1
|
Kennedy SP, Treacy O, Allott EH, Eustace AJ, Lynam-Lennon N, Buckley N, Robson T. Precision Medicine and Novel Therapeutic Strategies in Detection and Treatment of Cancer: Highlights from the 58th IACR Annual Conference. Cancers (Basel) 2022; 14:6213. [PMID: 36551698 PMCID: PMC9777219 DOI: 10.3390/cancers14246213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Innovation in both detection and treatment of cancer is necessary for the constant improvement in therapeutic strategies, especially in patients with novel or resistant variants of cancer. Cancer mortality rates have declined by almost 30% since 1991, however, depending on the cancer type, acquired resistance can occur to varying degrees. To combat this, researchers are looking towards advancing our understanding of cancer biology, in order to inform early detection, and guide novel therapeutic approaches. Through combination of these approaches, it is believed that a more complete and thorough intervention on cancer can be achieved. Here, we will discuss the advances and approaches in both detection and treatment of cancer, presented at the 58th Irish Association for Cancer Research (IACR) annual conference.
Collapse
Affiliation(s)
- Sean P. Kennedy
- School of Biological, Health and Sports Sciences, Technological University Dublin, D07 ADY7 Dublin, Ireland
| | - Oliver Treacy
- Discipline of Pharmacology and Therapeutics, College of Medicine, Nursing and Health Sciences, University of Galway, H91 TK33 Galway, Ireland
| | - Emma H. Allott
- Patrick G. Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, D08 HD53 Dublin, Ireland
| | - Alex J. Eustace
- National Institute for Cellular Biotechnology, Dublin City University, D09 NR58 Dublin, Ireland
| | - Niamh Lynam-Lennon
- Department of Surgery, Trinity St James’s Cancer Institute, Trinity Translational Medicine Institute, St James’s Hospital, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Niamh Buckley
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK
| | - Tracy Robson
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons Ireland, D02 YN77 Dublin, Ireland
| |
Collapse
|
2
|
Vlachostergios PJ, Karathanasis A, Papandreou CN, Tzortzis V. Early mRNA Expression of Neuroendocrine Differentiation Signals Predicts Recurrence After Radical Prostatectomy: A Transcriptomic Analysis. World J Oncol 2022; 12:232-239. [PMID: 35059083 PMCID: PMC8734499 DOI: 10.14740/wjon1423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 11/30/2021] [Indexed: 12/04/2022] Open
Abstract
Background Neuroendocrine differentiation (NED) of prostate cancer (PC) is a process that often occurs under evolutionary pressure from pharmacologic blockade of androgen receptor signaling at advanced stages of the disease. Identifying a subset of early PC that has a higher likelihood to evolve into this entity is key for developing therapeutic strategies that could more effectively target this phenotype. This study aimed to assess the prognostic relevance of mRNA expression of major players involved in NED of primary prostate tumors. Methods RNA sequencing data from 122 patients with localized PC were analyzed. Transcript levels of key genes involved in NED, with a focus on endothelin axis and nuclear factor kappa B (NF-κB), were assessed and were correlated with time to prostate specific antigen (PSA) recurrence. Copy number alteration of tumor suppressor genes and gene expression of additional signals hallmarking NED was compared between altered and unaltered groups, including lineage determining transcription factors, transcriptional repressors, cell cycle and epigenetic regulators. Results The presence of altered mRNA expression using a z-score threshold of 2 in NFKB1, RELA, EDN1, EDNRA, and EDNRB genes was associated with a higher Gleason score (P < 0.001) and a shorter time to biochemical recurrence (BCR) (P = 0.029). There was a significant direct correlation between NFKB1 and RELA (P < 0.001), NFKB1 and EDNRA (P < 0.001), NFKB1 and EDNRB (P < 0.001), EDNRA and EDNRB expression (P < 0.001). ASCL1 (q < 0.001), ONECUT2 (q < 0.001), DLL3 (q = 0.019), AURKA (q = 0.013), AURKB (q = 0.014), PLK1 (q < 0.001), and EZH2 (q < 0.001) were enriched in patients with tumors harboring alterations in endothelin axis and NF-κB subunit genes whereas REST was downregulated (q < 0.001). Conclusions This analysis suggests that altered mRNA expression of NF-κB and endothelin axis genes in early PC is not only a harbinger of a more aggressive clinical course but is also associated with aberrant gene expression of several transcription factors, transcriptional repressors, cell cycle and epigenetic regulators that are directly involved in NED, in line with their biological roles. This may have implications for closer follow-up and potential use of targeted therapeutic approaches postoperatively in the adjuvant setting to improve outcomes of these patients.
Collapse
Affiliation(s)
- Panagiotis J Vlachostergios
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Athanasios Karathanasis
- Department of Urology, University of Thessaly School of Health Sciences Faculty of Medicine, University Hospital of Larissa, Larissa 41100, Greece
| | - Christos N Papandreou
- Department of Medical Oncology, Faculty of Medicine Papageorgiou Hospital, Aristotle University of Thessaloniki, School of Health Sciences, Thessaloniki 54124, Greece
| | - Vassilios Tzortzis
- Department of Urology, University of Thessaly School of Health Sciences Faculty of Medicine, University Hospital of Larissa, Larissa 41100, Greece
| |
Collapse
|
3
|
Wang GC, Huang TR, Wang KY, Wu ZL, Xie JB, Zhang HL, Yin L, Tang WL, Peng B. Inflammation induced by lipopolysaccharide advanced androgen receptor expression and epithelial-mesenchymal transition progress in prostatitis and prostate cancer. Transl Androl Urol 2022; 10:4275-4287. [PMID: 34984192 PMCID: PMC8661260 DOI: 10.21037/tau-21-964] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/18/2021] [Indexed: 12/15/2022] Open
Abstract
Background To explore the mechanism of prostatic inflammation on prostate cancer (PCa) by comparing the changes of prostate epithelial cells and PCa cells in an inflammatory environment. Methods First, immunohistochemistry (IHC) was used to compare the level of expression of TNF-α, IL-1β, IL-6, and TGF-β between benign prostatic hyperplasia (BPH), prostatitis, and PCa. Then primary prostate epithelial cells were sampled from patients who were suspected of PCa and had histological prostatitis (HP) confirmed by pathological biopsy. Lipopolysaccharide (LPS) or BAY11-7082 were used to investigate the change of androgen receptor (AR) and AR-mediated transcription, epithelial-mesenchymal transition (EMT) in primary prostate epithelial cells, and lymph node carcinoma of the prostate (LNCap) cells. Results TNF-α, IL-1β, IL-6, and TGF-β were significantly increased in HP and PCa compared with those in BPH patients. The proliferation of primary prostate epithelial cells and LNCap cells got the inflection point at LPS 10 µg/mL. In an inflammatory environment with 10 µg/mL LPS, both primary prostate epithelial cell and LNCap cell viability increased, and AR, AR-mediated transcription, and EMT processes were significantly increased. Inhibitors of NF-κB with 10 nM BAY11-7082 decreased AR, AR-mediated transcription, and EMT processes. Conclusions NF-κB regulates AR expression and EMT in prostatitis and PCa, and NF-κB inhibitors may have potential therapeutic value.
Collapse
Affiliation(s)
- Guang-Chun Wang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tian-Run Huang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Urology, Shanghai Traditional Chinese Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ke-Yi Wang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zong-Lin Wu
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jin-Bo Xie
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hou-Liang Zhang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lei Yin
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen-Long Tang
- Department of Urology, People's Hospital of Lincang, Lincang, China
| | - Bo Peng
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Wang V, Geybels MS, Jordahl KM, Gerke T, Hamid A, Penney KL, Markt SC, Freedman M, Pomerantz M, Lee GSM, Rana H, Börnigen D, Rebbeck TR, Huttenhower C, Eeles RA, Stanford JL, Consortium P, Berndt SI, Claessens F, Sørensen KD, Park JY, Vega A, Usmani N, Mucci L, Sweeney CJ. A polymorphism in the promoter of FRAS1 is a candidate SNP associated with metastatic prostate cancer. Prostate 2021; 81:683-693. [PMID: 33956343 PMCID: PMC8491321 DOI: 10.1002/pros.24148] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/19/2021] [Accepted: 04/22/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND Inflammation and one of its mediators, NF-kappa B (NFκB), have been implicated in prostate cancer carcinogenesis. We assessed whether germline polymorphisms associated with NFκB are associated with the risk of developing lethal disease (metastases or death from prostate cancer). METHODS Using a Bayesian approach leveraging NFκB biology with integration of publicly available datasets we used a previously defined genome-wide functional association network specific to NFκB and lethal prostate cancer. A dense-module-searching method identified modules enriched with significant genes from a genome-wide association study (GWAS) study in a discovery data set, Physicians' Health Study and Health Professionals Follow-up Study (PHS/HPFS). The top 48 candidate single nucleotide polymorphisms (SNPs) from the dense-module-searching method were then assessed in an independent prostate cancer cohort and the one SNP reproducibly associated with lethality was tested in a third cohort. Logistic regression models evaluated the association between each SNP and lethal prostate cancer. The candidate SNP was assessed for association with lethal prostate cancer in 6 of 28 studies in the prostate cancer association group to investigate cancer associated alterations in the genome (PRACTICAL) Consortium where there was some medical record review for death ascertainment which also had SNP data from the ONCOARRAY platform. All men self-identified as Caucasian. RESULTS The rs1910301 SNP which was reproducibly associated with lethal disease was nominally associated with lethal disease (odds ratio [OR] = 1.40; p = .02) in the discovery cohort and the minor allele was also associated with lethal disease in two independent cohorts (OR = 1.35; p = .04 and OR = 1.35; p = .07). Fixed effects meta-analysis of all three cohorts found an association: OR = 1.37 (95% confidence interval [CI]: 1.15-1.62, p = .0003). This SNP is in the promoter region of FRAS1, a gene involved in epidermal-basement membrane adhesion and is present at a higher frequency in men with African ancestry. No association was found in the subset of studies from the PRACTICAL consortium studies which had a total of 106 deaths out total of 3263 patients and a median follow-up of 4.4 years. CONCLUSIONS Through its connection with the NFκB pathway, a candidate SNP with a higher frequency in men of African ancestry without cancer was found to be associated with lethal prostate cancer across three well-annotated independent cohorts of Caucasian men.
Collapse
Affiliation(s)
- Victoria Wang
- Department of Biostatistics & Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Milan S Geybels
- Division of Public Health Sciences, Fred Hutchison Cancer Research Center, Seattle, Washington, USA
- Department of Epidemiology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Kristina M Jordahl
- Division of Public Health Sciences, Fred Hutchison Cancer Research Center, Seattle, Washington, USA
| | - Travis Gerke
- Cancer Epidemiology, Moffitt Cancer Center, Tampa, Florida, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Anis Hamid
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Kathryn L Penney
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sarah C Markt
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Matthew Freedman
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Mark Pomerantz
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Gwo-Shu M Lee
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Huma Rana
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Daniela Börnigen
- University Medical Center Hamburg-Eppendorf, Bioinformatics Core, Hamburg, Germany
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Timothy R Rebbeck
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Ros A Eeles
- Oncogenetics, Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Foundation Trust, London, UK
| | - Janet L Stanford
- Division of Public Health Sciences, Fred Hutchison Cancer Research Center, Seattle, Washington, USA
| | - Practical Consortium
- Prostate Cancer Association Group to Investigate Cancer Associated Alterations in the Genome
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Frank Claessens
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Karina D Sørensen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus N, Denmark
| | - Jong Y Park
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Ana Vega
- Fundación Pública Galega Medicina Xenómica, Santiago de Compostela, Spain
| | - Nawaid Usmani
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Lorelei Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Christopher J Sweeney
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Jiang X, Li Y, Feng JL, Nik Nabil WN, Wu R, Lu Y, Liu H, Xi ZC, Xu HX. Safrana l Prevents Prostate Cancer Recurrence by Blocking the Re-activation of Quiescent Cancer Cells via Downregulation of S-Phase Kinase-Associated Protein 2. Front Cell Dev Biol 2021; 8:598620. [PMID: 33392189 PMCID: PMC7772204 DOI: 10.3389/fcell.2020.598620] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/19/2020] [Indexed: 01/01/2023] Open
Abstract
The re-proliferation of quiescent cancer cells is considered to be the primary contributor to prostate cancer (Pca) recurrence and progression. In this study, we investigated the inhibitory effect of safranal, a monoterpene aldehyde isolated from Crocus sativus (saffron), on the re-proliferation of quiescent Pca cells in vitro and in vivo. The results showed that safranal efficiently blocked the re-activation of quiescent Pca cells by downregulating the G0/G1 cell cycle regulatory proteins CDK2, CDK4, CDK6, and phospho-Rb at Ser807/811 and elevating the levels of cyclin-dependent kinase inhibitors, p21 and p27. Further investigation on the underlying mechanisms revealed that safranal suppressed the mRNA and protein expression levels of Skp2, possibly through the deregulation of the transcriptional activity of two major transcriptional factors, E2F1 and NF-κB subunits. Moreover, safranal inhibited AKT phosphorylation at Ser473 and deregulated both canonical and non-canonical NF-κB signaling pathways. Safranal suppressed the tumor growth of quiescent Pca cell xenografts in vivo. Furthermore, safranal-treated tumor tissues exhibited a reduction in Skp2, E2F1, NF-κB p65, p-IκBα (Ser32), c-MYC, p-Rb (Ser807), CDK4, CDK6, and CDK2 and an elevation of p27 and p21 protein levels. Therefore, our findings demonstrate that safranal suppresses cell cycle re-entry of quiescent Pca cells in vitro and in vivo plausibly by repressing the transcriptional activity of two major transcriptional activators of Skp2, namely, E2F1 and NF-κB, through the downregulation of AKT phosphorylation and NF-κB signaling pathways, respectively.
Collapse
Affiliation(s)
- Xue Jiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yang Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ji-Ling Feng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wan Najbah Nik Nabil
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Pharmaceutical Services Program, Ministry of Health, Petaling Jaya, Malaysia
| | - Rong Wu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yue Lu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Liu
- Hospital Management Office, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhi-Chao Xi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hong-Xi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
6
|
NF-κB signaling promotes castration-resistant prostate cancer initiation and progression. Pharmacol Ther 2020; 211:107538. [PMID: 32201312 DOI: 10.1016/j.pharmthera.2020.107538] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/17/2020] [Indexed: 12/14/2022]
Abstract
Prostate Cancer (PCa) is the second leading cause of cancer-related death in men. Adenocarcinoma of the prostate is primarily composed of Androgen Receptor-positive (AR+) luminal cells that require AR transcriptional activity for survival and proliferation. As a consequence, androgen deprivation and anti-androgens are used to treat PCa patients whose disease progresses following attempted surgical or radiation interventions. Unfortunately, patients with advanced PCa can develop incurable castration-resistant PCa (CRPCa) due to mutated, variant, or overexpressed AR. Conversely, low or no AR accumulation or activity can also underlie castration resistance. Whether CRPCa is due to aberrant AR activity or AR independence, NF-κB signaling is also implicated in the initiation and maintenance of CRPCa and, thus, the NF-κB pathway may be a promising alternative therapeutic target. In this review, we present evidence that NF-κB signaling promotes CRPCa initiation and progression, describe the dichotomic role of NF-κB in the regulation of AR expression and activity and outline studies that explore NF-κB inhibitors as PCa therapies.
Collapse
|
7
|
Pollard BS, Suckow MA, Wolter WR, Starr JM, Eidelman O, Dalgard CL, Kumar P, Battacharyya S, Srivastava M, Biswas R, Wilkerson MD, Zhang X, Yang Q, Pollard HB. Digitoxin Inhibits Epithelial-to-Mesenchymal-Transition in Hereditary Castration Resistant Prostate Cancer. Front Oncol 2019; 9:630. [PMID: 31428571 PMCID: PMC6687970 DOI: 10.3389/fonc.2019.00630] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 06/26/2019] [Indexed: 12/13/2022] Open
Abstract
Castration Resistant Prostate Cancer (CRPC) is thought to be driven by a collaborative mechanism between TNFα/NFκB and TGFβ signaling, leading to inflammation, Epithelial-to-Mesenchymal-Transition (EMT), and metastasis. Initially, TGFβ is a tumor suppressor, but in advanced metastatic disease it switches to being a tumor promoter. TGFBR2 may play a critical role in this collaboration, as its expression is driven by NFκB and it is the primary receptor for TGFβ. We have previously reported that the cardenolide drug digitoxin blocks TNFα/NFκB-driven proinflammatory signaling. We therefore hypothesized that digitoxin might break the collaborative process between NFκB and TGFβ by also inhibiting expression of TGFBR2. We therefore tested whether TGFβ-driven EMT and resulting metastases would be suppressed. Here we show, in vitro, that digitoxin inhibits NFκB-driven TGFBR2 expression, as well as Vimentin, while elevating E-cadherin expression. Digitoxin also significantly reduces HSPB1 mRNA and the HSPB1/RBFOX2 mRNA ratio in PC3 cells. In vivo, in a syngeneic, immune competent rat model of metastatic CRPC, we show that digitoxin also suppresses Tgfbr2 expression, as well as expression of other genes classically driven by NFκB, and of multiple EMT genes associated with metastasis. Concurrently, digitoxin suppresses tumor growth and metastasis in these animals, and prolongs survival. Gross tumor recurrence following tumor resection also appears prevented in ca 30% of cases. While the existence of a collaboration between NFκB and TGFβ to drive EMT and metastasis has previously been appreciated, we show here, for the first time, that chronic, low concentrations of digitoxin are able to block CRPC tumor progression, EMT and the ensuing metastatic disease.
Collapse
Affiliation(s)
| | - Mark A Suckow
- Lobund Institute, University of Notre Dame, Notre Dame, IN, United States
| | - William R Wolter
- Lobund Institute, University of Notre Dame, Notre Dame, IN, United States
| | - Joshua M Starr
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine-America's Medical School, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Ofer Eidelman
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine-America's Medical School, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Clifton L Dalgard
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine-America's Medical School, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.,Collaborative Health Initiative Research Program, Bethesda, MD, United States
| | - Parameet Kumar
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine-America's Medical School, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Sharmistha Battacharyya
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine-America's Medical School, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Meera Srivastava
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine-America's Medical School, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.,Collaborative Health Initiative Research Program, Bethesda, MD, United States
| | - Roopa Biswas
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine-America's Medical School, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Matthew D Wilkerson
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine-America's Medical School, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.,Collaborative Health Initiative Research Program, Bethesda, MD, United States
| | - Xijun Zhang
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine-America's Medical School, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.,Collaborative Health Initiative Research Program, Bethesda, MD, United States
| | - Qingfeng Yang
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine-America's Medical School, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Harvey B Pollard
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine-America's Medical School, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.,Collaborative Health Initiative Research Program, Bethesda, MD, United States
| |
Collapse
|
8
|
Wu XX, Yue GGL, Dong JR, Lam CWK, Wong CK, Qiu MH, Lau CBS. Actein Inhibits the Proliferation and Adhesion of Human Breast Cancer Cells and Suppresses Migration in vivo. Front Pharmacol 2018; 9:1466. [PMID: 30618758 PMCID: PMC6299023 DOI: 10.3389/fphar.2018.01466] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 11/30/2018] [Indexed: 12/12/2022] Open
Abstract
Background and purpose: Metastasis is an important cause of death in breast cancer patients. Anti-metastatic agents are urgently needed since standard chemotherapeutics cannot diminish the metastatic rate. Actein, a cycloartane triterpenoid, has been demonstrated to exhibit anti-angiogenic and anti-cancer activities. Its anti-metastatic activity and underlying mechanisms were evaluated in the present study. Methods: The effects of actein on the proliferation, cell cycle phase distribution, migration, motility and adhesion were evaluated using two human breast cancer cell lines, MDA-MB-231 (estrogen receptor-negative) and MCF-7 cells (estrogen receptor-positive) in vitro. Western blots and real-time PCR were employed to examine the protein and mRNA expression of relevant signaling pathways. A human metastatic breast cancer cell xenograft model was established in transparent zebrafish embryos to examine the anti-migration effect of actein in vivo. Results:In vitro results showed that actein treatment significantly decreased cell proliferation, migration and motility. Furthermore, actein significantly caused G1 phase cell cycle arrest and suppressed the protein expression of matrix metalloproteinases of MDA-MB-231 cells. In addition, actein inhibited breast cancer cell adhesion to collagen, also reduced the expression of integrins. Actein treatment down-regulated the protein expression of epidermal growth factor receptor (EGFR), AKT and NF-κB signaling proteins. In vivo results demonstrated that actein (60 μM) significantly decreased the number of zebrafish embryos with migrated cells by 74% and reduced the number of migrated cells in embryos. Conclusion: Actein exhibited anti-proliferative, anti-adhesion and anti-migration activities, with the underlying mechanisms involved the EGFR/AKT and NF-kappaB signalings. These findings shed light for the development of actein as novel anti-migration natural compound for advanced breast cancer.
Collapse
Affiliation(s)
- Xiao-Xiao Wu
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China
| | - Grace Gar-Lee Yue
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China.,State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Jin-Run Dong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - Christopher Wai-Kei Lam
- State Key Laboratory of Quality Research in Chinese Medicines, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, China
| | - Chun-Kwok Wong
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China.,Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China.,State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China.,Li Dak Sum Yip Yio Chin R & D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ming-Hua Qiu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - Clara Bik-San Lau
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China.,State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
9
|
Gerke T, Beltran H, Wang X, Lee GSM, Sboner A, Karnes RJ, Klein EA, Davicioni E, Yousefi K, Ross AE, Börnigen D, Huttenhower C, Mucci LA, Trock BJ, Sweeney CJ. Low Tristetraprolin Expression Is Associated with Lethal Prostate Cancer. Cancer Epidemiol Biomarkers Prev 2018; 28:584-590. [PMID: 30420441 DOI: 10.1158/1055-9965.epi-18-0667] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 09/07/2018] [Accepted: 11/05/2018] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Inflammation is linked to prostate cancer progression and is mediated by NF-κB. Tristetraprolin is a key node of NF-κB activation and we investigated its biological and prognostic role in lethal prostate cancer. METHODS In vitro assays assessed the function of tristetraprolin and the association between low mRNA tristetraprolin levels and lethal prostate cancer (metastatic disease or death) was assessed across independent prostatectomy cohorts: (i) nested case-control studies from Health Professionals Follow-up Study and Physicians' Health Study, and (ii) prostatectomy samples from Cleveland Clinic, Mayo Clinic, Johns Hopkins and Memorial Sloan Kettering Cancer Center. Tristetraprolin expression levels in prostatectomy samples from patients with localized disease and biopsies of metastatic castration-resistant prostate cancer (mCRPC) were assessed in a Cornell University cohort. RESULTS In vitro tristetraprolin expression was inversely associated with NF-κB-controlled genes, proliferation, and enzalutamide sensitivity. Men with localized prostate cancer and lower quartile of tumor tristetraprolin expression had a significant, nearly two-fold higher risk of lethal prostate cancer after adjusting for known clinical and histologic prognostic features (age, RP Gleason score, T-stage). Tristetraprolin expression was also significantly lower in mCRPC compared with localized prostate cancer. CONCLUSIONS Lower levels of tristetraprolin in human prostate cancer prostatectomy tissue are associated with more aggressive prostate cancer and may serve as an actionable prognostic and predictive biomarker. IMPACT There is a clear need for improved biomarkers to identify patients with localized prostate cancer in need of treatment intensification, such as adjuvant testosterone suppression, or treatment de-intensification, such as active surveillance. Tristetraprolin levels may serve as informative biomarkers in localized prostate cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Eric A Klein
- Cleveland Clinic Glickman Urological and Kidney Institute, Cleveland, Ohio
| | | | | | - Ashley E Ross
- James Buchanan Brady Urological Institute, Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Daniela Börnigen
- University Heart Center Hamburg, Clinic for General and Interventional Cardiology, Hamburg, Germany
| | | | - Lorelei A Mucci
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Bruce J Trock
- James Buchanan Brady Urological Institute, Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | |
Collapse
|
10
|
Chemopreventive action of non-steroidal anti-inflammatory drugs in 9,10-dimethylbenzanthracene induced lung carcinogenesis in BALB/C mice: Expression of COX-1, COX-2 and Nf-κB. J Appl Biomed 2018. [DOI: 10.1016/j.jab.2018.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
11
|
To PK, Do MH, Cho YS, Kwon SY, Kim MS, Jung C. Zinc Inhibits Expression of Androgen Receptor to Suppress Growth of Prostate Cancer Cells. Int J Mol Sci 2018; 19:E3062. [PMID: 30297600 PMCID: PMC6213098 DOI: 10.3390/ijms19103062] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/21/2018] [Accepted: 10/01/2018] [Indexed: 12/20/2022] Open
Abstract
The prostate gland contains a high level of intracellular zinc, which is dramatically diminished during prostate cancer (PCa) development. Owing to the unclear role of zinc in this process, therapeutic applications using zinc are limited. This study aimed to clarify the role of zinc and its underlying mechanism in the growth of PCa. ZnCl₂ suppressed the proliferation of androgen receptor (AR)-retaining PCa cells, whereas it did not affect AR-deficient PCa cells. In LNCaP and TRAMP-C2 cells, zinc downregulated the expression of AR in a dose- and time-dependent fashion. Zinc-mediated AR suppression accordingly inhibited the androgen-mediated transactivation and expression of the androgen target, prostate specific antigen (PSA). This phenomenon resulted from facilitated protein degradation, not transcriptional control. In studies using mice bearing TRAMP-C2 subcutaneous tumors, the intraperitoneal injection of zinc significantly reduced tumor size. Analyses of both xenograft tumors and normal prostates showed reduced expression of AR and increased cell death. Considering the significant loss of intracellular zinc and the dominant growth-modulating role of AR during PCa development, loss of zinc may be a critical step in the transformation of normal cells to cancer cells. This study provides the underlying mechanism by which zinc functions as a PCa suppressor, and forms the foundation for developing zinc-mediated therapeutics for PCa.
Collapse
Affiliation(s)
- Phuong Kim To
- Department of Anatomy, Chonnam National University Medical School, Gwangju 61469, Korea.
| | - Manh-Hung Do
- Department of Anatomy, Chonnam National University Medical School, Gwangju 61469, Korea.
| | - Young-Suk Cho
- Department of Anatomy, Chonnam National University Medical School, Gwangju 61469, Korea.
| | - Se-Young Kwon
- Department of Anatomy, Chonnam National University Medical School, Gwangju 61469, Korea.
| | - Min Soo Kim
- Department of Statistics, College of Natural Sciences, Chonnam National University, Gwangju 61186, Korea.
| | - Chaeyong Jung
- Department of Anatomy, Chonnam National University Medical School, Gwangju 61469, Korea.
| |
Collapse
|
12
|
Morel KL, Ormsby RJ, Solly EL, Tran LNK, Sweeney CJ, Klebe S, Cordes N, Sykes PJ. Chronic low dose ethanol induces an aggressive metastatic phenotype in TRAMP mice, which is counteracted by parthenolide. Clin Exp Metastasis 2018; 35:649-661. [PMID: 29936575 DOI: 10.1007/s10585-018-9915-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 06/18/2018] [Indexed: 11/29/2022]
Abstract
Despite advances in prostate cancer therapy, dissemination and growth of metastases results in shortened survival. Here we examined the potential anti-cancer effect of the NF-κB inhibitor parthenolide (PTL) and its water soluble analogue dimethylaminoparthenolide (DMAPT) on tumour progression and metastasis in the TRansgenic Adenocarcinoma of the Mouse Prostate (TRAMP) model of prostate cancer. Six-week-old male TRAMP mice received PTL (40 mg/kg in 10% ethanol/saline), DMAPT (100 mg/kg in sterile water), or vehicle controls by oral gavage thrice weekly until palpable tumour formation. DMAPT treatment slowed normal tumour development in TRAMP mice, extending the time-to-palpable prostate tumour by 20%. PTL did not slow overall tumour development, while the ethanol/saline vehicle used to administer PTL unexpectedly induced an aggressive metastatic tumour phenotype. Chronic ethanol/saline vehicle upregulated expression of NF-κB, MMP2, integrin β1, collagen IV, and laminin, and induced vascular basement membrane degradation in primary prostate tumours, as well as increased metastatic spread to the lung and liver. All of these changes were largely prevented by co-administration with PTL. DMAPT (in water) reduced metastasis to below that of water-control. These data suggest that DMAPT has the potential to be used as a cancer preventive and anti-metastatic therapy for prostate cancer. Although low levels of ethanol consumption have not been shown to strongly correlate with prostate cancer epidemiology, these results would support a potential effect of chronic low dose ethanol on metastasis and the TRAMP model provides a useful system in which to further explore the mechanisms involved.
Collapse
Affiliation(s)
- Katherine L Morel
- Molecular Medicine and Pathology, Flinders Centre for Innovation in Cancer, Flinders University and Medical Centre, Bedford Park, Adelaide, SA, Australia.
| | - Rebecca J Ormsby
- Molecular Medicine and Pathology, Flinders Centre for Innovation in Cancer, Flinders University and Medical Centre, Bedford Park, Adelaide, SA, Australia
| | - Emma L Solly
- Molecular Medicine and Pathology, Flinders Centre for Innovation in Cancer, Flinders University and Medical Centre, Bedford Park, Adelaide, SA, Australia
| | - Linh N K Tran
- Molecular Medicine and Pathology, Flinders Centre for Innovation in Cancer, Flinders University and Medical Centre, Bedford Park, Adelaide, SA, Australia
| | | | - Sonja Klebe
- Department of Anatomical Pathology, Flinders University and SA Pathology at Flinders Medical Centre, Bedford Park, SA, Australia
| | - Nils Cordes
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden; Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pamela J Sykes
- Molecular Medicine and Pathology, Flinders Centre for Innovation in Cancer, Flinders University and Medical Centre, Bedford Park, Adelaide, SA, Australia
| |
Collapse
|
13
|
Khurana N, Sikka SC. Targeting Crosstalk between Nrf-2, NF-κB and Androgen Receptor Signaling in Prostate Cancer. Cancers (Basel) 2018; 10:cancers10100352. [PMID: 30257470 PMCID: PMC6210752 DOI: 10.3390/cancers10100352] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/12/2018] [Accepted: 09/19/2018] [Indexed: 01/02/2023] Open
Abstract
Oxidative stress, inflammation and androgen receptor (AR) signaling play a pivotal role in the initiation, development and progression of prostate cancer (PCa). Numerous papers in the literature have documented the interconnection between oxidative stress and inflammation; and how antioxidants can combat the inflammation. It has been shown in the literature that both oxidative stress and inflammation regulate AR, the key receptor involved in the transition of PCa to castration resistant prostate cancer (CRPC). In this review, we discuss about the importance of targeting Nrf-2-antioxidant signaling, NF-κB inflammatory response and AR signaling in PCa. Finally, we discuss about the crosstalk between these three critical pathways as well as how the anti-inflammatory antioxidant phytochemicals like sulforaphane (SFN) and curcumin (CUR), which can also target AR, can be ideal candidates in the chemoprevention of PCa.
Collapse
Affiliation(s)
- Namrata Khurana
- Department of Internal Medicine-Medical Oncology, Washington University in St. Louis Medical Campus, 660 S Euclid Ave, St. Louis, MO 63110-1010, USA.
| | - Suresh C Sikka
- Department of Urology, Tulane University School of Medicine,1430 Tulane Avenue, New Orleans, LA 70112, USA.
| |
Collapse
|
14
|
Durairajan S, Jebaraj Walter CE, Samuel MD, Palani D, G DJD, C GPD, Pasupati S, Johnson T. Differential expression of NF-κB heterodimer RelA/p50 in human urothelial carcinoma. PeerJ 2018; 6:e5563. [PMID: 30225173 PMCID: PMC6139250 DOI: 10.7717/peerj.5563] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 08/11/2018] [Indexed: 12/30/2022] Open
Abstract
Background Urothelial carcinoma (UC) is the fifth most common malignancy that accounts for 5% of all cancers. Diagnostic markers that predict UC progressions are inadequate. NF-κB contributes towards disease progression upon constitutive activation in many solid tumors. The nuclear localization of NF-κB indicates increased transcriptional activity while cytoplasmic localization indicates the inactive protein repository that can be utilized readily by a malignant cell. This study delineates the nuclear and cytoplasmic differential expression of NF-κB heterodimers in UC progression. Methods The involvement of the NF-κB proteins in UC was analyzed in silico using cytoscape. The expression of NF-κB heterodimers was analyzed by immunohistochemistry. Results PINA4MS app in cytoscape revealed over expression of RelA and suppression of NF-κB1 (p50 precursor) in UC whereas the expression of NF-κB target proteins remained unhindered. Immunohistochemical localization showed nuclear RelA/p50 in low grade UC whereas in high grade only RelA expression was observed. Conversely, cytoplasmic expression of RelA/p50 remained extensive across high and low grade UC tissues (p < 0.005). RelA nuclear and cytoplasmic expression (p < 0.005) was directly proportional to the disease progression. In our study, some of the high-grade UC tissues with squamous differentiation and muscle invasion had extensive nuclear p50 localization. The phenomenon of RelA/p50 expression seen increased in low-grade UC than high grade UC might be due to their interaction with other members of NF-κB family of proteins. Thus, NF-κB RelA/p50 differential expression may play a unique role in UC pathogenesis and can serve as a biomarker for diagnosis.
Collapse
Affiliation(s)
- Sankari Durairajan
- Department of Biotechnology, Sri Ramachandra Medical College and Research Institute, Chennai, India
| | | | - Mary Divya Samuel
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (formerly Manipal University), Manipal, India
| | - Dinesh Palani
- Department of Biotechnology, Sri Ramachandra Medical College and Research Institute, Chennai, India
| | - Dicky John Davis G
- Department of Bioinformatics, Sri Ramachandra Medical College and Research Institute, Chennai, India
| | - George Priya Doss C
- Department of Integrative Biology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamilnadu, India
| | - Sneha Pasupati
- Department of Integrative Biology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamilnadu, India
| | - Thanka Johnson
- Department of Pathology, Sri Ramachandra Medical College and Research Institute, Chennai, India
| |
Collapse
|
15
|
Mendonca MS, Turchan WT, Alpuche ME, Watson CN, Estabrook NC, Chin-Sinex H, Shapiro JB, Imasuen-Williams IE, Rangel G, Gilley DP, Huda N, Crooks PA, Shapiro RH. DMAPT inhibits NF-κB activity and increases sensitivity of prostate cancer cells to X-rays in vitro and in tumor xenografts in vivo. Free Radic Biol Med 2017; 112:318-326. [PMID: 28782644 PMCID: PMC6322835 DOI: 10.1016/j.freeradbiomed.2017.08.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 07/20/2017] [Accepted: 08/01/2017] [Indexed: 01/22/2023]
Abstract
Constitutive activation of the pro-survival transcription factor NF-κB has been associated with resistance to both chemotherapy and radiation therapy in many human cancers, including prostate cancer. Our lab and others have demonstrated that the natural product parthenolide can inhibit NF-κB activity and sensitize PC-3 prostate cancers cells to X-rays in vitro; however, parthenolide has poor bioavailability in vivo and therefore has little clinical utility in this regard. We show here that treatment of PC-3 and DU145 human prostate cancer cells with dimethylaminoparthenolide (DMAPT), a parthenolide derivative with increased bioavailability, inhibits constitutive and radiation-induced NF-κB binding activity and slows prostate cancer cell growth. We also show that DMAPT increases single and fractionated X-ray-induced killing of prostate cancer cells through inhibition of DNA double strand break repair and also that DMAPT-induced radiosensitization is, at least partially, dependent upon the alteration of intracellular thiol reduction-oxidation chemistry. Finally, we demonstrate that the treatment of PC-3 prostate tumor xenografts with oral DMAPT in addition to radiation therapy significantly decreases tumor growth and results in significantly smaller tumor volumes compared to xenografts treated with either DMAPT or radiation therapy alone, suggesting that DMAPT might have a potential clinical role as a radiosensitizing agent in the treatment of prostate cancer.
Collapse
Affiliation(s)
- Marc S Mendonca
- Departments of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202 USA; Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA.
| | - William T Turchan
- Departments of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202 USA; Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Melanie E Alpuche
- Departments of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Christopher N Watson
- Departments of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202 USA; Richard L. Roudebush, VA Medical Center, Indianapolis, IN 46202 USA
| | - Neil C Estabrook
- Departments of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Helen Chin-Sinex
- Departments of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Jeremy B Shapiro
- Departments of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Imade E Imasuen-Williams
- Departments of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Gabriel Rangel
- Departments of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - David P Gilley
- Department of Chemistry and Applied Sciences, South Dakota School of Mines and Technology, Rapid City, SD 57701 USA
| | - Nazmul Huda
- Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Peter A Crooks
- College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Ronald H Shapiro
- Departments of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202 USA; Richard L. Roudebush, VA Medical Center, Indianapolis, IN 46202 USA
| |
Collapse
|
16
|
Janganati V, Ponder J, Thakkar S, Jordan CT, Crooks PA. Succinamide derivatives of melampomagnolide B and their anti-cancer activities. Bioorg Med Chem 2017; 25:3694-3705. [PMID: 28545815 PMCID: PMC5531864 DOI: 10.1016/j.bmc.2017.05.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 05/01/2017] [Accepted: 05/04/2017] [Indexed: 12/21/2022]
Abstract
A series of succinamide derivatives of melampomagnolide B have been synthesized by coupling MMB monosuccinate (2) with various heterocyclic amines to afford compounds 3a-3l. MMB monosuccinate was also reacted with terminal diaminoalkanes to afford dimeric succinamido analogs of MMB (4a-4h). These succinamide analogs of MMB were evaluated for their anti-cancer activity against a panel of sixty human cancer cell lines. Analogs 3d-3i and dimers 4f-4g exhibited promising anti-cancer activity with GI50 values ranging from 0.28 to 33.5µM against most of the cell lines in the panel. The dimeric analogs 4f and 4g were identified as lead compounds with GI50 values in the nanomolar range (GI50=280-980nM) against several cell lines in the panel; i.e. leukemia cell lines CCRF-CEM, HL-60(TB), K-562, MOLT-4, RPMI-8226 and SR; and solid tumor cell lines NCI-H522 (non-small cell lung cancer), SW-620 and HCT-116 (colon cancer), LOX IMVI (melanoma), RXF 393 (renal cancer), and MCF7, BT-549 and MDA-MB-468 (breast cancer). Succinamide analogs 3a, 3c-3l and 4b-4h were also evaluated for their apoptotic activity against M9-ENL1 acute myelogenous leukemia cells; compounds 3h-3j and 4g were equipotent with parthenolide, exhibiting LC50 values in the range 4.1-8.1μM. Molecular docking studies indicate that these molecules interact covalently with the highly conserved Cys-46 residue of the N-terminal lobe (1-109) of human IKKβ to inhibit the NFκB transcription factor complex, resulting in down-regulation of anti-apoptotic genes under NFκB control.
Collapse
Affiliation(s)
- Venumadhav Janganati
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jessica Ponder
- Department of Toxicology, University of Colorado, Aurora, CO 80045, USA
| | - Shraddha Thakkar
- National Center for Toxicological Research, Jefferson, AR 72079, USA
| | - Craig T Jordan
- Division of Hematology, University of Colorado, Aurora, CO 80045, USA
| | - Peter A Crooks
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
17
|
Bommagani S, Ponder J, Penthala NR, Janganati V, Jordan CT, Borrelli MJ, Crooks PA. Indole carboxylic acid esters of melampomagnolide B are potent anticancer agents against both hematological and solid tumor cells. Eur J Med Chem 2017; 136:393-405. [PMID: 28525840 DOI: 10.1016/j.ejmech.2017.05.031] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/08/2017] [Accepted: 05/10/2017] [Indexed: 01/08/2023]
Abstract
A series of novel, heteroaryl carboxylic acid conjugates of the sesquiterpene melampomagnolide-B (MMB, 3) has been evaluated as antitumor agents against an NCI panel of 64 human hematopoetic and solid tumor cell lines. The indole-3-acrylic acid conjugate 7j and the indole-3-carboxylic acid conjugate 7k were found to be the most potent analogs in the series. Compounds 7j and 7k exhibited remarkable growth inhibition, with GI50 values in the range 0.03-0.30 μM and 0.04-0.28 μM, respectively, against the cell lines in the leukemia sub-panel, and GI50 values of 0.05-0.40 μM and 0.04-0.61 μM, respectively, against 90% of the solid tumor cell lines in the NCI panel. Compound 7a was particularly effective against the sub-panel of breast cancer cell lines with GI50 values in the range <0.01-0.30 μM. Compounds 7j, 7a and its water soluble analog 7p also exhibited potent anticancer activity against rat 9L-SF gliosarcoma cells in culture. Compound 7j was the most potent compound in the series in the M9-ENL1 AML cell assay with a lethal dose concentration EC50 value of 720 nM, and exhibited the greatest cytotoxicity against a collection of primary AML stem cell specimens, which included a specimen that was unresponsive to PTL, affording EC50 values in the range 0.33-1.0 μM in three out of four specimens. The results from this study provide further evidence that analogs of the sesquiterpene MMB can be designed to afford molecules with significantly improved anticancer activity. Thus, both 7j and 7k are considered potential lead molecules in the search for new anticancer agents that can be used as treatments for both hematopoetic and solid tumors.
Collapse
Affiliation(s)
- Shobanbabu Bommagani
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jessica Ponder
- Division of Hematology, University of Colorado, Aurora, CO 80045, USA
| | - Narsimha R Penthala
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Venumadhav Janganati
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Craig T Jordan
- Division of Hematology, University of Colorado, Aurora, CO 80045, USA
| | - Michael J Borrelli
- Department of Radiology and Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Peter A Crooks
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
18
|
Shankar E, Zhang A, Franco D, Gupta S. Betulinic Acid-Mediated Apoptosis in Human Prostate Cancer Cells Involves p53 and Nuclear Factor-Kappa B (NF-κB) Pathways. Molecules 2017; 22:molecules22020264. [PMID: 28208611 PMCID: PMC5832059 DOI: 10.3390/molecules22020264] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 02/06/2017] [Accepted: 02/08/2017] [Indexed: 12/26/2022] Open
Abstract
Defects in p53 and nuclear factor-kappa B (NF-κB) signaling pathways are frequently observed in the initiation and development of various human malignancies, including prostate cancer. Clinical studies demonstrate higher expression of NF-κB/p65/RelA, NF-κB/p50/RelB, and cRel as well as downregulation of the p53 network in primary prostate cancer specimens and in metastatic tumors. Betulinic acid (BA), is a triterpenoid that has been reported to be an effective inducer of apoptosis through modification of several signaling pathways. Our objective was to investigate the pathways involved in BA-induced apoptosis in human prostate cancer cells. We employed the androgen-responsive LNCaP cells harboring wild-type p53, and androgen-refractory DU145 cells possessing mutated p53 with high constitutive NF-κB activity. Inhibition of cell survival by BA at 10 and 20 µM concentrations occurred as a result of alteration in Bax/Bcl-2 ratio in both cell lines that led to an increased cytochrome C release, caspase activation and poly(ADP)ribose polymerase (PARP) cleavage, leading to apoptosis. BA treatment resulted in stabilization of p53 through increase in phosphorylation at Ser15 in LNCaP cells, but not in DU145 cells, and induction of cyclin kinase inhibitor p21/Waf1 in both cell types. Furthermore, treatment of both prostate cancer cells with BA decreased the phosphorylation of IκB kinase (IKK)α and I-kappa-B-alpha (IκBα) inhibiting the nuclear location of NF-κB/p65 causing cytosolic accumulation and resulting in its decreased nuclear binding. We demonstrate that BA may induce apoptosis by stabilizing p53 and downregulating NF-κB pathway in human prostate cancer cells, irrespective of the androgen association, and therefore can potentially be developed as a molecule of interest in cancer chemoprevention.
Collapse
Affiliation(s)
- Eswar Shankar
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA.
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA.
| | - Ailin Zhang
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA.
| | - Daniel Franco
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA.
| | - Sanjay Gupta
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA.
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA.
- Department of Urology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA.
- Department of Nutrition, Case Western Reserve University, Cleveland, OH 44106, USA.
- Division of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA.
| |
Collapse
|
19
|
Yu FS, Lin ML, Hsu SC, Yu CC, Huang YP, Kuo YH, Chung JG. 4-Hydroxybutenolide impairs cell migration, and invasion of human oral cancer SCC-4 cells via the inhibition of NF-κB and MAPK signaling pathways. Int J Oncol 2016; 49:579-88. [DOI: 10.3892/ijo.2016.3537] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 04/27/2016] [Indexed: 11/05/2022] Open
|
20
|
Börnigen D, Tyekucheva S, Wang X, Rider JR, Lee GS, Mucci LA, Sweeney C, Huttenhower C. Computational Reconstruction of NFκB Pathway Interaction Mechanisms during Prostate Cancer. PLoS Comput Biol 2016; 12:e1004820. [PMID: 27078000 PMCID: PMC4831844 DOI: 10.1371/journal.pcbi.1004820] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 02/19/2016] [Indexed: 12/21/2022] Open
Abstract
Molecular research in cancer is one of the largest areas of bioinformatic investigation, but it remains a challenge to understand biomolecular mechanisms in cancer-related pathways from high-throughput genomic data. This includes the Nuclear-factor-kappa-B (NFκB) pathway, which is central to the inflammatory response and cell proliferation in prostate cancer development and progression. Despite close scrutiny and a deep understanding of many of its members’ biomolecular activities, the current list of pathway members and a systems-level understanding of their interactions remains incomplete. Here, we provide the first steps toward computational reconstruction of interaction mechanisms of the NFκB pathway in prostate cancer. We identified novel roles for ATF3, CXCL2, DUSP5, JUNB, NEDD9, SELE, TRIB1, and ZFP36 in this pathway, in addition to new mechanistic interactions between these genes and 10 known NFκB pathway members. A newly predicted interaction between NEDD9 and ZFP36 in particular was validated by co-immunoprecipitation, as was NEDD9's potential biological role in prostate cancer cell growth regulation. We combined 651 gene expression datasets with 1.4M gene product interactions to predict the inclusion of 40 additional genes in the pathway. Molecular mechanisms of interaction among pathway members were inferred using recent advances in Bayesian data integration to simultaneously provide information specific to biological contexts and individual biomolecular activities, resulting in a total of 112 interactions in the fully reconstructed NFκB pathway: 13 (11%) previously known, 29 (26%) supported by existing literature, and 70 (63%) novel. This method is generalizable to other tissue types, cancers, and organisms, and this new information about the NFκB pathway will allow us to further understand prostate cancer and to develop more effective prevention and treatment strategies. In molecular research in cancer it remains challenging to uncover biomolecular mechanisms in cancer-related pathways from high-throughput genomic data, including the Nuclear-factor-kappa-B (NFκB) pathway. Despite close scrutiny and a deep understanding of many of the NFκB pathway members’ biomolecular activities, the current list of pathway members and a systems-level understanding of their interactions remains incomplete. In this study, we provide the first steps toward computational reconstruction of interaction mechanisms of the NFκB pathway in prostate cancer. We identified novel roles for 8 genes in this pathway and new mechanistic interactions between these genes and 10 known pathway members. We combined 651 gene expression datasets with 1.4M interactions to predict the inclusion of 40 additional genes in the pathway. Molecular mechanisms of interaction were inferred using recent advances in Bayesian data integration to simultaneously provide information specific to biological contexts and individual biomolecular activities, resulting in 112 interactions in the fully reconstructed NFκB pathway. This method is generalizable, and this new information about the NFκB pathway will allow us to further understand prostate cancer.
Collapse
Affiliation(s)
- Daniela Börnigen
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, United States of America.,The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Svitlana Tyekucheva
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Xiaodong Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jennifer R Rider
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, United States of America
| | - Gwo-Shu Lee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, United States of America
| | - Christopher Sweeney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, United States of America.,The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| |
Collapse
|
21
|
Casticin Inhibits A375.S2 Human Melanoma Cell Migration/Invasion through Downregulating NF-κB and Matrix Metalloproteinase-2 and -1. Molecules 2016; 21:384. [PMID: 27007357 PMCID: PMC6274196 DOI: 10.3390/molecules21030384] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 03/15/2016] [Accepted: 03/16/2016] [Indexed: 01/14/2023] Open
Abstract
Casticin is one of the main components from Fructus Viticis, which is widely used as an anti-inflammatory agent. The mechanism of how casticin affects melanoma cell migration and invasion is still not well known. Here we studied the anti-metastasis effects of casticin on A375.S2 melanoma cells by using a non-lethal concentration. First; we used an adhesion assay to test the A375.S2 cells’ adhesion ability after treatment with casticin. We next investigated the cell migration ability after casticin treatment by using a wound healing assay to prove that the migration of A375.S2 cells can be inhibited by casticin and double checked the results using the transwell-migration assay. The suppressive effects on matrix metalloproteinase-2; and -9 (MMP-2; and -9) activities were examined by gelatin zymography. Furthermore, western blotting was used to investigate the protein level changes in A375.S2 cells. We found that p-EGFR; Ras and p-ERK1/2 are decreased by casticin, indicating that casticin can down-regulate the migration and invasion ability of A375.S2 cells via the p-EGFR/Ras/p-ERK pathway. The NF-κB p65 and p-ERK levels in nuclear proteins are also decreased by treatment with casticin. An EMSA assay also discovered that the NF-κB p65 and DNA interaction is decreased. NF-κB p65 protein level was examined by immunofluorescence staining and also decreased. Our findings suggest that casticin has anti-metastatic potential by decreasing the invasiveness of A375.S2 cells. We also found that casticin suppressed A375.S2 cell proliferation and cell adhesion ability, but did not affect cell death, as examined using cytometry and a collagen adhesion assay. Based on these observations, casticin could be used as an inhibitor of migration and invasion of human melanoma cells in the future.
Collapse
|
22
|
Lycopene acts through inhibition of IκB kinase to suppress NF-κB signaling in human prostate and breast cancer cells. Tumour Biol 2016; 37:9375-85. [PMID: 26779636 DOI: 10.1007/s13277-016-4798-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 01/06/2016] [Indexed: 01/03/2023] Open
Abstract
We studied the effect of the potent dietary antioxidant lycopene on multiple points along the nuclear factor kappa B (NF-κB) signaling pathway in prostate and breast cancer cells. Lycopene significantly inhibited prostate and breast cancer cell growth at physiologically relevant concentrations of ≥1.25 μM. Similar concentrations also caused a 30-40 % reduction in inhibitor of kappa B (IκB) phosphorylation in the cells, as determined by western blotting. Furthermore, the same degree of inhibition by lycopene was observed for NF-κB transcriptional activity, as determined by reporter gene assay. Concomitant with this, immunofluorescence staining of lycopene-treated cells showed a significant suppression (≥25 %) of TNF-induced NF-κB p65 subunit nuclear translocation. Further probing of lycopene's effects on upstream elements of the NF-κB pathway showed a 25 % inhibition of both activity of recombinant IκB kinase β (IKKβ) kinase in a cell-free in vitro assay, as well as activity of IKKβ immunoprecipitated from MDA-MB-231 cells treated with lycopene. In conclusion, the anticancer properties of lycopene may occur through inhibition of the NF-κB signaling pathway, beginning at the early stage of cytoplasmic IKK kinase activity, which then leads to reduced NF-κB-responsive gene regulation. Furthermore, these effects in cancer cells were observed at concentrations of lycopene that are relevant and achievable in vivo.
Collapse
|
23
|
Janganati V, Ponder J, Jordan CT, Borrelli MJ, Penthala NR, Crooks PA. Dimers of Melampomagnolide B Exhibit Potent Anticancer Activity against Hematological and Solid Tumor Cells. J Med Chem 2015; 58:8896-906. [PMID: 26540463 DOI: 10.1021/acs.jmedchem.5b01187] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Novel carbamate (7a-7h) and carbonate (7i, 7j, and 8) dimers of melampomagnolide B have been synthesized by reaction of the melampomagnolide-B-triazole carbamate synthon 6 with various terminal diamino- and dihydroxyalkanes. Dimeric carbamate products 7b, 7c, and 7f exhibited potent growth inhibition (GI50 = 0.16-0.99 μM) against the majority of cell lines in the NCI panel of 60 human hematological and solid tumor cell lines. Compound 7f and 8 exhibited anticancer activity that was 300-fold and 1 × 10(6)-fold more cytotoxic than DMAPT, respectively, at a concentration of 10 μM against rat 9L-SF gliosarcoma cells. Compounds 7a-7j and 8 were also screened against M9-ENL1 and acute myelogenous leukemia (AML) primary cell lines and exhibited 2- to 10-fold more potent antileukemic activity against M9-ENL1 cells (EC50 = 0.57-2.90 μM) when compared to parthenolide (EC50 = 6.0) and showed potent antileukemic activity against five primary AML cell lines (EC50 = 0.76-7.3 μM).
Collapse
Affiliation(s)
- Venumadhav Janganati
- Department of Pharmaceutical Sciences, College of Pharmacy and ‡Department of Radiology and Neurology, University of Arkansas for Medical Sciences , Little Rock, Arkansas 72205, United States.,Division of Hematology and §Department of Toxicology, University of Colorado , Aurora, Colorado 80045, United States
| | - Jessica Ponder
- Department of Pharmaceutical Sciences, College of Pharmacy and ‡Department of Radiology and Neurology, University of Arkansas for Medical Sciences , Little Rock, Arkansas 72205, United States.,Division of Hematology and §Department of Toxicology, University of Colorado , Aurora, Colorado 80045, United States
| | - Craig T Jordan
- Department of Pharmaceutical Sciences, College of Pharmacy and ‡Department of Radiology and Neurology, University of Arkansas for Medical Sciences , Little Rock, Arkansas 72205, United States.,Division of Hematology and §Department of Toxicology, University of Colorado , Aurora, Colorado 80045, United States
| | - Michael J Borrelli
- Department of Pharmaceutical Sciences, College of Pharmacy and ‡Department of Radiology and Neurology, University of Arkansas for Medical Sciences , Little Rock, Arkansas 72205, United States.,Division of Hematology and §Department of Toxicology, University of Colorado , Aurora, Colorado 80045, United States
| | - Narsimha Reddy Penthala
- Department of Pharmaceutical Sciences, College of Pharmacy and ‡Department of Radiology and Neurology, University of Arkansas for Medical Sciences , Little Rock, Arkansas 72205, United States.,Division of Hematology and §Department of Toxicology, University of Colorado , Aurora, Colorado 80045, United States
| | - Peter A Crooks
- Department of Pharmaceutical Sciences, College of Pharmacy and ‡Department of Radiology and Neurology, University of Arkansas for Medical Sciences , Little Rock, Arkansas 72205, United States.,Division of Hematology and §Department of Toxicology, University of Colorado , Aurora, Colorado 80045, United States
| |
Collapse
|
24
|
Khazir J, Riley DL, Chashoo G, Mir BA, Liles D, Islam MA, Singh SK, Vishwakarma RA, Pilcher LA. Design, synthesis and anticancer activity of Michael-type thiol adducts of α-santonin analogue with exocyclic methylene. Eur J Med Chem 2015. [DOI: 10.1016/j.ejmech.2015.07.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
25
|
Leung HW, Wang Z, Yue GGL, Zhao SM, Lee JKM, Fung KP, Leung PC, Lau CBS, Tan NH. Cyclopeptide RA-V inhibits cell adhesion and invasion in both estrogen receptor positive and negative breast cancer cells via PI3K/AKT and NF-κB signaling pathways. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1827-40. [DOI: 10.1016/j.bbamcr.2015.04.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 04/21/2015] [Accepted: 04/27/2015] [Indexed: 11/26/2022]
|
26
|
Henning SM, Wang P, Said JW, Huang M, Grogan T, Elashoff D, Carpenter CL, Heber D, Aronson WJ. Randomized clinical trial of brewed green and black tea in men with prostate cancer prior to prostatectomy. Prostate 2015; 75:550-9. [PMID: 25545744 PMCID: PMC4334734 DOI: 10.1002/pros.22943] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 11/17/2014] [Indexed: 12/20/2022]
Abstract
BACKGROUND Preclinical and epidemiologic studies suggest chemopreventive effects of green tea (GT) and black tea (BT) in prostate cancer. In the current study we determined the effect of GT and BT consumption on biomarkers related to prostate cancer development and progression. METHODS In this exploratory, open label, phase II trial 113 men diagnosed with prostate cancer were randomized to consume six cups daily of brewed GT, BT or water (control) prior to radical prostatectomy (RP). The primary endpoint was prostate tumor markers of cancer development and progression determined by tissue immunostaining of proliferation (Ki67), apoptosis (Bcl-2, Bax, Tunel), inflammation (nuclear and cytoplasmic nuclear factor kappa B [NFκB]) and oxidation (8-hydroxydeoxy-guanosine [8OHdG]). Secondary endpoints of urinary oxidation, tea polyphenol uptake in prostate tissue, and serum prostate specific antigen (PSA) were evaluated by high performance liquid chromatography and ELISA analysis. RESULTS Ninety three patients completed the intervention. There was no significant difference in markers of proliferation, apoptosis and oxidation in RP tissue comparing GT and BT to water control. Nuclear staining of NFκB was significantly decreased in RP tissue of men consuming GT (P = 0.013) but not BT (P = 0.931) compared to water control. Tea polyphenols were detected in prostate tissue from 32 of 34 men consuming GT but not in the other groups. Evidence of a systemic antioxidant effect was observed (reduced urinary 8OHdG) only with GT consumption (P = 0.03). GT, but not BT or water, also led to a small but statistically significant decrease in serum prostate-specific antigen (PSA) levels (P = 0.04). CONCLUSION Given the GT-induced changes in NFκB and systemic oxidation, and uptake of GT polyphenols in prostate tissue, future longer-term studies are warranted to further examine the role of GT for prostate cancer prevention and treatment, and possibly for other prostate conditions such as prostatitis.
Collapse
Affiliation(s)
| | - Piwen Wang
- Division of Cancer Research and Training, Charles R. Drew University of Medicine and Science, Los Angeles, CA, USA
| | | | - Min Huang
- VA Medical Center Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Tristan Grogan
- Department of Medicine Statistics Core, University of California Los Angeles
| | - David Elashoff
- Department of Medicine Statistics Core, University of California Los Angeles
| | | | - David Heber
- Center for Human Nutrition, University of California Los Angeles
| | - William J. Aronson
- Department of Urology, David Geffen School of Medicine, University of California Los Angeles
- VA Medical Center Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| |
Collapse
|
27
|
Yu J, Wang L, Zhang T, Shen H, Dong W, Ni Y, Du J. Co-expression of β-arrestin1 and NF-кB is associated with cancer progression and poor prognosis in lung adenocarcinoma. Tumour Biol 2015; 36:6551-8. [PMID: 25820700 DOI: 10.1007/s13277-015-3349-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 03/16/2015] [Indexed: 01/05/2023] Open
Abstract
β-arrestin1 and NF-κB have been demonstrated to be associated with tumorigenesis, tumor progression, and metastasis. Thus far, there is nevertheless little study about these two molecules in lung adenocarcinoma. The aim of this study was to investigate the correlation between β-arrestin1 and NF-κB expression and the clinicopathological characteristics in lung adenocarcinoma. A total of 115 surgically resected lung adenocarcinoma patients were recruited for the study. Expression of β-arrestin1 and p65 were detected by immunohistochemistry (IHC) in lung adenocarcinoma tissue samples. Nuclear expression of β-arrestin1 and p65 were observed in 39.1 % (45/115) and 46.1 % (53/115) cases of lung adenocarcinoma, respectively. And high expression of β-arrestin1 had negative prognostic impact for overall survival (OS) and disease-free survival (DFS) (p = 0.034 and p = 0.033). In addition, overexpression of p65 indicated a significantly poor OS and DFS than those of lower-expression (p = 0.038 and p = 0.041). Furthermore, co-expression of nuclear β-arrestin1 and p65 correlated with poorer OS and DFS in lung adenocarcinoma patients. Multivariate analysis using the Cox regression model confirmed that co-expression of nuclear β-arrestin1 and p65 was an independent prognostic factor for tumor progression (p = 0.008). In conclusion, these data indicated that co-expression of nuclear β-arrestin1 and p65 was a novel predictor for worse prognosis in patients with lung adenocarcinoma.
Collapse
Affiliation(s)
- Jianyu Yu
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, 324 Jingwu Road, Jinan, 250021, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
28
|
Al-Fatlawi AA, Al-Fatlawi AA, Irshad M, Ahmad A. Effect of parthenolide on growth and apoptosis regulatory genes of human cancer cell lines. PHARMACEUTICAL BIOLOGY 2015; 53:104-109. [PMID: 25289524 DOI: 10.3109/13880209.2014.911919] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
CONTEXT Parthenolide (a sesquiterpene lactone), a bioactive compound of Tanacetum parthenium (L.) Schultz Bip. (Asteraceae) herb, has been reported for antioxidant and anticancer activities. OBJECTIVE The present study evaluated the effect of parthenolide on growth and apoptosis-regulatory genes of human cervical cancer (SiHa) and breast cancer (MCF-7) cell lines. MATERIALS AND METHODS The cytotoxic activity of parthenolide (3.5-21 µM) was examined by MTT and LDH assays at 24 and 48 h time intervals. Apoptotic activity was evaluated by expression analysis of multiple apoptosis-regulatory genes (i.e., p53, Bcl-2, Bax, caspase-3, -6, and -9) by reverse transcriptase-PCR and DNA fragmentation assay. RESULTS Parthenolide inhibited the growth of SiHa and MCF-7 cell lines in a concentration-dependent manner at 24 and 48 h time intervals (p < 0.001). The IC50 value of parthenolide against SiHa and MCF-7 cells were 8.42 ± 0.76 and 9.54 ± 0.82 μM, respectively. Parthenolide-treated cells showed up-regulation of p53, Bax, caspase-3, -6, and -3 genes and down-regulation of Bcl-2 gene (p ≤ 0.008). At IC50, the p53 gene was up-regulated by 9.67- and 3.15-fold in SiHa and MCF-7 cells, respectively. The Bax to Bcl-2 ratio was 3.4 and 2.3 for SiHa and MCF-7 cells, respectively. Also, the fragmented genomic DNA in parthenolide-treated cells showed the signs of apoptosis. CONCLUSION Our study endorsed the biological activity of parthenolide and demonstrated the parthenolide-induced growth inhibition and apoptosis in SiHa and MCF-7 cells by modulating the expression of apoptosis-regulatory genes.
Collapse
Affiliation(s)
- Anees A Al-Fatlawi
- Department of Pharmacology, Institute of Pharmacy, NIMS University , Jaipur, Rajasthan , India
| | | | | | | |
Collapse
|
29
|
Janganati V, Penthala NR, Madadi NR, Chen Z, Crooks PA. Anti-cancer activity of carbamate derivatives of melampomagnolide B. Bioorg Med Chem Lett 2014; 24:3499-502. [PMID: 24928404 PMCID: PMC4381805 DOI: 10.1016/j.bmcl.2014.05.059] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 05/15/2014] [Accepted: 05/16/2014] [Indexed: 11/21/2022]
Abstract
Melampomagnolide B (MMB) is a natural sesquiterpene structurally related to parthenolide (PTL). We have shown that MMB exhibits anti-leukemic properties similar to PTL. Unlike PTL, the presence of a primary hydroxyl group in the MMB molecule allows the opportunity for examining the biological activity of a variety of conjugated analogs of MMB. We have now synthesized a series of carbamate analogs of MMB and evaluated these derivatives for anti-cancer activity against a panel of sixty human cancer cell lines. Analogs 6a and 6e exhibited promising anti-leukemic activity against human leukemia cell line CCRF-CEM with GI50 values of 680 and 620 nM, respectively. Analog 6a also showed GI50 values of 1.98 and 1.38 μM respectively, against RPMI-8226 and SR leukemia cell lines and GI50 values of 460 and 570 nM against MDA-MB-435 melanoma and MDA-MB-468 breast cancer cell lines, respectively. Analog 6e had GI50 values of 650 and 900 nM against HOP-92 non-small cell lung and RXF 393 renal cancer cell lines.
Collapse
Affiliation(s)
- Venumadhav Janganati
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Narsimha Reddy Penthala
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Nikhil Reddy Madadi
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Zheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Peter A Crooks
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
30
|
Sun X, Fu X, Li J, Xing C, Frierson HF, Wu H, Ding X, Ju T, Cummings RD, Dong JT. Deletion of atbf1/zfhx3 in mouse prostate causes neoplastic lesions, likely by attenuation of membrane and secretory proteins and multiple signaling pathways. Neoplasia 2014; 16:377-89. [PMID: 24934715 PMCID: PMC4198693 DOI: 10.1016/j.neo.2014.05.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 04/30/2014] [Accepted: 05/06/2014] [Indexed: 01/14/2023] Open
Abstract
The ATBF1/ZFHX3 gene at 16q22 is the second most frequently mutated gene in human prostate cancer and has reduced expression or mislocalization in several types of human tumors. Nonetheless, the hypothesis that ATBF1 has a tumor suppressor function in prostate cancer has not been tested. In this study, we examined the role of ATBF1 in prostatic carcinogenesis by specifically deleting Atbf1 in mouse prostatic epithelial cells. We also examined the effect of Atbf1 deletion on gene expression and signaling pathways in mouse prostates. Histopathologic analyses showed that Atbf1 deficiency caused hyperplasia and mouse prostatic intraepithelial neoplasia (mPIN) primarily in the dorsal prostate but also in other lobes. Hemizygous deletion of Atbf1 also increased the development of hyperplasia and mPIN, indicating a haploinsufficiency of Atbf1. The mPIN lesions expressed luminal cell markers and harbored molecular changes similar to those in human PIN and prostate cancer, including weaker expression of basal cell marker cytokeratin 5 (Ck5), cell adhesion protein E-cadherin, and the smooth muscle layer marker Sma; elevated expression of the oncoproteins phospho-Erk1/2, phospho-Akt and Muc1; and aberrant protein glycosylation. Gene expression profiling revealed a large number of genes that were dysregulated by Atbf1 deletion, particularly those that encode for secretory and cell membrane proteins. The four signaling networks that were most affected by Atbf1 deletion included those centered on Erk1/2 and IGF1, Akt and FSH, NF-κB and progesterone and β-estradiol. These findings provide in vivo evidence that ATBF1 is a tumor suppressor in the prostate, suggest that loss of Atbf1 contributes to tumorigenesis by dysregulating membrane and secretory proteins and multiple signaling pathways, and provide a new animal model for prostate cancer.
Collapse
Affiliation(s)
- Xiaodong Sun
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA 30322
| | - Xiaoying Fu
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA 30322; Department of Pathology, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Jie Li
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA 30322
| | - Changsheng Xing
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA 30322
| | - Henry F Frierson
- Department of Pathology, University of Virginia Health System, Charlottesville, VA
| | - Hao Wu
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA 30322
| | - Xiaokun Ding
- Department of Biochemistry, Emory University, Atlanta, GA 30322
| | - Tongzhong Ju
- Department of Biochemistry, Emory University, Atlanta, GA 30322
| | | | - Jin-Tang Dong
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA 30322.
| |
Collapse
|
31
|
Sharma J, Gray KP, Harshman LC, Evan C, Nakabayashi M, Fichorova R, Rider J, Mucci L, Kantoff PW, Sweeney CJ. Elevated IL-8, TNF-α, and MCP-1 in men with metastatic prostate cancer starting androgen-deprivation therapy (ADT) are associated with shorter time to castration-resistance and overall survival. Prostate 2014; 74:820-8. [PMID: 24668612 DOI: 10.1002/pros.22788] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 01/16/2014] [Indexed: 12/16/2022]
Abstract
BACKGROUND Chemokines and cytokines have been implicated in progression to castration-resistant prostate cancer (CRPC). METHODS Retrospective data were accessed from 122 men with serum samples drawn at a median of 0.5 months after starting ADT for metastatic prostate cancer. MCP-1, IL-1-β, IL-2, IL-8, IL-6, and TNF-α levels were measured by multiplex electrochemiluminescence assays. A multivariable Cox model assessed the association of time to CRPC and overall survival by the protein levels and adjusted for clinical variables (age and prostate specific antigen (PSA) levels at start of ADT, race, ECOG status, and extent of metastases). Associations were reported as hazard ratio (HR) with 95% confidence interval (CI). RESULTS Median follow-up and overall survival were 44 and 42.2 months, respectively. ECOG performance status (≥1 vs. 0) was negatively associated with overall survival [HR = 2.8 (1.1-7.0), P = 0.03], and PSA nadir < 0.2 was predictive of longer time to development of CRPC [HR = 0.3 (0.2-0.5), P < 0.0001]. The HR for time to CRPC by protein above the median was 1.4 (95% CI: 0.9, 2.2, P = 0.13) for IL-8; 1.3 (95% CI: 0.8, 2, P = 0.18) for TNF-α; 1.0 (95% CI: 0.7, 1.6, P = 0.95) for MCP-1. The HR for median overall survival for protein levels above the median was: 1.9 (95% CI: 1.0, 3.5, P = 0.04) for IL-8; 2.0 (95% CI: 1.1, 3.5, P = 0.02) for TNF-α; 1.7 (95% CI: 1.7, 3.0, P = 0.08) for MCP-1. There was no association with IL-1-β, IL-2, or IL-6. CONCLUSION Higher levels of inflammation-associated cytokines correlate with poorer prostate cancer outcomes and may guide strategies to improve prostate cancer therapy.
Collapse
Affiliation(s)
- Jaya Sharma
- Department of Medicine, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Chondrogiannis G, Kastamoulas M, Kanavaros P, Vartholomatos G, Bai M, Baltogiannis D, Sofikitis N, Arvanitis D, Galani V. Cytokine effects on cell viability and death of prostate carcinoma cells. BIOMED RESEARCH INTERNATIONAL 2014; 2014:536049. [PMID: 24982891 PMCID: PMC4058150 DOI: 10.1155/2014/536049] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Revised: 04/09/2014] [Accepted: 05/06/2014] [Indexed: 01/01/2023]
Abstract
We analyzed the effects of IL-13, IFN- γ , and IL-1 β on cell viability and death of LNCaP and PC-3 cells and major signaling pathways involved in these effects. Significant increase of LNCaP cell death (apoptotic and necrotic) and increased levels of active caspase 3 were observed in cells treated with inhibitors of ERK 1/2 (UO126) and p38 (SB203580) prior to IL-1 β treatment in comparison to cells treated with UO126, SB203580, or IL-1 β alone. Significant increase of LNCaP but not PC-3 cell death was detected after treatment with LY-294002 (inhibitor of phosphatidylinositol 3-kinase). No significant increase of LNCaP and PC-3 cell death was observed after treatment with SP600125 (inhibitor of JNK), SB203580 (inhibitor of p38), UO126 (inhibitor of ERK 1/2), or BAY 11-7082 (inhibitor of NF- κ B). Reduced c-FLIPL expression was observed in LNCaP cells treated with LY-294002. The significant potentiation of LNCaP cell death by inhibition of ERK 1/2, p38, and PI3-K pathways may provide a rationale for therapeutic approach in androgen-dependent prostate cancer.
Collapse
Affiliation(s)
- Georgios Chondrogiannis
- Department of Anatomy-Histology-Embryology, Medical School, University of Ioannina, 45110 Ioannina, Greece
| | - Michalis Kastamoulas
- Department of Anatomy-Histology-Embryology, Medical School, University of Ioannina, 45110 Ioannina, Greece
| | - Panagiotis Kanavaros
- Department of Anatomy-Histology-Embryology, Medical School, University of Ioannina, 45110 Ioannina, Greece
| | | | - Maria Bai
- Department of Pathology, Medical School, University of Ioannina, 45110 Ioannina, Greece
| | | | - Nikolaos Sofikitis
- Department of Urology, Medical School, University of Ioannina, 45110 Ioannina, Greece
| | - Dimitrios Arvanitis
- Department of Anatomy, Medical School, University of Thessaly, 44110 Larisa, Greece
| | - Vasiliki Galani
- Department of Anatomy-Histology-Embryology, Medical School, University of Ioannina, 45110 Ioannina, Greece
| |
Collapse
|
33
|
Janganati V, Penthala NR, Cragle CE, MacNicol AM, Crooks PA. Heterocyclic aminoparthenolide derivatives modulate G(2)-M cell cycle progression during Xenopus oocyte maturation. Bioorg Med Chem Lett 2014; 24:1963-7. [PMID: 24656611 PMCID: PMC4121966 DOI: 10.1016/j.bmcl.2014.02.067] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 02/22/2014] [Accepted: 02/24/2014] [Indexed: 12/11/2022]
Abstract
Aminoparthenolide derivatives have been prepared by reaction of parthenolide with various heterocyclic amines to afford corresponding Michael addition products. These novel compounds were evaluated for their modulatory effects on Xenopus oocyte maturation. Two compounds, 6e and 6f, were identified that promote G2-M cell cycle progression.
Collapse
Affiliation(s)
- Venumadhav Janganati
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Narsimha Reddy Penthala
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Chad E Cragle
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Angus M MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Peter A Crooks
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
34
|
Ginzburg S, Golovine KV, Makhov PB, Uzzo RG, Kutikov A, Kolenko VM. Piperlongumine inhibits NF-κB activity and attenuates aggressive growth characteristics of prostate cancer cells. Prostate 2014; 74:177-86. [PMID: 24151226 PMCID: PMC4052841 DOI: 10.1002/pros.22739] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 09/15/2013] [Indexed: 11/06/2022]
Abstract
BACKGROUND Elevated NF-κB activity has been previously demonstrated in prostate cancer cell lines as hormone-independent or metastatic characteristics develop. We look at the effects of piperlongumine (PL), a biologically active alkaloid/amide present in piper longum plant, on the NF-κB pathway in androgen-independent prostate cancer cells. METHODS NF-κB activity was evaluated using Luciferase reporter assays and Western blot analysis of p50 and p65 nuclear translocation. IL-6, IL-8, and MMP-9 levels were assessed using ELISA. Cellular adhesion and invasiveness properties of prostate cancer cells treated with PL were also assessed. RESULTS NF-κB DNA-binding activity was directly down-regulated with increasing concentrations of PL, along with decreased nuclear translocation of p50 and p65 subunits. Expression of IL-6, IL-8, MMP-9, and ICAM-1 was attenuated, and a decrease of cell-to-matrix adhesion and invasiveness properties of prostate cancer cells were observed. CONCLUSIONS PL-mediated inhibition of NF-κB activity decreases aggressive growth characteristics of prostate cancer cells in vitro.
Collapse
Affiliation(s)
- Serge Ginzburg
- Department of Urologic Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | | | - Petr B. Makhov
- Department of Urologic Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - Robert G. Uzzo
- Department of Urologic Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - Alexander Kutikov
- Department of Urologic Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | | |
Collapse
|
35
|
Tripathi V, Popescu NC, Zimonjic DB. DLC1 suppresses NF-κB activity in prostate cancer cells due to its stabilizing effect on adherens junctions. SPRINGERPLUS 2014; 3:27. [PMID: 24683532 PMCID: PMC3967735 DOI: 10.1186/2193-1801-3-27] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 01/09/2014] [Indexed: 01/06/2023]
Abstract
DLC1 (Deleted in Liver Cancer 1) gene encodes a RhoGTPase-activating protein (RhoGAP), which exerts most of its tumor suppressor functions through suppression of small Rho GTPases proteins RhoA, RhoB, RhoC and to some degree Cdc42, but not Rac. RhoGTPases are implicated in NF-κB activation in highly invasive prostate carcinoma (PCA), with consequences on cell proliferation, survival and metastatic capacity. Here we demonstrate that DLC1 transduction in two androgen-independent (AI) and highly metastatic PCA cell lines negatively regulates NF-κB activity in a GAP- and α-catenin-dependent manner. Expressed DLC1 protein suppresses the phosphorylation of NF-κB inhibitor, IκBα, causes its relocation from membrane ruffles into cytoplasm and attenuates its ubiquitination and subsequent degradation. DLC1-mediated NF-kB suppression and its effects are comparable to NF-κB inhibition using either shRNA knockdown or peptide inhibitor. Expression of transduced DLC1 suppressed the expression of NF-κB mediated genes. Such effects were found to be reliant on presence of calcium, indicating that the observed modifications are dependent on, and enabled by DLC-mediated stabilization of adherens junctions. These results expand the multitude of DLC1 interactions with other genes that modulate its oncosuppressive function, and may have potential therapeutic implications.
Collapse
Affiliation(s)
- Veenu Tripathi
- Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, 37 Convent Drive, MSC 4262, Bethesda, Maryland 20892 USA
| | - Nicholas C Popescu
- Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, 37 Convent Drive, MSC 4262, Bethesda, Maryland 20892 USA
| | - Drazen B Zimonjic
- Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, 37 Convent Drive, MSC 4262, Bethesda, Maryland 20892 USA
| |
Collapse
|
36
|
HOXB13 downregulates intracellular zinc and increases NF-κB signaling to promote prostate cancer metastasis. Oncogene 2013; 33:4558-67. [PMID: 24096478 DOI: 10.1038/onc.2013.404] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 07/30/2013] [Accepted: 08/23/2013] [Indexed: 12/13/2022]
Abstract
Characteristically, prostate cancer (PCa) cells exhibit marked decrease in intracellular zinc; however, the mechanism responsible is not clearly understood. HOXB13 is involved in PCa progression and is overexpressed in castration-resistant PCa. DNA microarray analysis of LNCaP Pca cells showed that ZnT zinc output transporters were strikingly upregulated among androgen-independent HOXB13 target genes. Furthermore, exogenous HOXB13 caused intracellular zinc concentrations to fall in PCa cells, stimulated NF-κB-mediated signaling by reducing inhibitor of NF-κB alpha (IκBα) and enhanced the nuclear translocation of RelA/p65. Human prostate tumors also exhibited strong inverse correlation between the protein expressions of HOXB13 and IκBα. Consequently, HOXB13 stimulated PCa cell invasion, and this was inhibited by the suppression of ZnT4. In addition, studies in a PC3 orthotopic mouse model of PCa metastasis showed that HOXB13 is a strong metastatic stimulator. Taken together, these results show that HOXB13 promotes PCa invasion and metastasis by decreasing intracellular zinc levels, thus stimulating NF-κB signals, and suggest that HOXB13 acts as a modulator of intracellular zinc levels that promotes the malignant characteristics of PCa.
Collapse
|
37
|
Manna S, Singha B, Phyo SA, Gatla HR, Chang TP, Sanacora S, Ramaswami S, Vancurova I. Proteasome inhibition by bortezomib increases IL-8 expression in androgen-independent prostate cancer cells: the role of IKKα. THE JOURNAL OF IMMUNOLOGY 2013; 191:2837-46. [PMID: 23894194 DOI: 10.4049/jimmunol.1300895] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Expression of the proinflammatory and proangiogenic chemokine IL-8, which is regulated at the transcriptional level by NF-κB, is constitutively increased in androgen-independent metastatic prostate cancer and correlates with poor prognosis. Inhibition of NF-κB-dependent transcription was used as an anticancer strategy for the development of the first clinically approved 26S proteasome inhibitor, bortezomib (BZ). Even though BZ has shown remarkable antitumor activity in hematological malignancies, it has been less effective in prostate cancer and other solid tumors; however, the mechanisms have not been fully understood. In this article, we report that proteasome inhibition by BZ unexpectedly increases IL-8 expression in androgen-independent prostate cancer PC3 and DU145 cells, whereas expression of other NF-κB-regulated genes is inhibited or unchanged. The BZ-increased IL-8 expression is associated with increased in vitro p65 NF-κB DNA binding activity and p65 recruitment to the endogenous IL-8 promoter. In addition, proteasome inhibition induces a nuclear accumulation of IκB kinase (IKK)α, and inhibition of IKKα enzymatic activity significantly attenuates the BZ-induced p65 recruitment to IL-8 promoter and IL-8 expression, demonstrating that the induced IL-8 expression is mediated, at least partly, by IKKα. Together, these data provide the first evidence, to our knowledge, for the gene-specific increase of IL-8 expression by proteasome inhibition in prostate cancer cells and suggest that targeting both IKKα and the proteasome may increase BZ effectiveness in treatment of androgen-independent prostate cancer.
Collapse
Affiliation(s)
- Subrata Manna
- Department of Biological Sciences, St. John's University, New York, NY 11439, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Li Y, Kong D, Ahmad A, Bao B, Sarkar FH. Antioxidant function of isoflavone and 3,3'-diindolylmethane: are they important for cancer prevention and therapy? Antioxid Redox Signal 2013; 19:139-50. [PMID: 23391445 PMCID: PMC3689155 DOI: 10.1089/ars.2013.5233] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
SIGNIFICANCE Oxidative stress has been mechanistically linked with aging and chronic diseases, including cancer. In fact, oxidative stress status, chronic disease-related inflammation, and cancer occurred in the aging population are tightly correlated. It is well known that the activation of nuclear factor kappa B (NF-κB) plays important roles in oxidative stress, inflammation, and carcinogenesis. Therefore, targeting NF-κB is an important preventive or therapeutic strategy against oxidative stress, inflammation, and cancer. RECENT ADVANCES A variety of natural compounds has been found to reduce oxidative stress through their antioxidant activity. Among them, isoflavone, indole-3-carbinol (I3C), and its in vivo dimeric compound 3,3'-diindolylmethane (DIM) have shown their promising effects on the inhibition of NF-κB with corresponding reduction of oxidative stress. CRITICAL ISSUES It has been found that isoflavone, I3C, and DIM could inhibit cancer development and progression by regulating multiple cellular signaling pathways that are related to oxidative stress and significantly deregulated in cancer. FUTURE DIRECTIONS The antioxidative and anticancer effects of these natural agents make them strong candidates for chemoprevention and/or therapy against human malignancies. However, more clinical trials are needed to evaluate the effects of isoflavone and DIM for the prevention of cancer development and also for the treatment of cancer either alone or in combination with conventional cancer therapeutics.
Collapse
Affiliation(s)
- Yiwei Li
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | | | | | |
Collapse
|
39
|
Gannon PO, Lessard L, Stevens LM, Forest V, Bégin LR, Minner S, Tennstedt P, Schlomm T, Mes-Masson AM, Saad F. Large-scale independent validation of the nuclear factor-kappa B p65 prognostic biomarker in prostate cancer. Eur J Cancer 2013; 49:2441-8. [DOI: 10.1016/j.ejca.2013.02.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 02/09/2013] [Accepted: 02/25/2013] [Indexed: 01/29/2023]
|
40
|
Piperine, a Bioactive Component of Pepper Spice Exerts Therapeutic Effects on Androgen Dependent and Androgen Independent Prostate Cancer Cells. PLoS One 2013; 8:e65889. [PMID: 23824300 PMCID: PMC3688824 DOI: 10.1371/journal.pone.0065889] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 04/30/2013] [Indexed: 01/16/2023] Open
Abstract
Prostate cancer is the most common solid malignancy in men, with 32,000 deaths annually. Piperine, a major alkaloid constituent of black pepper, has previously been reported to have anti-cancer activity in variety of cancer cell lines. The effect of piperine against prostate cancer is not currently known. Therefore, in this study, we investigated the anti-tumor mechanisms of piperine on androgen dependent and androgen independent prostate cancer cells. Here, we show that piperine inhibited the proliferation of LNCaP, PC-3, 22RV1 and DU-145 prostate cancer cells in a dose dependent manner. Furthermore, Annexin-V staining demonstrated that piperine treatment induced apoptosis in hormone dependent prostate cancer cells (LNCaP). Using global caspase activation assay, we show that piperine-induced apoptosis resulted in caspase activation in LNCaP and PC-3 cells. Further studies revealed that piperine treatment resulted in the activation of caspase-3 and cleavage of PARP-1 proteins in LNCaP, PC-3 and DU-145 prostate cancer cells. Piperine treatment also disrupted androgen receptor (AR) expression in LNCaP prostate cancer cells. Our evaluations further show that there is a significant reduction of Prostate Specific Antigen (PSA) levels following piperine treatment in LNCaP cells. NF-kB and STAT-3 transcription factors have previously been shown to play a role in angiogenesis and invasion of prostate cancer cells. Interestingly, treatment of LNCaP, PC-3 and DU-145 prostate cancer cells with piperine resulted in reduced expression of phosphorylated STAT-3 and Nuclear factor-κB (NF-kB) transcription factors. These results correlated with the results of Boyden chamber assay, wherein piperine treatment reduced the cell migration of LNCaP and PC-3 cells. Finally, we show that piperine treatment significantly reduced the androgen dependent and androgen independent tumor growth in nude mice model xenotransplanted with prostate cancer cells. Taken together, these results support further investigation of piperine as a potential therapeutic agent in the treatment of prostate cancer.
Collapse
|
41
|
Lee ST, Wong PF, He H, Hooper JD, Mustafa MR. Alpha-tomatine attenuation of in vivo growth of subcutaneous and orthotopic xenograft tumors of human prostate carcinoma PC-3 cells is accompanied by inactivation of nuclear factor-kappa B signaling. PLoS One 2013; 8:e57708. [PMID: 23437404 PMCID: PMC3578807 DOI: 10.1371/journal.pone.0057708] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 01/25/2013] [Indexed: 11/24/2022] Open
Abstract
Background Nuclear factor-kappa B (NF-κB) plays a role in prostate cancer and agents that suppress its activation may inhibit development or progression of this malignancy. Alpha (α)-tomatine is the major saponin present in tomato (Lycopersicon esculentum) and we have previously reported that it suppresses tumor necrosis factor-alpha (TNF-α)-induced nuclear translocation of nuclear factor-kappa B (NF-κB) in androgen-independent prostate cancer PC-3 cells and also potently induces apoptosis of these cells. However, the precise mechanism by which α-tomatine suppresses NF-κB nuclear translocation is yet to be elucidated and the anti-tumor activity of this agent in vivo has not been examined. Methodology/ Principal Findings In the present study we show that suppression of NF-κB activation by α-tomatine occurs through inhibition of I kappa B alpha (IκBα) kinase activity, leading to sequential suppression of IκBα phosphorylation, IκBα degradation, NF-κB/p65 phosphorylation, and NF-κB p50/p65 nuclear translocation. Consistent with its ability to induce apoptosis, α-tomatine reduced TNF-α induced activation of the pro-survival mediator Akt and its inhibition of NF-κB activation was accompanied by significant reduction in the expression of NF-κB-dependent anti-apoptotic (c-IAP1, c-IAP2, Bcl-2, Bcl-xL, XIAP and survivin) proteins. We also evaluated the antitumor activity of α-tomatine against PC-3 cell tumors grown subcutaneously and orthotopically in mice. Our data indicate that intraperitoneal administration of α-tomatine significantly attenuates the growth of PC-3 cell tumors grown at both sites. Analysis of tumor material indicates that the tumor suppressing effects of α-tomatine were accompanied by increased apoptosis and lower proliferation of tumor cells as well as reduced nuclear translocation of the p50 and p65 components of NF-κB. Conclusion/ Significance Our study provides first evidence for in vivo antitumor efficacy of α-tomatine against the human androgen-independent prostate cancer. The potential usefulness of α-tomatine in prostate cancer prevention and therapy requires further investigation.
Collapse
Affiliation(s)
- Sui-Ting Lee
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur, Malaysia
| | - Pooi-Fong Wong
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur, Malaysia
| | - Hui He
- Mater Medical Research Institute, South Brisbane, Queensland, Australia
| | - John David Hooper
- Mater Medical Research Institute, South Brisbane, Queensland, Australia
| | - Mohd Rais Mustafa
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur, Malaysia
- * E-mail:
| |
Collapse
|
42
|
Shiu SYW, Leung WY, Tam CW, Liu VWS, Yao KM. Melatonin MT1 receptor-induced transcriptional up-regulation of p27(Kip1) in prostate cancer antiproliferation is mediated via inhibition of constitutively active nuclear factor kappa B (NF-κB): potential implications on prostate cancer chemoprevention and therapy. J Pineal Res 2013; 54:69-79. [PMID: 22856547 DOI: 10.1111/j.1600-079x.2012.01026.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Our laboratory has recently demonstrated a melatonin MT1 receptor-mediated antiproliferative signaling mechanism in androgen receptor (AR)-positive prostate epithelial cells which involves up-regulation of p27(Kip1) through dual activation of Gα(s)/protein kinase A (PKA) and Gα(q)/protein kinase C (PKC) in parallel, and down-regulation of activated AR signaling via PKC stimulation. The aim of the present investigation was to identify the transcription factor that mediates melatonin's up-regulatory effect on p27(Kip1) in LNCaP and 22Rv1 prostate cancer cells. Deletion mapping and reporter assays of the p27(Kip1) promoter revealed that the putative melatonin-responsive transcription factor binds to a 116 base-pair region of the promoter sequence, which contains a potential nuclear factor kappa B (NF-κB) binding site. When the NF-κB binding site was abolished by site-directed mutagenesis, the stimulatory effect of melatonin on p27(Kip1) promoter activity was mitigated. Notably, melatonin inhibited the DNA binding of activated NF-κB via MT1 receptor-induced PKA and PKC stimulation. Furthermore, melatonin's up-regulatory effect on p27(Kip1) transcription and consequent cell antiproliferation were abrogated by NF-κB activator but mimicked by NF-κB inhibitor. The results indicate that inhibition of constitutively active NF-κB via melatonin MT1 receptor-induced dual activation of (Gα(s)) PKA and (Gα(q)) PKC can de-repress the p27(Kip1) promoter leading to transcriptional up-regulation of p27(Kip1). MT1 receptor-mediated inhibition of activated NF-κB signaling provides a novel mechanism supporting the use of melatonin in prostate cancer chemoprevention and therapy.
Collapse
Affiliation(s)
- Stephen Y W Shiu
- Department of Physiology, The University of Hong Kong, Hong Kong, China Department of Biochemistry, The University of Hong Kong, Hong Kong, China Nursing, School of Science and Technology, Open University of Hong Kong, Hong Kong, China
| | | | | | | | | |
Collapse
|
43
|
Nadiminty N, Tummala R, Zhu Y, Gao AC. NF-kappaB2/p52 in Prostate Cancer. Prostate Cancer 2013. [DOI: 10.1007/978-1-4614-6828-8_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
44
|
Involvement of interleukin-1β mediated nuclear factor κB signalling pathways to down-regulate prostate-specific antigen and cell proliferation in LNCaP prostate cancer cells. Cell Biol Int 2012; 36:449-54. [PMID: 22103356 DOI: 10.1042/cbi20100922] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Involvement of NF-κB (nuclear factor κB) mediated by IL-1β (interleukin-1β) on cell proliferation and PSA (prostate-specific antigen) production of LNCaP prostate cell lines and the possible cross-talk with Akt (also known as protein kinase B) signalling pathway has been investigated. NF-κB and Akt were analysed by Western blotting from LNCaP cells treated by IL-1β before proliferation and PSA production were measured. IL-1β inhibited proliferation and decreased PSA production. The Akt pathway was not sensitive, whereas NF-κB phosphorylation occurred as a result of treatment. PSA production and proliferation of LNCaP cells were down-regulated by NF-κB mediated by IL-1β promoting anti-apoptotic signalling and co-suppressor factors of PSA expression. IL-1β through NF-κB activation provides a rationale for therapeutic approaches in the anticancer treatment of prostate.
Collapse
|
45
|
IκB kinases modulate the activity of the androgen receptor in prostate carcinoma cell lines. Neoplasia 2012; 14:178-89. [PMID: 22496618 DOI: 10.1593/neo.111444] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Revised: 02/27/2012] [Accepted: 02/27/2012] [Indexed: 01/21/2023] Open
Abstract
Enhanced nuclear localization of nuclear factor κB (NF-κB) in prostate cancer (PCa) samples and constitutive NF-κB signaling in a class of PCa cell lines with low androgen receptor (AR) expression (PC3 and DU-145) imply an important role of the IκB kinase (IKK)/NF-κB system in PCa. However, most PCa and PCa cell lines depend on the activity of the AR, and the role of NF-κB in these AR-expressing PCa remains unclear. Here, we demonstrate that inhibition of NF-κB signaling by the IKK inhibitor BMS345541 reduced proliferation and increased apoptosis in AR-expressing PCa cell lines. Furthermore, AR activity and target gene expression were distinctively reduced, whereas AR protein levels remained unaltered on BMS345541 treatment. Similar effects were observed particularly after small interfering RNA (siRNA)-mediated knockdown of IKK1, but not by siRNA-mediated suppression of IKK2. Moreover, IKK1 overexpression augmented 5α-dihydrotestosterone-induced nuclear AR translocation, whereas nuclear AR was reduced by IKK1 knockdown or BMS345541. However, because IKK1 also enhances the activity of a chronically nuclear AR mutant, modulation of the subcellular distribution seems not to be the only mechanism by which IKK1 enhances AR activity. Finally, reduced in vivo AR phosphorylation after BMS345541 treatment and in vitro AR phosphorylation by IKK1 or IKK2 imply that AR constitutes a novel IKK target. Taken together, our data identify IKK1 as a potentially target structure for future therapeutic intervention in PCa.
Collapse
|
46
|
Hwang JW, Oh JH, Yoo HS, Lee YW, Cho CK, Kwon KR, Yoon JH, Park J, Her S, Lee ZW, Jang IS, Choi JS. Mountain ginseng extract exhibits anti-lung cancer activity by inhibiting the nuclear translocation of NF-κB. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2012; 40:187-202. [PMID: 22298458 DOI: 10.1142/s0192415x12500152] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Administration of mountain ginseng (MG) extract can restore advanced cancer to a normal state. To elucidate the mechanism by which MG extract prevents the progression of lung cancer, the processes of proliferation and death of lung cancer cells (A549) were examined after treatment with MG extract. Butanol-extracted MG (BX-MG) showed a high inhibitory effect (IC(50) = 2 mg/ml) by attenuating proliferation and inducing apoptosis in lung cancer cells. By HPLC-UV analysis of BX-MG, ginsenosides, Rb1 was identified as the most abundant ginsenoside, followed by Rg1, Re, Rc and Rb2. BX-MG induced caspase-3 dependent apoptosis by inhibiting NF-κB. In addition, BX-MG activated p53 and p21, resulting in the attenuated proliferation of A549 cells. Reduced activity of the NF-κB promoter and increased activity of the p53 promoter indicate that BX-MG regulates apoptosis at the level of transcription in lung cancer cells. Furthermore, BX-MG blocked the nuclear translocation of RelA and the associated reduction in surviving. These results suggest that BX-MG inhibits lung cancer cell growth by activating tumor suppressors and inhibiting nuclear translocation of NF-κB.
Collapse
Affiliation(s)
- Jeong Won Hwang
- Division of Life Science, Korea Basic Science Institute, Gwahangno 169-148, Yuseong-gu, Daejeon, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Expression of neutral endopeptidase, endothelin-1, and nuclear factor kappa B in prostate cancer: interrelations and associations with prostate-specific antigen recurrence after radical prostatectomy. Prostate Cancer 2012; 2012:452795. [PMID: 22666602 PMCID: PMC3362215 DOI: 10.1155/2012/452795] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 02/27/2012] [Indexed: 11/19/2022] Open
Abstract
Objective. To study the impact of the neutral endopeptidase (NEP)/neuropeptides (NPs) axis and nuclear factor kappa B (NFκB) as predictors of prostate-specific antigen (PSA) recurrence after radical prostatectomy (RP). Patients and Methods. 70 patients with early-stage PC were treated with RP and their tumor samples were evaluated for expression of NEP, endothelin-1 (ET-1) and NFκB (p65). Time to PSA recurrence was correlated with the examined parameters and combined with preoperative PSA level, Gleason score, pathological TNM (pT) stage, and surgical margin (SM) assessment. Results and Limitations. Membranous expression of NEP (P < 0.001), cytoplasmic ET-1 (P = 0.002), and cytoplasmic NFκB (P < 0.001) were correlated with time to PSA relapse. NEP was associated with ET-1 (P < 0.001) and NFκB (P < 0.001). ET-1 was also correlated with NFκB (P < 0.001). NEP expression (P = 0.017), pT stage (P = 0.013), and SMs (P = 0.036) were independent predictors of time to PSA recurrence.
Conclusions. There seems to be a clinical model of NEP/NPs and NFκB pathways interconnection, with their constituents following inverse patterns of expression in accordance with their biological roles and molecular interrelations.
Collapse
|
48
|
MacKenzie L, McCall P, Hatziieremia S, Catlow J, Adams C, McArdle P, Seywright M, Tanahill C, Paul A, Underwood M, Mackay S, Plevin R, Edwards J. Nuclear factor κB predicts poor outcome in patients with hormone-naive prostate cancer with high nuclear androgen receptor. Hum Pathol 2012; 43:1491-500. [PMID: 22406367 DOI: 10.1016/j.humpath.2011.11.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Revised: 11/11/2011] [Accepted: 11/16/2011] [Indexed: 10/28/2022]
Abstract
Despite recent advances in prostate cancer treatments, disease recurrence is common and associated with significant morbidity and mortality. The need for more effective antitumor agents has led researchers to target signaling pathways that drive tumorigenesis by modulating or bypassing androgen receptor signaling--attenuation or blockade of which current treatments aim to effect. The transcription factor nuclear factor κB/p65 has been implicated in prostate cancer progression; however, few studies have examined the involvement of nuclear factor κB in hormone-naive disease. We used immunohistochemistry to investigate expression of p65, androgen receptor, Ki-67, and phosphorylation status of p65 at serine 536, in 154 tumor samples taken from patients before hormone ablation or radical treatment. Nuclear p65 expression was significantly associated with disease-specific mortality: P = .005; hazard ratio, 2.2. When patients were stratified according to androgen receptor status, this relationship was abolished in low androgen receptor-expressing patients and potentiated in high androgen receptor-expressing patients: P = .002; hazard ratio, 3.1. Ki-67 expression was also prognostic of shorter disease-specific mortality: P = .001; hazard ratio, 2.3. When the cohort was stratified according to androgen receptor status, this relationship held for high androgen receptor expressers but not low expressers: P = .0003; hazard ratio, 3.5. Neither androgen receptor nor p65 phosphorylated at S536 were significantly prognostic when considered individually. These data suggest that future prostate cancer treatments that target nuclear factor κB signaling should be assigned primarily to patients with concomitant high nuclear p65 and androgen receptor expression.
Collapse
Affiliation(s)
- Lewis MacKenzie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Mimeault M, Johansson SL, Batra SK. Pathobiological implications of the expression of EGFR, pAkt, NF-κB and MIC-1 in prostate cancer stem cells and their progenies. PLoS One 2012; 7:e31919. [PMID: 22384099 PMCID: PMC3285632 DOI: 10.1371/journal.pone.0031919] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2011] [Accepted: 01/20/2012] [Indexed: 02/07/2023] Open
Abstract
The progression of prostate cancers (PCs) to locally invasive, androgen-independent and metastatic disease states is generally associated with treatment resistance and disease relapse. The present study was undertaken to establish the possibility of using a combination of specific oncogenic products, including epidermal growth factor receptor (EGFR), pAkt, nuclear factor-kappaB (NF-κB) and macrophage inhibitory cytokine-1 (MIC-1) as biomarkers and therapeutic targets for optimizing the management of patients with localized PC at earlier disease stages. The immunohistochemical and immunofluorescence data have revealed that the expression levels of EGFR, Ser473-pAkt, NF-κB p65 and MIC-1 proteins were significantly enhanced in the same subset of 76 cases of prostatic adenocarcinoma specimens during the disease progression and these biomarkers were expressed in a small subpopulation of CD133+ PC cells and the bulk tumor mass of CD133− PC cells. Importantly, all of these biomarkers were also overexpressed in 80–100% of 30 PC metastasis bone tissue specimens. Moreover, the results have indicated that the EGF-EGFR signaling pathway can provide critical functions for the self-renewal of side population (SP) cells endowed with stem cell-like features from highly invasive WPE1-NB26 cells. Of therapeutic interest, the targeting of EGFR, pAkt, NF-κB or MIC-1 was also effective at suppressing the basal and EGF-promoted prostasphere formation by SP WPE1-NB26 cells, inducing disintegration of SP cell-derived prostaspheres and decreasing the viability of SP and non-SP WPE1-NB26 cell fractions. Also, the targeting of these oncogenic products induced the caspase-dependent apoptosis in chemoresistant SP WPE1-NB26 cells and enhanced their sensibility to the cytotoxic effects induced by docetaxel. These findings suggest that the combined use of EGFR, pAkt, NF-κB and/or MIC-1 may represent promising strategies for improving the accuracy of current diagnostic and prognostic methods and efficacy of treatments of PC patients in considering the disease heterogeneity, thereby preventing PC progression to metastatic and lethal disease states.
Collapse
Affiliation(s)
- Murielle Mimeault
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail: (MM); (SKB)
| | - Sonny L. Johansson
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail: (MM); (SKB)
| |
Collapse
|
50
|
Tsapakidis K, Vlachostergios PJ, Voutsadakis IA, Befani CD, Patrikidou A, Hatzidaki E, Daliani DD, Moutzouris G, Liakos P, Papandreou CN. Bortezomib reverses the proliferative and antiapoptotic effect of neuropeptides on prostate cancer cells. Int J Urol 2012; 19:565-74. [PMID: 22324515 DOI: 10.1111/j.1442-2042.2012.02967.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Neuropeptides are important signal initiators in advanced prostate cancer, partially acting through activation of nuclear factor kappa B. Central to nuclear factor kappa B regulation is the ubiquitin-proteasome system, pharmacological inhibition of which has been proposed as an anticancer strategy. We investigated the putative role of the proteasome inhibitor bortezomib in neuropeptides signaling effects on prostate cancer cells. METHODS Human prostate cancer cell lines, LNCaP and PC-3, were used to examine cell proliferation, levels of proapoptotic (caspase-3, Bad) and cell cycle regulatory proteins (p53, p27, p21), as well as total and phosphorylated Akt and p44/42 mitogen-activated protein kinase proteins. Furthermore, 20S proteasome activity, subcellular localization of nuclear factor kappa B and transcription of nuclear factor kappa B target genes, interleukin-8 and vascular endothelial growth factor, were assessed. RESULTS Neuropeptides (endothelin-1, bombesin) increased cell proliferation, whereas bortezomib decreased proliferation and induced apoptosis, an effect maintained after cotreatment with neuropeptides. Bad, p53, p21 and p27 were downregulated by neuropeptides in PC-3, and these effects were reversed with the addition of bortezomib. Neuropeptides increased proteasomal activity and nuclear factor kappa B levels in PC-3, and these effects were prevented by bortezomib. Interleukin-8 and vascular endothelial growth factor transcripts were induced after neuropeptides treatment, but downregulated by bortezomib. These results coincided with the ability of bortezomib to reduce mitogen-activated protein kinase signaling in both cell lines. CONCLUSIONS These findings are consistent with bortezomib-mediated abrogation of neuropeptides-induced proliferative and antiapoptotic signaling. Thus, the effect of the drug on the neuropeptides axis needs to be further investigated, as neuropeptide action in prostate cancer might entail involvement of the proteasome.
Collapse
|