1
|
Tian C, Deng S, Yang M, Bai B, Pan Y, Xie G, Zhao D, Wei L. Indole-3-carbinol and its main derivative 3,3'-diindolylmethane: Regulatory roles and therapeutic potential in liver diseases. Biomed Pharmacother 2024; 180:117525. [PMID: 39388997 DOI: 10.1016/j.biopha.2024.117525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
Indole-3-carbinol (I3C), a compound found in cruciferous vegetables, has shown significant efficacy in treating both cancerous and non-cancerous diseases. Its primary derivative, 3,3'-diindolylmethane (DIM), formed during digestion, also exhibits similar therapeutic benefits. In liver disorders, I3C and DIM exhibit dual roles by inhibiting and promoting hepatocellular carcinoma (HCC) and providing relief for nonmalignant liver diseases, such as acute liver injury (ALI), hepatic fibrosis, nonalcoholic fatty liver disease (NAFLD), and alcohol-related liver disease (ALD). Mechanistically, I3C and DIM modulate various pathophysiological processes, including cell proliferation, apoptosis, oxidative stress, and lipogenesis. This review aims to enhance researchers' understanding of the regulatory roles of I3C and DIM in these liver diseases and explore the potential of plant-derived substances in liver disease treatment.
Collapse
Affiliation(s)
- Chao Tian
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China
| | - Shizhou Deng
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China; Research and Development Department, Guangdong Longsee Biomedical Corporation, Guangzhou 510700, China
| | - Ming Yang
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China
| | - Baochen Bai
- Department of Cardiology, Peking University People's hospital, Beijing 100044, China
| | - Yi Pan
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China
| | - Gangqiao Xie
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China; Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Dongliang Zhao
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China
| | - Lai Wei
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China.
| |
Collapse
|
2
|
Singh RD, Avadhesh A, Sharma G, Dholariya S, Shah RB, Goyal B, Gupta SC. Potential of Cytochrome P450, a Family of Xenobiotic Metabolizing Enzymes, in Cancer Therapy. Antioxid Redox Signal 2023; 38:853-876. [PMID: 36242099 DOI: 10.1089/ars.2022.0116] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Significance: Targeted cancer therapy with minimal off-target consequences has shown promise for some cancer types. Although cytochrome P450 (CYP) consists of 18 families, CYP1-4 families play key role in metabolizing xenobiotics and cancer drugs. This eventually affects the process of carcinogenesis, treatment outcomes, and cancer drug resistance. Differential overexpression of CYPs in transformed cells, together with phenotypic alterations in tumors, presents a potential for therapeutic intervention. Recent Advances: Recent advances in molecular tools and information technology have helped utilize CYPs as cancer targets. The precise expression in various tumors, X-ray crystal structures, improved understanding of the structure-activity relationship, and new approaches in the development of prodrugs have supported the ongoing efforts to develop CYP-based drugs with a better therapeutic index. Critical Issues: Narrow therapeutic index, off-target effects, drug resistance, and tumor heterogeneity limit the benefits of CYP-based conventional cancer therapies. In this review, we address the CYP1-4 families as druggable targets in cancer. An emphasis is given to the CYP expression, function, and the possible mechanisms that drive expression and activity in normal and transformed tissues. The strategies that inhibit or activate CYPs for therapeutic benefits are also discussed. Future Directions: Efforts are needed to develop more selective tools that will help comprehend molecular and metabolic alterations in tumor tissues with biological end-points in relation to CYPs. This will eventually translate to developing more specific CYP inhibitors/inducers. Antioxid. Redox Signal. 38, 853-876.
Collapse
Affiliation(s)
- Ragini D Singh
- Department of Biochemistry, All India Institute of Medical Sciences, Rajkot, India
| | - Avadhesh Avadhesh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Gaurav Sharma
- Department of Physiology, All India Institute of Medical Sciences, Rajkot, India
| | - Sagar Dholariya
- Department of Biochemistry, All India Institute of Medical Sciences, Rajkot, India
| | - Rima B Shah
- Department of Pharmacology, All India Institute of Medical Sciences, Rajkot, India
| | - Bela Goyal
- Department of Biochemistry, All India Institute of Medical Sciences, Rishikesh, India
| | - Subash Chandra Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
- Department of Biochemistry, All India Institute of Medical Sciences, Guwahati, India
| |
Collapse
|
3
|
Toydemir G. Screening of the AhR- and Nrf2-linked transcriptional activities of some cruciferous vegetables and nuts in human intestinal epithelial cells as foods containing endogenous AhR ligand precursors. FOOD BIOTECHNOL 2022. [DOI: 10.1080/08905436.2022.2028263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Gamze Toydemir
- Department of Food Engineering, Faculty of Engineering, Alanya Alaaddin Keykubat University, Antalya, Turkey
| |
Collapse
|
4
|
Zhang J, Yan E, Zhang L, Wang T, Wang C. Curcumin reduces oxidative stress and fat deposition in longissimus dorsi muscle of intrauterine growth-retarded finishing pigs. Anim Sci J 2022; 93:e13741. [PMID: 35707899 DOI: 10.1111/asj.13741] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 03/14/2022] [Accepted: 05/02/2022] [Indexed: 11/28/2022]
Abstract
Dietary curcumin possessing multiple biological activities may be an effective way to alleviate oxidative damage and fat deposition in intrauterine growth retardation (IUGR) finishing pigs. Therefore, this study was conducted to evaluate effects of dietary curcumin on meat quality, antioxidant capacity, and fat deposition of longissimus dorsi muscle in IUGR finishing pigs. Twelve normal birth weight (NBW) and 24 IUGR female piglets at 26 days of age were divided into 3 dietary groups: NBW (basal diet), IUGR (basal diet), and IUGR + Cur (basal diet supplemented with 200 mg/kg curcumin). The trial lasted for 169 days. Results showed that IUGR increased concentrations of malondialdehyde (MDA) and protein carbonyls (PC) and fat deposition in longissimus dorsi muscle. However, curcumin decreased the intramuscular fat content and the levels of MDA and PC and improved meat quality in IUGR pigs. Furthermore, curcumin inhibited the decrease of nuclear factor erythroid 2-related factor 2 (Nrf2) protein expression and decreased peroxisome pro liferator-activated receptors γ (PPARγ) expression in IUGR pigs. These findings suggested that dietary addition of 200 mg/kg curcumin could improve meat quality, alleviate oxidative stress through activating Nrf2 signaling pathway, and reduce fat deposition via inhibiting PPARγ expression in longissimus dorsi muscle of IUGR finishing pigs.
Collapse
Affiliation(s)
- Jiaqi Zhang
- College of Animal Science and Technology, National Experimental Teaching Demonstration Center for Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Enfa Yan
- College of Animal Science and Technology, National Experimental Teaching Demonstration Center for Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Lili Zhang
- College of Animal Science and Technology, National Experimental Teaching Demonstration Center for Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Tian Wang
- College of Animal Science and Technology, National Experimental Teaching Demonstration Center for Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Chao Wang
- College of Animal Science and Technology, National Experimental Teaching Demonstration Center for Animal Science, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
5
|
Indole-3-Carbinol (I3C) enhances the sensitivity of murine breast adenocarcinoma cells to doxorubicin (DOX) through inhibition of NF-κβ, blocking angiogenesis and regulation of mitochondrial apoptotic pathway. Chem Biol Interact 2018; 290:19-36. [DOI: 10.1016/j.cbi.2018.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/06/2018] [Accepted: 05/14/2018] [Indexed: 12/21/2022]
|
6
|
Mescher M, Haarmann-Stemmann T. Modulation of CYP1A1 metabolism: From adverse health effects to chemoprevention and therapeutic options. Pharmacol Ther 2018; 187:71-87. [PMID: 29458109 DOI: 10.1016/j.pharmthera.2018.02.012] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The human cytochrome P450 (CYP) 1A1 gene encodes a monooxygenase that metabolizes multiple exogenous and endogenous substrates. CYP1A1 has become infamous for its oxidative metabolism of benzo[a]pyrene and related polycyclic aromatic hydrocarbons, converting these chemicals into very potent human carcinogens. CYP1A1 expression is mainly controlled by the aryl hydrocarbon receptor (AHR), a transcription factor whose activation is induced by binding of persistent organic pollutants, including polycyclic aromatic hydrocarbons and dioxins. Accordingly, induction of CYP1A1 expression and activity serves as a biomarker of AHR activation and associated xenobiotic metabolism as well as toxicity in diverse animal species and humans. Determination of CYP1A1 activity is integrated into modern toxicological concepts and testing guidelines, emphasizing the tremendous importance of this enzyme for risk assessment and regulation of chemicals. Further, CYP1A1 serves as a molecular target for chemoprevention of chemical carcinogenesis, although present literature is controversial on whether its inhibition or induction exerts beneficial effects. Regarding therapeutic applications, first anti-cancer prodrugs are available, which require a metabolic activation by CYP1A1, and thus enable a specific elimination of CYP1A1-positive tumors. However, the application range of these drugs may be limited due to the frequently observed downregulation of CYP1A1 in various human cancers, probably leading to a reduced metabolism of endogenous AHR ligands and a sustained activation of AHR and associated tumor-promoting responses. We here summarize the current knowledge on CYP1A1 as a key player in the metabolism of exogenous and endogenous substrates and as a promising target molecule for prevention and treatment of human malignancies.
Collapse
Affiliation(s)
- Melina Mescher
- IUF - Leibniz-Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | | |
Collapse
|
7
|
Lopez-Vazquez A, Garcia-Banuelos JJ, Gonzalez-Garibay AS, Urzua-Lozano PE, Del Toro-Arreola S, Bueno-Topete MR, Sanchez-Enriquez S, Munoz-Valle JF, Jave-Suarez LF, Armendariz-Borunda J, Bastidas-Ramirez BE. IRS-1 pY612 and Akt-1/PKB pT308 Phosphorylation and Antiinflammatory Effect of Diindolylmethane in Adipocytes Cocultured with Macrophages. Med Chem 2017; 13:727-733. [PMID: 28934926 PMCID: PMC5744426 DOI: 10.2174/1573406413666170922095011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Affiliation(s)
- Alfonso Lopez-Vazquez
- Instituto de Investigacion en Enfermedades Cronico Degenerativas, Departamento de Biologia Molecular y Genomica, Universidad de Guadalajara, Guadalajara, Jalisco. Mexico
| | - Jesus J Garcia-Banuelos
- Instituto de Biologia Molecular en Medicina y Terapia Genica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, puerta 7, 3er. piso, Calle Sierra Mojada # 950, Col. Independencia, Guadalajara, Jalisco, C.P. 44340. Mexico
| | - Angelica S Gonzalez-Garibay
- Instituto de Investigacion en Enfermedades Cronico Degenerativas, Departamento de Biologia Molecular y Genomica, Universidad de Guadalajara, Guadalajara, Jalisco. Mexico
| | - Pedro E Urzua-Lozano
- Instituto de Investigacion en Enfermedades Cronico Degenerativas, Departamento de Biologia Molecular y Genomica, Universidad de Guadalajara, Guadalajara, Jalisco. Mexico
| | - Susana Del Toro-Arreola
- Instituto de Investigacion en Enfermedades Cronico Degenerativas, Departamento de Biologia Molecular y Genomica, Universidad de Guadalajara, Guadalajara, Jalisco. Mexico
| | - Miriam R Bueno-Topete
- Instituto de Investigacion en Enfermedades Cronico Degenerativas, Departamento de Biologia Molecular y Genomica, Universidad de Guadalajara, Guadalajara, Jalisco. Mexico
| | - Sergio Sanchez-Enriquez
- Laboratorio de Bioquimica, Departamento de Biologia Molecular y Genomica, Universidad de Guadalajara, Guadalajara, Jalisco. Mexico
| | - Jose F Munoz-Valle
- Instituto de Investigacion en Ciencias Biomedicas, Departamento de Biologia Molecular y Genomica, Universidad de Guadalajara, Guadalajara, Jalisco. Mexico
| | - Luis F Jave-Suarez
- Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco. Mexico
| | - Juan Armendariz-Borunda
- Instituto de Biologia Molecular en Medicina y Terapia Genica, Departamento de Biologia Molecular y Genomica, Universidad de Guadalajara, Guadalajara, Jalisco. Mexico
| | - Blanca E Bastidas-Ramirez
- Instituto de Investigacion en Enfermedades Cronico Degenerativas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, puerta 7, 2do. piso., Calle Sierra Mojada # 950, Col. Independencia, Guadalajara, Jalisco, C.P. 44340. Mexico
| |
Collapse
|
8
|
Subastri A, Suyavaran A, Preedia Babu E, Nithyananthan S, Barathidasan R, Thirunavukkarasu C. Troxerutin with copper generates oxidative stress in cancer cells: Its possible chemotherapeutic mechanism against hepatocellular carcinoma. J Cell Physiol 2017. [PMID: 28628229 DOI: 10.1002/jcp.26061] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Troxerutin (TXER) a rutin derivative is known for its anticancer effect against hepatocellular carcinoma (HCC). As part of large study, recently we have shown TXER interact with genetic material and its anti-mutagenic property. In the present study we have explored its possible mode of action in HCC. Since TXER alone did not show significant anticancer effect on Huh-7 cells, in vitro biochemical assays were performed for determining anticancer efficacy of TXER + metal complex using transition metals such as Cu, Zn, and Fe. The anticancer efficacy of TXER + Cu on Huh-7 cells were evaluated using MTT assay, DCFDA, JC-1 staining, comet assay, cell cycle analysis, immunocytochemistry, and Western blotting. Non-toxic nature of TXER was analyzed on primary rat hepatocytes. The in vivo efficacy of TXER was tested in N-nitrosodiethylamine initiated and γ-benzene hexachloride and partial hepatectomy promoted rat liver cancer. Liver markers, transition metal levels, histopathological examination, and expression levels of GST-P, 8-OHdG and Ki-67 were studied to assess the in vivo anticancer effect of TXER. We observed that TXER + Cu induced extensive cellular death on Huh-7 cells through generating free radicals and did not possess any toxic effect on normal hepatocytes. The in vivo studies revealed that TXER possess significant anti-cancer effect as assessed through improved liver markers and suppressed GST-P, 8-OHdG, and Ki-67 expression. TXER treatment reduced the hepatic Cu level in cancer bearing animals. Current study brings the putative mechanism involved in anti-cancer effect of TXER, further it will help to formulate phytoconstituents coupled anti-cancer drug for effective treatment of HCC.
Collapse
Affiliation(s)
- Ariraman Subastri
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, India
| | - Arumugam Suyavaran
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, India
| | | | | | - Rajamani Barathidasan
- Centre for Animal Research, Training and Services, CIDRF-DBT, Sri Balaji Vidyapeeth University, Puducherry, India
| | | |
Collapse
|
9
|
Segawa R, Hayashi H, Morita R, Ogawa T, Takimoto N, Hara S, Suzuki K, Shibutani M, Mitsumori K. Modifying effects of liver tumor promotion in rats subjected to co-administration of indole-3-carbinol and phenobarbital. J Toxicol Sci 2014; 39:129-40. [PMID: 24418717 DOI: 10.2131/jts.39.129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Indole-3-carbinol (I3C) and phenobarbital (PB) are cytochrome P450 (CYP) 1A and CYP2B inducers, respectively, and have liver tumor-promoting effects in rats. In this study, we investigated the modifying effects on tumor promotion by I3C and PB co-administration. Six-week-old male F344 rats received a single intraperitoneal injection of N-diethylnitrosamine for initiation treatment. Two weeks after the initiation, rats were given no tumor-promoting agents (DEN alone), I3C (2,500 or 5,000 ppm in diet), PB (60 or 120 ppm in drinking water), or 2,500 ppm I3C + 60 ppm PB for 6 weeks. One week after the I3C/PB treatments, all animals underwent a two-thirds partial hepatectomy. The number and area of liver cell foci positive for glutathione S-transferase placental form (GST-P(+) foci) were not significantly fluctuated in the PB+I3C group in the isoadditive statistical model. On the contrary, the mRNA levels of Cyp2b1/2 and Nqo1 were suppressed and enhanced, respectively, in the PB+I3C group in the isoadditive model, but there was no enhancement in the microsomal reactive oxygen species (ROS) production, thiobarbituric acid-reactive substance levels, and Ki-67(+) cell ratio in this group. The results suggest that the co-administration of I3C and PB causes no modifying effects in liver tumor promotion in rats.
Collapse
Affiliation(s)
- Risa Segawa
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
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]
|
11
|
Yamamoto R, Shimamoto K, Ishii Y, Kimura M, Fujii Y, Morita R, Suzuki K, Shibutani M, Mitsumori K. Involvement of PTEN/Akt signaling and oxidative stress on indole-3-carbinol (I3C)-induced hepatocarcinogenesis in rats. ACTA ACUST UNITED AC 2013; 65:845-52. [PMID: 23290887 DOI: 10.1016/j.etp.2012.12.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 12/06/2012] [Indexed: 10/27/2022]
Abstract
We previously reported that indole-3-carbinol (I3C) had hepatocellular tumor-promoting activity in a short-term (8 weeks) two-stage liver carcinogenesis model in rats. It was suggested that this effect was related to the production of reactive oxygen species (ROS) caused by cytochrome P450 1A (CYP1A) induction. In the present study, 0.5% I3C was administered to DEN-initiated rats for 26 weeks to examine the effect of prolonged administration of I3C and to clarify the possible mechanisms of I3C-induced hepatocarcinogenesis. The number and area of GST-P positive foci, ROS production, TBARS level, 8-OHdG content and mRNA levels of Ahr and Nrf2 gene batteries significantly increased in the DEN-I3C group compared with the DEN-alone group. Furthermore, some GST-P positive preneoplastic foci progressed to hepatocellular adenomas with the prolongation of I3C administration. Lack of PTEN and phospho-Smad2/3 expression and translocations of PDPK1 and phospho-Akt substrates to underneath the cell membrane were observed in the majority of hepatocellular adenomas. In addition, the number of Ki-67 positive cells increased in adenomas compared with the preneoplastic foci. These results suggest that the administration of I3C for 26 weeks in DEN-initiated rats induces tumor progression from hepatocellular altered foci to hepatocellular adenomas by ROS-mediated Akt activation that inhibits the TGF-β/Smad signaling and results in the increased cell proliferation.
Collapse
Affiliation(s)
- Ryuichi Yamamoto
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Hayashi H, Shimamoto K, Taniai E, Ishii Y, Morita R, Suzuki K, Shibutani M, Mitsumori K. Liver tumor promoting effect of omeprazole in rats and its possible mechanism of action. J Toxicol Sci 2012; 37:491-501. [PMID: 22687989 DOI: 10.2131/jts.37.491] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Omeprazole (OPZ), a proton pump inhibitor, is a cytochrome P450 (CYP) 1A1/2 inducer. Some CYP1A inducers are known to have liver tumor promoting effects in rats and the ability to enhance oxidative stress. In this study, we performed a two-stage liver carcinogenesis bioassay in rats to examine the tumor promoting effect of OPZ (Experiment 1) and to clarify a possible mechanism of action (Experiment 2). In Experiment 1, male F344 rats were subjected to a two-third partial hepatectomy, and treated with 0, 138 or 276 mg/kg OPZ by oral gavage once a day for six weeks after an intraperitoneal injection of N-diethylnitrosamine (DEN). Liver weights significantly increased in the DEN+OPZ groups, and the number and area of glutathione S-transferase placental form (GST-P) positive foci significantly increased in the DEN+276 mg/kg OPZ group. In Experiment 2, the same experiment as Experiment 1 was performed, but the dosage of OPZ was 0 or 276 mg/kg. The number and area of GST-P positive foci as well as liver weights significantly increased in the DEN+276 mg/kg OPZ group. The number of proliferative cell nuclear antigen (PCNA)-positive cells also significantly increased in the same group. Real-time RT-PCR showed that the expression of AhR battery genes including Cyp1a1, Cyp1a2, Ugt1a6 and Nqo1, and Nrf2 battery genes including Gpx2, Yc2, Akr7a3, Aldh1a1 Me1 and Ggt1 were significantly upregulated in this group. However, the production of microsomal reactive oxygen species (ROS) and formation of thiobarbituric acid-reactive substances (TBARS) decreased, and 8-hydroxydeoxyguanosine (8-OHdG) content remained unchanged in this group. These results indicate that OPZ, CYP1A inducer, is a liver tumor promoter in rats, but oxidative stress is not involved in the liver tumor promoting effect of OPZ.
Collapse
Affiliation(s)
- Hitomi Hayashi
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Hayashi H, Taniai E, Morita R, Hayashi M, Nakamura D, Wakita A, Suzuki K, Shibutani M, Mitsumori K. Enhanced liver tumor promotion but not liver initiation activity in rats subjected to combined administration of omeprazole and β-naphthoflavone. J Toxicol Sci 2012; 37:969-85. [PMID: 23038005 DOI: 10.2131/jts.37.969] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Omeprazole (OPZ) and β-naphthoflavone (BNF) are cytochrome P450 (CYP)1A inducers and have liver tumor promoting effects. In this study, we investigated the co-promoting and co-initiating effects of OPZ and BNF in rats. In Experiment 1, male rats were subjected to partial hepatectomy (PH), and given oral doses of 138 or 276 mg/kg OPZ, 0.125% or 0.25% BNF or 138 mg/kg OPZ+0.125% BNF (n = 9~12) for 6 weeks after N-diethylnitrosamine (DEN) initiation. In Experiment 2, male rats were treated with oral doses of 138 or 276 mg/kg OPZ, 0.03% or 0.06% BNF or 138 mg/kg OPZ+0.03% BNF (n = 11~12) for 9 days starting 1 week before initiating treatment. As an initiating treatment, 2-Amino-3,4-dimethylimidazo[4,5-f]quinolone (MeIQx) was orally administered 12 hr after PH. The rats were fed a basal diet for 15 days, followed by a diet containing 0.015% 2-acetylaminofluorene for the next 10 days with a single oral dose of carbon tetrachloride. In Experiment 1, the number and area of glutathione S-transferase placental form-positive foci in the OPZ+BNF group were significantly higher than the average values of the High OPZ or the High BNF group. The expression of cyclooxygenase-2 (Cox-2) and COX-2 protein in the liver significantly increased in the OPZ+BNF group. In Experiment 2, liver initiation activity was not enhanced by the co-administration of OPZ+BNF. The results of our studies suggest that the co-administration of OPZ and BNF results in synergistic effects in the liver tumor promotion probably owing to increased COX-2 expression, but no modifying effect in the liver initiation activity of MeIQx in rats.
Collapse
Affiliation(s)
- Hitomi Hayashi
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, Fuchu, Tokyo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Xie XL, Wei M, Kakehashi A, Yamano S, Okabe K, Tajiri M, Wanibuchi H. Dammar resin, a non-mutagen, inducts oxidative stress and metabolic enzymes in the liver of gpt delta transgenic mouse which is different from a mutagen, 2-amino-3-methylimidazo[4,5-f]quinoline. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2012; 748:29-35. [DOI: 10.1016/j.mrgentox.2012.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 06/06/2012] [Accepted: 06/23/2012] [Indexed: 10/28/2022]
|
15
|
Tasaki M, Kuroiwa Y, Inoue T, Hibi D, Matsushita K, Ishii Y, Maruyama S, Nohmi T, Nishikawa A, Umemura T. Oxidative DNA damage andin vivomutagenicity caused by reactive oxygen species generated in the livers ofp53-proficient or -deficientgptdelta mice treated with non-genotoxic hepatocarcinogens. J Appl Toxicol 2012; 33:1433-41. [DOI: 10.1002/jat.2807] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 06/28/2012] [Accepted: 06/28/2012] [Indexed: 12/22/2022]
Affiliation(s)
- Masako Tasaki
- Division of Pathology; National Institute of Health Sciences; 1-18-1, Kamiyoga, Setagaya-ku Tokyo 158-8501 Japan
| | - Yuichi Kuroiwa
- Division of Pathology; National Institute of Health Sciences; 1-18-1, Kamiyoga, Setagaya-ku Tokyo 158-8501 Japan
| | - Tomoki Inoue
- Division of Pathology; National Institute of Health Sciences; 1-18-1, Kamiyoga, Setagaya-ku Tokyo 158-8501 Japan
| | - Daisuke Hibi
- Division of Pathology; National Institute of Health Sciences; 1-18-1, Kamiyoga, Setagaya-ku Tokyo 158-8501 Japan
| | - Kohei Matsushita
- Division of Pathology; National Institute of Health Sciences; 1-18-1, Kamiyoga, Setagaya-ku Tokyo 158-8501 Japan
| | - Yuji Ishii
- Division of Pathology; National Institute of Health Sciences; 1-18-1, Kamiyoga, Setagaya-ku Tokyo 158-8501 Japan
| | - Soichi Maruyama
- Laboratory of Veterinary Public Health, Department of Veterinary Medicine, College of Bioresource Science; Nihon University; 1866, Kameino Fujisawa-city Kanagawa 252-8510 Japan
| | - Takehiko Nohmi
- Division of Genetics and Mutagenesis; National Institute of Health Sciences; 1-18-1, Kamiyoga, Setagaya-ku Tokyo 158-8501 Japan
| | - Akiyoshi Nishikawa
- Biological Safety Research Center; National Institute of Health Sciences; 1-18-1, Kamiyoga, Setagaya-ku Tokyo 158-8501 Japan
| | - Takashi Umemura
- Division of Pathology; National Institute of Health Sciences; 1-18-1, Kamiyoga, Setagaya-ku Tokyo 158-8501 Japan
| |
Collapse
|
16
|
Tsuchiya T, Wang L, Yafune A, Kimura M, Ohishi T, Suzuki K, Mitsumori K, Shibutani M. Disruptive cell cycle regulation involving epigenetic downregulation of Cdkn2a (p16Ink4a) in early-stage liver tumor-promotion facilitating liver cell regeneration in rats. Toxicology 2012; 299:146-54. [DOI: 10.1016/j.tox.2012.05.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 05/14/2012] [Accepted: 05/21/2012] [Indexed: 11/29/2022]
|
17
|
Shimamoto K, Hayashi H, Taniai E, Morita R, Imaoka M, Ishii Y, Suzuki K, Shibutani M, Mitsumori K. Antioxidant N-acetyl-L-cysteine (NAC) supplementation reduces reactive oxygen species (ROS)-mediated hepatocellular tumor promotion of indole-3-carbinol (I3C) in rats. J Toxicol Sci 2012; 36:775-86. [PMID: 22129741 DOI: 10.2131/jts.36.775] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Indole-3-carbinol (I3C) has a liver tumor promoting activity in rats, and is also known as a cytochrome p450 1A (CYP1A) inducer. The generation of reactive oxygen species (ROS) resulting from CYP1A induction due to I3C, is probably involved in the tumor promotion. To clarify whether ROS generation contributes to I3C's induction of hepatocellular altered foci, partially hepatectomized rats were fed a diet containing 0.5% of I3C for 8 weeks with or without 0.3% N-acetyl-L-cysteine (NAC), an antioxidant, in their drinking water after N-diethylnitrosamine (DEN) initiation. Immunohistochemical analysis showed that the glutathione-S-transferase placental form (GST-P) positive foci promoted by I3C were suppressed by the administration of NAC. The mRNAs of members of the phase II nuclear factor, erythroid derived 2, like 2 (Nrf2) gene batteries, whose promoter region is called as antioxidant response element (ARE), were down-regulated in the DEN-I3C-NAC group compared to the DEN-I3C group, but Cyp1a1 was not suppressed in the DEN-I3C-NAC group compared to the DEN-I3C group. There was no marked difference in production of microsomal ROS and genomic 8-hydroxy-2'-deoxygunosine (8-OHdG) as an oxidative DNA marker between the DEN-I3C-NAC and DEN-I3C groups, while mapkapk3 and Myc were decreased by the NAC treatment. These results indicate that oxidative stress plays an important role for I3C's tumor promotion, and NAC suppresses induction of hepatocellular altered foci with suppressed cytoplasmic oxidative stress.
Collapse
Affiliation(s)
- Keisuke Shimamoto
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, Fuchu, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Mikkelsen MD, Buron LD, Salomonsen B, Olsen CE, Hansen BG, Mortensen UH, Halkier BA. Microbial production of indolylglucosinolate through engineering of a multi-gene pathway in a versatile yeast expression platform. Metab Eng 2012; 14:104-11. [PMID: 22326477 DOI: 10.1016/j.ymben.2012.01.006] [Citation(s) in RCA: 197] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 01/17/2012] [Accepted: 01/26/2012] [Indexed: 11/18/2022]
Abstract
Epidemiological studies have shown that consumption of cruciferous vegetables, such as, broccoli and cabbages, is associated with a reduced risk of developing cancer. This phenomenon has been attributed to specific glucosinolates among the ~30 glucosinolates that are typically present as natural products characteristic of cruciferous plants. Accordingly, there has been a strong interest to produce these compounds in microbial cell factories as it will allow production of selected beneficial glucosinolates. We have developed a versatile platform for stable expression of multi-gene pathways in the yeast, Saccharomyces cerevisiae. Introduction of the seven-step pathway of indolylglucosinolate from Arabidopsis thaliana to yeast resulted in the first successful production of glucosinolates in a microbial host. The production of indolylglucosinolate was further optimized by substituting supporting endogenous yeast activities with plant-derived enzymes. Production of indolylglucosinolate serves as a proof-of-concept for our expression platform, and provides a basis for large-scale microbial production of specific glucosinolates for the benefit of human health.
Collapse
Affiliation(s)
- Michael Dalgaard Mikkelsen
- University of Copenhagen, Faculty of Life Sciences, Department of Plant Biology and Biotechnology, Molecular Plant Biology, VKR Research Centre for Pro-Active Plants, 40 Thorvaldsensvej, Frederiksberg C, Copenhagen, Denmark
| | | | | | | | | | | | | |
Collapse
|
19
|
Timme-Laragy AR, Karchner SI, Franks DG, Jenny MJ, Harbeitner RC, Goldstone JV, McArthur AG, Hahn ME. Nrf2b, novel zebrafish paralog of oxidant-responsive transcription factor NF-E2-related factor 2 (NRF2). J Biol Chem 2011; 287:4609-27. [PMID: 22174413 DOI: 10.1074/jbc.m111.260125] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
NF-E2-related factor 2 (NRF2; also called NFE2L2) and related NRF family members regulate antioxidant defenses by activating gene expression via antioxidant response elements (AREs), but their roles in embryonic development are not well understood. We report here that zebrafish (Danio rerio), an important developmental model species, possesses six nrf genes, including duplicated nrf1 and nrf2 genes. We cloned a novel zebrafish nrf2 paralog, nrf2b. The predicted Nrf2b protein sequence shares several domains with the original Nrf2 (now Nrf2a) but lacks the Neh4 transactivation domain. Zebrafish-human comparisons demonstrate conserved synteny involving nrf2 and hox genes, indicating that nrf2a and nrf2b are co-orthologs of human NRF2. nrf2a and nrf2b displayed distinct patterns of expression during embryonic development; nrf2b was more highly expressed at all stages. Embryos in which Nrf2a expression had been knocked down with morpholino oligonucleotides were more sensitive to tert-butylhydroperoxide but not tert-butylhydroquinone, whereas knockdown of Nrf2b did not affect sensitivity of embryos to either chemical. Gene expression profiling by microarray identified a specific role for Nrf2b as a negative regulator of several genes, including p53, cyclin G1, and heme oxygenase 1, in embryos. Nrf2a and Nrf2b exhibited different mechanisms of cross-talk with the Ahr2 signaling pathway. Together, these results demonstrate distinct roles for nrf2a and nrf2b, consistent with subfunction partitioning, and identify a novel negative regulatory role for Nrf2b during development. The identification of zebrafish nrf2 co-orthologs will facilitate new understanding of the multiple roles of NRF2 in protecting vertebrate embryos from oxidative damage.
Collapse
Affiliation(s)
- Alicia R Timme-Laragy
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Kang HJ, Hong YB, Kim HJ, Wang A, Bae I. Bioactive food components prevent carcinogenic stress via Nrf2 activation in BRCA1 deficient breast epithelial cells. Toxicol Lett 2011; 209:154-60. [PMID: 22192953 DOI: 10.1016/j.toxlet.2011.12.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Revised: 12/05/2011] [Accepted: 12/06/2011] [Indexed: 01/11/2023]
Abstract
Although BRCA1 is the most prevalent genetic factor in breast cancer, the pathologic mechanism of tumorigenesis caused by its deficiency has not been elucidated. We have previously demonstrated that BRCA1 can modulate responses to xenobiotic stress by regulating expression of genes involved in metabolic activation, detoxification and antioxidant reactions. In this study, we examined whether BRCA1 deficiency is more vulnerable to xenobiotic stress by employing an in vitro cell model system. Benzo[a]pyrene (B[a]P), used as a xenobiotic insult, increased intracellular reactive oxygen species (ROS) levels in breast epithelial cells. Accumulation of ROS upon B[a]P exposure was significantly augmented by abrogation of BRCA1 compared to the control. Overexpression of Nrf2 in BRCA1 deficient cells reduced elevated ROS to the control levels. Bioactive food components such as sulforaphane (SFN) and resveratrol (RSV) significantly reduced B[a]P-induced ROS accumulation regardless of BRCA1 presence. In addition, these bioactive food components increased Nrf2 levels and Nrf2 transcriptional activity, which led to attenuation of B[a]P-induced DNA damages. Likewise, incubation with bioactive food components reduced B[a]P-mediated DNA damage in BRCA1 deficient cells. In conclusion, we demonstrated that the lack of BRCA1 renders cells more susceptible to ROS-induced DNA damage, which may eventually result in tumorigenesis, and that administration of Nrf2-activating bioactive food components can reduce those risks.
Collapse
Affiliation(s)
- Hyo Jin Kang
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3970 Reservoir Road, NW, Washington, DC 20057, USA
| | | | | | | | | |
Collapse
|
21
|
Ahmad A, Sakr WA, Rahman KW. Mechanisms and therapeutic implications of cell death induction by indole compounds. Cancers (Basel) 2011; 3:2955-74. [PMID: 24212940 PMCID: PMC3759180 DOI: 10.3390/cancers3032955] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 07/07/2011] [Accepted: 07/14/2011] [Indexed: 11/16/2022] Open
Abstract
Indole compounds, obtained from cruciferous vegetables, are well-known for their anti-cancer properties. In particular, indole-3-carbinol (I3C) and its dimeric product, 3,3′-diindolylmethane (DIM), have been widely investigated for their effectiveness against a number of human cancers in vitro as well as in vivo. These compounds are effective inducers of apoptosis and the accumulating evidence documenting their ability to modulate multiple cellular signaling pathways is a testimony to their pleiotropic behavior. Here we attempt to update current understanding on the various mechanisms that are responsible for the apoptosis-inducing effects by these compounds. The significance of apoptosis-induction as a desirable attribute of anti-cancer agents such as indole compounds cannot be overstated. However, an equally intriguing property of these compounds is their ability to sensitize cancer cells to standard chemotherapeutic agents. Such chemosensitizing effects of indole compounds can potentially have major clinical implications because these non-toxic compounds can reduce the toxicity and drug-resistance associated with available chemotherapies. Combinational therapy is increasingly being realized to be better than single agent therapy and, through this review article, we aim to provide a rationale behind combination of natural compounds such as indoles with conventional therapeutics.
Collapse
Affiliation(s)
- Aamir Ahmad
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | | | | |
Collapse
|