1
|
Martins RX, Carvalho M, Maia ME, Flor B, Souza T, Rocha TL, Félix LM, Farias D. 2,4-D Herbicide-Induced Hepatotoxicity: Unveiling Disrupted Liver Functions and Associated Biomarkers. TOXICS 2024; 12:35. [PMID: 38250991 PMCID: PMC10818579 DOI: 10.3390/toxics12010035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/22/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024]
Abstract
2,4-dichlorophenoxyacetic acid (2,4-D) is a widely used herbicide worldwide and is frequently found in water samples. This knowledge has prompted studies on its effects on non-target organisms, revealing significant alterations to liver structure and function. In this review, we evaluated the literature on the hepatotoxicity of 2,4-D, focusing on morphological damages, toxicity biomarkers and affected liver functions. Searches were conducted on PubMed, Web of Science and Scopus and 83 articles were selected after curation. Among these studies, 72% used in vivo models and 30% used in vitro models. Additionally, 48% used the active ingredient, and 35% used commercial formulations in exposure experiments. The most affected biomarkers were related to a decrease in antioxidant capacity through alterations in the activities of catalase, superoxide dismutase and the levels of malondialdehyde. Changes in energy metabolism, lipids, liver function, and xenobiotic metabolism were also identified. Furthermore, studies about the effects of 2,4-D in mixtures with other pesticides were found, as well as hepatoprotection trials. The reviewed data indicate the essential role of reduction in antioxidant capacity and oxidative stress in 2,4-D-induced hepatotoxicity. However, the mechanism of action of the herbicide is still not fully understood and further research in this area is necessary.
Collapse
Affiliation(s)
- Rafael Xavier Martins
- Post-Graduation Program in Biochemistry, Department of Biochemistry and Molecular Biology, Building 907, Campus Pici, Federal University of Ceará, Fortaleza 60455-970, Brazil; (R.X.M.); (M.E.M.)
- Laboratory for Risk Assessment of Novel Technologies, Department of Molecular Biology, Federal University of Paraiba, João Pessoa 58050-085, Brazil; (M.C.); (B.F.); (T.S.)
| | - Matheus Carvalho
- Laboratory for Risk Assessment of Novel Technologies, Department of Molecular Biology, Federal University of Paraiba, João Pessoa 58050-085, Brazil; (M.C.); (B.F.); (T.S.)
| | - Maria Eduarda Maia
- Post-Graduation Program in Biochemistry, Department of Biochemistry and Molecular Biology, Building 907, Campus Pici, Federal University of Ceará, Fortaleza 60455-970, Brazil; (R.X.M.); (M.E.M.)
- Laboratory for Risk Assessment of Novel Technologies, Department of Molecular Biology, Federal University of Paraiba, João Pessoa 58050-085, Brazil; (M.C.); (B.F.); (T.S.)
| | - Bruno Flor
- Laboratory for Risk Assessment of Novel Technologies, Department of Molecular Biology, Federal University of Paraiba, João Pessoa 58050-085, Brazil; (M.C.); (B.F.); (T.S.)
| | - Terezinha Souza
- Laboratory for Risk Assessment of Novel Technologies, Department of Molecular Biology, Federal University of Paraiba, João Pessoa 58050-085, Brazil; (M.C.); (B.F.); (T.S.)
| | - Thiago Lopes Rocha
- Laboratory of Environmental Biotechnology and Ecotoxicology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia 74055-110, Brazil;
| | - Luís M. Félix
- Centre for the Research and Technology of Agro-Environment and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal;
- Inov4Agro, Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
| | - Davi Farias
- Post-Graduation Program in Biochemistry, Department of Biochemistry and Molecular Biology, Building 907, Campus Pici, Federal University of Ceará, Fortaleza 60455-970, Brazil; (R.X.M.); (M.E.M.)
- Laboratory for Risk Assessment of Novel Technologies, Department of Molecular Biology, Federal University of Paraiba, João Pessoa 58050-085, Brazil; (M.C.); (B.F.); (T.S.)
| |
Collapse
|
2
|
von Stackelberg K. A Systematic Review of Carcinogenic Outcomes and Potential Mechanisms from Exposure to 2,4-D and MCPA in the Environment. J Toxicol 2013; 2013:371610. [PMID: 23533401 PMCID: PMC3600329 DOI: 10.1155/2013/371610] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 05/18/2012] [Accepted: 07/11/2012] [Indexed: 12/21/2022] Open
Abstract
Chlorophenoxy compounds, particularly 2,4-dichlorophenoxyacetic acid (2,4-D) and 4-chloro-2-methylphenoxy)acetic acid (MCPA), are amongst the most widely used herbicides in the United States for both agricultural and residential applications. Epidemiologic studies suggest that exposure to 2,4-D and MCPA may be associated with increased risk non-Hodgkins lymphoma (NHL), Hodgkin's disease (HD), leukemia, and soft-tissue sarcoma (STS). Toxicological studies in rodents show no evidence of carcinogenicity, and regulatory agencies worldwide consider chlorophenoxies as not likely to be carcinogenic or unclassifiable as to carcinogenicity. This systematic review assembles the available data to evaluate epidemiologic, toxicological, pharmacokinetic, exposure, and biomonitoring studies with respect to key cellular events noted in disease etiology and how those relate to hypothesized modes of action for these constituents to determine the plausibility of an association between exposure to environmentally relevant concentrations of 2,4-D and MCPA and lymphohematopoietic cancers. The combined evidence does not support a genotoxic mode of action. Although plausible hypotheses for other carcinogenic modes of action exist, a comparison of biomonitoring data to oral equivalent doses calculated from bioassay data shows that environmental exposures are not sufficient to support a causal relationship. Genetic polymorphisms exist that are known to increase the risk of developing NHL. The potential interaction between these polymorphisms and exposures to chlorophenoxy compounds, particularly in occupational settings, is largely unknown.
Collapse
Affiliation(s)
- Katherine von Stackelberg
- E Risk Sciences, LLP, 12 Holton Street, Allston, MA 02134, USA
- Harvard Center for Risk Analysis, 401 Park Drive, Landmark 404J, Boston, MA 02215, USA
| |
Collapse
|
3
|
Butenhoff JL, Bjork JA, Chang SC, Ehresman DJ, Parker GA, Das K, Lau C, Lieder PH, van Otterdijk FM, Wallace KB. Toxicological evaluation of ammonium perfluorobutyrate in rats: twenty-eight-day and ninety-day oral gavage studies. Reprod Toxicol 2011; 33:513-530. [PMID: 21878386 DOI: 10.1016/j.reprotox.2011.08.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 06/20/2011] [Accepted: 08/11/2011] [Indexed: 11/18/2022]
Abstract
Sequential 28-day and 90-day oral toxicity studies were performed in male and female rats with ammonium perfluorobutyrate (NH(4)(+)PFBA) at doses up to 150 and 30mg/kg-d, respectively. Ammonium perfluorooctanoate was used as a comparator at a dose of 30mg/kg-d in the 28-day study. Female rats were unaffected by NH(4)(+)PFBA. Effects in males included: increased liver weight, slight to minimal hepatocellular hypertrophy; decreased serum total cholesterol; and reduced serum thyroxin with no change in serum thyrotropin. During recovery, liver weight, histological, and cholesterol effects were resolved. Results of RT-qPCR were consistent with increased transcriptional expression of the xenosensor nuclear receptors PPARα and CAR as well as the thyroid receptor, and decreased expression of Cyp1A1 (Ah receptor-regulated). No observable adverse effect levels (NOAELs) were 6 and >150mg/kg-d for male and female rats in the 28-day study and 6 and >30mg/kg-d in the 90-dat study, respectively.
Collapse
Affiliation(s)
- John L Butenhoff
- Medical Department, 3M Company, 3M Center 220-06-W-08, St. Paul, MN, USA.
| | - James A Bjork
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Minnesota, Duluth, MN, USA.
| | - Shu-Ching Chang
- Medical Department, 3M Company, 3M Center 220-06-W-08, St. Paul, MN, USA.
| | - David J Ehresman
- Medical Department, 3M Company, 3M Center 220-06-W-08, St. Paul, MN, USA.
| | | | - Kaberi Das
- United States Environmental Protection Agency, National Health and Environmental Effects Laboratory, Research Triangle Park, NC, USA.
| | - Christopher Lau
- United States Environmental Protection Agency, National Health and Environmental Effects Laboratory, Research Triangle Park, NC, USA.
| | - Paul H Lieder
- Medical Department, 3M Company, 3M Center 220-06-W-08, St. Paul, MN, USA.
| | | | - Kendall B Wallace
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Minnesota, Duluth, MN, USA.
| |
Collapse
|
4
|
High throughput systems for screening biomembrane interactions on fabricated mercury film electrodes. J APPL ELECTROCHEM 2011. [DOI: 10.1007/s10800-011-0319-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
5
|
Uptake of phenoxyacetic acid derivatives into Caco-2 cells by the monocarboxylic acid transporters. Toxicol Lett 2009; 189:102-9. [DOI: 10.1016/j.toxlet.2009.05.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Revised: 05/14/2009] [Accepted: 05/14/2009] [Indexed: 11/19/2022]
|
6
|
Bjork JA, Wallace KB. Structure-activity relationships and human relevance for perfluoroalkyl acid-induced transcriptional activation of peroxisome proliferation in liver cell cultures. Toxicol Sci 2009; 111:89-99. [PMID: 19407336 DOI: 10.1093/toxsci/kfp093] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Perfluoroalkyl acids (PFAAs) are widely distributed and environmentally persistent agents whose potential toxicity is not yet fully characterized. Perfluorooctanoic acid (PFOA) and perfluorooctane sulfonic acid elicit a number of potential toxicities in rodents, the most prevalent of which are governed by activation of the peroxisome proliferator-activated receptor alpha (PPARalpha). The purpose of this investigation was twofold: (1) To conduct a structure-activity relationship study of the transcriptional activation of peroxisome proliferation in primary rat liver cell cultures for PFAA-related carboxylic and sulfonic acids of varying carbon chain length and (2) to explore whether this activity can be translated to human liver cells in culture. Exposure to PFOA caused a dose-dependent stimulation of the expression of acyl-CoA oxidase (Acox), Cte/Acot1, and Cyp4a1/11 transcripts that are indicative of peroxisome proliferation in primary rat hepatocytes. PFOA concentrations of 30 microM and above caused cell injury characterized by the expression of Ddit3. Perfluorobutanoic acid (PFBA), on the other hand, stimulated Acox, Cte/Acot1, and Cyp4a1/11 gene expression in primary rat hepatocytes only at concentrations of 100 microM and above. Neither PFOA nor PFBA at concentrations up to 200 microM stimulated PPARalpha-related gene expression in either primary or HepG2 human liver cells. These data demonstrate that (1) PFFAs cause a concentration- and chain length-dependent increase in expression of gene targets related to cell injury and PPARalpha activation in primary rat hepatocytes, (2) the sulfonates are less potent than the corresponding carboxylates in stimulating PPARalpha-related gene expression in rat hepatocytes, and (3) stimulation of PPARalpha-mediated gene transcription is a mechanism that is not shared by human liver cells, adding further substantiation that PPARalpha-dependent liver toxicity in rodents does not extrapolate to assessing human health concerns.
Collapse
Affiliation(s)
- James A Bjork
- Department of Biochemistry and Molecular Biology, University of Minnesota Medical School, Duluth, Minnesota 55812, USA
| | | |
Collapse
|
7
|
O'Brien ML, Spear BT, Glauert HP. Role of Oxidative Stress in Peroxisome Proliferator-Mediated Carcinogenesis. Crit Rev Toxicol 2008; 35:61-88. [PMID: 15742903 DOI: 10.1080/10408440590905957] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
In this review, the evidence about the role of oxidative stress in the induction of hepatocellular carcinomas by peroxisome proliferators is examined. The activation of PPAR-alpha by peroxisome proliferators in rats and mice may produce oxidative stress, due to the induction of enzymes like fatty acyl coenzyme A (CoA) oxidase (AOX) and cytochrome P-450 4A1. The effect of peroxisome proliferators on the antioxidant defense system is reviewed, as is the effect on endpoints resulting from oxidative stress that may be important in carcinogenesis, such as lipid peroxidation, oxidative DNA damage, and transcription factor activation. Peroxisome proliferators clearly inhibit several enzymes in the antioxidant defense system, but studies examining effects on lipid peroxidation and oxidative DNA damage are conflicting. There is a profound species difference in the induction of hepatocellular carcinomas by peroxisome proliferators, with rats and mice being sensitive, whereas species such as nonhuman primates and guinea pigs are not susceptible to the effects of peroxisome proliferators. The possible role of oxidative stress in these species differences is also reviewed. Overall, peroxisome proliferators produce changes in oxidative stress, but whether these changes are important in the carcinogenic process is not clear at this time.
Collapse
Affiliation(s)
- Michelle L O'Brien
- Graduate Centerfor Toxicology, University of Kentucky, Lexington, Kentucky 40506-0054, USA
| | | | | |
Collapse
|
8
|
Perfluorooctane sulfonate (PFOS), perfluorooctanoic acid (PFOA) and their salts Scientific Opinion of the Panel on Contaminants in the Food chain. EFSA J 2008; 6:653. [PMID: 37213838 PMCID: PMC10193653 DOI: 10.2903/j.efsa.2008.653] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
9
|
Chang SC, Das K, Ehresman DJ, Ellefson ME, Gorman GS, Hart JA, Noker PE, Tan YM, Lieder PH, Lau C, Olsen GW, Butenhoff JL. Comparative Pharmacokinetics of Perfluorobutyrate in Rats, Mice, Monkeys, and Humans and Relevance to Human Exposure via Drinking Water. Toxicol Sci 2008; 104:40-53. [DOI: 10.1093/toxsci/kfn057] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
10
|
Abstract
It is well known that various kinds of hypolipidemic drugs induce marked changes in the livers of rats and mice. The initial hepatic responses in rodents are marked hepatomegaly, proliferation of peroxisomes in association with changes in peroxisome structure and enzyme composition. Furthermore, since many of hypolipidemic peroxisome proliferators induce hepatocellular carcinomas in both rats and mice, the relationship between peroxisome proliferation and hepatocarcinogenicity of these drugs has become extremely important. However, it has not yet been established whether there are any direct relationships among pharmacological action, peroxisome proliferation and carcinogenicity of these drugs. In order to clarify this task, we have studied the involvement of HGF in hepatocarcinogenesis caused by peroxisome proliferators. After male F-344 rats were orally given Wy-14,643, hepatocarcinomas and (pre) neoplastic nodules were observed in the livers. At that time, the content of HGF and the expression of HGF mRNA were significantly decreased in the liver tumors. These findings may indicate that decreases in hepatic HGF levels are specific events induced by peroxisome proliferators but not by genotoxic carcinogenesis, and that those changes play an important role in the promotion of neoplastic or preneoplastic cell growth induced by peroxisome proliferators. Decrease in HGF induced by peroxisome proliferators such as Wy-14,643 would inhibit the growth of normal hepatocytes and then lend an advantageous circumstance for the selective growth of neoplastic or preneoplastic cells, resulting in the development of growth of tumors.
Collapse
Affiliation(s)
- Tetsuya Suga
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Science, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| |
Collapse
|
11
|
Intrasuksri U, Rangwala SM, O'Brien M, Noonan DJ, Feller DR. Mechanisms of peroxisome proliferation by perfluorooctanoic acid and endogenous fatty acids. GENERAL PHARMACOLOGY 1998; 31:187-97. [PMID: 9688458 DOI: 10.1016/s0306-3623(98)00029-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
1. The effects of endogenous fatty acids and perfluorooctanoic acid (PFOA) and its analogs on peroxisomal acyl CoA oxidase (ACO) and microsomal laurate hydroxylase (LH) activities were evaluated in primary cultures of rat hepatocytes and activation of peroxisome proliferator-activated receptor alpha (PPARalpha) in CV-1 cells. The rank order for the stimulation of ACO activity in hepatocytes for selected compounds was PFOA >> octanoic acid>octanedioic acid, perfluorooctanol (inactive). Increases in ACO activity by PFOA, like those of ciprofibrate, were associated with a marked increase in peroxisome number and cytosolic occupancy volume. Maximal effects of ciprofibrate and PFOA on the stimulation of ACO activity were not additive, suggesting that these two compounds share a common pathway of peroxisome proliferation. 2. Saturated monocarboxylic acids of C4 to C18 chain length were inactive, and, among dicarboxylic acids, only small elevations (40-45%) in ACO activity were observed with the long-chain C12 and C16 dioic acids. Of the C18 fatty acids tested, only oleic and linoleic acids, at 1 mM, produced a two- to three-fold elevation in ACO and LH activities. In comparison with endogenous fatty acids, PFOA was more potent and exhibited a different time course and greater magnitude of stimulation of ACO and LH activities in cultured hepatocytes. 3. Addition of mitochondrial beta-oxidation inhibitors (3-mercaptopropionic and 2-bromooctanoic acids) did not alter ACO activity in the presence of octanoic acid or octanedioic acid; nor did they modify the stimulation of ACO activity by PFOA. The carnitine palmitoyltransferase I inhibitor 2-bromopalmitic acid produced a 2.5-fold increase in ACO stimulatory activity and reduced both ciprofibrate- and PFOA-mediated stimulations of ACO activity. 4. Cycloheximide treatment reduced PFOA- and ciprofibrate-induced ACO activities; however, the response to oleic acid was not blocked and increased slightly. 5. In rat and human PPARalpha transactivation assays, the rank order of activation was ciprofibrate > PFOA > oleic acid > or = octanoic acid > octanedioic acid or perfluorooctanol (inactive). PFOA, ciprofibrate and oleic acid were activators of rPPARalpha at concentrations that correlated favorably with the changes in ACO activity in cell culture. Octanoic acid did not increase ACO activity and was a weak activator of PPARalpha. 6. Our findings suggest that fatty acids such as oleic acid (endogenous fatty acids) and PFOA (a stable fatty acid) act through more than one pathway to increase ACO activity in rat hepatocytes. We conclude that the potent effects of PFOA are primarily mediated by a mechanism that includes the activation of liver PPARalpha.
Collapse
Affiliation(s)
- U Intrasuksri
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus 43210, USA
| | | | | | | | | |
Collapse
|
12
|
Eacho PI, Foxworthy PS, Lawrence JW, Herron DK, Noonan DJ. Common structural requirements for peroxisome proliferation by tetrazole and carboxylic acid-containing compounds. Ann N Y Acad Sci 1996; 804:387-402. [PMID: 8993558 DOI: 10.1111/j.1749-6632.1996.tb18630.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- P I Eacho
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, USA
| | | | | | | | | |
Collapse
|
13
|
Berthou L, Duverger N, Emmanuel F, Langouët S, Auwerx J, Guillouzo A, Fruchart JC, Rubin E, Denèfle P, Staels B, Branellec D. Opposite regulation of human versus mouse apolipoprotein A-I by fibrates in human apolipoprotein A-I transgenic mice. J Clin Invest 1996; 97:2408-16. [PMID: 8647932 PMCID: PMC507325 DOI: 10.1172/jci118687] [Citation(s) in RCA: 210] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The regulation of liver apolipoprotein (apo) A-I gene expression by fibrates was studied in human apo A-I transgenic mice containing a human genomic DNA fragment driving apo A-I expression in liver. Treatment with fenofibrate (0.5% wt/wt) for 7 d increased plasma human apo A-I levels up to 750% and HDL-cholesterol levels up to 200% with a shift to larger particles. The increase in human apo A-I plasma levels was time and dose dependent and was already evident after 3 d at the highest dose (0.5% wt/wt) of fenofibrate. In contrast, plasma mouse apo A-I concentration was decreased after fenofibrate in nontransgenic mice. The increase in plasma human apo A-I levels after fenofibrate treatment was associated with a 97% increase in hepatic human apo A-I mRNA, whereas mouse apo A-I mRNA levels decreased to 51%. In nontransgenic mice, a similar down-regulation of hepatic apo A-I mRNA levels was observed. Nuclear run-on experiments demonstrated that the increase in human apo A-I and the decrease in mouse apo A-I gene expression after fenofibrate occurred at the transcriptional level. Since part of the effects of fibrates are mediated through the nuclear receptor PPAR (peroxisome proliferator-activated receptor), the expression of the acyl CoA oxidase (ACO) gene was measured as a control of PPAR activation. Both in transgenic and nontransgenic mice, fenofibrate induced ACO mRNA levels up to sixfold. When transgenic mice were treated with gemfibrozil (0.5% wt/wt) plasma human apo A-I and HDL-cholesterol levels increased 32 and 73%, respectively, above control levels. The weaker effect of this compound on human apo A-I and HDL-cholesterol levels correlated with a less pronounced impact on ACO mRNA levels (a threefold increase) suggesting that the level of induction of human apo A-I gene is related to the PPAR activating potency of the fibrate used. Treatment of human primary hepatocytes with fenofibric acid (500 microM) provoked an 83 and 50% increase in apo A-I secretion and mRNA levels, respectively, supporting that a direct action of fibrates on liver human apo A-I production leads to the observed increase in plasma apo A4 and HDL-cholesterol.
Collapse
Affiliation(s)
- L Berthou
- Department of Biotechnology C.R.V.A. Rhône-Poulenc Rorer Gencell, Vitry-sur-Seine, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Labelle M, Belley M, Gareau Y, Gauthier J, Guay D, Gordon R, Grossman S, Jones T, Leblanc Y, McAuliffe M, McFarlane C, Masson P, Metters K, Ouimet N, Patrick D, Piechuta H, Rochette C, Sawyer N, Xiang Y, Pickett C, Ford-Hutchinson A, Zamboni R, Young R. Discovery of MK-0476, a potent and orally active leukotriene D4 receptor antagonist devoid of peroxisomal enxyme induction. Bioorg Med Chem Lett 1995. [DOI: 10.1016/0960-894x(95)00023-m] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
15
|
Tomaszewski KE, Melnick RL. In vitro evidence for involvement of CoA thioesters in peroxisome proliferation and hypolipidaemia. BIOCHIMICA ET BIOPHYSICA ACTA 1994; 1220:118-24. [PMID: 7906145 DOI: 10.1016/0167-4889(94)90126-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The mechanisms of peroxisomal induction and hypolipidaemia caused by treatment with peroxisome proliferators, such as nafenopin and clofibrate, remain to be elucidated. Proposed mechanisms include receptor-mediated processes or adaptations resulting from disruption of hepatic lipid metabolism. The latter mechanism was investigated in a series of in vitro studies. Incubation of primary rat hepatocytes with various carboxyl-containing compounds revealed no clear common factor which imparted potency as a peroxisomal inducer. Inhibitors of fatty acyl-CoA synthetase, norepinephrine and desulpho-CoA, however, decreased the level of peroxisomal induction by nafenopin in rat hepatocytes, suggesting that activation of carboxyl-containing compounds to their CoA thioesters may be a necessary step in initiating peroxisome proliferation. Coenzyme A thioesters of nafenopin, clofibric acid and other carboxyl-containing chemicals were synthesised and found to inhibit the activity of acetyl-CoA carboxylase to varying degrees. The CoA thioester of nafenopin was the most potent inhibitor among this group (Ki = 1.45 x 10(-5) M), but weaker than palmitoyl-CoA (Ki = 2.22 x 10(-6) M), the feedback inhibitor of acetyl-CoA carboxylase. Hypolipidaemia caused by treatment with peroxisome proliferators may, therefore, be related to inhibition of fatty-acid synthesis by the corresponding CoA thioester derivative.
Collapse
Affiliation(s)
- K E Tomaszewski
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709
| | | |
Collapse
|
16
|
Ahmad D, Caldwell J. Lack of stereoselectivity of the peroxisome proliferation induced by 2-phenylpropionic acid: evidence against a role for lipid disturbance in peroxisome proliferation. Chirality 1994; 6:365-71. [PMID: 8068495 DOI: 10.1002/chir.530060502] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The significance of disturbances of lipid metabolism caused by xenobiotic acyl-CoAs as possible causes of peroxisomal proliferation has been studied with the enantiomers of 2-phenylpropionic acid (2-PPA), the (R)-enantiomer of which is converted to the acyl-CoA in rats while its (S)-antipode is not. rac-2-PPA (250 mg/kg/day ip x 3) was shown to be an hepatic peroxisomal proliferator in male Sprague-Dawley rats on the basis of increases in microsomal cytochrome P-450 content and lauric acid hydroxylation and hepatic CN(-)-insensitive palmitoyl-CoA oxidation, a peroxisomal marker activity, while electron microscopy revealed a rise in the peroxisome/mitochondria ratio in hepatocytes. Further studies established the dose-response relationships for these biochemical changes. The (R)- and (S)-enantiomers were administered at a dose of 50 mg/kg/day ip x 3 and both were peroxisome proliferators of very similar potency. The effects of 100 mg/kg/day ip x 3 of the racemate, a dose giving ca. 75% of maximal response, were essentially additive of those of 50 mg/kg/day ip x 3 of its two component isomers. The stereoselectivity of acyl-CoA formation from the enantiomers of 2-PPA was confirmed by their differential inhibition of microsomal palmitoyl-CoA synthesis. Taken together, these data indicate that it is very unlikely that the acyl-CoA of 2-PPA plays any role in the peroxisomal proliferation which this compound causes in the rat.
Collapse
Affiliation(s)
- D Ahmad
- Department of Pharmacology and Toxicology, St. Mary's Hospital Medical School, Imperial College of Science, Technology and Medicine, London, England
| | | |
Collapse
|
17
|
Hosokawa M, Satoh T. Differences in the induction of carboxylesterase isozymes in rat liver microsomes by perfluorinated fatty acids. Xenobiotica 1993; 23:1125-33. [PMID: 8259694 DOI: 10.3109/00498259309059427] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
1. Differences in the ability of metabolically-inert peroxisome proliferators (perfluoro-n-decanoic acid (PFDA, C10), perfluoro-n-octanoic acid (PFOA, C8), perfluorooctane sulphonic acid (PFOS, C8) and 1-H,1-H-pentadecafluoro-n-octanol (PFOL, C8)) to induce three forms of hepatic microsomal carboxylesterase, namely RL1, RL2 and RH1, in the male rat were studied by measuring changes in hydrolytic activities towards p-nitrophenyl acetate (PNPA), isocarboxazid (ISOC) and butanilicaine (BUTA), which are thought to be specific substrates for RL1, RL2 and RH1, respectively, and by evaluating changes in the contents of the three isozymes by radial immunodiffusion assay with specific antibodies. 2. The administration of PFDA rather specifically decreases PNPA hydrolase activity and RL1 content. On the other hand, PFOA, PFOS and PFOL markedly increase all three hydrolase activities and the content of all three isozymes (except RH1 in the case of PFOA, where the increase was not statistically significant). 3. The correlations between hydrolase activities and isozyme contents supported specificity of the three substrates, with the exception that the content of the predominant isozyme, RL2, showed a higher correlation with BUTA hydrolase activity than with ISOC hydrolase activity. 4. In conclusion, we have demonstrated that metabolically-inert perfluorinated fatty acids induce hepatic microsomal carboxylesterase isozymes, as determined by radial immunodiffusion analysis using specific antibodies. This is the first report that perfluorinated fatty acid affect carboxylesterase isozymes in rat liver microsomes, and is indicative of the importance of peroxisome proliferators in hepatic metabolism of xenobiotics. Further work is needed to determine the regulatory mechanisms involved.
Collapse
Affiliation(s)
- M Hosokawa
- Laboratory of Biochemical Pharmacology and Biotoxicology, Faculty of Pharmaceutical Sciences, Chiba University, Japan
| | | |
Collapse
|
18
|
Watanabe T, Okawa S, Itoga H, Imanaka T, Suga T. Involvement of calmodulin- and protein kinase C-related mechanism in an induction process of peroxisomal fatty acid oxidation-related enzymes by hypolipidemic peroxisome proliferators. BIOCHIMICA ET BIOPHYSICA ACTA 1992; 1135:84-90. [PMID: 1591274 DOI: 10.1016/0167-4889(92)90170-g] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Trifluoperazine, a calmodulin antagonist, suppressed the clofibric acid-evoked induction of the peroxisomal cyanide-insensitive fatty acyl-CoA oxidizing system and carnitine acetyltransferase in rat liver and also in cultured rat hepatocytes. H-7, a potent inhibitor of protein kinase C, also suppressed the induction of these enzymes by clofibric acid, bezafibrate, Wyl4,643 or mono(2-ethylhexyl)phthalate in cultured rat hepatocytes. This suppressive effect was also confirmed by the protein composition of hepatocytes treated with clofibric acid and these antagonists, where the increase in the amount of peroxisomal bifunctional enzyme by peroxisome proliferator was markedly suppressed by above two antagonists. Profile of the time-dependent changes in the activities of the two enzymes after clofibric acid treatment showed that there might be two phases in the induction process. The initial phase (0-3 days after the treatment) showed a relative low inducing rate and subsequent phase (3-5 days after the treatment) showed an abrupt induction. The suppressive effect of the above two antagonists was significant in the later phase. In a time course study of the induction process of peroxisomal catalase, bifunctional enzyme or 69 kDa integral membrane protein using immunochemical detection, the induction of the membrane protein by clofibric acid was delayed compared with that of the bifunctional enzyme, where the induction was inhibited almost completely by nicardipine. These experimental results suggest that calmodulin- and protein kinase C-dependent processes play an important role in the process of marked induction of peroxisomal enzymes and membrane protein by drugs in rat liver.
Collapse
Affiliation(s)
- T Watanabe
- Department of Clinical Biochemistry, Tokyo College of Pharmacy, Hachioji, Japan
| | | | | | | | | |
Collapse
|