1
|
Chen H, Hu XB, Zhou J, He CY, Wang K, Yi Q. Association of chronic obstructive pulmonary disease with risk of lung cancer in individuals aged 40 years and older: A cross-sectional study based on NHANES 2013-2018. PLoS One 2024; 19:e0311537. [PMID: 39441785 PMCID: PMC11498685 DOI: 10.1371/journal.pone.0311537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND It remains unclear whether chronic obstructive pulmonary disease (COPD) is an independent risk factor for lung cancer after excluding confounding factors such as smoking, age, sex, body mass index (BMI), comorbidities, etc. METHODS Data from 11,440 participants (≥ 40 years old) in the National Health and Nutrition Examination Survey (NHANES) 2013-2018 were analyzed. Weighted multivariable logistic regression models were used to assess the association between COPD and lung cancer risk. Subgroup analyses were based on age, sex, BMI, and smoking. RESULTS This study included 660 patients with COPD and 10,780 participants without COPD. The prevalence of lung cancer was significantly higher in patients with COPD compared to participants without COPD (3.39% vs 0.14%). After adjusting for confounding factors, COPD was associated with a significantly increased risk of lung cancer (OR, 12.24, 95% CI, 4.99-30.06, p < 0.001). This association remained significant in all subgroups, particularly in individuals aged > 65 years (OR, 20.05, 95% CI, 6.85-58.72, p < 0.001), smokers (OR, 19.38, 95% CI, 2.02-185.66, p = 0.010), males (OR, 17.39, 95% CI, 5.28-57.31, p < 0.001), individuals who quit smoking within 10 years (OR, 12.86, 95% CI, 2.59, 63.99, p = 0.002), and individuals with a BMI > 25 kg/m2 (OR, 14.56, 95% CI, 3.88-54.69, p < 0.001). CONCLUSIONS COPD is an independent risk factor for lung cancer, especially in certain subgroups. The combination of COPD and smoking greatly amplifies the lung cancer risk. These findings highlight the importance of early lung cancer screening in patients with COPD.
Collapse
Affiliation(s)
- Hong Chen
- Department of Respiratory and Critical Care Medicine, Chengdu Second People’s Hospital, Chengdu, China
| | - Xiao-Bo Hu
- Department of Respiratory and Critical Care Medicine, Chengdu Second People’s Hospital, Chengdu, China
| | - Jin Zhou
- Department of Medical Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Chen-Yun He
- Department of Respiratory and Critical Care Medicine, Chengdu Second People’s Hospital, Chengdu, China
| | - Ke Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Qun Yi
- Department of Medical Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Papavassiliou KA, Anagnostopoulos N, Papavassiliou AG. Glucocorticoid Receptor Signaling in NSCLC: Mechanistic Aspects and Therapeutic Perspectives. Biomolecules 2023; 13:1286. [PMID: 37759686 PMCID: PMC10526876 DOI: 10.3390/biom13091286] [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: 07/25/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Recent advances in non-small cell lung cancer (NSCLC) biology and the discovery of novel therapeutic targets have led to the development of new pharmacological agents that may improve the clinical outcome of patients with NSCLC. The glucocorticoid receptor (GR) is an evolutionarily conserved protein belonging to the nuclear receptor superfamily of transcription factors and mediates the diverse actions of glucocorticoids in cells. Data suggest that the GR may play a relevant role in the molecular mechanisms of NSCLC tumorigenesis and malignant progression. Additionally, evidence indicates that glucocorticoids may affect the efficacy of standard treatment, including chemotherapy, immune checkpoint inhibitors, and targeted therapy. Furthermore, several findings show that GR expression may probably be associated with NSCLC patient survival. Finally, glucocorticoids may be used as therapeutic agents for the clinical management of NSCLC patients. Here, we briefly review the latest advances on the biological role of GR signaling in NSCLC and discuss the potential use of the GR as a prognostic and predictive biomarker. Importantly, we explore the therapeutic potential of glucocorticoids and the effect of adding such drugs to standard therapies for NSCLC.
Collapse
Affiliation(s)
- Kostas A. Papavassiliou
- First Department of Respiratory Medicine, ‘Sotiria’ Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (N.A.)
| | - Nektarios Anagnostopoulos
- First Department of Respiratory Medicine, ‘Sotiria’ Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (N.A.)
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
3
|
Motafeghi F, Mortazavi P, Ghassemi-Barghi N, Zahedi M, Shokrzadeh M. Dexamethasone as an anti-cancer or hepatotoxic. Toxicol Mech Methods 2023; 33:161-171. [PMID: 35866224 DOI: 10.1080/15376516.2022.2105183] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The linkage between inflammation and oxidative stress in liver damage has been proven and is undeniable; dexamethasone with some antioxidants can reduce the toxicity of liver tissue. Due to the importance of cancer treatment, glucocorticoids' synergistic effect in inhibiting cancer cell growth is also investigated. Dexamethasone alone and combined with etoposide were tested at concentrations of 1, 5, and 10 μM to evaluate the potency of dexamethasone in inhibiting the growth of A549 cells using oxidative stress factors and DNA damage. Also, intraperitoneal injection of dexamethasone in rats was used to induce liver toxicity. Coenzyme Q10 at different concentrations (1, 10, and 50 mg/kg) was used as an antioxidant to assess the oxidative stress factors and measure Caspase-3 activity. The results showed that dexamethasone combined with etoposide could significantly inhibit the growth of cancer cells and induce apoptosis. Treatment of A549 cells using dexamethasone also inhibits cancer cells' growth by inducing oxidative stress and DNA damage. Dexamethasone also, by inducing oxidative stress and activation of caspase 3, ultimately causes hepatotoxicity. Treatment with different concentrations of CoQ10 showed improved mitochondrial function, antioxidant defense, and liver enzyme. The best effect of coenzyme Q10 on dexamethasone-induced hepatotoxicity is 50 mg/kg. As a result, dexamethasone (alone and combined with etoposide) has an anti-cancer effect by damaging DNA and inducing oxidative stress. Also, CoQ10 has antioxidant effects against dexamethasone-induced hepatotoxicity by improving mitochondrial function and reducing caspase-3 activity.
Collapse
Affiliation(s)
- Farzaneh Motafeghi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Parham Mortazavi
- Department of Pharmacology and Toxicology, Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nasrin Ghassemi-Barghi
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Zahedi
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.,Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Shokrzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
4
|
Targeting Nuclear Receptors in Lung Cancer—Novel Therapeutic Prospects. Pharmaceuticals (Basel) 2022; 15:ph15050624. [PMID: 35631448 PMCID: PMC9145966 DOI: 10.3390/ph15050624] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 01/27/2023] Open
Abstract
Lung cancer, the second most commonly diagnosed cancer, is the major cause of fatalities worldwide for both men and women, with an estimated 2.2 million new incidences and 1.8 million deaths, according to GLOBOCAN 2020. Although various risk factors for lung cancer pathogenesis have been reported, controlling smoking alone has a significant value as a preventive measure. In spite of decades of extensive research, mechanistic cues and targets need to be profoundly explored to develop potential diagnostics, treatments, and reliable therapies for this disease. Nuclear receptors (NRs) function as transcription factors that control diverse biological processes such as cell growth, differentiation, development, and metabolism. The aberrant expression of NRs has been involved in a variety of disorders, including cancer. Deregulation of distinct NRs in lung cancer has been associated with numerous events, including mutations, epigenetic modifications, and different signaling cascades. Substantial efforts have been made to develop several small molecules as agonists or antagonists directed to target specific NRs for inhibiting tumor cell growth, migration, and invasion and inducing apoptosis in lung cancer, which makes NRs promising candidates for reliable lung cancer therapeutics. The current work focuses on the importance of various NRs in the development and progression of lung cancer and highlights the different small molecules (e.g., agonist or antagonist) that influence NR expression, with the goal of establishing them as viable therapeutics to combat lung cancer.
Collapse
|
5
|
Prekovic S, Schuurman K, Mayayo-Peralta I, Manjón AG, Buijs M, Yavuz S, Wellenstein MD, Barrera A, Monkhorst K, Huber A, Morris B, Lieftink C, Chalkiadakis T, Alkan F, Silva J, Győrffy B, Hoekman L, van den Broek B, Teunissen H, Debets DO, Severson T, Jonkers J, Reddy T, de Visser KE, Faller W, Beijersbergen R, Altelaar M, de Wit E, Medema R, Zwart W. Glucocorticoid receptor triggers a reversible drug-tolerant dormancy state with acquired therapeutic vulnerabilities in lung cancer. Nat Commun 2021; 12:4360. [PMID: 34272384 PMCID: PMC8285479 DOI: 10.1038/s41467-021-24537-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 06/24/2021] [Indexed: 12/13/2022] Open
Abstract
The glucocorticoid receptor (GR) regulates gene expression, governing aspects of homeostasis, but is also involved in cancer. Pharmacological GR activation is frequently used to alleviate therapy-related side-effects. While prior studies have shown GR activation might also have anti-proliferative action on tumours, the underpinnings of glucocorticoid action and its direct effectors in non-lymphoid solid cancers remain elusive. Here, we study the mechanisms of glucocorticoid response, focusing on lung cancer. We show that GR activation induces reversible cancer cell dormancy characterised by anticancer drug tolerance, and activation of growth factor survival signalling accompanied by vulnerability to inhibitors. GR-induced dormancy is dependent on a single GR-target gene, CDKN1C, regulated through chromatin looping of a GR-occupied upstream distal enhancer in a SWI/SNF-dependent fashion. These insights illustrate the importance of GR signalling in non-lymphoid solid cancer biology, particularly in lung cancer, and warrant caution for use of glucocorticoids in treatment of anticancer therapy related side-effects.
Collapse
Affiliation(s)
- Stefan Prekovic
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Karianne Schuurman
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Isabel Mayayo-Peralta
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Anna G Manjón
- Division of Cell Biology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Mark Buijs
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Selçuk Yavuz
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Max D Wellenstein
- Division of Tumour Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alejandro Barrera
- Department of Biostatistics & Bioinformatics, and Centre for Genomic & Computational Biology, Duke University Medical Centre, Durham, NC, USA
| | - Kim Monkhorst
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Anne Huber
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
| | - Ben Morris
- Division of Molecular Carcinogenesis and Robotics and Screening Centre, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Cor Lieftink
- Division of Molecular Carcinogenesis and Robotics and Screening Centre, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Theofilos Chalkiadakis
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ferhat Alkan
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Joana Silva
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Balázs Győrffy
- Semmelweis University Department of Bioinformatics and 2nd Department of Pediatrics, Budapest, Hungary.,TTK Cancer Biomarker Research Group, Institute of Enzymology, Budapest, Hungary
| | - Liesbeth Hoekman
- Mass spectrometry/Proteomics Facility, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Bram van den Broek
- Division of Cell Biology and BioImaging Facility, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Hans Teunissen
- Division of Gene Regulation, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Donna O Debets
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Tesa Severson
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Timothy Reddy
- Department of Biostatistics & Bioinformatics, and Centre for Genomic & Computational Biology, Duke University Medical Centre, Durham, NC, USA
| | - Karin E de Visser
- Division of Tumour Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - William Faller
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Roderick Beijersbergen
- Division of Molecular Carcinogenesis and Robotics and Screening Centre, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Maarten Altelaar
- Mass spectrometry/Proteomics Facility, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Elzo de Wit
- Division of Gene Regulation, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Rene Medema
- Division of Cell Biology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wilbert Zwart
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands. .,Laboratory of Chemical Biology and Institute for Complex Molecular Systems, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
| |
Collapse
|
6
|
D'Ippolito AM, McDowell IC, Barrera A, Hong LK, Leichter SM, Bartelt LC, Vockley CM, Majoros WH, Safi A, Song L, Gersbach CA, Crawford GE, Reddy TE. Pre-established Chromatin Interactions Mediate the Genomic Response to Glucocorticoids. Cell Syst 2018; 7:146-160.e7. [PMID: 30031775 DOI: 10.1016/j.cels.2018.06.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/27/2018] [Accepted: 06/12/2018] [Indexed: 10/28/2022]
Abstract
The glucocorticoid receptor (GR) is a hormone-inducible transcription factor involved in metabolic and anti-inflammatory gene expression responses. To investigate what controls interactions between GR binding sites and their target genes, we used in situ Hi-C to generate high-resolution, genome-wide maps of chromatin interactions before and after glucocorticoid treatment. We found that GR binding to the genome typically does not cause new chromatin interactions to target genes but instead acts through chromatin interactions that already exist prior to hormone treatment. Both glucocorticoid-induced and glucocorticoid-repressed genes increased interactions with distal GR binding sites. In addition, while glucocorticoid-induced genes increased interactions with transcriptionally active chromosome compartments, glucocorticoid-repressed genes increased interactions with transcriptionally silent compartments. Lastly, while the architectural DNA-binding proteins CTCF and RAD21 were bound to most chromatin interactions, we found that glucocorticoid-responsive chromatin interactions were depleted for CTCF binding but enriched for RAD21. Together, these findings offer new insights into the mechanisms underlying GC-mediated gene activation and repression.
Collapse
Affiliation(s)
- Anthony M D'Ippolito
- University Program in Genetics and Genomics, Duke University, Durham, NC 27708, USA; Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA
| | - Ian C McDowell
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA; Computational Biology and Bioinformatics Graduate Program, Duke University, Durham, NC 27708, USA
| | - Alejandro Barrera
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA; Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC 27710, USA
| | - Linda K Hong
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA; Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Sarah M Leichter
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA
| | - Luke C Bartelt
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA
| | - Christopher M Vockley
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA; Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC 27710, USA
| | - William H Majoros
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA; Computational Biology and Bioinformatics Graduate Program, Duke University, Durham, NC 27708, USA
| | - Alexias Safi
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA; Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Lingyun Song
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA; Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Charles A Gersbach
- University Program in Genetics and Genomics, Duke University, Durham, NC 27708, USA; Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Department of Orthopedic Surgery, Duke University Medical Center, Durham, NC 27708, USA
| | - Gregory E Crawford
- University Program in Genetics and Genomics, Duke University, Durham, NC 27708, USA; Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA; Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Timothy E Reddy
- University Program in Genetics and Genomics, Duke University, Durham, NC 27708, USA; Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA; Computational Biology and Bioinformatics Graduate Program, Duke University, Durham, NC 27708, USA; Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC 27710, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
7
|
Liu SF, Kuo HC, Lin MC, Ho SC, Tu ML, Chen YM, Chen YC, Fang WF, Wang CC, Liu GH. Inhaled corticosteroids have a protective effect against lung cancer in female patients with chronic obstructive pulmonary disease: a nationwide population-based cohort study. Oncotarget 2017; 8:29711-29721. [PMID: 28412726 PMCID: PMC5444697 DOI: 10.18632/oncotarget.15386] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 01/31/2017] [Indexed: 01/01/2023] Open
Abstract
Whether the use of inhaled corticosteroids (ICS) protects patients with chronic obstructive pulmonary disease (COPD) from lung cancer remains undetermined. In this retrospective nationwide population-based cohort study, we extracted data of 13,686 female COPD patients (ICS users, n = 1,290, ICS non-users, n = 12,396) diagnosed between 1997 and 2009 from the Taiwan's National Health Insurance database. These patients were followed-up until 2011, and lung cancer incidence was determined. Cox regression analysis was used to estimate hazard ratios (HRs) for lung cancer incidence. The time to lung cancer diagnosis was significantly different between ICS users and non-users (10.75 vs. 9.68 years, P < 0.001). Per 100,000 person-years, the lung cancer incidence rate was 235.92 for non-users and 158.67 for users [HR = 0.70 (95% confidence interval {CI}: 0.46-1.09)]. After adjusting for patients' age, income, and comorbidities, a cumulative ICS dose > 39.48 mg was significantly associated with a lower risk of lung cancer [ICS users > 39.48 mg, HR = 0.45 (95% CI: 0.21-0.96)]. Age ≥ 60 years, pneumonia, diabetes mellitus, and hypertension decreased lung cancer risk, whereas pulmonary tuberculosis increased the risk. Our results suggest that ICS have a potential role in lung cancer prevention among female COPD patients.
Collapse
Affiliation(s)
- Shih-Feng Liu
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung, Taiwan
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ho-Chang Kuo
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Meng-Chih Lin
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung, Taiwan
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shu-Chen Ho
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Mei-Lien Tu
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yu-Mu Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung, Taiwan
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yung-Che Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung, Taiwan
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Wen-Feng Fang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung, Taiwan
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chin-Chou Wang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung, Taiwan
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Guan-Heng Liu
- Department of Senior High School, Li-Chih Valuable School, Kaohsiung, Taiwan
| |
Collapse
|
8
|
McBeth L, Nwaneri AC, Grabnar M, Demeter J, Nestor-Kalinoski A, Hinds TD. Glucocorticoid receptor beta increases migration of human bladder cancer cells. Oncotarget 2016; 7:27313-24. [PMID: 27036026 PMCID: PMC5053652 DOI: 10.18632/oncotarget.8430] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/14/2016] [Indexed: 12/21/2022] Open
Abstract
Bladder cancer is observed worldwide having been associated with a host of environmental and lifestyle risk factors. Recent investigations on anti-inflammatory glucocorticoid signaling point to a pathway that may impact bladder cancer. Here we show an inverse effect on the glucocorticoid receptor (GR) isoform signaling that may lead to bladder cancer. We found similar GRα expression levels in the transitional uroepithelial cancer cell lines T24 and UMUC-3. However, the T24 cells showed a significant (p < 0.05) increased expression of GRβ compared to UMUC-3, which also correlated with higher migration rates. Knockdown of GRβ in the T24 cells resulted in a decreased migration rate. Mutational analysis of the 3' untranslated region (UTR) of human GRβ revealed that miR144 might positively regulate expression. Indeed, overexpression of miR144 increased GRβ by 3.8 fold. In addition, miR144 and GRβ were upregulated during migration. We used a peptide nucleic acid conjugated to a cell penetrating-peptide (Sweet-P) to block the binding site for miR144 in the 3'UTR of GRβ. Sweet-P effectively prevented miR144 actions and decreased GRβ expression, as well as the migration of the T24 human bladder cancer cells. Therefore, GRβ may have a significant role in bladder cancer, and possibly serve as a therapeutic target for the disease.
Collapse
Affiliation(s)
- Lucien McBeth
- Center for Hypertension and Personalized Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Assumpta C. Nwaneri
- Center for Hypertension and Personalized Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Maria Grabnar
- Center for Hypertension and Personalized Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Jonathan Demeter
- Center for Hypertension and Personalized Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Andrea Nestor-Kalinoski
- Advanced Microscopy and Imaging Center, Department of Surgery, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Terry D. Hinds
- Center for Hypertension and Personalized Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| |
Collapse
|
9
|
Taylor KM, Ray DW, Sommer P. Glucocorticoid receptors in lung cancer: new perspectives. J Endocrinol 2016; 229:R17-28. [PMID: 26795718 DOI: 10.1530/joe-15-0496] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 01/21/2016] [Indexed: 12/19/2022]
Abstract
Proper expression of the glucocorticoid receptor (GR) plays an essential role in the development of the lung. GR expression and signalling in the lung is manipulated by administration of synthetic glucocorticoids (Gcs) for the treatment of neonatal, childhood and adult lung diseases. In lung cancers, Gcs are also commonly used as co-treatment during chemotherapy. This review summarises the effect of Gc monotherapy and co-therapy on lung cancers in vitro, in mouse models of lung cancer, in xenograft, ex vivo and in vivo The disparity between the effects of pre-clinical and in vivo Gc therapy is commented on in light of the recent discovery of GR as a novel tumour suppressor gene.
Collapse
Affiliation(s)
- Kerryn M Taylor
- Division of GeneticsSchool of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - David W Ray
- Manchester Centre for Nuclear Hormone Research and DiseaseInstitute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom
| | - Paula Sommer
- Division of GeneticsSchool of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
10
|
Yenerall P, Kittler R. Minireview: Familiar Faces in Unfamiliar Places: The Emerging Role of Nuclear Receptors in Lung Cancer. Mol Endocrinol 2015; 29:1675-83. [PMID: 26484581 DOI: 10.1210/me.2015-1199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Nuclear hormone receptors (NRs) are a superfamily of 48 transcription factors that are frequently modulated by ligands and control various cancer-relevant cellular pathways, such as differentiation, proliferation, migration, and metabolism. These properties make them excellent therapeutic targets in cancers dependent upon their activity, and as such, 3 NRs, estrogen receptor-α, androgen receptor, and retinoic acid receptor-α (more specifically, the promyelocytic leukemia-retinoic acid receptor-α translocation), have been targeted clinically in breast cancer, prostate cancer, and acute promyelocytic leukemia, respectively. Recently, a number of studies have highlighted a putative role for NRs in nonsmall cell lung cancer (NSCLC), a highly lethal type of lung cancer with relatively few targeted agents. Here, we review the potential roles of selected NRs in NSCLC and offer insights on how NRs may be leveraged in NSCLC to improve patient outcomes.
Collapse
Affiliation(s)
- Paul Yenerall
- Eugene McDermott Center for Human Growth and Development (P.Y., R.K.), Hamon Center for Therapeutic Oncology Research (P.Y., R.K.), Simmons Comprehensive Cancer Center (R.K.), Department of Pharmacology (R.K.), and Green Center for Reproductive Biology Sciences (R.K.), The University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Ralf Kittler
- Eugene McDermott Center for Human Growth and Development (P.Y., R.K.), Hamon Center for Therapeutic Oncology Research (P.Y., R.K.), Simmons Comprehensive Cancer Center (R.K.), Department of Pharmacology (R.K.), and Green Center for Reproductive Biology Sciences (R.K.), The University of Texas Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|
11
|
Singh N, Taylor K, Mjoli PB, Poolman T, Ray DW, Sommer P. The N-terminal transactivation domain of the glucocorticoid receptor mediates apoptosis of human small cell lung cancer cells. Genes Chromosomes Cancer 2014; 53:999-1007. [DOI: 10.1002/gcc.22209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 07/24/2014] [Indexed: 12/20/2022] Open
Affiliation(s)
- Nimisha Singh
- Division of Genetics; School of Life Sciences, University of KwaZulu-Natal; Durban South Africa
| | - Kerryn Taylor
- Division of Genetics; School of Life Sciences, University of KwaZulu-Natal; Durban South Africa
| | - Phiwokuhle B. Mjoli
- Division of Genetics; School of Life Sciences, University of KwaZulu-Natal; Durban South Africa
| | - Toryn Poolman
- Centre in Endocrinology and Diabetes; Institute of Human Development, University of Manchester; Manchester UK
| | - David W. Ray
- Centre in Endocrinology and Diabetes; Institute of Human Development, University of Manchester; Manchester UK
| | - Paula Sommer
- Division of Genetics; School of Life Sciences, University of KwaZulu-Natal; Durban South Africa
| |
Collapse
|
12
|
Sommer P, Cowen RL, Berry A, Cookson A, Telfer BA, Williams KJ, Stratford IJ, Kay P, White A, Ray DW. Glucocorticoid receptor over-expression promotes human small cell lung cancer apoptosis in vivo and thereby slows tumor growth. Endocr Relat Cancer 2010; 17:203-13. [PMID: 20015838 PMCID: PMC2828806 DOI: 10.1677/erc-09-0241] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Small cell lung cancer (SCLC) is an aggressive tumor, associated with ectopic ACTH syndrome. We have shown that SCLC cells are glucocorticoid receptor (GR) deficient, and that restoration of GR expression confers glucocorticoid sensitivity and induces apoptosis in vitro. To determine the effects of GR expression in vivo, we characterized a mouse SCLC xenograft model that secretes ACTH precursor peptides, and so drives high circulating corticosterone concentrations (analogous to the ectopic ACTH syndrome). Infection of SCLC xenografts with GR-expressing adenovirus significantly slowed tumor growth compared with control virus infection. Time to fourfold initial tumor volume increased from a median of 9 days to 16 days (P=0.05; n=7 per group). Post-mortem analysis of GR-expressing tumors revealed a threefold increase in apoptotic (TUNEL positive) cells (P<0.01). Infection with the GR-expressing adenovirus caused a significant reduction in Bcl-2 and Bcl-xL transcripts. Furthermore, in both the GR-expressing adenovirus-infected cells and tumors, a significant number of uninfected cells underwent apoptosis, supporting a bystander cell killing effect. Therefore, GR expression is pro-apoptotic for human SCLCs in vivo, as well as in vitro, suggesting that loss of GR confers a survival advantage to SCLCs.
Collapse
Affiliation(s)
| | - Rachel L Cowen
- School of Pharmacy and Pharmaceutical SciencesUniversity of ManchesterAV Hill Building, Manchester, M13 9PTUK
| | - Andrew Berry
- Endocrine Sciences Research GroupUniversity of ManchesterAV Hill Building, Manchester, M13 9PTUK
| | - Ann Cookson
- Endocrine Sciences Research GroupUniversity of ManchesterAV Hill Building, Manchester, M13 9PTUK
| | - Brian A Telfer
- School of Pharmacy and Pharmaceutical SciencesUniversity of ManchesterAV Hill Building, Manchester, M13 9PTUK
| | - Kaye J Williams
- School of Pharmacy and Pharmaceutical SciencesUniversity of ManchesterAV Hill Building, Manchester, M13 9PTUK
| | - Ian J Stratford
- School of Pharmacy and Pharmaceutical SciencesUniversity of ManchesterAV Hill Building, Manchester, M13 9PTUK
| | - Paul Kay
- Endocrine Sciences Research GroupUniversity of ManchesterAV Hill Building, Manchester, M13 9PTUK
| | - Anne White
- Endocrine Sciences Research GroupUniversity of ManchesterAV Hill Building, Manchester, M13 9PTUK
- Faculty of Medical and Human SciencesUniversity of ManchesterAV Hill Building, Manchester, M13 9PTUK
- (Correspondence should be addressed to D W Ray; ; A White; )
| | - David W Ray
- Endocrine Sciences Research GroupUniversity of ManchesterAV Hill Building, Manchester, M13 9PTUK
- Faculty of Medical and Human SciencesUniversity of ManchesterAV Hill Building, Manchester, M13 9PTUK
- (Correspondence should be addressed to D W Ray; ; A White; )
| |
Collapse
|
13
|
Wang Y, Wen W, Yi Y, Zhang Z, Lubet RA, You M. Preventive effects of bexarotene and budesonide in a genetically engineered mouse model of small cell lung cancer. Cancer Prev Res (Phila) 2009; 2:1059-64. [PMID: 19934342 PMCID: PMC6001362 DOI: 10.1158/1940-6207.capr-09-0221] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In the present study, we examined the effect of bexarotene (Targretin) and budesonide in the chemoprevention of small cell lung carcinoma using a lung-specific knockout model of Rb1 and p53. Upon treatment with bexarotene, tumor incidence, number, and load were significantly reduced (P < 0.05). Budesonide treatment trended to inhibition, but the effect was not statistically significant (P > 0.05). Immunohistochemical staining indicated that bexarotene treatment decreased cell proliferation and increased apoptosis in tumors. The Rb1/p53 gene-targeted mouse seems to be a valuable model for chemopreventive studies on human small cell lung cancer. Our results indicate that the retinoid X receptor agonist bexarotene may be a potent chemopreventive agent in this cancer type.
Collapse
Affiliation(s)
- Yian Wang
- Department of Surgery and The Alvin J. Siteman Cancer Center and Division of Comparative Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Weidong Wen
- Department of Surgery and The Alvin J. Siteman Cancer Center and Division of Comparative Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Yijun Yi
- Department of Surgery and The Alvin J. Siteman Cancer Center and Division of Comparative Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Zhongqiu Zhang
- Department of Surgery and The Alvin J. Siteman Cancer Center and Division of Comparative Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Ronald A. Lubet
- Department of Pathology and Chemoprevention Agent Development Research Group, National Cancer Institute, Rockville, Maryland
| | - Ming You
- Department of Surgery and The Alvin J. Siteman Cancer Center and Division of Comparative Medicine, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
14
|
Li M, Chen F, Liu CP, Li DM, Li X, Wang C, Li JC. Dexamethasone enhances trichosanthin-induced apoptosis in the HepG2 hepatoma cell line. Life Sci 2009; 86:10-6. [PMID: 19891978 DOI: 10.1016/j.lfs.2009.10.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Revised: 10/11/2009] [Accepted: 10/16/2009] [Indexed: 11/18/2022]
Abstract
AIMS Trichosanthin (TCS) is a type I ribosome-inactivating protein (RIP) with antitumor activities for various cancers. In this paper, we aimed to investigate whether dexamethasone, an important synthetic member of the glucocorticoid steroids, in combination with TCS can be a potential therapy in treating hepatoma. MAIN METHODS Cell viability was investigated using MTT assay, and apoptosis was evaluated with Hoechst 33258 staining. Western blot analysis was used to examine the changes in the expression levels of IkappaB-alpha, NF-kappaB p65 subunit and Cox-2. Additionally, we took advantage of dominant-negative IkappaB (IkappaB-DM) over-expression and chemical inhibitor PDTC to inhibit NF-kappaB activation. KEY FINDINGS Our results demonstrated that dexamethasone could enhance TCS-induced apoptosis in the hepatoma cell line HepG2, decreasing IC50 values from in excess of 200microg/ml to 50microg/ml. In addition, our results demonstrated that TCS could induce rapid degradation of IkappaB-alpha, nuclear translocation of NF-kappaB and decrease of COX-2 expression in HepG2 cells. Inhibition of NF-kappaB by biological (IkappaB-DM) or chemical inhibitor (PDTC) increased HepG2 cells' sensitivity to TCS, resulting in cell viability rate decreasing and apoptotic rate increasing. Simultaneously, dexamethasone increased the level of IkappaB-alpha protein and effectively inhibited TCS-induced degradation of IkappaB-alpha. SIGNIFICANCE These results suggest that dexamethasone could enhance trichosanthin-induced apoptosis in the HepG2, at least in part, by inhibiting the NF-kappaB signaling pathway and thus strengthening the antitumor effects of TCS, which highlights the possibility of combined drug application of TCS and dexamethasone in the clinical treatment of hepatoma.
Collapse
Affiliation(s)
- Meng Li
- Institute of Cell Biology, Zhejiang University Medical School, Hangzhou 310058, China
| | | | | | | | | | | | | |
Collapse
|
15
|
Stuhr LEB, Salnikov AV, Iversen VV, Salvesen G, Rubin K, Reed RK. High‐dose, short‐term, anti‐inflammatory treatment with dexamethasone reduces growth and augments the effects of 5‐fluorouracil on dimethyl‐α‐benzanthracene‐induced mammary tumors in rats. Scandinavian Journal of Clinical and Laboratory Investigation 2009; 66:477-86. [PMID: 17000555 DOI: 10.1080/00365510600788332] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OBJECTIVE To evaluate the effects of dexamethasone (DXM) alone or in combination with 5-fluorouracil (5-FU) on dimethyl-alpha-benzanthracene (DMBA)-induced mammary tumors in rats. MATERIAL AND METHODS Female Sprague-Dawley rats were divided into 4 groups receiving: 1) saline (controls), 2) DXM (3 mg/kg), 3) 5-FU (1.5 mg/kg) and 4) DXM and 5-FU combined. The drugs were given i.p. every day for 4 days. Interstitial fluid pressure (Pif) and tumor growth were determined in all tumors on days 1, 5 and 7 using the "wick-in-the needle" technique and by external size measurements, respectively. Vessel density and inflammatory cell infiltration of tumor tissue were analyzed by immunohistochemistry. RESULTS DXM treatment significantly retarded tumor growth and reduced Pif. Treatment with a combination of DXM and 5-FU reduced tumor size significantly more than any of the agents alone (p<0.01-0.001). Enhanced uptake of 5-FU by DXM treatment was demonstrated by microdialysis. There were no differences in the density of CD31-positive vessels after DXM or 5-FU treatment, but inflammatory cell infiltration of tumor tissue was significantly reduced after DXM treatment. CONCLUSIONS Our data suggest that DXM may be beneficial as an adjuvant to chemotherapy in the treatment of mammary cancer by increasing the uptake of 5-FU in the tumor.
Collapse
Affiliation(s)
- L E B Stuhr
- Department of Biomedicine, University of Bergen, Bergen, Norway.
| | | | | | | | | | | |
Collapse
|
16
|
Békási S, Zalatnai A. Overexpression of glucocorticoid receptor in human pancreatic cancer and in xenografts. An immunohistochemical study. Pathol Oncol Res 2009; 15:561-6. [PMID: 19253003 DOI: 10.1007/s12253-009-9154-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Accepted: 02/12/2009] [Indexed: 12/31/2022]
Abstract
Glucocorticoid receptor overexpression has been reported in a variety of human solid tumors, but much less in known about its presence in pancreatic cancer. Only one report is available in the literature, back to 1994, since that no peculiar attention has been paid to this issue. Immunohistochemical analysis of paraffin-embedded tissue sections was performed in human normal pancreata and well differentiated pancreatic adenocarcinomas (monoclonal primary antibody, ABCAM, Cambridge, UK). As positive control invasive ductal adenocarcinoma of the breast was used. In the normal non-tumorous pancreas a strong positivity was detected in all acinar cells, typically in the cytoplasm. Nuclear staining was not visible. The distribution of the positive reaction was homogenous. The ductal pancreatic carcinoma cells also displayed a strong positivity. The location of the immune reaction was mainly cytoplasmic but in some tumors a strong nuclear reaction was also noticed. In some slides acini remained also positive in the close vicinity of the tumor. Although the positivity of the ductal tumor cells was a constant finding in our samples, surprisingly, the liver metastasis was completely negative. Strong glucocorticoid receptor expression was also found in xenografted human pancreatic cancer showing a diffuse, mainly cytoplasmic positivity. Our studies have shown that the human pancreatic carcinomas do overexpress a strong glucocorticoid receptor positivity, but its significance is not clear. However, this finding might have a clinical relevance.
Collapse
Affiliation(s)
- Sándor Békási
- First Department of Pathology and Experimental Cancer Research, Faculty of Medicine, Semmelweis University, Ulloi út 26, 1085, Budapest, Hungary
| | | |
Collapse
|
17
|
Herr I, Büchler MW, Mattern J. Glucocorticoid-mediated apoptosis resistance of solid tumors. Results Probl Cell Differ 2009; 49:191-218. [PMID: 19132324 DOI: 10.1007/400_2008_20] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
More than a quarter of a century ago, the phenomenon of glucocorticoid-induced apoptosis in the majority of hematological cells was first recognized. More recently, glucocorticoid-induced antiapoptotic signaling associated with apoptosis resistance towards cytotoxic therapy has been identified in cells of epithelial origin, most of malignant solid tumors and some other tissues. Despite these huge amounts of data demonstrating differential pro- and anti-apoptotic effects of glucocorticoids, the underlying mechanisms of cell type-specific glucocorticoid signaling are just beginning to be described. This review summarizes our present understanding of cell type-specific pro- and anti-apoptotic signaling induced by glucocorticoids. We shortly introduce mechanisms of glucocorticoid resistance of hematological cells. We highlight and discuss the emerging molecular evidence of a general induction of survival signaling in epithelial cells and carcinoma cells by glucocorticoids. We give a summary of our current knowledge of decreased proliferation rates in response to glucocorticoid pre- and combination treatment, which are suspicious to be involved not only in protection of normal tissues, but also in protection of solid tumors from cytotoxic effects of anticancer agents.
Collapse
Affiliation(s)
- Ingrid Herr
- Department of Surgery, University of Heidelberg, Germany.
| | | | | |
Collapse
|
18
|
Rice L, Waters CE, Eccles J, Garside H, Sommer P, Kay P, Blackhall FH, Zeef L, Telfer B, Stratford I, Clarke R, Singh D, Stevens A, White A, Ray DW. Identification and functional analysis of SKA2 interaction with the glucocorticoid receptor. J Endocrinol 2008; 198:499-509. [PMID: 18583474 PMCID: PMC2518725 DOI: 10.1677/joe-08-0019] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Glucocorticoid (GC) receptors (GRs) have profound anti-survival effects on human small cell lung cancer (SCLC). To explore the basis of these effects, protein partners for GRs were sought using a yeast two-hybrid screen. We discovered a novel gene, FAM33A, subsequently identified as a SKA1 partner and involved in mitosis, and so renamed Ska2. We produced an anti-peptide antibody that specifically recognized full-length human SKA2 to measure expression in human cell lines and tissues. There was a wide variation in expression across multiple cell lines, but none was detected in the liver cell line HepG2. A xenograft model of human SCLC had intense staining and archival tissue revealed SKA2 in several human lung and breast tumours. SKA2 was found in the cytoplasm, where it co-localized with GR, but nuclear expression of SKA2 was seen in breast tumours. SKA2 overexpression increased GC transactivation in HepG2 cells while SKA2 knockdown in A549 human lung epithelial cells decreased transactivation and prevented dexamethasone inhibition of proliferation. GC treatment decreased SKA2 protein levels in A549 cells, as did Staurosporine, phorbol ester and trichostatin A; all agents that inhibit cell proliferation. Overexpression of SKA2 potentiated the proliferative response to IGF-I exposure, and knockdown with shRNA caused cells to arrest in mitosis. SKA2 has recently been identified in HeLa S3 cells as part of a complex, which is critical for spindle checkpoint silencing and exit from mitosis. Our new data show involvement in cell proliferation and GC signalling, with implications for understanding how GCs impact on cell fate.
Collapse
Affiliation(s)
- Lisa Rice
- Faculty of Medical and Human Sciences, School of Medicine, University of ManchesterManchester M13 9PTUK
- Faculty of Life Sciences, University of ManchesterManchester M13 9PTUK
| | - Charlotte E Waters
- Faculty of Medical and Human Sciences, School of Medicine, University of ManchesterManchester M13 9PTUK
| | - Jennifer Eccles
- Faculty of Medical and Human Sciences, School of Medicine, University of ManchesterManchester M13 9PTUK
| | - Helen Garside
- Faculty of Medical and Human Sciences, School of Medicine, University of ManchesterManchester M13 9PTUK
| | - Paula Sommer
- Faculty of Medical and Human Sciences, School of Medicine, University of ManchesterManchester M13 9PTUK
| | - Paul Kay
- Faculty of Medical and Human Sciences, School of Medicine, University of ManchesterManchester M13 9PTUK
| | - Fiona H Blackhall
- Faculty of Medical and Human Sciences, School of Medicine, University of ManchesterManchester M13 9PTUK
| | - Leo Zeef
- Faculty of Life Sciences, University of ManchesterManchester M13 9PTUK
| | - Brian Telfer
- Faculty of Medical and Human Sciences, School of Pharmacy, University of ManchesterManchester M13 9PTUK
| | - Ian Stratford
- Faculty of Medical and Human Sciences, School of Pharmacy, University of ManchesterManchester M13 9PTUK
| | - Rob Clarke
- Faculty of Medical and Human Sciences, School of Medicine, University of ManchesterManchester M13 9PTUK
| | - Dave Singh
- Faculty of Medical and Human Sciences, School of Medicine, University of ManchesterManchester M13 9PTUK
| | - Adam Stevens
- Faculty of Medical and Human Sciences, School of Medicine, University of ManchesterManchester M13 9PTUK
| | - Anne White
- Faculty of Medical and Human Sciences, School of Medicine, University of ManchesterManchester M13 9PTUK
- Faculty of Life Sciences, University of ManchesterManchester M13 9PTUK
- (Correspondence should be addressed to D Ray, Centre for Molecular Medicine, and Endocrine Sciences Research Group, Faculty of Medical and Human Sciences, University of Manchester, Stopford Building, Manchester M13 9PT, UK; )
| | - David W Ray
- Faculty of Medical and Human Sciences, School of Medicine, University of ManchesterManchester M13 9PTUK
| |
Collapse
|
19
|
Sommer P, Le Rouzic P, Gillingham H, Berry A, Kayahara M, Huynh T, White A, Ray DW. Glucocorticoid receptor overexpression exerts an antisurvival effect on human small cell lung cancer cells. Oncogene 2007; 26:7111-21. [PMID: 17496926 DOI: 10.1038/sj.onc.1210524] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Small cell lung cancer (SCLC) is an aggressive tumour with an abysmal prognosis. These cancers are characteristically resistant to glucocorticoid (Gc) action, owing to impaired expression of the glucocorticoid receptor (GR). We identified reduced GR expression in human SCLC cell lines, compared to a non-SCLC cell line. The SCLC cells also showed no Gc inhibition of proliferation, in contrast to non-SCLC cells. Retroviral overexpression of GR resulted in significantly increased cell death, which was partially blocked by the GR antagonist, RU486. Indeed, in cells sorted for GR expression, there was rapid, near complete loss of live cells by 72 h, in contrast to control cells that proliferated as expected. Flow cytometry using Annexin V revealed that cell death was by apoptosis. In addition, confocal analysis of fixed cells showed that cells overexpressing GR displayed a significant increase in fragmenting apoptotic nuclei. Microarray studies showed that transgenic GR expression upregulated the proapoptotic genes, BAD and BAX. We have, therefore, identified a profound apoptotic effect of GR in SCLC cells, which may explain the low levels of endogenous GR in SCLC cells. Understanding how GR overexpression leads to apoptotic cell death in SCLC cells may uncover new therapeutic strategies.
Collapse
Affiliation(s)
- P Sommer
- Faculty of Medical and Human Sciences, Department of Medicine, Centre for Molecular Medicine and Endocrine Sciences Research Group, University of Manchester, Manchester, UK
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Zhao X, Teng LZ, Wang ZG, Xin T, Wei SC. Expression and significance of glucocorticoid receptor alpha in meningiomas. J Clin Neurosci 2007; 14:359-63. [PMID: 17236775 DOI: 10.1016/j.jocn.2006.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2005] [Revised: 02/19/2006] [Accepted: 02/22/2006] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To explore the expression of the glucocorticoid receptor alpha (GRalpha) and its significance in the occurrence and progress of meningiomas. MATERIALS AND METHODS By the use of flow cytometry, the proliferative index (PI), S-phase fraction (SPF) and DNA ploidy were detected to evaluate the proliferation of tumor cells in 58 meningioma specimens. The expressions of GRalpha in all meningiomas and seven normal dura samples were studied by means of reverse transcription-polymerase chain reaction to compare the difference in GRalpha between meningiomas and normal dura. The relation between GRalpha and histological grades, PI, SPF and DNA ploidy were also analyzed. RESULTS The mean PI was 9.32%+/-4.41% while the mean SPF was 2.79%+/-2.43% in 58 meningioma specimens. DNA was diploid in 51 cases and aneuploid in the remaining seven cases, with the aneuploid rate being 12.1%. Nine of 58 meningiomas were GRalpha- negative and the rest were GRalpha-positive with a GRalpha-positive rate of 84.5%. The GRalpha-positive meningiomas included 13 '+', 15 '++', 9 '+++' and 12 '++++'. GRalpha was negative in normal dura samples. The GRalpha-positive rate of meningiomas was significantly greater than that of normal dura. There were no significant differences in PI and SPF among GRalpha-negative, GRalpha-weak positive and GRalpha-strong positive meningiomas. The difference in aneuploid rate between GRalpha-positive and GRalpha-negative meningiomas was also not significant. CONCLUSION GRalpha is of significance in meningiomas, which are a target tissue for glucocorticoid. However, GRalpha's expression had no obvious effect on the proliferative activity of meningiomas, so it may not be a major control of this process.
Collapse
Affiliation(s)
- Xu Zhao
- Department of Neurosurgery, The Second Hospital of Shandong University, 247 Beiyuan Road, Jinan, ShanDong, 250033, China.
| | | | | | | | | |
Collapse
|
21
|
Cavallaro G, Maniscalco L, Giammona G, Civiale C, Mazzone M, Enea V. Chemical conjugation of dexamethasone to a polyaspartamide and in vitro evaluation studies. J Drug Deliv Sci Technol 2004. [DOI: 10.1016/s1773-2247(04)50066-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
22
|
Gamarra F, Lingk P, Marmarova A, Edelmann M, Hautmann H, Stepp H, Baumgartner R, Huber RM. 5-Aminolevulinic acid-induced fluorescence in bronchial tumours: dependency on the patterns of tumour invasion. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2004; 73:35-42. [PMID: 14732249 DOI: 10.1016/j.jphotobiol.2003.09.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
5-ALA-induced protoporphyrin IX (PPIX) fluorescence kinetics was quantified by fluorescence microscopy in three-dimensional organ co-cultures of human bronchial epithelium, which were infiltrated by four different lung tumour cell lines (EPLC-M31, LCLC-103H, NCI-H125 and NCI-H841). Corresponding fluorescence measurements were performed in monolayer cultures of these tumour cell lines and BEAS-2B cells as a model for normal bronchial epithelium by flow cytometry. Significant differences of fluorescence intensities (FI) between the tumours were detected in organ co-cultures as well as in single cell measurements. Relative FI values in organ co-cultures (FI(EPLC-32M1)>FI(LCLC-H103)>FI(NCI-H125)>FI(NCI-H841)) did not correspond to the measurements in single cells (FI(LCLC-H103)>FI(NCI-H125)>FI(NCI-H841)>FI(EPLC-32M1)). Histology of organ co-cultures revealed different patterns of invasion and tumour cell densities depending on the tumour type. After correction of FI in the co-cultures to tumour cell density the correlation coefficient for fluorescence values between both models increased considerably. Thus, additionally to distinctive features of 5-ALA metabolism, patterns of tumour invasion may be a factor determining 5-ALA-induced fluorescence. Considering these results, a pronounced heterogeneity of 5-ALA-induced fluorescence might be expected in different bronchial tumours in vivo. This could interfere with the diagnostic reliability of 5-ALA-induced fluorescence for early tumour detection.
Collapse
Affiliation(s)
- F Gamarra
- Medizinische Klinik--Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Ziemssenstr. 1, 80336 München, Germany
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Novak J, Schleman S, Scott J, Balderman VL, Krech L, Kane MA. Dexamethasone regulation of gastrin-releasing peptide receptor in human lung cells. Lung Cancer 2004; 43:17-28. [PMID: 14698533 DOI: 10.1016/j.lungcan.2003.07.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We investigated the effects of the glucocorticoid, dexamethasone (Dex), on expression of the gastrin-releasing peptide (GRP) receptor by human small cell lung carcinoma (SCLC) SHP77 cells. After 12h of 10nM Dex exposure, a six-fold increase in the peak of GRP receptor mRNA compared with untreated controls (10.5+/-4 versus 1.65+/-0.15 attomols/microg total RNA, respectively, P<0.05) occurred. GRP receptor mRNA levels fell to less than 0.5 attomols/microg total RNA after 24h; in Dex-treated cells, these levels rose to 1.2 compared with 0.12 attomols/microg total RNA in the absence of Dex after 7 days. A significant increase (P<0.05) in the GRP receptor-specific binding was also found. Stimulation of SHP77 cell proliferation (25-35% in the presence of 10-100 nM Dex; P<0.0001) was observed after 4-8 days of exposure; this stimulation was inhibited by GRP receptor antagonists. SHP77 cell content and concentration of bombesin-like peptides (BLP) in conditioned medium (approximately 4 nM) was unchanged by Dex. Stimulation of human SCLC SHP77 cell proliferation by Dex may, in part, occur via effects on the GRP autocrine system in these cells.
Collapse
Affiliation(s)
- John Novak
- Section of Medical Oncology, Denver Veterans Affairs Medical Center, University of Colorado Health Sciences Center and University of Colorado Cancer Center, 1055 Clermont Street, Denver, CO 80220, USA
| | | | | | | | | | | |
Collapse
|
24
|
Greenberg AK, Hu J, Basu S, Hay J, Reibman J, Yie TA, Tchou-Wong KM, Rom WN, Lee TC. Glucocorticoids inhibit lung cancer cell growth through both the extracellular signal-related kinase pathway and cell cycle regulators. Am J Respir Cell Mol Biol 2002; 27:320-8. [PMID: 12204894 DOI: 10.1165/rcmb.4710] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Glucocorticoids inhibit the proliferation of various cell types, but the mechanism of this inhibition remains unclear. We investigated the effect of dexamethasone on non-small cell lung cancer cell growth and cell cycle progression. We showed that dexamethasone suppresses the proliferation of A549 and Calu-1 cells, with accumulation of cells in G1/G0 stage of the cell cycle, as determined by fluorescence-activated cell sorter analysis. Western blot analysis confirmed that this is associated with hypophosphorylation of retinoblastoma protein. Using Western blot analysis and in vitro kinase assays, we found that dexamethasone results in decreased activity of CDK2 and 4, decreased levels of cyclin D, E2F, and Myc, and increased levels of the CDK inhibitor p21(Cip1). In addition, we found that dexamethasone decreases activity of extracellular signal-related kinase (ERK)/mitogen-activated protein kinase (MAPK). The kinetics of all these changes indicate that inhibition of the ERK/MAPK pathway precedes the cell cycle effects, suggesting that regulation of this MAPK-signaling pathway may be an alternative mechanism for glucocorticoid-induced cell cycle arrest and growth inhibition.
Collapse
Affiliation(s)
- Alissa K Greenberg
- Department of Medicine, New York University School of Medicine, New York 10016, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Kudawara I, Ueda T, Yoshikawa H, Miyama T, Yamamoto T, Nishizawa Y. In vivo inhibition of tumour growth by dexamethasone in murine osteosarcomas. Eur J Cancer 2001; 37:1703-8. [PMID: 11527699 DOI: 10.1016/s0959-8049(01)00185-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
This study was performed to determine whether glucocorticoid (GC) is an effective inhibitor of tumour growth in murine osteosarcoma (OS) in vivo. The effects of dexamethasone (DEX) on the growth of this tumour were studied in male C3H/He mice. The animals received a dose of 1.25 or 5 microg/g of DEX in 0.1 ml of steroid solution daily intraperitoneally (i.p.) for 14 days. In each DEX-treated group, significant inhibition of the tumour growth curve was seen in a dose- dependent manner compared with the control group (P<0.0001). The percentage of proliferative cell nuclear antigen (PCNA)-positive cells was 22.7% in the 5 microg/g DEX treatment group compared with 67.6% in the control group (P=0.009). Furthermore, mifepristone, a GC receptor antagonist, blocked the inhibition of tumour growth induced by DEX. In the control group, tumour cells showed positive reactivity for nuclear glucocorticoid receptors (GR) by immunohistochemistry. The results of this study indicate that tumour growth inhibition by DEX in murine osteosarcoma may be via GR.
Collapse
Affiliation(s)
- I Kudawara
- Department of Orthopaedic Surgery, Osaka National Hospital, 2-1-14, Hoenzaka, Chuo-ku, 540-0006, Osaka, Japan.
| | | | | | | | | | | |
Collapse
|
26
|
Bergman AM, Pinedo HM, Peters GJ. Steroids affect collateral sensitivity to gemcitabine of multidrug-resistant human lung cancer cells. Eur J Pharmacol 2001; 416:19-24. [PMID: 11282108 DOI: 10.1016/s0014-2999(01)00858-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Gemcitabine is phosphorylated by deoxycytidine kinase and thymidine kinase 2 and during S-phase incorporated into DNA. The steroids cortisol and dexamethasone, which regulate cell proliferation and gene expression, are pumped out of the cell by the membrane efflux pumps P-glycoprotein and multidrug resistance-associated protein (MRP), which are blocked by verapamil. In parental non-small cell lung cancer (NSCLC) cells (SW1573), 5 microM cortisol and 100 nM dexamethasone decreased sensitivity to gemcitabine. However, both cortisol and dexamethasone only decreased sensitivity with verapamil in MRP (2R120) and P-glycoprotein (2R160) overexpressing variants. Cortisol decreased deoxycytidine kinase activity in SW1573 cells and cortisol with verapamil in 2R120 and 2R160 cells. Dexamethasone with verapamil decreased deoxycytidine kinase activity in 2R160. Cortisol decreased thymidine kinase 2 activity in 2R120 and 2R160 cells. Dexamethasone decreased thymidine kinase 2 activity in SW1573, 2R120 and 2R160 cells. In conclusion, since dexamethasone is frequently used to treat side effects of oncolytic therapy, a decrease of sensitivity to gemcitabine by steroids might be clinically relevant.
Collapse
Affiliation(s)
- A M Bergman
- Department of Oncology, VU Medical Center, P.O. Box 7057, 1007 MB, Amsterdam, Netherlands
| | | | | |
Collapse
|
27
|
Santoro JC, Harris G, Sitlani A. Colorimetric detection of glutamine synthetase-catalyzed transferase activity in glucocorticoid-treated skeletal muscle cells. Anal Biochem 2001; 289:18-25. [PMID: 11161290 DOI: 10.1006/abio.2000.4911] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Induction of the enzyme glutamine synthetase (GS) by corticosteroids correlates with muscle wasting and gluconeogenesis, characteristic side effects of chronic glucocorticoid treatment. This highlights the importance of developing robust high-throughput assays to measure drug-induced GS in whole cells. We have optimized a colorimetric method to measure GS-catalyzed gamma-glutamyltransferase (GT) activity in rat L6 skeletal muscle cells (96-well-plate format) and human skeletal muscle cells (24-well-plate format). We observe a fourfold increase in GT activity in dexamethasone treated L6 cells, as compared to untreated cells, with good reproducibility in the measurements (errors of less than 5%). This assay can distinguish between partial agonists such as halopredone acetate and complete agonists such as prednisolone and measure the potency of known glucocorticoid receptor (GR) antagonists like mifepristone. Importantly, the ability of corticosteroids to induce GS-catalyzed GT activity correlates well with their whole cell GR binding potency, indicating a GR-specific effect. Interestingly, in general, induction of GT activity by commonly administered anti-inflammatory corticosteroid drugs is comparable in rat and human skeletal muscle cells, which emphasizes the potential of a rat model system to study GS induction and muscle wasting by these drugs in humans.
Collapse
Affiliation(s)
- J C Santoro
- Department of Molecular Endocrinology, Merck Research Laboratories, Rahway, New Jersey 07065, USA
| | | | | |
Collapse
|
28
|
Honma Y, Yamamoto-Yamaguchi Y, Kanatani Y. Vesnarinone and glucocorticoids cooperatively induce G1 arrest and have an anti-tumour effect on human non-small cell lung carcinoma cells grown in nude mice. Br J Cancer 1999; 80:96-103. [PMID: 10389984 PMCID: PMC2363000 DOI: 10.1038/sj.bjc.6690327] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Vesnarinone, an oral cardiotonic, inhibited the growth of several human non-small cell lung carcinoma cell lines, and its anti-proliferative effects in vitro and in vivo were greatly enhanced by combination with glucocorticoids, but not other steroids. Simultaneous treatment with vesnarinone and dexamethasone is the most effective to evoke the synergistic effect in the growth inhibition of lung carcinoma EBC-1 cells. Dexamethasone and other glucocorticoids induced morphological changes in EBC-1 cells and these agents together with vesnarinone induced alkaline phosphatase activity, which is a typical marker of type II pneumocyte maturation. This treatment arrested the growth of the cells at the G1 phase, indicating that this treatment is cytostatic rather than cytotoxic. These results suggest that vesnarinone plus glucocorticoid might be useful in lung cancer therapy.
Collapse
Affiliation(s)
- Y Honma
- Department of Chemotherapy, Saitama Cancer Center Research Institute, Ina, Japan
| | | | | |
Collapse
|
29
|
Kaiser U, Schilli M, Wegmann B, Barth P, Wedel S, Hofmann J, Havemann K. Expression of vitamin D receptor in lung cancer. J Cancer Res Clin Oncol 1996; 122:356-9. [PMID: 8642046 DOI: 10.1007/bf01220803] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The active metabolite of vitamin D 1,25-dihydroxycholecalciferol is a hormone-like agent that regulates cell differentiation and proliferation. Various vitamin D derivatives have been shown to induce differentiation in neoplastic cells. The prerequisite for any hormone action is the presence of its receptor. We studied the expression of vitamin D receptor in human lung cancer cell lines and in primary lung cancer tissue. Employing the polymerase chain reaction, 10 out of 11 cell lines stemming from small-cell lung cancer and 15 out of 15 cell lines stemming from non-small-cell lung cancer demonstrated vitamin D receptor expression. An immunohistochemical analysis, using a specific monoclonal antibody, demonstrated vitamin D receptor protein expression in 31 out of 117 (26%) primary small-cell lung cancer cases tested. Positive cells exhibited a nuclear reaction pattern. Twenty-one out of 37 primary non-small-cell lung cancer cases, particularly adenocarcinomas (9/14) and squamous-cell carcinomas (10/15), exhibited vitamin D receptor. Results indicate that a subset of lung cancer cases may be susceptible to the differentiating effects of vitamin D analogues.
Collapse
Affiliation(s)
- U Kaiser
- Zentrum Innere Medizin, Abt. Hämatologie/Onkologie, Philipps-Universität Marburg, Germany
| | | | | | | | | | | | | |
Collapse
|