cereal acute hepatotoxicity.18,31,32,42−44 However, based on a consid-
production to combine grain from many farms and from eration of their well-established genotoxicity8,45,46 and on
different cropping areas. Such bulking dilutes and reduces the recent risk assessments suggesting that to make cancer unlikely
level of unwanted contaminants in the grain, including a maximum tolerable daily intake of 0.007 μg of dehydroPAs/
dehydroPAs, but it results in a wider distribution of any kg body weight (bw) should not be exceeded,31,42−44 the
dehydroPAs that were present and thus increases the dehydroPAs in these foods could be contributing to somatic
population exposed to low levels of these hazardous chemicals mutations, leading to a wide range of cancers.8,46 They may also
in grain-based foods. While these measures and practices are be initiating other chronic diseases such as pulmonary arterial
apparently sufficient to prevent the acute dehydroPA poisoning hypertension (PAH), leading to enlargement of the right
that regularly occurs in countries in which such measures and ventricle and right heart congestive failure,8,20,47 and may be a
practices are not applied, it is still possible that occasional, low- cause of some congenital anomalies.8,20,47−49
level dietary exposure in grain-based products could be This Perspective provides a comprehensive review of the
contributing worldwide to the incidence of several slowly characteristics of acute and chronic dehydroPA toxicity and
developing, chronic diseases.18,19 suggests some associative considerations, or indicators, for
Other food items increasingly being reported to be linking specific cases of cancer, PAH, congenital anomalies, and
periodically contaminated by low levels of dehydroPAs include liver diseases with intermittent, low-level dietary exposure to
milk, eggs, meat, honey, pollen, teas, salads, and foods dehydroPAs. Some of these indicators are then used to identify
containing these as ingredients.18,20−40 Both Echium spp. cases of chronic diseases reported in the literature that may
shown in Figure 1, and a number of other dehydroPA- have resulted from dietary exposure to dehydroPAs. The
producing plants, are widely used in commercial honey etiology of these cases warrants further investigation to confirm
production, 41 and the honey produced is significantly or negate this possibility.
5 DOI: 10.1021/tx500403t
Chem. Res. Toxicol. 2015, 28, 4−20
Chemical Research in Toxicology Perspective

Many cases of cancer, PAH, congenital anomalies, and liver monoesters (e.g., heliotrine), (b) open-chain diesters (e.g.,
disease are of unknown etiology, so establishing that they are lasiocarpine), (c) macrocyclic diesters (e.g., retrorsine), and (d)
sometimes caused by dietary exposure to dehydroPAs should seco-alkaloids (e.g., senkirkine). This complexity is further
lead to ways of reducing the incidence of these costly and amplified by their existence in plants as both free bases and as
debilitating medical conditions. The challenge is to identify N-oxides (Figure 2), the latter predominating in many plant
specific cases that have been initiated or exacerbated by dietary species.4,52
exposure to dehydroPAs. 2.2. Bioactivation and Mechanism of Toxicity.
DehydroPA free bases are pro-toxins. Following ingestion,
2. TOXICOLOGY they are absorbed from the gut and converted in the liver, by
cytochrome P450 monooxygenases (CYP450), specifically the
2.1. Occurrence, Evolution, and Chemistry. It has been CYP3A and CYP2B isoforms in humans, to 1-hydroxymethyl-7-
estimated that 3% of all flowering plants produce poisonous hydroxy-6,7-dihydropyrrolizine ester metabolites (DHP esters)
dehydroPAs.50 They are very effective insect-feeding deterrents (Figure 3).8,46,52−75 These “pyrrolic” metabolites are bifunc-
and consequently have evolved independently on at least four tional biological alkylating agents capable of causing tissue
occasions in a number of different plant families.51 Approx- damage and inducing genetic mutations.52 In vivo N-oxidation
imately 500 potentially toxic dehydroPAs are known.4,52 of dehydroPAs by CYP450 enzymes and flavin-containing
Varying in the character and/or stereochemistry of the necine mono-oxygenases68 is a detoxifying mechanism that leads to
base (Figure 2) and the esterifying acids (necic acids), they can excretion of the water-soluble dehydroPAs N-oxides produced
be classified into four main categories (Figure 2): (a) (Figure 3);52,76 however, a significant proportion of ingested
dehydroPA N-oxides are reduced to their free base forms
during passage through the gut and in the liver52 and thus,
when they are present in food, they too contribute to hepatic
formation of genotoxic, mutagenic, tissue-damaging DHP esters
(Figure 3).8,68,77−83
While the DHP ester metabolites, e.g., dehydroriddelliine
shown in Figure 3 and dehydromonocrotaline (Figure 4), have
been chemically synthesized from dehydroPAs,84−87 they have
never been isolated from in vivo or in vitro metabolic studies
due to their extremely high chemical reactivity and very short
half-lives, especially in aqueous environments.52,88 After
formation in hepatocytes, the DHP ester metabolites react
rapidly with nucleophilic functional groups (sulfhydryl, amino,
hydroxyl) on DNA, proteins, and other substances, e.g.,
glutathione (GSH), in the liver to produce mixtures of C7
and C9 mono and di DHP adducts, the latter forming cross-
links between −SH, −OH, and −NH groups on different
molecules or on the same macromolecule (Figures 3 and
4).46,52,57−75,89−91
Rapid, spontaneous in vivo hydrolysis of the DHP esters
produces the relatively more stable enantiomers of the DHP
diol, (±)-1-hydroxymethyl-7-hydroxy-6,7-dihydropyrrolizine
(DHP) (Figure 3), namely, dehydroheliotridine55 and dehy-
droretronecine,92 or, more likely, a racemic mixture of these
two enantiomers.52 Unlike the highly reactive DHP esters, the
effects of which are largely if not entirely confined to the
liver,8,52 DHP survives for a longer time in the body.52 It
escapes from the liver and has been isolated and identified in
both in vivo and in vitro studies of dehydroPA metabo-
lism.54,55,92 While less chemically reactive than the precursor
DHP esters, DHP retains significant bifunctional, biological
alkylating potential, especially under mildly acid conditions,
and, as with the DHP esters, it forms C7 and C9 DHP adducts
in vivo (Figure 3).52,87,93−97 DHP adducts have been detected
in the liver, lung, kidney, blood, bone marrow, gastrointestinal
tract, brain, muscles, pancreas, thymus, heart, spleen, and testes
of animals exposed to dehydroPAs and following intra-
peritoneal or subcutaneous injection of DHP.4,25,52,90,98−103
DHP, the final and common toxic metabolite of all
Figure 2. Three most common necine bases defining dehydroPAs, viz., dehydroPAs, is carcinogenic104−106 and immunosuppressive107
retronecine, heliotridine, and otonecine; a general structure represent- and mimics the antimitotic effects of radiation in tissues
ing dehydroPA N-oxides; and structures representing the four main displaying high rates of mitosis.108−111 Circulating DHP is
categories of dehydroPA pro-toxins. considered to be responsible for the effects of dehydroPAs
6 DOI: 10.1021/tx500403t
Chem. Res. Toxicol. 2015, 28, 4−20
Chemical Research in Toxicology Perspective

Figure 3. Liver metabolism of dehydroPAs, using the macrocyclic diester riddelliine as an example.

outside of the liver. It is, however, highly water-soluble, and any 3. PATHOLOGICAL EFFECTS
DHP that remains unadducted is excreted (Figure 3).52 3.1. Sinusoidal Obstruction Syndrome and Cirrhosis.
The potential harm of dietary exposure to dehydroPAs does Following exposure to relatively high doses of dehydroPAs, e.g.,
not end with formation of the initial DHP adducts or excretion tens of milligrams of dehydroPAs per kilogram of bodyweight,
of dehydroPA N-oxides and DHP. Mono DHP adducts, the liver sinusoidal endothelial cells have been shown, in animal
retaining one active alkylating site at C7 or C9, remain models, to be the first to display the effects of the DHP ester
potentially hazardous as alkylating agents in vivo.112 In accord metabolites produced in adjacent hepatocytes. They have been
with this, a mixture of preformed galectin-1-DHP adducts observed to round-up, swell, and slough off, leading to blockage
(Figure 4), shown by mass spectrometry to be mainly of the microcirculation in the sinusoids.114−118 The resulting
monoadducted, was found to be cytotoxic in human pulmonary hepatic sinusoidal obstruction syndrome (HSOS) (also referred
artery endothelial cells.112 More importantly, it has been to as hepatic veno-occlusive disease or VOD) is highly
established that some DHP adducts, involving DHP attached to characteristic of acute dehydroPA poisoning seen in people8,14
weak nucleophiles, can dissociate and release DHP or transfer and leads to necrosis of surrounding liver tissue, fibrosis,
DHP to stronger nucleophilic sites.52,95−97,109 The in vivo nodular hyperplasia, bile duct proliferation, and eventually to
persistence of “a reservoir of stabilized pyrrolic alkylating agents cirrhosis and liver failure.8,110,117−119 DehydroPAs are the only
(DHP adducts) which could subsequently interact with other naturally occurring agents in the environment known to
produce HSOS when consumed, and they are frequently used
tissue constituents”52 is one reason why the toxic effects of
to produce animal models of clinical cases of HSOS and
dehydroPAs continue to develop slowly over an extended chronic, progressive liver disease that are used to study the
period even from a single exposure to these hazardous development of, and potential treatments for, these dis-
substances.52,95−97,109−111,113 The reversibility of the reaction eases.115,118,120,121
of dehydroPA metabolites with certain weak nucleophiles The particular susceptibility of the sinusoidal endothelial cells
prolongs the effects and enhances the hazard of dietary is thought to be, in part, associated with increased activity of
exposure to dehydroPAs relative to some other genotoxic matrix metalloproteinases (MMPs), e.g., MMP-9 (gelatinase
substances, and this fact should be taken into account in B), that degrade the extracellular matrix, leading to the
assessing the risk of foods contaminated by these alkaloids. endothelial cells being released.116,117,122 MMPs are normally
7 DOI: 10.1021/tx500403t
Chem. Res. Toxicol. 2015, 28, 4−20
Chemical Research in Toxicology Perspective

Figure 4. A typical mixture of DHP adducts produced by reaction of galectin-1 with dehydromonocrotaline.112

suppressed by GSH. As sinusoidal GSH levels rapidly decrease experimental models of PAH.134−139 Doses of dehydroPAs
due to alkylation by dehydroPA metabolites and the redox state required to produce PAH in experimental animals comparable
of the sinusoids changes, the activity of the MMPs increases, to clinical cases of PAH in humans are generally lower than
and HSOS develops. In support of this mechanism, it has been those required to produce HSOS. This may explain, therefore,
demonstrated, for example, that GSH infused into the portal the occasional development of PAH, without notable liver
vein prevents HSOS from developing in animals exposed to pathology, in people taking herbal medicines containing
dehydroPAs.116,123 dehydroPAs.140−142
While HSOS is considered to be highly indicative of Children form a significant cohort of PAH cases of unknown
dehydroPA exposure8,111,119 and has developed in humans etiology. The PAH produced in nonhuman primates by
consuming levels as low as tens of micrograms of dehydroPAs monocrotaline shows very similar pathology to PAH diagnosed
per kilogram of bodyweight per day,17,110,124,125 it is not always in children.130−132 Rodents given monocrotaline are apparently
readily apparent. For example, when the liver damage develops a less appropriate model of the human disease,131,132,136 but the
over an extended period, due to relatively low-level exposure to monocrotaline-rodent model has become the most widely used
dehydroPAs, HSOS may not be apparent or may be obscured to study the development of PAH and to test treatments for
by hepatic fibrosis and nodular regeneration.110 Under these this condition.137,138 Stenmark et al.136 and Gomez-Arroyo et
circumstances, the resulting cirrhosis may be indistinguishable al.139 have discussed some of the differences between the
from cirrhosis caused by other agents, e.g., alcohol, aflatoxins, monocrotaline-rodent models of PAH and PAH seen in
and hepatitis viruses, and the etiology can be ambiguous.110,119 humans. These differences appear to relate to the initial acute
Thus, as well as clinical cases of HSOS, some cases of cirrhosis effects of dehydroPAs that are not seen in clinical cases of PAH.
of unknown cause and without obvious HSOS could also have a These discordant effects could be explained, in part, by the
dietary dehydroPA etiology. rodent model of PAH typically being generated using a single,
3.2. Pulmonary Arterial Hypertension (PAH). Recog- relatively high dose of monocrotaline, sufficient to produce
nition that ingestion of dehydroPAs causes PAH leading to PAH relatively quickly in a high proportion of animals and not
enlargement of the right ventricle and right heart failure (cor just the most susceptible. A long-term, intermittent, lower level
pulmonale) was established many years ago in experimental of exposure, similar to that expected from the levels of
animals,56,126−133 and, since then, dehydroPAs such as dehydroPAs currently being detected in food, may avoid the
monocrotaline and fulvine have been deliberately administered early discordant acute effects sometimes seen in the standard
by medical researchers to various animal species to generate monocrotaline-rodent model of PAH136,139 and may be
8 DOI: 10.1021/tx500403t
Chem. Res. Toxicol. 2015, 28, 4−20
Chemical Research in Toxicology Perspective

expected to slowly produce PAH more typical of the disease dehydroPA N-oxide, indicine-N-oxide, was at one time clinically
normally seen in humans and in the nonhuman primate model investigated for use as a cancer chemotherapeutic agent.148,149
developed by Chesney and Allen.130−132 The antimitotic effect of dehydroPAs exposure can, therefore,
Several experiments with nonhuman primates provide be expected to initially inhibit both cancer development and the
insights into what can be expected from long-term, low-level development of the quasi-malignant endothelial cells in the
dietary exposure to dehydroPAs.130−132 A highly significant, pulmonary arterioles that characterize PAH.150 The develop-
age-related difference was observed following subcutaneous ment of both cancer and PAH will, therefore, be particularly
injection of the relatively high dose of 30 mg/kg bw favored by extended, low-level, and intermittent periods of
monocrotaline, followed by three further injections, 2 months dehydroPA exposure, as is expected from current knowledge of
apart, of 60 mg/kg bw, to 4 week old and 15 month old foods contaminated by dehydroPAs, as this type of exposure
monkeys.130 The older animals, as expected from the very high allows intervening periods of normal mitotic activity that
doses administered, developed HSOS and showed typical provide opportunity for cancers and progressive PAH to
dehydroPA liver pathology, whereas the young monkeys develop.8,52,110,150−155
showed minimal liver pathology but developed PAH typical 3.4. Teratogenicity. Exposure of pregnant women to
of that seen in children. It was established that the young dehydroPAs in herbal teas, herbal medicines, and spices has
monkeys displayed much lower cytochrome CYP450 activity in caused fatal HSOS in neonates.8,52,110,125 DehydroPAs and
their livers and would therefore have produced insufficient their DHP metabolites are teratogenic in rats, and it has been
levels of the monocrotaline DHP ester metabolite (dehydro- shown that they cross the placenta and form DHP adducts in
monocrotaline) to rapidly deplete sinusoidal GSH, activate rat embryos.48,49,52 Growth retardation, abnormal skeletal
MMPs, release sinusoidal endothelial cells, and cause HSOS. development, and deficiencies in ossification are common
They must, however, have produced sufficient low levels of features. Congenital anomalies have, however, not yet been
circulating, labile DHP adducts and DHP to damage the extensively studied or considered as a possible consequence of
endothelium of the pulmonary arteries and cause PAH. The low-level dietary exposure to dehydroPAs. Nonetheless, as well
response of the younger monkeys represents the situation that as stillbirths and HSOS, congenital anomalies are to be
would most likely pertain if a human diet regularly delivered expected in surviving neonates following dietary exposure of
very small amounts of dehydroPAs that yielded insufficient their mothers to dehydroPAs during pregnancy.
toxic metabolites to cause HSOS but sufficient to initiate PAH
and perhaps cause somatic mutations leading to other 4. POSSIBLE INDICATORS OF
genotoxic conditions such as cancer. Thus, PAH and cancer, DEHYDROPYRROLIZIDINE ALKALOID
rather than HSOS, are the most likely outcomes from INVOLVEMENT
circulating low microgram per kilogram of bodyweight levels Growing evidence of significant dehydroPA contamination of a
of labile DHP adducts and free DHP. This would be especially wide range of common foods suggests that they could be a
so if dietary exposure to dehydroPAs is intermittent since this common cause of several chronic diseases.20 The ability of
would allow the well-established antimitotic effects of dehydroPAs to cause disparate chronic diseases, viz., cancers,
dehydroPA metabolites to be relatively short-lived and enable PAH, HSOS, and genetic anomalies, suggests that disease
cancers and progressive PAH, both requiring cell proliferation, complexes involving these diseases could be used to indicate
to develop. This type of intermittent, low-level dietary exposure the involvement of dehydroPAs. This section considers the
seems increasingly likely to have occurred in the past and to identification of diseases potentially caused by dietary
continue to the present day to varying degrees in many regions dehydroPAs in this context.
throughout the world.20,47 Current knowledge of the relatively 4.1. Cancer. Linking a diverse range of cancers with somatic
low but significant levels of dehydroPAs in food and an mutations arising from relatively constant or, more effectively,
expectation of intermittent dietary exposure, insufficient to intermittent, low-level, long-term dietary exposure to dehy-
produce HSOS, suggest that dietary dehydroPAs could be droPAs is likely to be difficult, but the highly characteristic
responsible for at least some cases of clinical PAH of currently ability of dehydroPAs to produce HSOS, PAH, and genetic
unknown etiology reported in the medical literature. anomalies could be used as clinical signs suggesting such a link.
3.3. Carcinogenesis. It has been shown, using exper- Evidence of one or more of these other conditions can be
imental animals and human cell cultures, that one of several expected in at least some patients with cancers caused by
genetic targets for toxic dehydroPA metabolites is the gene dehydroPAs.
TP53, encoding the tumor suppressor protein p53.46,143,144 As well as exposure to dehydroPAs, another clinically
Signature mutations of dehydroPAs include G:C → T:A recognized and common cause of dehydroPA-like HSOS is
transversion and tandem base substitutions.46 Cancers of the associated with chemotherapy of childhood cancers such as
liver, lung, kidney, skin, intestines, bladder, brain and spinal rhabdomyosarcoma (RMS), nephroblastoma (Wilms tumor),
cord, pancreas, adrenal gland, muscle (rhabdomyosarcoma, metastatic colorectal cancer, and leukemia using alkylating
RMS), and blood (leukemia) have been produced by agents such as cyclophosphamide or busulfan.156−164 Treat-
dehydroPAs and their metabolites in experimental ani- ment of hematologic cancers in neonates by allogenic stem cell
mals.8,46,145,146 These are also the tissues in which DHP transplantation (SCT) following myeloablation using these
adducts have been detected. A very similar, wide range of agents can also cause HSOS as a complication.164−177
cancers is associated with Li-Fraumeni syndrome caused by Like dehydroPAs, cyclophosphamide and busulfan are
germline mutations of TP53,147 a known target of dehydroPA biological alkylating agents and thus may simply mimic
metabolites. dehydroPAs in causing HSOS, 178−180 although, unlike
As previously mentioned, dehydroPAs and their DHP dehydroPAs, neither of these agents is used to produce animal
metabolites display strong antimitotic effects8,52,106 typical of models of HSOS. Indeed, cyclophosphamide is not generally
many other biological alkylating agents used to treat cancer. A considered to be hepatotoxic,181 so it is somewhat unexpected
9 DOI: 10.1021/tx500403t
Chem. Res. Toxicol. 2015, 28, 4−20
Chemical Research in Toxicology Perspective

that it causes HSOS in some childhood cancer patients. It is, PAH.185 The dehydroPA monocrotaline and its DHP ester
however, possible that if the cancer being treated developed as metabolite (dehydromonocrotaline, “monocrotaline pyrrole”,
a consequence of dietary exposure to dehydroPAs, for example Figure 4) cause mutation of BMP signaling genes in animal
when fetuses are exposed to dehydroPA metabolites in utero or models of PAH,185−187 and their ability to cause mutations of
as infants consuming dehydroPA-contaminated breast milk BMP-signaling genes is clearly a significant factor in the ability
and/or dehydroPA-contaminated food, then asymptomatic of dehydroPAs to cause PAH.
HSOS may co-occur with their cancers. Overt HSOS can A hallmark feature of both clinical PAH and dehydroPA-
then be precipitated by the additional insult to the sinusoids induced PAH are the proliferative, quasi-malignant, hyper-
from chemotherapeutic agents not normally capable of causing trophic (megalocytic), apoptosis-resistant cells, in particular
HSOS. Dietary exposure to low levels of dehydroPAs may be, smooth muscle cells, that contribute to the characteristic
like radiotherapy, a priming event for HSOS when cancers plexiform (onionskin-like) lesions responsible for progressive
caused by dehydroPAs are treated with therapeutic alkylating vascular remodeling and vasoconstriction of the pulmonary
agents such as cyclophosphamide and busulfan. arterioles.138,188−194 The previously mentioned susceptibility of
DehydroPAs have been shown to induce biosynthesis of muscle cells to DHP alkylation leading to RMS may have a
GSH in the liver in response to the depletion of GSH they parallel in the development of the quasi-malignant smooth
cause.182 The potential genotoxicity associated with continued muscle cells in the pathogenesis of PAH.
dietary exposure to dehydroPAs during induction of GSH In regard to the hypertrophic character of the pulmonary
biosynthesis could cause mutations in the upregulated GSH artery endothelial cells involved in PAH, similar long-lived,
biosynthesis genes. In turn, this could lead to compromise of morphologically and functionally viable megalocytes have long
the ability of some dehydroPA-affected livers to biosynthesize been recognized as a highly characteristic aberration of
GSH and thus dehydroPA-exposed livers could become hepatocytes in dehydroPA-poisoned animals.4,52,195−198 They
sensitized to other alkylating agents such as cyclophosphamide have also been described in the lungs,152 kidney,4,152 and
or busulfan that normally do not cause HSOS. In this regard, it duodenum111 of animals exposed to dehydroPAs. While
would be of interest to compare the development of HSOS in functionally viable and long-lived, the hepatic megalocytes
experimental animals using cyclophosphamide and busulfan that develop in surviving animals following acute dehydroPA-
with and without priming by dietary exposure to very low levels poisoning initially appear to be incapable of division.196,197 This
of dehydroPAs and also to investigate whether the GSH characteristic is, however, believed to be due to the persistent
biosynthesis capability of livers is compromised by such antimitotic effects of dehydroPAs, and, given time, some of
priming. these hypertrophic cells are expected to “escape from mitotic
Rhabdomyosarcoma (RMS) is a cancer originating from inhibition.”197 In some cases, the proliferative, hypertrophic,
mutations in muscle progenitor cells.183 DHP becomes a more apoptosis-resistant smooth muscle cells, seen in the pulmonary
aggressive alkylating agent under mildly acid conditions due to arterioles of patients with PAH, could arise in this way and be
protonation of the alcohol groups, providing a good leaving indicative of intermittent, low-level dietary exposure to
entity (H2O) for generating the carbonium ions involved in dehydroPAs.
alkylation.52,87,95 Therefore, accumulation of lactic acid in Twenty percent of idiopathic PAH cases are associated with
muscle tissue could quite conceivably result in myoblasts being BMP receptor II (BMPR2) mutations, and 75% of familial PAH
a favored site for DHP alkylation, causing mutations and cases display germline mutations of BMPR2.199,200 These
resulting in RMS. In accord with this possibility, while mutations alone are, however, insufficient to cause PAH.199−201
intraperitoneal injection of DHP into rats produces a range The low penetrance (20%) of PAH among carriers of BMPR2
of cancers similar to those produced by ingestion of the parent mutations has led to the suggestion that additional environ-
dehydroPAs,106 subcutaneous injection of the DHP enantiomer mental factors and further somatic mutations may be needed
dehydroretronecine produced RMS at the site of injection in before PAH develops in people carrying BMPR2 mutations. It
65% of rats (39 of 60).104,105 A dehydroPA etiology for some has therefore been suggested that a so-called second hit is
cases of RMS is, therefore, in accord with this observation and required before PAH develops.200,201 Dietary exposure to
also with HSOS being a complication in a significant dehydroPAs could deliver such a second hit in carriers of
proportion of RMS patients when they undergo chemotherapy BMPR2 mutations and lead to the development of PAH in
involving alkylating agents such as cyclophosphamide and these individuals. Thus, dietary exposure to dehydroPAs may
busulfan. not only be responsible for the development of PAH in
Cancer clusters involving several types of cancer could also previously mutation-negative individuals but also could be an
be indicative of a dietary dehydroPA origin. Such heteroge- important contributor to the development of PAH in carriers of
neous clusters are often dismissed as unlikely to have a BMPR2 mutations.
common etiology. However, Daughton184 has drawn attention Portopulmonary hypertension (POPH) is a relatively rare
to the co-occurrence of acute lymphoblastic leukemia and RMS form of PAH associated with cirrhosis and portal hyper-
in Nevada and Arizona and has suggested that this coincidence tension.202−204 The exact pathogenesis and etiology of POPH
could indicate a dietary dehydroPA etiology. It would be of are poorly understood.205 One clinical study of 2459 patients
interest to determine if there was concurrently or subsequently with cirrhosis showed that 15 (0.61%) exhibited POPH and
an increased incidence of cirrhosis, HSOS, and PAH in those concluded that an unknown dietary cause was “the most
communities. attractive explanation for the association of portal and
4.2. Pulmonary Arterial Hypertension. Dysregulation of pulmonary hypertension.”206 Another study207 has suggested
bone morphogenetic protein (BMP) signaling is a feature of that the “prevalence of primary pulmonary hypertension
clinical cases of both sporadic (idiopathic) and inherited associated with cirrhosis may be greater than previously
(familial) PAH and has been implicated in the vascular cell reported.” The combination of cirrhosis, portal hypertension,
proliferation and remodeling of pulmonary arterioles seen in and PAH, characteristic of POPH, resembles the chronic
10 DOI: 10.1021/tx500403t
Chem. Res. Toxicol. 2015, 28, 4−20
Chemical Research in Toxicology Perspective

condition sometimes caused by dehydroPAs in animals, so Environmental copper is known to exacerbate the
dietary dehydroPAs could conceivably be, in some cases, a hepatotoxicity of dehydroPAs in livestock and to result in
differential consideration in determining the etiology of POPH- accumulation of very high levels of copper in the cirrhotic
like diseases. liver.4,52,77,227−230 The poisoning of livestock by dehydroPAs
All cases of PAH of uncertain etiology need to be was at one time called chronic copper toxicity until it was
investigated as being possibly caused, initiated, or exacerbated shown that this particular condition also required exposure to
by dietary exposure to dehydroPAs. plants containing dehydroPAs.4,227 Numerous investigations
4.3. Congenital Anomalies. The skeletal defects seen in have sought to explain individual examples and clusters of
rat fetuses whose mothers were exposed to heliotrine or cirrhosis in young children that have occurred in Germany,
DHP48,49 strongly suggest that the alkaloids are causing Austria, India, the USA, Australia, and elsewhere. Such cases are
deficiencies in BMP signaling.208,209 As previously discussed, sometimes associated with copper accumulation.222−226,231−242
mutation of BMP signaling genes by dehydroPA metabolites Many investigations have concluded, however, that high levels
contributes to their ability to cause PAH in experimental of environmental copper are not the only cause.235,237,238 Some
animals, so it is not surprising that ineffective bone formation is have suggested that other environmental agents, including
among the congenital anomalies seen in animals exposed to exposure to dehydroPAs240,241 and perhaps a non-Wilsonian
dehydroPA metabolites in utero.48,49 genetic predisposition to copper accumulation and toxicity, may
HSOS is commonly experienced as a complication of be required.234,236−240,242−244 This situation is very reminiscent
conditioning regimes involving cyclophosphamide and busulfan of the historic debate in the 1950s on the etiology of chronic
used for prior marrow ablation when, for example, congenital copper toxicity in livestock.4,52,227 Cirrhosis accompanied by
anomalies such as malignant infantile osteopetrosis (MIOP), copper accumulation is, therefore, another important indication
primary immunodeficiency (PID), and severe immune for suspecting possible dietary dehydroPA involvement.
dysregulatory disorders (SIDs) are treated by SCT.210,211 If A form of HSOS called hepatic veno-occlusive disease with
these congenital anomalies developed as a consequence of immunodeficiency (VODI) is associated with germline
maternal exposure to dehydroPAs, then such exposure could mutations of the SP110 nuclear body protein gene.245−250
prime the livers of the neonates for development of HSOS SP110 nuclear body protein is involved in regulation of the
during myeloablation. body’s immune response, so a nonfunctional version of SP110
As well as displaying a predisposition to developing HSOS nuclear body protein will lead to impairment of the immune
following SCT, some MIOP patients also frequently develop system.251 It is, however, less clear how the loss of SP110
severe PAH as a complication.210 PAH was also the most nuclear body protein can cause HSOS.252 This suggests a
possible role for another factor such as dietary dehydroPAs.
common cause of death in a study of 48 patients with
The known immunotoxicity of dehydroPAs and DHP107,108,253
fibrodysplasia ossification progressiva (FOP).212 Of the 48 FOP
could conceivably contribute to the development of a VODI-
patients studied, 26 (54%) died prematurely (age range 8−58,
like disease in susceptible individuals with or without a pre-
median life span 42 years) from PAH. Another possible
existing germline mutation of the SP110 gene. It is even
indication of a dehydroPA etiology is that FOP is characterized
possible that mutation of the SP110 gene by dehydroPA
by congenital toe malformations and ossification issues metabolites could contribute to or account for some of the
involving BMP signaling abnormalities.213,214 immune system impairment associated with dehydroPA
Beckwith−Wiedemann syndrome (BWS), some features of exposure.107
which involve faulty BMP signaling,215,216 is another congenital 4.5. Exacerbating Factors. Modern bulking of commod-
disorder that could sometimes have a dehydroPA etiology. ities such as grain, milk, and honey for wide national and
BWS is associated with significantly increased risk of tumors international distribution leads to dilution of dehydroPA
such as RMS, hepatoblastoma, and Wilms tumor,217−219 contaminants and ensures that dietary exposure to acutely
including, in some cases, the co-occurrence of RMS and the hazardous levels of dehydroPAs is relatively rare in the many
anaplastic subtype of Wilms tumor associated with mutations of countries where such bulking normally occurs. Thus, hazardous
the TP53 gene.220,221 The deficiencies in BMP signaling as well levels of dietary exposure to dehydroPAs are likely to occur
as the susceptibility to developing certain cancers, including more commonly in particular localities and communities or
RMS, and concurrent susceptibility to HSOS during chemo- households due to specific items of food produced and
therapy could quite conceivably indicate that the congenital consumed locally being contaminated by dehydroPAs at certain
defects of BWS, the cancers that develop, and the susceptibility times. Regional and/or familial clusters of dehydroPA-
to HSOS may sometimes all have a common dehydroPA origin. associated chronic diseases are particularly likely in areas
In combination, these observations suggest that a dehydroPA where locally produced and consumed honey, milk, eggs, grain,
etiology should be considered as a possible cause of some cases and other foods are occasionally contaminated via dehydroPA-
of BWS, MIOP, FOP, PID, and SID and possibly other producing plants that grow in that region and especially during
congenital conditions. times when incidents of livestock poisoning are being
4.4. Liver Disease. Dietary exposure to dehydroPAs should reported.254 A familial genetic propensity for hepatic activation
also be considered for all cases of cirrhosis of unknown of dehydroPAs, rather than detoxication, associated with
etiology, especially those showing evidence of HSOS. While inherited CYP450 profiles can also be expected to result in
clinicians dealing with cases of HSOS, especially in neonates family clusters of dehydroPA-initiated chronic diseases.
and young children, often question the mothers of the patients Some medications, e.g., rifampicin, St. Johns wort, and
involved in regard to their consumption of herbal medicines phenobarbital, and environmental contaminants, e.g., poly-
containing dehydroPAs,222−226 the potential for dehydroPAs chlorinated biphenyls, induce cytochrome CYP3A enzymes, so
being present in foods they have consumed, such as milk, concurrent consumption of or exposure to these could increase
honey, flour, tea, and eggs, is not normally considered. an individual’s susceptibility to dehydroPA toxicity via
11 DOI: 10.1021/tx500403t
Chem. Res. Toxicol. 2015, 28, 4−20
Chemical Research in Toxicology Perspective

enhanced bioactivation. Viruses, bacterial endotoxins, and (HSOS),238 and rare mitotic figures223 that are indicative of a
aflatoxins have also been shown to exacerbate liver damage dehydroPA etiology.
and cancers caused by dehydroPAs.255,256 If several of the indicators suggested above are present in an
The particular susceptibility of fetuses and nursing neonates individual case, then the possibility that dietary exposure to
to toxic effects when their mothers consume foods or herbal dehydroPAs is involved deserves further investigation,
medicines containing dehydroPAs is well-established.8,20,257,258 especially if other features of dehydroPA poisoning are also
Fetal and neonate liver CYP450 enzymes are generally less present such as hypertrophic (megalocytic) cells, antimitotic
effective in activating dehydroPA,52,130 and it is thought that the effects, and evidence of immunotoxicity.
DHP metabolites produced by and transferred from the mother
are largely responsible for the harm caused to fetuses and breast
feeding neonates.52 In recognition of the susceptibility of 6. CONCLUSIONS AND FUTURE DIRECTIONS
fetuses and nursing neonates, German regulations for Recent recognition that some individuals and communities may
phytopharmaceuticals specify that dehydroPA-containing herb- be intermittently exposed to hazardous levels of mutagenic
al medicines cannot be sold to pregnant or breast feeding dehydroPAs, naturally present or present as contaminants in a
women.257,258 For other consumers in Germany, the daily dose number of common foods, raises the possibility that these
of dehydroPAs in a small number of specified dehydroPA- natural toxins may be important contributors to the develop-
containing phytopharmaceuticals must not exceed 1 μg, ment of certain chronic diseases in those individuals and
reduced to 0.1 μg if the medication is taken daily for more communities. However, epidemiological support is required,
than 6 weeks per year.257 These regulations are not currently and considerable research is needed to establish the veracity
applied to food, although a significant number of retail food and extent of this possibility. As well as increased regional
items have been shown to exceed the maximum levels of analyses of food to determine current levels of dietary exposure
dehydroPAs allowed in phytopharmaceuticals in Ger-
to dehydroPAs, awareness among clinicians of this possibility
many.21,27−29,33−40
should lead to questioning of patients about their dietary habits
5. SELECTED INDICATORS SUGGESTING A POSSIBLE and the sources of the foods they consume.
DEHYDROPYRROLIZIDINE ALKALOID ETIOLOGY The chronic diseases that could sometimes be caused,
initiated, or precipitated by the current intermittent, low levels
It will be necessary to eventually develop criteria, well
of dietary exposure to dehydroPAs include (a) a wide range of
supported by targeted research and epidemiological studies,
cancers, (b) PAH, (c) progressive liver disease leading to
which can be used to assist in identifying diseases that may be
associated with dietary exposure to dehydroPAs. Until this is cirrhosis, and (d) congenital anomalies, or a combination of
achieved, the following indicators, rationally based on these. PAH and HSOS, in particular, are highly characteristic
associative considerations described above, are recommended and indicative responses to dehydroPA exposure. All cases of
as possible indications of a dietary dehydroPA etiology and chronic illness involving or accompanied by evidence of either
suggest the need for further investigations into possible sources or both of these relatively rarely co-occurring conditions could
of exposure: indicate a possible dietary dehydroPA etiology.160,207
(1) Latent or overt HSOS and PAH of unknown or The availability of methods for detecting and quantifying
uncertain etiology. DHP−DNA71,261 and DHP−protein adducts262 provides
means of measuring levels of dietary exposure to dehydroPAs
(2) Cirrhosis, especially if associated with HSOS and/or in different populations and allows the level of DHP adducts
accumulation of copper in the liver. detected to be correlated with the incidence of certain chronic
(3) Cancers and/or congenital anomalies where there is diseases. Such analysis is, for example, particularly called for in
evidence of overt or asymptomatic HSOS, PAH, bone regions where honeys from dehydroPA-producing plants are
deformities, or immunological deficiencies. harvested and sold locally or when flour from locally harvested
(4) PAH accompanied by evidence of hepatotoxicity. grain could sometimes be contaminated. These data can then
(5) Any of the above occurring in consanguineous or be correlated with the relative incidence of associated chronic
regional clusters, especially in areas where dehydroPA-
diseases in these particular regions.
producing weeds grow and where locally produced The current animal models of PAH produced by exposure to
dehydroPA-contaminated foods are likely to occasionally
dehydroPAs are aimed at generating high numbers of affected
occur and especially if there is concurrent evidence of
animals relatively quickly for experimental purposes, and some
livestock being poisoned by dehydroPAs.254
acute effects of dehydroPAs are, therefore, also produced.
(6) Any of the above involving fetuses, neonates, and
While these animal models do indeed reflect all of the
children.
important aspects of the pathophysiology of PAH, they do not
In considering and selecting the six indicators comprising the
above list, several cases of liver disease and cancers with a mimic likely low level and intermittent dietary exposure to
p o s s i b l e d e h ydr o P A e t i o l o g y ha v e b e e n iden t i - these natural toxins. There is a need to explore these animal
fied.158−160,163,168,176,222−226,231−234,236−238,245,247−249,259,260 Of models using levels of dehydroPAs equivalent to those expected
these, ten cases159,160,176,223,237,238,245,247−249 display at least from current levels and patterns of food contamination in at-
four of the suggested indicators, nine risk communities. Congenital anomalies in fetuses and
others158,163,222,224,225,232,233,258,260 have three indicators, and neonates, caused by dehydroPA at levels equivalent to those
five other cases168,226,231,234,236 have two of the suggested currently occurring in the food supply, also need to be explored
indicators. Some of these cases also showed other features, in animal models, as does the apparent role of defective BMP
including hypertrophic hepatocytes (ballooned, swel- signaling as a common mechanism in, and cause of, several
ling),222,232,233,237,238 inconsistent veno-occlusive disease possible dehydroPA-associated diseases.
12 DOI: 10.1021/tx500403t
Chem. Res. Toxicol. 2015, 28, 4−20
Chemical Research in Toxicology


Perspective

AUTHOR INFORMATION (15) Kleiman, R., Rentz, E. D., Teshale, E., Thompson, N., and
Schurz-Rogers, H. (2008) Update on research and activities at the
Corresponding Author Centers for Disease Control and Prevention, and the Agency for Toxic
*Tel: +61 2 9490 5000; Fax: +61 2 9490 8499; E-mail: john. Substances and Disease Registry. J. Med. Toxicol. 4, 197−200.
edgar@csiro.au. (16) Kakar, F., Akbarian, Z., Leslie, T., Mustafa, M. L., Watson, J., van
Notes Egmond, H. P., Omar, M. F., and Mofleh, J. (2010) An outbreak of
hepatic veno-occlusive disease in western Afghanistan associated with
The authors declare no competing financial interest. exposure to wheat flour contaminated with pyrrolizidine alkaloids. J.

■ ABBREVIATIONS
HSOS, hepatic sinusoidal obstruction syndrome; dehydroPAs,
Toxicol. 2010, 313280.
(17) Edgar, J. A. (2003) Pyrrolizidine alkaloids and food safety.
Chem. Aust. 70, 4−7.
(18) Food Standards Australia New Zealand (FSANZ) (2001)
1,2-dehydropyrrolizidine ester alkaloids; PAH, pulmonary Pyrrolizidine alkaloids in food. A toxicological review and risk assessment,
arterial hypertension; bw, body weight; CYP450, cytochrome Tech. Report series No. 2, FSANZ, Canberra, Australia, http://www.
P450 monoamine oxidases; DHP, 1-hydroxymethyl-7-hydroxy- foodstandards.gov.au/publications/documents/TR2.pdf.
6,7-dihydropyrrolizine; GSH, glutathione; MMPs, metallopro- (19) Azadbakht, M., and Talavaki, M. (2003) Qualitative and
teinases; POPH, portopulmonary hypertension; BMPR2, bone quantitative determination of pyrrolizidine alkaloids of wheat and flour
morphogenic protein receptor II; VODI, hepatic veno-occlusive contaminated with Senecio in Mazandaran Province farms. Iran. J.
disease with immunodeficiency; RMS, rhabdomyosarcoma; Pharm. Res. 2, 179−183.
SCT, allogeneic stem cell transplantation; MIOP, malignant (20) Edgar, J. A., Colegate, S. M., Boppré, M., and Molyneux, R. J.
infantile osteopetrosis; PID, primary immunodeficiency; SIDs, (2011) Pyrrolizidine alkaloids in food: a spectrum of potential health
consequences. Food Addit. Contam. 28, 308−324.
severe immune dysregulatory disorders; FOP, fibrodysplasia
(21) Bodi, D., Ronczka, S., Gottschalk, C., Behr, N., Skibba, A.,
ossification progressive; BWS, Beckwith-Wiedemann Syndrome


Wagner, M., Lahrssen-Wiederholt, M., Preiss-Weigert, A., and These,
A. (2014) Determination of pyrrolizidine alkaloids in tea, herbal drugs
REFERENCES and honey. Food Addit. Contam. 31, 1886−1895.
(1) Willmot, F. C., and Robertson, G. W. (1920) Senecio disease, or (22) Dickinson, J. O., and King, R. R. (1978) The transfer of
cirrhosis of the liver due to Senecio poisoning. Lancet, 848−849. pyrrolizidine alkaloids from Senecio jacobaea into milk of lactating cows
(2) Steyn, D. G. (1933) Poisoning of human beings by weeds and goats, in Effects of Poisonous Plants on Livestock (Keeler, I.,
contained in cereals (Bread Poisoning). Onderstepoort J. Vet. Sci. Anim. VanKampen, K. R., and James, L. F., Eds.) pp 201−206, Academic
Ind. 1, 219−266. Press, New York.
(3) Selzer, G., and Parker, R. G. F. (1951) Senecio poisoning as (23) Deinzer, M. L., Thomson, P. A., Burgett, D. M., and Isaacson, D.
Chiari’s syndrome. A report on twelve cases. Am. J. Pathol. 27, 885− L. (1977) Pyrrolizidine alkaloids: their occurrence in honey from tansy
907. ragwort (Senecio jacobaea L.). Science 195, 497−499.
(4) Bull, L. B., Culvenor, C. C. J., and Dick, A. J. (1968) The (24) Culvenor, C. C. J., Edgar, J. A., and Smith, L. W. (1981)
Pyrrolizidine Alkaloids. Their Chemistry, Pathogenicity and Other Pyrrolizidine alkaloids in honey from Echium plantagineum L. J. Agric.
Biological Properties, North Holland Publishing Co., Amsterdam, The Food Chem. 29, 958−960.
Netherlands. (25) Lüthy, J., Heim, T., and Schlatter, C. (1983) Transfer of
(5) Tandon, B. N., Tandon, R. K., Tandon, H. D., Narndranathan, [3H]pyrrolizidine alkaloids from Senecio vulgaris L. and metabolites
M., and Joshi, Y. K. (1976) An epidemic of veno-occlusive disease of into rat milk and tissues. Toxicol. Lett. 17, 283−288.
liver in central India. Lancet 2, 271−272. (26) Molyneux, R. J., and James, L. F. (1990) Pyrrolizidine alkaloids
(6) Tandon, R. K., Tandon, B. N., Tandon, H. D., Bhatia, M. L., in milk: thresholds of in toxication. Vet. Hum. Toxicol. 32 (Suppl.),
Bhargava, S., Lal, P., and Arora, R. R. (1976) Study of an epidemic of 94S−103S.
venoocclusive disease in India. Gut 17, 849−855. (27) Edgar, J. A., and Smith, L. W. (2000) Transfer of pyrrolizidine
(7) Mohabbat, O., Younos, M. S., Merzad, A. A., Srivastava, R. N., alkaloids into eggs: food safety implications, in Natural and Selected
Sediq, G. G., and Aram, G. N. (1976) An outbreak of hepatic veno- Synthetic Toxins, Biological Implications. (Tu, A. T., and Gaffield, W.,
occlusive disease in North-Western Afghanistan. Lancet 308, 269−271. Eds.) ACS Symposium Series 745, Chapter 8, pp 118−128, American
(8) International Programme on Chemical Safety (IPCS) (1988) Chemical Society, Washington, DC.
Pyrrolizidine alkaloids: Environmental Health Criteria 80, WHO, (28) Beales, K. A., Betteridge, K., Colegate, S. M., and Edgar, J. A.
Geneva, Switzerland, www.inchem.org/documents/ehc/ehc/ehc080. (2004) Solid phase extraction and LCMS analysis of pyrrolizidine
htm. alkaloids in honeys. J. Agric. Food Chem. 52, 6664−6672.
(9) International Programme on Chemical Safety (IPCS) (1989) (29) Beales, K, Betteridge, K, Boppré, M, Cao, Y, Colegate, S. M.,
Pyrrolizidine alkaloids: health and safety guide no. 26; WHO, Geneva, Edgar, J. A. (2007) Hepatotoxic pyrrolizidine alkaloids and their N-
Switzerland, http://www.inchem.org/documents/hsg/hsg/hsg026. oxides in honey and pollen, in Poisonous Plants: Global Research and
htm. Solutions (Panter, K., Wierenga, T., and Pfister, J., Eds.) Chapter 16, pp
(10) Chauvin, P., Dillon, J., Moren, A., Talbak, S., and Barakaev, S. 94−100, CABI, Wallingford.
(1993) Heliotrope poisoning in Tadjikistan. Lancet 341, 1663. (30) Bundesamt für Risiokobewertung (BfR) (2007) Salatmischung
(11) Chauvin, P., Dillon, J. C., and Moren, A. (1994) An outbreak of mit pyrrolizidinalkaloid-haltigem Greiskraut verunreinigt, Stellungnahme
heliotrope food poisoning, Tadjikistan, November 1992−March 1993. Nr.028/2007 des BfR vom 10, http://www.bfr.bund.de/cm/208/
Sante 4, 263−268. salatmischung_mit_pyrrolizidinalkaloid_haltigem_geiskraut_
(12) Mayer, F., and Lüthy, J. (1993) Heliotrope poisoning in verunreinigt.pdf.
Tadjikistan. Lancet 342, 246−247. (31) Bundesamt für Risiokobewertung (BfR) (2011) Chemical
(13) Altaee, M. Y., and Mahmood, M. H. (1998) An outbreak of analysis and toxicity of pyrrolizidine alkaloids and assessment of the
veno-occlusive disease of the liver in northern Iraq. East. Mediterr. health risk posed by their occurrence in honey, BfR opinion no. 038/2011
Health J. 4, 142−148. 11, http://www.bfr.bund.de/cm/349/chemical-analysis-and-toxicity-
(14) Integrated Regional Information Networks (IRIN) (2008) of-pyrrolizidine-alkaloids-and-assessment-of-the-health-risks-posed-by-
Afghanistan: “Charmak” disease still killing people, livestock in west, UN their-occurence-in-honey.pdf.
Office for the Coordination of Humanitarian Affairs, http://www. (32) Bundesamt für Risiokobewertung (BfR) (2013) Pyrrolizidine
irinnews.org/Report.aspx?ReportId=81971. alkaloids in herbal teas and teas, BfR opinion no. 018/2013, http://

13 DOI: 10.1021/tx500403t
Chem. Res. Toxicol. 2015, 28, 4−20
Chemical Research in Toxicology Perspective

www.bfr.bund.de/cm/349/pyrrolizidine-alkaloids-in-herbal-teas-and- defense system in separate angiosperm lineages. Plant Cell 16, 2772−
teas.pdf. 2784.
(33) Kempf, M., Heil, S., Hasslauer, I., Schmidt, L., von der Ohe, K., (52) Mattocks, A. R. (1986) Chemistry and Toxicology of Pyrrolizidine
Theuring, C., Reinhard, A., Scheier, P., and Beuerle, T. (2010) Alkaloids, Academic Press, London, UK.
Pyrrolizidine alkaloids in pollen and pollen products. Mol. Nutr. Food (53) Mattocks, A. R. (1968) Toxicity of pyrrolizidine alkaloids.
Res. 54, 1−9. Nature 217, 723−728.
(34) Hoogenboom, L. A. P., Mulder, P. P. J., Zeilmaker, M. J., van (54) Culvenor, C. C. J., Downing, D. T., Edgar, J. A., and Jago, M. V.
den Top, H. J., Remmelink, G. J., Brandon, E. F. A., Klijnstra, M., (1969) Pyrrolizidine alkaloids as alkylating and antimitotic agents.
Meijer, G. A. L., Schothorst, R., and van Egmond, H. P. (2011) Carry- Ann. N.Y. Acad. Sci. 163, 837−847.
over of pyrrolizidine alkaloids from feed in dairy cows. Food Addit. (55) Jago, M. V., Edgar, J. A., Smith, L. W., and Culvenor, C. C. J.
Contam. 28, 359−372. (1970) Metabolic conversion of heliotridine-based pyrrolizidine
(35) Dübecke, A., Beckh, G., and Lüllmann, C. (2011) Pyrrolizidine alkaloids to dehydroheliotridine. Mol. Pharmacol. 6, 402−406.
alkaloids in honey and bee pollen. Food Addit. Contam. 28, 348−358. (56) Butler, W. H., Mattocks, A. R., and Barnes, J. M. (1970) Lesions
(36) Fletcher, M. T., McKenzie, R. A., Reichmann, K. G., and Blaney, in the liver and lungs of rats given pyrrole derivatives of pyrrolizidine
B. J. (2011) Risks from plants containing pyrrolizidine alkaloids for alkaloids. J. Pathol. 100, 169−175.
livestock and meat quality in northern Australia, in Poisonings by Plants, (57) Williams, D. E., Reed, R. L., Kedzierski, B., Dannan, G. A.,
Mycotoxins and Related Toxins (Riet-Correa, F., Pfister, J., Schild, A. L., Guengerich, F. P., and Buhler, D. R. (1989) Bioactivation and
and Wierenga, T., Eds.) pp 208−218, CAB International, Wallingford, detoxication of the pyrrolizidine alkaloid senecionine by cytochrome
UK. P-450 enzymes in rat liver. Drug Metab. Dispos. 17, 387−392.
(37) Kempf, M., Wittig, M., Schönfeld, K., Cramer, L., Schreier, P., (58) Miranda, C. L., Reed, R. L., Guengerich, F. P., and Buhler, D. R.
and Beuerle, T. (2011) Pyrrolizidine alkaloids in food: downstream (1991) Role of cytochrome P450IIIA4 in the metabolism of the
contamination in the food chain caused by honey and pollen. Food pyrrolizidine alkaloid senecionine in human liver. Carcinogenesis 12,
Addit. Contam. 28, 325−331. 515−519.
(38) Kempf, M., Wittig, M., Reinhard, A., von der Ohe, K., (59) Hincks, J. R., Kim, H.-Y., Segall, H. J., Molyneux, R. J., Stermitz,
Blacquiére, T., Raezke, K.-P., Michel, R., Scheier, P., and Beuerle, T. F. R., and Coulombe, R. A. (1991) DNA cross-linking in mammalian
(2011) Pyrrolizidine alkaloids in honey: comparison of analytical cells by pyrrolizidine alkaloids: structure−activity relationships.
methods. Food Addit. Contam. 28, 332−347. Toxicol. Appl. Pharmacol. 111, 90−98.
(39) Cao, Y., Colegate, S. M., and Edgar, J. A. (2013) Persistence of (60) Chung, W. G., and Buhler, D. R. (1994) The effect of
spironolactone treatment on the cytochrom P450-mediated metabo-
echimidine, a hepatotoxic pyrrolizidine alkaloid, from honey into
lism of the pyrrolizidine alkaloid senecionine by hepatic microsomes
mead. J. Food Compos. Anal. 29, 106−109.
from rats and guinea pigs. Toxicol. Appl. Pharmacol. 127, 314−319.
(40) Griffin, C. T., Danaher, M., Elliott, C. T., Kennedy, D. G., and
(61) Chung, W. G., Miranda, C. L., and Buhler, D. R. (1995) A
Furey, A. (2013) Detection of pyrrolizidine alkaloids in commercial
cytochrome P450B form is the major bioactivation enzyme for the
honey using liquid chromatography-ion trap mass spectrometry. Food
pyrrolizidine alkaloid senecionine in guinea pig. Xenobiotica 25, 929−
Chem. 136, 1577−1583.
939.
(41) Edgar, J. A., Roeder, E., and Molyneux, R. J. (2002) Honey from
(62) Kim, H.-Y., Stermitz, F. R., and Coulombe, R. A. (1995)
plants containing pyrrolizidine alkaloids: a potential threat to health. J. Pyrrolizidine alkaloid-induced DNA-protein cross-links. Carcinogenesis
Agric. Food Chem. 50, 2719−2730. 16, 2691−2697.
(42) Committee on Toxicity of Chemicals in Food, Consumer (63) Kasahara, Y., Kiyatake, K., Tatsumi, K., Sugito, K., Kakusaka, I.,
Products and the Environment (COT) (2008) COT statement on Yamagata, S., Ohmori, S., Kitada, M., and Kuriyama, T. (1997)
pyrrolizidine alkaloids in food, http://cot.food.gov.uk/pdfs/ Bioactivation of monocrotaline by P450 3A in rat liver. J. Cardiovasc.
cotstatementpa200806.pdf. Pharmacol. 30, 124−129.
(43) WHO (2011) Discussion paper on pyrrolizidine alkaloids, Joint (64) Reid, M. J., Lamé, M. W., Morin, D., Wilson, D. W., and Segall,
FAO/WHO food standards programme, Codex committee on H. J. (1998) Involvement of cytochrome P450 3A in the metabolism
contaminants in foods, 5th session, The Hague, The Netherlands, and covalent binding of 14C-monocrotaline in rat liver microsomes. J.
21−25 March, ftp://ftp.fao.org/codex/meetings/cccf/cccf5/cf05_14e. Biochem. Mol. Toxicol. 12, 157−166.
pdf. (65) Pereira, T. N., Webb, R. I., Reilly, P. E. B., Seawright, A. A., and
(44) European Food Safety Authority (EFSA) Panel on Prakash, A. S. (1998) Dehydromonocrotaline generates sequence-
Contaminants in the Food Chain (CONTAM) (2011) Scientific selective N-7 guanine alkylation and heat and alkali stable multiple
opinion on pyrrolizidine alkaloids in food and feed. EFSA J. 9, 2406− fragment DNA crosslinks. Nucl. Acid Res. 26, 5441−5447.
2540. (66) Tepe, J. J., and Williams, R. M. (1999) DNA cross-linking by a
(45) The U.S. National Toxicology Program (2011) 12th Report on phototriggered dehydromonocrotaline progenitor. J. Am. Chem. Soc.
Carcinogens (RoC), pp iii−499, U.S. Department of Health and Human 121, 2951−2955.
Services, Washington, DC. (67) Lin, G., Cui, Y. Y., and Hawes, E. M. (2000) Characterization of
(46) Chen, T., Mei, N., and Fu, P. P. (2010) Genotoxicity of rat liver microsomal metabolites of clivorine, an hepatotoxic
pyrrolizidine alkaloids. J. Appl. Toxicol. 30, 183−196. otonecine-type pyrrolizidine alkaloid. Drug Metab. Dispos. 28, 1475−
(47) Edgar, J. A. (2014) Food contaminants capable of causing 1483.
cancer, pulmonary hypertension and cirrhosis. Med. J. Aust. 200, 73− (68) Fu, P. P., Xia, Q., Lin, G., and Chou, M. W. (2004) Pyrrolizidine
74. alkaloidsgenotoxicity, metabolism enzymes, metabolic activation,
(48) Green, C. R., and Christie, G. S. (1961) Malformation in foetal and mechanisms. Drug Metab. Rev. 36, 1−55.
rats induced by the pyrrolizidine alkaloid, heliotrine. Br. J. Exp. Pathol. (69) Fu, P. P., Xia, Q., Lin, G., and Chou, M. W. (2002) Genotoxic
42, 369−378. pyrrolizidine alkaloidsmechanism leading to DNA adduct formation
(49) Peterson, J. E., and Jago, M. V. (1980) Comparison of the toxic and tumorigenicity. Int. J. Mol. Sci. 3, 948−964.
effects of dehydroheliotridine and heliotrine in pregnant rats and their (70) Yang, Y.-C., Yan, J., Doerge, D. R., Chan, P.-C., Fu, P. P., and
embryos. J. Pathol. 131, 339−355. Chou, M. W. (2001) Metabolic activation of the tumorigenic
(50) Smith, L. W., and Culvenor, C. C. J. (1981) Plant sources of pyrrolizidine alkaloid, riddelliine, leading to DNA adduct formation
hepatotoxic pyrrolizidine alkaloids. J. Nat. Prod. 44, 129−152. in vivo. Chem. Res. Toxicol. 14, 101−109.
(51) Reimann, A., Nurhayati, N., Backenköhler, A., and Ober, D. (71) Yang, Y.-C., Yan, J., Churchwell, M., Beger, R., Chan, P.-C.,
(2004) Repeated evolution of the pyrrolizidine alkaloid-mediated Doerge, D. R., Fu, P. P., and Chou, M. W. (2001) Development of a

14 DOI: 10.1021/tx500403t
Chem. Res. Toxicol. 2015, 28, 4−20
Chemical Research in Toxicology Perspective
32
P-postlabeling/HPLC method for detection of dehydroretronecine- excretion, and transfer into milk of lactating mice. Drug Metab. Dispos.
derived DNA adducts in vivo and in vitro. Chem. Res. Toxicol. 14, 91− 10, 236−240.
100. (91) Chou, M. W., and Fu, P. P. (2006) Formation of DHP-derived
(72) Xia, Q., Chou, M. W., Kadlubar, F. F., Chan, P., and Fu, P. P. DNA adducts in vivo from dietary supplements and Chinese herbal
(2003) Human liver microsomal metabolism and DNA adduct plant extracts containing carcinogenic pyrrolizidine alkaloids. Toxicol.
formation of the tumorigenic pyrrolizidine alkaloid, riddelliine. Ind. Health 22, 321−327.
Chem. Res. Toxicol. 16, 66−73. (92) Hsu, I. C., Allen, J. R., and Chesney, C. F. (1973) Identification
(73) Chou, M. W., Yan, J., Nichols, J., Xia, Q., Beland, F. A., Chan, P. and toxicological effects of dehydroretronecinemetabolite of
C., and Fu, P. P. (2003) Correlation of DNA adduct formation and monocrotaline. Proc. Soc. Exp. Biol. Med. 144, 834−838.
riddelliine-induced liver tumorigenesis in F344 rats and B6C3F1 mice. (93) Hsu, I. C., Robertson, K. A., Shumaker, R. C., and Allen, J. R.
Cancer Lett. 193, 119−125. (1975) Binding of tritiated dehydroretronecine to macromolecules.
(74) Xia, Q., Chou, M. W., Lin, G., and Fu, P. P. (2004) Metabolic Res. Commun. Chem. Pathol. Pharmacol. 11, 99−106.
formation of DHP-derived DNA adducts from a representative (94) Curtain, C. C., and Edgar, J. A. (1976) The binding of
otonecine type pyrrolizidine alkaloid clivorine and the extract of dehydroheliotridine to DNA and the effect of it and other compounds
Ligularia hodgsonnii Hook. Chem. Res. Toxicol. 17, 702−708. on repair synthesis in main and satellite band DNA. Chem.−Biol.
(75) Xia, Q., Chou, M. W., Edgar, J. A., Doerge, D. R., and Fu, P. P. Interact. 13, 243−256.
(2006) Formation of DHP-derived DNA adducts from metabolic (95) Sun, P. S., Hsia, M-T. S., Chu, F. S., and Allen, J. R. (1977)
activation of the prototype heliotridine-type pyrrolizidine alkaloid, Inhibition of yeast alcohol dehydrogenase by dehydroretronecine.
lasiocarpine. Cancer Lett. 231, 138−145. Food Cosmet. Toxicol. 15, 419−422.
(76) Mattocks, A. R., and Bird, I. (1983) Pyrrolic and N-oxide (96) Seawright, A. A. (1992) Potential toxicity problems with herbal
metabolites formed from pyrrolizidine alkaloids by hepatic micro- medicines and foodnew observations with pyrrolizidine alkaloids.
somes in vitrorelevance to in vivo hepatotoxicity. Chem.−Biol. Proc. Nutr. Soc. Aust. 17, 20−25.
Interact. 43, 209−222. (97) Seawright, A. A. (1994) Toxic plant residues in meat, in Plant-
(77) Bull, L. B., and Dick, A. T. (1959) The chronic pathological Associated Toxins; Agricultural, Phytochemical and Ecological Aspects
effects on the liver of the rat of the pyrrolizidine alkaloids heliotrine, (Colegate, S. M., and Dorling, P. R., Eds.) Chapter 15, pp 77−84, CAB
lasiocarpine and their N-oxides. J. Pathol. Bacteriol. 78, 483−502. International, Wallingford UK.
(78) Mattocks, A. R. (1971) Hepatotoxic effects due to pyrrolizidine (98) Hsu, I. C., Schumaker, R. C., and Allen, J. R. (1974) Tissue
alkaloid N-oxides. Xenobiotica 1, 563−565. distribution of tritium-labeled dehydroretronecine. Chem.−Biol.
(79) Molyneux, R. J., Johnson, A. E., Olson, J. D., and Baker, D. C. Interact. 8, 163−170.
(1991) Toxicity of pyrrolizidine alkaloids from Riddell groundsel (99) Hsu, I. C., Robertson, K. A., and Allen, J. R. (1976) Tissue
(Senecio riddellii) to cattle. Am. J. Vet. Res. 52, 146−151. distribution, binding properties and lesions produced by dehydrore-
(80) Chou, M. W., Wang, J. Y., Yang, Y.-C., Beger, R. D., Williams, L. tronecine in the non-human primate. Chem.−Biol. Interact. 12, 19−28.
D., Doerge, D. R., and Fu, P. P. (2003) Riddelliine N-oxide is a (100) Shumaker, R. C., Hsu, I. C., and Allen, J. R. (1976)
phytochemical and mammalian metabolite with genotoxic activity that Localisation and tissue effects of tritiated dehydroretronecine in young
is comparable to the parent pyrrolizidine alkaloid riddelliine. Toxicol. rats. J. Pathol. 119, 21−28.
Lett. 145, 239−247. (101) Mattocks, A. R., and White, I. N. H. (1976) The distribution of
(81) Wang, Y.-P., Yan, J., Fu, P. P., and Chou, M. W. (2005) Human [3H]synthanecine a bis-N-ethylcarbamate and its metabolites in the
liver microsomal reduction of pyrrolizidine alkaloid N-oxides to form rat. Chem.−Biol. Interact. 15, 173−184.
the corresponding carcinogenic parent alkaloid. Toxicol. Lett. 155, (102) Candrian, U., Lüthy, J., and Schlatter, C. (1985) In vivo
411−420. covalent binding of retronecine-labeled [3H] seneciphylline and [3H]
(82) Yan, J., Xia, Q., Chou, M. W., and Fu, P. P. (2008) Metabolic senecionine to DNA of rat liver, lung and kidney. Chem.−Biol. Interact.
activation of retronecine and retronecine-N-oxideformation of 54, 57−69.
DHP-derived DNA adducts. Toxicol. Ind. Health 24, 181−188. (103) Mattocks, A. R., and Jukes, R. (1990) Recovery of the pyrrolic
(83) Cao, Y., Colegate, S. M., and Edgar, J. A. (2008) Safety nucleus of pyrrolizidine alkaloid metabolites from sulphur conjugates
assessment of food and herbal products containing hepatotoxic in tissues and body fluids. Chem.−Biol. Interact. 75, 225−239.
pyrrolizidine alkaloids: inter-laboratory consistency and the impor- (104) Allen, J. R., Hsu, I.-C., and Carstens, L. A. (1975)
tance of N-oxide determination. Phytochem. Anal. 19, 526−533. Dehydroretronecine induced rhabdomyosarcomas in rats. Cancer Res.
(84) Mattocks, A. R. (1968) Iron (II)-catalysed conversion of 35, 997−1002.
unsaturated pyrrolizidine alkaloid N-oxides to pyrrole derivatives. (105) Shumaker, R. C., Robertson, K. A., Hsu, I. C., and Allen, J. R.
Nature 219, 480. (1976) Neoplastic transformation in tissues of rats exposed to
(85) Culvenor, C. C. J., Edgar, J. A., Smith, L. W., and Tweedale, H. J. monocrotaline or dehydroretronecine. J. Natl. Cancer Inst. 56, 787−
(1969) Dihydropyrrolizine analogues of pyrrolizidine alkaloids. Aust. J. 789.
Chem. 41, 3599−3602. (106) Peterson, J. E., Jago, M. V., Reddy, J. K., and Jarrett, R. G.
(86) Mattocks, A. R. (1969) Dihydropyrrolizine derivatives from (1983) Neoplasia and chronic disease associated with the prolonged
unsaturated pyrrolizidine alkaloids. J. Chem. Soc. C, 1155−1162. administration of dehydroheliotridine to rats. J. Natl. Cancer Inst. 70,
(87) Culvenor, C. C. J., Edgar, J. A., Smith, L. W., and Tweedale, H. J. 381−386.
(1970) Dihydropyrrolizines III. Preparation and reactions of (107) Percy, J., and Pierce, A. E. (1971) Immunosuppressive activity
derivatives related to pyrrolizidine alkaloids. Aust. J. Chem. 23, of the pyrrolizidine alkaloid metabolite dehydroheliotridine. Immunol-
1853−1867. ogy 21, 273−280.
(88) Mattocks, A. R., and Jukes, R. (1990) Trapping and (108) Peterson, J. E., Samuel, A., and Jago, M. V. (1972) Pathological
measurement of short-lived alkylating agents in a recirculating flow effects of dehydroheliotridine, a metabolite of heliotridine-based
system. Chem.−Biol. Interact. 76, 19−30. pyrrolizidine alkaloids in the young rat. J. Pathol. 107, 175−189.
(89) Mattocks, A. R., Driver, H. E., Barbour, R. H., and Robins, D. J. (109) Mattocks, A. R., and Bird, I. (1983) Alkylation by
(1986) Metabolism and toxicity of synthetic analogues of macrocyclic dehydroretronecine, a cytotoxic metabolite of some pyrrolizidine
diester pyrrolizidine alkaloids. Chem.−Biol. Interact. 58, 95−108. alkaloids: an in vivo test. Toxicol. Lett. 16, 1−8.
(90) Eastman, D. F., Dimenna, G. P., and Segall, H. J. (1982) (110) Prakash, A. S., Pereira, T. N., Reilly, P. E. B., and Seawright, A.
Covalent binding of two pyrrolizidine alkaloids, senecionine and A. (1999) Pyrrolizidine alkaloids in human diet. Mutat. Res. 443, 53−
seneciphylline, to hepatic macromolecules and their distribution, 67.

15 DOI: 10.1021/tx500403t
Chem. Res. Toxicol. 2015, 28, 4−20
Chemical Research in Toxicology Perspective

(111) Hooper, P. T. (1975) Experimental acute gastrointestinal non-human primates (Macaca arctoides). Am. J. Vet. Res. 34, 1577−
disease caused by the pyrrolizidine alkaloid, lasiocarpine. J. Comp. 1581.
Pathol. 85, 341−349. (132) Chesney, C. F., and Allen, J. R. (1973) Monocrotaline induced
(112) Wong, L. S.N., Lamé, M. W., Jones, A. D., and Wilson, D. W. pulmonary vascular lesions in non-human primates. Cardiovasc. Res. 7,
(2010) Differential cellular responses to protein adducts of 508−518.
naphthoquinone and monocrotaline pyrrole. Chem. Res. Toxicol. 23, (133) Fishman, A. P. (1974) Dietary pulmonary hypertension. Circ.
1504−1513. Res. 35, 657−660.
(113) Molyneux, R. J., Johnson, A. E., and Stuart, L. D. (1988) (134) Pan, L. C., Wilson, D. W., Lamé, M. W., Jones, A. D., and
Delayed manifestation of Senecio-induced pyrrolizidine alkaloidosis in Segall, H. J. (1993) Cor pulmonale is caused by monocrotaline and
cattle: case reports. Vet. Hum. Toxicol. 30, 201−205. dehydromonocrotaline, but not by glutathione or cysteine conjugates
(114) DeLeve, L. D., Wang, X., Kuhlenkamp, J. F., and Kaplowitz, N. of dihydropyrrolizine. Toxicol. Appl. Pharmacol. 118, 87−97.
(1996) Toxicity of azathioprine and monocrotaline in murine (135) Sehgal, P. B., and Mukhopadhyay, S. (2007) Dysfunctional
sinusoidal endothelial cells and hepatocytes: the role of glutathione intracellular trafficking in the pathobiology of pulmonary arterial
and relevance to hepatic venooclusive disease. Hepatology 23, 589− hypertension. Am. J. Respir. Cell Mol. Biol. 37, 31−37.
599. (136) Stenmark, K. R., Meyrick, B., Galie, N., Mooi, W. J., and
(115) DeLeve, L. D., McCuskey, R. S., Wang, X., Hu, L., McCuskey, McMurtry, I. F. (2009) Animal models of pulmonary arterial
M. K., Epstein, R. B., and Kanel, G. C. (1999) Characterization of a hypertension: the hope for etiological discovery and pharmacological
reproducible rat model of hepatic veno-occlusive disease. Hepatology cure. Am. J. Physiol.: Lung Cell. Mol. Physiol. 297, L1013−1032.
29, 1779−1791. (137) Yen, C. H., Tsai, T. H., Leu, S., Chen, Y. L., Chang, L. T., Chai,
(116) DeLeve, L. D., Shulman, H. M., and McDonald, G. B. (2002) H. T., Chung, S. Y., Chua, S., Tsai, C. Y., Chang, H. W., Ko, S. F., Sun,
Toxic injury to hepatic sinusoids: sinusoidal obstruction syndrome C. K., and Yip, H. K. (2013) Sildenafil improves long term effect of
(veno-occlusive disease). Semin. Liver Dis. 22, 27−41. endothelial progenitor cell-based treatment for monocrotaline-induced
(117) DeLeve, L. D., Ito, Y., Bethea, N. W., McCuskey, M. K., Wang, rat pulmonary arterial hypertension. Cytotherapy 15, 209−223.
X., and McCuskey, R. S. (2003) Embolization by sinusoidal lining cells (138) Gupta, V., Gupta, N., Shaik, I. H., Mehvar, R., Nozik-Grayck,
obstructs the microcirculation in rat sinusoidal obstruction syndrome. E., McMurtry, I. F., Oka, M., Komatsu, M., and Ahsan, F. (2013)
Am. J. Physiol.: Gastrointest. Liver Physiol. 284, G1045−G1052. Inhaled PLGA particles of prostaglandin E1 ameliorate symptoms and
(118) DeLeve, L. D., Valla, D.-C., and Garcia-Tsao, G. (2009) progression of pulmonary hypertension at a reduced dosing frequency.
Vascular disorders of the liver. Hepatology 49, 1729−1764. Mol. Pharmaceutics 10, 1655−1667.
(119) Huxtable, R. J. (1989) Human health implications of (139) Gomez-Arroyo, J. G., Farkas, L., Alhussaini, A. A., Farkas, D.,
pyrrolizidine alkaloids and herbs containing them, in Toxicants of Kraskauskas, D., Voelkel, N. F., and Bogaard, H. J. (2012) The
Plant Origin (Cheeke, P. R., Ed). Vol. 1, Chapter 3, pp 42−86, CRC monocrotaline model of pulmonary hypertension in perspective. Am. J.
Press, Inc., Boca Raton, FL. Physiol.: Lung Cell. Mol. Physiol. 302, L363−369.
(120) Nolan, J. P., Scheig, R. L., and Klatskin, G. (1966) Delayed (140) Kay, J. M., Heath, D., Smith, P., Bras, G., and Summerell, J.
hepatitis and cirrhosis in weanling rats following a single small dose of (1971) Fulvine and pulmonary circulation. Thorax 26, 249−261.
the Senecio alkaloid, lasiocarpine. Am. J. Pathol. 49, 129−151. (141) Heath, D., Shaba, J., Williams, A., Smith, P., and Kombe, A.
(121) Gordon, G. J., Coleman, W. B., and Grisham, J. W. (2000) (1975) A pulmonary hypertension-producing plant from Tanzania.
Temporal analysis of hepatocyte differentiation by small hepatocyte- Thorax 30, 399−404.
like progenitor cells during liver regeneration in retrorsine-exposed (142) Györika, S., and Stricker, H. (2009) Severe pulmonary
rats. Am. J. Pathol. 157, 771−786. hypertension possibly due to pyrrolizidine alkaloids in polyphytother-
(122) Hanumegowda, U. M., Copple, B. L., Shibuya, M., Malle, E., apy. Swiss Med. Wkly. 139, 210−211.
Ganey, P. E., and Roth, R. A. (2003) Basement membrane and matrix (143) Couet, C. E., Hanley, A. B., MacDonald, S., and Mayes, L.
metalloproteinases in monocrotaline-induced liver injury. Toxicol. Sci. (1993) The biological assay of natural mutagens using p53, in Food
76, 237−246. and Cancer Prevention (Waldron, K. W., Johnston, I. T., and Fenwick,
(123) Wang, X., Kanel, G. C., and DeLeve, L. D. (2000) Support of G. R., Eds.) pp 426−428, Royal Society of Chemistry, London.
sinusoidal endothelial cell glutathione prevents hepatic veno-occlusive (144) Ji, L., Zhang, M., Sheng, Y., and Wang, Z. (2005) Pyrrolizidine
disease in the rat. Hepatology 31, 428−434. alkaloid clivorine induces apoptosis in human normal liver L-20 cells
(124) Ridker, P. M., Ohkuma, S., McDermott, W. V., Trey, C., and and reduces the expression of p53 protein. Toxicol. In Vitro 19, 41−46.
Huxtable, R. J. (1985) Hepatic venooclusive disease associated with (145) Hirono, I. (1993) Edible plants containing naturally occurring
the consumption of pyrrolizidine-containing dietary supplements. carcinogens in Japan. Jpn. J. Cancer Res. 84, 997−1006.
Gastroenterology 88, 1050−1054. (146) Chan, P. C., Haseman, J. K., Prejean, J. D., and Nyska, A.
(125) Rasenack, R., Müller, C., Kleinschmidt, M., Rasenack, J., and (2003) Toxicity and carcinogenicity of riddelliine in rats and mice.
Wiedenfeld, H. (2003) Veno-occlusive disease in a fetus caused by Toxicol. Lett. 144, 295−311.
pyrrolizidine alkaloids of food origin. Fetal Diagn. Ther. 18, 223−225. (147) Malkin, D. (2011) Li-Fraumeni syndrome. Genes Cancer 2,
(126) Kay, J. M., and Heath, D. (1966) Observations on the 475−484.
pulmonary arteries and heart weight of rats fed on Crotalaria spectabilis (148) LeTendre, L., Ludwig, J., Perrault, J., Smithson, W. A., and
seeds. J. Pathol. Bacteriol. 92, 385−394. Kovach, J. S. (1984) Hepatocellular toxicity during treatment of
(127) Heath, D., and Kay, J. M. (1967) Medial thickness of refractory acute leukemia with indicine N-oxide. Cancer 54, 1256−
pulmonary trunk in rats with cor pulmonale induced by ingestion of 1259.
Crotalaria spectabilis seeds. Cardiovasc. Res. 1, 74−79. (149) Miser, J. S., Smithson, W. A., Krivit, W., Hughes, C., Davis, D.,
(128) Kay, J. M., Harris, P., and Heath, D. (1967) Pulmonary Krailo, M., and Hammond, D. (1991) Phase II trial of indicine N-oxide
hypertension produced in rats by ingestion of Crotalaria spectabilis in relapsed pediatric solid tumors. Invest. New Drugs 9, 339−342.
seeds. Thorax 22, 176−179. (150) Rai, P. R., Cool, C. D., King, J. A. C., Stevens, T., Burns, N.,
(129) Burns, J. (1972) The heart and pulmonary arteries in rats fed Winn, R. A., Kasper, M., and Voelkel, N. F. (2008) The cancer
on Senecio jacobaea. J. Pathol. 106, 187−194. paradigm of severe pulmonary arterial hypertension. Am. J. Respir. Crit.
(130) Allen, J. R., and Chesney, C. F. (1972) Effect of age on Care Med. 178, 558−564.
development of cor pulmonale in non-human primates following (151) Schoental, R. (1968) Toxicology and carcinogenic action of
pyrrolizidine alkaloid intoxication. Exp. Mol. Pathol. 17, 220−232. pyrrolizidine alkaloids. Cancer Res. 28, 2237−2246.
(131) Chesney, C. F., and Allen, J. R. (1973) Endocardial fibrosis (152) McLean, E. K. (1970) The toxic actions of pyrrolizidine
associated with monocrotaline induced pulmonary hypertension in (Senecio) alkaloids. Pharmacol. Rev. 22, 429−483.

16 DOI: 10.1021/tx500403t
Chem. Res. Toxicol. 2015, 28, 4−20
Chemical Research in Toxicology Perspective

(153) Svoboda, D. J., and Reddy, J. K. (1972) Malignant tumors in (170) Barker, C. C., Butzner, J. D., Anderson, R. A., Brant, R., and
rats given lasiocarpine. Cancer Res. 32, 908−912. Sauve, R. S. (2003) Incidence, survival and risk factors for the
(154) Hirono, I., Shimizu, M., Fushimi, K., Mori, H., and Kato, K. development of veno-occlusive disease in pediatric hematopoietic stem
(1973) Carcinogenic activity of Petasites japonicus Maxim, a kind of cell transplant recipients. Bone Marrow Transplant. 32, 79−87.
coltsfoot. Gann Monogr. Cancer Res. 64, 527−528. (171) Kraemer, D. M., Waschke, J., and Kunzmann, V. (2003)
(155) Hirono, I., Mori, H., and Haga, M. (1978) Carcinogenic Venoocclusive disease in a male patient with Marfam syndrome and
activity of Symphytum of f icinale. J. Natl. Cancer Inst. 61, 865−869. common acute lymphoblastic leukemia during induction therapy. Ann.
(156) Kanwar, V. S., Albuquerque, M. L., Ribeiro, R. C., Kauffman, Hematol. 82, 444−447.
W. M., and Furman, W. L. (1995) Veno-occlusive disease of the liver (172) Bajwa, R. P. S., Cant, A. J., Abinun, M., Flood, T. J., Hodges, S.,
after chemotherapy for rhabdomyosarcoma: case report with a review Hale, J. P., and Skinner, R. (2003) Recombinant tissue plasminogen
of the literature. Med. Pediatr. Oncol. 24, 334−340. activator for treatment of hepatic veno-occlusive disease following
(157) Ortega, J. A., Donaldson, S. S., Ivy, S. P., Pappo, A., and bone marrow transplantation in children: effectiveness and a scoring
Maurer, H. M. (1997) Venoocclusive disease of the liver after system for initiating treatment. Bone Marrow Transplant. 31, 591−597.
chemotherapy with vincristine, actinomycin D, and cyclophosphamide (173) Kalayoglu-Besisik, S., Yenerel, M. N., Caliskan, Y., Ozturk, S.,
for the treatment of rhabdomyosarcoma. Cancer 79, 2435−2439. Besisik, F., and Sargin, D. (2005) Time-related changes in the
(158) Bisogno, G., de Kraker, J., Weirich, A., Masiero, L., Ludwig, R., incidence severity and clinical outcome of hepatic veno-occlusive
Tornade, M. F., and Carli, M. (1997) Veno-occlusive disease of the disease in hematopoietic stem cell transplantation patients during the
liver in children treated for Wilms tumor. Med. Pediatr. Oncol. 29, past ten years. Transplant. Proc. 37, 2285−2289.
245−251. (174) Papadopoulos, A., Ntaios, G., Kaiafa, G., Girtovitis, Z. S.,
(159) Ludwig, R., Weirich, A., Abel, U., Hofmann, W., Graf, N., and Kontoninas, Z., Diamantidis, M. D., Savopoulos, C., and Hatzitolios, A.
Tournade, M. F. (1999) Hepatotoxicity in patients treated according (2008) Veno-occlusive disease of the liver during induction therapy for
to the nephroblastoma trial and study SIOP-9/GPOH. Med. Pediatr. acute lymphoblastic leukemia. Int. J. Hematol. 88, 441−442.
Oncol. 33, 462−469. (175) Dulley, F. L., Kanfer, E. J., Appelbaum, F. R., Amos, D., Hill, R.
(160) Burkhardt, A., and Klöppel, A. (1977) Unusual obliterative S., Buckner, C. D., Shulman, H. M., McDonald, G. B., and Thomas, E.
disease of the hepatic veins in an infant. Virchows Arch. A: Pathol. Anat. D. (1987) Veno-occlusive disease of the liver after chemoradiotherapy
Histol. 375, 225−232. and autologous bone marrow transplantation. Transplantation 43,
(161) Rubbia-Brandt, L., Audard, V., Sartoretti, P., Roth, A. D., 870−873.
Brezault, C., Le Charpentier, M., Dousset, B., Morel, P., Soubrane, O., (176) Carreras, E. (2000) Veno-occlusive disease of the liver after
Chausade, S., Mentha, G., and Terris, B. (2004) Severe hepatic hemopoietic cell transplantation. Eur. J. Haematol. 64, 281−291.
sinusoidal obstruction associated with oxaliplatin-based chemotherapy (177) Kumar, S., DeLeve, L. D., Kamath, P. S., and Tefferi, A. (2003)
in patients with metastatic colorectal cancer. Ann. Oncol. 15, 460−466. Hepatic veno-occlusive disease (sinusoidal obstruction syndrome)
(162) Boula, A. M., Mantadakis, E., Xilouri, I. M., Christoforidou, A. after hematopoietic stem cell transplantation. Mayo Clin. Proc. 78,
V., Foudoulakis, A. M., and Samonis, G. (2005) Veno-occlusive disease 589−598.
of the liver associated with chronic myelomonocytic leukemia treated (178) Gharib, M. I., Bulley, S. R., Doyle, J. J., and Wynn, R. F. (2006)
with vincristine and standard doses of cytarabine. Am. J. Hematol. 79, Venous occlusive disease in children. Thromb. Res. 118, 27−38.
216−219. (179) Rubbia-Brandt, L., Tauzin, S., Brezault, C., Delucinge-Vivier,
(163) Elli, M., Pinarli, F. G., Dagdemir, A., and Acar, S. (2006) Veno- C., Descombes, P., Dousset, P. E., Mentha, G., and Terris, B. (2011)
occlusive disease of the liver in a child after chemotherapy for brain Gene expression profiling provides insights into pathways of
tumor. Pediatr. Blood Cancer 46, 521−523. oxaliplatin-related sinusoidal obstruction syndrome in humans. Mol.
(164) Matute-Bello, G., McDonald, G. D., Hinds, M. S., Schoch, H. Cancer Ther. 10, 687−696.
G., and Crawford, S. W. (1998) Association of pulmonary function (180) De Jonge, M. E., Huitema, A. D. R., Beijnen, J. H., and
testing abnormalities and severe veno-occlusive disease of the liver Rodenhuis, S. (2006) High exposures to bioactivated cyclophospha-
after marrow transplantation. Bone Marrow Transplant. 21, 1152− mide are related to the occurrence of veno-occlusive disease of the
1130. liver following high-dose chemotherapy. Br. J. Cancer 94, 1226−1230.
(165) McDonald, G. B., Hinds, M. S., Fisher, L. D., Schoch, H. G., (181) Plapp, F. V., and Chiga, M. (1972) Disaggregation of mouse
Wolford, J. L., Banaji, M., Hardin, B. J., Shulman, H. M., and Clift, R. liver polysomes by cyclophosphamide. Arch. Pharmacol 272, 351−353.
A. (1993) Veno-occlusive disease of the liver and multiorgan failure (182) Yan, C. C., and Huxtable, R. J. (1996) Effects of
after bone marrow transplantation: a cohort study of 355 patients. Ann. monocrotaline, a pyrrolizidine alkaloid, on glutathione metabolism in
Int. Med. 118, 255−267. the rat. Biochem. Pharmacol. 51, 375−379.
(166) Strasser, S. I., Sullivan, K. M., Myerson, D., Spurgeon, C. L., (183) Hettmer, S., and Wagers, A. J. (2010) Uncovering the origins
Storer, B., Schoch, H. G., Murakami, C. S., and McDonald, G. B. of rhabdomyosarcoma. Nat. Med. 16, 171−173.
(1999) Cirrhosis of the liver in long-term marrow transplant survivors. (184) Daughton, C. G. (2005) Overlooked in Fallon? Environ. Health
Blood 93, 3259−3266. Perspect. 113, A224.
(167) Copelan, E. A., Penza, S. L., Elder, P. J., Ezzone, S. A., Scholl, (185) Morty, R. E., Nejman, B., Kwapiszewska, G., Hecker, M.,
M. D., Bechtel, T. P., Belt, P. S., and Avalos, B. R. (2000) Analysis of Zakrzewicz, A., Kouri, F. M., Peters, D. M., Dumitrascu, R., Seeger, W.,
prognostic factors for allogenic marrow transplantation following Knaus, P., Schermuly, R. T., and Eickelberg, O. (2007) Dysregulated
busulfan and cyclophosphamide in myelodysplastic syndrome and bone morphogenetic Protein signaling in monocrotaline-induced
after leukemic transformation. Bone Marrow Transplant. 25, 1219− pulmonary arterial hypertension. Arterioscler., Thromb., Vasc. Biol. 27,
1222. 1072−1078.
(168) Horn, B., Reiss, U., Matthay, K., McMillan, A., and Cowan, M. (186) Murakami, K., Mathew, R., Huang, J., Farahani, R., Peng, H.,
(2002) Veno-occlusive disease of the liver in children with solid Olson, S. C., and Etlinger, J. D. (2010) Smurf1 ubiquitin ligase causes
tumors undergoing autologous hematopoietic progentitor cell trans- downregulation of BMP receptors and is induced in monocrotaline
plantation: a high incidence in patients with neuroblastoma. Bone and hypoxia models of pulmonary arterial hypertension. Exp. Biol.
Marrow Transplant. 29, 409−415. Med. 235, 805−813.
(169) Bairey, O., Kirgner, I., Yakobi, M., Hamdan, A., Ben-Ari, Z., (187) Ramos, M., Lamé, M. W., Segall, H. J., and Wilson, D. W.
and Shaklai, M. (2002) Clinical severe hepatic venooclusive disease (2007) Monocrotaline pyrrole induces Smad nuclear accumulation
during induction treatment of acute monoblastic leukemia managed and altered signaling expression in human pulmonary arterial
with defibrotide. Am. J. Hematol. 69, 281−284. endothelial cells. Vasc. Pharmacol. 46, 439−448.

17 DOI: 10.1021/tx500403t
Chem. Res. Toxicol. 2015, 28, 4−20
Chemical Research in Toxicology Perspective

(188) Owens, G. K., Rabinovitch, P. S., and Schwartz, S. M. (1981) (207) Murata, K., Shimizo, A., Takase, K., Nakano, T., and Tameda,
Smooth muscle hypertrophy versus hyperplasia in hypertension. Proc. Y. (1997) Asymptomatic primary pulmonary hypertension associated
Natl. Acad. Sci. U.S.A. 78, 7759−7763. with liver cirrhosis. J. Gastroenterol. 32, 102−104.
(189) Veyssier-Belot, C., and Cacoub, P. (1999) Role of endothelial (208) Bragdon, B., Moseychuk, O., Saldanha, S., King, D., Julian, J.,
and smooth muscle cells in the physiopathology and treatment and Nohe, A. (2011) Bone morphogenetic proteins: a critical review.
management of pulmonary hypertension. Cardiovasc. Res. 44, 274− Cell. Signalling 23, 609−620.
282. (209) Okamoto, M., Murai, J., Yoshikawa, H., and Tsumali, N.
(190) Tajsic, T., and Morrell, N. W. (2011) Smooth muscle cell (2006) Bone morphogenetic proteins in bone stimulate osteoclasts
hypertrophy, proliferation, migration and apoptosis in pulmonary and osteoblasts during bone development. J. Bone Miner. Res. 21,
hypertension. Comp. Physiol. 1, 295−317. 1022−1033.
(191) Lappin, P. B., and Roth, R. A. (1997) Hypertrophy and (210) Steward, C. G., Pellier, I., Mahajan, A., Ashworth, M. T., Stuart,
prolonged DNA synthesis in smooth muscle cells characterize A. G., Fasth, A., Lang, D., Fischer, A., Friedrich, W., and Schulz, A. S.
pulmonary arterial wall thickening after monocrotaline pyrrole (2004) Severe pulmonary hypertension: a frequent complication of
administration. Toxicol. Pathol. 25, 372−380. stem cell transplantation for malignant infantile osteopetrosis. Br. J.
(192) Lappin, P. B., Ross, K. L., King, L. E., Fraker, P. J., and Roth, R. Hamaetol. 124, 63−71.
(211) Slatter, M. A., Rao, K., Amrolia, P., Flood, T., Abinun, M.,
A. (1998) The response of pulmonary vascular endothelial cells to
Hambleton, S., Nademi, Z., Goulden, N., Davies, G., Qasim, W.,
monocrotaline pyrrole: cell proliferation and DNA synthesis in vitro
Gaspar, H. B., Cant, A., Gennery, A. R., and Veys, P. (2011)
and in vivo. Toxicol. Appl. Pharmacol. 150, 37−48.
Treosulfan-based conditioning regimens for hematopoietic stem cell
(193) Lee, J., Reich, R., Xu, F., and Sehgal, P. B. (2009) Golgi,
transplantation in children with primary immunodeficiency (PID): UK
trafficking, and mitosis dysfunction in pulmonary arterial endothelial experience. Blood 117, 4367−4375.
cells exposed to monocrotaline pyrrole and NO scavenging. Am. J. (212) Kaplan, F. S., Zasloff, M. A., Kitterman, J. A., Shore, E. M.,
Physiol.: Lung Cell. Mol. Physiol. 297, L715−L728. Hong, C. X., and Rocke, D. M. (2010) Early mortality and
(194) Mukhopadhyay, S., Shah, M., Xu, F., Patel, K., Tuder, R. M., cardiorespiratory failure in patients with fibrodysplasia ossificans
and Sehgal, P. B. (2008) Cytoplasmic provenance of STAT3 and PY- progressiva. J. Bone Jt. Surg., Am. Vol. 92, 686−691.
STAT3 in the endolysomal compartments in pulmonary arterial (213) Xu, M.-Q., Feldman, G., LeMerrer, M., Shugart, Y. Y., Glaser,
endothelial and smooth muscle cells: implications in pulmonary D. L., Urtizberea, J. A., Fardeau, M., Connor, J. M., Triffitt, J., Smith,
arterial hypertension. Am. J. Physiol.: Lung Cell Mol. Physiol. 249, R., Shore, E. M., and Kaplan, F. S. (2000) Linkage exclusion and
L449−L468. mutational analysis of the noggin gene in patients with fibrodysplasia
(195) Bull, L. B. (1955) The histological evidence of liver damage ossificans progressive (FOP). Clin. Genet. 58, 291−298.
from pyrrolizidine alkaloids: megalocytosis of the liver cells and (214) Gannon, F. H., Kaplan, F. S., Olmsted, E., Finkel, G. C.,
inclusion globules. Aust. Vet. J. 31, 33−40. Zasloff, M. A., and Shore, E. S. (1997) Bone morphogenetic protein 2/
(196) Jago, M. V. (1969) The development of the hepatic 4 in early fibromatous lesions of fibrodysplasia ossificans progressive.
megalocytosis of chronic pyrrolizidine alkaloid poisoning. Am. J. Human Pathol. 28, 339−343.
Pathol. 56, 405−422. (215) Enklaar, T., Zabel, B. U., and Prawitt, D. (2006) Beckwith-
(197) Svoboda, D., Reddy, J., and Bunyaratvej, S. (1971) Hepatic Wiedemann syndrome: multiple molecular mechanisms. Expert Rev.
megalocytosis in chronic lasiocarpine poisoning. Am. J. Pathol. 65, Mol. Med. 8, 1−19.
399−408. (216) Sun, J., Liu, Y.-H., Chen, H., Nguyen, M. P., Mishina, Y.,
(198) Samuel, A., and Jago, M. V. (1975) Localization in the cell Upperman, J. S., Ford, H. R., and Shi, W. (2007) Deficient Alk3-
cycle of the antimitotic action of the pyrrolizidine alkaloid lasiocarpine mediated BMP signaling causes prenatal omphalocele-like defect.
and of its metabolite, dehydroheliotridine. Chem.−Biol. Interact. 10, Biochem. Biophys. Res. Commun. 360, 238−243.
185−197. (217) Sotelo-Avila, C., and Gooch, W. M. (1976) Neoplasms
(199) Runo, J. R., and Loyd, J. E. (2003) Primary pulmonary associated with the Beckwith-Weidemann syndrome. Perspect. Pediatr.
hypertension. Lancet 361, 1533−1544. Pathol. 3, 255−272.
(200) Aldred, M. A., Comhair, S. A., Varella-Garcia, M., Asosingh, K., (218) Rump, P., Zeegers, M. P. A., and van Essen, A. J. (2005)
Xu, W., Noon, G. P., Thistlewaite, P. A., Tuder, R. M., Erzurum, S. C., Tumor risk in Beckwith-Wiedemann syndrome: a review and meta-
Geraci, M. W., and Coldren, C. D. (2010) Somatic chromosome analysis. Am. J. Med. Gen. 136A, 95−104.
abnormalities in the lungs of patients with pulmonary arterial (219) Spector, L. G., and Birch, J. (2012) The epidemiology of
hepatoblastoma. Pediatr. Blood Cancer 59, 776−779.
hypertension. Am. J. Respir. Crit. Care Med. 182, 1153−1160.
(220) Bardeesy, N., Falkoff, D., Petruzzi, M., Nowak, N., Zabel, B.,
(201) Austin, E. D., Hamid, R., and Ahmed, F. (2010) Somatic
Adam, M., Agiar, M. C., Grundy, P., Shows, T., and Pelletier, J. (1994)
mutations in pulmonary arterial hypertension. Am. J. Respir. Crit. Care
Anaplastic Wilm’s tumour, a subtype displaying poor prognosis,
Med. 182, 1095−1095.
harbours p53 gene mutations. Nat. Genet. 7, 91−97.
(202) Taura, P., Garcia-Valdecasas, J. C., Beltran, J., Izquierdo, E.,
(221) Muwakkit, S., Antillon, F., Valverde, P., Jenkins, J. J., Zaatari,
Navasa, M., Sala-Blanch, J., Mas, A., Balust, J., Grande, L., and Visa, J. G., and Dome, J. S. (2002) Letter to the Editor: Simultaneous
(1996) Moderate primary pulmonary hypertension in patients occurrence of Wilms tumor and rhabdomyosarcoma in two patients.
undergoing liver transplantation. Anesth. Analg. 83, 675−680. Med. Pediatr. Oncol. 39, 143−145.
(203) Hoeper, M. M., Krowka, M. J., and Strassburg, C. P. (2004) (222) Müller-Höcker, J., Weiss, M., Meyer, U., Schramel, P.,
Portopulmonary hypertension and hepatopulmonary syndrome. Wiebecke, B., Belohradsky, B. H., and Hübner, G. (1987) Fatal
Lancet 363, 1461−1468. copper storage disease of the liver in a German infant resembling
(204) Herve, P., Le Pavec, J., Sztrymf, B., Decante, B., Savale, L., and Indian childhood cirrhosis. Virchows Arch. A: Pathol. Anat. Histopathol.
Sitbon, O. (2007) Pulmonary vascular abnormalities in cirrhosis. Best 411, 379−385.
Pract. Res., Clin. Gastroenterol. 21, 141−159. (223) Price, L. A., Walker, N. I., Clague, A. E., Pullen, I. D., Smits, S.
(205) Kochar, R., Moises, I., Rubin, N., and Fallon, M. B. (2011) J., Ong, T.-H., and Patrick, M. (1996) Chronic copper toxicosis
Pulmonary complications of cirrhosis. Curr. Gastroenterol. Rep. 13, 34− presenting as liver failure in an Australian child. Pathology 28, 316−
39. 320.
(206) McDonnell, P. J., Toye, P. A., and Hutchins, G. M. (1983) (224) Seibold-Weiger, K., Vochem, M., Mackensen-Haen, S., and
Primary pulmonary hypertension and cirrhosis: are they related? Am. Speer, C. (1997) Fatal hepatic veno-occlusive disease in a newborn
Rev. Respir. Dis. 127, 437−441. infant. Am. J. Perinatol. 14, 107−111.

18 DOI: 10.1021/tx500403t
Chem. Res. Toxicol. 2015, 28, 4−20
Chemical Research in Toxicology Perspective

(225) Sergi, C., Beedgen, B., Linderkamp, O., and Hofmann, W. J. (246) Etzioni, A., Benderly, A., Rosenthal, E., Shehadah, V.,
(1999) Fatal course of veno-occlusive disease of the liver Auslander, L., Lahat, N., and Pollack, S. (1987) Defective humoral
(Endophlebitis Hepatica obliterans) in a preterm infant. Pathol., Res. and cellular immune functions associated with veno-occlusive disease
Pract. 195, 847−851. of the liver. J. Pediatr. 110, 549−554.
(226) Trollmann, R., Neureiter, D., Lang, T., Dörr, H. G., and (247) Roscioli, T., Cliffe, S. T., Bloch, D. B., Bell, C. G., Mullan, G.,
Behrens, R. (1999) Late manifestation of Indian childhood cirrhosis in Taylor, P. J., Sarris, M., Wang, J., Donald, J. A., Kirk, E. P., Ziegler, J. B.,
a 3-year-old German girl. Eur. J. Pediatr. 158, 375−378. Salzer, U., McDonald, G. B., Wong, M., Lindeman, R., and Buckley, M.
(227) Bull, L. B., Dick, A. T., Keast, J. C., and Edgar, G. (1956) An F. (2006) Mutations in the gene encoding the PML nuclear body
experimental investigation of the hepatotoxic and other effects on protein Sp110 are associated with immunodeficiency and hepatic
sheep of consumption of Heliotropium europaeum L.: heliotrope veno-occlusive disease. Nat. Genet. 38, 620−622.
poisoning of sheep. Aust. J. Agric. Res. 7, 281−331. (248) Cliffe, S. T., Wong, M., Taylor, P. J., Ruga, E., Wilcken, B.,
(228) St George-Grambauer, T. D., and Rac, R. (1962) Lindeman, R., Buckley, M. F., and Roscioli, T. (2007) The first
Hepatogenous chronic copper poisoning in sheep in South Australia prenatal diagnosis for veno-occlusive disease and immunodeficiency
due to the consumption of Echium plantagineum L. (Salvation Jane). syndrome, an autosomal recessive condition associated with mutation
Aust. Vet. J. 38, 288−293. in SP110. Prenatal Diagn. 27, 674−676.
(229) Miranda, C. L., Henderson, M. C., and Buhler, D. R. (1981) (249) Cliffe, S. T., Bloch, D. B., Suryani, S., Kamsteeg, E.-J., Avery, D.
Dietary copper enhances the hepatotoxicity of Senecio jacobaea in rats. T., Palendira, U., Church, J. A., Wainstein, B. K., Trizzino, A., Lefranc,
Toxicol. Appl. Pharmacol. 60, 418−423. G., Akatcherian, C., Megarbane, A., Gilissen, C., Moshous, D.,
(230) Howell, J. M., Deol, H. S., Dorling, P. R., and Thomas, J. B. Reichenbach, J., Ziegler, J. B., Wong, M., Tangye, S. G., Lindeman,
(1991) Experimental copper and heliotrope intoxication in sheep: R., Buckley, M. F., and Roscioli, T. (2012) Clinical, molecular, and
morphological changes. J. Comp. Pathol. 105, 49−74. cellular immunological findings in patients with SP110-associated
(231) Walker-Smith, J., and Blomfield, J. (1973) Wilson’s disease or veno-occlusive disease with immunodeficiency syndrome. J. Allergy
chronic copper poisoning? Arch. Dis. Child. 48, 476−479. Clin. Immunol. 130, 735−742.
(232) Müller-Höcker, J., Meyer, U., Wiebecke, B., Hübner, G., Eife, (250) Wang, T., Ong, P., Roscioli, T., Cliffe, S. T., and Church, J. A.
R., Kellner, M., and Schramel, P. (1988) Copper storage disease of the (2012) Hepatic veno-occlusive disease with immunodeficiency
liver and chronic dietary copper intoxication in two further German
(VODI): first reported case in the U.S. and identification of a unique
infants mimicking Indian Childhood Cirrhosis. Pathol., Res. Pract. 183,
mutation in Sp110. Clin. Immunol. 145, 102−107.
39−45. (251) Ganaiem, H., Eisenstein, E. M., Tenenbaum, A., Somech, R.,
(233) Müller-Höcker, J., Summer, K. H., Schramel, P., and Rodeck,
Simanovsky, N., Roscioli, T., Weintraub, M., and Stepensky, P. (2013)
B. (1998) Different pathomorphic patterns in exogenic infantile
The role of hematopoietic stem cell transplantation in SP110
copper intoxication of the liver. Pathol., Res. Pract. 194, 377−384.
associated veno-occlusive disease with immunodeficiency syndrome.
(234) Von Mühlendahl, K. E., and Lange, H. (1994) Copper and
Pediatr. Allergy Immunol. 24, 250−256.
childhood cirrhosis. Lancet 344, 1515−1516.
(252) (2009) Genetics Home Reference, US National Library of
(235) Scheinberg, I. H., and Sternlieb, I. (1994) Is non-Indian
childhood cirrhosis caused by excess dietary copper? Lancet 344, Medicine, http://ghr.nlm.nih.gov/gene/SP110.
(253) Deyo, J. A., and Kerkvliet, N. I. (1990) Immunotoxicity of the
1002−1004.
(236) Baker, A., Gormally, S., Saxena, R., Baldwin, D., Drumm, B., pyrrolizidine alkaloid monocrotaline following subchronic adminis-
Bonham, J., Potmann, B., and Mowat, A. P. (1995) Copper-associated tration to C57B1/6 mice. Fundam. Appl. Toxicol. 14, 842−849.
liver disease in childhood. J. Hepatol. 23, 538−543. (254) Molyneux, R. J., Gardner, D. L., Colegate, S. M., and Edgar, J.
(237) Müller, T., Feichtinger, H., Berger, H., and Müller, W. (1996) A. (2011) Pyrrolizidine alkaloid toxicity in livestock: a paradigm for
Endemic Tyrolean infantile cirrhosis: an ecogenetic disorder. Lancet human poisoning? Food Addit. Contam., Part A 28, 293−307.
347, 877−880. (255) Newberne, P. M., and Rogers, A. E. (1973) Nutrition,
(238) Müller, T., Müller, W., and Feichtinger, H. (1998) Idiopathic monocrotaline and aflatoxin B1 in liver carcinogenesis. Plant Foods
copper toxicosis. Am. J. Clin. Nutr. 67, 1082S−1986S. Man 1, 23−31.
(239) Wijmenga, C., Müller, T., Brunt, T., Feichtinger, H., (256) Yee, S. B., Kinser, S., Hill, D. A., Barton, C. C., Hotchkis, J. A.,
Schönitzer, D., Houwen, R. H. J., Müller, W., Sandkuijl, L. A., and Harkema, J. R., Ganey, P. E., and Roth, R. A. (2000) Synergistic
Pearson, P. L. (1998) Endemic Tyrolean infantile cirrhosis is not an hepatotoxicity from coexposure to bacterial endotoxin and the
allelic variant of Wilson’s disease. Eur. J. Hum. Genet. 6, 624−628. pyrrolizidine alkaloid monocrotaline. Toxicol. Appl. Pharmacol. 166,
(240) Tanner, M. S. (1998) Role of copper in Indian childhood 173−185.
cirrhosis. Am. J. Clin. Nutr. 67, 1074S−1081S. (257) (1992) Abwehr von Arzneimittelrisiken-Stufe II. Bundesan-
(241) Aston, N. S., Morris, P. A., Tanner, M. S., and Variend, S. zeiger 17 June, 4805; cited by Dtsch. Apoth. Ztg. 132, 1406−1408.
(1998) An animal model for copper-associated cirrhosis in infancy. J. (258) European Medicines Agency (2013) Public statement on the use
Pathol. 186, 215−221. of herbal medicinal products containing toxic, unsaturated pyrrolizidine
(242) Dieter, H. H., Schimmelpfennig, W., Meyer, E., and Tabert, M. alkaloids (PAs), EMA/HMPC/893108/2011, 2nd draft, 6 Nov,
(1999) Early childhood cirrhosis (ECC) in Germany between 1982 EMA,London, http://www.ema.europa.eu/docs/en_GB/document_
and 1994 with special consideration of copper etiology. Eur. J. Med. library/Public_statement/2013/11/WC500154224.pdf.
Res. 28, 233−242. (259) Czauderna, P., Katski, K., Kowalczyk, J., Kurylak, A., Lopatka,
(243) Zietz, B. P., de Vergara, J. D., and Dunkelberg, H. (2003) B., Skotnicka-Klonowicz, G., Sawicz-Birkoska, K., and Godzinski, J.
Copper concentrations in tap water and possible effects on infant’s (2000) Venoocclusive liver disease (VOD) as a complication of
healthresults of a study in Lower Saxony, Germany. Environ. Res. 92, Wilms’ tumour management in the series of consecutive 206 patients.
129−138. Eur. J. Pediatr. Surg. 10, 300−303.
(244) Zietz, B. P., Dieter, H. H., Lakomek, M., Schneider, H., (260) Cecen, E., Uysal, K. M., Ozguven, A., and Gunes, D. (2007)
Kessler-Gaedke, B., and Dunkelberg, H. (2003) Epidemiological Veno-occlusive disease in a child with rhabdomyosarcoma after
investigation on chronic copper toxicity to children exposed via the conventional chemotherapy. Pediatr. Hematol. Oncol. 24, 615−621.
public drinking water supply. Sci. Total Environ. 302, 127−144. (261) Xia, Q., Zhao, Y., Von Tungeln, L. S., Doerge, D. R., Lin, G.,
(245) Mellis, C., and Bales, P. M. (1976) Familial hepatic Cai, L., and Fu, P. P. (2013) Pyrrolizidine alkaloid-derived DNA
venoocclusive disease with probable immune deficiency. J. Pediatr. adducts as a common biological biomarker of pyrrolizidine alkaloid-
88, 236−242. induced tumorigenicity. Chem. Res. Toxicol. 26, 1384−1396.

19 DOI: 10.1021/tx500403t
Chem. Res. Toxicol. 2015, 28, 4−20
Chemical Research in Toxicology Perspective

(262) Gao, H., Li, N., Wang, J. Y., Zhang, S. C., and Lin, G. (2012)
Definitive diagnosis of hepatic obstruction syndrome induced by
pyrrolizidine alkaloids. J. Dig. Dis. 13, 33−39.

20 DOI: 10.1021/tx500403t
Chem. Res. Toxicol. 2015, 28, 4−20

Anda mungkin juga menyukai