1
|
Yoda T, Tochitani T, Usui T, Kouchi M, Inada H, Hosaka T, Kanno Y, Miyawaki I, Yoshinari K. Involvement of the CYP1A1 inhibition-mediated activation of aryl hydrocarbon receptor in drug-induced hepatotoxicity. J Toxicol Sci 2022; 47:359-373. [PMID: 36047110 DOI: 10.2131/jts.47.359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Hepatotoxicity is one of the most common toxicities observed in non-clinical safety studies of drug candidates, and it is important to understand the hepatotoxicity mechanism to assess the risk of drug-induced liver injury in humans. In this study, we investigated the mechanism of hepatotoxicity caused by 2-[2-Methyl-1-(oxan-4-yl)-1H-benzimidazol-5-yl]-1,3-benzoxazole (DSP-0640), a drug candidate that showed hepatotoxicity characterized by centrilobular hypertrophy and vacuolation of hepatocytes in a 4-week oral repeated-dose toxicity study in male rats. In the liver of rats treated with DSP-0640, the expression of aryl hydrocarbon receptor (AHR) target genes, including Cyp1a1, was upregulated. In in vitro reporter assays, however, DSP-0640 showed only minimal AHR-activating potency. Therefore, we investigated the possibility that DSP-0640 indirectly activated AHR by inhibiting the CYP1 enzyme-dependent clearance of endogenous AHR agonists. In in vitro assays, DSP-0640 showed inhibitory effects on both rat and human CYP1A1 and enhanced rat and human AHR-mediated reporter gene expression induced by 6-formylindolo[3,2-b]carbazole, a well-known endogenous AHR agonist. The possible involvement of CYP1A1 inhibition in AHR activation was also demonstrated with other hepatotoxic compounds tacrine and albendazole. These results suggest that CYP1A1 inhibition-mediated AHR activation is involved in the hepatotoxicity caused by DSP-0640 and that DSP-0640 might induce hepatotoxicity in humans as well. We propose that CYP1A1 inhibition-mediated AHR activation is a novel mechanism for drug-induced hepatotoxicity.
Collapse
Affiliation(s)
- Tomomi Yoda
- Preclinical Research Unit, Sumitomo Pharma Co., Ltd.,Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka
| | | | - Toru Usui
- Preclinical Research Unit, Sumitomo Pharma Co., Ltd
| | - Mami Kouchi
- Preclinical Research Unit, Sumitomo Pharma Co., Ltd
| | | | - Takuomi Hosaka
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka
| | - Yuichiro Kanno
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka
| | | | - Kouichi Yoshinari
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka
| |
Collapse
|
2
|
Coe KJ, Feinstein M, Higgins JW, Leung P, Scott BP, Skaptason J, Tam Y, Volak LP, Kinong J, Bittner A, McAllister H, Lim NM, Hack M, Koudriakova T. Characterization of JNJ-2482272 [4-(4-Methyl-2-(4-(Trifluoromethyl)Phenyl)Thiazole-5-yl) Pyrimidine-2-Amine] As a Strong Aryl Hydrocarbon Receptor Activator in Rat and Human. Drug Metab Dispos 2022; 50:1064-1076. [PMID: 35680134 DOI: 10.1124/dmd.121.000825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 05/18/2022] [Indexed: 11/22/2022] Open
Abstract
[4-(4-Methyl-2-(4-(trifluoromethyl)phenyl)thiazole-5-yl)pyrimidine-2-amine] (JNJ-2482272), under investigation as an anti-inflammatory agent, was orally administered to rats once daily at 60 mg/kg for 6 consecutive days. Despite high plasma exposure after single administration (Cmax of 7.1 μM), JNJ-2482272 had plasma concentrations beneath the lower limit of quantification (3 ng/ml) after 6 consecutive days of dosing. To determine if JNJ-2482272 is an autoinducer in rats, plated rat hepatocytes were treated with JNJ-2482272 for 2 days. The major hydroxylated metabolites of JNJ-2482272 were isolated and characterized by mass spectrometry and NMR analyses. Compared with the vehicle-treated cells, a concentration-dependent increase was observed in the formation of phase I- and II-mediated metabolites coinciding with greater expression of cytochrome P450s (P450s) and UDP-glucuronosyltransferases (UGTs) in rat hepatocytes. CYP1A1, CYP1A2, CYP1B1, and UGT1A6 transcripts were predominantly induced, suggesting that JNJ-2482272 is an activator of the aryl hydrocarbon receptor (AhR). In a human AhR reporter assay, JNJ-2482272 demonstrated potent AhR activation with an EC50 value of 0.768 nM, a potency more comparable to the strong AhR activator and toxin 2,3,7,8-tetrachloro-dibenzodioxin than to weaker AhR activators 3-methylcholanthrene, β-naphthoflavone, and omeprazole. In plated human hepatocytes, JNJ-2482272 induced CYP1A1 gene expression with an EC50 of 20.4 nM and increased CYP1A activity >50-fold from basal levels. In human recombinant P450s, JNJ-2482272 was exclusively metabolized by the CYP1 family of enzymes and most rapidly by CYP1A1. The summation of these in vitro findings bridges the in vivo conclusion that JNJ-2482272 is a strong autoinducer in rats and potentially in humans through potent AhR activation. SIGNIFICANCE STATEMENT: Drugs that induce their own metabolism (autoinducers) can lack sustained exposures for pharmacology and safety assessment hindering their development. JNJ-2482272 is demonstrated herein as a strong aryl hydrocarbon receptor (AhR) activator and CYP1A autoinducer, explaining its near complete loss of exposure after repeat administration in rat, which is likely translatable to human (if progressed further) considering its nanomolar potency comparable to "classical" AhR ligands like 2,3,7,8-tetrachloro-dibenzo-dioxin despite bearing a "nonclassical" drug structure.
Collapse
Affiliation(s)
- Kevin J Coe
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Mark Feinstein
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - J William Higgins
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Perry Leung
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Brian P Scott
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Judy Skaptason
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Yuen Tam
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Laurie P Volak
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Jennifer Kinong
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Anton Bittner
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Heather McAllister
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Nathan M Lim
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Michael Hack
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Tatiana Koudriakova
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| |
Collapse
|
3
|
Ma X, Jin H, Chu X, Dai W, Tang W, Zhu J, Wang F, Yang X, Li W, Liu G, Yang X, Liang H. The Host CYP1A1-Microbiota Metabolic Axis Promotes Gut Barrier Disruption in Methicillin-Resistant Staphylococcus aureus-Induced Abdominal Sepsis. Front Microbiol 2022; 13:802409. [PMID: 35572636 PMCID: PMC9093654 DOI: 10.3389/fmicb.2022.802409] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 03/08/2022] [Indexed: 11/30/2022] Open
Abstract
Background Host-microbiota crosstalk has been implicated in multiple host metabolic pathway axes that regulate intestinal barrier function. Although constitutive cytochrome P4501A1 (CYP1A1) expression perturbs the microbiome-derived autoregulatory loop following enteric infection, little is known about the role of host CYP1A1 in modulating gut microbiome-mediated signaling during methicillin-resistant Staphylococcus aureus (MRSA)-induced abdominal sepsis and its effects on intestinal barrier integrity. Methods Abdominal sepsis was induced by the intraperitoneal injection of MRSA in mice. The effect of CYP1A1 deficiency on gut barrier integrity was investigated using RNA sequencing, microbiome analyses, and targeted metabolomics. The microbiota-produced metabolites were validated in patients with sepsis and persistent MRSA infection. Results Mice lacking CYP1A1 exhibited an altered gut microbiome, a reduced metabolic shift from lysine to cadaverine in the caecal contents and antimicrobial molecule production (Retnlb, Gbp7, and Gbp3), and they were protected against gut barrier disruption when subjected to MRSA challenge. These beneficial effects were validated in aryl hydrocarbon receptor (AHR) knockout (KO) mice by cohousing with CYP1A1 KO mice and abrogated after supplementation with cadaverine or Enterococcus faecalis, the primary microbiota genus for cadaverine synthesis. Antibiotic-driven gut dysbacteriosis impaired the survival benefit and disrupted the intestinal barrier integrity in CYP1A1 KO mice after MRSA infection. Furthermore, increased cadaverine levels in feces and serum were detected in critically ill patients with gut leakiness during persistent MRSA infection, whereas cadaverine was not detected in healthy controls. Additionally, microbiota-derived cadaverine induced enterocyte junction disruption by activating the histamine H4 receptor/nuclear factor-κB/myosin light-chain kinase signaling pathway. Conclusion This study revealed the unexpected function of host CYP1A1 in microbiota-mediated cadaverine metabolism, with crucial consequences for dysbacteriosis following MRSA-induced abdominal sepsis, indicating that inhibiting CYP1A1 or blocking cadaverine-histamine H4 receptor signaling could be a potential therapeutic target against abdominal sepsis. Clinical Trial Registration [http://www.chictr.org.cn/index.aspx], identifier [ChiCTR1800018646].
Collapse
Affiliation(s)
- Xiaoyuan Ma
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China
| | - Huaijian Jin
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China.,Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiang Chu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China
| | - Weihong Dai
- Trauma Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Wanqi Tang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China
| | - Junyu Zhu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China
| | - Fangjie Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China
| | - Xue Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China
| | - Wei Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China
| | - Guodong Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burn and Combined Injuries, Medical Center of Trauma and War Injuries, Daping Hospital, Army Medical University, Chongqing, China
| | - Xia Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China
| | - Huaping Liang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
4
|
Yu X, Chu Z, Li J, He R, Wang Y, Cheng C. Pharmacokinetic Drug-drug Interaction of Antibiotics Used in Sepsis Care in China. Curr Drug Metab 2021; 22:5-23. [PMID: 32990533 DOI: 10.2174/1389200221666200929115117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/17/2020] [Accepted: 07/07/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Many antibiotics have a high potential for interactions with drugs, as a perpetrator and/or victim, in critically ill patients, and particularly in sepsis patients. METHODS The aim of this review is to summarize the pharmacokinetic drug-drug interaction (DDI) of 45 antibiotics commonly used in sepsis care in China. Literature search was conducted to obtain human pharmacokinetics/ dispositions of the antibiotics, their interactions with drug-metabolizing enzymes or transporters, and their associated clinical drug interactions. Potential DDI is indicated by a DDI index ≥ 0.1 for inhibition or a treatedcell/ untreated-cell ratio of enzyme activity being ≥ 2 for induction. RESULTS The literature-mined information on human pharmacokinetics of the identified antibiotics and their potential drug interactions is summarized. CONCLUSION Antibiotic-perpetrated drug interactions, involving P450 enzyme inhibition, have been reported for four lipophilic antibacterials (ciprofloxacin, erythromycin, trimethoprim, and trimethoprim-sulfamethoxazole) and three antifungals (fluconazole, itraconazole, and voriconazole). In addition, seven hydrophilic antibacterials (ceftriaxone, cefamandole, piperacillin, penicillin G, amikacin, metronidazole, and linezolid) inhibit drug transporters in vitro. Despite no clinical PK drug interactions with the transporters, caution is advised in the use of these antibacterials. Eight hydrophilic antibiotics (all β-lactams; meropenem, cefotaxime, cefazolin, piperacillin, ticarcillin, penicillin G, ampicillin, and flucloxacillin), are potential victims of drug interactions due to transporter inhibition. Rifampin is reported to perpetrate drug interactions by inducing CYP3A or inhibiting OATP1B; it is also reported to be a victim of drug interactions, due to the dual inhibition of CYP3A4 and OATP1B by indinavir. In addition, three antifungals (caspofungin, itraconazole, and voriconazole) are reported to be victims of drug interactions because of P450 enzyme induction. Reports for other antibiotics acting as victims in drug interactions are scarce.
Collapse
Affiliation(s)
- Xuan Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zixuan Chu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jian Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Rongrong He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yaya Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Chen Cheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
5
|
Rannug A. How the AHR Became Important in Intestinal Homeostasis-A Diurnal FICZ/AHR/CYP1A1 Feedback Controls Both Immunity and Immunopathology. Int J Mol Sci 2020; 21:ijms21165681. [PMID: 32784381 PMCID: PMC7461111 DOI: 10.3390/ijms21165681] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 12/12/2022] Open
Abstract
Ever since the 1970s, when profound immunosuppression caused by exogenous dioxin-like compounds was first observed, the involvement of the aryl hydrocarbon receptor (AHR) in immunomodulation has been the focus of considerable research interest. Today it is established that activation of this receptor by its high-affinity endogenous ligand, 6-formylindolo[3,2-b]carbazole (FICZ), plays important physiological roles in maintaining epithelial barriers. In the gut lumen, the small amounts of FICZ that are produced from L-tryptophan by microbes are normally degraded rapidly by the inducible cytochrome P4501A1 (CYP1A1) enzyme. This review describes how when the metabolic clearance of FICZ is attenuated by inhibition of CYP1A1, this compound passes through the intestinal epithelium to immune cells in the lamina propria. FICZ, the level of which is thus modulated by this autoregulatory loop involving FICZ itself, the AHR and CYP1A1, plays a central role in maintaining gut homeostasis by potently up-regulating the expression of interleukin 22 (IL-22) by group 3 innate lymphoid cells (ILC3s). IL-22 stimulates various epithelial cells to produce antimicrobial peptides and mucus, thereby both strengthening the epithelial barrier against pathogenic microbes and promoting colonization by beneficial bacteria. Dietary phytochemicals stimulate this process by inhibiting CYP1A1 and causing changes in the composition of the intestinal microbiota. The activity of CYP1A1 can be increased by other microbial products, including the short-chain fatty acids, thereby accelerating clearance of FICZ. In particular, butyrate enhances both the level of the AHR and CYP1A1 activity by stimulating histone acetylation, a process involved in the daily cycle of the FICZ/AHR/CYP1A1 feedback loop. It is now of key interest to examine the potential involvement of FICZ, a major physiological activator of the AHR, in inflammatory disorders and autoimmunity.
Collapse
Affiliation(s)
- Agneta Rannug
- Karolinska Institutet, Institute of Environmental Medicine, 171 77 Stockholm, Sweden
| |
Collapse
|
6
|
Yamazoe Y, Yoshinari K. Prediction of regioselectivity and preferred order of CYP1A1-mediated metabolism: Solving the interaction of human and rat CYP1A1 forms with ligands on the template system. Drug Metab Pharmacokinet 2020; 35:165-185. [DOI: 10.1016/j.dmpk.2019.10.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 07/28/2019] [Accepted: 10/28/2019] [Indexed: 10/25/2022]
|
7
|
Forsch K, Schöning V, Assmann GM, Moser C, Siewert B, Butterweck V, Drewe J. In vitro hepatotoxicity of Petasites hybridus extract (Ze 339) depends on the concentration, the cytochrome activity of the cell system, and the species used. Phytother Res 2019; 34:184-192. [PMID: 31631423 PMCID: PMC7004140 DOI: 10.1002/ptr.6516] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/19/2019] [Accepted: 09/05/2019] [Indexed: 12/24/2022]
Abstract
Ze 339, a CO2 extract prepared from the leaves of Petasites hybridus, possesses antispasmodic and anti‐inflammatory effects and is proven to be effective in the treatment of allergic rhinitis. To study possible hepatotoxic effects of Ze 339, its main constituents and metabolites, a series of in vitro investigations were performed. Furthermore, different reconstituted fractions of extract (petasins and fatty acid fraction) were examined in three in vitro test systems using hepatocytes: Two human cell lines, with lower and higher activity of cytochrome P450 enzymes (HepG2, HepaRG) as well as a rodent cell line with high cytochrome P450 activity (H‐4‐II‐E), were used. Metabolic activity, assessed by the WST‐1 assay, was chosen as indicator of cytotoxicity. To assess potential bioactivation of Ze 339 compounds, metabolic experiments using S9 fractions from rats, dogs, and humans and isolated cytochromes (human/rat) were performed, and the formation of reactive metabolites was assessed by measuring cellular concentrations of glutathione and glutathione disulphide. Our data revealed that the cytotoxicity of Ze 339, its single constituents, and main metabolites depends on the concentration, the cytochrome activity of the cell system, and the species used.
Collapse
Affiliation(s)
- Kristina Forsch
- Preclinical Research, Max Zeller Söhne AG, Romanshorn, Switzerland
| | - Verena Schöning
- Preclinical Research, Max Zeller Söhne AG, Romanshorn, Switzerland
| | | | - Christin Moser
- Preclinical Research, Max Zeller Söhne AG, Romanshorn, Switzerland
| | - Beate Siewert
- Preclinical Research, Max Zeller Söhne AG, Romanshorn, Switzerland
| | | | - Jürgen Drewe
- Preclinical Research, Max Zeller Söhne AG, Romanshorn, Switzerland
| |
Collapse
|
8
|
Yamazoe Y, Yoshinari K. Prediction of regioselectivity and preferred order of metabolisms on CYP1A2-mediated reactions part 3: Difference in substrate specificity of human and rodent CYP1A2 and the refinement of predicting system. Drug Metab Pharmacokinet 2019; 34:217-232. [DOI: 10.1016/j.dmpk.2019.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/04/2018] [Accepted: 02/01/2019] [Indexed: 12/11/2022]
|
9
|
Liu X, Zhang L, Li J, Wang J, Meng G, Chi M, Zhao Y, Wu Y. Relative Effect Potency Estimates for Dioxin-Like Compounds in Pregnant Women with Gestational Diabetes Mellitus and Blood Glucose Outcomes Based on a Nested Case-control Study. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2019; 53:7792-7802. [PMID: 31149810 DOI: 10.1021/acs.est.9b00988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
To improve the applicability of the toxic equivalents principle for human health risk assessment, systemic relative effect potencies (REPs) for dioxin-like compounds (DLCs) deriving from human in vivo data are required. A prospective nested case-control study was performed to determine REPs from the human serum concentration of DLCs using gestational diabetes mellitus (GDM) and fasting blood glucose (FBG) as the end points of concern. Serum concentration of 29 DLCs from 77 cases and 154 controls were measured. Logistic and linear regression were used to estimate the effects of individual congeners on GDM and FBG, respectively. The REPs based on GDM and FBG were calculated from the ratios of regression coefficients, βi (DLCs)/βTCDD. Two sets of consistent human serum-based REPs, that is, GDM-REP and FBG-REP, were established and largely agree with REPs from other human studies. These human-serum REPs show much smaller variation compared to the 4 to 5 orders of magnitude span in REPs database for the present WHO-TEF determination. Moreover, the established REPs fitted well with WHO-TEFs, especially for polychlorinated dibenzo- p-dioxins, furans. These REPs reflecting real human exposure scenarios exhibited validity and could be used to improve health risk assessment of human body burden of DLCs.
Collapse
Affiliation(s)
- Xin Liu
- NHC Key Laboratory of Food Safety Risk Assessment , China National Center for Food Safety Risk Assessment , Beijing 100021 , China
- State Key Laboratory of Food Science and Technology , Nanchang University , Nanchang 330047 , China
| | - Lei Zhang
- NHC Key Laboratory of Food Safety Risk Assessment , China National Center for Food Safety Risk Assessment , Beijing 100021 , China
| | - Jingguang Li
- NHC Key Laboratory of Food Safety Risk Assessment , China National Center for Food Safety Risk Assessment , Beijing 100021 , China
- State Key Laboratory of Food Science and Technology , Nanchang University , Nanchang 330047 , China
| | - Jun Wang
- Shenzhen Center for Chronic Disease Control , Shenzhen 518020 , China
| | - Guimin Meng
- Beijing Fengtai Hospital obstetrics and gynecology , Beijing 100071 , China
| | - Min Chi
- NHC Key Laboratory of Food Safety Risk Assessment , China National Center for Food Safety Risk Assessment , Beijing 100021 , China
- Taiyuan Center for Disease Control and Prevention , Taiyuan 030000 , China
| | - Yunfeng Zhao
- NHC Key Laboratory of Food Safety Risk Assessment , China National Center for Food Safety Risk Assessment , Beijing 100021 , China
| | - Yongning Wu
- NHC Key Laboratory of Food Safety Risk Assessment , China National Center for Food Safety Risk Assessment , Beijing 100021 , China
- State Key Laboratory of Food Science and Technology , Nanchang University , Nanchang 330047 , China
| |
Collapse
|
10
|
Brown MR, Garside H, Thompson E, Atwal S, Bean C, Goodall T, Sullivan M, Graham MJ. From the Cover: Development and Application of a Dual Rat and Human AHR Activation Assay. Toxicol Sci 2018; 160:408-419. [PMID: 29029351 DOI: 10.1093/toxsci/kfx188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Significant prolonged aryl hydrocarbon receptor (AHR) activation, classically exhibited following exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin, can cause a variety of undesirable toxicological effects. Novel pharmaceutical chemistries also have the potential to cause activation of AHR and consequent toxicities in pre-clinical species and man. Previous methods either employed relatively expensive and low-throughput primary hepatocyte dosing with PCR endpoint, or low resolution overexpressing reporter gene assays. We have developed, validated and applied an in vitro microtitre plate imaging-based medium throughput screening assay for the assessment of endogenous species-specific AHR activation potential via detection of induction of the surrogate transcriptional target Cytochrome P450 CYP1A1. Routine testing of pharmaceutical drug development candidate chemistries using this assay can influence the chemical design process and highlight AHR liabilities. This assay should be introduced such that human AHR activation liability is flagged early for confirmatory testing.
Collapse
Affiliation(s)
- Martin R Brown
- Discovery Sciences, AstraZeneca R&D Darwin, Cambridge CB4 0WG, UK
| | - Helen Garside
- Discovery Safety & Metabolism, AstraZeneca R&D, Macclesfield, Cheshire SK10 4TG, UK
| | - Emma Thompson
- Discovery Safety & Metabolism, AstraZeneca R&D, Macclesfield, Cheshire SK10 4TG, UK
| | - Saseela Atwal
- Discovery Safety & Metabolism, AstraZeneca R&D, Macclesfield, Cheshire SK10 4TG, UK
| | - Chloe Bean
- AstraZeneca R&D Charnwood, Loughborough, Leics LE11 5RH, UK
| | - Tony Goodall
- AstraZeneca R&D Charnwood, Loughborough, Leics LE11 5RH, UK
| | | | - Mark J Graham
- AstraZeneca R&D Charnwood, Loughborough, Leics LE11 5RH, UK
| |
Collapse
|
11
|
Roth AD, Lama P, Dunn S, Hong S, Lee MY. Polymer coating on a micropillar chip for robust attachment of PuraMatrix peptide hydrogel for 3D hepatic cell culture. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 90:634-644. [PMID: 29853133 DOI: 10.1016/j.msec.2018.04.092] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 12/11/2017] [Accepted: 04/27/2018] [Indexed: 12/12/2022]
Abstract
For better mimicking tissues in vivo and developing predictive cell models for high-throughput screening (HTS) of potential drug candidates, three-dimensional (3D) cell cultures have been performed in various hydrogels. In this study, we have investigated several polymer coating materials to robustly attach PuraMatrix peptide hydrogel on a micropillar chip for 3D culture of Hep3B human hepatic cells, which can be used as a tool for high-throughput assessment of compound hepatotoxicity. Among several amphiphilic polymers with maleic anhydride groups tested, 0.01% (w/v) poly(maleic anhydride-alt-1-octadecene) (PMA-OD) provided superior coating properties with no PuraMatrix spot detachment from the micropillar chip and no air bubble entrapment in a complementary microwell chip. To maintain Hep3B cell viability in PuraMatrix gel on the chip, gelation conditions were optimized in the presence of additional salts, at different seeding densities, and for growth medium washes. As a result, salts in growth media were sufficient for gelation, and relatively high cell seeding at 6 million cells/mL and two media washes for pH neutralization were required. With optimized 3D cell culture conditions, controlled gene expression and compound toxicity assessment were successfully demonstrated by using recombinant adenoviruses carrying genes for green and red fluorescent proteins as well as six model compounds. Overall, PuraMatrix hydrogel on the chip was suitable for 3D cell encapsulation, gene expression, and rapid toxicity assessment.
Collapse
Affiliation(s)
- Alexander David Roth
- Department of Chemical and Biomedical Engineering, Cleveland State University, Fenn Hall Room 455, 1960 East 24th Street, Cleveland, OH 44115, United States
| | - Pratap Lama
- Department of Chemical and Biomedical Engineering, Cleveland State University, Fenn Hall Room 455, 1960 East 24th Street, Cleveland, OH 44115, United States
| | - Stephen Dunn
- Department of Chemical and Biomedical Engineering, Cleveland State University, Fenn Hall Room 455, 1960 East 24th Street, Cleveland, OH 44115, United States
| | - Stephen Hong
- Department of Chemical and Biomedical Engineering, Cleveland State University, Fenn Hall Room 455, 1960 East 24th Street, Cleveland, OH 44115, United States
| | - Moo-Yeal Lee
- Department of Chemical and Biomedical Engineering, Cleveland State University, Fenn Hall Room 455, 1960 East 24th Street, Cleveland, OH 44115, United States.
| |
Collapse
|
12
|
Fernández I, Gavaia PJ, Laizé V, Cancela ML. Fish as a model to assess chemical toxicity in bone. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2018; 194:208-226. [PMID: 29202272 DOI: 10.1016/j.aquatox.2017.11.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/14/2017] [Accepted: 11/22/2017] [Indexed: 06/07/2023]
Abstract
Environmental toxicology has been expanding as growing concerns on the impact of produced and released chemical compounds over the environment and human health are being demonstrated. Among the toxic effects observed in organisms exposed to pollutants, those affecting skeletal tissues (osteotoxicity) have been somehow overlooked in comparison to hepato-, immune-, neuro- and/or reproductive toxicities. Nevertheless, sub-lethal effects of toxicants on skeletal development and/or bone maintenance may result in impaired growth, reduced survival rate, increased disease susceptibility and diminished welfare. Osteotoxicity may occur by acute or chronic exposure to different environmental insults. Because of biologically and technically advantagous features - easy to breed and inexpensive to maintain, external and rapid rate of development, translucent larvae and the availability of molecular and genetic tools - the zebrafish (Danio rerio) has emerged in the last decade as a vertebrate model system of choice to evaluate osteotoxicity. Different experimental approaches in fish species and analytical tools have been applied, from in vitro to in vivo systems, from specific to high throughput methodologies. Current knowledge on osteotoxicity and underlying mechanisms gained using fish, with a special emphasis on zebrafish systems, is reviewed here. Osteotoxicants have been classified into four categories according to the pathway involved in the transduction of the osteotoxic effects: activation/inhibition of membrane and/or nuclear receptors, alteration of redox condition, mimicking of bone constituents and unknown pathways. Knowledge on these pathways is also reported here as it may provide critical insights into the development, production and release of future chemical compounds with none or low osteotoxicity, thus promoting the green/environmental friendly chemistry.
Collapse
Affiliation(s)
- Ignacio Fernández
- Centre of Marine Sciences (CCMAR), University of Algarve, Campus de Gambelas, Faro, Portugal.
| | - Paulo J Gavaia
- Centre of Marine Sciences (CCMAR), University of Algarve, Campus de Gambelas, Faro, Portugal; Department of Biomedical Sciences and Medicine, University of Algarve, Campus de Gambelas, Faro, Portugal
| | - Vincent Laizé
- Centre of Marine Sciences (CCMAR), University of Algarve, Campus de Gambelas, Faro, Portugal
| | - M Leonor Cancela
- Centre of Marine Sciences (CCMAR), University of Algarve, Campus de Gambelas, Faro, Portugal; Department of Biomedical Sciences and Medicine, University of Algarve, Campus de Gambelas, Faro, Portugal; Algarve Biomedical Center (ABC), Universidade do Algarve, Campus de Gambelas, Faro, Portugal
| |
Collapse
|
13
|
One TEF concept does not fit all: The case for human risk assessment of polychlorinated biphenyls. CURRENT OPINION IN TOXICOLOGY 2017. [DOI: 10.1016/j.cotox.2017.01.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
14
|
van Ede KI, van Duursen MBM, van den Berg M. Evaluation of relative effect potencies (REPs) for dioxin-like compounds to derive systemic or human-specific TEFs to improve human risk assessment. Arch Toxicol 2016; 90:1293-305. [PMID: 27161441 PMCID: PMC4873528 DOI: 10.1007/s00204-016-1724-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 04/21/2016] [Indexed: 01/20/2023]
Abstract
Toxic equivalency factors (TEFs) are generally applied for estimating human risk of dioxins and dioxin-like compounds using systemic (e.g., blood) levels, even though these TEFs are established based on intake doses in rodent studies. This review shows that systemic relative effect potencies (REPs) can deviate substantially from intake REPs, but are similar to in vitro-derived REPs. Interestingly, the in vitro REPs for 1,2,3,4,6,7,8-heptachlorodibenzo-p-dioxin (HpCDD) and 2,3,4,7,8-pentachlorodibenzofuran (4-PeCDF) are up to one order of magnitude higher than their in vivo REPs and WHO-TEFs, based on oral intake. In addition, clear species-differences in in vitro REPs were apparent for some congeners. Especially the human-derived REP for polychlorinated biphenyl 126 is one to two orders of magnitude lower than rodent REPs and its current WHO-TEF. Next, suggested adapted systemic or human-specific TEFs for these congeners were applied to calculate changes in systemic TEQ concentrations in studies from the USA, Germany and Japan and compared with either the JECFA TDI or USEPA RfD of TCDD. Overall, the effect of such TEF changes for these three congeners on total TEQ roughly balances each other out in the general population. However, results may be different for situations in which a specific group of congeners dominates. For those congeners that show a distinct deviation between either intake and systemic REPs or between rodent- and human-based in vitro REPs, we propose that especially REPs derived from human-based in vitro models are weighted more heavily in establishing systemic or human-specific TEF values to improve human health risk assessment.
Collapse
Affiliation(s)
- Karin I van Ede
- Division of Toxicology and Veterinary Pharmacology, Institute for Risk Assessment Sciences (IRAS), Utrecht University, P.O. Box 80.177, NL-3508 TD, Utrecht, The Netherlands.
| | - Majorie B M van Duursen
- Division of Toxicology and Veterinary Pharmacology, Institute for Risk Assessment Sciences (IRAS), Utrecht University, P.O. Box 80.177, NL-3508 TD, Utrecht, The Netherlands
| | - Martin van den Berg
- Division of Toxicology and Veterinary Pharmacology, Institute for Risk Assessment Sciences (IRAS), Utrecht University, P.O. Box 80.177, NL-3508 TD, Utrecht, The Netherlands
| |
Collapse
|
15
|
Determining the IC 50 Values for Vorozole and Letrozole, on a Series of Human Liver Cytochrome P450s, to Help Determine the Binding Site of Vorozole in the Liver. Enzyme Res 2015; 2015:321820. [PMID: 26635974 PMCID: PMC4655258 DOI: 10.1155/2015/321820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 10/26/2015] [Indexed: 11/18/2022] Open
Abstract
Vorozole and letrozole are third-generation aromatase (cytochrome P450 19A1) inhibitors. [11C]-Vorozole can be used as a radiotracer for aromatase in living animals but when administered by IV, it collects in the liver. Pretreatment with letrozole does not affect the binding of vorozole in the liver. In search of finding the protein responsible for the accumulation of vorozole in the liver, fluorometric high-throughput screening assays were used to test the inhibitory capability of vorozole and letrozole on a series of liver cytochrome P450s (CYP1A1, CYP1A2, CYP2A6, and CYP3A4). It was determined that vorozole is a potent inhibitor of CYP1A1 (IC50 = 0.469 μM) and a moderate inhibitor of CYP2A6 and CYP3A4 (IC50 = 24.4 and 98.1 μM, resp.). Letrozole is only a moderate inhibitor of CYP1A1 and CYP2A6 (IC50 = 69.8 and 106 μM) and a very weak inhibitor of CYP3A4 (<10% inhibition at 1 mM). Since CYP3A4 makes up the majority of the CYP content found in the human liver, and vorozole inhibits it moderately well but letrozole does not, CYP3A4 is a good candidate for the protein that [11C]-vorozole is binding to in the liver.
Collapse
|
16
|
Becker RA, Patlewicz G, Simon TW, Rowlands JC, Budinsky RA. The adverse outcome pathway for rodent liver tumor promotion by sustained activation of the aryl hydrocarbon receptor. Regul Toxicol Pharmacol 2015; 73:172-90. [PMID: 26145830 DOI: 10.1016/j.yrtph.2015.06.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 06/19/2015] [Accepted: 06/22/2015] [Indexed: 12/29/2022]
Abstract
An Adverse Outcome Pathway (AOP) represents the existing knowledge of a biological pathway leading from initial molecular interactions of a toxicant and progressing through a series of key events (KEs), culminating with an apical adverse outcome (AO) that has to be of regulatory relevance. An AOP based on the mode of action (MOA) of rodent liver tumor promotion by dioxin-like compounds (DLCs) has been developed and the weight of evidence (WoE) of key event relationships (KERs) evaluated using evolved Bradford Hill considerations. Dioxins and DLCs are potent aryl hydrocarbon receptor (AHR) ligands that cause a range of species-specific adverse outcomes. The occurrence of KEs is necessary for inducing downstream biological responses and KEs may occur at the molecular, cellular, tissue and organ levels. The common convention is that an AOP begins with the toxicant interaction with a biological response element; for this AOP, this initial event is binding of a DLC ligand to the AHR. Data from mechanistic studies, lifetime bioassays and approximately thirty initiation-promotion studies have established dioxin and DLCs as rat liver tumor promoters. Such studies clearly show that sustained AHR activation, weeks or months in duration, is necessary to induce rodent liver tumor promotion--hence, sustained AHR activation is deemed the molecular initiating event (MIE). After this MIE, subsequent KEs are 1) changes in cellular growth homeostasis likely associated with expression changes in a number of genes and observed as development of hepatic foci and decreases in apoptosis within foci; 2) extensive liver toxicity observed as the constellation of effects called toxic hepatopathy; 3) cellular proliferation and hyperplasia in several hepatic cell types. This progression of KEs culminates in the AO, the development of hepatocellular adenomas and carcinomas and cholangiolar carcinomas. A rich data set provides both qualitative and quantitative knowledge of the progression of this AOP through KEs and the KERs. Thus, the WoE for this AOP is judged to be strong. Species-specific effects of dioxins and DLCs are well known--humans are less responsive than rodents and rodent species differ in sensitivity between strains. Consequently, application of this AOP to evaluate potential human health risks must take these differences into account.
Collapse
Affiliation(s)
- Richard A Becker
- Regulatory and Technical Affairs Department, American Chemistry Council (ACC), Washington, DC 20002, USA.
| | - Grace Patlewicz
- DuPont Haskell Global Centers for Health and Environmental Sciences, Newark, DE 19711, USA
| | - Ted W Simon
- Ted Simon LLC, 4184 Johnston Road, Winston, GA 30187, USA
| | - J Craig Rowlands
- The Dow Chemical Company, Toxicology & Environmental Research & Consulting, 1803 Building Washington Street, Midland, MI 48674, USA
| | - Robert A Budinsky
- The Dow Chemical Company, Toxicology & Environmental Research & Consulting, 1803 Building Washington Street, Midland, MI 48674, USA
| |
Collapse
|
17
|
Shi F, Zhao P, Li X, Pan H, Ma S, Ding L. Cytotoxicity of luteolin in primary rat hepatocytes: the role of CYP3A-mediatedortho-benzoquinone metabolite formation and glutathione depletion. J Appl Toxicol 2015; 35:1372-80. [DOI: 10.1002/jat.3106] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 11/20/2014] [Accepted: 11/21/2014] [Indexed: 11/10/2022]
Affiliation(s)
- Fuguo Shi
- Department of Pharmaceutical Analysis, Key Laboratory of Drug Quality Control and Pharmacovigilance affiliated to Ministry of Education; China Pharmaceutical University; 24 Tongjiaxiang Nanjing 210009 China
| | - Peng Zhao
- Department of Pharmacology of Chinese Materia Medica; China Pharmaceutical University; 24 Tongjiaxiang Nanjing 210009 China
| | - Xiaobing Li
- Department of Pharmaceutical Analysis, Key Laboratory of Drug Quality Control and Pharmacovigilance affiliated to Ministry of Education; China Pharmaceutical University; 24 Tongjiaxiang Nanjing 210009 China
| | - Hong Pan
- Department of Pharmaceutical Analysis, Key Laboratory of Drug Quality Control and Pharmacovigilance affiliated to Ministry of Education; China Pharmaceutical University; 24 Tongjiaxiang Nanjing 210009 China
| | - Shiping Ma
- Department of Pharmacology of Chinese Materia Medica; China Pharmaceutical University; 24 Tongjiaxiang Nanjing 210009 China
| | - Li Ding
- Department of Pharmaceutical Analysis, Key Laboratory of Drug Quality Control and Pharmacovigilance affiliated to Ministry of Education; China Pharmaceutical University; 24 Tongjiaxiang Nanjing 210009 China
| |
Collapse
|
18
|
van Ede KI, Gaisch KPJ, van den Berg M, van Duursen MBM. Differential relative effect potencies of some dioxin-like compounds in human peripheral blood lymphocytes and murine splenic cells. Toxicol Lett 2014; 226:43-52. [PMID: 24472611 DOI: 10.1016/j.toxlet.2014.01.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 01/16/2014] [Accepted: 01/17/2014] [Indexed: 11/29/2022]
Abstract
Human risk assessment for dioxin-like compounds is typically based on the concentration measured in blood serum multiplied by their assigned toxic equivalency factor (TEF). Consequently, the actual value of the TEF is very important for accurate human risk assessment. In this study we investigated the effect potencies of three polychlorinated dibenzo-p-dioxins (PCDDs), six polychlorinated dibenzofurans (PCDFs) and 10 polychlorinated biphenyls (PCBs) relative to the reference congener 2,3,7,8-tetrachloro-dibenzo-p-dioxin (TCDD) in in vitro exposed primary human peripheral blood lymphocytes (PBLs) and mouse splenic cells. REPs were determined based on cytochrome P450 (CYP) 1A1, 1B1 and aryl hydrocarbon receptor repressor (AhRR) gene expression as well as CYP1A1 activity in human PBLs and Cyp1a1 gene expression in murine splenic cells. Estimated median human REPs for 1,2,3,4,6,7,8-heptachlorodibenzo-p-dioxin (1234678-HpCDD), 2,3,4,7,8,-pentachlorodibenzofuran (23478-PeCDF), 1,2,3,4,7,8-hexachlorodibenzofuran (123478-HxCDF) and 1,2,3,4,7,8,9-heptachlorodibenzofuran (1234789-HpCDF) were with 0.1, 1.1, 1 and 0.09, respectively, significantly higher compared to those estimated for mouse with REPs of 0.05, 0.45, 0.09 and 0.04, respectively. Opposite to these results, the estimated median human REP of 3,3',4,4',5-pentachlorobiphenyl (PCB 126), was with 0.001 30-fold lower compared to the mouse REP of 0.03. Furthermore, human REPs for 1234678-HpCDD, 23478-PeCDF, 123478-HxCDF, 1234789-HpCDF and PCB 126 were all outside the ± half log uncertainty range that is taken into account in the WHO-assigned TEFs. Together, these data show congener- and species-specific differences in REPs for some, but not all dioxin-like congeners tested. This suggests that, more emphasis should be placed on human-tissue derived REPs in the establishment of a TEF for human risk assessment.
Collapse
Affiliation(s)
- Karin I van Ede
- Institute for Risk Assessment Sciences, Utrecht University, Yalelaan 104, 3584 CM Utrecht, The Netherlands.
| | - Konrad P J Gaisch
- Institute for Risk Assessment Sciences, Utrecht University, Yalelaan 104, 3584 CM Utrecht, The Netherlands.
| | - Martin van den Berg
- Institute for Risk Assessment Sciences, Utrecht University, Yalelaan 104, 3584 CM Utrecht, The Netherlands.
| | - Majorie B M van Duursen
- Institute for Risk Assessment Sciences, Utrecht University, Yalelaan 104, 3584 CM Utrecht, The Netherlands.
| |
Collapse
|
19
|
Budinsky RA, Schrenk D, Simon T, Van den Berg M, Reichard JF, Silkworth JB, Aylward LL, Brix A, Gasiewicz T, Kaminski N, Perdew G, Starr TB, Walker NJ, Rowlands JC. Mode of action and dose–response framework analysis for receptor-mediated toxicity: The aryl hydrocarbon receptor as a case study. Crit Rev Toxicol 2013; 44:83-119. [DOI: 10.3109/10408444.2013.835787] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
20
|
Liu Y, Flynn TJ, Ferguson MS, Hoagland EM. Use of the Combination Index to determine interactions between plant-derived phenolic acids on hepatotoxicity endpoints in human and rat hepatoma cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2013; 20:461-8. [PMID: 23380082 DOI: 10.1016/j.phymed.2012.12.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 11/27/2012] [Accepted: 12/25/2012] [Indexed: 06/01/2023]
Abstract
The beneficial or adverse effects of isolated phytochemicals are not always concordant with effects of the botanical dietary supplements from which they were derived. This disparity could be due to interactions between the various phytochemicals present in the whole plant. The phenolic acids, rosmarinic acid (RA), caffeic acid (CA) and ferulic acid (FA) are widely present in foods and dietary supplements, and they are assumed to exert various beneficial biological effects. However, there is little data on the potential biological interactions of these three phenolic acids which commonly occur together and are linked metabolically. In the present study, liver toxicity of the three phenolic acids was assessed on the three compounds singly and in various binary and one ternary combinations. A series of in vitro endpoints relevant to liver toxicity were evaluated in both a human (HepG2/C3A) and rat (MH1C1) hepatocyte cell line. The Combination Index (CI) was calculated for each endpoint from both the concentration responses of the single compounds and the responses of the various binary and ternary mixtures. Both synergistic and antagonistic interactions were observed for some endpoints and some combinations of test agents. Interactions were most prevalent in measures of oxidative stress and cytochrome P450 activities in both cell types. There was only a 53% concordance between the rat and human cells which may be suggestive of species differences. The data suggest an approach for better characterizing the beneficial or adverse effects of complex botanical products through evaluation of interactions between individual phytochemical components.
Collapse
Affiliation(s)
- Yitong Liu
- Division of Toxicology, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD 20708, USA
| | | | | | | |
Collapse
|
21
|
Knudsen TB, Houck KA, Sipes NS, Singh AV, Judson RS, Martin MT, Weissman A, Kleinstreuer NC, Mortensen HM, Reif DM, Rabinowitz JR, Setzer RW, Richard AM, Dix DJ, Kavlock RJ. Activity profiles of 309 ToxCast™ chemicals evaluated across 292 biochemical targets. Toxicology 2011; 282:1-15. [PMID: 21251949 DOI: 10.1016/j.tox.2010.12.010] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Revised: 12/10/2010] [Accepted: 12/21/2010] [Indexed: 12/01/2022]
Abstract
Understanding the potential health risks posed by environmental chemicals is a significant challenge elevated by the large number of diverse chemicals with generally uncharacterized exposures, mechanisms, and toxicities. The present study is a performance evaluation and critical analysis of assay results for an array of 292 high-throughput cell-free assays aimed at preliminary toxicity evaluation of 320 environmental chemicals in EPA's ToxCast™ project (Phase I). The chemicals (309 unique, 11 replicates) were mainly precursors or the active agent of commercial pesticides, for which a wealth of in vivo toxicity data is available. Biochemical HTS (high-throughput screening) profiled cell and tissue extracts using semi-automated biochemical and pharmacological methodologies to evaluate a subset of G-protein coupled receptors (GPCRs), CYP450 enzymes (CYPs), kinases, phosphatases, proteases, HDACs, nuclear receptors, ion channels, and transporters. The primary screen tested all chemicals at a relatively high concentration 25 μM concentration (or 10 μM for CYP assays), and a secondary screen re-tested 9132 chemical-assay pairs in 8-point concentration series from 0.023 to 50 μM (or 0.009-20 μM for CYPs). Mapping relationships across 93,440 chemical-assay pairs based on half-maximal activity concentration (AC50) revealed both known and novel targets in signaling and metabolic pathways. The primary dataset, summary data and details on quality control checks are available for download at http://www.epa.gov/ncct/toxcast/.
Collapse
Affiliation(s)
- Thomas B Knudsen
- National Center for Computational Toxicology (B205-01), Office of Research & Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Fujimura H, Murakami N, Miwa S, Aruga C, Toriumi W. The suitability of rat hepatoma cell line H4IIE for evaluating the potentials of compounds to induce CYP3A23 expression. ACTA ACUST UNITED AC 2010; 64:527-33. [PMID: 21146967 DOI: 10.1016/j.etp.2010.11.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2009] [Accepted: 11/12/2010] [Indexed: 01/22/2023]
Abstract
To investigate the suitability of H4IIE cells for detecting cytochrome P450 (CYP) induction in vitro, we compared CYP induction by typical CYP inducers in H4IIE cells and rat primary hepatocytes by examining gene expression and enzyme activity, and by immunocytochemistry. The cells were preincubated with 0.1 μM of dexamethasone (DEX) for 24 h, followed by 48 h of exposure to 10 μM of beta-naphthoflavone (bNF), 100 μM of phenobarbital (PB) and 10 μM of DEX. Cyp1a1, Cyp2b1/2 and Cyp3a23/3a1 (Cyp3a23) expressions in H4IIE cells were up-regulated 280-, 1.5- and 65-fold relative to those in vehicle-treated cells, respectively. The fold inductions of those expressions in rat primary hepatocytes were 80-, 33- and 152-fold, respectively. Comprehensive gene expression analysis using DNA microarrays showed that Cyp3a23, Gsta2, Ugt2b12, Udpgt and Sult2a1 expressions were up-regulated in H4IIE cells exposed to 10 μM of DEX. CYP3A activity was not increased, but some H4IIE cells exposed to DEX were stained strongly with anti-CYP3A antibody. We cloned these cells and obtained cloned H4IIE (cH4IIE) cells with expression level of Cyp3a23 higher than those of vehicle-treated cells. It was confirmed that preincubation with 0.1 μM of DEX increased pregnane X receptor (Pxr) expression level and enhanced the Cyp3a23 induction effects of test compounds significantly. Retrospective examination of in vitro CYP induction assay using cH4IIE cells resulted in 80% correlation with the data from in vivo rat toxicity studies. These results suggested that cH4IIE cells are suitable for evaluating the potentials of a compound to induce CYP3A23 expression.
Collapse
Affiliation(s)
- Hisako Fujimura
- Safety Research Laboratory, Mitsubishi Tanabe Pharma Co., 2-50, Kawagishi, 2-Chome, Toda, Saitama 335-8505, Japan.
| | | | | | | | | |
Collapse
|
23
|
Bolt HM, Hengstler JG. Most cited articles: metal toxicity, oxidative stress control and induction as well as inhibition of cytochrome P450 enzymes. Arch Toxicol 2010. [DOI: 10.1007/s00204-010-0624-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
24
|
Audebert M, Riu A, Jacques C, Hillenweck A, Jamin EL, Zalko D, Cravedi JP. Use of the γH2AX assay for assessing the genotoxicity of polycyclic aromatic hydrocarbons in human cell lines. Toxicol Lett 2010; 199:182-92. [PMID: 20832459 DOI: 10.1016/j.toxlet.2010.08.022] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Revised: 08/27/2010] [Accepted: 08/31/2010] [Indexed: 01/18/2023]
Abstract
The development of in vitro genotoxic assays as an alternative method to animal experimentation is of growing interest in the context of the implementation of new regulations on chemicals. However, extrapolation of toxicity data from in vitro systems to in vivo models is hampered by the fact that in vitro systems vary in their capability to metabolize chemicals, and that biotransformation can greatly influence the experimental results. Therefore, much attention has to be paid to the cellular models used and experimental conditions. Polycyclic aromatic hydrocarbons (PAHs) are carcinogenic ubiquitous pollutants. Human exposure to PAHs is mainly from food origin. In this study, a detailed analysis of the biotransformation capabilities of three human cell lines commonly used for in vitro testing (HepG2, ACHN and Caco-2) was undertaken using 3 model PAHs (benzo(a)pyrene [B(a)P], fluoranthene [FLA] and 3-methylcholanthrene [3-MC]). Concomitantly the genotoxicity of these PAHs was investigated in different cell lines, using a new genotoxic assay (H2AX) in 96-well plates. The metabolic rates of B(a)P, FLA and 3-MC were similar in HepG2 and Caco-2 cell lines, respectively, though with the production of different metabolites. The ACHN cell line was shown to express very limited metabolic capabilities. We demonstrated that the PAHs having a high metabolic rate (B(a)P and 3-MC) were genotoxic from 10(-7) molar in both HepG2 and Caco-2 cells. The present study shows that H2AX measurement in human cell lines competent for the metabolism, is an efficient and sensitive genotoxic assay requiring less cells and time than other currently available tests.
Collapse
Affiliation(s)
- M Audebert
- INRA, UMR 1089 Xénobiotiques INRA-ENVT, Toulouse, France.
| | | | | | | | | | | | | |
Collapse
|
25
|
Sutter CH, Bodreddigari S, Sutter TR, Carlson EA, Silkworth JB. Analysis of the CYP1A1 mRNA dose-response in human keratinocytes indicates that relative potencies of dioxins, furans, and PCBs are species and congener specific. Toxicol Sci 2010; 118:704-15. [PMID: 20819910 DOI: 10.1093/toxsci/kfq262] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Reports indicate that toxic equivalency factors (TEFs) based primarily on rodent data do not accurately predict in vitro human responsiveness to certain dioxin-like chemicals (DLCs). To investigate this in cells responsive to dioxins and relevant to chloracne, normal human epidermal keratinocytes were treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and several DLCs, each with a TEF value of 0.1, representing three classes of congeners. We estimated half maximal effective concentration (EC50)-based donor-specific relative potency (REP) values for cytochrome P450 1A1 (CYP1A1) messenger RNA (mRNA) induction for TCDD, 1,2,3,6,7,8-hexachlorodibenzo-p-dioxin (HxCDD), 2,3,7,8-tetrachlorodibenzofuran (TCDF), 1,2,3,6,7,8-hexachlorodibenzofuran (HxCDF), and 3,3',4,4',5-pentachlorobiphenyl (PCB 126). We also determined EC50-based population-level REP values (n = 4) for CYP1A1 mRNA induction for TCDD, HxCDF, and PCB 126. Furthermore, an alternative factor, the relative threshold factor (RTF) based on the low end (threshold) of the dose-response curve, was calculated. Our results demonstrated that HxCDF had a population-based REP value of 0.98, 9.8-fold higher than its assigned TEF value of 0.1. Conversely, PCB 126 had an REP value of 0.0027 and an RTF of 0.0022, 37-fold and 45-fold less than its assigned TEF of 0.1, respectively. The REP values for HxCDD and TCDF were 0.24 and 0.10, respectively, similar to their assigned value of 0.1. Therefore, although the DLCs tested in the current study all possessed the same assigned TEF value of 0.1, congener-specific differences in REPs and RTFs were observed for human keratinocytes. These congener-specific discrepancies are likely because of differences in interspecies factors that have yet to be defined.
Collapse
Affiliation(s)
- Carrie H Sutter
- Department of Biological Sciences and W. Harry Feinstone Center for Genomic Research, University of Memphis, Memphis, Tennessee 38152-3560, USA.
| | | | | | | | | |
Collapse
|
26
|
Sato W, Suzuki H, Sasaki T, Kumagai T, Sakaguchi S, Mizugaki M, Miyairi S, Yamazoe Y, Nagata K. Construction of a system that simultaneously evaluates CYP1A1 and CYP1A2 induction in a stable human-derived cell line using a dual reporter plasmid. Drug Metab Pharmacokinet 2010; 25:180-9. [PMID: 20460824 DOI: 10.2133/dmpk.25.180] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Human CYP1A1 and CYP1A2 genes are in a head-to-head orientation on chromosome 15 and are separated by a 23-kb intergenic space. To our knowledge, this is the first report on a stable cell line that contains the 23-kb full-length regulatory region and is able to simultaneously assess the transcriptional activation of CYP1A1 and CYP1A2 genes. The stable cell line that constitutively expresses the reporter activities was constructed by inserting the dual reporter plasmid containing the 23-kb region between the CYP1A1 and CYP1A2 genes into the chromosome. Transcriptional activation of the CYP1A1 and CYP1A2 genes was measured simultaneously using luciferase (Luc) and secreted alkaline phosphatase (SEAP) activities, respectively. To demonstrate the utility of the stable cell line, CYP1A1/1A2 induction by the majority of compounds previously identified as CYP1A1/1A2 inducers was measured. The results clearly show that all compounds caused induction of reporter activities. In addition to assessing transcriptional activation of the CYP1A1 and CYP1A2 genes by measuring reporter activities, we determined the intrinsic CYP1A1 and CYP1A2 mRNA levels by treating them with the same compounds. The results suggest that this stable cell line may be used to rapidly and accurately predict CYP1A1/1A2 induction.
Collapse
Affiliation(s)
- Wataru Sato
- Department of Environmental and Health Science, Tohoku Pharmaceutical University, Sendai, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Smith EM, Wilson JY. Assessment of cytochrome P450 fluorometric substrates with rainbow trout and killifish exposed to dexamethasone, pregnenolone-16alpha-carbonitrile, rifampicin, and beta-naphthoflavone. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2010; 97:324-333. [PMID: 20167382 DOI: 10.1016/j.aquatox.2010.01.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 12/23/2009] [Accepted: 01/05/2010] [Indexed: 05/28/2023]
Abstract
Cytochrome P450s (CYPs) are important xenobiotic metabolizing proteins. While their functions are well understood in mammals, CYP function in non-mammalian vertebrate systems is much less defined, with function often inferred from mammalian data, assuming similar function across vertebrate species. In this study, we investigate whether in vivo treatment with known mammalian CYP inducers can alter the in vitro catalytic activity of fish microsomes using eleven fluorescent CYP-mediated substrates. We investigate the basal metabolism and induction potential for hepatic CYPs in two fish species, rainbow trout (Oncorhynchus mykiss) and killifish (Fundulus heteroclitus). Species differences were found in the baseline metabolism of these substrates. Killifish have significantly higher metabolic rates for all tested substrates except 7-benzyloxyquinoline and 7-benzyloxy-4-trifluoromethylcoumarin (both mammalian CYP3A substrates); significant differences were also seen between male and female killifish. Treatment with dexamethasone, pregnenolone-16alpha-carbonitrile, and rifampicin did not cause broad, measurable CYP induction in either fish species. In trout, dexamethasone (100 mg kg(-1)) significantly induced 3-cyano-7-ethoxycoumarin metabolism and rifampicin (100 mg kg(-1)) induced the dealkylation of 7-methoxyresorufin, although both were highly variable. Female killifish exposed to pregnenolone-16alpha-carbonitrile (100 mg kg(-1)) showed significantly higher metabolism of 7-pentoxyresorufin. Overall, dexamethasone, pregnenolone-16alpha-carbonitrile and rifampicin did not appear to consistently increase CYP activity in fish. Trout treated with 10 or 50 mg kg(-1) beta-naphthoflavone (BNF), a CYP1A inducer, showed significantly induced activity across almost all substrates tested, exceptions being 7-benzyloxyquinoline, 7-benzyloxy-4-trifluoromethylcoumarin and dibenzylfluorescein. 7-Methoxy-4-(aminomethyl)coumarin, a typical CYP2D substrate in mammals, was not metabolized by untreated fish liver microsomes; however, treatment with BNF significantly induced the metabolism of this substrate in trout. Induced substrate metabolism in BNF-treated microsomes was only correlated across selective substrates, suggesting that BNF induces multiple CYPs in fish liver. These include the known BNF inducible CYP1s plus a number of as yet unidentified fish CYPs. Overall, many of these catalytic assays could be valuable tools for identification of the function of specific CYP subfamilies and individual isoforms in fish.
Collapse
Affiliation(s)
- Emily M Smith
- Department of Biology, McMaster University, Hamilton, Ontario L8S4K1, Canada
| | | |
Collapse
|
28
|
Khalaf H, Larsson A, Berg H, McCrindle R, Arsenault G, Olsson PE. Diastereomers of the brominated flame retardant 1,2-dibromo-4-(1,2 dibromoethyl)cyclohexane induce androgen receptor activation in the hepg2 hepatocellular carcinoma cell line and the lncap prostate cancer cell line. ENVIRONMENTAL HEALTH PERSPECTIVES 2009; 117:1853-9. [PMID: 20049203 PMCID: PMC2799458 DOI: 10.1289/ehp.0901065] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2009] [Accepted: 08/03/2009] [Indexed: 05/02/2023]
Abstract
BACKGROUND Reported incidences of prostate cancer and masculinization of animals indicate a release of compounds with androgenic properties into the environment. Large numbers of environmental pollutants have been screened to identify such compounds; however, not until recently was 1,2-dibromo-4-(1,2-dibromoethyl)cyclohexane (TBECH) identified as the first potent activator of the human androgen receptor (hAR). TBECH has been found in beluga whales and bird eggs and has also been found to be maternally transferred in zebrafish. OBJECTIVES In the present study we investigated interaction energies between TBECH diastereomers (alpha, beta, gamma, and delta) and the hAR, and their ability to activate the receptor and induce prostate-specific antigen (PSA) expression in vitro. METHODS We performed computational modeling to determine interaction energies between the ligand and the AR ligand-binding site, and measured in vitro competitive binding assays for AR by polarization fluorometry analysis. We used enzyme-linked immunosorbent assays to determine PSA activity in LNCaP and HepG2 cells. RESULTS We found the gamma and delta diastereomers to be more potent activators of hAR than the alpha and beta diastereomers, which was confirmed in receptor binding studies. All TBECH diastereomers induced PSA expression in LNCaP cells even though the AR present in these cells is mutated (T877A). Modeling studies of LNCaP AR revealed that TBECH diastereomers bound to the receptor with a closer distance to the key amino acids in the ligand-binding domain, indicating stronger binding to the mutated receptor. CONCLUSIONS The present study demonstrates the ability of TBECH to activate the hAR, indicating that it is a potential endocrine disruptor.
Collapse
Affiliation(s)
- Hazem Khalaf
- Örebro Life Science Center, Academy of Science and Technology, Örebro University, Örebro, Sweden
| | - Anders Larsson
- Örebro Life Science Center, Academy of Science and Technology, Örebro University, Örebro, Sweden
| | - Håkan Berg
- Örebro Life Science Center, Academy of Science and Technology, Örebro University, Örebro, Sweden
| | - Robert McCrindle
- Wellington Laboratories Inc., Research Division, Guelph, Ontario, Canada
| | - Gilles Arsenault
- Wellington Laboratories Inc., Research Division, Guelph, Ontario, Canada
| | - Per-Erik Olsson
- Örebro Life Science Center, Academy of Science and Technology, Örebro University, Örebro, Sweden
- Address correspondence to P.-E. Olsson, Biology, Örebro Life Science Center, School of Science and Technology, Örebro University, SE-701 82 Örebro, Sweden. Telephone: 46-19-301244. Fax: 46-19-303566. E-mail:
| |
Collapse
|
29
|
Carlson EA, McCulloch C, Koganti A, Goodwin SB, Sutter TR, Silkworth JB. Divergent transcriptomic responses to aryl hydrocarbon receptor agonists between rat and human primary hepatocytes. Toxicol Sci 2009; 112:257-72. [PMID: 19692669 DOI: 10.1093/toxsci/kfp200] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Toxicogenomics has great potential for enhancing our understanding of environmental chemical toxicity, hopefully leading to better informed human health risk assessments. This study employed toxicogenomic technology to reveal species differences in response to two prototypical aryl hydrocarbon receptor (AHR) agonists 2,3,7,8-tetrachlorodibenzo-p-dioxin and the polychlorinated biphenyl (PCB) congener PCB 126. Dose-responses of primary cultures of rat and human hepatocytes were determined using species-specific microarrays sharing over 4000 gene orthologs. Forty-seven human and 79 rat genes satisfied dose-response criteria for both chemicals and were subjected to further analysis including the calculation of the 50% effective concentration and the relative potency (REP) of PCB 126 for each gene. Only five responsive orthologous genes were shared between the two species; yet, the geometric mean of the REPs for all rat and human modeled responsive genes were 0.06 (95% confidence interval [CI]; 0.03-0.1) and 0.002 (95% CI; 0.001-0.005), respectively, suggesting broad species differences in the initial events that follow AHR activation but precede toxicity. This indicates that there are species differences in both the specific genes that responded and the agonist potency and REP for those genes. This observed insensitivity of human cells to PCB 126 is consistent with more traditional measurements of AHR activation (i.e., cytochrome P450 1A1 enzyme activity) and suggests that the species difference in PCB 126 sensitivity is likely due to certain aspects of AHR function. That a species divergence also exists in this expanded AHR-regulated gene repertoire is a novel finding and should help when extrapolating animal data to humans.
Collapse
Affiliation(s)
- Erik A Carlson
- General Electric Company, Global Research Center, Environmental Technology Laboratory, One Research Circle, Niskayuna, New York 12309, USA
| | | | | | | | | | | |
Collapse
|
30
|
|
31
|
Cytotoxicity of ginkgolic acid in HepG2 cells and primary rat hepatocytes. Toxicol Lett 2009; 187:131-6. [DOI: 10.1016/j.toxlet.2009.02.012] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2008] [Revised: 02/04/2009] [Accepted: 02/12/2009] [Indexed: 11/23/2022]
|
32
|
Benfenati E, Benigni R, Demarini DM, Helma C, Kirkland D, Martin TM, Mazzatorta P, Ouédraogo-Arras G, Richard AM, Schilter B, Schoonen WGEJ, Snyder RD, Yang C. Predictive models for carcinogenicity and mutagenicity: frameworks, state-of-the-art, and perspectives. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, ENVIRONMENTAL CARCINOGENESIS & ECOTOXICOLOGY REVIEWS 2009; 27:57-90. [PMID: 19412856 DOI: 10.1080/10590500902885593] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Mutagenicity and carcinogenicity are endpoints of major environmental and regulatory concern. These endpoints are also important targets for development of alternative methods for screening and prediction due to the large number of chemicals of potential concern and the tremendous cost (in time, money, animals) of rodent carcinogenicity bioassays. Both mutagenicity and carcinogenicity involve complex, cellular processes that are only partially understood. Advances in technologies and generation of new data will permit a much deeper understanding. In silico methods for predicting mutagenicity and rodent carcinogenicity based on chemical structural features, along with current mutagenicity and carcinogenicity data sets, have performed well for local prediction (i.e., within specific chemical classes), but are less successful for global prediction (i.e., for a broad range of chemicals). The predictivity of in silico methods can be improved by improving the quality of the data base and endpoints used for modelling. In particular, in vitro assays for clastogenicity need to be improved to reduce false positives (relative to rodent carcinogenicity) and to detect compounds that do not interact directly with DNA or have epigenetic activities. New assays emerging to complement or replace some of the standard assays include Vitotox, GreenScreenGC, and RadarScreen. The needs of industry and regulators to assess thousands of compounds necessitate the development of high-throughput assays combined with innovative data-mining and in silico methods. Various initiatives in this regard have begun, including CAESAR, OSIRIS, CHEMOMENTUM, CHEMPREDICT, OpenTox, EPAA, and ToxCast. In silico methods can be used for priority setting, mechanistic studies, and to estimate potency. Ultimately, such efforts should lead to improvements in application of in silico methods for predicting carcinogenicity to assist industry and regulators and to enhance protection of public health.
Collapse
Affiliation(s)
- E Benfenati
- Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
|
34
|
Hart SN, Cui Y, Klaassen CD, Zhong XB. Three patterns of cytochrome P450 gene expression during liver maturation in mice. Drug Metab Dispos 2009; 37:116-21. [PMID: 18845660 PMCID: PMC2683655 DOI: 10.1124/dmd.108.023812] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2008] [Accepted: 10/06/2008] [Indexed: 01/03/2023] Open
Abstract
The neonatal period of liver development is an often overlooked phase of development. For instance, ontogeny of xenobiotic-metabolizing enzymes can markedly affect biotransformation as the liver matures. To systematically examine the ontogenic gene expression patterns of cytochrome P450 genes (P450) in mice, the gene expression profiles of 19 xenobiotic-metabolizing P450 in Cyp1 to 4 families were determined. The mRNA levels in C57BL/6 mouse livers were quantified using branched DNA technology at the following ages: gestational day 17 (2 days before birth) and postnatal days 0, 1, 3, 5, 10, 15, 20, 30, and 45. Among the 13 P450 genes expressed in mouse livers, three distinct ontogenic expression patterns were identified by cluster analysis. Genes in group 1 (Cyp3a16 as well as 3a41b in male) were expressed in the perinatal period, but they were essentially nondetectable by 30 days of age. Genes in group 2 (Cyp2e1, 3a11, and 4a10 as well as 3a41b in female) quickly increased after birth and reached maximal expression levels by day 5. Genes in group 3 (Cyp1a2, 2a4, 2b10, 2c29, 2d22, 2f2, 3a13, and 3a25) were expressed at low levels until days 10 to 15, but they markedly increased at day 20 to a high and stable level. In conclusion, the developmental expression of P450 in mouse liver can be divided into three patterns, suggesting that different mechanisms are responsible for the expression of P450 during liver maturation.
Collapse
Affiliation(s)
- Steven N Hart
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, Kansas 66160, USA
| | | | | | | |
Collapse
|