1
|
Das DN, Ravi N. Influences of polycyclic aromatic hydrocarbon on the epigenome toxicity and its applicability in human health risk assessment. ENVIRONMENTAL RESEARCH 2022; 213:113677. [PMID: 35714684 DOI: 10.1016/j.envres.2022.113677] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 06/15/2023]
Abstract
The existence of polycyclic aromatic hydrocarbons (PAHs) in ambient air is an escalating concern worldwide because of their ability to cause cancer and induce permanent changes in the genetic material. Growing evidence implies that during early life-sensitive stages, the risk of progression of acute and chronic diseases depends on epigenetic changes initiated by the influence of environmental cues. Several reports deciphered the relationship between exposure to environmental chemicals and epigenetics, and have known toxicants that alter the epigenetic states. Amongst PAHs, benzo[a]pyrene (B[a]P) is accepted as a group 1 cancer-causing agent by the International Agency for the Research on Cancer (IARC). B[a]P is a well-studied pro-carcinogen that is metabolically activated by the aryl hydrocarbon receptor (AhR)/cytochrome P450 pathway. Cytochrome P450 plays a pivotal role in the stimulation step, which is essential for DNA adduct formation. Accruing evidence suggests that epigenetic alterations assume a fundamental part in PAH-promoted carcinogenesis. This interaction between PAHs and epigenetic factors results in an altered profile of these marks, globally and locus-specific. Some of the epigenetic changes due to exposure to PAHs lead to increased disease susceptibility and progression. It is well understood that exposure to environmental carcinogens, such as PAH triggers disease pathways through changes in the genome. Several evidence reported due to the epigenome-wide association studies, that early life adverse environmental events may trigger widespread and persistent variations in transcriptional profiling. Moreover, these variations respond to DNA damage and/or a consequence of epigenetic modifications that need further investigation. Growing evidence has associated PAHs with epigenetic variations involving alterations in DNA methylation, histone modification, and micro RNA (miRNA) regulation. Epigenetic alterations to PAH exposure were related to chronic diseases, such as pulmonary disease, cardiovascular disease, endocrine disruptor, nervous system disorder, and cancer. This hormetic response gives a novel perception concerning the toxicity of PAHs and the biological reaction that may be a distinct reliance on exposure. This review sheds light on understanding the latest evidence about how PAHs can alter epigenetic patterns and human health. In conclusion, as several epigenetic change mechanisms remain unclear yet, further analyses derived from PAHs exposure must be performed to find new targets and disease biomarkers. In spite of the current limitations, numerous evidence supports the perception that epigenetics grips substantial potential for advancing our knowledge about the molecular mechanisms of environmental toxicants, also for predicting health-associated risks due to environmental circumstances exposure and individual susceptibility.
Collapse
Affiliation(s)
- Durgesh Nandini Das
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Nathan Ravi
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis, St. Louis, MO, 63110, USA; Department of Energy, Environmental & Chemical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA; Institute for Public Health, Washington University in St. Louis, St. Louis, MO, 63110, USA; Veterans Affairs St. Louis Hospital, St. Louis, MO, 63106, USA.
| |
Collapse
|
2
|
Zhang Q, He X, Yu Q, Liu Y, Zhu Y, Yang L, Shang P, Zhang J, Liu H, Feng F. Endoplasmic reticulum stress regulates pyroptosis in BPDE-induced BEAS-2B cells. ENVIRONMENTAL TOXICOLOGY 2022; 37:1768-1780. [PMID: 35297523 DOI: 10.1002/tox.23524] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/23/2022] [Accepted: 03/06/2022] [Indexed: 06/14/2023]
Abstract
Benzo(a)pyrene(B(a)P), as the main representative of polycyclic aromatic hydrocarbons, can promote inflammation and many chronic pulmonary diseases. However, the underlying mechanism of Benzo(a)pyrene-7,8-diol-9,10-epoxide (BPDE)-induced human bronchial epithelial cell pyroptosis related to endoplasmic reticulum stress (ERS) has not been elucidated. This study focused on the effects of BPDE on ERS and pyroptosis in human bronchial epithelial cells (BEAS-2B), and explored the relationship between ERS and pyroptosis. BEAS-2B cells were stimulated with 0.50, 0.75, and 1.00 μmol/L BPDE for 24 h to detect ERS and pyroptosis. After inhibition of ERS with 4-phenylbutyrate (4-PBA), pyroptosis of BEAS-2B cells was tested. The results showed that BPDE decreased the cell viability, changed the morphological structure of endoplasmic reticulum and increased the expression levels of GRP78 and p-PERK. After BPDE treatment, the cell membrane was damaged and incomplete under transmission electron microscope; Hoechst 33342/PI fluorescence staining showed that the number of PI-positive cells was enhanced. The expression levels of GSDMD-N, cleaved-caspase 1, and cleaved-IL-1β were elevated, and the expression levels of IL-1β, IL-18, and NLRP3 protein were improved. In BPDE combined with 4-PBA intervention group, the rate of PI-positive cells was reduced, the expression levels of GRP78, GSDMD-N, and cleaved-caspase 1 were decreased, and the expression levels of IL-1β, IL-18, and NLRP3 were decreased. In conclusion, BPDE could induce ERS and pyroptosis in BEAS-2B cells, and ERS may promote the occurrence of BPDE-induced pyroptosis.
Collapse
Affiliation(s)
- Qiao Zhang
- Department of Toxicology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Xi He
- Department of Toxicology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Qi Yu
- Department of Toxicology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Yitong Liu
- College of Public Health, University of Southern California, Los Angeles, California, USA
| | - Yonghang Zhu
- Department of Toxicology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Liu Yang
- Department of Toxicology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Pingping Shang
- Key Laboratory of Tobacco Chemistry, Zhengzhou Tobacco Research Institute, CNC, Zhengzhou, Henan, China
| | - Jiatong Zhang
- Department of Disease Control and Prevention, Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hong Liu
- Department of Pulmonary Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Feifei Feng
- Department of Toxicology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
3
|
Naproxen inhibits spontaneous lung adenocarcinoma formation in Kras G12V mice. Neoplasia 2021; 23:574-583. [PMID: 34091121 PMCID: PMC8187931 DOI: 10.1016/j.neo.2021.05.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 02/07/2023] Open
Abstract
Naproxen inhibits the adenocarcinoma by 64% in KrasG12V mice. Naproxen inhibits serum PGE2/CXCR4 levels in KrasG12V mice. Naproxen inhibits the progression of adenocarcinoma in KrasG12V mice.
Lung cancer is the leading cause of cancer related deaths worldwide. The present study investigated the effects of naproxen (NSAID) on lung adenocarcinoma in spontaneous lung cancer mouse model. Six-week-old transgenic KrasG12V mice (n = 20; male + female) were fed modified AIN-76A diets containing naproxen (0/400 ppm) for 30 wk and euthanized at 36 wk of age. Lungs were evaluated for tumor incidence, multiplicity, and histopathological stage (adenoma and adenocarcinoma). Lung tumors were noticeable as early as 12 wk of age exclusively in the KrasG12V mice. By 36 wk age, 100% of KrasG12V mice on control diet developed lung tumors, mostly adenocarcinomas. KrasG12V mice fed control diet developed 19.8 ± 0.96 (Mean ± SEM) lung tumors (2.5 ± 0.3 adenoma, 17.3 ± 0.7 adenocarcinoma). Administration of naproxen (400 ppm) inhibited lung tumor multiplicity by ∼52% (9.4 ± 0.85; P < 0001) and adenocarcinoma by ∼64% (6.1 ± 0.6; P < 0001), compared with control-diet-fed mice. However, no significant difference was observed in the number of adenomas in either diet, suggesting that naproxen was more effective in inhibiting tumor progression to adenocarcinoma. Biomarker analysis showed significantly reduced inflammation (COX-2, IL-10), reduced tumor cell proliferation (PCNA, cyclin D1), and increased apoptosis (p21, caspase-3) in the lung tumors exposed to naproxen. Decreased serum levels of PGE2 and CXCR4 were observed in naproxen diet fed KrasG12V mice. Gene expression analysis of tumors revealed a significant increase in cytokine modulated genes (H2-Aa, H2-Ab1, Clu), which known to further modulate the cytokine signaling pathways. Overall, the results suggest a chemopreventive role of naproxen in inhibiting spontaneous lung adenocarcinoma formation in KrasG12V mice.
Collapse
|
4
|
Increased expression of IGF-1Ec with increasing colonic polyp dysplasia and colorectal cancer. J Cancer Res Clin Oncol 2020; 146:2861-2870. [PMID: 32772171 DOI: 10.1007/s00432-020-03345-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE IGF-1Ec is an isoform of Insulin-like growth factor 1 (IGF-1) and has recently been identified to be overexpressed in cancers including prostate and neuroendocrine tumours. The aim of this paper is to investigate the expression of IGF-1Ec in colorectal cancer and polyps compared to normal colon tissues and its association with recurrent disease using semi-quantitative immunohistochemistry. METHODS Immunohistochemistry for IGF-1Ec expression was performed for colorectal cancer, colorectal polyps and normal colonic tissues. The quantification of IGF-1Ec expression was performed with the use of Image J software and the IHC profiler plugin. Following ethics approval from the National Research Ethics Service (Reference 11/LO/1521), clinical information including recurrent disease on follow-up was collected for patients with colorectal cancer. RESULTS Immunohistochemistry was performed in 16 patients with colorectal cancer and 11 patients with colonic polyps and compared to normal colon tissues and prostate adenocarcinoma (positive control) tissues. Significantly increased expression of IGF-1Ec was demonstrated in colorectal cancer (p < 0.001) and colorectal polyps (p < 0.05) compared to normal colonic tissues. Colonic adenomas with high-grade dysplasia had significantly higher expression of IGF-1Ec compared to low-grade dysplastic adenomas (p < 0.001). Colorectal cancers without lymph node metastases at the time of presentation had significantly higher IGF-1Ec expression compared to lymph node-positive disease (p < 0.05). No correlation with recurrent disease was identified with IGF-1Ec expression. CONCLUSION IGF-1Ec is significantly overexpressed in colorectal cancer and polyps compared to normal colon tissues offering a potential target to improve colonoscopic identification of colorectal polyps and cancer and intraoperative identification of colorectal tumours.
Collapse
|
5
|
Das L, Patel B, Patri M. Adolescence benzo[a]pyrene treatment induces learning and memory impairment and anxiolytic like behavioral response altering neuronal morphology of hippocampus in adult male Wistar rats. Toxicol Rep 2019; 6:1104-1113. [PMID: 31720231 PMCID: PMC6838974 DOI: 10.1016/j.toxrep.2019.10.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 10/11/2019] [Accepted: 10/15/2019] [Indexed: 12/11/2022] Open
Abstract
Intraperitoneal B[a]P administration induces anxiolytic like behavior in rats. B[a]P induces oxidative stress and reduces antioxidant enzyme activity. Exposure to B[a]P-induces decrease in dendrite length and spine density through oxidative stress affecting antioxidant defence system. Alteration in the neuronal architecture of the hippocampal cells after B[a]P administration is associated with learning and memory defict.
Exposure to benzo[a]pyrene (B[a]P), a prototype of polycyclic aromatic hydrocarbons (PAHs) easily cross blood brain barrier (BBB) and is associated with impaired learning and memory in adult rats. However, there is no symmetric study reported on association between B[a]P exposure during the early development and hippocampal dendritic architecture causing behavioral changes like learning and memory deficit of adult rats. We investigated a fourteen day consecutive B[a]P administration, intraperitonial (i.p.), with two different doses (0.1 and 1μM) during early adolescence at PND30-44 and learning behavior assessed between PND 45-60 in adult male rats. The anxiolytic like behavioural analysis was done by LDPT. Depressive like behaviour was estimated through sucrose preference and learning and memory by T-maze. After B[a]P administration oxidative stress markers like glutathione S-transferase (GST), glutathione reductase (GR), glutathione peroxidase (GPx), reduced (GSH) and oxidized glutathione (GSSG) were evaluated. To parallel these behavioral and antioxidant level changes to alteration in dendritic morphology, Golgi-Cox staining was performed in the hippocampus. Our study showed anxiolytic like behavioral response with significant increase in time spent in light zone and significant (p < 0.05) decrease in preference for sucrose and a reduction in percentage of spontaneous responses in T-maze test in B[a]P administered group as compared to vehicle control. B[a]P exposed male rats showed significant increase in GST activity (p < 0.05) and concentration of GSSG with a decay in GSH, GPx and GR in both the groups as compared to control. B[a]P administered rats showed significant loss in total dendritic length and number (28%) with reduced spine density (18%) in both higher and lower doses. These results suggested that B[a]P administration can be associated with an increase ROS production showing altered antioxidant defence system through glutathione biosynthesis and causing profound alterations in dendritic length and spine density of hippocampal neurons leading towards learning and memory deficits in adult rats.
Collapse
Affiliation(s)
- Lipsa Das
- Neurobiology Laboratory, Department of Zoology, School of Life Sciences, Ravenshaw University, Cuttack, 753003, Odisha, India
| | - Bhupesh Patel
- Neurobiology Laboratory, Department of Zoology, School of Life Sciences, Ravenshaw University, Cuttack, 753003, Odisha, India
| | - Manorama Patri
- Neurobiology Laboratory, Department of Zoology, School of Life Sciences, Ravenshaw University, Cuttack, 753003, Odisha, India
| |
Collapse
|
6
|
Lewis CW, Bukhari AB, Xiao EJ, Choi WS, Smith JD, Homola E, Mackey JR, Campbell SD, Gamper AM, Chan GK. Upregulation of Myt1 Promotes Acquired Resistance of Cancer Cells to Wee1 Inhibition. Cancer Res 2019; 79:5971-5985. [PMID: 31594837 DOI: 10.1158/0008-5472.can-19-1961] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/04/2019] [Accepted: 10/04/2019] [Indexed: 11/16/2022]
Abstract
Adavosertib (also known as AZD1775 or MK1775) is a small-molecule inhibitor of the protein kinase Wee1, with single-agent activity in multiple solid tumors, including sarcoma, glioblastoma, and head and neck cancer. Adavosertib also shows promising results in combination with genotoxic agents such as ionizing radiation or chemotherapy. Previous studies have investigated molecular mechanisms of primary resistance to Wee1 inhibition. Here, we investigated mechanisms of acquired resistance to Wee1 inhibition, focusing on the role of the Wee1-related kinase Myt1. Myt1 and Wee1 kinases were both capable of phosphorylating and inhibiting Cdk1/cyclin B, the key enzymatic complex required for mitosis, demonstrating their functional redundancy. Ectopic activation of Cdk1 induced aberrant mitosis and cell death by mitotic catastrophe. Cancer cells with intrinsic adavosertib resistance had higher levels of Myt1 compared with sensitive cells. Furthermore, cancer cells that acquired resistance following short-term adavosertib treatment had higher levels of Myt1 compared with mock-treated cells. Downregulating Myt1 enhanced ectopic Cdk1 activity and restored sensitivity to adavosertib. These data demonstrate that upregulating Myt1 is a mechanism by which cancer cells acquire resistance to adavosertib. SIGNIFICANCE: Myt1 is a candidate predictive biomarker of acquired resistance to the Wee1 kinase inhibitor adavosertib.
Collapse
Affiliation(s)
- Cody W Lewis
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada.,Experimental Oncology, Cross Cancer Institute, Edmonton, Alberta, Canada.,Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, Alberta, Canada
| | - Amirali B Bukhari
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada.,Experimental Oncology, Cross Cancer Institute, Edmonton, Alberta, Canada.,Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, Alberta, Canada
| | - Edric J Xiao
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada.,Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, Alberta, Canada
| | - Won-Shik Choi
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada.,Experimental Oncology, Cross Cancer Institute, Edmonton, Alberta, Canada.,Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, Alberta, Canada
| | - Joanne D Smith
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada.,Experimental Oncology, Cross Cancer Institute, Edmonton, Alberta, Canada.,Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, Alberta, Canada
| | - Ellen Homola
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - John R Mackey
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada.,Medical Oncology, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Shelagh D Campbell
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Armin M Gamper
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada.,Experimental Oncology, Cross Cancer Institute, Edmonton, Alberta, Canada.,Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, Alberta, Canada
| | - Gordon K Chan
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada. .,Experimental Oncology, Cross Cancer Institute, Edmonton, Alberta, Canada.,Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
7
|
Abstract
Purpose of review This article provides a brief overview of cancer-preventive phytochemicals specifically targeting pancreatic cancer (PC) stem cells for prevention and treatment. Recent findings Cancer stem cells (CSCs) represent a small proportion of the total cells of a given tumor, and contribute to tumor growth, recurrence, metastasis, and treatment resistance. Many intertwined pathways, including hedgehog, Wnt Signaling, and NOTCH, have been shown to play a role in the formation of CSCs. Recently, numerous chemopreventive agents, such as genistein, (-)-epigallocatechin-3-gallate (EGCG), sulforaphane, curcumin, resveratrol, and quercetin, have been shown to target CSCs mediated through the inhibition of multiple signalling pathways, to avoid toxicity and the side effects of chemical compounds. Summary A growing body of research suggests that CSCs are the drivers in treatment resistance, cancer recurrence, and metastasis, in addition to tumor initiation and heterogeneity. Patient survival depends on these CSCs, which are one cause of tumor recurrence after surgery and chemotherapy. Therefore, target selection; an improved understanding of CSC biology, the genetic and molecular profiles of CSCs, and their key signaling pathways, and; appropriate clinical trials endpoints that are designed to target CSCs will help in the development of drugs that will specifically target this small population of stem cells.
Collapse
|
8
|
Janakiram NB, Mohammed A, Madka V, Kumar G, Rao CV. Prevention and treatment of cancers by immune modulating nutrients. Mol Nutr Food Res 2016; 60:1275-94. [PMID: 26833775 PMCID: PMC6038926 DOI: 10.1002/mnfr.201500884] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 01/16/2016] [Accepted: 01/18/2016] [Indexed: 12/11/2022]
Abstract
Epidemiological and laboratory data support the protective effects of bioactive nutrients in our diets for various diseases. Along with various factors, such as genetic history, alcohol, smoking, exercise, and dietary choices play a vital role in affecting an individual's immune responses toward a transforming cell, by either preventing or accelerating a neoplastic transformation. Ample evidence suggests that dietary nutrients control the inflammatory and protumorigenic responses in immune cells. Immunoprevention is usually associated with the modulation of immune responses that help in resolving the inflammation, thus improving clinical outcome. Various metabolic pathway-related nutrients, including glutamine, arginine, vitamins, minerals, and long-chain fatty acids, are important components of immunonutrient mixes. Epidemiological studies related to these substances have reported different results, with no or minimal effects. However, several studies suggest that these nutrients may have immune-modulating effects that may lower cancer risk. Preclinical studies submit that most of these components may provide beneficial effects. The present review discusses the available data, the immune-modulating functions of these nutrients, and how these substances could be used to study immune modulation in a neoplastic environment. Further research will help to determine whether the mechanistic signaling pathways in immune cells altered by nutrients can be exploited for cancer prevention and treatment.
Collapse
Affiliation(s)
- Naveena B. Janakiram
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology Oncology Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Altaf Mohammed
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology Oncology Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Venkateshwar Madka
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology Oncology Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Gaurav Kumar
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology Oncology Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Chinthalapally V. Rao
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology Oncology Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
9
|
Kumar G, Patlolla JMR, Madka V, Mohammed A, Li Q, Zhang Y, Biddick L, Singh A, Gillaspy A, Lightfoot S, Steele VE, Kopelovich L, Rao CV. Simultaneous targeting of 5-LOX-COX and ODC block NNK-induced lung adenoma progression to adenocarcinoma in A/J mice. Am J Cancer Res 2016; 6:894-909. [PMID: 27293987 PMCID: PMC4889708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 03/02/2016] [Indexed: 06/06/2023] Open
Abstract
Lung cancer is the leading cause of cancer deaths worldwide. Targeting complementary pathways will achieve better treatment efficacy than a single agent high-dose strategy that could increase risk of side effects and tumor resistance. To target COX-2, 5-LOX, and ODC simultaneously, we tested the effects of a dual 5-LOX-COX inhibitor, licofelone, and an ODC inhibitor, DFMO, alone and in combination, on NNK-induced lung tumors in female A/J mice. Seven-week-old mice were treated with NNK (10 μmol/mouse, single dose, i.p.) and randomized to different treatment groups. Three weeks after injection, mice were fed control or experimental diets (DFMO 1500/3000 ppm, licofelone 200/400 ppm, or a low-dose combination of 1500 ppm DFMO and 200 ppm licofelone) for 17 or 34 weeks. Both agents significantly inhibited tumor formation in a dose-dependent manner. As anticipated more adenomas and adenocarcinomas were observed at 17 and 34 weeks, respectively. Importantly, low dose combination of DFMO and licofelone showed more pronounced effects at 17 or 34 weeks in inhibiting the total tumor formation (~60%, p < 0.0001) and adenocarcinoma (~65%, p < 0.0001) compared to individual high dose of DFMO (~44% and 46%, p < 0.0001) and licofelone (~48% and 55%, p < 0.0001). DFMO and combination-treated mice lung tumors exhibited modulated ODC pathway components (Oat, Oaz, SRM, SMS, and SAT, p < 0.05) along with decreased proliferation (PCNA, Cyclin D1 and Cyclin A) and increased expression of p53, p21 and p27 compared to mice fed control diet. Both DFMO and licofelone significantly inhibited tumor inflammatory markers. Our findings suggest that a low-dose combined treatment targeting inflammation and polyamine synthesis may provide effective chemoprevention.
Collapse
Affiliation(s)
- Gaurav Kumar
- Center for Cancer Prevention and Drug Development, Hematology-Oncology Section, Department of Medicine, Stephenson Cancer Center, University of Oklahoma Health Sciences CenterOklahoma City, OK 73104, USA
| | - Jagan Mohan R Patlolla
- Center for Cancer Prevention and Drug Development, Hematology-Oncology Section, Department of Medicine, Stephenson Cancer Center, University of Oklahoma Health Sciences CenterOklahoma City, OK 73104, USA
| | - Venkateshwar Madka
- Center for Cancer Prevention and Drug Development, Hematology-Oncology Section, Department of Medicine, Stephenson Cancer Center, University of Oklahoma Health Sciences CenterOklahoma City, OK 73104, USA
| | - Altaf Mohammed
- Center for Cancer Prevention and Drug Development, Hematology-Oncology Section, Department of Medicine, Stephenson Cancer Center, University of Oklahoma Health Sciences CenterOklahoma City, OK 73104, USA
| | - Qian Li
- Center for Cancer Prevention and Drug Development, Hematology-Oncology Section, Department of Medicine, Stephenson Cancer Center, University of Oklahoma Health Sciences CenterOklahoma City, OK 73104, USA
| | - Yuting Zhang
- Center for Cancer Prevention and Drug Development, Hematology-Oncology Section, Department of Medicine, Stephenson Cancer Center, University of Oklahoma Health Sciences CenterOklahoma City, OK 73104, USA
| | - Laura Biddick
- Center for Cancer Prevention and Drug Development, Hematology-Oncology Section, Department of Medicine, Stephenson Cancer Center, University of Oklahoma Health Sciences CenterOklahoma City, OK 73104, USA
| | - Anil Singh
- Center for Cancer Prevention and Drug Development, Hematology-Oncology Section, Department of Medicine, Stephenson Cancer Center, University of Oklahoma Health Sciences CenterOklahoma City, OK 73104, USA
| | - Allison Gillaspy
- Division of Cancer Prevention, Chemoprevention Agent Development Research Group, National Cancer InstituteBethesda, MD, USA
| | - Stanley Lightfoot
- Center for Cancer Prevention and Drug Development, Hematology-Oncology Section, Department of Medicine, Stephenson Cancer Center, University of Oklahoma Health Sciences CenterOklahoma City, OK 73104, USA
| | - Vernon E Steele
- Division of Cancer Prevention, Chemoprevention Agent Development Research Group, National Cancer InstituteBethesda, MD, USA
| | - Levy Kopelovich
- Division of Cancer Prevention, Chemoprevention Agent Development Research Group, National Cancer InstituteBethesda, MD, USA
| | - Chinthalapally V Rao
- Center for Cancer Prevention and Drug Development, Hematology-Oncology Section, Department of Medicine, Stephenson Cancer Center, University of Oklahoma Health Sciences CenterOklahoma City, OK 73104, USA
| |
Collapse
|
10
|
PATLOLLA JAGANM, KOPELOVICH LEVY, QIAN LI, ZHANG YUTING, KUMAR GAURAV, MADKA VENKATESHWAR, MOHAMMED ALTAF, BIDDICK LAURA, SADEGHI MICHAEL, LIGHTFOOT STAN, RAO CHINTHALAPALLYV. Early and delayed intervention with rapamycin prevents NNK-induced lung adenocarcinoma in A/J mice. Oncol Rep 2015; 34:2925-34. [PMID: 26397133 PMCID: PMC4735698 DOI: 10.3892/or.2015.4277] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 07/09/2015] [Indexed: 12/18/2022] Open
Abstract
In tobacco-associated lung cancers, the protein kinase B/mammalian target of rapamycin (Akt/mTOR) pathway frequently is activated by nicotine and its metabolite 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK). The aim of the present study was to examine the effects of early or late intervention with rapamycin in NNK-induced lung adenoma and progression to adenocarcinoma in female A/J mice. At 7 weeks of age, 40 mice/each carcinogen group received one dose of 10 μmol NNK i.p. Three weeks later, the early intervention groups (25/group) were fed diets containing 0, 8 or 16 ppm rapamycin. The mice were sacrificed after 17 or 34 weeks of drug exposure and tumors were evaluated via histopathology. For late intervention (late adenoma and adenocarcinoma stage), groups of 15 mice were administered diets containing 8 or 16 ppm rapamycin starting 20 weeks after NNK treatment and continuing for 17 weeks before evaluation of tumor progression. Administration of 8 or 16 ppm rapamycin as an early or a late stage intervention significantly suppressed lung adenoma and adenocarcinoma formation (p<0.01-0.0001) after 17 or 34 weeks of exposure. The effect was more pronounced (>50‑60% tumor inihibition; p<0.0001) at the early intervention and the size of NNK-induced tumors decreased from >2.10 to <~0.75 mm3 (p=0.0056). Lung tumors harvested from mice exposed to rapamycin showed a significant decrease in p-mTOR, p-S6K1, PCNA and Bcl-xL as compared with controls in the early and late stage intervention studies. These observations suggest that rapamycin is highly effective even with administration after dysplastic adenoma or early adenocarcinoma stages and is useful for high-risk lung cancer patients.
Collapse
Affiliation(s)
- JAGAN M.R. PATLOLLA
- Center for Chemoprevention and Cancer Drug Development, Department of Medicine, Hem-Onc Section, PCS Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - LEVY KOPELOVICH
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD 20892-9788, USA
| | - LI QIAN
- Center for Chemoprevention and Cancer Drug Development, Department of Medicine, Hem-Onc Section, PCS Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - YUTING ZHANG
- Center for Chemoprevention and Cancer Drug Development, Department of Medicine, Hem-Onc Section, PCS Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - GAURAV KUMAR
- Center for Chemoprevention and Cancer Drug Development, Department of Medicine, Hem-Onc Section, PCS Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - VENKATESHWAR MADKA
- Center for Chemoprevention and Cancer Drug Development, Department of Medicine, Hem-Onc Section, PCS Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - ALTAF MOHAMMED
- Center for Chemoprevention and Cancer Drug Development, Department of Medicine, Hem-Onc Section, PCS Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - LAURA BIDDICK
- Center for Chemoprevention and Cancer Drug Development, Department of Medicine, Hem-Onc Section, PCS Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - MICHAEL SADEGHI
- Center for Chemoprevention and Cancer Drug Development, Department of Medicine, Hem-Onc Section, PCS Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - STAN LIGHTFOOT
- Center for Chemoprevention and Cancer Drug Development, Department of Medicine, Hem-Onc Section, PCS Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - CHINTHALAPALLY V. RAO
- Center for Chemoprevention and Cancer Drug Development, Department of Medicine, Hem-Onc Section, PCS Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
11
|
Puliyappadamba VT, Thulasidasan AKT, Vijayakurup V, Antony J, Bava SV, Anwar S, Sundaram S, Anto RJ. Curcumin inhibits B[a]PDE-induced procarcinogenic signals in lung cancer cells, and curbs B[a]P-induced mutagenesis and lung carcinogenesis. Biofactors 2015; 41:431-42. [PMID: 26643788 DOI: 10.1002/biof.1244] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 10/19/2015] [Indexed: 12/22/2022]
Abstract
Benzo[a]pyrene is a procarcinogen present in environment and cigarette smoke, which could be bio-transformed in vivo to B[a]PDE, a potent carcinogen known to form DNA adducts and induce mutations. We observed that curcumin, a known chemopreventive, could significantly inhibit the survival of lung cancer cells exposed to B[a]PDE. It also downregulates B[a]PDE-induced nuclear translocation of NF-κB as assessed by Electrophoretic Mobility Shift Assay (EMSA) and NF-κB-dependent reporter gene assay. Ames assay demonstrated its ability to revert the mutagenic property of benzo[a]pyrene. These observations prompted us to evaluate the efficacy of curcumin in preventing B[a]P-induced lung carcinogenesis in vivo and to explore the molecular mechanism associated with it. The average number of tumor nodules present in the lungs of the Swiss albino mice, which received benzo[a]pyrene, was significantly high compared to that received curcumin as 2% diet along with B[a]P. Curcumin treatment significantly reverted histopathological deviations in the lung tissues due to benzo[a]pyrene ingestion. Moreover, curcumin diet reduced benzo[a]pyrene-induced activation of NF-κB and MAPK signaling and Cox-2 transcription in lung tissues of mice. Taken together, this study illustrates multifaceted efficacy of curcumin in preventing lung cancer.
Collapse
Affiliation(s)
| | - Arun Kumar T Thulasidasan
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - Vinod Vijayakurup
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - Jayesh Antony
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - Smitha V Bava
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - Shabna Anwar
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - Sankar Sundaram
- Department of Pathology, Government Medical College, Thiruvananthapuram, Kerala, 695011, India
| | - Ruby John Anto
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| |
Collapse
|