1
|
Figueroa‐González G, Quintas‐Granados LI, Reyes‐Hernández OD, Caballero‐Florán IH, Peña‐Corona SI, Cortés H, Leyva‐Gómez G, Habtemariam S, Sharifi‐Rad J. Review of the anticancer properties of 6-shogaol: Mechanisms of action in cancer cells and future research opportunities. Food Sci Nutr 2024; 12:4513-4533. [PMID: 39055196 PMCID: PMC11266911 DOI: 10.1002/fsn3.4129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 02/05/2024] [Accepted: 03/13/2024] [Indexed: 07/27/2024] Open
Abstract
Cancer is a major global health challenge that affects every nation and accounts for a large portion of the worldwide disease burden. Furthermore, cancer cases will rise significantly in the next few decades. The Food and Drug Administration has approved more than 600 drugs for treating diverse types of cancer. However, many conventional anticancer medications cause side effects, and drug resistance develops as the treatment proceeds with a concomitant impact on patients' quality of life. Thus, exploring natural products with antitumor properties and nontoxic action mechanisms is essential. Ginger (Zingiber officinale Roscoe) rhizome has a long history of use in traditional medicine, and it contains biologically active compounds, gingerols and shogaols. The main ginger shogaol is 6-shogaol, whose concentration dramatically increases during the processing of ginger, primarily due to the heat-induced conversion of 6-gingerol. Some studies have demonstrated that 6-shogaol possesses biological and pharmacological properties, such as antioxidant, anti-inflammatory, and anticancer activities. The mechanism of action of 6-shogaol as an anticancer drug includes induction of paraptosis, induction of apoptosis, increase in the production of reactive oxygen species, induction of autophagy, and the inhibition of AKT/mTOR signaling. Despite this knowledge, the mechanism of action of 6-shogaol is not fully understood, and the scientific data on its therapeutic dose, safety, and toxicity are not entirely described. This review article examines the potential of 6-shogaol as an anticancer drug, addressing the limitations of current medications; it covers 6-shogaol's attributes, mechanism of action in cancer cells, and opportunities for future research.
Collapse
Affiliation(s)
- Gabriela Figueroa‐González
- Laboratorio de Farmacogenética, UMIEZ, Facultad de Estudios Superiores ZaragozaUniversidad Nacional Autónoma de MéxicoCiudad de MéxicoMexico
| | - Laura Itzel Quintas‐Granados
- Colegio de Ciencias y Humanidades, Plantel CuautepecUniversidad Autónoma de la Ciudad de MéxicoCiudad de MéxicoMexico
| | - Octavio Daniel Reyes‐Hernández
- Laboratorio de Biología Molecular del Cáncer, UMIEZ, Facultad de Estudios Superiores ZaragozaUniversidad Nacional Autónoma de MéxicoCiudad de MéxicoMexico
| | - Isaac H. Caballero‐Florán
- Departamento de Farmacia, Facultad de QuímicaUniversidad Nacional Autónoma de MéxicoCiudad de MéxicoMexico
| | - Sheila I. Peña‐Corona
- Departamento de Farmacia, Facultad de QuímicaUniversidad Nacional Autónoma de MéxicoCiudad de MéxicoMexico
| | - Hernán Cortés
- Laboratorio de Medicina Genómica, Departamento de GenómicaInstituto Nacional de Rehabilitación Luis Guillermo Ibarra IbarraCiudad de MexicoMexico
| | - Gerardo Leyva‐Gómez
- Departamento de Farmacia, Facultad de QuímicaUniversidad Nacional Autónoma de MéxicoCiudad de MéxicoMexico
| | - Solomon Habtemariam
- Pharmacognosy Research & Herbal Analysis Services UKCentral Avenue, Chatham‐MaritimeKentME4 4TBUK
| | | |
Collapse
|
2
|
Amormino C, Russo E, Tedeschi V, Fiorillo MT, Paiardini A, Spallotta F, Rosanò L, Tuosto L, Kunkl M. Targeting staphylococcal enterotoxin B binding to CD28 as a new strategy for dampening superantigen-mediated intestinal epithelial barrier dysfunctions. Front Immunol 2024; 15:1365074. [PMID: 38510259 PMCID: PMC10951378 DOI: 10.3389/fimmu.2024.1365074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 02/26/2024] [Indexed: 03/22/2024] Open
Abstract
Staphylococcus aureus is a gram-positive bacterium that may cause intestinal inflammation by secreting enterotoxins, which commonly cause food-poisoning and gastrointestinal injuries. Staphylococcal enterotoxin B (SEB) acts as a superantigen (SAg) by binding in a bivalent manner the T-cell receptor (TCR) and the costimulatory receptor CD28, thus stimulating T cells to produce large amounts of inflammatory cytokines, which may affect intestinal epithelial barrier integrity and functions. However, the role of T cell-mediated SEB inflammatory activity remains unknown. Here we show that inflammatory cytokines produced by T cells following SEB stimulation induce dysfunctions in Caco-2 intestinal epithelial cells by promoting actin cytoskeleton remodelling and epithelial cell-cell junction down-regulation. We also found that SEB-activated inflammatory T cells promote the up-regulation of epithelial-mesenchymal transition transcription factors (EMT-TFs) in a nuclear factor-κB (NF-κB)- and STAT3-dependent manner. Finally, by using a structure-based design approach, we identified a SEB mimetic peptide (pSEB116-132) that, by blocking the binding of SEB to CD28, dampens inflammatory-mediated dysregulation of intestinal epithelial barrier.
Collapse
Affiliation(s)
- Carola Amormino
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
| | - Emanuela Russo
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
| | - Valentina Tedeschi
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
| | - Maria Teresa Fiorillo
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
| | - Alessandro Paiardini
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, Rome, Italy
| | - Francesco Spallotta
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
- Laboratory affiliated to Instituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Laura Rosanò
- Institute of Molecular Biology and Pathology, CNR, Rome, Italy
| | - Loretta Tuosto
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
| | - Martina Kunkl
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
3
|
Han G, Zhou S, Shen J, Yang Y, Bian X, Li Y, Ling R, Liang R, Tao M. The role of TMEM26 in disrupting tight junctions and activating NF-κB signaling to promote epithelial-mesenchymal transition in esophageal squamous cell carcinoma. Clinics (Sao Paulo) 2023; 78:100276. [PMID: 37611445 PMCID: PMC10466919 DOI: 10.1016/j.clinsp.2023.100276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/20/2023] [Accepted: 07/31/2023] [Indexed: 08/25/2023] Open
Abstract
OBJECTIVES Metastasis is one of the biggest challenges in the management of Esophageal Squamous Cell Carcinoma (ESCC), of which molecular mechanisms remain elusive. The present study aimed to explore the roles and underlying mechanisms of Transmembrane protein 26 (TMEM26) in ESCC. METHOD TMEM26 expressions in tumorous and adjacent tissues from patients with ESCC and in normal esophageal epithelial and ESCC cell lines were detected by immunostaining and western blotting, respectively. The Epithelial-Mesenchymal Transition (EMT), a critical process during metastasis, was investigated by wound healing and Transwell assays, and EMT-related proteins were examined after the TMEM26 alteration in ESCC cell lines. NF-κB signaling activation and Tight Junction (TJ) protein expression were analyzed by western blotting and immunofluorescence, respectively. In vivo verification was performed on the liver metastatic murine model. RESULTS Compared with non-cancerous esophageal tissues and cells, the TMEM26 expression level was higher in ESCC samples and cell lines, where the plasma membrane localization of TMEM26 was observed. The EMT-related processes of ESCC cells were suppressed by RNAi depletion of TMEM26 but aggravated by TMEM26 overexpression. Mechanistically, TMEM26 promoted NF-κB signaling to accelerate EMT in ESCC cells. The plasma membrane presentation and assembly of TJ proteins were impaired by TMEM26. CONCLUSION Overall, TMEM26 acts as a critical determinant for EMT in ESCC cells by disrupting TJ formation and promoting NF-κB signaling, which may be a potential therapeutic target for treating metastatic ESCC.
Collapse
Affiliation(s)
- Guohu Han
- Department of Oncology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China; Department of Oncology, Jingjiang People's Hospital Affiliated with Yangzhou University, Jingjiang, Jiangsu, China
| | - Shuangshuang Zhou
- Department of Oncology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China
| | - Junjun Shen
- Department of Oncology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China
| | - Yuanyuan Yang
- Department of Oncology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China
| | - Xuyu Bian
- Department of Oncology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China
| | - Yahu Li
- Department of Oncology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China
| | - Rui Ling
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Rongrui Liang
- Department of Oncology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China; Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Min Tao
- Department of Oncology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China; Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
4
|
Wang AG, Son M, Kenna E, Thom N, Tay S. NF-κB memory coordinates transcriptional responses to dynamic inflammatory stimuli. Cell Rep 2022; 40:111159. [PMID: 35977475 PMCID: PMC10794069 DOI: 10.1016/j.celrep.2022.111159] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/11/2022] [Accepted: 07/13/2022] [Indexed: 12/13/2022] Open
Abstract
Many scenarios in cellular communication require cells to interpret multiple dynamic signals. It is unclear how exposure to inflammatory stimuli alters transcriptional responses to subsequent stimulus. Using high-throughput microfluidic live-cell analysis, we systematically profile the NF-κB response to different signal sequences in single cells. We find that NF-κB dynamics store the short-term history of received signals: depending on the prior pathogenic or cytokine signal, the NF-κB response to subsequent stimuli varies from no response to full activation. Using information theory, we reveal that these stimulus-dependent changes in the NF-κB response encode and reflect information about the identity and dose of the prior stimulus. Small-molecule inhibition, computational modeling, and gene expression profiling show that this encoding is driven by stimulus-dependent engagement of negative feedback modules. These results provide a model for how signal transduction networks process sequences of inflammatory stimuli to coordinate cellular responses in complex dynamic environments.
Collapse
Affiliation(s)
- Andrew G Wang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA; Medical Scientist Training Program, University of Chicago, Chicago, IL 60637, USA
| | - Minjun Son
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Emma Kenna
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Nicholas Thom
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Savaş Tay
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
5
|
Wu J, Feng Z, Chen L, Li Y, Bian H, Geng J, Zheng ZH, Fu X, Pei Z, Qin Y, Yang L, Zhao Y, Wang K, Chen R, He Q, Nan G, Jiang X, Chen ZN, Zhu P. TNF antagonist sensitizes synovial fibroblasts to ferroptotic cell death in collagen-induced arthritis mouse models. Nat Commun 2022; 13:676. [PMID: 35115492 PMCID: PMC8813949 DOI: 10.1038/s41467-021-27948-4] [Citation(s) in RCA: 101] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022] Open
Abstract
Ferroptosis is a nonapoptotic cell death process that requires cellular iron and the accumulation of lipid peroxides. In progressive rheumatoid arthritis (RA), synovial fibroblasts proliferate abnormally in the presence of reactive oxygen species (ROS) and elevated lipid oxidation. Here we show, using a collagen-induced arthritis (CIA) mouse model, that imidazole ketone erastin (IKE), a ferroptosis inducer, decreases fibroblast numbers in the synovium. Data from single-cell RNA sequencing further identify two groups of fibroblasts that have distinct susceptibility to IKE-induced ferroptosis, with the ferroptosis-resistant fibroblasts associated with an increased TNF-related transcriptome. Mechanistically, TNF signaling promotes cystine uptake and biosynthesis of glutathione (GSH) to protect fibroblasts from ferroptosis. Lastly, low dose IKE together with etanercept, a TNF antagonist, induce ferroptosis in fibroblasts and attenuate arthritis progression in the CIA model. Our results thus imply that the combination of TNF inhibitors and ferroptosis inducers may serve as a potential candidate for RA therapy. Expansion of synovial fibroblast is associated with rheumatoid arthritis (RA) progression, but how this expansion is regulated is still not clear. Here the authors use a mouse RA model, single cell RNA sequencing and in vitro analyses to show that inducing ferroptosis and suppressing TNF signaling reduce fibroblast numbers and ameliorate experimental arthritis.
Collapse
Affiliation(s)
- Jiao Wu
- Department of Clinical Immunology, Xijing Hospital, Fourth Military Medical University, Xi'an, China. .,National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, China. .,Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
| | - Zhuan Feng
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Liang Chen
- School of Medicine, Shanghai University, Shanghai, China
| | - Yong Li
- Xijing 986 Hospital Department, Fourth Military Medical University, Xi'an, China.,The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Huijie Bian
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Jiejie Geng
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Zhao-Hui Zheng
- Department of Clinical Immunology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xianghui Fu
- Department of Clinical Immunology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhuo Pei
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Yifei Qin
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, China.,School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Liu Yang
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Yilin Zhao
- Department of Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ke Wang
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Ruo Chen
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Qian He
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Gang Nan
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
| | - Zhi-Nan Chen
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, China.
| | - Ping Zhu
- Department of Clinical Immunology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
6
|
Kandasamy M. NF-κB signalling as a pharmacological target in COVID-19: potential roles for IKKβ inhibitors. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:561-567. [PMID: 33394134 PMCID: PMC7780215 DOI: 10.1007/s00210-020-02035-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022]
Abstract
Coronavirus disease 2019 (COVID-19) has been characterized by lymphopenia as well as a proinflammatory cytokine storm, which are responsible for the poor prognosis and multiorgan defects. The transcription factor nuclear factor-κB (NF-κB) modulates the functions of the immune cells and alters the gene expression profile of different cytokines in response to various pathogenic stimuli, while many proinflammatory factors have been known to induce NF-κB signalling cascade. Besides, NF-κB has been known to potentiate the production of reactive oxygen species (ROS) leading to apoptosis in various tissues in many diseases and viral infections. Though the reports on the involvement of the NF-κB signalling pathway in COVID-19 are limited, the therapeutic benefits of NF-κB inhibitors including dexamethasone, a synthetic form of glucocorticoid, have increasingly been realized. Considering the fact, the abnormal activation of the NF-κB resulting from severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection might be associated with the pathogenic profile of immune cells, cytokine storm and multiorgan defects. Thus, the pharmacological inactivation of the NF-κB signalling pathway can strongly represent a potential therapeutic target to treat the symptomatology of COVID-19. This article signifies pharmacological blockade of the phosphorylation of inhibitor of nuclear factor kappa B kinase subunit beta (IKKβ), a key downstream effector of NF-κB signalling, for a therapeutic consideration to attenuate COVID-19.
Collapse
Affiliation(s)
- Mahesh Kandasamy
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620024, India.
- Faculty Recharge Programme, University Grants Commission (UGC-FRP), New Delhi, India.
| |
Collapse
|
7
|
Mashukova A, Forteza R, Shah VN, Salas PJ. The cell polarity kinase Par1b/MARK2 activation selects specific NF-kB transcripts via phosphorylation of core mediator Med17/TRAP80. Mol Biol Cell 2021; 32:690-702. [PMID: 33596087 PMCID: PMC8108508 DOI: 10.1091/mbc.e20-10-0646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Par1b/MARK2 is a Ser/Thr kinase with pleiotropic effects that participates in the generation of apico-basal polarity in Caenorhabditis elegans. It is phosphorylated by atypical PKC(ι/λ) in Thr595 and inhibited. Because previous work showed a decrease in atypical protein kinase C (aPKC) activity under proinflammatory conditions, we analyzed the hypothesis that the resulting decrease in Thr595-MARK2 with increased kinase activity may also participate in innate immunity. We confirmed that pT595-MARK2 was decreased under inflammatory stimulation. The increase in MARK2 activity was verified by Par3 delocalization and rescue with a specific inhibitor. MARK2 overexpression significantly enhanced the transcriptional activity of NF-kB for a subset of transcripts. It also resulted in phosphorylation of a single band (∼Mr 80,000) coimmunoprecipitating with RelA, identified as Med17. In vitro phosphorylation showed direct phosphorylation of Med17 in Ser152 by recombinant MARK2. Expression of S152D-Med17 mimicked the effect of MARK2 activation on downstream transcriptional regulation, which was antagonized by S152A-Med17. The decrease in pThr595 phosphorylation was validated in aPKC-deficient mouse jejunal mucosae. The transcriptional effects were confirmed in transcriptome analysis and transcript enrichment determinations in cells expressing S152D-Med17. We conclude that theMARK2-Med17 axis represents a novel form of cross-talk between polarity signaling and transcriptional regulation including, but not restricted to, innate immunity responses.
Collapse
Affiliation(s)
- Anastasia Mashukova
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL 33136.,Department of Medical Education, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314
| | - Radia Forteza
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Viraj N Shah
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Pedro J Salas
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL 33136
| |
Collapse
|
8
|
Munir S, Frøsig TM, Hansen M, Svane IM, Andersen MH. Characterization of T-cell responses against IκBα in cancer patients. Oncoimmunology 2021; 1:1290-1296. [PMID: 23243592 PMCID: PMC3518501 DOI: 10.4161/onci.21625] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The nuclear factor κ light chain enhancer of activated B cells (NFκB) is constitutively active in most cancers, controlling multiple cellular processes including proliferation, invasion and resistance to therapy. NFκB is primarily regulated through the association with inhibitory proteins that are known as inhibitors of NFκB (IκBs). Increased NFκB activity in tumor cells has been correlated with decrease stability of IκB proteins, in particular IκBα. In responso to a large number of stimuli, IκB proteins are degraded by the proteasome. Cytotoxic T lymphocytes (CTLs) recognize HLA-restricted antigenic peptides that are generated by proteasomal degradation in target cells. In the present study, we demonstrate the presence of naturally occurring IκBα -specific T cells in the peripheral blood of patients suffering from several unrelated tumor types, i.e., breast cancer, malignant melanoma and renal cell carcinoma, but not of healthy controls. Furthermore, we show that such IBα-specific T cells are granzyme B-releasing, cytotoxic cells. Hence, the increased proteasomal degradation of IκBα in cancer induces IκBα-specific CTLs.
Collapse
Affiliation(s)
- Shamaila Munir
- Center for Cancer Immune Therapy (CCIT); Department of Hematology and Oncology; Copenhagen University Hospital; Herlev, Denmark
| | | | | | | | | |
Collapse
|
9
|
Inhibition of kinase IKKβ suppresses cellular abnormalities induced by the human papillomavirus oncoprotein HPV 18E6. Sci Rep 2021; 11:1111. [PMID: 33441820 PMCID: PMC7807017 DOI: 10.1038/s41598-020-80193-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 12/15/2020] [Indexed: 11/14/2022] Open
Abstract
Human papillomavirus (HPV) is the leading cause of cervical cancer and has been implicated in several other cancer types including vaginal, vulvar, penile, and oropharyngeal cancers. Despite the recent availability of a vaccine, there are still over 310,000 deaths each year worldwide. Current treatments for HPV-mediated cancers show limited efficacy, and would benefit from improved understanding of disease mechanisms. Recently, we developed a Drosophila ‘HPV 18 E6’ model that displayed loss of cellular morphology and polarity, junctional disorganization, and degradation of the major E6 target Magi; we further provided evidence that mechanisms underlying HPV E6-induced cellular abnormalities are conserved between humans and flies. Here, we report a functional genetic screen of the Drosophila kinome that identified IKK\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\beta$$\end{document}β—a regulator of NF-κB—as an enhancer of E6-induced cellular defects. We demonstrate that inhibition of IKK\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\beta$$\end{document}β reduces Magi degradation and that this effect correlates with hyperphosphorylation of E6. Further, the reduction in IKK\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\beta$$\end{document}β suppressed the cellular transformation caused by the cooperative action of HPVE6 and the oncogenic Ras. Finally, we demonstrate that the interaction between IKK\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\beta$$\end{document}β and E6 is conserved in human cells: inhibition of IKK\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\beta$$\end{document}β blocked the growth of cervical cancer cells, suggesting that IKK\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\beta$$\end{document}β may serve as a novel therapeutic target for HPV-mediated cancers.
Collapse
|
10
|
Kunkl M, Amormino C, Frascolla S, Sambucci M, De Bardi M, Caristi S, Arcieri S, Battistini L, Tuosto L. CD28 Autonomous Signaling Orchestrates IL-22 Expression and IL-22-Regulated Epithelial Barrier Functions in Human T Lymphocytes. Front Immunol 2020; 11:590964. [PMID: 33178223 PMCID: PMC7592429 DOI: 10.3389/fimmu.2020.590964] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/22/2020] [Indexed: 12/21/2022] Open
Abstract
IL-22 is a member of the IL-10 cytokine family involved in host protection against extracellular pathogens, by promoting epithelial cell regeneration and barrier functions. Dysregulation of IL-22 production has also frequently been observed in acute respiratory distress syndrome (ARDS) and several chronic inflammatory and autoimmune diseases. We have previously described that human CD28, a crucial co-stimulatory receptor necessary for full T cell activation, is also able to act as a TCR independent signaling receptor and to induce the expression of IL-17A and inflammatory cytokines related to Th17 cells, which together with Th22 cells represent the main cellular source of IL-22. Here we characterized the role of CD28 autonomous signaling in regulating IL-22 expression in human CD4+ T cells. We show that CD28 stimulation in the absence of TCR strongly up-regulates IL-22 gene expression and secretion. As recently observed for IL-17A, we also found that CD28-mediated regulation of IL-22 transcription requires the cooperative activities of both IL-6-activated STAT3 and RelA/NF-κB transcription factors. CD28-mediated IL-22 production also promotes the barrier functions of epithelial cells by inducing mucin and metalloproteases expression. Finally, by using specific inhibitory drugs, we also identified CD28-associated class 1A phosphatidylinositol 3-kinase (PI3K) as a pivotal mediator of CD28-mediated IL-22 expression and IL-22-dependent epithelial cell barrier functions.
Collapse
Affiliation(s)
- Martina Kunkl
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Carola Amormino
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Simone Frascolla
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Manolo Sambucci
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Marco De Bardi
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Silvana Caristi
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Stefano Arcieri
- Department of Surgical Sciences, Sapienza University of Rome, Rome, Italy
| | - Luca Battistini
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Loretta Tuosto
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| |
Collapse
|
11
|
Singh A, Srivastava N, Yadav A, Ateeq B. Targeting AGTR1/NF-κB/CXCR4 axis by miR-155 attenuates oncogenesis in glioblastoma. Neoplasia 2020; 22:497-510. [PMID: 32896760 PMCID: PMC7481885 DOI: 10.1016/j.neo.2020.08.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 08/01/2020] [Accepted: 08/05/2020] [Indexed: 01/05/2023]
Abstract
Glioblastoma (GBM) represents the most aggressive malignancy of the central nervous system. Increased expression of Angiotensin II Receptor Type 1 (AGTR1) has been associated with proliferative and infiltrative properties of glioma cells. However, the underlying mechanism of AGTR1 upregulation in GBM is still unexplored. To understand the post-transcriptional regulation of AGTR1 in GBM, we screened 3'untranslated region (3'UTR) of AGTR1 for putative miRNA binding by using prediction algorithms. Interestingly, miR-155 showed conserved binding on the 3'UTR of AGTR1, subsequently confirmed by luciferase reporter assay. Furthermore, miR-155 overexpressing GBM cells show decrease in AGTR1 expression accompanied with reduced cell proliferation, invasion, foci formation and anchorage-independent growth. Strikingly, immunodeficient mice implanted with stable miR-155 overexpressing SNB19 cells show negligible tumor growth. Notably, miR-155 attenuates NF-κB signaling downstream of AGTR1 leading to reduced CXCR4 as well as AGTR1 levels. Mechanistically, miR-155 mitigates AGTR1-mediated angiogenesis, epithelial-to-mesenchymal transition, stemness, and MAPK signaling. Similar effects were observed by using pharmacological inhibitor of IκB Kinase (IKK) complex in multiple cell-based assays. Taken together, we established that miRNA-155 post-transcriptionally regulates AGTR1 expression, abrogates AGTR1/NF-κB/CXCR4 signaling axis and elicits pleiotropic anticancer effects in GBM. This study opens new avenues for using IKK inhibitors and miRNA-155 replacement therapies for the treatment of AGTR1-positive malignancies.
Collapse
MESH Headings
- Animals
- Apoptosis
- Brain Neoplasms/genetics
- Brain Neoplasms/metabolism
- Brain Neoplasms/pathology
- Cell Movement
- Cell Proliferation
- Female
- Gene Expression Regulation, Neoplastic
- Glioblastoma/genetics
- Glioblastoma/metabolism
- Glioblastoma/pathology
- Humans
- Mice
- Mice, Inbred NOD
- Mice, SCID
- MicroRNAs/genetics
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptors, CXCR4/genetics
- Receptors, CXCR4/metabolism
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Anukriti Singh
- Molecular Oncology Laboratory, Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, U.P., India; Department of Bioscience and Biotechnology, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India
| | - Nidhi Srivastava
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India
| | - Anjali Yadav
- Molecular Oncology Laboratory, Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, U.P., India
| | - Bushra Ateeq
- Molecular Oncology Laboratory, Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, U.P., India; Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, U.P., India.
| |
Collapse
|
12
|
Espinosa-Sánchez A, Suárez-Martínez E, Sánchez-Díaz L, Carnero A. Therapeutic Targeting of Signaling Pathways Related to Cancer Stemness. Front Oncol 2020; 10:1533. [PMID: 32984007 PMCID: PMC7479251 DOI: 10.3389/fonc.2020.01533] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/16/2020] [Indexed: 12/11/2022] Open
Abstract
The theory of cancer stem cells (CSCs) proposes that the different cells within a tumor, as well as metastasis deriving from it, are originated from a single subpopulation of cells with self-renewal and differentiation capacities. These cancer stem cells are supposed to be critical for tumor expansion and metastasis, tumor relapse and resistance to conventional therapies, such as chemo- and radiotherapy. The acquisition of these abilities has been attributed to the activation of alternative pathways, for instance, WNT, NOTCH, SHH, PI3K, Hippo, or NF-κB pathways, that regulate detoxification mechanisms; increase the metabolic rate; induce resistance to apoptotic, autophagic, and senescence pathways; promote the overexpression of drug transporter proteins; and activate specific stem cell transcription factors. The elimination of CSCs is an important goal in cancer therapeutic approaches because it could decrease relapses and metastatic dissemination, which are main causes of mortality in oncology patients. In this work, we discuss the role of these signaling pathways in CSCs along with their therapeutic potential.
Collapse
Affiliation(s)
- Asunción Espinosa-Sánchez
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
- CIBER de Cancer, Madrid, Spain
| | - Elisa Suárez-Martínez
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
- CIBER de Cancer, Madrid, Spain
| | - Laura Sánchez-Díaz
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
- CIBER de Cancer, Madrid, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
- CIBER de Cancer, Madrid, Spain
| |
Collapse
|
13
|
Altonsy MO, Kurwa HA, Lauzon GJ, Amrein M, Gerber AN, Almishri W, Mydlarski PR. Corynebacterium tuberculostearicum, a human skin colonizer, induces the canonical nuclear factor-κB inflammatory signaling pathway in human skin cells. Immun Inflamm Dis 2020; 8:62-79. [PMID: 31912662 PMCID: PMC7016847 DOI: 10.1002/iid3.284] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 12/19/2019] [Accepted: 12/23/2019] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Corynebacterium tuberculostearicum (C. t.) is a ubiquitous bacterium that colonizes human skin. In contrast to other members of the genus Corynebacterium, such as toxigenic Corynebacterium diphtheriae or the opportunistic pathogen Corynebacterium jeikeium, several studies suggest that C. t. may play a role in skin health and disease. However, the mechanisms underlying these effects remain poorly understood. METHODS To investigate whether C. t. induces inflammatory pathways in primary human epidermal keratinocytes (HEKs) and human cutaneous squamous carcinoma cells (SCCs), cell culture, reverse transcription-polymerase chain reaction (PCR), enzyme-linked immunosorbent assay, immunofluorescence microscopy, Western blot, chromatin immunoprecipitation-PCR, small interfering RNA knockdown and luciferase reporter expression system were used. RESULTS Herein, we demonstrate that C. t. upregulates the messenger RNA (mRNA) and protein levels of inflammatory mediators in two human skin cell lines, HEKs and SCCs. We further show activation of the canonical nuclear factor-κB (NF-κB) pathway in response to C. t. infection, including phosphorylation of the inhibitor of κB (IκB), the nuclear translocation of NF-κB subunit (NF-κB-P65 ) and the recruitment of NF-κB-P65 and RNA polymerase to the NF-κB response elements at the promoter region of the inflammatory genes. Lastly, the data confirm that C. t.-induced tumor necrosis factor mRNA expression in HEKs is toll-like receptor 2 (TLR2 ) dependent. CONCLUSION Our results offer a mechanistic model for C. t.-induced inflammation in human keratinocytes via TLR2 and activation of IκB kinase and downstream signaling through the canonical NF-κB pathway. Relevance to chronic inflammatory diseases of the skin and cutaneous oncology is discussed.
Collapse
Affiliation(s)
- Mohammed O. Altonsy
- Division of Dermatology, Department of MedicineUniversity of CalgaryCalgaryCanada
- Department of Zoology, Faculty of ScienceSohag UniversitySohagEgypt
| | - Habib A. Kurwa
- Division of Dermatology, Department of MedicineUniversity of CalgaryCalgaryCanada
| | - Gilles J. Lauzon
- Division of Dermatology, Department of MedicineUniversity of CalgaryCalgaryCanada
| | - Matthias Amrein
- Department of Cell Biology and AnatomyUniversity of CalgaryCalgaryCanada
| | - Anthony N. Gerber
- Department of MedicineNational Jewish HealthDenverColorado
- Department of MedicineUniversity of ColoradoDenverColorado
| | - Wagdi Almishri
- Division of Gastroenterology, Department of MedicineUniversity of CalgaryCalgaryCanada
| | | |
Collapse
|
14
|
Chen A, Koehler AN. Transcription Factor Inhibition: Lessons Learned and Emerging Targets. Trends Mol Med 2020; 26:508-518. [PMID: 32359481 DOI: 10.1016/j.molmed.2020.01.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/10/2020] [Accepted: 01/21/2020] [Indexed: 12/15/2022]
Abstract
Transcription factors have roles at focal points in signaling pathways, controlling many normal cellular processes, such as cell growth and proliferation, metabolism, apoptosis, immune responses, and differentiation. Their activity is frequently deregulated in disease and targeting this class of proteins is a major focus of interest. However, the structural disorder and lack of binding pockets have made design of small molecules for transcription factors challenging. Here, we review some of the most recent developments for small molecule inhibitors of transcription factors emphasized in James Darnell's vision 17 years ago. We also discuss the progress so far on transcription factors recently nominated by genome-scale loss-of-function screens from the cancer dependency map project.
Collapse
Affiliation(s)
- Andrew Chen
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, MA 02142, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, MA 02139, USA; MIT Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts, MA 02142, USA
| | - Angela N Koehler
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, MA 02142, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, MA 02139, USA; MIT Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts, MA 02142, USA; Broad Institute of MIT and Harvard, Cambridge, Massachusetts, MA 02142, USA.
| |
Collapse
|
15
|
Lung HL, Kan R, Chau WY, Man OY, Mak NK, Fong CH, Shuen WH, Tsao SW, Lung ML. The anti-tumor function of the IKK inhibitor PS1145 and high levels of p65 and KLF4 are associated with the drug resistance in nasopharyngeal carcinoma cells. Sci Rep 2019; 9:12064. [PMID: 31427673 PMCID: PMC6700134 DOI: 10.1038/s41598-019-48590-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 08/06/2019] [Indexed: 11/09/2022] Open
Abstract
We and others have previously shown that the canonical nuclear factor kappa-B (NF-κB) pathway is essential to nasopharyngeal carcinoma (NPC) tumor development and angiogenesis, suggesting that the NF-κB pathway, including its upstream modulators and downstream effectors, are potential therapeutic targets for NPC. The inhibitor of upstream IκB kinase (IKK), PS1145, is a small molecule which can specifically inhibit the IκB phosphorylation and degradation and the subsequent nuclear translocation of NF-κB. The present study aims to determine the anti-tumor activity of PS1145 on NPC. Our results showed that PS1145 significantly inhibited the growth of tumorigenic NPC cell lines, but not in the normal nasopharyngeal epithelial cell line. Results in the in vivo study showed that low concentration of PS1145 (3 mg/kg) could significantly suppress the subcutaneous tumor formation in the nude mice bearing NPC xenografts. Apparent adverse effects were not observed in the animal study. Drug resistance against PS1145 seems to be associated with the increased levels of active NF-kB p65 and change of expression levels of kruppel-like factor 4. As can be seen, PS1145 appears to be a safe agent for animal experiments and its effects are tumor-specific, and the proteins associated with the drug resistance of PS1145 are implied.
Collapse
Affiliation(s)
- Hong Lok Lung
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong (SAR), P.R. China.
| | - Rebecca Kan
- Department of Clinical Oncology, University of Hong Kong, Pokfulam, Hong Kong (SAR), P.R. China.,Ketchum Pte. Ltd., 30 Merchant Road, Riverside Point, #03-12, Singapore, Singapore
| | - Wai Yin Chau
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong (SAR), P.R. China
| | - On Ying Man
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong (SAR), P.R. China
| | - Nai Ki Mak
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong (SAR), P.R. China.,Center for Nasopharyngeal Carcinoma Research, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pok Fu Lam, Hong Kong (SAR), P.R. China
| | - Chun Hung Fong
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong (SAR), P.R. China.,Department of Clinical Oncology, University of Hong Kong, Pokfulam, Hong Kong (SAR), P.R. China
| | - Wai Ho Shuen
- Department of Clinical Oncology, University of Hong Kong, Pokfulam, Hong Kong (SAR), P.R. China.,Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Sai Wah Tsao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, P.R. China.,Center for Nasopharyngeal Carcinoma Research, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pok Fu Lam, Hong Kong (SAR), P.R. China
| | - Maria Li Lung
- Department of Clinical Oncology, University of Hong Kong, Pokfulam, Hong Kong (SAR), P.R. China. .,Center for Nasopharyngeal Carcinoma Research, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pok Fu Lam, Hong Kong (SAR), P.R. China.
| |
Collapse
|
16
|
Marino S, Bishop RT, Carrasco G, Logan JG, Li B, Idris AI. Pharmacological Inhibition of NFκB Reduces Prostate Cancer Related Osteoclastogenesis In Vitro and Osteolysis Ex Vivo. Calcif Tissue Int 2019; 105:193-204. [PMID: 30929064 DOI: 10.1007/s00223-019-00538-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 02/20/2019] [Indexed: 12/12/2022]
Abstract
NFκB is implicated in cancer and bone remodelling, and we have recently reported that the verified NFκB inhibitor Parthenolide (PTN) reduced osteolysis and skeletal tumour growth in models of metastatic breast cancer. Here, we took advantage of in vitro and ex vivo bone cell and organ cultures to study the effects of PTN on the ability of prostate cancer cells and their derived factors to regulate bone cell activity and osteolysis. PTN inhibited the in vitro growth of a panel of human, mouse and rat prostate cancer cells in a concentration-dependent manner with a varying degree of potency. In prostate cancer cell-osteoclast co-cultures, the rat Mat-Ly-Lu, but not human PC3 or mouse RM1-BT, enhanced RANKL stimulated osteoclast formation and PTN reduced these effects without affecting prostate cancer cell viability. In the absence of cancer cells, PTN reduced the support of Mat-Ly-Lu conditioned medium for the adhesion and spreading of osteoclast precursors, and survival of mature osteoclasts. Pre-exposure of osteoblasts to PTN prior to the addition of conditioned medium from Mat-Ly-Lu cells suppressed their ability to support the formation of osteoclasts by inhibition of RANKL/OPG ratio. PTN enhanced the ability of Mat-Ly-Lu derived factors to increase calvarial osteoblast differentiation and growth. Ex vivo, PTN enhanced bone volume in calvaria organ-Mat-Ly-Lu cell co-culture, without affecting Mat-Ly-Lu viability or apoptosis. Mechanistic studies in osteoclasts and osteoblasts confirmed that PTN inhibit NFκB activation related to derived factors from Mat-Ly-Lu cells. Collectively, these findings suggest that pharmacological inhibition of the skeletal NFκB signalling pathway reduces prostate cancer related osteolysis, but further studies in the therapeutic implications of NFκB inhibition in cells of the osteoblastic lineage are needed.
Collapse
Affiliation(s)
- Silvia Marino
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
- Bone and Cancer Group, Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XR, UK
| | - Ryan T Bishop
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Giovana Carrasco
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - John G Logan
- Bone and Cancer Group, Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XR, UK
| | - Boya Li
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Aymen I Idris
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK.
- Bone and Cancer Group, Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XR, UK.
| |
Collapse
|
17
|
Kunkl M, Mastrogiovanni M, Porciello N, Caristi S, Monteleone E, Arcieri S, Tuosto L. CD28 Individual Signaling Up-regulates Human IL-17A Expression by Promoting the Recruitment of RelA/NF-κB and STAT3 Transcription Factors on the Proximal Promoter. Front Immunol 2019; 10:864. [PMID: 31068940 PMCID: PMC6491678 DOI: 10.3389/fimmu.2019.00864] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 04/04/2019] [Indexed: 01/22/2023] Open
Abstract
CD28 is an important co-stimulatory receptor for T lymphocytes that, in humans, delivers TCR-independent signal leading to the up-regulation of pro-inflammatory cytokines. We have recently reported that CD28 autonomous signaling induces the expression of IL-17A in peripheral CD4+ T lymphocytes from healthy donors, multiple sclerosis, and type 1 diabetes patients. Due to the relevance of IL-17A in the pathophysiology of several inflammatory and autoimmune diseases, we characterized the mechanisms and signaling mediators responsible for CD28-induced IL-17A expression. Here we show that CD28-mediated up-regulation of IL-17A gene expression depends on RelA/NF-κB and IL-6-associated STAT3 transcriptions factors. In particular, we found that CD28-activated RelA/NF-κB induces the expression of IL-6 that, in a positive feedback loop, mediates the activation and nuclear translocation of tyrosine phosphorylated STAT3 (pSTAT3). pSTAT3 in turn cooperates with RelA/NF-κB by binding specific sequences within the proximal promoter of human IL-17A gene, thus inducing its expression. Finally, by using specific inhibitory drugs, we also identified class 1A phosphatidylinositol 3-kinase (PI3K) as a critical upstream regulator of CD28-mediated RelA/NF-κB and STAT3 recruitments and trans-activation of IL-17A promoter. Our findings reveal a novel mechanism by which human CD28 may amplify IL-17A expression in human T lymphocytes and provide biological bases for immunotherapeutic approaches targeting CD28-associated class 1A PI3K to dampen IL-17A-mediated inflammatory response in autoimmune/inflammatory disorders.
Collapse
Affiliation(s)
- Martina Kunkl
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Marta Mastrogiovanni
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy.,Lymphocyte Cell Biology Unit, INSERM U1221, Department of Immunology, Pasteur Institute, Paris, France
| | - Nicla Porciello
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Silvana Caristi
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Emanuele Monteleone
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Stefano Arcieri
- Department of Surgical Sciences, Sapienza University of Rome, Rome, Italy
| | - Loretta Tuosto
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| |
Collapse
|
18
|
Longitudinal tracking of subpopulation dynamics and molecular changes during LNCaP cell castration and identification of inhibitors that could target the PSA-/lo castration-resistant cells. Oncotarget 2017; 7:14220-40. [PMID: 26871947 PMCID: PMC4924710 DOI: 10.18632/oncotarget.7303] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 01/29/2016] [Indexed: 12/02/2022] Open
Abstract
We have recently demonstrated that the undifferentiated PSA−/lo prostate cancer (PCa) cell population harbors self-renewing long-term tumor-propagating cells that are refractory to castration, thus representing a therapeutic target. Our goals here are, by using the same lineage-tracing reporter system, to track the dynamic changes of PSA−/lo and PSA+ cells upon castration in vitro, investigate the molecular changes accompanying persistent castration, and develop large numbers of PSA−/lo PCa cells for drug screening. To these ends, we treated LNCaP cells infected with the PSAP-GFP reporter with three regimens of castration, i.e., CDSS, CDSS plus bicalutamide, and MDV3100 continuously for up to ~21 months. We observed that in the first ~7 months, castration led to time-dependent increases in PSA−/lo cells, loss of AR and PSA expression, increased expression of cancer stem cell markers, and many other molecular changes. Meanwhile, castrated LNCaP cells became resistant to high concentrations of MDV3100, chemotherapeutic drugs, and other agents. However, targeted and medium-throughput library screening identified several kinase (e.g., IGF-1R, AKT, PI3K/mTOR, Syk, GSK3) inhibitors as well as the BCL2 inhibitor that could effectively sensitize the LNCaP-CRPC cells to killing. Of interest, LNCaP cells castrated for >7 months showed evidence of cyclic changes in AR and the mTOR/AKT signaling pathways potentially involving epigenetic mechanisms. These observations indicate that castration elicits numerous molecular changes and leads to enrichment of PSA−/lo PCa cells. The ability to generate large numbers of PSA−/lo PCa cells should allow future high-throughput screening to identify novel therapeutics that specifically target this population.
Collapse
|
19
|
Jin R, Chen Q, Yao S, Bai E, Fu W, Wang L, Wang J, Du X, Wei T, Xu H, Jiang C, Qiu P, Wu J, Li W, Liang G. Synthesis and anti-tumor activity of EF24 analogues as IKKβ inhibitors. Eur J Med Chem 2017; 144:218-228. [PMID: 29351887 DOI: 10.1016/j.ejmech.2017.11.077] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/26/2017] [Accepted: 11/27/2017] [Indexed: 12/16/2022]
Abstract
EF24 is an IKKβ inhibitor (IC50: 72 μM) containing various anti-tumor activities. In this study, a series of EF24 analogs targeting IKKβ were designed and synthesized. Several IKKβ inhibitors with better activities than EF24 were screened out and B3 showed best IKKβ inhibitory (IC50: 6.6 μM). Molecular docking and dynamic simulation experiments further confirmed this inhibitory effect. B3 obviously suppressed the viability of Hela229, A549, SGC-7901 and MGC-803 cells. Then, in SGC-7901 and MGC-803 cells, B3 blocked the NF-κB signal pathway by inhibiting IKKβ phosphorylation, and followed arrested the cell cycle at G2/M phase by suppressing the Cyclin B1 and Cdc2 p34 expression, induced the cell apoptosis by down-regulating Bcl-2 protein and up-regulating cleaved-caspase3. Moreover, B3 significantly reduced tumor growth and suppressed the IKKβ-NF-κB signal pathway in SGC-7901 xenograft model. In total, this study present a potential IKKβ inhibitor as anti-tumor precursor.
Collapse
Affiliation(s)
- Rong Jin
- Department of Digestive Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Qiuxiang Chen
- Department of Ultrasound, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Song Yao
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Encheng Bai
- Department of Digestive Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Weitao Fu
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Ledan Wang
- Department of Gynecology and Obstetrics, The 2nd Affiliated Hospital of the Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jiabing Wang
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaojing Du
- Department of Digestive Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Tao Wei
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Haineng Xu
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Wenzhou Biomedical Innovation Center, Wenzhou University and Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Chengxi Jiang
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Wenzhou Biomedical Innovation Center, Wenzhou University and Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Peihong Qiu
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jianzhang Wu
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Wenzhou Biomedical Innovation Center, Wenzhou University and Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Wulan Li
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; College of Information Science and Computer Engineering, The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Guang Liang
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Wenzhou Biomedical Innovation Center, Wenzhou University and Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| |
Collapse
|
20
|
Identification of 11(13)-dehydroivaxillin as a potent therapeutic agent against non-Hodgkin's lymphoma. Cell Death Dis 2017; 8:e3050. [PMID: 28906487 PMCID: PMC5636986 DOI: 10.1038/cddis.2017.442] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 07/15/2017] [Accepted: 07/31/2017] [Indexed: 12/11/2022]
Abstract
Despite great advancements in the treatment of non-Hodgkin lymphoma (NHL), sensitivity of different subtypes to therapy varies. Targeting the aberrant activation NF-κB signaling pathways in lymphoid malignancies is a promising strategy. Here, we report that 11(13)-dehydroivaxillin (DHI), a natural compound isolated from the Carpesium genus, induces growth inhibition and apoptosis of NHL cells. Multiple signaling cascades are influenced by DHI in NHL cells. PI3K/AKT and ERK are activated or inhibited in a cell type dependent manner, whereas NF-κB signaling pathway was inhibited in all the NHL cells tested. Applying the cellular thermal shift assay, we further demonstrated that DHI directly interacts with IKKα/IKKβ in NHL cells. Interestingly, DHI treatment also reduced the IKKα/IKKβ protein level in NHL cells. Consistent with this finding, knockdown of IKKα/IKKβ inhibits cell proliferation and enhances DHI-induced proliferation inhibition. Overexpression of p65, p52 or RelB partially reverses DHI-induced cell growth inhibition. Furthermore, DHI treatment significantly inhibits the growth of NHL cell xenografts. In conclusion, we demonstrate that DHI exerts anti-NHL effect in vitro and in vivo, through a cumulative effect on NF-κB and other pathways. DHI may serve as a promising lead compound for the therapy of NHL.
Collapse
|
21
|
Pippione AC, Federico A, Ducime A, Sainas S, Boschi D, Barge A, Lupino E, Piccinini M, Kubbutat M, Contreras JM, Morice C, Al-Karadaghi S, Lolli ML. 4-Hydroxy- N-[3,5-bis(trifluoromethyl)phenyl]-1,2,5-thiadiazole-3-carboxamide: a novel inhibitor of the canonical NF-κB cascade. MEDCHEMCOMM 2017; 8:1850-1855. [PMID: 30108896 DOI: 10.1039/c7md00278e] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 08/22/2017] [Indexed: 12/21/2022]
Abstract
The NF-κB signaling pathway is a validated oncological target. Here, we applied scaffold hopping to IMD-0354, a presumed IKKβ inhibitor, and identified 4-hydroxy-N-[3,5-bis(trifluoromethyl)phenyl]-1,2,5-thiadiazole-3-carboxamide (4) as a nM-inhibitor of the NF-κB pathway. However, both 4 and IMD-0354, being potent inhibitors of the canonical NF-κB pathway, were found to be inactive in human IKKβ enzyme assays.
Collapse
Affiliation(s)
- Agnese C Pippione
- Department of Science and Drug Technology , University of Torino , via Pietro Giuria 9 , 10125 Torino , Italy
| | - Antonella Federico
- Department of Science and Drug Technology , University of Torino , via Pietro Giuria 9 , 10125 Torino , Italy
| | - Alex Ducime
- Department of Science and Drug Technology , University of Torino , via Pietro Giuria 9 , 10125 Torino , Italy
| | - Stefano Sainas
- Department of Science and Drug Technology , University of Torino , via Pietro Giuria 9 , 10125 Torino , Italy
| | - Donatella Boschi
- Department of Science and Drug Technology , University of Torino , via Pietro Giuria 9 , 10125 Torino , Italy
| | - Alessandro Barge
- Department of Science and Drug Technology , University of Torino , via Pietro Giuria 9 , 10125 Torino , Italy
| | - Elisa Lupino
- Department of Oncology , University of Torino , via Michelangelo 27/B , 10126 Torino , Italy
| | - Marco Piccinini
- Department of Oncology , University of Torino , via Michelangelo 27/B , 10126 Torino , Italy
| | | | - Jean-Marie Contreras
- Prestwick Chemical , 220 Boulevard Gonthier d'Andernach , 67400 Illkirch , France
| | - Christophe Morice
- Prestwick Chemical , 220 Boulevard Gonthier d'Andernach , 67400 Illkirch , France
| | - Salam Al-Karadaghi
- SARomics Biostructures and Department of Biochemistry & Structural Biology , Lund University , Lund , Sweden
| | - Marco L Lolli
- Department of Science and Drug Technology , University of Torino , via Pietro Giuria 9 , 10125 Torino , Italy
| |
Collapse
|
22
|
Wang YX, Liu L, Zeng QX, Fan TY, Jiang JD, Deng HB, Song DQ. Synthesis and Identification of Novel Berberine Derivatives as Potent Inhibitors against TNF-α-Induced NF-κB Activation. Molecules 2017; 22:molecules22081257. [PMID: 28749438 PMCID: PMC6152030 DOI: 10.3390/molecules22081257] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 07/24/2017] [Accepted: 07/25/2017] [Indexed: 02/04/2023] Open
Abstract
Twenty-three new berberine (BBR) analogues defined on substituents of ring D were synthesized and evaluated for their activity for suppression of tumor necrosis factor (TNF)-α-induced nuclear factor (NF)-κB activation. Structure–activity relationship (SAR) analysis indicated that suitable tertiary/quaternary carbon substitutions at the 9-position or rigid fragment at position 10 might be beneficial for enhancing their anti-inflammatory potency. Among them, compounds 2d, 2e, 2i and 2j exhibited satisfactory inhibitory potency against NF-κB activation, with an inhibitory rate of around 90% (5 μM), much better than BBR. A preliminary mechanism study revealed that all of them could inhibit TNF-α-induced NF-κB activation via impairing IκB kinase (IKK) phosphorylation as well as cytokines interleukin (IL)-6 and IL-8 induced by TNF-α. Therefore, the results provided powerful information on further structural modifications and development of BBR derivatives into a new class of anti-inflammatory candidates for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Yan-Xiang Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China.
| | - Lu Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China.
| | - Qing-Xuan Zeng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China.
| | - Tian-Yun Fan
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China.
| | - Jian-Dong Jiang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China.
| | - Hong-Bin Deng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China.
| | - Dan-Qing Song
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
23
|
Tan ZH, Zhang Y, Tian Y, Tan W, Li YH. IκB kinase b Mediating the Downregulation of p53 and p21 by Lipopolysaccharide in Human Papillomavirus 16 + Cervical Cancer Cells. Chin Med J (Engl) 2017; 129:2703-2707. [PMID: 27824003 PMCID: PMC5126162 DOI: 10.4103/0366-6999.193463] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background: Cervical cancer is the second most common cancer of woman in the world, and human papillomavirus (HPV) infection plays an important role in the development of most of the cases. IκB kinase β (IKKβ) is a kinase-mediating nuclear factor kappa B (NF-κB) activation by phosphorylating the inhibitor of NF-κB (IκB) and is related by some diseases caused by virus infection. However, there is little known about the correlation between IKKβ and HPV infection in cervical cancer. This study aimed to investigate the expression of IKKβ protein in cervical cancer tissues and effects of inflammation on HPV positive or negative cervical cancer cells through detecting the expression of IKKβ, IκBα, p53, and p21 proteins after treated with lipopolysaccharide (LPS) to mimic bacterial infection. We also examined the effects of LPS on cervical cancer cells after blocking IKKβ with pharmacological inhibitor. Methods: Thirty-six matched specimens of cervical cancer and adjacent normal tissues were collected and analyzed in the study. The expression of IKKβ in the tissue specimens was determined by immunohistochemical staining. In addition, Western blot was used to detect the expression level changes of IKKβ, IκBα, p53, and p21 after LPS stimulated in the HPV16+ (SiHa) and HPV16− (C33A) cervical cancer cell lines. Furthermore, the effects of IKKβ inhibitor SC-514 on LPS-induced expression change of these proteins were investigated. Results: The expression of IKKβ was higher in cervical cancer than adjacent normal tissues, and there was no significant difference between tumor differentiation, size, and invasive depth with IKKβ expression. The LPS, which increased the expression level of IKKβ protein but decreased in the IκBα, p53 and p21 proteins, was illustrated in HPV16+ (SiHa) but not in HPV16− (C33A) cells. Moreover, IKKβ inhibitor SC-514 totally reversed the upregulation of IKKβ and downregulation of p53 and p21 by LPS in SiHa cells. Conclusions: IKKβ may mediate the downregulation of p53 and p21 by LPS in HPV16+ cervical cancer cells.
Collapse
Affiliation(s)
- Zhi-Hui Tan
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China
| | - Yu Zhang
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China
| | - Yan Tian
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China
| | - Wei Tan
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China
| | - Ying-Hua Li
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China
| |
Collapse
|
24
|
Shukla S, Kanwal R, Shankar E, Datt M, Chance MR, Fu P, MacLennan GT, Gupta S. Apigenin blocks IKKα activation and suppresses prostate cancer progression. Oncotarget 2016; 6:31216-32. [PMID: 26435478 PMCID: PMC4741599 DOI: 10.18632/oncotarget.5157] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 08/24/2015] [Indexed: 12/27/2022] Open
Abstract
IKKα has been implicated as a key regulator of oncogenesis and driver of the metastatic process; therefore is regarded as a promising therapeutic target in anticancer drug development. In spite of the progress made in the development of IKK inhibitors, no potent IKKα inhibitor(s) have been identified. Our multistep approach of molecular modeling and direct binding has led to the identification of plant flavone apigenin as a specific IKKα inhibitor. Here we report apigenin, in micro molar range, inhibits IKKα kinase activity, demonstrates anti-proliferative and anti-invasive activities in functional cell based assays and exhibits anticancer efficacy in experimental tumor model. We found that apigenin directly binds with IKKα, attenuates IKKα kinase activity and suppresses NF-ĸB/p65 activation in human prostate cancer PC-3 and 22Rv1 cells much more effectively than IKK inhibitor, PS1145. We also showed that apigenin caused cell cycle arrest similar to knockdown of IKKα in prostate cancer cells. Studies in xenograft mouse model indicate that apigenin feeding suppresses tumor growth, lowers proliferation and enhances apoptosis. These effects correlated with inhibition of p-IKKα, NF-ĸB/p65, proliferating cell nuclear antigen and increase in cleaved caspase 3 expression in a dose-dependent manner. Overall, our results suggest that inhibition of cell proliferation, invasiveness and decrease in tumor growth by apigenin are mediated by its ability to suppress IKKα and downstream targets affecting NF-ĸB signaling pathways.
Collapse
Affiliation(s)
- Sanjeev Shukla
- Department of Urology, Case Western Reserve University & University Hospitals Case Medical Center, Cleveland, Ohio 44106, USA.,The Urology Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106, USA
| | - Rajnee Kanwal
- Department of Urology, Case Western Reserve University & University Hospitals Case Medical Center, Cleveland, Ohio 44106, USA.,The Urology Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106, USA
| | - Eswar Shankar
- Department of Urology, Case Western Reserve University & University Hospitals Case Medical Center, Cleveland, Ohio 44106, USA.,The Urology Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106, USA
| | - Manish Datt
- Center for Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Mark R Chance
- Center for Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Pingfu Fu
- Department of Epidemiology & Biostatistics, Case Western Reserve University & University Hospitals Case Medical Center, Cleveland, Ohio 44106, USA
| | - Gregory T MacLennan
- Department of Pathology, Case Western Reserve University & University Hospitals Case Medical Center, Cleveland, Ohio 44106, USA
| | - Sanjay Gupta
- Department of Urology, Case Western Reserve University & University Hospitals Case Medical Center, Cleveland, Ohio 44106, USA.,The Urology Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106, USA.,Department of Nutrition, Case Western Reserve University & University Hospitals Case Medical Center, Cleveland, Ohio 44106, USA.,Divison of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, Ohio 44106, USA.,Department of Urology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio 44106, USA
| |
Collapse
|
25
|
Jayaraman S, Doucet M, Lau WM, Kominsky SL. CITED2 Modulates Breast Cancer Metastatic Ability through Effects on IKKα. Mol Cancer Res 2016; 14:730-9. [PMID: 27216153 PMCID: PMC4987170 DOI: 10.1158/1541-7786.mcr-16-0081] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/14/2016] [Indexed: 12/31/2022]
Abstract
UNLABELLED Previously, we identified the transcriptional coactivator CITED2 as a potential facilitator of bone metastasis using a murine mammary cancer model. Extending these studies to human breast cancer, it was observed that CITED2 mRNA expression was significantly elevated in patient specimens of metastatic breast cancer relative to primary tumors, with highest levels in metastasis to bone relative to non-bone sites. To further evaluate CITED2 functions in breast cancer metastasis, CITED2 expression was stably reduced in the human breast cancer cell lines MDA-MB-231 and MDA-MB-468, which are metastatic in animal models. While CITED2 knockdown had no effect on cell proliferation, cell migration and invasion were significantly reduced, as was the establishment of metastasis following intracardiac administration in athymic nude mice. To explore the mechanism behind these effects, gene expression following CITED2 knockdown in MDA-MB-231 cells by cDNA microarray was performed. As confirmed at the mRNA and protein levels in both MDA-MB-231 and MDA-MB-468 cells, expression of the NF-κB regulator IKKα was significantly reduced, along with several NF-κB targets with known roles in metastasis (OPN, MMP9, uPA, SPARC, IL11, and IL1β). Furthermore, ChIP assay revealed recruitment of CITED2 to the promoter of IKKα, indicating a direct role in regulating its expression. Consistent with reduced IKKα expression, CITED2 knockdown inhibited both canonical and noncanonical NF-κB signaling. Finally, restoration of IKKα expression following CITED2 knockdown in MDA-MB-231 and MDA-MB-468 cells rescued their invasive ability. Collectively, these data demonstrate that CITED2 modulates metastatic ability in human breast cancer cells, at least in part, through the regulation of IKKα. IMPLICATIONS The current study highlights the role of CITED2 in facilitating breast cancer metastasis, partly via regulation of IKKα. Mol Cancer Res; 14(8); 730-9. ©2016 AACR.
Collapse
Affiliation(s)
- Swaathi Jayaraman
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michele Doucet
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Wen Min Lau
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Scott L Kominsky
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
26
|
Han C, Wu W, Ale A, Kim MS, Cai D. Central Leptin and Tumor Necrosis Factor-α (TNFα) in Diurnal Control of Blood Pressure and Hypertension. J Biol Chem 2016; 291:15131-42. [PMID: 27226618 DOI: 10.1074/jbc.m116.730408] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Indexed: 12/21/2022] Open
Abstract
Leptin and TNFα can individually work in the brain to affect blood pressure; however, it remains unknown whether these two cytokines might have an interactive role in this process and, if so, how. In this work, we found that leptin stimulation led to TNFα production under both in vitro and in vivo conditions, and diurnal fluctuation of leptin concentrations in the cerebrospinal fluid predicted the circadian changes of TNFα gene expression in the hypothalamus. Signaling analysis showed that leptin stimulation led to a rapid and strong STAT3 activation followed by a second-phase moderate STAT3 activation, which was selectively abolished by anti-inflammatory chemical PS1145 or TNFα antagonist WP9QY. Physiological study in normal mice revealed that diurnal rise of blood pressure was abrogated following central administration of PS1145 or a leptin receptor antagonist. Central TNFα pretreatment was found to potentiate the effect of leptin in elevating blood pressure in normal mice. In pathophysiology, dietary obesity mimicked TNFα pretreatment in promoting leptin-induced blood pressure rise, and this effect was blocked by central treatment with either PS1145 or WP9QY. Hence, central leptin employs TNFα to mediate the diurnal blood pressure elevation in physiology while enhancement of this mechanism can contribute to hypertension development.
Collapse
Affiliation(s)
- Cheng Han
- From the Department of Molecular Pharmacology, Diabetes Research Center, Institute of Aging, Albert Einstein College of Medicine, Bronx, New York 10461 and
| | - Wenhe Wu
- From the Department of Molecular Pharmacology, Diabetes Research Center, Institute of Aging, Albert Einstein College of Medicine, Bronx, New York 10461 and Key Laboratory of Laboratory Medicine, Zhejiang Provincial Key Laboratory of Medical Genetics, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Albert Ale
- From the Department of Molecular Pharmacology, Diabetes Research Center, Institute of Aging, Albert Einstein College of Medicine, Bronx, New York 10461 and
| | - Min Soo Kim
- From the Department of Molecular Pharmacology, Diabetes Research Center, Institute of Aging, Albert Einstein College of Medicine, Bronx, New York 10461 and
| | - Dongsheng Cai
- From the Department of Molecular Pharmacology, Diabetes Research Center, Institute of Aging, Albert Einstein College of Medicine, Bronx, New York 10461 and
| |
Collapse
|
27
|
Zhang Y, Lapidus RG, Liu P, Choi EY, Adediran S, Hussain A, Wang X, Liu X, Dan HC. Targeting IκB Kinase β/NF-κB Signaling in Human Prostate Cancer by a Novel IκB Kinase β Inhibitor CmpdA. Mol Cancer Ther 2016; 15:1504-14. [PMID: 27196761 DOI: 10.1158/1535-7163.mct-15-0999] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 03/28/2016] [Indexed: 12/13/2022]
Abstract
NF-κB plays an important role in many types of cancer, including prostate cancer, but the role of the upstream kinase of NF-κB, IKKβ, in prostate cancer has neither been fully documented nor are there any effective IKKβ inhibitors used in clinical settings. Here, we have shown that IKKβ activity is mediated by multiple kinases including IKKα in human prostate cancer cell lines that express activated IKKβ. IHC analysis (IHC) of human prostate cancer tissue microarrays (TMA) demonstrates that phosphorylation of IKKα/β within its activation loop gradually increases in low to higher stage tumors as compared with normal tissue. The expression of cell proliferation and survival markers (Ki-67, Survivin) and epithelial-to-mesenchymal transition (EMT) markers (Slug, Snail), as well as cancer stem cell (CSC)-related transcription factors (Nanog, Sox2, Oct-4), also increase in parallel among the respective TMA samples analyzed. IKKβ, but not NF-κB, is found to regulate Nanog, which, in turn, modulates the levels of Oct4, Sox2, Snail, and Slug, indicating an essential role of IKKβ in regulating CSCs and EMT. The novel IKKβ inhibitor CmpdA inhibits constitutively activated IKKβ/NF-κB signaling, leading to induction of apoptosis and inhibition of proliferation, migration, and stemness in these cells. CmpdA also significantly inhibits tumor growth in xenografts without causing apparent in vivo toxicity. Furthermore, CmpdA and docetaxel act synergistically to inhibit proliferation of prostate cancer cells. These results indicate that IKKβ plays a pivotal role in prostate cancer, and targeting IKKβ, including in combination with docetaxel, may be a potentially useful strategy for treating advanced prostate cancer. Mol Cancer Ther; 15(7); 1504-14. ©2016 AACR.
Collapse
Affiliation(s)
- Yanting Zhang
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Rena G Lapidus
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Peiyan Liu
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Eun Yong Choi
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Samusi Adediran
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Arif Hussain
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland. Baltimore VA Medical Center, Baltimore, Maryland
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| | - Xuefeng Liu
- Department of Pathology, Georgetown University Medical Center, Washington, DC
| | - Han C Dan
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland. Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland.
| |
Collapse
|
28
|
Jin R, Xia Y, Chen Q, Li W, Chen D, Ye H, Zhao C, Du X, Shi D, Wu J, Liang G. Da0324, an inhibitor of nuclear factor-κB activation, demonstrates selective antitumor activity on human gastric cancer cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:979-95. [PMID: 27042000 PMCID: PMC4780725 DOI: 10.2147/dddt.s90081] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background The transcription factor nuclear factor-κB (NF-κB) is constitutively activated in a variety of human cancers, including gastric cancer. NF-κB inhibitors that selectively kill cancer cells are urgently needed for cancer treatment. Curcumin is a potent inhibitor of NF-κB activation. Unfortunately, the therapeutic potential of curcumin is limited by its relatively low potency and poor cellular bioavailability. In this study, we presented a novel NF-κB inhibitor named Da0324, a synthetic asymmetric mono-carbonyl analog of curcumin. The purpose of this study is to research the expression of NF-κB in gastric cancer and the antitumor activity and mechanism of Da0324 on human gastric cancer cells. Methods The expressions between gastric cancer tissues/cells and normal gastric tissues/cells of NF-κB were evaluated by Western blot. The inhibition viability of compounds on human gastric cancer cell lines SGC-7901, BGC-823, MGC-803, and normal gastric mucosa epithelial cell line GES-1 was assessed with the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide assay. Absorption spectrum method and high-performance liquid chromatography method detected the stability of the compound in vitro. The compound-induced changes of inducible NF-κB activation in the SGC-7901 and BGC-823 cells were examined by Western blot analysis and immunofluorescence methods. The antitumor activity of compound was performed by clonogenic assay, matrigel invasion assay, flow cytometric analysis, Western blot analysis, and Hoechst 33258 staining assay. Results High levels of p65 were found in gastric cancer tissues and cells. Da0324 displayed higher growth inhibition against several types of gastric cancer cell lines and showed relatively low toxicity to GES-1. Moreover, Da0324 was more stable than curcumin in vitro. Western blot analysis and immunofluorescence methods showed that Da0324 blocked NF-κB activation. In addition, Da0324 significantly inhibited tumor proliferation and invasion, arrested the cell cycle, and induced apoptosis in vitro. Conclusion The asymmetric mono-carbonyl analog of curcumin Da0324 exhibited significantly improved antigastric cancer activity. Da0324 may be a promising NF-κB inhibitor for the selective targeting of cancer cells. However, further studies are needed in animals to validate these findings for the therapeutic use of Da0324.
Collapse
Affiliation(s)
- Rong Jin
- Department of Digestive Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China; Department of Epidemiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Yiqun Xia
- Department of Digestive Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Qiuxiang Chen
- Department of Digestive Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Wulan Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China; College of Information Science and Computer Engineering, Wenzhou Medical College, Wenzhou, People's Republic of China
| | - Dahui Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Hui Ye
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China; School of Basic Medical Sciences, Wenzhou Medical College, Wenzhou, People's Republic of China
| | - Chengguang Zhao
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Xiaojing Du
- Department of Digestive Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Dengjian Shi
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Jianzhang Wu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| |
Collapse
|
29
|
Rajmani RS, Gandham RK, Gupta SK, Sahoo AP, Singh PK, Saxena S, Kumar R, Chaturvedi U, Tiwari AK. Administration of IκB-kinase inhibitor PS1145 enhances apoptosis in DMBA-induced tumor in male Wistar rats. Cell Biol Int 2015; 39:1317-28. [DOI: 10.1002/cbin.10510] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 06/30/2015] [Indexed: 11/11/2022]
Affiliation(s)
- R. S. Rajmani
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Ravi Kumar Gandham
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Shishir Kumar Gupta
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Aditya P. Sahoo
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Prafull Kumar Singh
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Shikha Saxena
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Rajiv Kumar
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Uttara Chaturvedi
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Ashok K. Tiwari
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| |
Collapse
|
30
|
Matteucci C, Minutolo A, Marino-Merlo F, Grelli S, Frezza C, Mastino A, Macchi B. Characterization of the enhanced apoptotic response to azidothymidine by pharmacological inhibition of NF-kB. Life Sci 2015; 127:90-7. [PMID: 25744407 DOI: 10.1016/j.lfs.2015.01.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 11/10/2014] [Accepted: 01/28/2015] [Indexed: 12/26/2022]
Abstract
AIMS The present study addresses the issue of enhanced apoptotic response to AZT following co-treatment with an NF-kB inhibitor. MAIN METHODS To investigate this issue, different cell lines were assayed for susceptibility to AZT-mediated apoptosis without or with the addition of the NF-kB inhibitor Bay-11-7085. For further investigation, U937 cells were selected as good-responder cells to the combination treatment with 32 or 128 μM AZT, and 1 μM Bay-11-7085. Inhibition of NF-kB activation by Bay-11-7085 in cells treated with AZT was assayed through Western blot analysis of p65 expression and by EMSA. Involvement of the mitochondrial pathway of apoptosis in mechanisms underlying the improved effect of AZT following Bay-11-7085 co-treatment, was evaluated by assaying the cytochrome c release and the mitochondrial membrane potential (MMP) status using the JC-1 dye. Moreover, the transcriptional activity of both anti- and pro-apoptotic genes in U937 cells after combination treatment was quantitatively evaluated through real-time PCR. KEY FINDINGS We found that the combined treatment induced high levels of cytochrome c release and of MMP collapse in association with evident changes in the expression of both anti- and pro-apoptotic genes of the Bcl-2 family. Overexpression of Bcl-2 significantly suppressed the sensitization of U937 cells to an enhanced apoptotic response to AZT following co-treatment with the NF-kB inhibitor. SIGNIFICANCE The new findings suggest that a combination regimen based on AZT plus an NF-kB inhibitor could represent a new chemotherapeutic tool for retrovirus-related pathologies.
Collapse
Affiliation(s)
- Claudia Matteucci
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Antonella Minutolo
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Francesca Marino-Merlo
- Department of Biological and Environmental Sciences, University of Messina, Messina, Italy
| | - Sandro Grelli
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Caterina Frezza
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Antonio Mastino
- Department of Biological and Environmental Sciences, University of Messina, Messina, Italy; The Institute of Translational Pharmacology, CNR, Rome, Italy.
| | - Beatrice Macchi
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
31
|
Colony stimulating factor-1 receptor signaling networks inhibit mouse macrophage inflammatory responses by induction of microRNA-21. Blood 2015; 125:e1-13. [PMID: 25573988 DOI: 10.1182/blood-2014-10-608000] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Macrophage polarization between the M2 (repair, protumorigenic) and M1 (inflammatory) phenotypes is seen as a continuum of states. The detailed transcriptional events and signals downstream of colony-stimulating factor 1 receptor (CSF-1R) that contributes to amplification of the M2 phenotype and suppression of the M1 phenotype are largely unknown. Macrophage CSF-1R pTyr-721 signaling promotes cell motility and enhancement of tumor cell invasion in vitro. Combining analysis of cellular systems for CSF-1R gain of function and loss of function with bioinformatic analysis of the macrophage CSF-1R pTyr-721-regulated transcriptome, we uncovered microRNA-21 (miR-21) as a downstream molecular switch controlling macrophage activation and identified extracellular signal-regulated kinase1/2 and nuclear factor-κB as CSF-1R pTyr-721-regulated signaling nodes. We show that CSF-1R pTyr-721 signaling suppresses the inflammatory phenotype, predominantly by induction of miR-21. Profiling of the miR-21-regulated messenger RNAs revealed that 80% of the CSF-1-regulated canonical miR-21 targets are proinflammatory molecules. Additionally, miR-21 positively regulates M2 marker expression. Moreover, miR-21 feeds back to positively regulate its own expression and to limit CSF-1R-mediated activation of extracellular signal-regulated kinase1/2 and nuclear factor-κB. Consistent with an anti-inflammatory role of miRNA-21, intraperitoneal injection of mice with a miRNA-21 inhibitor increases the recruitment of inflammatory monocytes and enhances the peritoneal monocyte/macrophage response to lipopolysaccharide. These results identify the CSF-1R-regulated miR-21 network that modulates macrophage polarization.
Collapse
|
32
|
Wang X, Wang J, Zheng H, Xie M, Hopewell EL, Albrecht RA, Nogusa S, García-Sastre A, Balachandran S, Beg AA. Differential requirement for the IKKβ/NF-κB signaling module in regulating TLR- versus RLR-induced type 1 IFN expression in dendritic cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:2538-45. [PMID: 25057006 DOI: 10.4049/jimmunol.1400675] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Host innate-immune responses are tailored by cell type to control and eradicate specific infectious agents. For example, an acute RNA virus infection can result in high-level expression of type 1 IFNs by both conventional dendritic cells (cDCs) and plasmacytoid dendritic cells (pDCs), but whereas cDCs preferentially use RIG-I-like receptor (RLR) signaling to produce type 1 IFNs, pDCs predominantly use TLRs to induce these cytokines. We previously found that the IκB kinase β (IKKβ)/NF-κB pathway regulates early IFN-β expression, but not the magnitude of type 1 IFN expression following RLR engagement. In this study, we use IKKβ inhibition and mice deficient in IKKβ or canonical NF-κB subunits (p50, RelA/p65, and cRel) to demonstrate that the IKKβ/NF-κB axis is critical for virus-induced type 1 IFN expression in pDCs, but not in cDCs. We also reveal a crucial and more general requirement for IKKβ/NF-κB in TLR- but not RLR-induced expression of type 1 IFNs and inflammatory cytokines. Together, these findings reveal a previously unappreciated specificity of the IKKβ/NF-κB signaling axis in regulation of antimicrobial responses by different classes of pattern recognition receptors, and therefore by individual cell types reliant on particular pattern recognition receptors for their innate-immune transcriptional responses.
Collapse
Affiliation(s)
- Xingyu Wang
- Department of Immunology, Moffitt Cancer Center, Tampa, FL 33612
| | - Junmei Wang
- Department of Immunology, Moffitt Cancer Center, Tampa, FL 33612
| | - Hong Zheng
- Department of Immunology, Moffitt Cancer Center, Tampa, FL 33612
| | - Mengyu Xie
- Department of Immunology, Moffitt Cancer Center, Tampa, FL 33612
| | - Emily L Hopewell
- Department of Immunology, Moffitt Cancer Center, Tampa, FL 33612; Cancer Biology Ph.D. Program, Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612
| | - Randy A Albrecht
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Shoko Nogusa
- Fox Chase Cancer Center, Philadelphia, PA 19111; and
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | | | - Amer A Beg
- Department of Immunology, Moffitt Cancer Center, Tampa, FL 33612;
| |
Collapse
|
33
|
Saha A, Blando J, Silver E, Beltran L, Sessler J, DiGiovanni J. 6-Shogaol from dried ginger inhibits growth of prostate cancer cells both in vitro and in vivo through inhibition of STAT3 and NF-κB signaling. Cancer Prev Res (Phila) 2014; 7:627-38. [PMID: 24691500 DOI: 10.1158/1940-6207.capr-13-0420] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Despite much recent progress, prostate cancer continues to represent a major cause of cancer-related mortality and morbidity in men. Prostate cancer is the most common nonskin neoplasm and second leading cause of death in men. 6-Shogaol (6-SHO), a potent bioactive compound in ginger (Zingiber officinale Roscoe), has been shown to possess anti-inflammatory and anticancer activity. In the present study, the effect of 6-SHO on the growth of prostate cancer cells was investigated. 6-SHO effectively reduced survival and induced apoptosis of cultured human (LNCaP, DU145, and PC3) and mouse (HMVP2) prostate cancer cells. Mechanistic studies revealed that 6-SHO reduced constitutive and interleukin (IL)-6-induced STAT3 activation and inhibited both constitutive and TNF-α-induced NF-κB activity in these cells. In addition, 6-SHO decreased the level of several STAT3 and NF-κB-regulated target genes at the protein level, including cyclin D1, survivin, and cMyc and modulated mRNA levels of chemokine, cytokine, cell cycle, and apoptosis regulatory genes (IL-7, CCL5, BAX, BCL2, p21, and p27). 6-SHO was more effective than two other compounds found in ginger, 6-gingerol, and 6-paradol at reducing survival of prostate cancer cells and reducing STAT3 and NF-κB signaling. 6-SHO also showed significant tumor growth inhibitory activity in an allograft model using HMVP2 cells. Overall, the current results suggest that 6-SHO may have potential as a chemopreventive and/or therapeutic agent for prostate cancer and that further study of this compound is warranted.
Collapse
Affiliation(s)
- Achinto Saha
- Authors' Affiliations: Division of Pharmacology and Toxicology and Department of Nutritional Sciences, Dell Pediatric Research Institute; and Department of Chemistry, The University of Texas at Austin, Austin, Texas
| | - Jorge Blando
- Authors' Affiliations: Division of Pharmacology and Toxicology and Department of Nutritional Sciences, Dell Pediatric Research Institute; and Department of Chemistry, The University of Texas at Austin, Austin, Texas
| | - Eric Silver
- Authors' Affiliations: Division of Pharmacology and Toxicology and Department of Nutritional Sciences, Dell Pediatric Research Institute; and Department of Chemistry, The University of Texas at Austin, Austin, Texas
| | - Linda Beltran
- Authors' Affiliations: Division of Pharmacology and Toxicology and Department of Nutritional Sciences, Dell Pediatric Research Institute; and Department of Chemistry, The University of Texas at Austin, Austin, Texas
| | - Jonathan Sessler
- Authors' Affiliations: Division of Pharmacology and Toxicology and Department of Nutritional Sciences, Dell Pediatric Research Institute; and Department of Chemistry, The University of Texas at Austin, Austin, Texas
| | - John DiGiovanni
- Authors' Affiliations: Division of Pharmacology and Toxicology and Department of Nutritional Sciences, Dell Pediatric Research Institute; and Department of Chemistry, The University of Texas at Austin, Austin, TexasAuthors' Affiliations: Division of Pharmacology and Toxicology and Department of Nutritional Sciences, Dell Pediatric Research Institute; and Department of Chemistry, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
34
|
Nguyen DP, Li J, Yadav SS, Tewari AK. Recent insights into NF-κB signalling pathways and the link between inflammation and prostate cancer. BJU Int 2014; 114:168-76. [PMID: 24215139 DOI: 10.1111/bju.12488] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Inflammation is involved in regulation of cellular events in prostate carcinogenesis through control of the tumour micro-environment. A variety of bone marrow-derived cells, including CD4+ lymphocytes, macrophages and myeloid-derived suppressor cells, are integral components of the tumour micro-environment. On activation by inflammatory cytokines, NF-κB complexes are capable of promoting tumour cell survival through anti-apoptotic signalling in prostate cancer (PCa). Positive feedback loops are able to maintain NF-κB activation. NF-κB activation is also associated with the metastatic phenotype and PCa progression to castration-resistant prostate cancer (CRPC). A novel role for inhibitor of NF-κB kinase (IKK)-α in NF-κB-independent PCa progression to metastasis and CRPC has recently been uncovered, providing a new mechanistic link between inflammation and PCa. Expansion of PCa progenitors by IKK-α may be involved in this process. In this review, we offer the latest evidence regarding the role of the NF-κB pathway in PCa and discuss therapeutic attempts to target the NF-κB pathways. We point out the need to further dissect inflammatory pathways in PCa in order to develop appropriate preventive measures and design novel therapeutic strategies.
Collapse
Affiliation(s)
- Daniel P Nguyen
- Laboratory of Urological Oncology, Department of Urology, Weill Cornell Medical College-New York Presbyterian Hospital, New York, NY, USA; Department of Urology, University of Berne, Inselspital, Berne, Switzerland
| | | | | | | |
Collapse
|
35
|
Singha B, Gatla HR, Manna S, Chang TP, Sanacora S, Poltoratsky V, Vancura A, Vancurova I. Proteasome inhibition increases recruitment of IκB kinase β (IKKβ), S536P-p65, and transcription factor EGR1 to interleukin-8 (IL-8) promoter, resulting in increased IL-8 production in ovarian cancer cells. J Biol Chem 2013; 289:2687-700. [PMID: 24337575 DOI: 10.1074/jbc.m113.502641] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Proinflammatory and pro-angiogenic chemokine interleukin-8 (IL-8, CXCL8) contributes to ovarian cancer progression through its induction of tumor cell proliferation, survival, angiogenesis, and metastasis. Proteasome inhibition by bortezomib, which has been used as a frontline therapy in multiple myeloma, has shown only limited effectiveness in ovarian cancer and other solid tumors. However, the responsible mechanisms remain elusive. Here, we show that proteasome inhibition dramatically increases the IL-8 expression and release in ovarian cancer cells. The responsible mechanism involves an increased nuclear accumulation of IκB kinase β (IKKβ) and an increased recruitment of the nuclear IKKβ, p65-phosphorylated at Ser-536, and the transcription factor early growth response-1 (EGR-1) to the endogenous IL-8 promoter. Coimmunoprecipitation studies identified the nuclear EGR-1 associated with IKKβ and with p65, with preferential binding to S536P-p65. Both IKKβ activity and EGR-1 expression are required for the increased IL-8 expression induced by proteasome inhibition in ovarian cancer cells. Interestingly, in multiple myeloma cells the IL-8 release is not increased by bortezomib. Together, these data indicate that the increased IL-8 release may represent one of the underlying mechanisms responsible for the decreased effectiveness of proteasome inhibition in ovarian cancer treatment and identify IKKβ and EGR-1 as potential new targets in ovarian cancer combination therapies.
Collapse
|
36
|
Park DW, Jiang S, Tadie JM, Stigler WS, Gao Y, Deshane J, Abraham E, Zmijewski JW. Activation of AMPK enhances neutrophil chemotaxis and bacterial killing. Mol Med 2013; 19:387-98. [PMID: 24091934 DOI: 10.2119/molmed.2013.00065] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 09/20/2013] [Indexed: 01/04/2023] Open
Abstract
An inability of neutrophils to eliminate invading microorganisms is frequently associated with severe infection and may contribute to the high mortality rates associated with sepsis. In the present studies, we examined whether metformin and other 5' adenosine monophosphate-activated protein kinase (AMPK) activators affect neutrophil motility, phagocytosis and bacterial killing. We found that activation of AMPK enhanced neutrophil chemotaxis in vitro and in vivo, and also counteracted the inhibition of chemotaxis induced by exposure of neutrophils to lipopolysaccharide (LPS). In contrast, small interfering RNA (siRNA)-mediated knockdown of AMPKα1 or blockade of AMPK activation through treatment of neutrophils with the AMPK inhibitor compound C diminished neutrophil chemotaxis. In addition to their effects on chemotaxis, treatment of neutrophils with metformin or aminoimidazole carboxamide ribonucleotide (AICAR) improved phagocytosis and bacterial killing, including more efficient eradication of bacteria in a mouse model of peritonitis-induced sepsis. Immunocytochemistry showed that, in contrast to LPS, metformin or AICAR induced robust actin polymerization and distinct formation of neutrophil leading edges. Although LPS diminished AMPK phosphorylation, metformin or AICAR was able to partially decrease the effects of LPS/toll-like receptor 4 (TLR4) engagement on downstream signaling events, particularly LPS-induced IκBα degradation. The IκB kinase (IKK) inhibitor PS-1145 diminished IκBα degradation and also prevented LPS-induced inhibition of chemotaxis. These results suggest that AMPK activation with clinically approved agents, such as metformin, may facilitate bacterial eradication in sepsis and other inflammatory conditions associated with inhibition of neutrophil activation and chemotaxis.
Collapse
Affiliation(s)
- Dae Won Park
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America Division of Infectious Diseases, Korea University Ansan Hospital, Ansan, Republic of Korea
| | - Shaoning Jiang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jean-Marc Tadie
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America Service des Maladies Infectieuses et Réanimation Médicale, Centre Hospitalier Universitaire, Rennes, France
| | - William S Stigler
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Yong Gao
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jessy Deshane
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Edward Abraham
- Office of the Dean, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Jaroslaw W Zmijewski
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
37
|
Synthesis and in vitro characterization of ionone-based compounds as dual inhibitors of the androgen receptor and NF-κB. Invest New Drugs 2013; 32:227-34. [DOI: 10.1007/s10637-013-0040-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 10/08/2013] [Indexed: 10/26/2022]
|
38
|
|
39
|
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
|
40
|
Straus DS. Design of small molecules targeting transcriptional activation by NF-κB: overview of recent advances. Expert Opin Drug Discov 2013; 4:823-36. [PMID: 23496269 DOI: 10.1517/17460440903143739] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The transcription factor NF-κB plays a central role in immune signaling and the inflammatory response. It also activates transcription of antiapoptotic factors in tumor cells, leading to enhanced cell survival. Because of the importance of NF-κB in inflammation and cancer, there is considerable interest in development of drugs that inhibit NF-κB activation or NF-κB-directed transcription. Recent elucidation of the intracellular pathways that activate NF-κB and mechanisms for transcriptional regulation by NF-κB has identified molecular targets for rational design of such drugs. OBJECTIVE This review provides an update on NF-κB signaling and an overview of three classes of NF-κB inhibitors: i) inhibitors of IκB kinase-β (also called IKK2), an essential link in the inflammatory response; ii) agents that react with NF-κB and prevent its binding to DNA; and iii) ligands for nuclear receptors such as the glucocorticoid receptor, PPARs and liver X receptor, which interfere with NF-κB-mediated transcription through a mechanism termed ligand-dependent transrepression. Recent progress in development of glucocorticoid receptor, PPAR and liver X receptor ligands with dissociated activity, which retain transrepression but have reduced transactivation potency, is also described. CONCLUSIONS NF-κB inhibitors have yielded promising results in rodent models of inflammatory disease and cancer. Some of these are currently advancing into clinical trials.
Collapse
Affiliation(s)
- Daniel S Straus
- University of California, Division of Biomedical Sciences, Riverside, CA 92521-0121, USA +1 951 827 5612 ; +1 951 827 5504 ;
| |
Collapse
|
41
|
Aberrant IKKα and IKKβ cooperatively activate NF-κB and induce EGFR/AP1 signaling to promote survival and migration of head and neck cancer. Oncogene 2013; 33:1135-47. [PMID: 23455325 PMCID: PMC3926900 DOI: 10.1038/onc.2013.49] [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: 09/25/2011] [Revised: 01/07/2013] [Accepted: 01/18/2013] [Indexed: 12/23/2022]
Abstract
The Inhibitor-κB Kinase-Nuclear Factor-κB (IKK-NF-κB) and Epidermal Growth Factor Receptor-Activator Protein-1 (EGFR-AP-1) pathways are often co-activated and promote malignant behavior, but the underlying basis for this relationship is unclear. Resistance to inhibitors of IKKβ or EGFR is observed in head and neck squamous cell carcinomas (HNSCC). Here, we reveal that both IKKα and β contribute to nuclear activation of canonical and alternate NF-κB/REL family transcription factors, and overexpression of signal components enhancing co-activation of the EGFR-AP1 pathway. We observed that IKKα and IKKβ exhibit increased protein expression, nuclear localization and phosphorylation in HNSCC tissues and cell lines. Individually, IKK activity varied amongst different cell lines, but overexpression of both IKKs induced the strongest NF-κB activation. Conversely, siRNA knockdown of both IKKs significantly decreased nuclear localization and phosphorylation of canonical RELA and IκBα, and alternative p52 and RELB subunits. Knockdown of both IKKs more effectively inhibited NF-κB activation, broadly modulated gene expression, and suppressed cell proliferation and migration. Global expression profiling revealed that NF-κB, cytokine, inflammatory response, and growth factor signaling are among the top pathways and networks regulated by IKKs. Importantly, IKKα and IKKβ together promoted the expression and activity of TGFα, EGFR, and AP1 transcription factors cJun, JunB, and Fra1. Knockdown of AP1 subunits individually decreased 8/15 (53%) of IKK-targeted genes sampled, and similarly inhibited cell proliferation and migration. Mutations of NF-κB and AP1 binding sites abolished or decreased IKK-induced IL-8 promoter activity. Compounds such as wedelactone with dual IKK inhibitory activity, and geldanomycins that block IKKα/β and EGFR pathways were more active than IKKβ-specific inhibitors in suppressing NF-κB activation and proliferation, and inducing cell death. We conclude that IKKα and IKKβ cooperatively activate NF-κB and EGFR/AP1 networks of signaling pathways, and contribute to the malignant phenotype and the intrinsic or acquired therapeutic resistance of HNSCC.
Collapse
|
42
|
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
|
43
|
Raifer H, Mahiny AJ, Bollig N, Petermann F, Hellhund A, Kellner K, Guralnik A, Reinhard K, Bothur E, Huber M, Bauer S, Löhning M, Kiss EA, Ganal SC, Diefenbach A, Korn T, Lohoff M. Unlike αβ T cells, γδ T cells, LTi cells and NKT cells do not require IRF4 for the production of IL-17A and IL-22. Eur J Immunol 2012; 42:3189-201. [PMID: 22961652 DOI: 10.1002/eji.201142155] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Revised: 08/07/2012] [Accepted: 09/03/2012] [Indexed: 12/15/2022]
Abstract
Apart from conventional CD4(+) Th17 cells, the cytokines IL-17A and IL-22 can also be produced by γδ T cells, NK cells and lymphoid tissue inducer (LTi) cells. Th17 cells develop from precursor cells after T-cell receptor stimulation in the presence of TGF-β, IL-6 and IL-23. In contrast, a subset of γδ T cells ("γδT17") is committed for fast IL-17 production already in the thymus; however, γδ T cells can also produce IL-17 after prolonged in vitro stimulation via their γδ T-cell receptor plus IL-23. Here, we show that γδ T-, LTi- and NKT cells differ extensively from Th17 cells in their signalling requirements for the generation of IL-17A and IL-22. While production of these cytokines by Th17 cells totally depends on the transcription factor interferon regulatory factor 4 (IRF4), IRF4 is irrelevant in the other cell types. As for γδ T cells, this finding pertains to both thymic commitment and prolonged in vitro culture. Furthermore, IL-17A-producing γδ T cells accumulate in the central nervous system of IRF4 deficient (Irf4(-/-)) mice during experimental autoimmune encephalomyelitis. IL-17A-producing WT and Irf4(-/-) γδ T cells equally express CCR6 and lack CD27. The underlying IRF4-independent pathway partially involves STAT3 during in vitro stimulation.
Collapse
MESH Headings
- Animals
- Central Nervous System/immunology
- Central Nervous System/metabolism
- Central Nervous System/pathology
- Encephalomyelitis, Autoimmune, Experimental
- Gene Expression Regulation/genetics
- Gene Expression Regulation/immunology
- Interferon Regulatory Factors/genetics
- Interferon Regulatory Factors/immunology
- Interferon Regulatory Factors/metabolism
- Interleukin-17/biosynthesis
- Interleukin-17/genetics
- Interleukin-17/immunology
- Interleukins/biosynthesis
- Interleukins/genetics
- Interleukins/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Natural Killer T-Cells/immunology
- Natural Killer T-Cells/metabolism
- Natural Killer T-Cells/pathology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, CCR6/biosynthesis
- Receptors, CCR6/genetics
- Receptors, CCR6/immunology
- STAT3 Transcription Factor/genetics
- STAT3 Transcription Factor/immunology
- STAT3 Transcription Factor/metabolism
- Th17 Cells/immunology
- Th17 Cells/metabolism
- Th17 Cells/pathology
- Tumor Necrosis Factor Receptor Superfamily, Member 7/biosynthesis
- Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics
- Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
- Interleukin-22
Collapse
Affiliation(s)
- Hartmann Raifer
- Institut für medizinische Mikrobiologie und Krankenhaushygiene, Philipps Universität Marburg, Marburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Garg R, Blando J, Perez CJ, Wang H, Benavides FJ, Kazanietz MG. Activation of nuclear factor κB (NF-κB) in prostate cancer is mediated by protein kinase C epsilon (PKCepsilon). J Biol Chem 2012; 287:37570-82. [PMID: 22955280 DOI: 10.1074/jbc.m112.398925] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Protein kinase C ε (PKCε) has emerged as an oncogenic kinase and plays important roles in cell survival, mitogenesis and invasion. PKCε is up-regulated in most epithelial cancers, including prostate, breast, and lung cancer. Here we report that PKCε is an essential mediator of NF-κB activation in prostate cancer cells. A strong correlation exists between PKCε overexpression and NF-κB activation status in prostate cancer cells. Moreover, transgenic overexpression of PKCε in the mouse prostate causes preneoplastic lesions that display significant NF-κB hyperactivation. PKCε RNAi depletion or inhibition in prostate cancer cells diminishes NF-κB translocation to the nucleus with subsequent impairment of both activation of NF-κB transcription and induction of NF-κB responsive genes in response to the proinflammatory cytokine tumor necrosis factor α (TNFα). On the other hand, PKCε overexpression in normal prostate cells enhances activation of the NF-κB pathway. A mechanistic analysis revealed that TNFα activates PKCε via a C1 domain/diacylglycerol-dependent mechanism that involves phosphatidylcholine-phospholipase C. Moreover, PKCε facilitates the assembly of the TNF receptor-I signaling complex to trigger NF-κB activation. Our studies identified a molecular link between PKCε and NF-κB that controls key responses implicated in prostate cancer progression.
Collapse
Affiliation(s)
- Rachana Garg
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | |
Collapse
|
45
|
Kuo PL, Shen KH, Hung SH, Hsu YL. CXCL1/GROα increases cell migration and invasion of prostate cancer by decreasing fibulin-1 expression through NF-κB/HDAC1 epigenetic regulation. Carcinogenesis 2012; 33:2477-87. [PMID: 23027620 DOI: 10.1093/carcin/bgs299] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Inflammatory tumor microenvironments play pivotal roles in the development of cancer. Inflammatory cytokines such as CXCL1/GROα exert cancer-promoting activities by increasing tumor angiogenesis. However, whether CXCL1/GROα also plays a role in the progression of prostate cancer, particularly in highly invasive castration-resistant prostate cancer (CRPC), has not been investigated. We explored whether CXCL1/GROα enhances cell migration and invasion in PC-3 and DU145 CRPC. Induction of PC-3 and DU145 cancer progression by CXCL1/GROα is associated with increased AKT activation and IκB kinase α (IKKα) phosphorylation, resulting in nuclear factor-kappaB (NF-κB) activation. Activated NF-κB interacts with histone deacetylase 1 (HDAC1) to form a gene-silencing complex, which represses the expression of fibulin-1D by decreasing the acetylation of histone H3 and H4 on the NF-κB-binding site of the fibulin-1D promoter. Blockade of AKT2 by small hairpin RNA (shRNA) decreases IKKα phosphorylation, NF-κB nuclear translocation and cell migration, indicating that AKT is required in CXCL1/GROα-mediated NF-κB activation and cell migration. In addition, NF-κB and HDAC1 shRNA decrease the effect of CXCL1/GROα on fibulin-1D downregulation, migration and invasion, suggesting that the NF-κB/HDAC1 complex is also involved in CXCL1/GROα-mediated cancer progression. Our findings provide the first evidence that CXCL1/GROα decreases fibulin-1D expression in prostate cancer cells and also reveals novel insights into the mechanism by which CXCL1/GROα regulates NF-κB activation through the AKT pathway. Our results also clearly establish that co-operation of NF-κB and HDAC1 regulates fibulin-1D expression by epigenetic modification. Our study suggests that inhibition of CXCL1/GROα-mediated AKT/NF-κB signaling may be an attractive therapeutic target for CRPC.
Collapse
Affiliation(s)
- Po-Lin Kuo
- Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | | | | | | |
Collapse
|
46
|
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
|
47
|
Liu T, Liu D, Liu J, Song JT, Gao SL, Li H, Hu LH, Liu BR. Effect of NF-κB inhibitors on the chemotherapy-induced apoptosis of the colon cancer cell line HT-29. Exp Ther Med 2012; 4:716-722. [PMID: 23170132 PMCID: PMC3501445 DOI: 10.3892/etm.2012.647] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 06/06/2012] [Indexed: 12/14/2022] Open
Abstract
This study aimed to investigate the impact of the combined use of the nuclear factor-κB (NF-κB) inhibitors pyrrolidine dithiocarbamate (PDTC), bortezomib or SN50, and the chemotherapy agents arsenic acid (As2O3), fluorouracil (5FU), oxaliplatin or paclitaxel on the growth and apoptosis of HT-29 cells. Cell morphology was observed using inverted microscopy, and cell viability and apoptosis were assessed using the MTT assay and flow cytometry, respectively. The activities of NF-κB were analyzed by western blotting and electrophoretic mobility shift assay (EMSA). Cell growth was significantly inhibited by As2O3, oxaliplatin and paclitaxel in a time- and concentration-dependent manner (P<0.05), while 5FU inhibited cell growth in a time-dependent manner only (P<0.05). The growth inhibition rate and apoptosis induction ratio were increased following the combined treatment of the chemotherapy agent and NF-κB inhibitor. The expression of NF-κB p65 was upregulated when cells were treated with a chemotherapy drug, however it was downregulated following combined treatment or treatment with an NF-κB inhibitor alone. In conclusion, an NF-κB inhibitor combined with a chemotherapy drug effectively inhibited cell proliferation, induced cell apoptosis and inhibited NF-κB activity to enhance the chemotherapeutic sensitivity of HT-29 cells.
Collapse
Affiliation(s)
- Ting Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
The inhibitor of nuclear factor-κB (IκB) kinase (IKK) complex is the master regulator of the NF-κB signaling pathway. The activation of the IKK complex is a tightly regulated, highly stimulus-specific, and target-specific event that is essential for the plethora of functions attributed to NF-κB. More recently, NF-κB-independent roles of IKK members have brought increased complexity to its biological function. This review highlights some of the major advances in the studies of the process of IKK activation and the biological roles of IKK family members, with a focus on NF-κB-independent functions. Understanding these complex processes is essential for targeting IKK for therapeutics.
Collapse
Affiliation(s)
- Fei Liu
- Laboratory of Genetics, The Salk Institute, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
49
|
Histone methyltransferase NSD2/MMSET mediates constitutive NF-κB signaling for cancer cell proliferation, survival, and tumor growth via a feed-forward loop. Mol Cell Biol 2012; 32:3121-31. [PMID: 22645312 DOI: 10.1128/mcb.00204-12] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Constitutive NF-κB activation by proinflammatory cytokines plays a major role in cancer progression. However, the underlying mechanism is still unclear. We report here that histone methyltransferase NSD2 (also known as MMSET or WHSC1), a target of bromodomain protein ANCCA/ATAD2, acts as a strong coactivator of NF-κB by directly interacting with NF-κB for activation of target genes, including those for interleukin-6 (IL-6), IL-8, vascular endothelial growth factor A (VEGFA), cyclin D, Bcl-2, and survivin, in castration-resistant prostate cancer (CRPC) cells. NSD2 is recruited to the target gene promoters upon induction and mediates NF-κB activation-associated elevation of histone H3K36me2 and H3K36me3 marks at the promoter, which involves its methylase activity. Interestingly, we found that NSD2 is also critical for cytokine-induced recruitment of NF-κB and acetyltransferase p300 and histone hyperacetylation. Importantly, NSD2 is overexpressed in prostate cancer tumors, and its overexpression correlates with NF-κB activation. Furthermore, NSD2 expression is strongly induced by tumor necrosis factor alpha (TNF-α) and IL-6 via NF-κB and plays a crucial role in tumor growth. These results identify NSD2 to be a key chromatin regulator of NF-κB and mediator of the cytokine autocrine loop for constitutive NF-κB activation and emphasize the important roles played by NSD2 in cancer cell proliferation and survival and tumor growth.
Collapse
|
50
|
Elkind E, Vaisid T, Kornspan JD, Barnoy S, Rottem S, Kosower NS. Calpastatin upregulation in Mycoplasma hyorhinis-infected cells is promoted by the mycoplasma lipoproteins via the NF-κB pathway. Cell Microbiol 2012; 14:840-51. [PMID: 22288381 DOI: 10.1111/j.1462-5822.2012.01760.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Mycoplasma hyorhinis frequently contaminates cultured cells, with effects on synthetic and metabolic pathways. We demonstrated for the first time that contamination of cells by a strain of M. hyorhinis (NDMh) results in increased levels of calpastatin (the endogenous inhibitor of the ubiquitous Ca(2+) -dependent protease calpain). We now show that the calpastatin upregulation by NDMh in neuroblastoma SH-SY5Y cells resides in the NDMh lipoprotein fraction (LPP), via the NF-κB transcription pathway. NF-κB activation requires dissociation of the cytoplasmic NF-κB/IκB complex followed by NF-κB translocation to the nucleus. NDMh-LPP induced translocation of the NF-κB RelA subunit to the nucleus and upregulated calpastatin. RelA translocation and calpastatin elevation were prevented when dissociation of the NF-κB/IκB complex was inhibited either by transfection with the non-phosphorylatable IκB mutant ΔNIκBα, or by using PS1145, an inhibitor of the IκB kinase (IKK complex). Increased calpastatin levels attenuate calpain-related amyloid-β-peptide and Ca(2+) -toxicity (these are central to the pathogenesis of Alzheimer's Disease). LPP-induced elevation of calpastatin provides an example of effects on non-inflammatory intracellular proteins, the outcome being significant alterations in host cell functions. Since calpastatin level is important in the control of calpain activity, mycoplasmal LPP may be of interest in treating some pathological processes involving excessive calpain activation.
Collapse
Affiliation(s)
- Esther Elkind
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel-Aviv University, Ramat-Aviv, Tel-Aviv 69978, Israel
| | | | | | | | | | | |
Collapse
|