1
|
Gregoris F, Minervini G, Tosatto SCE. In Silico Exploration of AHR-HIF Pathway Interplay: Implications for Therapeutic Targeting in ccRCC. Genes (Basel) 2024; 15:1167. [PMID: 39336758 PMCID: PMC11431742 DOI: 10.3390/genes15091167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
The oxygen-sensing pathway is a crucial regulatory circuit that defines cellular conditions and is extensively exploited in cancer development. Pathogenic mutations in the von Hippel-Lindau (VHL) tumour suppressor impair its role as a master regulator of hypoxia-inducible factors (HIFs), leading to constitutive HIF activation and uncontrolled angiogenesis, increasing the risk of developing clear cell renal cell carcinoma (ccRCC). HIF hyperactivation can sequester HIF-1β, preventing the aryl hydrocarbon receptor (AHR) from correctly activating gene expression in response to endogenous and exogenous ligands such as TCDD (dioxins). In this study, we used protein-protein interaction networks and gene expression profiling to characterize the impact of VHL loss on AHR activity. Our findings reveal specific expression patterns of AHR interactors following exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and in ccRCC. We identified several AHR interactors significantly associated with poor survival rates in ccRCC patients. Notably, the upregulation of the androgen receptor (AR) and retinoblastoma-associated protein (RB1) by TCDD, coupled with their respective downregulation in ccRCC and association with poor survival rates, suggests novel therapeutic targets. The strategic activation of the AHR via selective AHR modulators (SAhRMs) could stimulate its anticancer activity, specifically targeting RB1 and AR to reduce cell cycle progression and metastasis formation in ccRCC. Our study provides comprehensive insights into the complex interplay between the AHR and HIF pathways in ccRCC pathogenesis, offering novel strategies for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Francesco Gregoris
- Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121 Padova, Italy
| | - Giovanni Minervini
- Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121 Padova, Italy
| | - Silvio C E Tosatto
- Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121 Padova, Italy
| |
Collapse
|
2
|
Chaudhry KA, Bianchi-Smiraglia A. The aryl hydrocarbon receptor as a tumor modulator: mechanisms to therapy. Front Oncol 2024; 14:1375905. [PMID: 38807762 PMCID: PMC11130384 DOI: 10.3389/fonc.2024.1375905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 05/03/2024] [Indexed: 05/30/2024] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that is widely recognized to play important, but complex, modulatory roles in a variety of tumor types. In this review, we comprehensively summarize the increasingly controversial role of AhR as a tumor regulator and the mechanisms by which it alters tumor progression based on the cancer cell type. Finally, we discuss new and emerging strategies to therapeutically modulate AhR, focusing on novel agents that hold promise in current human clinical trials as well as existing FDA-approved drugs that could potentially be repurposed for cancer therapy.
Collapse
Affiliation(s)
| | - Anna Bianchi-Smiraglia
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, NY, United States
| |
Collapse
|
3
|
Elson DJ, Nguyen BD, Korjeff NA, Wilferd SF, Puig-Sanvicens V, Sang Jang H, Bernales S, Chakravarty S, Belmar S, Ureta G, Finlay D, Plaisier CL, Kolluri SK. Suppression of Ah Receptor (AhR) increases the aggressiveness of TNBC cells and 11-Cl-BBQ-activated AhR inhibits their growth. Biochem Pharmacol 2023; 215:115706. [PMID: 37506922 DOI: 10.1016/j.bcp.2023.115706] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/21/2023] [Accepted: 07/25/2023] [Indexed: 07/30/2023]
Abstract
Triple-negative breast cancer (TNBC) represents around 15% of the 2.26 million breast cancers diagnosed worldwide annually and has the worst outcome. Despite recent therapeutic advances, there remains a lack of targeted therapies for this breast cancer subtype. The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor with biological roles in regulating development, xenobiotic metabolism, cell cycle progression and cell death. AhR activation by select ligands can promote tumor suppression in multiple cancer types. AhR can negatively regulate the activity of different oncogenic signaling pathways and can directly upregulate tumor suppressor genes such as p27Kip1. To determine the role of AhR in TNBC, we generated AhR-deficient cancer cells and investigated the impact of AhR loss on TNBC cell growth phenotypes. We found that AhR-deficient MDA-MB-468 TNBC cells have increased proliferation and formed significantly more colonies compared to AhR expressing cells. These cells without AhR expression grew aggressively in vivo. To determine the molecular targets driving this phenotype, we performed transcriptomic profiling in AhR expressing and AhR knockout MDA-MB-468 cells and identified tyrosine receptor kinases, as well as other genes involved in proliferation, survival and clonogenicity that are repressed by AhR. In order to determine therapeutic targeting of AhR in TNBC, we investigated the anti-cancer effects of the novel AhR ligand 11-chloro-7H-benzimidazo[2,1-a]benzo[de]iso-quinolin-7-one (11-Cl-BBQ), which belongs to a class of high affinity, rapidly metabolized AhR ligands called benzimidazoisoquinolines (BBQs). 11-Cl-BBQ induced AhR-dependent cancer cell-selective growth inhibition and strongly inhibited colony formation in TNBC cells.
Collapse
Affiliation(s)
- Daniel J Elson
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, United States
| | - Bach D Nguyen
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, United States
| | - Nicholas A Korjeff
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, United States
| | - Sierra F Wilferd
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, United States
| | - Veronica Puig-Sanvicens
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, United States
| | - Hyo Sang Jang
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, United States
| | - Sebastian Bernales
- Praxis Biotech, San Francisco, CA 94158, United States; Fundación Ciencia & Vida, Centro Científico y Tecnológico Ciencia & Vida, Avda. Del valle Norte 725, Santiago, Chile
| | | | - Sebastián Belmar
- Praxis Biotech, San Francisco, CA 94158, United States; Merken Biotech, Avda. Del valle Norte 725, Santiago, Chile
| | - Gonzalo Ureta
- Praxis Biotech, San Francisco, CA 94158, United States; Merken Biotech, Avda. Del valle Norte 725, Santiago, Chile
| | - Darren Finlay
- Sanford Burnham Prebys Medical Discovery Institute, NCI Designated Cancer Center, La Jolla, CA 92037, United States
| | - Christopher L Plaisier
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, United States
| | - Siva K Kolluri
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, United States; Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, United States.
| |
Collapse
|
4
|
Chong ZX, Yong CY, Ong AHK, Yeap SK, Ho WY. Deciphering the roles of aryl hydrocarbon receptor (AHR) in regulating carcinogenesis. Toxicology 2023; 495:153596. [PMID: 37480978 DOI: 10.1016/j.tox.2023.153596] [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: 06/27/2023] [Revised: 07/13/2023] [Accepted: 07/16/2023] [Indexed: 07/24/2023]
Abstract
Aryl hydrocarbon receptor (AHR) is a ligand-dependent receptor that belongs to the superfamily of basic helix-loop-helix (bHLH) transcription factors. The activation of the canonical AHR signaling pathway is known to induce the expression of cytochrome P450 enzymes, facilitating the detoxification metabolism in the human body. Additionally, AHR could interact with various signaling pathways such as epidermal growth factor receptor (EGFR), signal transducer and activator of transcription 3 (STAT3), hypoxia-inducible factor-1α (HIF-1α), nuclear factor ekappa B (NF-κβ), estrogen receptor (ER), and androgen receptor (AR) signaling pathways. Over the past 30 years, several studies have reported that various chemical, physical, or biological agents, such as tobacco, hydrocarbon compounds, industrial and agricultural chemical wastes, drugs, UV, viruses, and other toxins, could affect AHR expression or activity, promoting cancer development. Thus, it is valuable to overview how these factors regulate AHR-mediated carcinogenesis. Current findings have reported that many compounds could act as AHR ligands to drive the expressions of AHR-target genes, such as CYP1A1, CYP1B1, MMPs, and AXL, and other targets that exert a pro-proliferation or anti-apoptotic effect, like XIAP. Furthermore, some other physical and chemical agents, such as UV and 3-methylcholanthrene, could promote AHR signaling activities, increasing the signaling activities of a few oncogenic pathways, such as the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathways. Understanding how various factors regulate AHR-mediated carcinogenesis processes helps clinicians and scientists plan personalized therapeutic strategies to improve anti-cancer treatment efficacy. As many studies that have reported the roles of AHR in regulating carcinogenesis are preclinical or observational clinical studies that did not explore the detailed mechanisms of how different chemical, physical, or biological agents promote AHR-mediated carcinogenesis processes, future studies should focus on conducting large-scale and functional studies to unravel the underlying mechanism of how AHR interacts with different factors in regulating carcinogenesis processes.
Collapse
Affiliation(s)
- Zhi Xiong Chong
- Faculty of Science and Engineering, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia
| | - Chean Yeah Yong
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, 43900 Sepang, Selangor, Malaysia
| | - Alan Han Kiat Ong
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, 43000 Kajang, Malaysia
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, 43900 Sepang, Selangor, Malaysia.
| | - Wan Yong Ho
- Faculty of Science and Engineering, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia.
| |
Collapse
|
5
|
Elson D, Nguyen BD, Bernales S, Chakravarty S, Jang HS, Korjeff NA, Zhang Y, Wilferd SF, Castro DJ, Plaisier CL, Finlay D, Oshima RG, Kolluri SK. Induction of Aryl Hydrocarbon Receptor-Mediated Cancer Cell-Selective Apoptosis in Triple-Negative Breast Cancer Cells by a High-Affinity Benzimidazoisoquinoline. ACS Pharmacol Transl Sci 2023; 6:1028-1042. [PMID: 37470014 PMCID: PMC10353065 DOI: 10.1021/acsptsci.2c00253] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Indexed: 07/21/2023]
Abstract
Triple-negative breast cancer (TNBC) remains a disease with a paucity of targeted treatment opportunities. The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that is involved in a wide range of physiological processes, including the sensing of xenobiotics, immune function, development, and differentiation. Different small-molecule AhR ligands drive strikingly varied cellular and organismal responses. In certain cancers, AhR activation by select small molecules induces cell cycle arrest or apoptosis via activation of tumor-suppressive transcriptional programs. AhR is expressed in triple-negative breast cancers, presenting a tractable therapeutic opportunity. Here, we identify a novel ligand of the aryl hydrocarbon receptor that potently and selectively induces cell death in triple-negative breast cancer cells and TNBC stem cells via the AhR. Importantly, we found that this compound, Analog 523, exhibits minimal cytotoxicity against multiple normal human primary cells. Analog 523 represents a high-affinity AhR ligand with potential for future clinical translation as an anticancer agent.
Collapse
Affiliation(s)
- Daniel
J. Elson
- Cancer
Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, 97331, United States
| | - Bach D. Nguyen
- Cancer
Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, 97331, United States
| | - Sebastian Bernales
- Praxis
Biotech, San Francisco, California, 94158, United States
- Centro Ciencia
& Vida, Avda. Del
Valle Norte 725, Santiago, 8580702, Chile
| | | | - Hyo Sang Jang
- Cancer
Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, 97331, United States
| | - Nicholas A. Korjeff
- Cancer
Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, 97331, United States
| | - Yi Zhang
- Cancer
Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, 97331, United States
| | - Sierra F. Wilferd
- School
of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
| | - David J. Castro
- Sanford
Burnham Prebys Medical Discovery Institute, NCI Designated Cancer
Center, La Jolla, California, 92037, United States
- Oregon Health
& Science University, Portland, Oregon, 97239, United States
| | - Christopher L. Plaisier
- School
of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
| | - Darren Finlay
- Sanford
Burnham Prebys Medical Discovery Institute, NCI Designated Cancer
Center, La Jolla, California, 92037, United States
| | - Robert G. Oshima
- Sanford
Burnham Prebys Medical Discovery Institute, NCI Designated Cancer
Center, La Jolla, California, 92037, United States
| | - Siva K. Kolluri
- Cancer
Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, 97331, United States
- Linus
Pauling Institute, Oregon State University, Corvallis, Oregon, 97331, United
States
- The
Pacific Northwest Center for Translational Environmental Health Research, Oregon State University, Corvallis, Oregon, 97331, United States
| |
Collapse
|
6
|
Nguyen BD, Stevens BL, Elson DJ, Finlay D, Gamble J, Kopparapu P, Tanguay RL, Buermeyer AB, Kerkvliet NI, Kolluri SK. 11-Cl-BBQ, a select modulator of AhR-regulated transcription, suppresses lung cancer cell growth via activation of p53 and p27 Kip1. FEBS J 2023; 290:2064-2084. [PMID: 36401795 PMCID: PMC10807707 DOI: 10.1111/febs.16683] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 09/01/2022] [Accepted: 11/17/2022] [Indexed: 11/21/2022]
Abstract
Aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor and functions as a tumour suppressor in different cancer models. In the present study, we report detailed characterization of 11-chloro-7H-benzimidazo[2,1-a]benzo[de]iso-quinolin-7-one (11-Cl-BBQ) as a select modulator of AhR-regulated transcription (SMAhRT) with anti-cancer actions. Treatment of lung cancer cells with 11-Cl-BBQ induced potent and sustained AhR-dependent anti-proliferative effects by promoting G1 phase cell cycle arrest. Investigation of 11-Cl-BBQ-induced transcription in H460 cells with or without the AhR expression by RNA-sequencing revealed activation of p53 signalling. In addition, 11-Cl-BBQ suppressed multiple pathways involved in DNA replication and increased expression of cyclin-dependent kinase inhibitors, including p27Kip1 , in an AhR-dependent manner. CRISPR/Cas9 knockout of individual genes revealed the requirement for both p53 and p27Kip1 for the AhR-mediated anti-proliferative effects. Our results identify 11-Cl-BBQ as a potential lung cancer therapeutic, highlight the feasibility of targeting AhR and provide important mechanistic insights into AhR-mediated-anticancer actions.
Collapse
Affiliation(s)
- Bach D. Nguyen
- Cancer Research Laboratory, Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331
| | - Brenna L. Stevens
- Cancer Research Laboratory, Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331
| | - Daniel J. Elson
- Cancer Research Laboratory, Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331
| | - Darren Finlay
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - John Gamble
- Cancer Research Laboratory, Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331
| | - Prasad Kopparapu
- Cancer Research Laboratory, Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331
| | - Robyn L. Tanguay
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
- The Pacific Northwest Center for Translational Environmental Health Research, Oregon State University, Corvallis, OR, 97331, USA
| | - Andrew B. Buermeyer
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331
| | - Nancy I. Kerkvliet
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331
| | - Siva K. Kolluri
- Cancer Research Laboratory, Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
- The Pacific Northwest Center for Translational Environmental Health Research, Oregon State University, Corvallis, OR, 97331, USA
| |
Collapse
|
7
|
Elson DJ, Kolluri SK. Tumor-Suppressive Functions of the Aryl Hydrocarbon Receptor (AhR) and AhR as a Therapeutic Target in Cancer. BIOLOGY 2023; 12:526. [PMID: 37106727 PMCID: PMC10135996 DOI: 10.3390/biology12040526] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/25/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor involved in regulating a wide range of biological responses. A diverse array of xenobiotics and endogenous small molecules bind to the receptor and drive unique phenotypic responses. Due in part to its role in mediating toxic responses to environmental pollutants, AhR activation has not been traditionally viewed as a viable therapeutic approach. Nonetheless, the expression and activation of AhR can inhibit the proliferation, migration, and survival of cancer cells, and many clinically approved drugs transcriptionally activate AhR. Identification of novel select modulators of AhR-regulated transcription that promote tumor suppression is an active area of investigation. The development of AhR-targeted anticancer agents requires a thorough understanding of the molecular mechanisms driving tumor suppression. Here, we summarized the tumor-suppressive mechanisms regulated by AhR with an emphasis on the endogenous functions of the receptor in opposing carcinogenesis. In multiple different cancer models, the deletion of AhR promotes increased tumorigenesis, but a precise understanding of the molecular cues and the genetic targets of AhR involved in this process is lacking. The intent of this review was to synthesize the evidence supporting AhR-dependent tumor suppression and distill insights for development of AhR-targeted cancer therapeutics.
Collapse
Affiliation(s)
- Daniel J. Elson
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - Siva K. Kolluri
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
8
|
Wu J, Pang T, Lin Z, Zhao M, Jin H. The key player in the pathogenesis of environmental influence of systemic lupus erythematosus: Aryl hydrocarbon receptor. Front Immunol 2022; 13:965941. [PMID: 36110860 PMCID: PMC9468923 DOI: 10.3389/fimmu.2022.965941] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/01/2022] [Indexed: 11/28/2022] Open
Abstract
The aryl hydrocarbon receptor was previously known as an environmental receptor that modulates the cellular response to external environmental changes. In essence, the aryl hydrocarbon receptor is a cytoplasmic receptor and transcription factor that is activated by binding to the corresponding ligands, and they transmit relevant information by binding to DNA, thereby activating the transcription of various genes. Therefore, we can understand the development of certain diseases and discover new therapeutic targets by studying the regulation and function of AhR. Several autoimmune diseases, including systemic lupus erythematosus (SLE), have been connected to AhR in previous studies. SLE is a classic autoimmune disease characterized by multi-organ damage and disruption of immune tolerance. We discuss here the homeostatic regulation of AhR and its ligands among various types of immune cells, pathophysiological roles, in addition to the roles of various related cytokines and signaling pathways in the occurrence and development of SLE.
Collapse
|
9
|
Elson DJ, Nguyen BD, Wood R, Zhang Y, Puig-Sanvicens V, Kolluri SK. The cyclin-dependent kinase inhibitor p27 Kip1 interacts with the aryl hydrocarbon receptor and negatively regulates its transcriptional activity. FEBS Lett 2022; 596:2056-2071. [PMID: 35735777 DOI: 10.1002/1873-3468.14434] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 11/11/2022]
Abstract
p27Kip1 functions to coordinate cell cycle progression through the inhibition of cyclin-dependent kinase (CDK) complexes. p27Kip1 also exerts distinct activities beyond CDK-inhibition, including functioning as a transcriptional regulator. The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor with diverse biological roles. The regulatory inputs that control AhR-mediated transcriptional responses are an active area of investigation. AhR was previously established as a direct regulator of p27Kip1 transcription. Here, we report the physical interaction of AhR and p27Kip1 and show that p27Kip1 expression negatively regulates AhR-mediated transcription. p27Kip1 knockout cells display increased AhR nuclear localisation and significantly higher expression of AhR target genes. This work thus identifies new regulatory cross-talk between p27Kip1 and AhR.
Collapse
Affiliation(s)
- Daniel J Elson
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, USA
| | - Bach D Nguyen
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, USA
| | - Rhand Wood
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, USA
| | - Yi Zhang
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, USA
| | - Veronica Puig-Sanvicens
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, USA
| | - Siva K Kolluri
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, USA.,Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| |
Collapse
|
10
|
Teriflunomide Loaded SPION Nanoparticles Induced Apoptosis in MDA-MB-231 Breast Cancer Cells. J CLUST SCI 2022. [DOI: 10.1007/s10876-022-02327-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
11
|
Luo C, Ai J, Ren E, Li J, Feng C, Li X, Luo X. Research progress on evodiamine, a bioactive alkaloid of Evodiae fructus: Focus on its anti-cancer activity and bioavailability (Review). Exp Ther Med 2021; 22:1327. [PMID: 34630681 DOI: 10.3892/etm.2021.10762] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 08/17/2021] [Indexed: 12/24/2022] Open
Abstract
Evodiae fructus (Wu-Zhu-Yu in Chinese) can be isolated from the dried, unripe fruits of Tetradium ruticarpum and is a well-known traditional Chinese medicine that is applied extensively in China, Japan and Korea. Evodiae fructus has been traditionally used to treat headaches, abdominal pain and menorrhalgia. In addition, it is widely used as a dietary supplement to provide carboxylic acids, essential oils and flavonoids. Evodiamine (EVO) is one of the major bioactive components contained within Evodiae fructus and is considered to be a potential candidate anti-cancer agent. EVO has been reported to exert anti-cancer effects by inhibiting cell proliferation, invasion and metastasis, whilst inducing apoptosis in numerous types of cancer cells. However, EVO is susceptible to metabolism and may inhibit the activities of metabolizing enzymes, such as cytochrome P450. Clinical application of EVO in the treatment of cancers may prove difficult due to poor bioavailability and potential toxicity due to metabolism. Currently, novel drug carriers involving the use of solid dispersion techniques, phospholipids and nanocomplexes to deliver EVO to improve its bioavailability and mitigate side effects have been tested. The present review aims to summarize the reported anti-cancer effects of EVO whilst discussing the pharmacokinetic behaviors, characteristics and effective delivery systems of EVO.
Collapse
Affiliation(s)
- Chaodan Luo
- Subtropical Agricultural Products Processing Engineering Technology Center, Guangxi Institute of Subtropical Agricultural Products Processing, Guangxi Subtropical Crops Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, Guangxi 530001, P.R. China
| | - Jingwen Ai
- Subtropical Agricultural Products Processing Engineering Technology Center, Guangxi Institute of Subtropical Agricultural Products Processing, Guangxi Subtropical Crops Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, Guangxi 530001, P.R. China
| | - Erfang Ren
- Subtropical Agricultural Products Processing Engineering Technology Center, Guangxi Institute of Subtropical Agricultural Products Processing, Guangxi Subtropical Crops Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, Guangxi 530001, P.R. China
| | - Jianqiang Li
- Subtropical Agricultural Products Processing Engineering Technology Center, Guangxi Institute of Subtropical Agricultural Products Processing, Guangxi Subtropical Crops Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, Guangxi 530001, P.R. China
| | - Chunmei Feng
- Subtropical Agricultural Products Processing Engineering Technology Center, Guangxi Institute of Subtropical Agricultural Products Processing, Guangxi Subtropical Crops Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, Guangxi 530001, P.R. China
| | - Xinrong Li
- Subtropical Agricultural Products Processing Engineering Technology Center, Guangxi Institute of Subtropical Agricultural Products Processing, Guangxi Subtropical Crops Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, Guangxi 530001, P.R. China
| | - Xiaojie Luo
- Subtropical Agricultural Products Processing Engineering Technology Center, Guangxi Institute of Subtropical Agricultural Products Processing, Guangxi Subtropical Crops Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, Guangxi 530001, P.R. China
| |
Collapse
|
12
|
O'Donnell EF, Jang HS, Liefwalker DF, Kerkvliet NI, Kolluri SK. Discovery and Mechanistic Characterization of a Select Modulator of AhR-regulated Transcription (SMAhRT) with Anti-cancer Effects. Apoptosis 2021; 26:307-322. [PMID: 33893898 DOI: 10.1007/s10495-021-01666-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2021] [Indexed: 02/07/2023]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor and a member of the bHLH/PAS (basic Helix-Loop-Helix/Per-Arnt-Sim) family of proteins. The AhR was cloned and characterized for its role in mediating the toxicity of dioxins. Subsequent research has identified the role of AhR in suppression of cancer cell growth. We hypothesized that the AhR is a molecular target for therapeutic intervention in cancer, and that activation of the AhR by unique AhR ligands in cancer cells could have anti-cancer effects including induction of cell death. This study describes the discovery and characterization of a new class of anti-cancer agents targeting the AhR, that we designate as Select Modulators of AhR-regulated Transcription (SMAhRTs). We employed two independent small molecule screening approaches to identify potential SMAhRTs. We report the identification of CGS-15943 that activates AhR signaling and induces apoptosis in an AhR-dependent manner in liver and breast cancer cells. Investigation of the downstream signaling pathway of this newly identified SMAhRT revealed upregulation of Fas-ligand (FasL), which is required for AhR-mediated apoptosis. Our results provide a basis for further development of a new class of anti-cancer therapeutics targeting an underappreciated molecular target, the AhR.
Collapse
Affiliation(s)
- Edmond Francis O'Donnell
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, 97331, USA
- Department of Orthopaedic Surgery, University of California Davis Medical Center, Davis, CA, USA
| | - Hyo Sang Jang
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, 97331, USA
| | - Daniel F Liefwalker
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, 97331, USA
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Nancy I Kerkvliet
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, 97331, USA
- The Pacific Northwest Center for Translational Environmental Health Research, Oregon State University, Corvallis, OR, 97331, USA
| | - Siva Kumar Kolluri
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, 97331, USA.
- Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331, USA.
- Center for Genome Research and Biocomputing, Oregon State University, Corvallis, OR, 97331, USA.
- The Pacific Northwest Center for Translational Environmental Health Research, Oregon State University, Corvallis, OR, 97331, USA.
| |
Collapse
|
13
|
A New Insight into the Potential Role of Tryptophan-Derived AhR Ligands in Skin Physiological and Pathological Processes. Int J Mol Sci 2021; 22:ijms22031104. [PMID: 33499346 PMCID: PMC7865493 DOI: 10.3390/ijms22031104] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 12/31/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) plays a crucial role in environmental responses and xenobiotic metabolism, as it controls the transcription profiles of several genes in a ligand-specific and cell-type-specific manner. Various barrier tissues, including skin, display the expression of AhR. Recent studies revealed multiple roles of AhR in skin physiology and disease, including melanogenesis, inflammation and cancer. Tryptophan metabolites are distinguished among the groups of natural and synthetic AhR ligands, and these include kynurenine, kynurenic acid and 6-formylindolo[3,2-b]carbazole (FICZ). Tryptophan derivatives can affect and regulate a variety of signaling pathways. Thus, the interest in how these substances influence physiological and pathological processes in the skin is expanding rapidly. The widespread presence of these substances and potential continuous exposure of the skin to their biological effects indicate the important role of AhR and its ligands in the prevention, pathogenesis and progression of skin diseases. In this review, we summarize the current knowledge of AhR in skin physiology. Moreover, we discuss the role of AhR in skin pathological processes, including inflammatory skin diseases, pigmentation disorders and cancer. Finally, the impact of FICZ, kynurenic acid, and kynurenine on physiological and pathological processes in the skin is considered. However, the mechanisms of how AhR regulates skin function require further investigation.
Collapse
|
14
|
AhR and Cancer: From Gene Profiling to Targeted Therapy. Int J Mol Sci 2021; 22:ijms22020752. [PMID: 33451095 PMCID: PMC7828536 DOI: 10.3390/ijms22020752] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/04/2021] [Accepted: 01/08/2021] [Indexed: 02/08/2023] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that has been shown to be an essential regulator of a broad spectrum of biological activities required for maintaining the body’s vital functions. AhR also plays a critical role in tumorigenesis. Its role in cancer is complex, encompassing both pro- and anti-tumorigenic activities. Its level of expression and activity are specific to each tumor and patient, increasing the difficulty of understanding the activating or inhibiting roles of AhR ligands. We explored the role of AhR in tumor cell lines and patients using genomic data sets and discuss the extent to which AhR can be considered as a therapeutic target.
Collapse
|
15
|
Cortés H, Reyes-Hernández OD, Alcalá-Alcalá S, Bernal-Chávez SA, Caballero-Florán IH, González-Torres M, Sharifi-Rad J, González-Del Carmen M, Figueroa-González G, Leyva-Gómez G. Repurposing of Drug Candidates for Treatment of Skin Cancer. Front Oncol 2021; 10:605714. [PMID: 33489912 PMCID: PMC7821387 DOI: 10.3389/fonc.2020.605714] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 11/27/2020] [Indexed: 12/24/2022] Open
Abstract
Skin cancers are highly prevalent malignancies that affect millions of people worldwide. These include melanomas and nonmelanoma skin cancers. Melanomas are among the most dangerous cancers, while nonmelanoma skin cancers generally exhibit a more benign clinical pattern; however, they may sometimes be aggressive and metastatic. Melanomas typically appear in body regions exposed to the sun, although they may also appear in areas that do not usually get sun exposure. Thus, their development is multifactorial, comprising endogenous and exogenous risk factors. The management of skin cancer depends on the type; it is usually based on surgery, chemotherapy, immunotherapy, and targeted therapy. In this respect, oncological treatments have demonstrated some progress in the last years; however, current therapies still present various disadvantages such as little cell specificity, recurrent relapses, high toxicity, and increased costs. Furthermore, the pursuit of novel medications is expensive, and the authorization for their clinical utilization may take 10-15 years. Thus, repositioning of drugs previously approved and utilized for other diseases has emerged as an excellent alternative. In this mini-review, we aimed to provide an updated overview of drugs' repurposing to treat skin cancer and discuss future perspectives.
Collapse
Affiliation(s)
- Hernán Cortés
- Laboratorio de Medicina Genómica, Departamento de Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de México, Mexico
| | - Octavio D. Reyes-Hernández
- Laboratorio de Biología Molecular del Cáncer, UMIEZ, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Sergio Alcalá-Alcalá
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Sergio A. Bernal-Chávez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Isaac H. Caballero-Florán
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Maykel González-Torres
- CONACyT-Laboratorio de Biotecnología, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de México, Mexico
| | - Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Facultad de Medicina, Universidad del Azuay, Cuenca, Ecuador
| | | | - Gabriela Figueroa-González
- Laboratorio de Farmacogenética, UMIEZ, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
16
|
Phan T, Nguyen VH, Buettner R, Morales C, Yang L, Wong P, Tsai W, Salazar MD, Gil Z, Diamond DJ, Rabinowitz JD, Rosen S, Melstrom LG. Inhibition of de novo pyrimidine synthesis augments Gemcitabine induced growth inhibition in an immunocompetent model of pancreatic cancer. Int J Biol Sci 2021; 17:2240-2251. [PMID: 34239352 PMCID: PMC8241727 DOI: 10.7150/ijbs.60473] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/18/2021] [Indexed: 12/13/2022] Open
Abstract
Leflunomide (Lef) is an agent used in autoimmune disorders that interferes with DNA synthesis. De Novo pyrimidine synthesis is a mechanism of Gemcitabine (Gem) resistance in pancreatic cancer. This study aims to assess the efficacy and changes in the tumor microenvironment of Lef monotherapy and in combination with Gem, in a syngeneic mouse model of pancreatic cancer. Methods: MTS proliferation assays were conducted to assess growth inhibition by Gem (0-20 nM), Lef (0-40 uM) and Gem+Lef in KPC (KrasLSL.G12D/+;p53R172H/+; PdxCretg/+) cells in vitro. An in vivo heterotopic KPC model was used and cohorts were treated with: PBS (control), Gem (75 mg/kg/q3d), Lef (40 mg/kg/d), or Gem+Lef. At d28 post-treatment, tumor burden, proliferation index (Ki67), and vascularity (CD31) were measured. Changes in the frequency of peripheral and intratumoral immune cell subsets were evaluated via FACS. Liquid chromatography-mass spectrometry was used for metabolomics profiling. Results: Lef inhibits KPC cell growth and synergizes with Gem in vitro (P<0.05; Combination Index 0.44 (<1 indicates synergy). In vivo, Lef alone and in combination with Gem delays KPC tumor progression (P<0.001). CTLA-4+T cells are also significantly decreased in tumors treated with Lef, Gem or in combination (Gem+Lef) compared to controls (P<0.05). Combination therapy also decreased the Ki67 and vascularity (P<0.01). Leflunomide inhibits de novo pyrimidine synthesis both in vitro (p<0.0001) and in vivo (p<0.05). Conclusions: In this study, we demonstrated that Gem+Lef inhibits pancreatic cancer growth, decrease T cell exhaustion, vascularity and as proof of principle inhibits de novo pyrimidine synthesis. Further characterization of changes in adaptive immunity are necessary to characterize the mechanism of tumor growth inhibition and facilitate translation to a clinical trial.
Collapse
Affiliation(s)
- Thuy Phan
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010
| | - Vu H. Nguyen
- Department of Hematology, City of Hope National Medical Center, Duarte, CA 91010
| | - Ralf Buettner
- Department of Hematology, City of Hope National Medical Center, Duarte, CA 91010
| | - Corey Morales
- Department of Hematology, City of Hope National Medical Center, Duarte, CA 91010
| | - Lifeng Yang
- Lewis Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University, Princeton, NJ 08544
| | - Paul Wong
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010
| | - Weiman Tsai
- Department of Hematology, City of Hope National Medical Center, Duarte, CA 91010
| | | | - Ziv Gil
- Rambam Medical Center, Israel
| | - Don J Diamond
- Department of Hematology, City of Hope National Medical Center, Duarte, CA 91010
| | - Joshua D. Rabinowitz
- Lewis Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University, Princeton, NJ 08544
| | - Steven Rosen
- Department of Hematology, City of Hope National Medical Center, Duarte, CA 91010
| | - Laleh G. Melstrom
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010
- ✉ Corresponding author: Laleh Melstrom MD, City of Hope National Medical Center, Department of Surgery and Immuno-oncology, 1500 E Duarte Road, Duarte, CA 91010. E-mail: ; Phone: 626 218 0282; Fax: 626 218 1113
| |
Collapse
|
17
|
Dwyer AR, Kerkvliet CP, Krutilina RI, Playa HC, Parke DN, Thomas WA, Smeester BA, Moriarity BS, Seagroves TN, Lange CA. Breast Tumor Kinase (Brk/PTK6) Mediates Advanced Cancer Phenotypes via SH2-Domain Dependent Activation of RhoA and Aryl Hydrocarbon Receptor (AhR) Signaling. Mol Cancer Res 2020; 19:329-345. [PMID: 33172975 DOI: 10.1158/1541-7786.mcr-20-0295] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 09/08/2020] [Accepted: 11/04/2020] [Indexed: 11/16/2022]
Abstract
Protein tyrosine kinase 6 (PTK6; also called Brk) is overexpressed in 86% of patients with breast cancer; high PTK6 expression predicts poor outcome. We reported PTK6 induction by HIF/GR complexes in response to either cellular or host stress. However, PTK6-driven signaling events in the context of triple-negative breast cancer (TNBC) remain undefined. In a mouse model of TNBC, manipulation of PTK6 levels (i.e., via knock-out or add-back) had little effect on primary tumor volume, but altered lung metastasis. To delineate the mechanisms of PTK6 downstream signaling, we created kinase-dead (KM) and kinase-intact domain structure mutants of PTK6 via in-frame deletions of the N-terminal SH3 or SH2 domains. While the PTK6 kinase domain contributed to soft-agar colony formation, PTK6 kinase activity was entirely dispensable for cell migration. Specifically, TNBC models expressing a PTK6 variant lacking the SH2 domain (SH2-del PTK6) were unresponsive to growth factor-stimulated cell motility relative to SH3-del, KM, or wild-type PTK6 controls. Reverse-phase protein array revealed that while intact PTK6 mediates spheroid formation via p38 MAPK signaling, the SH2 domain of PTK6 limits this biology, and instead mediates TNBC cell motility via activation of the RhoA and/or AhR signaling pathways. Inhibition of RhoA and/or AhR blocked TNBC cell migration as well as the branching/invasive morphology of PTK6+/AhR+ primary breast tumor tissue organoids. Inhibition of RhoA also enhanced paclitaxel cytotoxicity in TNBC cells, including in a taxane-refractory TNBC model. IMPLICATIONS: The SH2-domain of PTK6 is a potent effector of advanced cancer phenotypes in TNBC via RhoA and AhR, identified herein as novel therapeutic targets in PTK6+ breast tumors.
Collapse
Affiliation(s)
- Amy R Dwyer
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | | | - Raisa I Krutilina
- Department of Pathology and Laboratory Medicine and Center for Cancer Research, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Hilaire C Playa
- Department of Pathology and Laboratory Medicine and Center for Cancer Research, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Deanna N Parke
- Department of Pathology and Laboratory Medicine and Center for Cancer Research, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Warner A Thomas
- Department of Pathology and Laboratory Medicine and Center for Cancer Research, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | | | | | - Tiffany N Seagroves
- Department of Pathology and Laboratory Medicine and Center for Cancer Research, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee.
| | - Carol A Lange
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.
- Departments of Medicine (Division of Hematology, Oncology, and Transplantation) and Pharmacology, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
18
|
Jin UH, Michelhaugh SK, Polin LA, Shrestha R, Mittal S, Safe S. Omeprazole Inhibits Glioblastoma Cell Invasion and Tumor Growth. Cancers (Basel) 2020; 12:E2097. [PMID: 32731514 PMCID: PMC7465678 DOI: 10.3390/cancers12082097] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 02/06/2023] Open
Abstract
Background: The aryl hydrocarbon receptor (AhR) is expressed in gliomas and the highest staining is observed in glioblastomas. A recent study showed that the AhR exhibited tumor suppressor-like activity in established and patient-derived glioblastoma cells and genomic analysis showed that this was due, in part, to suppression of CXCL12, CXCR4 and MMP9. Methods: Selective AhR modulators (SAhRMs) including AhR-active pharmaceuticals were screened for their inhibition of invasion using a spheroid invasion assay in patient-derived AhR-expressing 15-037 glioblastoma cells and in AhR-silenced 15-037 cells. Invasion, migration and cell proliferation were determined using spheroid invasion, Boyden chambers and scratch assay, and XTT metabolic assays for cell growth. Changes in gene and gene product expression were determined by real-time PCR and Western blot assays, respectively. In vivo antitumorigenic activity of omeprazole was determined in SCID mice bearing subcutaneous patient-derived 15-037 cells. Results: Results of a screening assay using patient-derived 15-037 cells (wild-type and AhR knockout) identified the AhR-active proton pump inhibitor omeprazole as an inhibitor of glioblastoma cell invasion and migration only AhR-expressing cells but not in cells where the AhR was downregulated. Omeprazole also enhanced AhR-dependent repression of the pro-invasion CXCL12, CXCR4 and MMP9 genes, and interactions and effectiveness of omeprazole plus temozolomide were response-dependent. Omeprazole (100 mg/kg/injection) inhibited and delayed tumors in SCID mice bearing patient-derived 15-037 cells injected subcutaneously. Conclusion: Our results demonstrate that omeprazole enhances AhR-dependent inhibition of glioblastoma invasion and highlights a potential new avenue for development of a novel therapeutic mechanism-based approach for treating glioblastoma.
Collapse
Affiliation(s)
- Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA;
| | - Sharon K. Michelhaugh
- Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, VA 24014, USA; (S.K.M.); (S.M.)
| | - Lisa A. Polin
- Department of Oncology, Wayne State University and Karmanos Cancer Institute, Detroit, MI 48201, USA;
| | - Rupesh Shrestha
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA;
| | - Sandeep Mittal
- Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, VA 24014, USA; (S.K.M.); (S.M.)
- Carilion Clinic-Neurosurgery, Roanoke, VA 24014, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA;
| |
Collapse
|
19
|
Zhu P, Zhou K, Lu S, Bai Y, Qi R, Zhang S. Modulation of aryl hydrocarbon receptor inhibits esophageal squamous cell carcinoma progression by repressing COX2/PGE2/STAT3 axis. J Cell Commun Signal 2020; 14:175-192. [PMID: 31925646 DOI: 10.1007/s12079-019-00535-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/30/2019] [Indexed: 12/11/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most common malignant tumors with poor prognosis. Aryl hydrocarbon receptor (AHR) is a ligand-dependent transcription factor and emerging evidence shows it is associated with tumor initiation and promotion. However, the relationship between AHR and ESCC is not clear and it is meaningful to explore whether AHR could be a therapeutic target. In the present study, immunohistochemistry was performed to determine AHR expression levels in ESCC tissues. Knockdown of AHR expression in ESCC cell lines genetically and modulation of AHR by 3, 3'-diindolylmethane (DIM) pharmacologically both in vitro and in vivo were utilized to examine the corresponding alterations in cell growth, migration and invasion. Our study indicated that AHR expression levels were elevated in ESCC and associated with poor prognosis. Both knockdown and modulation of AHR inhibited tumor progression through down-regulating expression levels of PCNA, Bcl-2, Cyclin D1, MMP1, MMP2, MMP9 and up-regulating expression levels of Bax, Cleaved-Caspase 3. Our findings also indicated that repressing COX2/PGE2/STAT3 axis exerted inhibitory effects on ESCC both in vitro and in vivo assays. Taken together, AHR plays the key role in ESCC progression and targeting AHR as a therapeutic strategy with DIM is deserved for further exploration.
Collapse
Affiliation(s)
- Peiyao Zhu
- Department of Thoracic Surgery, The First Hospital of China Medical University, No.155 North Nanjing Street, Shenyang, 110001, China
| | - Kun Zhou
- Department of Thoracic Surgery, The First Hospital of China Medical University, No.155 North Nanjing Street, Shenyang, 110001, China
| | - Shilong Lu
- Department of Otolaryngology, Dermatology, Pathology, University of Colorado Anschutz Medical Campus, 12700 E 19th Avenue, Aurora, CO, 80045, USA
| | - Yu Bai
- Department of Thoracic Surgery, The First Hospital of China Medical University, No.155 North Nanjing Street, Shenyang, 110001, China
| | - Ruiqun Qi
- Department of Dermatology, The First Hospital of China Medical University, No.155 North Nanjing Street, Shenyang, 110001, China.
| | - Shuguang Zhang
- Department of Thoracic Surgery, The First Hospital of China Medical University, No.155 North Nanjing Street, Shenyang, 110001, China.
| |
Collapse
|
20
|
Role of the Aryl Hydrocarbon Receptor in Environmentally Induced Skin Aging and Skin Carcinogenesis. Int J Mol Sci 2019; 20:ijms20236005. [PMID: 31795255 PMCID: PMC6928879 DOI: 10.3390/ijms20236005] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/22/2019] [Accepted: 11/27/2019] [Indexed: 12/19/2022] Open
Abstract
The skin is constantly exposed to a variety of environmental threats, including solar electromagnetic radiation, microbes, airborne particulate matter, and chemicals. Acute exposure to these environmental factors results in the activation of different signaling pathways that orchestrate adaptive stress responses to maintain cell and tissue homeostasis. Chronic exposure of skin to these factors, however, may lead to the accumulation of damaged macromolecules and loss of cell and tissue integrity, which, over time, may facilitate aging processes and the development of aging-related malignancies. One transcription factor that is expressed in all cutaneous cells and activated by various environmental stressors, including dioxins, polycyclic aromatic hydrocarbons, and ultraviolet radiation, is the aryl hydrocarbon receptor (AHR). By regulating keratinocyte proliferation and differentiation, epidermal barrier function, melanogenesis, and immunity, a certain degree of AHR activity is critical to maintain skin integrity and to adapt to acute stress situations. In contrast, a chronic activation of cutaneous AHR signaling critically contributes to premature aging and the development of neoplasms by affecting metabolism, extracellular matrix remodeling, inflammation, pigmentation, DNA repair, and apoptosis. This article provides an overview of the detrimental effects associated with sustained AHR activity in chronically stressed skin and pinpoints AHR as a promising target for chemoprevention.
Collapse
|
21
|
Hidaka T, Fujimura T, Aiba S. Aryl Hydrocarbon Receptor Modulates Carcinogenesis and Maintenance of Skin Cancers. Front Med (Lausanne) 2019; 6:194. [PMID: 31552251 PMCID: PMC6736988 DOI: 10.3389/fmed.2019.00194] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 08/20/2019] [Indexed: 12/20/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that responds to a wide range of chemicals, including chemical carcinogens such as dioxins and carcinogenic polyaromatic hydrocarbons, and induces a battery of genes associated with detoxification, proliferation, and immune regulation. Recent reports suggest that AHR plays an important role in carcinogenesis and maintenance of various types of skin cancers. Indeed, AHR is a susceptibility gene for squamous cell carcinoma and a prognostic factor for melanoma and Merkel cell carcinoma. In addition, the carcinogenic effects of ultraviolet (UV) and chemical carcinogens, both of which are major environmental carcinogenetic factors of skin, are at least partly mediated by AHR, which regulates UV-induced inflammation and apoptosis, the DNA repair system, and metabolic activation of chemical carcinogens. Furthermore, AHR modulates the efficacy of key therapeutic agents in melanoma. AHR activation induces the expression of resistance genes against the inhibitors of V600E mutated B-Raf proto-oncogene, serine/threonine kinase (BRAF) in melanoma and upregulation of programmed cell death protein 1 (PD-1) in tumor-infiltrating T cells surrounding melanoma. Taken together, these findings underscore the importance of AHR in the biology of skin cancers. Development of therapeutic agents that modulate AHR activity is a promising strategy to advance chemoprevention and chemotherapy for skin cancers.
Collapse
Affiliation(s)
- Takanori Hidaka
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Taku Fujimura
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Setsuya Aiba
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
22
|
Buettner R, Morales C, Wu X, Sanchez JF, Li H, Melstrom LG, Rosen ST. Leflunomide Synergizes with Gemcitabine in Growth Inhibition of PC Cells and Impairs c-Myc Signaling through PIM Kinase Targeting. MOLECULAR THERAPY-ONCOLYTICS 2019; 14:149-158. [PMID: 31211245 PMCID: PMC6562366 DOI: 10.1016/j.omto.2019.04.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/30/2019] [Indexed: 02/07/2023]
Abstract
The immunosuppressive agent leflunomide has been used in the treatment of over 300,000 patients with rheumatoid arthritis. Its active metabolite, teriflunomide (Ter), directly inhibits dihydroorotate dehydrogenase (DHODH), an enzyme involved in nucleoside synthesis. We report that Ter not only shows in vitro anti-proliferative activity in pancreatic cancer (PC) cells as a single agent but also synergizes with the chemotherapeutic gemcitabine (Gem) in growth inhibition of PC cells. The growth-inhibitory effects of Ter are not solely caused by inhibition of DHODH. Through a kinase screening approach, we identified the PIM-3 serine-threonine kinase as a novel direct target. Subsequent dose-response kinase assays showed that Ter directly inhibited all three PIM family members, with the highest activities against PIM-3 and -1. The PIM-3 kinase was the PIM family member most often associated with PC oncogenesis and was also the kinase inhibited the most by Ter among more than 600 kinases investigated. Ter in PC cells induced changes in phosphorylation and expression of PIM downstream targets, consistent with the effects achieved by overexpression or downregulation of PIM-3. Finally, pharmacological inhibition of PIM proteins not only diminished PC cell proliferation, but also small-molecule pan-PIM and PIM-3 inhibitors synergized with Gem in growth inhibition of PC cells.
Collapse
Affiliation(s)
- Ralf Buettner
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Corey Morales
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Xiwei Wu
- Integrative Genomics Core, Department of Molecular Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - James F Sanchez
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Hongzhi Li
- Department of Computational Therapeutics, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Laleh G Melstrom
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Steven T Rosen
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
23
|
Vorontsova JE, Cherezov RO, Kuzin BA, Simonova OB. Aryl-Hydrocarbon Receptor as a Potential Target for Anticancer Therapy. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES B: BIOMEDICAL CHEMISTRY 2019. [DOI: 10.1134/s1990750819010116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
24
|
Bian Y, Li Y, Shrestha G, Wen X, Cai B, Wang K, Wan X. ITE, an endogenous aryl hydrocarbon receptor ligand, suppresses endometrial cancer cell proliferation and migration. Toxicology 2019; 421:1-8. [PMID: 30953668 DOI: 10.1016/j.tox.2019.03.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 02/16/2019] [Accepted: 03/29/2019] [Indexed: 01/16/2023]
Abstract
BACKGROUND Identification of new molecular targets for the treatment of endometrial cancer (EC) is an important clinical goal, especially for the patients which were resistant to conventional therapies. The aryl hydrocarbon receptor (AhR) is a ligand- activated transcription factor known primarily as the mediator of dioxin toxicity. However, the AhR can also inhibit cellular proliferation in a ligand-dependent manner and act as a tumor suppressor in mice, thus may be a potential anticancer target. In this study, we investigated if the endogenous AhR ligand 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE) regulated proliferation and migration of EC cells via AhR. METHODS We used quantitative real-time PCR and western blot to assess the expression of AhR in EC tissues and paired adjacent normal tissues. In addition, we conducted transwell assay to test whether the treatment of ITE altered the locomotive potential and proliferation of EC cells. Next, we conducted mouse xenograft models to further explore the in vivo effect of ITE. RESULTS We found that the AhR protein and RNA levels were increased mildly in EC tissues relative to the para-tumor normal endometrial tissues. Besides, ITE suppressed EC cells proliferation and migration in vitro, and also suppressed EC cells xenograft growth in mice. CONCLUSIONS Our results strongly supported the possibility of using the ITE as a small molecular compound for the treatment of EC.
Collapse
Affiliation(s)
- Yiding Bian
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Yiran Li
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Garima Shrestha
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Xiaoli Wen
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Bailian Cai
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Kai Wang
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, PR China.
| | - Xiaoping Wan
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, PR China.
| |
Collapse
|
25
|
Ladds MJGW, Laín S. Small molecule activators of the p53 response. J Mol Cell Biol 2019; 11:245-254. [PMID: 30689917 PMCID: PMC6478124 DOI: 10.1093/jmcb/mjz006] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/21/2018] [Accepted: 01/18/2019] [Indexed: 01/10/2023] Open
Abstract
Drugging the p53 pathway has been a goal for both academics and pharmaceutical companies since the designation of p53 as the 'guardian of the genome'. Through growing understanding of p53 biology, we can see multiple routes for activation of both wild-type p53 function and restoration of mutant p53. In this review, we focus on small molecules that activate wild-type p53 and that do so in a non-genotoxic manner. In particular, we will describe potential approaches to targeting proteins that alter p53 stability and function through posttranslational modification, affect p53's subcellular localization, or target RNA synthesis or the synthesis of ribonucleotides. The plethora of pathways for exploitation of p53, as well as the wide-ranging response to p53 activation, makes it an attractive target for anti-cancer therapy.
Collapse
Affiliation(s)
- Marcus J G W Ladds
- Department of Microbiology, Tumor and Cell Biology, Biomedicum, Solnavägen 9, Karolinska Institutet, Stockholm, Sweden
- SciLifeLab, Tomtebodavägen 23A, Solna, Stockholm, Sweden
| | - Sonia Laín
- Department of Microbiology, Tumor and Cell Biology, Biomedicum, Solnavägen 9, Karolinska Institutet, Stockholm, Sweden
- SciLifeLab, Tomtebodavägen 23A, Solna, Stockholm, Sweden
| |
Collapse
|
26
|
Corre S, Tardif N, Mouchet N, Leclair HM, Boussemart L, Gautron A, Bachelot L, Perrot A, Soshilov A, Rogiers A, Rambow F, Dumontet E, Tarte K, Bessede A, Guillemin GJ, Marine JC, Denison MS, Gilot D, Galibert MD. Sustained activation of the Aryl hydrocarbon Receptor transcription factor promotes resistance to BRAF-inhibitors in melanoma. Nat Commun 2018; 9:4775. [PMID: 30429474 PMCID: PMC6235830 DOI: 10.1038/s41467-018-06951-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 09/12/2018] [Indexed: 12/20/2022] Open
Abstract
BRAF inhibitors target the BRAF-V600E/K mutated kinase, the driver mutation found in 50% of cutaneous melanoma. They give unprecedented anti-tumor responses but acquisition of resistance ultimately limits their clinical benefit. The master regulators driving the expression of resistance-genes remain poorly understood. Here, we demonstrate that the Aryl hydrocarbon Receptor (AhR) transcription factor is constitutively activated in a subset of melanoma cells, promoting the dedifferentiation of melanoma cells and the expression of BRAFi-resistance genes. Typically, under BRAFi pressure, death of BRAFi-sensitive cells leads to an enrichment of a small subpopulation of AhR-activated and BRAFi-persister cells, responsible for relapse. Also, differentiated and BRAFi-sensitive cells can be redirected towards an AhR-dependent resistant program using AhR agonists. We thus identify Resveratrol, a clinically compatible AhR-antagonist that abrogates deleterious AhR sustained-activation. Combined with BRAFi, Resveratrol reduces the number of BRAFi-resistant cells and delays tumor growth. We thus propose AhR-impairment as a strategy to overcome melanoma resistance.
Collapse
Affiliation(s)
- Sébastien Corre
- IGDR (Institut de Génétique et Développement de Rennes)-UMR6290, CNRS, Univ Rennes, F-35000, Rennes, France.
| | - Nina Tardif
- IGDR (Institut de Génétique et Développement de Rennes)-UMR6290, CNRS, Univ Rennes, F-35000, Rennes, France
| | - Nicolas Mouchet
- IGDR (Institut de Génétique et Développement de Rennes)-UMR6290, CNRS, Univ Rennes, F-35000, Rennes, France
| | - Héloïse M Leclair
- IGDR (Institut de Génétique et Développement de Rennes)-UMR6290, CNRS, Univ Rennes, F-35000, Rennes, France
| | - Lise Boussemart
- IGDR (Institut de Génétique et Développement de Rennes)-UMR6290, CNRS, Univ Rennes, F-35000, Rennes, France.,Department of Dermatology, Hospital University of Rennes (CHU Rennes), F-35000, Rennes, France
| | - Arthur Gautron
- IGDR (Institut de Génétique et Développement de Rennes)-UMR6290, CNRS, Univ Rennes, F-35000, Rennes, France
| | - Laura Bachelot
- IGDR (Institut de Génétique et Développement de Rennes)-UMR6290, CNRS, Univ Rennes, F-35000, Rennes, France
| | - Anthony Perrot
- IGDR (Institut de Génétique et Développement de Rennes)-UMR6290, CNRS, Univ Rennes, F-35000, Rennes, France
| | - Anatoly Soshilov
- Department of Environmental Toxicology, University of California, Meyer Hall, Davis, CA, 95616, USA
| | - Aljosja Rogiers
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, VIB, Leuven, 3000, Belgium.,Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, Leuven, 3000, Belgium
| | - Florian Rambow
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, VIB, Leuven, 3000, Belgium.,Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, Leuven, 3000, Belgium
| | - Erwan Dumontet
- MICMAC (MIcroenvironment, Cell differentiation, iMmunology And Cancer)-UMR_S 1236, Inserm, Univ Rennes, F-35000, Rennes, France
| | - Karin Tarte
- MICMAC (MIcroenvironment, Cell differentiation, iMmunology And Cancer)-UMR_S 1236, Inserm, Univ Rennes, F-35000, Rennes, France
| | | | - Gilles J Guillemin
- Neuroinflammation Group, MND and Neurodegenerative Diseases Research Center, Macquarie University, Sydney, NSW, 2109, Australia
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, VIB, Leuven, 3000, Belgium.,Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, Leuven, 3000, Belgium
| | - Michael S Denison
- Department of Environmental Toxicology, University of California, Meyer Hall, Davis, CA, 95616, USA
| | - David Gilot
- IGDR (Institut de Génétique et Développement de Rennes)-UMR6290, CNRS, Univ Rennes, F-35000, Rennes, France.
| | - Marie-Dominique Galibert
- IGDR (Institut de Génétique et Développement de Rennes)-UMR6290, CNRS, Univ Rennes, F-35000, Rennes, France. .,Department of Molecular Genetics and Genomics, Hospital University of Rennes (CHU Rennes), F-35000, Rennes, France.
| |
Collapse
|
27
|
Vorontsova JE, Cherezov RO, Kuzin BA, Simonova OB. [Aryl-hydrocarbon receptor as a potential target for anticancer therapy]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2018; 64:397-415. [PMID: 30378556 DOI: 10.18097/pbmc20186405397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Aryl-hydrocarbon receptor (Aryl Hydrocarbon Receptor, AHR) is a ligand-dependent transcription factor, whose functions are related to xenobiotic detoxification, response to inflammation, and maintenance of tissue homeostasis. Recent investigations suggest that AHR also plays an important role in the processes of carcinogenesis. Increased expression of AHR is observed in several types of tumors and tumor cell lines. In addition, it turned out that the composition of pharmaceutical drugs used in oncotherapy includes some ligands AHR. These facts allow us to consider an aryl-hydrocarbon receptor as a potential target for anticancer therapy, especially for the treatment of severe cancers whose treatment options are very limited or do not exist at all. In this review the examples of AHR ligands' effect on tumor cell cultures and on model mice lines with AHR-dependent response are discussed.
Collapse
Affiliation(s)
- J E Vorontsova
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, Russia
| | - R O Cherezov
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, Russia
| | - B A Kuzin
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, Russia
| | - O B Simonova
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
28
|
O'Donnell EF, Jang HS, Pearce M, Kerkvliet NI, Kolluri SK. The aryl hydrocarbon receptor is required for induction of p21cip1/waf1 expression and growth inhibition by SU5416 in hepatoma cells. Oncotarget 2018; 8:25211-25225. [PMID: 28424418 PMCID: PMC5421923 DOI: 10.18632/oncotarget.16056] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 02/06/2017] [Indexed: 11/25/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a potential clinical target for cancer and autoimmune dysfunction. Identifying selective AhR modulators that produce desirable clinical outcomes represents an opportunity for developing new anti-cancer agents. Repurposing clinically-used drugs with established safety profiles that activate the AhR represents a good starting place to pursue this goal. In this study, we characterized the AhR-dependent effects of SU5416 (Semaxanib) following its identification in a small-molecule library screen. SU5416 potently activated AhR-dependent reporter genes, induced AhR nuclear localization, facilitated AhR-DNA binding, and increased, expression of its endogenous target genes. SU5416 significantly inhibited proliferation of Hepa1 hepatoma cells in an AhR-dependent manner, but did not induce apoptosis. SU5416 also inhibited the growth of human HepG2 liver cancer cells. The effects of SU5416 correlated with an increased G1 population and increased expression of cell cycle inhibitor p21cip1/waf1 at both the mRNA and protein level. Increased expression of p21cip1/waf1 by SU5416 required expression of both AhR and Arnt. In addition, evidence for long-term activation of the AhR in vivo by a single dose of SU5416 was identified by analyzing published microarray data. Our results provide support for continued investigation of the AhR as therapeutic for cancers such as hepatocellular carcinoma. In addition, our findings raise the possibility that some of the previously observed anti-proliferative effects of SU5416 may be due to activation of the AhR.
Collapse
Affiliation(s)
- Edmond F O'Donnell
- Cancer Research Laboratory, Oregon State University, Corvallis, Oregon, USA.,Department of Environmental and Molecular Toxicology, and Environmental Health Sciences Center, Oregon State University, Corvallis, Oregon, USA
| | - Hyo Sang Jang
- Cancer Research Laboratory, Oregon State University, Corvallis, Oregon, USA.,Department of Environmental and Molecular Toxicology, and Environmental Health Sciences Center, Oregon State University, Corvallis, Oregon, USA
| | - Martin Pearce
- Cancer Research Laboratory, Oregon State University, Corvallis, Oregon, USA.,Department of Environmental and Molecular Toxicology, and Environmental Health Sciences Center, Oregon State University, Corvallis, Oregon, USA
| | - Nancy I Kerkvliet
- Department of Environmental and Molecular Toxicology, and Environmental Health Sciences Center, Oregon State University, Corvallis, Oregon, USA
| | - Siva Kumar Kolluri
- Cancer Research Laboratory, Oregon State University, Corvallis, Oregon, USA.,Department of Environmental and Molecular Toxicology, and Environmental Health Sciences Center, Oregon State University, Corvallis, Oregon, USA
| |
Collapse
|
29
|
Chen Y, Huang Q, Zhou H, Wang Y, Hu X, Li T. Inhibition of canonical WNT/β-catenin signaling is involved in leflunomide (LEF)-mediated cytotoxic effects on renal carcinoma cells. Oncotarget 2018; 7:50401-50416. [PMID: 27391060 PMCID: PMC5226591 DOI: 10.18632/oncotarget.10409] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 06/13/2016] [Indexed: 11/25/2022] Open
Abstract
Leflunomide (LEF), an inhibitor of dihydroorotate dehydrogenase (DHODH) in pyrimidine biosynthetic pathway, is an immunomodulatory agent approved for the treatment of rheumatoid arthritis. In this study, we show that LEF significantly reduced cell proliferation of renal carcinoma cells in a concentration-dependent manner. LEF at 50 μM induced S-phase arrest and autophagy. Higher doses of LEF (>50 μM) effectively induced cell apoptosis. Modulating the concentration of LEF resulted in distinct effects on the expression of regulatory proteins associated with cell cycle, apoptosis, and autophagy. In particular, high concentrations of LEF inhibited canonical WNT signaling by promoting nucleo-cytoplasmic shuttling and proteasome-dependent degradation of β-catenin. Mechanistic studies showed that the repression of AKT activation partly accounted for LEF-mediated WNT inhibition. Gene expression microarray revealed that LEF treatment greatly inhibited the expression of FZD10 gene, a receptor mediating WNT/β-catenin activation. In vivo xenograft study in NOD/SCID mice further validated the inhibitory effects of LEF on tumor growth and Wnt/β-catenin signaling. However, LEF treatment also triggered cell autophagy and elevated the expression of WNT3a, which ameliorated its cytotoxic effects. The combination of LEF with a WNT inhibitor IWP-2 or autophagy inhibitor HCQ could yield an enhanced anti-tumor outcome. Taken together, these results identify the potential utility and pharmacological feature of LEF in the chemotherapy of renal cell carcinoma (RCC).
Collapse
Affiliation(s)
- Yicheng Chen
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Qiaoli Huang
- Department of Biology, College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang 321004, China
| | - Hua Zhou
- Department of Biology, College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang 321004, China
| | - Yueping Wang
- Department of Urology, Wuyi First People's Hospital, Wuyi, Zhejiang 321200, China
| | - Xian Hu
- Department of Plastic Surgery, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Tao Li
- Department of Biology, College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang 321004, China
| |
Collapse
|
30
|
Identification of New Activators of Mitochondrial Fusion Reveals a Link between Mitochondrial Morphology and Pyrimidine Metabolism. Cell Chem Biol 2017; 25:268-278.e4. [PMID: 29290623 DOI: 10.1016/j.chembiol.2017.12.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 09/12/2017] [Accepted: 11/30/2017] [Indexed: 01/26/2023]
Abstract
Mitochondria are dynamic organelles that produce most of the cellular ATP, and are involved in many other cellular functions such as Ca2+ signaling, differentiation, apoptosis, cell cycle, and cell growth. One key process of mitochondrial dynamics is mitochondrial fusion, which is catalyzed by mitofusins (MFN1 and MFN2) and OPA1. The outer mitochondrial membrane protein MFN2 plays a relevant role in the maintenance of mitochondrial metabolism, insulin signaling, and mutations that cause neurodegenerative disorders. Therefore, modulation of proteins involved in mitochondrial dynamics has emerged as a potential pharmacological strategy. Here, we report the identification of small molecules by high-throughput screen that promote mitochondrial elongation in an MFN1/MFN2-dependent manner. Detailed analysis of their mode of action reveals a previously unknown connection between pyrimidine metabolism and mitochondrial dynamics. Our data indicate a link between pyrimidine biosynthesis and mitochondrial dynamics, which maintains cell survival under stress conditions characterized by loss of pyrimidine synthesis.
Collapse
|
31
|
Jang HS, Pearce M, O'Donnell EF, Nguyen BD, Truong L, Mueller MJ, Bisson WH, Kerkvliet NI, Tanguay RL, Kolluri SK. Identification of a Raloxifene Analog That Promotes AhR-Mediated Apoptosis in Cancer Cells. BIOLOGY 2017; 6:biology6040041. [PMID: 29194351 PMCID: PMC5745446 DOI: 10.3390/biology6040041] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/14/2017] [Accepted: 11/22/2017] [Indexed: 12/11/2022]
Abstract
We previously reported that raloxifene, an estrogen receptor modulator, is also a ligand for the aryl hydrocarbon receptor (AhR). Raloxifene induces apoptosis in estrogen receptor-negative human cancer cells through the AhR. We performed structure–activity studies with seven raloxifene analogs to better understand the structural requirements of raloxifene for induction of AhR-mediated transcriptional activity and apoptosis. We identified Y134 as a raloxifene analog that activates AhR-mediated transcriptional activity and induces apoptosis in MDA-MB-231 human triple negative breast cancer cells. Suppression of AhR expression strongly reduced apoptosis induced by Y134, indicating the requirement of AhR for Y134-induced apoptosis. Y134 also induced apoptosis in hepatoma cells without having an effect on cell cycle regulation. Toxicity testing on zebrafish embryos revealed that Y134 has a significantly better safety profile than raloxifene. Our studies also identified an analog of raloxifene that acts as a partial antagonist of the AhR, and is capable of inhibiting AhR agonist-induced transcriptional activity. We conclude that Y134 is a promising raloxifene analog for further optimization as an anti-cancer agent targeting the AhR.
Collapse
Affiliation(s)
- Hyo Sang Jang
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA.
| | - Martin Pearce
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA.
| | - Edmond F O'Donnell
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA.
| | - Bach Duc Nguyen
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA.
| | - Lisa Truong
- Department of Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA.
| | - Monica J Mueller
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA.
| | - William H Bisson
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA.
| | - Nancy I Kerkvliet
- Department of Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA.
| | - Robert L Tanguay
- Department of Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA.
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA.
- Center for Genome Research and Biocomputing, Oregon State University, Corvallis, OR 97331, USA.
| | - Siva Kumar Kolluri
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA.
- Department of Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA.
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA.
- Center for Genome Research and Biocomputing, Oregon State University, Corvallis, OR 97331, USA.
| |
Collapse
|
32
|
Carbidopa is an activator of aryl hydrocarbon receptor with potential for cancer therapy. Biochem J 2017; 474:3391-3402. [DOI: 10.1042/bcj20170583] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 08/25/2017] [Accepted: 08/30/2017] [Indexed: 02/08/2023]
Abstract
Carbidopa is used with l-DOPA (l-3,4-dihydroxyphenylalanine) to treat Parkinson's disease (PD). PD patients exhibit lower incidence of most cancers including pancreatic cancer, but with the notable exception of melanoma. The decreased cancer incidence is not due to l-DOPA; however, the relevance of Carbidopa to this phenomenon has not been investigated. Here, we tested the hypothesis that Carbidopa, independent of l-DOPA, might elicit an anticancer effect. Carbidopa inhibited pancreatic cancer cell proliferation both in vitro and in vivo. Based on structural similarity with phenylhydrazine, an inhibitor of indoleamine-2,3-dioxygenase-1 (IDO1), we predicted that Carbidopa might also inhibit IDO1, thus providing a molecular basis for its anticancer effect. The inhibitory effect was confirmed using human recombinant IDO1. To demonstrate the inhibition in intact cells, AhR (aryl hydrocarbon receptor) activity was monitored as readout for IDO1-mediated generation of the endogenous AhR agonist kynurenine in pancreatic and liver cancer cells. Surprisingly, Carbidopa did not inhibit but instead potentiated AhR signaling, evident from increased CYP1A1 (cytochrome P450 family 1 subfamily A member 1), CYP1A2, and CYP1B1 expression. In pancreatic and liver cancer cells, Carbidopa promoted AhR nuclear localization. AhR antagonists blocked Carbidopa-dependent activation of AhR signaling. The inhibitory effect on pancreatic cancer cells in vitro and in vivo and the activation of AhR occurred at therapeutic concentrations of Carbidopa. Chromatin immunoprecipitation assay further confirmed that Carbidopa promoted AhR binding to its target gene CYP1A1 leading to its induction. We conclude that Carbidopa is an AhR agonist and suppresses pancreatic cancer. Hence, Carbidopa could potentially be re-purposed to treat pancreatic cancer and possibly other cancers as well.
Collapse
|
33
|
Dorasamy MS, Choudhary B, Nellore K, Subramanya H, Wong PF. Dihydroorotate dehydrogenase Inhibitors Target c-Myc and Arrest Melanoma, Myeloma and Lymphoma cells at S-phase. J Cancer 2017; 8:3086-3098. [PMID: 28928900 PMCID: PMC5604460 DOI: 10.7150/jca.14835] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 03/05/2017] [Indexed: 02/04/2023] Open
Abstract
Dihydroorotate dehydrogenase (DHODH) is a rate-limiting enzyme in the de novo biosynthesis pathway of pyrimidines. Inhibition of this enzyme impedes cancer cell proliferation but the exact mechanisms of action of these inhibitors in cancer cells are poorly understood. In this study, we showed that cancer cells, namely melanoma, myeloma and lymphoma overexpressed DHODH protein and treatment with A771726 and Brequinar sodium resulted in cell cycle arrest at S-phase. Transfection with DHODH shRNA depleted DHODH protein expression and impeded the proliferation of melanoma cells. shRNA knockdown of DHODH in combination with DHODH inhibitors further reduced the cancer cell proliferation, suggesting that knockdown of DHODH had sensitized the cells to DHODH inhibitors. Cell cycle regulatory proteins, c-Myc and its transcriptional target, p21 were found down- and up-regulated, respectively, following treatment with DHODH inhibitors in melanoma, myeloma and lymphoma cells. Interestingly, knockdown of DHODH by shRNA had also similarly affected the expression of c-Myc and p21 proteins. Our findings suggest that DHODH inhibitors induce cell cycle arrest in cancer cells via additional DHODH-independent pathway that is associated with p21 up-regulation and c-Myc down-regulation. Hence, DHODH inhibitors can be explored as potential therapeutic agents in cancer therapy.
Collapse
Affiliation(s)
- Mathura Subangari Dorasamy
- Department of Pharmacology, Faculty of Medicine, 50603 University of Malaya, Kuala Lumpur, Malaysia.,Aurigene Discovery Technologies Limited, Bollaram Road, Miyapur Hyderabad - 500049, Telangana, India
| | - Bhavesh Choudhary
- Aurigene Discovery Technologies Limited, Bollaram Road, Miyapur Hyderabad - 500049, Telangana, India
| | - Kavitha Nellore
- Aurigene Discovery Technologies Limited, 39-40, KIADB Industrial Area, Phase II Electronic City, Hosur Road, Bangalore - 560100 Karnataka, India
| | - Hosahalli Subramanya
- Aurigene Discovery Technologies Limited, 39-40, KIADB Industrial Area, Phase II Electronic City, Hosur Road, Bangalore - 560100 Karnataka, India
| | - Pooi-Fong Wong
- Department of Pharmacology, Faculty of Medicine, 50603 University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
34
|
Liu L, Dong Z, Lei Q, Yang J, Hu H, Li Q, Ji Y, Guo L, Zhang Y, Liu Y, Cui H. Inactivation/deficiency of DHODH induces cell cycle arrest and programed cell death in melanoma. Oncotarget 2017; 8:112354-112370. [PMID: 29348830 PMCID: PMC5762515 DOI: 10.18632/oncotarget.19379] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 07/11/2017] [Indexed: 12/21/2022] Open
Abstract
Malignant melanoma (MM) is one of the most malignant tumors and has a very poor prognosis. However, there are no effective drugs to treat this disease. As a kind of iron flavin dependent enzyme, dihydroorotate dehydrogenase (DHODH, EC 1.3.3.1) is the fourth and a key enzyme in the de novo biosynthesis of pyrimidines. Herein, we found that DHODH inactivation/deficiency inhibited melanoma cell proliferation, induced cell cycle arrest at S phase and lead to autophagy in human melanoma cells. Meanwhile, leflunomide treatment induced cell apoptosis and deficiency of DHODH sensitized cells to drug-induced apoptosis in BCL-2 deficient melanoma cells, while not in BCL-2 abundant melanoma cells. Then we found that BCL-2 could rescue apoptosis induced by DHODH inactivation/deficiency. Moreover, BCL-2 also showed to promote cell cycle arrest and to inhibit autophagy induced by leflunomide. To explore the mechanisms underlying autophagy induced by DHODH inhibition, we found that AMPK-Ulk1 axis was activated in this process. Besides, JNK was phosphorylated and activated to phosphorylate BCL-2, which abrogated the interaction between BCL-2 and Beclin1 and then abolished autophagy. Our findings provided evidences for the potential of DHODH used as a drug target for melanoma treatment.
Collapse
Affiliation(s)
- Lichao Liu
- Department of Dermatology, The Third Hospital of Hebei Medical University, Shijiazhuang, 050000, China.,State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, China
| | - Zhen Dong
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, China
| | - Qian Lei
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, China
| | - Jie Yang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, China
| | - Huanrong Hu
- Department of Dermatology, The Third Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Qian Li
- Department of Dermatology, The Third Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Yacong Ji
- Department of Dermatology, The Third Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Leiyang Guo
- Department of Dermatology, The Third Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Yanli Zhang
- Department of Dermatology, The Third Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Yaling Liu
- Department of Dermatology, The Third Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, China
| |
Collapse
|
35
|
Abstract
Cancer is a major health issue worldwide, and the global burden of cancer is expected to increase in the coming years. Whereas the limited success with current therapies has driven huge investments into drug development, the average number of FDA approvals per year has declined since the 1990s. This unmet need for more effective anti-cancer drugs has sparked a growing interest for drug repurposing, i.e. using drugs already approved for other indications to treat cancer. As such, data both from pre-clinical experiments, clinical trials and observational studies have demonstrated anti-tumor efficacy for compounds within a wide range of drug classes other than cancer. Whereas some of them induce cancer cell death or suppress various aspects of cancer cell behavior in established tumors, others may prevent cancer development. Here, we provide an overview of promising candidates for drug repurposing in cancer, as well as studies describing the biological mechanisms underlying their anti-neoplastic effects.
Collapse
Affiliation(s)
- Linda Sleire
- Oncomatrix Research Group, Department of Biomedicine, University of Bergen, Jonas Lies vei 91 5009 Bergen, Norway
| | - Hilde Elise Førde
- Oncomatrix Research Group, Department of Biomedicine, University of Bergen, Jonas Lies vei 91 5009 Bergen, Norway
| | - Inger Anne Netland
- Oncomatrix Research Group, Department of Biomedicine, University of Bergen, Jonas Lies vei 91 5009 Bergen, Norway
| | - Lina Leiss
- Oncomatrix Research Group, Department of Biomedicine, University of Bergen, Jonas Lies vei 91 5009 Bergen, Norway
| | - Bente Sandvei Skeie
- Oncomatrix Research Group, Department of Biomedicine, University of Bergen, Jonas Lies vei 91 5009 Bergen, Norway; Department of Neurosurgery, Haukeland University Hospital, Jonas Lies vei, 71, 5021 Bergen, Norway
| | - Per Øyvind Enger
- Oncomatrix Research Group, Department of Biomedicine, University of Bergen, Jonas Lies vei 91 5009 Bergen, Norway; Department of Neurosurgery, Haukeland University Hospital, Jonas Lies vei, 71, 5021 Bergen, Norway.
| |
Collapse
|
36
|
Formosa R, Vassallo J. The Complex Biology of the Aryl Hydrocarbon Receptor and Its Role in the Pituitary Gland. Discov Oncol 2017. [PMID: 28634910 DOI: 10.1007/s12672-017-0300-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor best known for its ability to mediate the effects of environmental toxins such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD or dioxin), polycyclic aromatic hydrocarbons (PAHs), benzene, and polychlorinated biphenyls (PCBs) through the initiation of transcription of a number of metabolically active enzymes. Therefore, the AHR has been studied mostly in the context of xenobiotic signaling. However, several studies have shown that the AHR is constitutively active and plays an important role in general cell physiology, independently of its activity as a xenobiotic receptor and in the absence of exogenous ligands. Within the pituitary, activation of the AHR by environmental toxins has been implicated in disruption of gonadal development and fertility. Studies carried out predominantly in mouse models have revealed the detrimental influence of several environmental toxins on specific cell lineages of the pituitary tissue mediated by activation of AHR and its downstream effectors. Activation of AHR during fetal development adversely affected pituitary development while adult models exposed to AHR ligands demonstrated varying degrees of pituitary dysfunction. Such dysfunction may arise as a result of direct effects on pituitary cells or indirect effects on the hypothalamic-pituitary-gonadal axis. This review offers in-depth analysis of all aspects of AHR biology, with a particular focus on its role and activity within the adenohypophysis and specifically in pituitary tumorigenesis. A novel mechanism by which the AHR may play a direct role in pituitary cell proliferation and tumor formation is postulated. This review therefore attempts to cover all aspects of the AHR's role in the pituitary tissue, from fetal development to adult physiology and the pathophysiology underlying endocrine disruption and pituitary tumorigenesis.
Collapse
Affiliation(s)
- Robert Formosa
- Department of Medicine, Faculty of Medicine and Surgery, University of Malta, MSD 2080, Msida, Malta
| | - Josanne Vassallo
- Department of Medicine, Faculty of Medicine and Surgery, University of Malta, MSD 2080, Msida, Malta. .,Neuroendocrine Clinic, Department of Medicine, Mater Dei Hospital, Msida, Malta.
| |
Collapse
|
37
|
Role of the aryl hydrocarbon receptor in carcinogenesis and potential as an anti-cancer drug target. Arch Toxicol 2017; 91:2497-2513. [PMID: 28508231 DOI: 10.1007/s00204-017-1981-2] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 05/08/2017] [Indexed: 12/31/2022]
Abstract
The aryl hydrocarbon receptor (AhR) was initially identified as the receptor that binds and mediates the toxic effects induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and structurally related halogenated aromatics. Other toxic compounds including some polynuclear aromatic hydrocarbons act through the AhR; however, during the last 25 years, it has become apparent that the AhR plays an essential role in maintaining cellular homeostasis. Moreover, the scope of ligands that bind the AhR includes endogenous compounds such as multiple tryptophan metabolites, other endogenous biochemicals, pharmaceuticals and health-promoting phytochemicals including flavonoids, indole-3-carbinol and its metabolites. It has also been shown that like other receptors, the AhR is a drug target for multiple diseases including cancer, where both AhR agonists and antagonists effectively block many of the critical hallmarks of cancer in multiple tumor types. This review describes the anti-cancer activities of AhR ligands and demonstrates that it is time to separate the AhR from TCDD and exploit the potential of the AhR as a novel target for cancer chemotherapy.
Collapse
|
38
|
Zeng F, Qi T, Li C, Li T, Li H, Li S, Zhu L, Xu X. Synthesis, structure-activity relationship and binding mode analysis of 4-thiazolidinone derivatives as novel inhibitors of human dihydroorotate dehydrogenase. MEDCHEMCOMM 2017; 8:1297-1302. [PMID: 30108840 DOI: 10.1039/c7md00081b] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Accepted: 04/17/2017] [Indexed: 01/21/2023]
Abstract
A series of 4-thiazolidinone derivatives were synthesized and evaluated as novel human dihydroorotate dehydrogenase (hDHODH) inhibitors. Compounds 26 and 31 displayed IC50 values of 1.75 and 1.12 μM, respectively. The structure-activity relationship was summarized. Further binding mode analysis revealed that compound 31 could form a hydrogen bond with Tyr38 and a water-mediated hydrogen bond with Ala55, which may be necessary for maintaining the bioactivities of the compounds in this series. Further structural optimization of the para- or meta-position of the phenyl group at R will lead to the identification of more potent hDHODH inhibitors.
Collapse
Affiliation(s)
- Fanxun Zeng
- Shanghai Key Laboratory of Chemical Biology , School of Pharmacy , East China University of Science and Technology , 130 Meilong Road , Shanghai 200237 , China . ; ; ; Fax: +86 21 64252603 ; Tel: +86 21 64250213 ; Tel: +86 21 64252945
| | - Tiantian Qi
- Shanghai Key Laboratory of New Drug Design , State Key Laboratory of Bioreactor Engineering , School of Pharmacy , East China University of Science & Technology , Shanghai 200237 , China . ; ; Tel: +86 21 64253379
| | - Chunyan Li
- Shanghai Key Laboratory of Chemical Biology , School of Pharmacy , East China University of Science and Technology , 130 Meilong Road , Shanghai 200237 , China . ; ; ; Fax: +86 21 64252603 ; Tel: +86 21 64250213 ; Tel: +86 21 64252945
| | - Tingfang Li
- Shanghai Key Laboratory of Chemical Biology , School of Pharmacy , East China University of Science and Technology , 130 Meilong Road , Shanghai 200237 , China . ; ; ; Fax: +86 21 64252603 ; Tel: +86 21 64250213 ; Tel: +86 21 64252945
| | - Honglin Li
- Shanghai Key Laboratory of New Drug Design , State Key Laboratory of Bioreactor Engineering , School of Pharmacy , East China University of Science & Technology , Shanghai 200237 , China . ; ; Tel: +86 21 64253379
| | - Shiliang Li
- Shanghai Key Laboratory of Chemical Biology , School of Pharmacy , East China University of Science and Technology , 130 Meilong Road , Shanghai 200237 , China . ; ; ; Fax: +86 21 64252603 ; Tel: +86 21 64250213 ; Tel: +86 21 64252945.,Shanghai Key Laboratory of New Drug Design , State Key Laboratory of Bioreactor Engineering , School of Pharmacy , East China University of Science & Technology , Shanghai 200237 , China . ; ; Tel: +86 21 64253379
| | - Lili Zhu
- Shanghai Key Laboratory of New Drug Design , State Key Laboratory of Bioreactor Engineering , School of Pharmacy , East China University of Science & Technology , Shanghai 200237 , China . ; ; Tel: +86 21 64253379
| | - Xiaoyong Xu
- Shanghai Key Laboratory of Chemical Biology , School of Pharmacy , East China University of Science and Technology , 130 Meilong Road , Shanghai 200237 , China . ; ; ; Fax: +86 21 64252603 ; Tel: +86 21 64250213 ; Tel: +86 21 64252945.,Shanghai Collaborative Innovation Center for Biomanufacturing Technology , 130 Meilong Road , Shanghai 200237 , China
| |
Collapse
|
39
|
Shrestha AK, Patel A, Menon RT, Jiang W, Wang L, Moorthy B, Shivanna B. Leflunomide induces NAD(P)H quinone dehydrogenase 1 enzyme via the aryl hydrocarbon receptor in neonatal mice. Biochem Biophys Res Commun 2017; 485:195-200. [PMID: 28192119 DOI: 10.1016/j.bbrc.2017.02.051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 02/09/2017] [Indexed: 01/28/2023]
Abstract
Aryl hydrocarbon receptor (AhR) has been increasingly recognized to play a crucial role in normal physiological homeostasis. Additionally, disrupted AhR signaling leads to several pathological states in the lung and liver. AhR activation transcriptionally induces detoxifying enzymes such as cytochrome P450 (CYP) 1A and NAD(P)H quinone dehydrogenase 1 (NQO1). The toxicity profiles of the classical AhR ligands such as 3-methylcholanthrene and dioxins limit their use as a therapeutic agent in humans. Hence, there is a need to identify nontoxic AhR ligands to develop AhR as a clinically relevant druggable target. Recently, we demonstrated that leflunomide, a FDA approved drug, used to treat rheumatoid arthritis in humans, induces CYP1A enzymes in adult mice via the AhR. However, the mechanisms by which this drug induces NQO1 in vivo are unknown. Therefore, we tested the hypothesis that leflunomide will induce pulmonary and hepatic NQO1 enzyme in neonatal mice via AhR-dependent mechanism(s). Leflunomide elicited significant induction of pulmonary CYP1A1 and NQO1 expression in neonatal mice. Interestingly, the dose at which leflunomide increased NQO1 was significantly higher than that required to induce CYP1A1 enzyme. Likewise, it also enhanced hepatic CYP1A1, 1A2 and NQO1 expression in WT mice. In contrast, leflunomide failed to induce these enzymes in AhR-null mice. Our results indicate that leflunomide induces pulmonary and hepatic CYP1A and NQO1 enzymes via the AhR in neonatal mice. These findings have important implications to prevent and/or treat disorders such as bronchopulmonary dysplasia in human infants where AhR may play a crucial role in the disease pathogenesis.
Collapse
Affiliation(s)
- Amrit Kumar Shrestha
- Section of Neonatology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ananddeep Patel
- Section of Neonatology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Renuka T Menon
- Section of Neonatology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Weiwu Jiang
- Section of Neonatology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Lihua Wang
- Section of Neonatology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Bhagavatula Moorthy
- Section of Neonatology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Binoy Shivanna
- Section of Neonatology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
40
|
Ehrlich AK, Kerkvliet NI. Is chronic AhR activation by rapidly metabolized ligands safe for the treatment of immune-mediated diseases? CURRENT OPINION IN TOXICOLOGY 2017; 2:72-78. [PMID: 28944315 DOI: 10.1016/j.cotox.2017.01.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
There is a long standing perception that AhR ligands are automatically disqualified from pharmaceutical development due to their induction of Cyp1a1 as well as their potential for causing "dioxin-like" toxicities. However, recent discoveries of new AhR ligands with potential therapeutic applications have been reported, inviting reconsideration of this policy. One area of exploration is focused on the activation of AhR to promote the generation of regulatory T cells, which control the intensity and duration of immune responses. Rapidly metabolized AhR ligands (RMAhRLs), which do not bioaccumulate in the same manner as 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) have been discovered that induce Tregs and display impressive therapeutic efficacy in a broad range of preclinical models of immune-mediated diseases. Given the promise of these RMAhRLs, is the bias against AhR activators still valid? Can RMAhRLs be given chronically to maintain therapeutic levels of AhR activation without producing the same toxicity profile as dioxin-like compounds? Based on our review of the data, there is little evidence to support the indiscriminate exclusion of AhR activators/Cyp1a1 inducers from early drug developmental pipelines. We also found no evidence that short-term treatment with RMAhRLs produce "dioxin-like toxicity" and, in fact, were well tolerated. However, safety testing of individual RMAhRLs under therapeutic conditions, as performed with all promising new drugs, will be needed to reveal whether or not chronic activation of AhR leads to unacceptable adverse outcomes.
Collapse
Affiliation(s)
- Allison K Ehrlich
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, 97331, United States
| | - Nancy I Kerkvliet
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, 97331, United States
| |
Collapse
|
41
|
Safe S, Cheng Y, Jin UH. The Aryl Hydrocarbon Receptor (AhR) as a Drug Target for Cancer Chemotherapy. CURRENT OPINION IN TOXICOLOGY 2017; 2:24-29. [PMID: 28459113 DOI: 10.1016/j.cotox.2017.01.012] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The aryl hydrocarbon receptor (AhR) is overexpressed in some patients with different tumor types, and the receptor can be a negative or positive prognostic factor. There is also evidence from both in vivo and in vitro cell culture models that the AhR can exhibit tumor-specific pro-oncogenic and tumor suppressor-like functions and therefore can be treated with AhR antagonists or agonists, respectively. Successful clinical applications of AhR ligands will require the synthesis and development of selective AhR modulators (SAhRMs) with tumor-specific AhR agonist or antagonist activity, and some currently available compounds such as indole-3-carbinol and diindolylmethane-(DIM) and synthetic AhR antagonists are potential drug candidates. There is also evidence that some AhR-active pharmaceuticals, including tranilast, flutamide, hydroxytamoxifen and omeprazole or their derivatives, may be effective AhR-dependent anticancer agents for single or combination cancer chemotherapies for treatment of breast and pancreatic cancers.
Collapse
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX 77843
| | - Yating Cheng
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX 77843
| | - Un-Ho Jin
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX 77843
| |
Collapse
|
42
|
Ma LL, Wu ZT, Wang L, Zhang XF, Wang J, Chen C, Ni X, Lin YF, Cao YY, Luan Y, Pan GY. Inhibition of hepatic cytochrome P450 enzymes and sodium/bile acid cotransporter exacerbates leflunomide-induced hepatotoxicity. Acta Pharmacol Sin 2016; 37:415-24. [PMID: 26806301 DOI: 10.1038/aps.2015.157] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 11/03/2015] [Indexed: 11/09/2022] Open
Abstract
AIM Leflunomide is an immunosuppressive agent marketed as a disease-modifying antirheumatic drug. But it causes severe side effects, including fatal hepatitis and liver failure. In this study we investigated the contributions of hepatic metabolism and transport of leflunomide and its major metabolite teriflunomide to leflunomide induced hepatotoxicity in vitro and in vivo. METHODS The metabolism and toxicity of leflunomide and teriflunomide were evaluated in primary rat hepatocytes in vitro. Hepatic cytochrome P450 reductase null (HRN) mice were used to examine the PK profiling and hepatotoxicity of leflunomide in vivo. The expression and function of sodium/bile acid cotransporter (NTCP) were assessed in rat and human hepatocytes and NTCP-transfected HEK293 cells. After Male Sprague-Dawley (SD) rats were administered teriflunomide (1,6, 12 mg · kg(-1) · d(-1), ig) for 4 weeks, their blood samples were analyzed. RESULTS A nonspecific CYPs inhibitor aminobenzotriazole (ABT, 1 mmol/L) decreased the IC50 value of leflunomide in rat hepatocytes from 409 to 216 μmol/L, whereas another nonspecific CYPs inhibitor proadifen (SKF, 30 μmol/L) increased the cellular accumulation of leflunomide to 3.68-fold at 4 h. After oral dosing (15 mg/kg), the plasma exposure (AUC0-t) of leflunomide increased to 3-fold in HRN mice compared with wild type mice. Administration of leflunomide (25 mg·kg(-1) · d(-1)) for 7 d significantly increased serum ALT and AST levels in HRN mice; when the dose was increased to 50 mg·kg(-1) · d(-1), all HRN mice died on d 6. Teriflunomide significantly decreased the expression of NTCP in human hepatocytes, as well as the function of NTCP in rat hepatocytes and NTCP-transfected HEK293 cells. Four-week administration of teriflunomide significantly increased serum total bilirubin and direct bilirubin levels in female rats, but not in male rats. CONCLUSION Hepatic CYPs play a critical role in detoxification process of leflunomide, whereas the major metabolite teriflunomide suppresses the expression and function of NTCP, leading to potential cholestasis.
Collapse
|
43
|
Chemopreventive effect of leflunomide against Ehrlich's solid tumor grown in mice: Effect on EGF and EGFR expression and tumor proliferation. Life Sci 2015; 141:193-201. [DOI: 10.1016/j.lfs.2015.10.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 08/07/2015] [Accepted: 10/02/2015] [Indexed: 02/07/2023]
|
44
|
Patel A, Zhang S, Paramahamsa M, Jiang W, Wang L, Moorthy B, Shivanna B. Leflunomide Induces Pulmonary and Hepatic CYP1A Enzymes via Aryl Hydrocarbon Receptor. Drug Metab Dispos 2015; 43:1966-70. [PMID: 26417045 DOI: 10.1124/dmd.115.066084] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/25/2015] [Indexed: 11/22/2022] Open
Abstract
Emerging evidence indicates that the aryl hydrocarbon receptor (AhR) plays a crucial role in normal physiologic homeostasis. Additionally, aberrant AhR signaling leads to several pathologic states in the lung and liver. Activation of AhR transcriptionally induces phase I (CYP1A) detoxifying enzymes. Although the effects of the classic AhR ligands such as 3-methylcholanthrene and dioxins on phase 1 enzymes are well studied in rodent lung, liver, and other organs, the toxicity profiles limit their use as therapeutic agents in humans. Hence, there is a need to identify and investigate nontoxic AhR ligands not only to understand the AhR biology but also to develop the AhR as a clinically relevant therapeutic target. Leflunomide is a Food and Drug Administration-approved drug in humans that is known to have AhR agonist activity in vitro. Whether it activates AhR and induces phase 1 enzymes in vivo is unknown. Therefore, we tested the hypothesis that leflunomide will induce pulmonary and hepatic CYP1A enzymes in C57BL/6J wild-type mice, but not in AhR-null mice. We performed real-time reverse-transcription polymerase chain reaction analyses for CYP1A1/2 mRNA expression, western blot assays for CYP1A1/2 protein expression, and ethoxyresorufinO-deethylase assay for CYP1A1 catalytic activity. Leflunomide increased CYP1A1/A2 mRNA, protein, and enzymatic activities in wild-type mice. In contrast, leflunomide failed to increase pulmonary and hepatic CYP1A enzymes in AhR-null mice. In conclusion, we provide evidence that leflunomide induces pulmonary and hepatic CYP1A enzymes via the AhR.
Collapse
Affiliation(s)
- Ananddeep Patel
- Section of Neonatology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Shaojie Zhang
- Section of Neonatology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Maturu Paramahamsa
- Section of Neonatology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Weiwu Jiang
- Section of Neonatology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Lihua Wang
- Section of Neonatology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Bhagavatula Moorthy
- Section of Neonatology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Binoy Shivanna
- Section of Neonatology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
45
|
Ochieng J, Nangami GN, Ogunkua O, Miousse IR, Koturbash I, Odero-Marah V, McCawley LJ, Nangia-Makker P, Ahmed N, Luqmani Y, Chen Z, Papagerakis S, Wolf GT, Dong C, Zhou BP, Brown DG, Colacci AM, Hamid RA, Mondello C, Raju J, Ryan EP, Woodrick J, Scovassi AI, Singh N, Vaccari M, Roy R, Forte S, Memeo L, Salem HK, Amedei A, Al-Temaimi R, Al-Mulla F, Bisson WH, Eltom SE. The impact of low-dose carcinogens and environmental disruptors on tissue invasion and metastasis. Carcinogenesis 2015; 36 Suppl 1:S128-59. [PMID: 26106135 DOI: 10.1093/carcin/bgv034] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The purpose of this review is to stimulate new ideas regarding low-dose environmental mixtures and carcinogens and their potential to promote invasion and metastasis. Whereas a number of chapters in this review are devoted to the role of low-dose environmental mixtures and carcinogens in the promotion of invasion and metastasis in specific tumors such as breast and prostate, the overarching theme is the role of low-dose carcinogens in the progression of cancer stem cells. It is becoming clearer that cancer stem cells in a tumor are the ones that assume invasive properties and colonize distant organs. Therefore, low-dose contaminants that trigger epithelial-mesenchymal transition, for example, in these cells are of particular interest in this review. This we hope will lead to the collaboration between scientists who have dedicated their professional life to the study of carcinogens and those whose interests are exclusively in the arena of tissue invasion and metastasis.
Collapse
Affiliation(s)
- Josiah Ochieng
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA, Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA, Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA, Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA, Department of Pathology, Wayne State University, Detroit, MI 48201, USA, Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia, Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA, Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA, Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy, Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia, Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy, Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA, Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India, Mediterranean Institute of Oncology, Viagrande 95029, Italy, Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt, Department of Experimental and
| | - Gladys N Nangami
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA, Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA, Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA, Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA, Department of Pathology, Wayne State University, Detroit, MI 48201, USA, Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia, Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA, Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA, Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy, Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia, Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy, Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA, Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India, Mediterranean Institute of Oncology, Viagrande 95029, Italy, Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt, Department of Experimental and
| | - Olugbemiga Ogunkua
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA, Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA, Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA, Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA, Department of Pathology, Wayne State University, Detroit, MI 48201, USA, Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia, Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA, Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA, Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy, Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia, Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy, Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA, Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India, Mediterranean Institute of Oncology, Viagrande 95029, Italy, Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt, Department of Experimental and
| | - Isabelle R Miousse
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Valerie Odero-Marah
- Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
| | - Lisa J McCawley
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| | | | - Nuzhat Ahmed
- Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia
| | - Yunus Luqmani
- Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | - Zhenbang Chen
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA, Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA, Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA, Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA, Department of Pathology, Wayne State University, Detroit, MI 48201, USA, Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia, Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA, Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA, Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy, Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia, Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy, Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA, Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India, Mediterranean Institute of Oncology, Viagrande 95029, Italy, Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt, Department of Experimental and
| | - Silvana Papagerakis
- Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA
| | - Gregory T Wolf
- Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA
| | - Chenfang Dong
- Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Binhua P Zhou
- Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Dustin G Brown
- Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Anna Maria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Roslida A Hamid
- Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia
| | - Chiara Mondello
- Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy
| | - Jayadev Raju
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Jordan Woodrick
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - A Ivana Scovassi
- Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy
| | - Neetu Singh
- Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Rabindra Roy
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Stefano Forte
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Lorenzo Memeo
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Hosni K Salem
- Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze 50134, Italy and
| | - Rabeah Al-Temaimi
- Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | - Fahd Al-Mulla
- Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | - William H Bisson
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA
| | - Sakina E Eltom
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA, Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA, Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA, Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA, Department of Pathology, Wayne State University, Detroit, MI 48201, USA, Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia, Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA, Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA, Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy, Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia, Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy, Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA, Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India, Mediterranean Institute of Oncology, Viagrande 95029, Italy, Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt, Department of Experimental and
| |
Collapse
|
46
|
Contador-Troca M, Alvarez-Barrientos A, Merino JM, Morales-Hernández A, Rodríguez MI, Rey-Barroso J, Barrasa E, Cerezo-Guisado MI, Catalina-Fernández I, Sáenz-Santamaría J, Oliver FJ, Fernandez-Salguero PM. Dioxin receptor regulates aldehyde dehydrogenase to block melanoma tumorigenesis and metastasis. Mol Cancer 2015; 14:148. [PMID: 26242870 PMCID: PMC4524442 DOI: 10.1186/s12943-015-0419-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 07/22/2015] [Indexed: 01/16/2023] Open
Abstract
Background The dioxin (AhR) receptor can have oncogenic or tumor suppressor activities depending on the phenotype of the target cell. We have shown that AhR knockdown promotes melanoma primary tumorigenesis and lung metastasis in the mouse and that human metastatic melanomas had reduced AhR levels with respect to benign nevi. Methods Mouse melanoma B16F10 cells were engineered by retroviral transduction to stably downregulate AhR expression, Aldh1a1 expression or both. They were characterized for Aldh1a1 activity, stem cell markers and migration and invasion in vitro. Their tumorigenicity in vivo was analyzed using xenografts and lung metastasis assays as well as in vivo imaging. Results Depletion of aldehyde dehydrogenase 1a1 (Aldh1a1) impairs the pro-tumorigenic and pro-metastatic advantage of melanoma cells lacking AhR expression (sh-AhR). Thus, Aldh1a1 knockdown in sh-AhR cells (sh-AhR + sh-Aldh1a1) diminished their migration and invasion potentials and blocked tumor growth and metastasis to the lungs in immunocompetent AhR+/+ recipient mice. However, Aldh1a1 downmodulation in AhR-expressing B16F10 cells did not significantly affect tumor growth in vivo. Aldh1a1 knockdown reduced the high levels of CD133+/CD29+/CD44+ cells, melanosphere size and the expression of the pluripotency marker Sox2 in sh-AhR cells. Interestingly, Sox2 increased Aldh1a1 expression in sh-AhR but not in sh-AhR + sh-Aldh1a1 cells, suggesting that Aldh1a1 and Sox2 may be co-regulated in melanoma cells. In vivo imaging revealed that mice inoculated with AhR + Aldh1a1 knockdown cells had reduced tumor burden and enhanced survival than those receiving Aldh1a1-expressing sh-AhR cells. Conclusions Aldh1a1 overactivation in an AhR-deficient background enhances melanoma progression. Since AhR may antagonize the protumoral effects of Aldh1a1, the AhRlow-Aldh1a1high phenotype could be indicative of bad outcome in melanoma. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0419-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- María Contador-Troca
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, 06071, Badajoz, Spain.
| | | | - Jaime M Merino
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, 06071, Badajoz, Spain.
| | | | - María I Rodríguez
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, 18016, Granada, Spain.
| | - Javier Rey-Barroso
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, 06071, Badajoz, Spain.
| | - Eva Barrasa
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, 06071, Badajoz, Spain.
| | - María I Cerezo-Guisado
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, 06071, Badajoz, Spain.
| | | | - Javier Sáenz-Santamaría
- Servicio de Anatomía Patológica, Hospital Universitario Infanta Cristina, 06071, Badajoz, Spain.
| | - Francisco J Oliver
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, 18016, Granada, Spain.
| | | |
Collapse
|
47
|
Xie G, Raufman JP. Role of the Aryl Hydrocarbon Receptor in Colon Neoplasia. Cancers (Basel) 2015; 7:1436-46. [PMID: 26264025 PMCID: PMC4586780 DOI: 10.3390/cancers7030847] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 07/27/2015] [Accepted: 07/29/2015] [Indexed: 12/27/2022] Open
Abstract
For both men and women, colorectal cancer (CRC) is the second leading cause of cancer death in the United States, primarily as a consequence of limited therapies for metastatic disease. The aryl hydrocarbon receptor (AhR) is a ligand-dependent transcription factor with diverse functions in detoxification of xenobiotics, inflammatory responses, and tissue homeostasis. Emerging evidence indicates that AhR also plays an important role in regulating intestinal cell proliferation and tumorigenesis. Here, we review both the pro- and anti-carcinogenic properties of AhR signaling and its potential role as a therapeutic target in CRC.
Collapse
Affiliation(s)
- Guofeng Xie
- Division of Gastroenterology and Hepatology, Veterans Administration Maryland Health Care System, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Jean-Pierre Raufman
- Division of Gastroenterology and Hepatology, Veterans Administration Maryland Health Care System, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
48
|
Anti-androgen flutamide suppresses hepatocellular carcinoma cell proliferation via the aryl hydrocarbon receptor mediated induction of transforming growth factor-β1. Oncogene 2015; 34:6092-104. [PMID: 25867062 DOI: 10.1038/onc.2015.55] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 12/07/2014] [Accepted: 12/22/2014] [Indexed: 12/17/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor and a member of the basic helix-loop-helix PER/ARNT/SIM family of chemosensors and developmental regulators. The AhR is widely known as a mediator of dioxin toxicity; however, it also suppresses cancer cell proliferation and recent findings have implicated its role as a tumor suppressor. We conducted a chemical library screen to identify nontoxic AhR ligands with anti-cancer effects and discovered flutamide (Eulexin) as a putative AhR ligand. Flutamide is an androgen receptor (AR) antagonist approved by the United States Food and Drug Administration for the treatment of prostate cancer. We found that flutamide inhibited the growth of several cancer cell lines independent of AR status, and that suppression of AhR expression reversed the anti-proliferative effects of flutamide. We investigated the AhR-dependent mechanism of action of flutamide in human hepatocellular carcinoma cells and identified that transforming growth factor-β1 (TGF-β1) is induced by flutamide in an AhR-dependent manner. In contrast, the potent AhR agonist 2,3,7,8-Tetrachlorodibenzo-p-dioxin had no effect on TGF-β1 expression, indicating the ligand specificity of AhR activation. We also determined that TGF-β1 induction is required for the AhR-dependent growth inhibitory effects of flutamide. Therefore, flutamide may be effective in AhR-positive cancers that are sensitive to TGF-β1 signaling, such as hepatocellular carcinoma.
Collapse
|
49
|
Pund S, Pawar S, Gangurde S, Divate D. Transcutaneous delivery of leflunomide nanoemulgel: Mechanistic investigation into physicomechanical characteristics, in vitro anti-psoriatic and anti-melanoma activity. Int J Pharm 2015; 487:148-56. [PMID: 25869452 DOI: 10.1016/j.ijpharm.2015.04.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Revised: 04/08/2015] [Accepted: 04/09/2015] [Indexed: 11/18/2022]
Abstract
The present study is a mechanistic validation of 'proof of concept' of effective topical delivery of leflunomide (LFD) nanoemulgel for localized efficient treatment of psoriatic lesions as well as melanoma affected skin regions. Hyperproliferation of keratinocytes in psoriasis and symbiotic relationship between keratinocytes and melanocytes, justifies the need of dual acting treatment. LFD is recently introduced significantly effective disease modifying anti-rheumatic drug and has been considered valuable for the treatment of psoriatic arthritis as well as melanoma. Current available treatments for psoriasis and melanoma are inefficient due to systemic side effects, poor transcutaneous permeation and thus present a challenge for development of novel colloidal carriers. We newly reformulated LFD as a nanoemulgel based on self nanoemulsifying technique using Capryol 90, Cremophor EL, Transcutol HP as nanoemulsifying components and Pluronic F127 as a gelling agent. This thermodynamically stable nanoemuslsifying preconcentrate after gelation showed mean globule size, 123.7 nm and viscosity 9620 ± 93 cp. Complete mechanical characterization was carried out using Texture Analyzer and hardness, adhesiveness and springiness index were found to be 523 gms, 431 gms and 1.02, respectively. Ex vivo permeation through rat abdominal skin revealed significant improvement in flux, apparent permeability coefficient, steady state diffusion coefficient and drug deposition in skin due to nanoemulsification of LFD. The in vitro cytoxicity of LFD nanoemulgel in human HaCaT, melanoma A375 and SK-MEL-2 cell lines showed significantly enhanced therapeutic response. In gist, LFD nanoemulgel for trancutaneous delivery will reduce the overall dose and drug consumption, by effectively localizing at the applied target site and will ultimately minimize systemic side effects.
Collapse
Affiliation(s)
- Swati Pund
- Department of Pharmaceutics, STES's Sinhgad Institute of Pharmacy, Narhe, Pune 411041, India.
| | - Satish Pawar
- Department of Pharmaceutics, STES's Sinhgad Institute of Pharmacy, Narhe, Pune 411041, India
| | - Shashikant Gangurde
- Department of Pharmaceutics, STES's Sinhgad Institute of Pharmacy, Narhe, Pune 411041, India
| | - Deepali Divate
- Department of Pharmaceutics, STES's Sinhgad Institute of Pharmacy, Narhe, Pune 411041, India
| |
Collapse
|
50
|
Doscas ME, Williamson AJ, Usha L, Bogachkov Y, Rao GS, Xiao F, Wang Y, Ruby C, Kaufman H, Zhou J, Williams JW, Li Y, Xu X. Inhibition of p70 S6 kinase (S6K1) activity by A77 1726 and its effect on cell proliferation and cell cycle progress. Neoplasia 2014; 16:824-34. [PMID: 25379019 PMCID: PMC4212247 DOI: 10.1016/j.neo.2014.08.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 08/13/2014] [Accepted: 08/15/2014] [Indexed: 11/16/2022] Open
Abstract
Leflunomide is a novel immunomodulatory drug prescribed for treating rheumatoid arthritis. It inhibits the activity of protein tyrosine kinases and dihydroorotate dehydrogenase, a rate-limiting enzyme in the pyrimidine nucleotide synthesis pathway. Here, we report that A77 1726, the active metabolite of leflunomide, inhibited the phosphorylation of ribosomal protein S6 and two other substrates of S6K1, insulin receptor substrate-1 and carbamoyl phosphate synthetase 2, in an A375 melanoma cell line. A77 1726 increased the phosphorylation of AKT, p70 S6 (S6K1), ERK1/2, and MEK through the feedback activation of the IGF-1 receptor–mediated signaling pathway. Invitro kinase assay revealed that leflunomide and A77 1726 inhibited S6K1 activity with IC50 values of approximately 55 and 80 μM, respectively. Exogenous uridine partially blocked A77 1726–induced inhibition of A375 cell proliferation. S6K1 knockdown led to the inhibition of A375 cell proliferation but did not potentiate the antiproliferative effect of A77 1726. A77 1726 stimulated bromodeoxyuridine incorporation in A375 cells but arrested the cell cycle in the S phase, which was reversed by addition of exogenous uridine or by MAP kinase pathway inhibitors but not by rapamycin and LY294002 (a phosphoinositide 3-kinase inhibitor). These observations suggest that A77 1726 accelerates cell cycle entry into the S phase through MAP kinase activation and that pyrimidine nucleotide depletion halts the completion of the cell cycle. Our study identified a novel molecular target of A77 1726 and showed that the inhibition of S6K1 activity was in part responsible for its antiproliferative activity. Our study also provides a novel mechanistic insight into A77 1726–induced cell cycle arrest in the S phase.
Collapse
Affiliation(s)
- Michelle E Doscas
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL 60612
| | | | - Lydia Usha
- Section of Hematology/Oncology, Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612
| | - Yedida Bogachkov
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL 60612
| | - Geetha S Rao
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL 60612
| | - Fei Xiao
- Cinkate Corporation, Oak Park, IL 60302
| | - Yimin Wang
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL 60612
| | - Carl Ruby
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL 60612 ; Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612
| | - Howard Kaufman
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL 60612 ; Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612
| | - Jingsong Zhou
- Department of Molecular Biophysics and Physiology, Rush University Medical Center, Chicago, IL 60612
| | | | - Yi Li
- Baylor College of Medicine, Lester and Sue Smith Breast Center, Department of Molecular and Cell Biology, Houston, TX 77030
| | - Xiulong Xu
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL 60612 ; Department of General Surgery, Rush University Medical Center, Chicago, IL 60612
| |
Collapse
|