Pyrrolizidine Alkaloids: Metabolic Activation Pathways Leading to

Nov 7, 2016 - It was recently determined that a genotoxic mechanism of liver tumor initiation mediated by PA-derived DNA adducts is a common metabolic...
0 downloads 9 Views 580KB Size
Subscriber access provided by BOSTON UNIV

Perspective

Pyrrolizidine Alkaloids: Metabolic Activation Pathways Leading to Liver Tumor Initiation Peter P. Fu Chem. Res. Toxicol., Just Accepted Manuscript • DOI: 10.1021/acs.chemrestox.6b00297 • Publication Date (Web): 07 Nov 2016 Downloaded from http://pubs.acs.org on November 14, 2016

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Chemical Research in Toxicology is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 39

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Pyrrolizidine Alkaloids: Metabolic Activation Pathways Leading to Liver Tumor Initiation Peter P. Fu National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079 USA

1 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Table of Contents Graphic:

2 ACS Paragon Plus Environment

Page 2 of 39

Page 3 of 39

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

ABSTRACT: Pyrrolizidine alkaloids (PAs) and PA N-oxides are a class of phytochemical carcinogens contained in over 6000 plant species widespread in the world. It has been estimated that approximately half of the 660 PAs and PA-N-oxides that have been characterized are cytotoxic, genotoxic, and tumorigenic. It was recently determined that a genotoxic mechanism of liver tumor initiation mediated by PA-derived DNA adducts is a common metabolic activation pathway of a number of PAs. We proposed this set of PA-derived DNA adducts could be a common biological biomarker of PA exposure and a potential biomarker of PA-induced liver tumor formation. We have also found that several reactive secondary pyrrolic metabolites can dissociate and inter-convert to other secondary pyrrolic metabolites, resulting in the formation of the same exogenous DNA adducts. This present perspective reports the current progress on these new findings and proposes the future research needed for obtaining a greater understanding on the role of this activation pathway, and validating the use of this set of PA-derived DNA adducts as a biological biomarker of PA-induced liver tumor initiation.

3 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

■ CONTENTS 1. Introduction 1.1. Livestock and Human Poisoning 1.2. Mechanism of Toxicity 1.3. Carcinogenicity 2. Current Understanding on Metabolic Activation 2.1. Mechanism of Liver Tumor Initiation 2.1.1. Formation of DNA Cross-linking and DNA–Protein Cross-linking 2.1.2. Exogenous DNA Adduct Formation 2.2. DNA Adducts as Biomarkers 2.3. Quantitation of Protein Adducts 3. Perspectives and Future Research 3.1. DHP-DNA Adducts – Liver Tumor Initiation 3.1.1. More PAs for metabolic activation studies 3.1.2. Other potential carcinogenic metabolites 3.1.3. Multiple activation pathways 3.1.4. Inter-convertibility and reversibility between pyrrolic metabolites 3.1.5. DHP-DNA adducts – A surrogate of PA carcinogenicity determination 3.1.6. Site and gene sequence-specificity of DNA adducts formation 3.2. DHP-Protein Adducts for Clinical Assay 3.3. Regulation of PA-Containing Products 3.4. DNA-DNA Cross Links and DNA Protein-Cross Links 3.5. Other Type of Reactive Pyrrolic Metabolite(s) 4. Conclusion Author Information Corresponding Author Funding Notes Biograph Acknowledgements Abbreviations References

4 ACS Paragon Plus Environment

Page 4 of 39

Page 5 of 39

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

1. INTRODUCTION Pyrrolizidine alkaloids (PAs), more formally by another nomenclature system called dehydropyrrolizidine alkaloids or 1,2-dehydro-PAs, and PA N-oxides are a class of rodent liver carcinogens.1-3 Recent findings have revealed that a number of PAs and PA N-oxides exert liver tumorigenicity mediated by a set of exogenous DNA adducts. In this present review, we address this mechanism, the discovery of the new reactive pyrrolic metabolites, their dissociation and interconversion leading to multiple metabolic activation pathways of PAs, and proposed future research directions. Part of the experimental results discussed in this perspective was generated from our laboratory.

1.1. Livestock and Human Poisoning PAs and PA N-oxides are common plant secondary metabolites.2, 4 There are over 660 PAs and PA N-oxides present in many plants around the world.5, 6 It has been estimated that approximately half of the identified PAs and PA N-oxides are hepatotoxic, mutagenic, carcinogenic, teratogenic, and occasionally pneumotoxic.2, 6-9 The acute hepatotoxicity caused by PAs results in haemorrhagic necrosis, hepatomegaly, ascites, necrosis, dysfunction, and death.2, 4, 6-8, 10, 11 The PA-induced liver damage results in the veno-occlusion disease (VOD).2, 7, 8, 11

Both PAs and PA-containing plant extracts exhibit a variety of genotoxicities in different

systems in vivo.2, 3, 7, 8, 12-24 There are two major routes of human exposure to PAs; (i) the consumption of food staples which contain the PAs constituents; including herbal medicines,18, 25-28 herbal dietary supplements,10, 11, 18, 29 teas,25, 30 herbal teas,18, 27, 31, 32 honey,7, 25, 33-37 and milk;38, 39 and (ii) the intake of food contaminated with toxic PAs7, 31, 40 Exposure of humans and livestock to toxic PAs through the food contamination pathway has resulted in numerous severe poisoning epidemics, with high mortality and morbidity rates.1, 2, 6, 7, 9, 18, 31, 38, 40-44 The first report on livestock poisoning by PAs was in 1787.31 Since then, many PAcontaining plant species have been found to cause numerous poisoning events in humans and livestock, including in cattle, sheep, chickens, ducks, and pigs.2, 9, 31, 43-45 The first large scale food poisoning in humans was recognized in 1920 in South Africa.46 The use of herbal products has been progressively increasing. Human poisonings caused by intake of PA-containing medicinal plants and herbal dietary supplements have been reported.28, 32 In 1989 the 5 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

International Program on Chemical Safety (IPCS) concluded that ‘‘consumption of contaminated grain or the use of PA-containing plants as herbal medicine, beverages, or food by man, or grazing on contaminated pastures by animals, may cause acute or chronic disease’’.40

1.2. Mechanism of Toxicity PAs consist of a necic acid and a necine base.2, 4, 47 There are four common necine bases of PAs, the retronecine, heliotridine, otonecine, and platynecine types, among which the first three types of necine bases possess a double bond at the C1 and C2 positions and exhibit hepatotoxicity, genotoxic, and potentially carcinogenicity (Figure 1).2, 4, 6-8, 48 Among the PAs, macrocyclic diester PAs exhibit the highest hepatotoxicity, genotoxicity, and tumorigenicity, followed by monoester PAs.2, 4, 22 Retronecine-type PAs possess a 7R stereochemistry, while the heliotridine-type PAs have a 7S absolute configuration. Retronecinetype PAs are the most abundant PAs and therefore are most widely studied.2, 4, 40 Platynecinetype PAs which possess a saturated necine base are in general not toxic.2, 22, 49

6 ACS Paragon Plus Environment

Page 6 of 39

Page 7 of 39

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Figure 1. The common necine bases of PAs; structures of DHR, DHH, and DHP; and the necic acid and necine base of riddelliine. DHP is a molecule that contains DHR and DHH in an equal ratio.

PAs are biologically inactive and require metabolic activation to exert toxicity, including carcinogenicity.2, 4, 7, 8 The retronecine-type and heliotridine-type PAs have three major metabolic pathways: (i) hydrolysis of the ester groups to form the corresponding necines and necic acids; (ii) oxidation of the nitrogen atom of the necine bases to produce PA N-oxides; and (iii) enzymatic hydroxylation of the necine base to produce the 3- or 8-hydroxy-PAs which are rapidly dehydrated to the reactive (+/-)-6,7-dihydro-7-hydroxy-1-hydroxymethyl-5H-pyrrolizine esters (DHP esters; or dehydro-PAs) (Figure 2). The otonecine-type PAs have only two major metabolic pathways: (i) hydrolysis of the ester groups to form the corresponding necine bases 7 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

and necic acids; and (ii) enzymatic oxidative N-demethylation of the nitrogen atom of the necine base, resulting in the ring closure, and dehydration to form DHP esters (dehydro-PAs).2, 4

Figure 2. Major metabolic pathways of retronecine-type and heliotridine-type PAs.

Cytochrome P-450 (CYP) 2B and CYP3A are the principal metabolizing enzymes responsible for the metabolism of PAs.4, 7, 50, 51 The enzymes that catalyze the metabolism of PAs to PA N-oxides are both cytochrome P-450 and flavin-containing monooxygenases.4, 52 Hydrolysis of the ester groups of PAs to form necine base and necic acids is catalyzed by the hepatic microsomal carboxylesterases.4, 52 Because DHP esters (dehydro-PA) metabolites are biologically active and can lead to liver tumor initiation, the hepatic metabolism of PAs to DHP esters (dehydro-PAs) is considered the principal metabolic activation pathway (Figure 2).2 It has been found that the PA N-oxide 8 ACS Paragon Plus Environment

Page 8 of 39

Page 9 of 39

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

metabolites, formed from metabolism of the parent PAs in vivo, can be enzymatically reduced to the parent PAs in the gut and liver (Figure 2).2, 4, 7, 8, 36, 53-55 Consequently, the metabolism of PAs to PA N-oxides should be considered a potential, but minor, activation pathway.2, 22, 53

1.3. Carcinogenicity In 1954, Schoental et al.56 reported that retrorsine and the PA-containing plant S. jacobaea lin induced liver tumors in rats. In the early 1970, a series of PAs were found to induce tumors, mainly the liver tumors, in rats and other experimental rodents. To date, more than 20 purified plant PAs, a PA N-oxide, dehydro-PAs, and plant extracts have been demonstrated to induce tumors in rodents. The PAs and retrorsine N-oxide that are known to induce liver tumors in rats are listed in Table 1, and their structures are shown in Figure 3. Since PAs are widespread in the world, human exposure to genotoxic and carcinogenic PAs is a concern. Among the carcinogenic PAs, lasiocarpine and riddelliine have been tested by the U.S. National Toxicology Program (NTP) in chronic tumorigenicity bioassays and shown to be rodent carcinogens.77, 78 Riddelliine is listed as a "reasonably anticipated to be human carcinogen” in the NTP 13rd Report on Carcinogens.79 The IARC listed monocrotaline, riddelliine, and lasiocarpine as Group 2B, possible human carcinogens.79-81

9 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Table 1. Carcinogenic Pyrrolizidine Alkaloids (1,2-Dehydro-PAs) and a Pyrrolizidine Alkaloid N-Oxide that Induce Liver Tumors in Rats. Pyrrolizidine Alkaloids Plant species (Family) References Retronecine - Type Pyrrolizidine Alkaloids 56-59 Retrorsine 1 Senecio (Compositae) 57, 58, 60 Riddelliine 2 Senecio (Compositae), Crotalaria (Leguminosae) 61, 62 Monocrotaline 3 Crotalaria (Leguminosae) 56, 58, 63 Senecionine 4 Senecio (Compositae) 57, 63 Seneciphylline 5 Senecio (Compositae) 56, 64 Jacobine 6 Senecio L. (Compositae) Symphytine 7 Symphytum officinale L (Boraginaceae) 65, 66 Retronecine – Type Pyrrolizidine Alkaloid N-Oxide 56, 58, 59 Retrorsine N-oxide Senecio (Compositae), (Isatidine) 8 Crotalaria (Leguminosae) Heliotridine - Type Pyrrolizidine Alkaloids 67-70 Lasiocarpine 9 Heliotropium (Boraginaceae) 71 Heliotrine 10 Heliotropium (Boraginaceae) Otonecine - Type Pyrrolizidine Alkaloids 72, 73 Ligularia dentata Hara (Compositae) Clivorine 11 63, 65, 74 Senecio (Compositae) Senkirkine 12 Petasites (Compositae) 74-76 Patasitenine 13 Senecio (Compositae)

10 ACS Paragon Plus Environment

Page 10 of 39

Page 11 of 39

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

HO

HO

CH2OH O

Me H O

O

Me

Me O

O

H

N

Retrorsine 1

O

O

H O

O

CH2OH H Me

CH2

H

H

H

O

O

HO

CH(Me) 2 O

Me

O O

O

O CH2

H

N

Symphytine 7

HO

CH(Me) 2

MeO

O Me

HO

O CH2

H

CH2

H

O CH2

Senecionine 4

HO Me

HO Me HO C Me MeO O Me H Me O

O

O

O

Me

N

CH2

N

O

H

Me

Me

Jacobine 6

Me

CH2

Me

N

H

O

O

O

Me O

HO

Seneciphylline 5 HO

H O

Monocrotaline 3

Me

CH2

H

O

O

O O

Me

Me

N

Me

CH2

HO O

O

CH2

Riddelliine 2

Me

Me

O

N

HO

Me HO

H CH2

O

HO Me

O

CH2

H

CH2OH

N O

Me

H

HO O

H3 C O

O

O

Me

CH2

N Me

Clivorine 11

O

O

Heliotrine 10

Me

Me

Me O

Me

O CH2

O

HO

O O

H

H O

Lasiocarpine 9

OAc

H2 C

N

N

Retrorsine N-oxide (Isatidine) 8

H O

O

Me O

N

N

Me

Me

Senkirkine 12

O CH2

Petasitenine 13

Figure 3. Structures and names of the liver carcinogens of PAs and retrorsine N-oxide.

11 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

2. CURRENT UNDERSTANDING ON METABOLIC ACTIVATION 2.1. Mechanism of Liver Tumor Initiation Segall and co-workers determined that trans-4-hydroxy-2-hexenal was formed from the liver microsomal metabolism of senecionine.82-85 This metabolite was cytotoxic in primary cultures of rat hepatocytes83 and could bind with deoxyguanosine to form adducts.85 However, the mechanism of its formation was still uncertain. Further studies are required to find out whether or not the formation of this metabolite is involved in the senecionine-induced liver tumor initiation.

2.1.1. Formation of DNA Cross-linking and DNA–Protein Cross-linking Dehydro-PAs and DHP possess two electrophilic sites at the C7 and C9 positions of the necine base, both of which can bind to cellular DNA and proteins to form DNA-DNA cross-links and DNA-protein cross-links.13-15, 86-92 Coulombe and coworkers studied the formation of DNA-DNA and DNA-protein links using eight representative PAs in cultured bovine kidney epithelial cells in the presence of an external metabolizing system and determined the structure–activity relationships.14, 93 The eight PAs included five macrocyclic diesters, two open chain diesters, and one necine base (retronecine). The level of the DNA-protein cross-links was determined by Western immunoblotting, and the results indicated a positive structure-activity relationship, with the macrocyclic diester PAs producing higher quantities of DNA-protein cross linking than the other types of PAs. Dehydrosenecionine, dehydroseneciphylline, dehydroriddelliine, and dehydromonocrotaline are reactive primary DHP esters metabolites of the parent PAs senecionine, seneciphylline, riddelliine, and monocrotaline, all of which belong to macrocyclic diesters PAs. Kim et al.15 determined that, compared to DHR, these four DHP esters (dehydro-PAs) induced more proteinDNA cross-links. These results are in general consistent with mutagenicity and hepatocarcinogenicity of the parent PAs.15 Because the tested PAs are carcinogens, it is highly possible that DNA-DNA and DNA-protein cross-links can cause tumor initiation.

2.1.2. Exogenous DNA Adduct Formation It has long been determined that dehydromonocrotaline, dehydroretrorsine, DHR, and dehydroheliotridine (DHH) can bind to DNA, nucleosides, and nucleotides.2, 94-97 These results 12 ACS Paragon Plus Environment

Page 12 of 39

Page 13 of 39

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

indicate that DHP esters metabolites could bind with cellular DNA in vivo, resulting in liver tumor initiation mediated by exogenous DNA adducts formation. The 2001 and the more recent research findings provided the first direct correlation demonstrating that PA induced liver tumors in experimental rodents are mediated by DNA-DHP adduct formation.23 Yang et al. determined that riddelliine induced liver tumors in rats and mice mediated by DHP-derived DNA adducts that were produced in the livers in vivo in a doseresponse manner.23, 98, 99 Described below is the mechanism-based evidence about this genotoxic mechanism, which should potentially be applicable to livestock and humans: 1. There is a good correlation between the levels of the DHP-DNA adducts and the tumorigenic potencies in rats fed different doses of riddelliine.23, 98, 99 2. The DHP-DNA adducts formation in vivo is cell type specific, generating more DHPDNA adducts in the hepatic endothelial cells than in the parenchymal cells.98 3. Using transgenic Big Blue rats for study, Mei et al.100 determined that riddelliine induced mutations at guanine bases in the liver; and mutations occurred mainly in the endothelial cells. 4. Hong et al.101 reported that B6C3F1 mice treated with riddelliine resulted in G to T mutations at codon 12 of the K-ras oncogene. 5. Using the cII gene mutation assay in transgenic Big Blue rats, Mei et al.100, 102 determined that riddelliine induced mutants, predominantly the G:C to T:A transversions. These mutation patterns suggested that metabolism of riddelliine generated DHP-dG adducts in the endothelial cells, which induce mutations leading to liver tumor initiation.100, 103 6. The metabolic pattern and DNA adduct profile produced from the human and rat liver microsomal metabolism of riddelliine were highly similar, which indicated that the studies using experimental rodents are highly relevant to humans and that riddelliine can be a human carcinogen.104 Subsequently, the U.S. NTP classified riddelliine as "reasonably anticipated to be a human carcinogen”.79 The success of the mechanistic studies on riddelliine relied on the development of a 32Ppostlabeling/HPLC method that could accurately quantify the exogenous DNA adducts formed in the liver of riddelliine-treated rats and mice.105 By using this analytical method, PA-DNA adducts were detected and quantified in vivo and in vitro from the metabolism of a series of

13 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

PAs,23, 98, 99, 104, 106-112 PA N-oxides,53, 55, 112 PA-containing herbal plant extracts,29, 109 and dietary supplements extracts.29 Since the 32P-postlabeling/HPLC method could not provide information about the molecular structures of the PA-derived DNA adducts, an HPLC-ES-MS/MS method was then developed for the further studies.16, 19-22, 24 By this method, a set of four DHP-DNA adducts, designated as DHP-dG-3, DHP-dG-4, DHP-dA-3, and DHP-dA-4, was detected and quantified in the livers of rats treated with riddelliine and monocrotaline (Figure 4).16, 22, 24

O

O

N

N

NH

N

N

1

N H

dR

8

9

N

7

2

6 3

4

N

N

5

DHP-dG-4

N

7 8

2

OH

OH

1 9

9

NH N N dR

OH

N H

6

4 3

N

dR

N

DHP-dG-3

5

NH

OH

9

NH N

N

N

N

N N

dR

DHP-dA-3

DHP-dA-4

Figure 4. Structures of DHP-dG-3, DHP-dG-4, DHP-dA-3, and DHP-dA-4 adducts.

2.2. DNA Adducts as Biomarkers Xia et al.22 studied DHP-DNA adduct formation in the livers of rats treated with a series of PAs in vivo. The results indicated that all the seven hepatocarcinogenic PAs and riddelline Noxide produced the same set of DHP-dG-3, DHP-dG-4, DHP-dA-3, and DHP-dA-4 adducts in the livers of treated rats in a dose-response manner. The levels of DHP-dG adducts were higher 14 ACS Paragon Plus Environment

Page 14 of 39

Page 15 of 39

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

than those of DHP-dA adducts. The levels of DHP-DNA adducts formed in the liver of PAtreated rats were in the order: retrorsine > lasiocarpine > riddelliine ~ monocrotaline > riddelliine N-oxide > senkirkine > heliotrine ≥ clivorine >>> lycopsamine ~ retronecine ~ platyphylliine ~ control. Lycopsamine and retronecine, which are not liver tumorigens, formed the lowest levels of DHP-DNA adducts. These results indicate that the level of DHP-DNA adduct formation is in the order: di-ester PAs > mono-ester PAs ~ di-ester PA N-oxide >>> retronecine (which does not have an ester linkage).22 Based on these and our previously published results,16, 24, 104, 109, 110 a general metabolic activation pathway is proposed (Figure 5). This represents the first finding that an identical set of exogenous (not endogenous) DNA adducts is generated from individual members of a large class of chemical carcinogens, and these DNA adducts can be potential biomarkers of PA exposure and liver tumor initiation.

15 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Figure 5. Proposed general mechanism of DHP-DNA adducts formation.

2.3. Quantitation of Protein Adducts Pyrrole-protein (DHP-protein) adducts have been proposed as a biological biomarker of exposure to toxic PAs.2, 7,113 A qualitative analytical method was developed by Mattocks and coworkers in the early 1990’s to detect these adducts in experimental animals and livestock.114-117 This method used silver nitrate in ethanol to cleave the thiol linkage of the adducts, replacing the protein moiety with ethoxyl groups. To enhance the detection sensibility, the 7,9-di-C2H5ODHP formed was reacted with the Ehrlich reagent to form the product that was measured colorimetrically.118 In 2011, Lin and co-workers modified this method, using the developed LC/MS/MS method to detect DHP-protein adducts in the blood of patients with liver disease.11 This method was further modified to allow quantification of DHP-protein adducts formed in vivo, including human blood samples.51

16 ACS Paragon Plus Environment

Page 16 of 39

Page 17 of 39

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Xia et al.119 recently developed an alternate LC/MS/MS method which was used to accurately quantify the DHP-protein adducts formed in the blood of rats treated with a series of PAs.22

3. PERSPECTIVES AND FUTURE RESEARCH 3.1. DHP-DNA Adducts - Liver Tumor Initiation Nearly half of the 660 PAs and PA N-oxides currently identified contain a unsaturated necine base and are cytotoxic, genototoxic, and tumorigenic.6, 8 As such, determination of the mechanism leading to liver tumor initiation for so many PAs is challenging. Recent findings have shown that the metabolic activation of a number of hepatocarcinogenic PAs is by the same mechanism mediated by DHP-DNA adducts formation leading to the liver tumor initiation. Consequently, much of the future research should focus on this mechanism. 3.1.1. More PAs for metabolic activation studies At the present, only seven hepatocarcinogenic PAs and retrorsine N-oxide have been shown to initiate liver tumors through this general metabolic activation pathway. In order to establish the generality of this genotoxic mechanism, more representative PAs and PA N-oxides will need to be studied.

3.1.2. Other potential carcinogenic metabolites To date, DHP esters (dehydro-PAs) and DHP are the only two reactive metabolites that have been shown to be able to cause liver tumor initiation.2, 7, 23 DHP esters are extremely unstable,120 and therefore, may not be the principal metabolites to initiate PA-induced carcinogenicity. On the other hand, although DHP is more stable, it is much less reactive and thus may not be the principal metabolite for liver tumor initiation.7, 23 Thus, other metabolites may be involved in generating DNA adducts in vivo and causing liver tumor initiation.2 7-Glutathione-DHP (7-GS-DHP) (Figure 6) is a metabolite formed in vivo and in vitro.21, 121125

Estep et al. reported that 7-N-acetylcysteine-DHP (Figure 6) was present in the urine of male

Sprague-Dawley rats treated with 14C-monocrotaline or 14C-senecionine.126 7-Cysteine-DHP (Figure 6) was formed from the rat liver microsomal metabolism of 7-GS-DHP.20 We have determined that these three metabolites (7-GS-DHP, 7-cysteine-DHP, and 7-N-acetylcysteine-

17 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

DHP) can react with calf thymus DNA to form DNA adducts.19-21 Therefore, these three metabolites are potential carcinogenic metabolites, capable of initiating liver tumors.19-21 Segall et al.127 reported that 7-methoxy-DHP (7-CH3O-DHP) (Figure 6) was a metabolite formed from the mouse liver microsomal metabolism of senecionine.127 7-Lysine-DHP was recently found to be a metabolite formed in mice administered with monocrotaline.128 Chen et al. reported that 9-GS-DHP is a metabolite in the male Sprague-Dawley rats administered with isoline, retrorsine, or monocrotaline.129 7-CH3O-DHP, 7-lysine-DHP, and 9-GS-DHP all possess the same type of DHP moiety as that of the metabolites described above. It is important to determine whether or not they are hepatocarcinogens, and are also mediated by this activation pathway.

Figure 6. Structures of 7-GS-DHP, 9-GS-DHP, 7-cysteine-DHP, 7-N-acetylcysteine-DHP, 7CH3O-DHP, 9-CHO-DHP, and (3H-pyrrolizin-7-yl)methanol.

3.1.3. Multiple activation pathways 18 ACS Paragon Plus Environment

Page 18 of 39

Page 19 of 39

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

At present, dehydro-PAs and DHP are the two types of metabolites known to bind to DNA in cells to form DHP-DNA adducts. As described previously, 7-GS-DHP, 7-cysteine-DHP, and 7N-acetylcysteine-DHP are able to bind to DNA to form DHP-DNA adducts, these secondary metabolites should be able to bind to cellular DNA in vivo, leading to liver tumor initiation. Similarly, both 7-CH3O-DHP and 7-lysine-DHP may also transfer the DHP moiety to cellular DNA. Consequently, it is highly possible that there exists multiple metabolic activation pathways resulting in the DHP-DNA adducts formation and PA-induced liver tumors.

3.1.4. Inter-conversion and reversibility between pyrrolic metabolites The primary DHP esters (dehydro-PAs) can react with water and cellular constituents to produce secondary pyrrolic metabolites. The reversibility of the secondary metabolites was first observed by Curtain and Edgar in 1976, who discovered that the binding of dehydroheliotridine (DHH) with DNA was reversible.97 In 1997, Sun et al. determined that the binding of DHR with albumin was reversible,130 and in 1983, Mattocks and Bird determined that the reaction product of DHR and nicotinamide was also reversible131 The inter-conversion and the reversible reactions proceed through an SN1 mechanism, and the resulting carbonium ion can be stabilized by resonance.2 Partly based on these observations, Edgar et al. proposed that in vivo, DHP, DHP-DNA adducts, and DHP-protein adducts can be dissociated in the liver, resulting in a pooled secondary metabolites and forming multiple activation pathways leading to the PA-induced liver tumor initiation.7 While this proposed mechanism is promising, the structures of the above-described secondary metabolites have not been fully characterized.7, 97, 130, 131 It is also not known whether or not these reaction products are indeed metabolites formed in vivo and are involved in the liver tumor initiation. As such, for the support of this proposed mechanism, it is timely and important to determine that some identified secondary metabolites can indeed undergo dissociation, interconversion, and reversible reactions. The following recently published experimental results support this proposed mechanism. 7-GS-DHP, 7-cysteine-DHP, and 7-N-acetylcysteine-DHP are secondary pyrrolic metabolites produced from the reactions of DHP esters (dehydro-PAs) with glutathione, cysteine, and Nacetylcysteine, respectively, in vivo and/or in vitro.19-21, 121-126, 132 We have demonstrated that upon incubation with human and rat liver microsomes, 7-GS-DHP, 7-cysteine-DHP, and 7-N19 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

acetylcysteine-DHP dissociated to DHP.19-21 We have also demonstrated that reaction of 7-GSDHP, 7-cysteine-DHP, or 7-N-acetylcysteine-DHP with dG produced DHP-dG-3 and DHP-dG4; and reaction with dA generated DHP-dA-3 and DHP-dA-4. Upon incubation with calf thymus DNA, these three secondary metabolites were inter-converted into DHP-dG and DHP-dA adducts19-21 In all these reactions, the glutathionyl, cysteinyl, and N-acetylcysteinyl functional groups were disassociated (removed).19-21 We have determined that DHP can also react with glutathione, cysteine, and N-acetylcysteine to form the corresponding 7-GS-DHP, 7-cysteineDHP, and 7-N-acetylcysteine-DHP, but as expected, the reactions are much slower than the dissociation of 7-GS-DHP, 7-cysteine-DHP, and 7-N-acetylcysteine-DHP into DHP.19-21 Also, 7-GS-DHP was metabolically dissociated into 7-cysteine-DHP.20 In addition, we have found that DHP-dA-3 and DHP-dA-4, as well as DHP-dG-3 and DHP-dG-4, are inter-convertible; and these inter-conversion reactions were through an SN1 mechanism.24 Recently, Chen et al. found that 9-GS-DHP, a predominant glutathione-DHP metabolite in the bile of rats treated with PAs, was inter-converted to 7-GS-DHP.129 Thus, the inter-conversion and dissociation reactions of the secondary metabolites strongly support the mechanism proposed by Edgar et al.7 Similar to the mechanism proposed by Edgar at al., we herewith propose a mechanism using the identified secondary metabolites 7-GS-DHP, 9-GS-DHP, 7-cysteine-DHP, and 7-N-acetylcysteine-DHP as examples (Figure 7).

20 ACS Paragon Plus Environment

Page 20 of 39

Page 21 of 39

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

R

R O

O

O

O

9

O 7

O

P450

CH2

CH2

1

8

6

2

N 4

5

Dehydro-PA GSH NA C

CH2OH

SG

HO

N

3

PA

NAC

O O

HO

H2O CH2OH

N

N

9-GS-DHP

GS H, CY S

DNA

SG

N

7-NAC-DHP

DHP DN A

DNA

+

N

A DN

CYS

CH2OH

7-GS-DHP

CH2OH

N

7-CYS-DHP

DHP-dG-3 DHP-dG-4 DHP-dA-3 DNP-dA-4

DNA

HO

CH2OH

N

DHP

Liver Tumors

Figure 7. Proposed multiple metabolic activation pathways of PA-induced tumor formation, involving the inter-conversion of secondary metabolites. Based on this proposed mechanism, secondary metabolites should play a critical role on the PA-induced liver tumor initiation. The significant new findings can provide inside of the mechanistic activation. The proposed new research directions are as follows: (i)

Besides DHP esters (dehydro-PAs) and DHP, other reactive carcinogenic metabolites are likely to exist.

(ii)

There are multiple activation pathways, with each pathway generating the same set of DHP-DNA adducts in vivo, leading to liver tumor initiation.

(iii) It is important to determine the principal carcinogenic metabolite(s) of PAs. (iv) The formation of 7-GS-DHP has long been considered as a detoxification pathway.121125, 133

Nevertheless, our findings suggest that metabolic pathways leading to the

formation of 7-GS-DHP, 9-GS-DHP, 7-cysteine-DHP, and 7-N-acetylcysteine-DHP are potential activation pathways leading to liver tumor initiation.

21 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(v)

As earlier described, DHP esters (dehydro-PAs) can bind to cellular DNA and proteins to produce DNA adducts,16, 19-24, 29, 98, 99, 104, 106-113 DHP-protein adducts,7, 11, 51, 114-119, 134 DNA-DNA cross links, and DNA-protein cross links in vivo and/or in vitro.13-15, 86-88, 90, 91, 119

It warrants to investigate whether or not DNA adducts, DHP-protein adducts,

DNA-DNA cross links, and DNA-protein cross links can undergo inter-conversion and reversible reactions.

3.1.5. DHP-DNA adducts – A surrogate of PA carcinogenicity determination It may not be possible to assess the carcinogenic potency for many PAs and PA N-oxides by animal tumorigenicity bioassays. As such, it is desirable to develop an alternative mechanismbased, convenient, and reliable bioassay for risk assessment. Since it has been shown that a series of carcinogenic PAs and a PA N-oxide (retrorsine N-oxide) induced liver tumors mediated by the same set of DHP-DNA adducts, we propose that this metabolic activation pathway can potentially serve as a mechanism-based biological assay for assessing the PAs-induced and PA N-oxides-induced carcinogenicity. For the method validation, it is necessary to use a variety of different types of carcinogenic PAs as substrates and many different biological systems for method development, so that a reliable and convenient in vitro system, such as liver microsomal metabolism or in the cultured cells, can be developed for use.

3.1.6. Site and gene sequence-specificity of DNA adducts formation It has been previously described that riddelliine induced hemangiosarcomas in B6C3F1 mice resulting in G to T mutations at codon 12 of the K-ras oncogene,101 and that G:C to T:A transversion was the major type of mutation in rats treated with riddelliine.103 These mutation patterns support that DHP-dG adducts can lead to mutations and progression of cells from normality to neoplasia. However, there are apparent knowledge gap concerning DNA adduct formation, its relationship with the specific gene mutations, and ultimately with the tumor initiation induced by PAs. These warrant further investigation.

3.2. DHP-Protein Adducts for Clinical Assay The analytical method developed by Lin et al. has been successfully used to quantify DHPprotein adducts present in the blood of HSOS patients.11, 51 Our developed LC/MS/MS analytical 22 ACS Paragon Plus Environment

Page 22 of 39

Page 23 of 39

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

method can accurately quantify DHP-protein adducts formed in rodent blood samples.119 It is our belief that, with further study, this newly developed LC/MS/MS analytical method can be used to serve as a convenient clinical bioassay. Furthermore, since the level of DHP-protein adducts well correlated with that of DHP-DNA adducts,119 with the further study for method development, we propose that DHP-protein adducts may potentially be used to serve as an invasive biomarker of PA carcinogenesis.

3.3. Regulation of PA-containing Products Many PAs and PA N-oxides present in herbal plants and herbal products are chemical carcinogens. Several PAs have been listed as probable human carcinogens by IARC and the NTP. Regulatory agencies around the world have issued bans and alerts on products containing PAs.7, 28, 135 In 1992, the Federal Health Department of Germany restricted that the herbal plants “may be sold and used only if daily external exposure to no more than 100 µg PAs and internal exposure to no more than 1 µg per day for no more than six weeks a year”.28 However, hundreds of toxic PAs and PA N-oxides are present in thousands of plants worldwide and their carcinogenic potency is drastically different. At the present, no practical analytical methods are available for quantifying the total quantity of toxic PAs. Furthermore, the regulation to limit the intake of the carcinogenic PAs by weight is not scientifically justified. Therefore, practical and reliable mechanism-based analytical methods must be developed to assess the risk posed by PAs. We propose that, upon metabolism of the herbal products in vivo, in vitro, or in the cultured cells, quantitation of the DHP-DNA adducts should be an assessable, reliable, and mechanism-based bioassay. For regulatory purpose, we believe this is a highly significant and worthy goal to pursue.

3.4. DNA-DNA Cross Links and DNA-Protein Cross Links As previously addressed, PAs can bind to DNA and proteins to form DNA-DNA links and DNA-protein crosslinks in cultured cells.13-15, 87, 93, 136 The experimental results obtained by Coulombe and co-workers14, 93 support the concept that the DNA-DNA and DNA-protein links are potential biomarkers of PA-induced tumorigenicity. Nevertheless, the structures of the DNA crosslink adducts have not been fully elucidated. The correlation between the levels of these cross links formed in vivo with the tumor potency of 23 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

treated animals has yet to be examined. It is important to validate the metabolic formation of DNA-DNA and DNA-protein crosslinks in vivo as another important metabolic activation pathway leading to the PA-induced liver tumor initiation. Future studies should include: (i) the structural characterization of these cross links formed in vivo; (ii) identification and quantitation of their formation in vivo; and (iii) study of the relationship between their formation and the liver tumor potency of PAs and PA N-oxides on a structure-activity basis.

3.5. Other Type of Reactive Pyrrolic Metabolite(s) Segall et al.127 found that 1-formyl-7-hydroxy-6,7-dihvdro-5H-pyrrolizine (9-CHO-DHP) was a metabolite formed from the mouse liver microsomal metabolism of senecionine (Figure 6). Fashe et al.137 reported that human liver microsomal metabolism of retrorsine generated (3Hpyrrolizin-7-yl)methanol which can form glutathione conjugates. These two metabolites contain a DHP moiety and may be able to transfer DHP to DNA to initiate liver tumor formation. In addition, there may be other active metabolites that can contribute to liver tumor initiation.

4. CONCLUSIONS The study of chemical carcinogens initially requires determination of the metabolic activation pathway leading to carcinogenesis, which involves the identification of the ultimate carcinogenic metabolite. In general, the most important and convincing pathway of cancer induction is the mechanism involving the exogenous DNA adduct formation. The detection and quantitation of the exogenous DNA adducts formed in vivo requires the development of analytical methods for detection and quantitation. With the analytical method developed available, the ultimate carcinogenic metabolite(s) can be determined. The determination of such an activation pathway that is responsible for tumor initiation is always challenging. Many carcinogenic PAs and PA N-oxides are carcinogenic, and PA-containing plants are the most common poisonous and possibly carcinogenic plants affecting livestock, wildlife, and humans. We recently determined that there is a common genotoxic mechanism mediated by DHP-DNA adducts. The levels of the DHP-DNA adducts correlated with the liver tumor potency of a series of PAs with different structural features. This finding is particularly important because this allows studying the mechanism for a whole family of PAs carcinogens, not a single carcinogenic PA. 24 ACS Paragon Plus Environment

Page 24 of 39

Page 25 of 39

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

The new findings that several secondary metabolites can be dissociated and inter-convertible in vitro strongly support the mechanism proposed by Edgar et al.7 Because of the all abovedescribed significant new findings, we therefore consider that part of the future research will be focused on these research areas. At the same time, we also believe that the activation pathway mediated by DNA-DNA and DNA-protein cross links is equally important, and should be the focus of further investigations as well.

AUTHOR INFORMATION Corresponding Author *(P.P.F.) E-mail: [email protected].

Notes The author declares no competing financial interest.

ACKNOWLEDGMENTS The author thanks Dr. Frederick A. Beland for critical review and Dr. Qingsu Xia and Ashley Groves for the preparation of this manuscript. The views presented in this paper do not necessarily represent those of the U.S. Food and Drug Administration (FDA). No official support or endorsement by the U.S. FDA is intended or should be inferred.

FUNDING SOURCES Funding was provided by an Internal FDA Fund.

ABBREVIATIONS PA, pyrrolizidine alkaloid; DHR, dehydroretronecine or (-)-R-6,7-dihydro-7-hydroxy-1hydroxymethyl-5H- pyrrolizine); DHP, (+/-)-6,7-dihydro-7-hydroxy-1-hydroxymethyl-5Hpyrrolizine; dG, 2’-deoxyguanosine; dA, 2’-deoxyadenosine; DHP-dG-3 and DHP-dG-4, a pair of epimers of 7-hydroxy-9-(deoxyguanosin-N2-yl)dehydrosupinidine adducts; DHP-dA-3 and DHP-dA-4, a pair of epimers of 7-hydroxy-9-(deoxyadenosin-N6-yl)dehydrosupinidine adducts; 25 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

HPLC-ES-MS/MS, high-performance liquid chromatography electrospray ionization tandem mass spectrometry; NCTR, National Center for Toxicological Research; NTP, National Toxicology Program; MRM, multiple reaction monitoring.

26 ACS Paragon Plus Environment

Page 26 of 39

Page 27 of 39

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

REFERENCES (1)

Bull, L. B., Culvenor, C. C., and Dick, A. J. (1968) The pyrrolizidine alkaloids. Their

chemistry, pathogenicity and other biological properties. North-Holland, Amsterdam. (2)

Mattocks, A. R. (1986) Chemistry and Toxicology of Pyrrolizidine Alkaloids., Academic

Press, London, NY. (3)

Mattocks, A. R. (1968) Toxicity of pyrrolizidine alkaloids. Nature, 217, 723-728.

(4)

Fu, P. P., Xia, Q., Lin, G., and Chou, M. W. (2004) Pyrrolizidine alkaloids--genotoxicity,

metabolism enzymes, metabolic activation, and mechanisms. Drug Metab. Rev., 36, 1-55. (5)

Wiedenfeld, H. (2013) Alkaloids derived from ornithine: pyrrolizidine alkaloids, In

Natural Products. Phytochemistry, Botany and Metabolism of Alkaloids, Phenolics and Terpenes (Ramawat, K. G., and Merillon, J. M., Eds.) pp 359-379, Springer-Verlag Berlin Heidelberg. (6)

Wiedenfeld, H., and Edgar, J. (2011) Toxicity of pyrrolizidine alkaloids to humans and

ruminants. Phytochem Rev, 10, 137-151. (7)

Edgar, J. A., Molyneux, R. J., and Colegate, S. M. (2015) Pyrrolizidine alkaloids:

potential role in the etiology of cancers, pulmonary hypertension, congenital anomalies, and liver disease. Chem. Res. Toxicol., 28, 4-20. (8)

IPCS. (1988) Pyrrolizidine alkaloids., In Environmental Health Criteria 80, International

Programme on Chemical Safety WHO: Geneva. (9)

Kakar, F., Akbarian, Z., Leslie, T., Mustafa, M. L., Watson, J., van Egmond, H. P., Omar,

M. F., and Mofleh, J. (2010) An outbreak of hepatic veno-occlusive disease in Western Afghanistan associated with exposure to wheat flour contaminated with pyrrolizidine alkaloids. J Toxicol, 2010, ID 313280. (10)

Fu, P. P., Xia, Q., Chou, M. W., and Lin, G. (2007) Detection, hepatotoxicity, and

tumorigenicity of pyrrolizidine alkaloids in Chinese herbal plants and herbal dietary supplements. J. Food Drug Anal., 15, 400-415. (11)

Lin, G., Wang, J. Y., Li, N., Li, M., Gao, H., Ji, Y., Zhang, F., Wang, H., Zhou, Y., Ye,

Y., Xu, H. X., and Zheng, J. (2011) Hepatic sinusoidal obstruction syndrome associated with consumption of Gynura segetum. J. Hepatol., 54, 666-673. (12)

Chen, T., Mei, N., and Fu, P. P. (2010) Genotoxicity of pyrrolizidine alkaloids. J Appl

Toxicol, 30, 183-196.

27 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(13)

Coulombe, R. A., Jr., Drew, G. L., and Stermitz, F. R. (1999) Pyrrolizidine alkaloids

crosslink DNA with actin. Toxicol. Appl. Pharmacol., 154, 198-202. (14)

Hincks, J. R., Kim, H.-Y., Segall, H. J., Molyneux, R. J., Stermitz, F. R., and Coulombe,

R. A., Jr. (1991) DNA cross-linking in mammalian cells by pyrrolizidine alkaloids: structureactivity relationships. Toxicol. Appl. Pharmacol., 111, 90-98. (15)

Kim, H.-Y., Stermitz, F. R., and Coulombe, R. A. J. (1995) Pyrrolizidine alkaloid-

induced DNA-protein cross-links. Carcinogenesis, 16, 2691-2697. (16)

Fu, P. P., Chou, M. W., Churchwell, M., Wang, Y., Zhao, Y., Xia, Q., Gamboa da Costa,

G., Marques, M. M., Beland, F. A., and Doerge, D. R. (2010) High-performance liquid chromatography electrospray ionization tandem mass spectrometry for the detection and quantitation of pyrrolizidine alkaloid-derived DNA adducts in vitro and in vivo. Chem. Res. Toxicol., 23, 637-652. (17)

Fu, P. P., Chou, M. W., Xia, Q., Yang, Y.-C., Yan, J., Doerge, D. R., and Chan, P. C.

(2001) Genotoxic pyrrolizidine alkaloids and pyrrolizidine alkaloid N-oxides—mechanisms leading to DNA adduct formation and tumorigenicity. J. Environ. Sci. Health, Part C, 19, 353385. (18)

Fu, P. P., Xia, Q., Lin, G., and Chou, M. W. (2002) Genotoxic pyrrolizidine alkaloids —

mechanisms leading to DNA adduct formation and tumorigenicity. Int. J. Mol. Sci., 3, 948-964. (19)

He, X., Ma, L., Xia, Q., and Fu, P. P. (2016) 7-N-Acetylcysteine-pyrrole conjugate - a

potent DNA reactive metabolite of pyrrolizidine alkaloids. Journal of Food and Drug Analysis, in press. (20)

He, X., Xia, Q., Ma, L., and Fu, P. P. (2016) 7-Cysteinyl-Pyrrole Conjugate-a New

Potential DNA Reactive Metabolite of Pyrrolizidine Alkaloids. Journal of Environmental Science and Health, Part C, 34, 57-76. (21)

Xia, Q., Ma, L., He, X., Cai, L., and Fu, P. P. (2015) 7-Glutathione pyrrole adduct: a

potential DNA reactive metabolite of pyrrolizidine alkaloids. Chem. Res. Toxicol., 28, 615-620. (22)

Xia, Q., Zhao, Y., Von Tungeln, L. S., Doerge, D. R., Lin, G., Cai, L., and Fu, P. P.

(2013) Pyrrolizidine alkaloid-derived DNA adducts as a common biological biomarker of pyrrolizidine alkaloid-induced tumorigenicity. Chem. Res. Toxicol., 26, 1384-1396.

28 ACS Paragon Plus Environment

Page 28 of 39

Page 29 of 39

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

(23)

Yang, Y.-C., Yan, J., Doerge, D. R., Chan, P.-C., Fu, P. P., and Chou, M. W. (2001)

Metabolic activation of the tumorigenic pyrrolizidine alkaloid, riddelliine, leading to DNA adduct formation in vivo. Chem. Res. Toxicol., 14, 101-109. (24)

Zhao, Y., Xia, Q., Gamboa da Costa, G., Yu, H., Cai, L., and Fu, P. P. (2012) Full

structure assignments of pyrrolizidine alkaloid DNA adducts and mechanism of tumor initiation. Chem. Res. Toxicol., 25, 1985-1996. (25)

Bodi, D., Ronczka, S., Gottschalk, C., Behr, N., Skibba, A., Wagner, M., Lahrssen-

Wiederholt, M., Preiss-Weigert, A., and These, A. (2014) Determination of pyrrolizidine alkaloids in tea, herbal drugs and honey. Food additives & contaminants. Part A, Chemistry, analysis, control, exposure & risk assessment, 31, 1884-1895. (26)

Edgar, J. A., Lin, H. J., Kumana, C. R., and Ng, M. M. (1992) Pyrrolizidine alkaloid

composition of three Chinese medicinal herbs, Eupatorium cannabinum, E. japonicum and Crotalaria assamica. Am J Chin Med, 20, 281-288. (27)

Griffin, C. T., Gosetto, F., Danaher, M., Sabatini, S., and Furey, A. (2014) Investigation

of targeted pyrrolizidine alkaloids in traditional Chinese medicines and selected herbal teas sourced in Ireland using LC-ESI-MS/MS. Food Addit Contam Part A Chem Anal Control Expo Risk Assess, 31, 940-961. (28)

Roeder, E. (2000) Medicinal plants in China containing pyrrolizidine alkaloids.

Pharmazie, 55, 711-726. (29)

Chou, M. W., and Fu, P. P. (2006) Formation of DHP-derived DNA adducts in vivo from

dietary supplements and Chinese herbal plant extracts containing carcinogenic pyrrolizidine alkaloids. Toxicol. Ind. Health, 22, 321-327. (30)

Mathon, C., Edder, P., Bieri, S., and Christen, P. (2014) Survey of pyrrolizidine alkaloids

in teas and herbal teas on the Swiss market using HPLC-MS/MS. Anal Bioanal Chem. (31)

Molyneux, R. J., Gardner, D. L., Colegate, S. M., and Edgar, J. A. (2011) Pyrrolizidine

alkaloid toxicity in livestock: a paradigm for human poisoning? Food Addit. Contam. Part A Chem Anal Control Expo Risk Assess, 28, 293-307. (32)

Roeder, E. (1995) Medicinal plants in Europe containing pyrrolizidine alkaloids.

Pharmazie, 50, 83-98.

29 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(33)

Edgar, J. A., and Smith, L. W., Eds. (2000) Transfer of pyrrolizidine alkaloids into eggs:

food safety implications., Vol. ACS Symposium Series 745, American Chemical Society, Washington D.C. (34)

Griffin, C. T., Danaher, M., Elliott, C. T., Glenn Kennedy, D., and Furey, A. (2013)

Detection of pyrrolizidine alkaloids in commercial honey using liquid chromatography–ion trap mass spectrometry. Food chemistry, 136, 1577-1583. (35)

Kempf, M., Reinhard, A., and Beuerle, T. (2010) Pyrrolizidine alkaloids (PAs) in honey

and pollen-legal regulation of PA levels in food and animal feed required. Mol Nutr Food Res, 54, 158-168. (36)

Molyneux, R. J., Johnson, A. E., Olsen, J. D., and Baker, D. C. (1991) Toxicity of

pyrrolizidine alkaloids from Riddell groundsel (Senecio riddellii) to cattle. Am J Vet Res, 52, 146-151. (37)

Puscas, A., Hosu, A., and Cimpoiu, C. (2013) Application of a newly developed and

validated high-performance thin-layer chromatographic method to control honey adulteration. Journal of Chromatography A, 1272, 132-135. (38)

Edgar, J. A., Colegate, S. M., Boppre, M., and Molyneux, R. J. (2011) Pyrrolizidine

alkaloids in food: a spectrum of potential health consequences. Food Addit Contam Part A Chem Anal Control Expo Risk Assess, 28, 308-324. (39)

Panter, K., and James, L. (1990) Natural plant toxicants in milk: a review. Journal of

animal science, 68, 892-904. (40)

IPCS. (1989) Pyrrolizidine Alkaloids Health and Safety Guide, In Health and Safety

Criteria Guide 26., International Programme on Chemical Safety, WHO: Geneva. (41)

Creeper, J. H., Mitchell, A. A., Jubb, T. F., and Colegate, S. M. (1999) Pyrrolizidine

alkaloid poisoning of horses grazing a native heliotrope (Heliotropium ovalifolium). Aust Vet J, 77, 401-402. (42)

Fu, P. P., Chiang, H. M., Xia, Q., Chen, T., Chen, B. H., Yin, J. J., Wen, K. C., Lin, G.,

and Yu, H. (2009) Quality assurance and safety of herbal dietary supplements. Journal of environmental science and health. Part C, Environmental carcinogenesis & ecotoxicology reviews, 27, 91-119.

30 ACS Paragon Plus Environment

Page 30 of 39

Page 31 of 39

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

(43)

Harper, P. A., Walker, K. H., Krahenbuhl, R. E., and Christie, B. M. (1985) Pyrrolizidine

alkaloid poisoning in calves due to contamination of straw by Heliotropium europeum. Aust Vet J, 62, 382-383. (44)

Hill, B. D., Gaul, K. L., and Noble, J. W. (1997) Poisoning of feedlot cattle by seeds of

Heliotropium europaeum. Aust Vet J, 75, 360-361. (45)

Shimshoni, J. A., Mulder, P. P., Bouznach, A., Edery, N., Pasval, I., Barel, S., Abd-El

Khaliq, M., and Perl, S. (2015) Heliotropium europaeum Poisoning in Cattle and Analysis of Its Pyrrolizidine Alkaloid Profile. Journal of agricultural and food chemistry, 63, 1664-1672. (46)

Willmot, F. C., and Robertson, G. W. (1920) Senecio disease, or cirrhosis of the liver due

to Senecio poisoning. Lancet, 196, 848-849. (47)

Schoental, R., Ed. (1976) Carcinogens in plants and microorganisms, Vol. ACS

Monograph 173, American Chemical Society, Washington, DC. (48)

Kedzierski, B., and Buhler, D. R. (1985) Configuration of necine pyrroles--toxic

metabolites of pyrrolizidine alkaloids. Toxicol. Lett., 25, 115-119. (49)

Ruan, J., Liao, C., Ye, Y., and Lin, G. (2014) Lack of metabolic activation and

predominant formation of an excreted metabolite of nontoxic platynecine-type pyrrolizidine alkaloids. Chem Res Toxicol, 27, 7-16. (50)

Reid, M. J., Lame, M. W., Morin, D., Wilson, D. W., and Segall, H. J. (1998)

Involvement of cytochrome P450 3A in the metabolism and covalent binding of 14Cmonocrotaline in rat liver microsomes. J Biochem Mol Toxicol, 12, 157-166. (51)

Ruan, J., Gao, H., Li, N., Xue, J., Chen, J., Ke, C., Ye, Y., Fu, P. P.-C., Zheng, J., Wang,

J., and Lin, G. (2015) Blood pyrrole-protein adducts–A biomarker of pyrrolizidine alkaloidinduced liver injury in humans. J. Environ. Sci. Health, Part C, 33, 404-421. (52)

Miranda, C. L., Chung, W., Reed, R. E., Zhao, X., Henderson, M. C., Wang, J. L.,

Williams, D. E., and Buhler, D. R. (1991) Flavin-containing monooxygenase: a major detoxifying enzyme for the pyrrolizidine alkaloid senecionine in guinea pig tissues. Biochem Biophys Res Commun, 178, 546-552. (53)

Chou, M. W., Wang, Y. P., Yan, J., Yang, Y.-C., Beger, R. D., Williams, L. D., Doerge,

D. R., and Fu, P. P. (2003) Riddelliine N-oxide is a phytochemical and mammalian metabolite with genotoxic activity that is comparable to the parent pyrrolizidine alkaloid riddelliine. Toxicol. Lett., 145, 239-247. 31 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(54)

Mattocks, A. (1971) Hepatotoxic effects due to pyrrolizidine alkaloid N-oxides.

Xenobiotica, 1, 563-565. (55)

Wang, Y. P., Yan, J., Fu, P. P., and Chou, M. W. (2005) Human liver microsomal

reduction of pyrrolizidine alkaloid N-oxides to form the corresponding carcinogenic parent alkaloid. Toxicol Lett, 155, 411-420. (56)

Schoental, R., Head, M. A., and Peacock, P. R. (1954) Senecio alkaloids: Primary liver

tumours in rats as a result of treatment with (1) a mixture of alkaloids from S. jacobaea lin.; (2) retrorsine; (3) isatidine. Br. J. Cancer, 8, 458-465. (57)

Harris, P. N., and Chen, K. K. (1970) Development of hepatic tumors in rats following

ingestion of senecio longilobus. Cancer Res., 30, 2881-2886. (58)

Schoental, R. (1957) Hepatotoxic action of pyrrolizidine (Senecio) alkaloids in relation to

their structure. Nature, 179, 361-363. (59)

Schoental, R., Hard, G. C., and Gibbard, S. (1971) Histopathology of renal lipomatous

tumors in rats treated with the "natural" products, pyrrolizidine alkaloids and , -unsaturated aldehydes. J Natl Cancer Inst, 47, 1037-1044. (60)

Brandange, S., Luning, B., Moberg, C., and Sjostrand, E. (1970) Studies on orchidaceae

alkaloids. XXIV. A pyrrolizidine alkaloid from Phalaenopsis cornu-cervi Rchb. f. Acta Chem Scand, 25, 349-350. (61)

Allen, J. R., Hsu, I. C., and Carstens, L. A. (1975) Dehydroretronecine-induced

rhabdomyosarcomas in rats. Cancer Res., 35, 997-1002. (62)

Shumaker, R. C., Robertson, K. A., Hsu, I. C., and Allen, J. R. (1976) Neoplastic

transformation in tissues of rats exposed to monocrotaline or dehydroretronecine. J Natl Cancer Inst, 56, 787-790. (63)

Hirono, I., Mori, H., and Culvenor, C. C. (1976) Carcinogenic activity of coltsfoot,

Tussilago farfara L. Gann, 67, 125-129. (64)

Cook, J. W., Duffy, E., and Schoental, R. (1950) Primary liver tumours in rats following

feeding with alkaloids of Senecio jacobaea. Br J Cancer, 4, 405-410. (65)

Hirono, I., Haga, M., Fujii, M., Matsuura, S., Matsubara, N., Nakayama, M., Furuya, T.,

Hikichi, M., Takanashi, H., Uchida, E., Hosaka, S., and Ueno, I. (1979) Induction of hepatic tumors in rats by senkirkine and symphytine. J. Natl. Cancer Inst., 63, 469-472.

32 ACS Paragon Plus Environment

Page 32 of 39

Page 33 of 39

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

(66)

Hirono, I., Mori, H., and Haga, M. (1978) Carcinogenic activity of symphytum officinale.

J. Natl. Cancer Inst., 61, 865-868. (67)

Rao, M. S., Jago, M. V., and Reddy, J. K. (1983) Effect of calorie restriction on the fate

of hyperplastic liver nodules induced by concurrent administration of lasiocarpine and thioacetamide. Hum Toxicol, 2, 15-26. (68)

Rao, M. S., and Reddy, J. K. (1978) Malignant neoplasms in rats fed lasiocarpine. Br J

Cancer, 37, 289-293. (69)

Svoboda, D. J., and Reddy, J. K. (1974) Lasiocarpine-induced, transplantable squamous

cell carcinoma of rat skin. J Natl Cancer Inst, 53, 1415-1418. (70)

Svoboda, D. J., and Reddy, J. K. (1972) Malignant tumors in rats given lasiocarpine.

Cancer Res, 32, 908-913. (71)

Schoental, R. (1975) Pancreatic islet-cell and other tumors in rats given heliotrine, a

monoester pyrrolizidine alkaloid, and nicotinamide. Cancer Res, 35, 2020-2024. (72)

Kuhara, K., Takanashi, H., Hirono, I., Furuya, T., and Asada, Y. (1980) Carcinogenic

activity of clivorine, a pyrrolizidine alkaloid isolated from Ligularia dentata. Cancer Lett., 10, 117-122. (73)

Lin, G., Cui, Y. Y., Liu, X. Q., and Wang, Z. T. (2002) Species differences in the in vitro

metabolic activation of the hepatotoxic pyrrolizidine alkaloid clivorine. Chem Res Toxicol, 15, 1421-1428. (74)

Hirono, I., Ueno, I., Aiso, S., Yamaji, T., and Haga, M. (1983) Carcinogenic activity of

Farfugium japonicum and Senecio cannabifolius. Cancer Lett, 20, 191-198. (75)

Furuya, T., Hikichi, M., and Iitaka, Y. (1976) Fukinotoxin, a new pyrrolizidine alkaloid

from Petasites japonicus. Chem Pharm Bull (Tokyo), 24, 1120-1122. (76)

Hirono, I., Mori, H., Yamada, K., Hirata, Y., and Haga, M. (1977) Carcinogenic activity

of petasitenine, a new pyrrolizidine alkaloid isolated from Petasites japonicus Maxim. J Natl Cancer Inst, 58, 1155-1157. (77)

NTP. (2003) TR-508 Toxicology and carcinogenesis studies of riddelliine (CAS No.

23246-96-0) in F344/N rats and B6C3F1 mice (gavage studies) (U.S. Department of Health and Human Services, P. H. S., National Institutes of Health, Ed.), NIH Publication No. 03-4442., Research Triangle Park, NC.

33 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(78)

NTP. (1978) TR-39 Bioassay of Lasiocarpine for Possible Carcinogenicity (CAS No.

303-34-4), (U.S. Department of Health and Human Services, P. H. S., National Institutes of Health, Ed.), NIH Publication No. 78-839, Research Triangle Park, NC. (79)

NTP. (2014) Riddelliine in Report on Carcinogens Thirteenth Edition National

Toxicololgy Program (Services, U. S. D. o. H. a. H., Ed.), , Research Triangle Park, NC. (80)

IARC. (2002) Some Traditional Herbal Medicines, Some Mycotoxins, Naphthalene and

Styrene, In IARC Monographs on the evaluation of carcinogenic risks of chemicals to humans pp 153-168, International Agency for Research in Cancer, Lyon, France. (81)

IARC. (1976) Pyrrolizidine alkaloids, In IARC Monograph on the evaluation of

carcinogenic risk of chemicals to man - Some naturally occurring substance, International Agency for Research in Cancer, Lyon, France. (82)

Griffin, D. S., and Segall, H. J. (1989) Effects of the pyrrolizidine alkaloid senecionine

and the alkenals trans-4-OH-hexenal and trans-2-hexenal on intracellular calcium compartmentation in isolated hepatocytes. Biochem Pharmacol, 38, 391-397. (83)

Griffin, D. S., and Segall, H. J. (1986) Genotoxicity and cytotoxicity of selected

pyrrolizidine alkaloids, a possible alkenal metabolite of the alkaloids, and related alkenals. Toxicol Appl Pharmacol, 86, 227-234. (84)

Segall, H. J., Wilson, D. W., Dallas, J. L., and Haddon, W. F. (1985) trans-4-Hydroxy-2-

hexenal: a reactive metabolite from the macrocyclic pyrrolizidine alkaloid senecionine. Science, 229, 472-475. (85)

Winter, C. K., Segall, H. J., and Haddon, W. F. (1986) Formation of cyclic adducts of

deoxyguanosine with the aldehydes trans- 4-hydroxy-2-hexenal and trans-4-hydroxy-2-nonenal in vitro. Cancer Res, 46, 5682-5686. (86)

Hincks, J. R., and Coulombe, R. A., Jr. (1989) Rapid detection of DNA-interstrand and

DNA-protein cross-links in mammalian cells by gravity-flow alkaline elution. Environ Mol Mutagen, 13, 211-217. (87)

Kim, H.-Y., Stermitz, F. R., Li, J. K.-K., and Coulombe, R. A. J. (1999) Comparative

DNA cross-linking by activated pyrrolizidine alkaloids. Food Chem. Toxicol., 37, 619-625. (88)

Pereira, T. N., Webb, R. I., Reilly, P. E., Seawright, A. A., and Prakash, A. S. (1998)

Dehydromonocrotaline generates sequence-selective N-7 guanine alkylation and heat and alkali stable multiple fragment DNA crosslinks. Nucleic Acids Res, 26, 5441-5447. 34 ACS Paragon Plus Environment

Page 34 of 39

Page 35 of 39

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

(89)

Petry, T. W., Bowden, G. T., Huxtable, R. J., and Sipes, I. G. (1984) Characterization of

hepatic DNA damage induced in rats by the pyrrolizidine alkaloid monocrotaline. Cancer Res, 44, 1505-1509. (90)

Reed, R. L., Ahern, K. G., Pearson, G. D., and Buhler, D. R. (1988) Crosslinking of DNA

by dehydroretronecine, a metabolite of pyrrolizidine alkaloids. Carcinogenesis, 9, 1355-1361. (91)

Tepe, J. J., and Williams, R. M. (1999) DNA cross-linking by phototriggered

dehydromonocrotaline progenitor. J of the American Chemical Society, 121, 2951-2955. (92)

White, I. N., and Mattocks, A. R. (1972) Reaction of dihydropyrrolizines with

deoxyribonucleic acids in vitro. Biochem J, 128, 291-297. (93)

Kim, H. Y., Stermitz, F. R., Molyneux, R. J., Wilson, D. W., Taylor, D., and Coulombe,

R. A. (1993) Structural influences on pyrrolizidine alkaloid-induced cytopathology. Toxicol. Appl. Pharmacol., 122, 61-69. (94)

Niwa, H., Ogawa, T., Okamoto, O., and Yamada, K. (1991) Alkylation of nucleosides by

dehydromonocrotaline, the putative toxic metabolite of the carcinogenic pyrrolizidine alkaloid monocrotaline. Tetrahedron letters, 32, 927-930. (95)

Robertson, K. A. (1982) Alkylation of N2 in deoxyguanosine by dehydroretronecine, a

carcinogenic metabolite of the pyrrolizidine alkaloid monocrotaline. Cancer Res, 42, 8-14. (96)

Wickrammanayake, P. P., Arbogast, B. L., and al., e. (1985) Alkylation of nucleosides

and nucleotides by dehydroretronecine; Characterization of covalent adducts by liquid secondary ion mass spectrometry. J. Am. Chem. Soc., 107, 2485-2488. (97)

Curtain, C. C., and Edgar, J. A. (1976) The binding of dehydroheliotridine to DNA and

the effect of it and other compounds on repair synthesis in main and satellite band DNA. Chem Biol Interact, 13, 243-256. (98)

Chou, M. W., Yan, J., Nichols, J., Xia, Q., Beland, F. A., Chan, P.-C., and Fu, P. P.

(2003) Correlation of DNA adduct formation and riddelliine-induced liver tumorigenesis in F344 rats and B6C3F1 mice. Cancer Lett., 193, 119-125. (99)

Chou, M. W., Yan, J., Williams, L., Xia, Q., Churchwell, M., Doerge, D. R., and Fu, P. P.

(2003) Identification of DNA adducts derived from riddelliine, a carcinogenic pyrrolizidine alkaloid. Chem. Res. Toxicol., 16, 1130-1137.

35 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(100) Mei, N., Chou, M. W., Fu, P. P., Heflich, R. H., and Chen, T. (2004) Differential mutagenicity of riddelliine in liver endothelial and parenchymal cells of transgenic big blue rats. Cancer Lett., 215, 151-158. (101) Hong, H. L., Ton, T. V., Devereux, T. R., Moomaw, C., Clayton, N., Chan, P., Dunnick, J. K., and Sills, R. C. (2003) Chemical-specific alterations in ras, p53, and β-catenin genes in hemangiosarcomas from B6C3F1 mice exposed to o-nitrotoluene or riddelliine for 2 years. Toxicol. Appl. Pharmacol., 191, 227-234. (102) Mei, N., Heflich, R. H., Chou, M. W., and Chen, T. (2004) Mutations induced by the carcinogenic pyrrolizidine alkaloid riddelliine in the liver cII gene of transgenic Big Blue rats. Chem. Res. Toxicol., 17, 814-818. (103) Nyska, A., Moomaw, C. R., Foley, J. F., Maronpot, R. R., Malarkey, D. E., Cummings, C. A., Peddada, S., Moyer, C. F., Allen, D. G., Travlos, G., and Chan, P. C. (2002) The hepatic endothelial carcinogen riddelliine induces endothelial apoptosis, mitosis, S phase, and p53 and hepatocytic vascular endothelial growth factor expression after short-term exposure. Toxicol Appl Pharmacol, 184, 153-164. (104) Xia, Q., Chou, M. W., Kadlubar, F. F., Chan, P.-C., and Fu, P. P. (2003) Human liver microsomal metabolism and DNA adduct formation of the tumorigenic pyrrolizidine alkaloid, riddelliine. Chem Res Toxicol, 16, 66-73. (105) Yang, Y., Yan, J., Churchwell, M., Beger, R., Chan, P., Doerge, D. R., Fu, P. P., and Chou, M. W. (2001) Development of a (32)P-postlabeling/HPLC method for detection of dehydroretronecine-derived DNA adducts in vivo and in vitro. Chem Res Toxicol, 14, 91-100. (106) Wang, Y. P., Fu, P. P., and Chou, M. W. (2005) Metabolic activation of the tumorigenic pyrrolizidine alkaloid, retrorsine, leading to DNA adduct formation in vivo. Int J Environ Res Public Health, 2, 74-79. (107) Wang, Y. P., Yan, J., Beger, R. D., Fu, P. P., and Chou, M. W. (2005) Metabolic activation of the tumorigenic pyrrolizidine alkaloid, monocrotaline, leading to DNA adduct formation in vivo. Cancer Lett, 226, 27-35. (108) Xia, Q., Chou, M. W., Edgar, J. A., Doerge, D. R., and Fu, P. P. (2006) Formation of DHP-derived DNA adducts from metabolic activation of the prototype heliotridine-type pyrrolizidine alkaloid, lasiocarpine. Cancer Lett., 231, 138-145.

36 ACS Paragon Plus Environment

Page 36 of 39

Page 37 of 39

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

(109) Xia, Q., Chou, M. W., Lin, G., and Fu, P. P. (2004) Metabolic formation of DHP-derived DNA adducts from a representative otonecine type pyrrolizidine alkaloid clivorine and the extract of Ligularia hodgsonnii hook. Chem. Res. Toxicol., 17, 702-708. (110) Xia, Q., Yan, J., Chou, M. W., and Fu, P. P. (2008) Formation of DHP-derived DNA adducts from metabolic activation of the prototype heliotridine-type pyrrolizidine alkaloid, heliotrine. Toxicol Lett, 178, 77-82. (111) Yan, J., Nichols, J., Yang, Y. C., Fu, P. P., and Chou, M. W. (2002) Detection of riddelliine-derived DNA adducts in blood of rats fed riddelliine. Intl J Mol Sci, 3, 1019-1026. (112) Yan, J., Xia, Q., Chou, M. W., and Fu, P. P. (2008) Metabolic activation of retronecine and retronecine N-oxide - formation of DHP-derived DNA adducts. Toxicol Ind Health, 24, 181188. (113)

DeLeve, L. D., Ito, Y., Bethea, N. W., McCuskey, M. K., Wang, X., and McCuskey, R.

S. (2003) Embolization by sinusoidal lining cells obstructs the microcirculation in rat sinusoidal obstruction syndrome. Am. J. Physiol. Gastrointest. Liver Physiol. 284, G1045–G1052. (114) Mattocks, A. R., and Jukes, R. (1992) Detection of sulphur-conjugated pyrrolic metabolites in blood and fresh or fixed liver tissue from rats given a variety of toxic pyrrolizidine alkaloids. Toxicol. Lett., 63, 47-55. (115) Mattocks, A. R., and Jukes, R. (1992) Chemistry of sulphur-bound pyrrolic metabolites in the blood of rats given different types of pyrrolizidine alkaloid. Nat. Toxins, 1, 89-95. (116) Mattocks, A. R., and Jukes, R. (1990) Recovery of the pyrrolic nucleus of pyrrolizidine alkaloid metabolites from sulphur conjugates in tissues and body fluids. Chem. Biol. Interact., 75, 225-239. (117) Winter, H., Seawright, A. A., Hrdlicka, J., Mattocks, A. R., Jukes, R., Wangdi, K., and Gurung, K. B. (1993) Pyrrolizidine alkaloid poisoning of yaks: diagnosis of pyrrolizidine alkaloid exposure by the demonstration of sulphur-conjugated pyrrolic metabolites of the alkaloid in circulating haemoglobin. Aust. Vet. J., 70, 312-313. (118) Mattocks, A. R., and White, I. N. H. (1970) Estimation of metabolites of pyrrolizidine alkaloids in animal tissues. Anal. Biochem., 38, 529-535. (119) Xia, Q., Zhao, Y., Lin, G., Beland, F. A., Cai, L., and Fu, P. P. (2016) Pyrrolizidine Alkaloid-Protein Adducts-Potential Non-Invasive Biomarkers of Pyrrolizidine Alkaloid-Induced Liver Toxicity and Exposure. Chemical Research in Toxicology, 29, 1282-1292. 37 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(120) Cooper, R. A., and Huxtable, R. J. (1996) A simple procedure for determining the aqueous half-lives of pyrrolic metabolites of pyrrolizidine alkaloids. Toxicon, 34, 604-607. (121) Buhler, D. R., Miranda, C. L., Kedzierski, B., and Reed, R. L. (1991) Mechanisms for pyrrolizidine alkaloid activation and detoxification. Adv Exp Med Biol, 283, 597-603. (122) Lame, M. W., Morin, D., Jones, A. D., Segall, H. J., and Wilson, D. W. (1990) Isolation and identification of a pyrrolic glutathione conjugate metabolite of the pyrrolizidine alkaloid monocrotaline. Toxicol Lett, 51, 321-329. (123) Lin, G., Zhou, K. Y., Zhao, X. G., Wang, Z. T., and But, P. P. (1998) Determination of hepatotoxic pyrrolizidine alkaloids by on-line high performance liquid chromatography mass spectrometry with an electrospray interface. Rapid Commun Mass Spectrom, 12, 1445-1456. (124) Mattocks, A. R., Croswell, S., Jukes, R., and Huxtable, R. J. (1991) Identity of a biliary metabolite formed from monocrotaline in isolated, perfused rat liver. Toxicon, 29, 409-415. (125) Reed, R. L., Miranda, C. L., Kedzierski, B., Henderson, M. C., and Buhler, D. R. (1992) Microsomal formation of a pyrrolic alcohol glutathione conjugate of the pyrrolizidine alkaloid senecionine. Xenobiotica, 22, 1321-1327. (126) Estep, J. E., Lame, M. W., Jones, A. D., and Segall, H. J. (1990) N-acetylcysteineconjugated pyrrole identified in rat urine following administration of two pyrrolizidine alkaloids, monocrotaline and senecionine. Toxicol Lett, 54, 61-69. (127) Segall, H. J., Dallas, J. L., and Haddon, W. F. (1984) Two dihydropyrrolizine alkaloid metabolites isolated from mouse hepatic microsomes in vitro. Drug Metab Dispos, 12, 68-71. (128) Li, W., Wang, K., Lin, G., Peng, Y., and Zheng, J. (2016) Lysine adduction by reactive metabolite (s) of monocrotaline. Chemical Research in Toxicology, 29, 333-341. (129) Chen, M., Li, L., Zhong, D., Shen, S., Zheng, J., and Chen, X. (2016) 9-Glutathionyl-6, 7-dihydro-1-hydroxymethyl-5 H-pyrrolizine Is the Major Pyrrolic Glutathione Conjugate of Retronecine-Type Pyrrolizidine Alkaloids in Liver Microsomes and in Rats. Chemical research in toxicology, 29, 180-189. (130) Sun, P. S., Hsia, M. T., Chu, F. S., and Allen, J. R. (1977) Inhibition of yeast alcohol dehydrogenase by dehydroretronecine. Food Cosmet Toxicol, 15, 419-422. (131) Mattocks, A. R., and Bird, I. (1983) Alkylation by dehydroretronecine, a cytotoxic metabolite of some pyrrolizidine alkaloids: an in vitro test. Toxicol Lett, 16, 1-8.

38 ACS Paragon Plus Environment

Page 38 of 39

Page 39 of 39

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

(132) Ma, L., Zhao, H., Xia, Q., Cai, L., and Fu, P. P. (2015) Synthesis and phototoxicity of isomeric 7, 9-diglutathione pyrrole adducts: Formation of reactive oxygen species and induction of lipid peroxidation. Journal of Food and Drug Analysis, 23, 577-586. (133) Tamta, H., Pawar, R. S., Wamer, W. G., Grundel, E., Krynitsky, A. J., and Rader, J. I. (2012) Comparison of metabolism-mediated effects of pyrrolizidine alkaloids in a HepG2/C3A cell-S9 co-incubation system and quantification of their glutathione conjugates. Xenobiotica, 42, 1038-1048. (134) Zhao, Y., Wang, S., Xia, Q., Gamboa da Costa, G., Doerge, D. R., Cai, L., and Fu, P. P. (2014) Reaction of dehydropyrrolizidine alkaloids with valine and hemoglobin. Chem. Res. Toxicol., 27, 1720-1731. (135) Merz, K. H., and Schrenk, D. (2016) Interim relative potency factors for the toxicological risk assessment of pyrrolizidine alkaloids in food and herbal medicines. Toxicol Lett. (136) Rieben, W. K., Jr., and Coulombe, R. A., Jr. (2004) DNA cross-linking by dehydromonocrotaline lacks apparent base sequence preference. Toxicol Sci, 82, 497-503. (137) Fashe, M. M., Juvonen, R. O., Petsalo, A., Rahnasto-Rilla, M., Auriola, S., Soininen, P., Vepsäläinen, J., and Pasanen, M. (2014) Identification of a New Reactive Metabolite of Pyrrolizidine Alkaloid Retrorsine:(3 H-Pyrrolizin-7-yl) methanol. Chemical research in toxicology, 27, 1950-1957.

Biography Peter P. Fu received his Ph.D. degree in Chemistry at the University of Illinois at Chicago in 1973, he worked at the Ben May Institute for Cancer Research, University of Chicago on the mechanism by which polycyclic aromatic hydrocarbons induce cancer. Since working at the Biochemical Toxicology Division, National Center for Toxicological Research (NCTR) in 1979, his mechanistic studies on tumor induction provided mechanism-based information highly useful for subsequent carcinogenic risk assessments and regulatory decisions by FDA. His on-going research focuses on mechanistic studies of the genotoxicity and tumorigenicity of pyrrolizidine alkaloids, nanomaterials, and herbal dietary supplements of FDA interest.

39 ACS Paragon Plus Environment