1
|
Wu J, Shyy M, Shyy JYJ, Xiao H. Role of inflammasomes in endothelial dysfunction. Br J Pharmacol 2024. [PMID: 38952037 DOI: 10.1111/bph.16479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/14/2024] [Accepted: 05/04/2024] [Indexed: 07/03/2024] Open
Abstract
The vascular endothelium dynamically responds to environmental cues and plays a pivotal role in maintaining vascular homeostasis by regulating vasomotor tone, blood cell trafficking, permeability and immune responses. However, endothelial dysfunction results in various pathological conditions. Inflammasomes are large intracellular multimeric complexes activated by pathogens or cellular damage. Inflammasomes in vascular endothelial cells (ECs) initiate innate immune responses, which have emerged as significant mediators in endothelial dysfunction, contributing to the pathophysiology of an array of diseases. This review summarizes the mechanisms and ramifications of inflammasomes in ECs and related vascular diseases such as atherosclerosis, abdominal aortic aneurysm, stroke, and lung and kidney diseases. We also discuss potential drugs targeting EC inflammasomes and their applications in treating vascular diseases.
Collapse
Affiliation(s)
- Jimin Wu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Haihe Laboratory of Cell Ecosystem, Beijing, China
| | - Melody Shyy
- Biological Sciences, University of California, Santa Barbara, Santa Barbara, California, USA
| | - John Y-J Shyy
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Han Xiao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Haihe Laboratory of Cell Ecosystem, Beijing, China
| |
Collapse
|
2
|
Medeiros M, Guenka S, Bastos D, Oliveira KL, Brassesco MS. Amicis Omnia Sunt Communia: NF-κB Inhibition as an Alternative to Overcome Osteosarcoma Heterogeneity. Pharmaceuticals (Basel) 2024; 17:734. [PMID: 38931401 PMCID: PMC11206879 DOI: 10.3390/ph17060734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Tumor heterogeneity poses a significant challenge in osteosarcoma (OS) treatment. In this regard, the "omics" era has constantly expanded our understanding of biomarkers and altered signaling pathways (i.e., PI3K/AKT/mTOR, WNT/β-catenin, NOTCH, SHH/GLI, among others) involved in OS pathophysiology. Despite different players and complexities, many commonalities have been described, among which the nuclear factor kappa B (NF-κB) stands out. Its altered activation is pervasive in cancer, with pleiotropic action on many disease-relevant traits. Thus, in the scope of this article, we highlight the evidence of NF-κB dysregulation in OS and its integration with other cancer-related pathways while we summarize the repertoire of compounds that have been described to interfere with its action. In silico strategies were used to demonstrate that NF-κB is closely coordinated with other commonly dysregulated signaling pathways not only by functionally interacting with several of their members but also by actively participating in the regulation of their transcription. While existing inhibitors lack selectivity or act indirectly, the therapeutic potential of targeting NF-κB is indisputable, first for its multifunctionality on most cancer hallmarks, and secondly, because, as a common downstream effector of the many dysregulated pathways influencing OS aggressiveness, it turns complex regulatory networks into a simpler picture underneath molecular heterogeneity.
Collapse
Affiliation(s)
- Mariana Medeiros
- Cell Biology Department, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirão Preto 14040-900, São Paulo, Brazil;
| | - Sophia Guenka
- Biology Department, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirão Preto 14040-900, São Paulo, Brazil; (S.G.); (D.B.)
| | - David Bastos
- Biology Department, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirão Preto 14040-900, São Paulo, Brazil; (S.G.); (D.B.)
| | - Karla Laissa Oliveira
- Regional Blood Center, University of São Paulo, Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirão Preto 14051-140, São Paulo, Brazil;
| | - María Sol Brassesco
- Biology Department, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirão Preto 14040-900, São Paulo, Brazil; (S.G.); (D.B.)
| |
Collapse
|
3
|
Gobrecht P, Gebel J, Hilla A, Gisselmann G, Fischer D. Targeting Vasohibins to Promote Axon Regeneration. J Neurosci 2024; 44:e2031232024. [PMID: 38429108 PMCID: PMC10993095 DOI: 10.1523/jneurosci.2031-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/13/2023] [Accepted: 02/07/2024] [Indexed: 03/03/2024] Open
Abstract
Treatments accelerating axon regeneration in the nervous system are still clinically unavailable. However, parthenolide promotes adult sensory neurons' axon growth in culture by inhibiting microtubule detyrosination. Here, we show that overexpression of vasohibins increases microtubule detyrosination in growth cones and compromises growth in culture and in vivo. Moreover, overexpression of these proteins increases the required parthenolide concentrations to promote axon regeneration. At the same time, the partial knockdown of endogenous vasohibins or their enhancer SVBP in neurons facilitates axon growth, verifying them as pharmacological targets for promoting axon growth. In vivo, repeated intravenous application of parthenolide or its prodrug di-methyl-amino-parthenolide (DMAPT) markedly facilitates the regeneration of sensory, motor, and sympathetic axons in injured murine and rat nerves, leading to acceleration of functional recovery. Moreover, orally applied DMAPT was similarly effective in promoting nerve regeneration. Thus, pharmacological inhibition of vasohibins facilitates axon regeneration in different species and nerves, making parthenolide and DMAPT the first promising drugs for curing nerve injury.
Collapse
Affiliation(s)
- Philipp Gobrecht
- Center of Pharmacology, Institute II, Medical Faculty, University of Cologne, Cologne D-50931, Germany
- Department of Cell Physiology, Ruhr University of Bochum, Bochum 44780, Germany
| | - Jeannette Gebel
- Center of Pharmacology, Institute II, Medical Faculty, University of Cologne, Cologne D-50931, Germany
- Department of Cell Physiology, Ruhr University of Bochum, Bochum 44780, Germany
| | - Alexander Hilla
- Department of Cell Physiology, Ruhr University of Bochum, Bochum 44780, Germany
| | - Günter Gisselmann
- Department of Cell Physiology, Ruhr University of Bochum, Bochum 44780, Germany
| | - Dietmar Fischer
- Center of Pharmacology, Institute II, Medical Faculty, University of Cologne, Cologne D-50931, Germany
- Department of Cell Physiology, Ruhr University of Bochum, Bochum 44780, Germany
| |
Collapse
|
4
|
Obulkasim H, Aji G, Abudoula A, Liu Y, Duan S. Parthenolide induces gallbladder cancer cell apoptosis via MAPK signalling. Ann Med Surg (Lond) 2024; 86:1956-1966. [PMID: 38576937 PMCID: PMC10990370 DOI: 10.1097/ms9.0000000000001828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 02/04/2024] [Indexed: 04/06/2024] Open
Abstract
Objective Parthenolide (PTL) has a wide range of clinical applications owing to its anti-inflammatory and antitumor effects. To date, the antitumor effect of PTL on gallbladder cancer (GBC) remains largely unknown. Therefore, we aimed to investigate the biological effects of PTL on GBC. Methods The cellular viability and proliferation of GBC-SD and NOZ cell lines after treatment with different concentrations of PTL were analyzed using the Cell Counting Kit-8 (CCK8)assay and colony formation assay. Apoptosis analysis was performed using flow cytometry. Hoechst staining was performed. RNA sequencing (RNA-seq) was performed to identify PTL-related genes and signalling pathways. Furthermore, we confirmed the involvement of these signalling pathways by qRT-PCR and western blotting. For the in-vivo experiments, a xenograft model was used to evaluate the effects of PTL on the proliferation of NOZ cells. Results PTL significantly inhibited GBC cell growth in vitro and induced apoptosis in the GBC-SD and NOZ cell lines in a dose-dependent manner. RNA sequencing data showed that the immune response and mitogen-activated protein kinase (MAPK) signalling pathways are closely associated with PTL-induced gallbladder cancer cell apoptosis. PTL upregulated BAX, cleaved PARP-1, cleaved caspase-3, cleaved caspase-9, P53 and decreased the expression of BCL-2, phosphorylated ERK, and phosphorylated MEK in vitro. Tumour volume and weight were also suppressed by PTL in vivo. Moreover, the effects of PTL on GBC cells might be mediated by the MAPK pathway. Conclusion PTL significantly inhibits gallbladder cancer cell proliferation and induces apoptosis through the MAPK pathway, which is a potential molecular reagent for treating GBC. However, further exploration is needed to verify the antitumor effects of PTL and its intracellular signalling mechanism.
Collapse
Affiliation(s)
- Halmurat Obulkasim
- Hospital of Traditional Chinese Medicine Affiliated to Xinjiang Medical University, Traditional Chinese Medicine Hospital of Xinjiang Uyghur Autonomous Region
- Postdoctoral Workstation of Traditional Chinese Medicine Hospital of Xinjiang Uygur Autonomous Region, Saybagh District
| | - Gulibositan Aji
- The First Affiliated Hospital of Xinjiang Medical University, Xinshi District, Urumqi, China
| | - Abudoukaher Abudoula
- Hospital of Traditional Chinese Medicine Affiliated to Xinjiang Medical University, Traditional Chinese Medicine Hospital of Xinjiang Uyghur Autonomous Region
| | - Yu Liu
- Hospital of Traditional Chinese Medicine Affiliated to Xinjiang Medical University, Traditional Chinese Medicine Hospital of Xinjiang Uyghur Autonomous Region
| | - Shaobin Duan
- Hospital of Traditional Chinese Medicine Affiliated to Xinjiang Medical University, Traditional Chinese Medicine Hospital of Xinjiang Uyghur Autonomous Region
| |
Collapse
|
5
|
Zhu S, Sun P, Bennett S, Charlesworth O, Tan R, Peng X, Gu Q, Kujan O, Xu J. The therapeutic effect and mechanism of parthenolide in skeletal disease, cancers, and cytokine storm. Front Pharmacol 2023; 14:1111218. [PMID: 37033622 PMCID: PMC10080395 DOI: 10.3389/fphar.2023.1111218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/17/2023] [Indexed: 03/12/2023] Open
Abstract
Parthenolide (PTL or PAR) was first isolated from Magnolia grandiflora and identified as a small molecule cancer inhibitor. PTL has the chemical structure of C15H20O3 with characteristics of sesquiterpene lactones and exhibits the biological property of inhibiting DNA biosynthesis of cancer cells. In this review, we summarise the recent research progress of medicinal PTL, including the therapeutic effects on skeletal diseases, cancers, and inflammation-induced cytokine storm. Mechanistic investigations reveal that PTL predominantly inhibits NF-κB activation and other signalling pathways, such as reactive oxygen species. As an inhibitor of NF-κB, PTL appears to inhibit several cytokines, including RANKL, TNF-α, IL-1β, together with LPS induced activation of NF-κB and NF-κB -mediated specific gene expression such as IL-1β, TNF-α, COX-2, iNOS, IL-8, MCP-1, RANTES, ICAM-1, VCAM-1. It is also proposed that PTL could inhibit cytokine storms or hypercytokinemia triggered by COVID-19 via blocking the activation of NF-κB signalling. Understanding the pharmacologic properties of PTL will assist us in developing its therapeutic application for medical conditions, including arthritis, osteolysis, periodontal disease, cancers, and COVID-19-related disease.
Collapse
Affiliation(s)
- Sipin Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
- *Correspondence: Sipin Zhu, ; Jiake Xu,
| | - Ping Sun
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
- Department of Endocrinology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Samuel Bennett
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Oscar Charlesworth
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Renxiang Tan
- The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Functional Biomolecules, Nanjing University, Nanjing, China
| | - Xing Peng
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Qiang Gu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Omar Kujan
- UWA Dental School, The University of Western Australia, Perth, WA, Australia
| | - Jiake Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
- *Correspondence: Sipin Zhu, ; Jiake Xu,
| |
Collapse
|
6
|
Yeh SJ, Chen BS. Systems Medicine Design based on Systems Biology Approaches and Deep Neural Network for Gastric Cancer. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:3019-3031. [PMID: 34232888 DOI: 10.1109/tcbb.2021.3095369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Gastric cancer (GC) is the third leading cause of cancer death in the world. It is associated with the stimulation of microenvironment, aberrant epigenetic modification, and chronic inflammation. However, few researches discuss the GC molecular progression mechanisms from the perspective of the system level. In this study, we proposed a systems medicine design procedure to identify essential biomarkers and find corresponding drugs for GC. At first, we did big database mining to construct candidate protein-protein interaction network (PPIN) and candidate gene regulation network (GRN). Second, by leveraging the next-generation sequencing (NGS) data, we performed system modeling and applied system identification and model selection to obtain real genome-wide genetic and epigenetic networks (GWGENs). To make the real GWGENs easy to analyze, the principal network projection method was used to extract the core signaling pathways denoted by KEGG pathways. Subsequently, based on the identified biomarkers, we trained a deep neural network of drug-target interaction (DeepDTI) with supervised learning and filtered our candidate drugs considering drug regulation ability and drug sensitivity. With the proposed systematic strategy, we not only shed the light on the progression of GC but also suggested potential multiple-molecule drugs efficiently.
Collapse
|
7
|
Kazantseva L, Becerra J, Santos-Ruiz L. Traditional Medicinal Plants as a Source of Inspiration for Osteosarcoma Therapy. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27155008. [PMID: 35956961 PMCID: PMC9370649 DOI: 10.3390/molecules27155008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 11/16/2022]
Abstract
Osteosarcoma is one of the most common types of bone cancers among paediatric patients. Despite the advances made in surgery, chemo-, and radiotherapy, the mortality rate of metastatic osteosarcoma remains unchangeably high. The standard drug combination used to treat this bone cancer has remained the same for the last 20 years, and it produces many dangerous side effects. Through history, from ancient to modern times, nature has been a remarkable source of chemical diversity, used to alleviate human disease. The application of modern scientific technology to the study of natural products has identified many specific molecules with anti-cancer properties. This review describes the latest discovered anti-cancer compounds extracted from traditional medicinal plants, with a focus on osteosarcoma research, and on their cellular and molecular mechanisms of action. The presented compounds have proven to kill osteosarcoma cells by interfering with different pathways: apoptosis induction, stimulation of autophagy, generation of reactive oxygen species, etc. This wide variety of cellular targets confer natural products the potential to be used as chemotherapeutic drugs, and also the ability to act as sensitizers in drug combination treatments. The major hindrance for these molecules is low bioavailability. A problem that may be solved by chemical modification or nano-encapsulation.
Collapse
Affiliation(s)
- Liliya Kazantseva
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29590 Málaga, Spain
| | - José Becerra
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29590 Málaga, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Cell Biology, Genetics and Physiology, Universidad de Málaga, 29071 Málaga, Spain
| | - Leonor Santos-Ruiz
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29590 Málaga, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Cell Biology, Genetics and Physiology, Universidad de Málaga, 29071 Málaga, Spain
- Correspondence:
| |
Collapse
|
8
|
Cui M, Wang Z, Huang LT, Wang JH. Parthenolide leads to proteomic differences in thyroid cancer cells and promotes apoptosis. BMC Complement Med Ther 2022; 22:99. [PMID: 35366876 PMCID: PMC8977004 DOI: 10.1186/s12906-022-03579-0] [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: 05/28/2021] [Accepted: 03/23/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Parthenolide has anti-inflammatory, immunomodulatory and anti-cancer activities. But its effect on thyroid cancer cells is still largely unknown.
Methods
Label-free quantitative proteomics and bioinformatics analysis were used to investigate the differentially expressed proteins and their functions in thyroid cancer treated with parthenolide and control pair. Hoechst 33258 fluorescent staining and Annexin V-FITC/PI double staining flow cytometry were used to detected BCPAP cells apoptosis. Parallel reaction monitoring (PRM) and quantitative real-time PCR were used to verify the expression of apoptosis-related differential proteins and their mRNA.
Results
Sixty up-regulated and 96 down-regulated differentially expressed proteins were identified in parthenolide treated thyroid cancer cells BCPAP compared with control thyroid cancer cells. The proteins were mainly relevant to various biological processes that included metabolic processes, response to extracellular stimulus and interaction with host. The molecular functions of most differentially expressed proteins were associated with binding functions and nucleotidyltransferase activity. According to the Kyoto Encyclopedia of Genes and Genomes, the differentially expressed proteins identified are primarily related to various types of metabolic pathways and DNA replication. In cell experiments in vitro, with the increase of the dose of parthenolide, the number of cells gradually decreased, the apoptosis rate gradually increased. PRM verified that the apoptosis-related proteins HMOX1 and GCLM were up-regulated and IL1B was down-regulated in BCPAP cells treated with parthenolide. The mRNA expressions of HMOX1, GCLM, ITGA6 and CASP8 were up-regulated and HSPA1A was down-regulated by PCR.
Conclusions
Parthenolide may influence the biological behavior of human thyroid cancer cells by affecting the expression of proteins related to cell metabolism and DNA replication. Parthenolide induced significant cellular morphological changes and apoptosis in human thyroid cancer cells, leading to an anti-proliferative effect.
Collapse
|
9
|
Recent and Ongoing Research into Metastatic Osteosarcoma Treatments. Int J Mol Sci 2022; 23:ijms23073817. [PMID: 35409176 PMCID: PMC8998815 DOI: 10.3390/ijms23073817] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 12/16/2022] Open
Abstract
The survival rate for metastatic osteosarcoma has not improved for several decades, since the introduction and refinement of chemotherapy as a treatment in addition to surgery. Over two thirds of metastatic osteosarcoma patients, many of whom are children or adolescents, fail to exhibit durable responses and succumb to their disease. Concerted efforts have been made to increase survival rates through identification of candidate therapies via animal studies and early phase trials of novel treatments, but unfortunately, this work has produced negligible improvements to the survival rate for metastatic osteosarcoma patients. This review summarizes data from clinical trials of metastatic osteosarcoma therapies as well as pre-clinical studies that report efficacy of novel drugs against metastatic osteosarcoma in vivo. Considerations regarding the design of animal studies and clinical trials to improve survival outcomes for metastatic osteosarcoma patients are also discussed.
Collapse
|
10
|
Das R, Mehta DK, Dhanawat M. Medicinal Plants in Cancer Treatment: Contribution of Nuclear Factor-Kappa B (NF-kB) Inhibitors. Mini Rev Med Chem 2022; 22:1938-1962. [PMID: 35260052 DOI: 10.2174/1389557522666220307170126] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/23/2021] [Accepted: 12/14/2021] [Indexed: 01/17/2023]
Abstract
Nuclear factor-kappa B (NF-κB) is one of the principal inducible proteins that is a predominant transcription factor known to control the gene expression in mammals and plays a pivotal role in regulating cell signalling in the body under certain physiological and pathological conditions. In cancer cells, such as colon, breast, pancreatic, ovarian, melanoma, and lymphoma, the NF-κB pathway has been reported to be active. In cellular proliferation, promoting angiogenesis, invasion, metastasis of tumour cells and blocking apoptosis, the constitutive activity of NF-κB signalling has been reported. Therefore, immense attention has been given to developing drugs targeting NF-κB signalling pathways to treat many types of tumours. They are a desirable therapeutic target for drugs, and many studies concentrated on recognizing compounds. They may be able to reverse or standstill the growth and spread of tumours that selectively interfere with this pathway. Recently, numerous substances derived from plants have been evaluated as possible inhibitors of the NF-κB pathway. These include various compounds, such as flavonoids, lignans, diterpenes, sesquiterpenes, polyphenols, etc. A study supported by folk medicine demonstrated that plant-derived compounds could suppress NF-κB signalling. Taking this into account, the present review revealed the anticancer potential of naturally occurring compounds which have been verified both by inhibiting the NF-κB signalling and suppressing growth and spread of cancer and highlighting their mechanism of NF-κB inhibition.
Collapse
Affiliation(s)
- Rina Das
- M.M.College of Pharmacy, Maharishi Markandeshwar (Deemed to be) University, Mullana, Ambala, HR, India
| | - Dinesh Kumar Mehta
- M.M.College of Pharmacy, Maharishi Markandeshwar (Deemed to be) University, Mullana, Ambala, HR, India
| | - Meenakshi Dhanawat
- M.M.College of Pharmacy, Maharishi Markandeshwar (Deemed to be) University, Mullana, Ambala, HR, India
| |
Collapse
|
11
|
Li W, Li Y, Tian W, Han X, Zhao J, Xin Z, Hu H, Li J, Hang K, Xu R. 2-methylbenzoyl berbamine, a multi-targeted inhibitor, suppresses the growth of human osteosarcoma through disabling NF-κB, ERK and AKT signaling networks. Aging (Albany NY) 2020; 12:15037-15049. [PMID: 32713851 PMCID: PMC7425514 DOI: 10.18632/aging.103565] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/08/2020] [Indexed: 12/15/2022]
Abstract
Osteosarcoma is the most common malignant bone tumor in children and young adults, and it has a survival rate of only 60% with current cytotoxic chemotherapy combined with aggressive surgery. The aim of this study was to evaluate the therapeutic efficacy of the berbamine derivative 2-methylbenzoyl berbamine (BBD24) for osteosarcoma in vitro and in vivo. We used human osteosarcoma cell lines, primary osteosarcoma cells and mouse models to evaluate the inhibitory effects of BBD24 on osteosarcoma and to determine the molecular mechanism. Our results showed that BBD24 inhibited the growth of the human osteosarcoma cell lines HOS and MG63 in a time- and dose-dependent manner. BBD24 also exhibited significant inhibitory effects on primary osteosarcoma cells. In contrast, BBD24 did not affect normal blood cells under the same conditions. Treatment with BBD24 induced apoptosis, necrosis and autophagy in osteosarcoma cells. Western blot analysis revealed that BBD24 activated the caspase-dependent pathway and downregulated the NF-kB, AKT, and ERK pathways. Finally, BBD24 treatment induced a significant inhibitory effect on the growth of osteosarcoma in nude mice. Our findings indicate that BBD24 is a multitarget inhibitor and may represent a new type of anticancer agent for osteosarcoma treatment.
Collapse
Affiliation(s)
- Weixu Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Yan Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Wenjia Tian
- Department of Endocrinology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310000, Zhejiang, China
| | - Xiuguo Han
- Department of Orthopaedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.,Department of Orthopedics, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Jie Zhao
- Department of Orthopedics, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Zengfeng Xin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China
| | - Hejia Hu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China
| | - Jun Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Kai Hang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Rongzhen Xu
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China.,Department of Hematology, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou 310009, Zhejiang, China
| |
Collapse
|
12
|
Yasuda N, Takenaka S, Nakai S, Nakai T, Yamada S, Imura Y, Outani H, Hamada K, Yoshikawa H, Naka N. TAS-115 inhibits PDGFRα/AXL/FLT-3 signaling and suppresses lung metastasis of osteosarcoma. FEBS Open Bio 2020; 10:767-779. [PMID: 32128992 PMCID: PMC7193166 DOI: 10.1002/2211-5463.12827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/21/2020] [Accepted: 02/28/2020] [Indexed: 11/22/2022] Open
Abstract
Osteosarcoma is the most common malignant bone tumor in adolescence and childhood. Metastatic osteosarcoma has a poor prognosis with an overall 5‐year survival rate of approximately 20%. TAS‐115 is a novel multiple receptor tyrosine kinase inhibitor that is currently undergoing clinical trials. Using the mouse highly lung‐metastatic osteosarcoma cell line, LM8, we showed that TAS‐115 suppressed the growth of subcutaneous grafted tumor and lung metastasis of osteosarcoma at least partially through the inhibition of platelet‐derived growth factor receptor alpha, AXL, and Fms‐like tyrosine kinase 3 phosphorylation. We also show that these signaling pathways are activated in various human osteosarcoma cell lines and are involved in proliferation. Our results suggest that TAS‐115 may have potential for development into a novel treatment for metastatic osteosarcoma.
Collapse
Affiliation(s)
- Naohiro Yasuda
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Satoshi Takenaka
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Sho Nakai
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Takaaki Nakai
- Department of Orthopaedic Surgery, Kawachi General Hospital, Higashiosaka, Japan
| | - Shutaro Yamada
- Department of Orthopaedic Surgery, Yao Municipal Hospital, Japan
| | - Yoshinori Imura
- Musculoskeletal Oncology Service, Osaka International Cancer Institute, Japan
| | - Hidetatsu Outani
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kenichiro Hamada
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hideki Yoshikawa
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Japan.,Department of Orthopaedic Surgery, Toyonaka Municipal Hospital, Japan
| | - Norifumi Naka
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Japan.,Musculoskeletal Oncology Service, Osaka International Cancer Institute, Japan
| |
Collapse
|
13
|
Che ST, Bie L, Li X, Qi H, Yu P, Zuo L. Parthenolide inhibits the proliferation and induces the apoptosis of human uveal melanoma cells. Int J Ophthalmol 2019; 12:1531-1538. [PMID: 31637187 DOI: 10.18240/ijo.2019.10.03] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 05/21/2019] [Indexed: 02/08/2023] Open
Abstract
AIM To explore the effect of parthenolide (PTL) on human uveal melanoma (UM) cells (C918 and SP6.5 cells) and its molecular mechanism. METHODS Carboxyfluorescein succinimidyl amino ester (CFSE) assays and cell counting kit-8 (CCK-8) were performed to detect the cell viability. Flow cytometry was used to analyze cell cycle and apoptosis. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot assays were performed to measure proliferation-related and apoptosis-related factors. RESULTS Firstly, PTL decreased the viability of C918 and SP6.5 cells in a dose-dependent manner, and the effect of PTL on C918 cells was stronger than on SP6.5; however, it did not affect normal cells. Secondly, PTL increased the proportion of cell number at cell cycle G1 phase in C918 cells, and decreased the proportion of cell number at S phase, but the proportion did not change at G2 phase. In addition, PTL induced the apoptosis of C918 cells, and decreased the expressions of Cyclin D1, B-cell lymphoma-2 (Bcl-2) and B-cell lymphoma-extra large (Bcl-XL). Also, PTL increased Cyclin inhibition protein 1 (P21), Bcl-2-associated X protein (Bax), Cysteinyl aspartate specific proteinas-3 (Caspase-3) and Caspase-9 expression. However, the expression of Caspase-8 was not changed. CONCLUSION PTL inhibites proliferation and induces apoptosis in UM cells by arresting G1 phase and regulating mitochondrial pathway, however, it does not affect normal cells.
Collapse
Affiliation(s)
- Song-Tian Che
- Department of Ocular Fundus Disease, the Second Hospital of Jilin University, Changchun 130022, Jilin Province, China
| | - Li Bie
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun 130022, Jilin Province, China
| | - Xu Li
- Department of Ocular Fundus Disease, the Second Hospital of Jilin University, Changchun 130022, Jilin Province, China
| | - Hui Qi
- Department of Ocular Fundus Disease, the Second Hospital of Jilin University, Changchun 130022, Jilin Province, China
| | - Peng Yu
- Department of Ocular Fundus Disease, the Second Hospital of Jilin University, Changchun 130022, Jilin Province, China
| | - Ling Zuo
- Department of Ocular Fundus Disease, the Second Hospital of Jilin University, Changchun 130022, Jilin Province, China
| |
Collapse
|
14
|
Dawood M, Ooko E, Efferth T. Collateral Sensitivity of Parthenolide via NF-κB and HIF-α Inhibition and Epigenetic Changes in Drug-Resistant Cancer Cell Lines. Front Pharmacol 2019; 10:542. [PMID: 31164821 PMCID: PMC6536578 DOI: 10.3389/fphar.2019.00542] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 04/30/2019] [Indexed: 12/18/2022] Open
Abstract
Parthenolide (PT) is a sesquiterpene lactone isolated from Tanacetum parthenium. In this study, PT showed varying cytotoxic effects against different solid tumor cell lines. HCT116 (p53+/+) colon carcinoma cells and their parental HCT116 knockout p53 (p53-/-) cell lines showed a resistance degree of 2.36. On the other hand, wild-type U87.MG cells or cells transfected with a deletion-activated EGFR cDNA (U87.MGΔEGFR) exhibited slight sensitivity toward PT. Multidrug-resistant MDA-MB-231-BCRP cells were even more sensitive toward PT than sensitive MDA-MB-231-pcDNA cells with a resistance degree of 0.07 (collateral sensitivity). To the best of our knowledge, hypersensitivity (collateral sensitivity) in MDA-MB-231-BCRP cell line is reported in this study for the first time. We attempted to identify the mechanism of collateral sensitivity. Firstly, we found that PT bound to IKK preventing IκBα degradation and eventually inhibition of the nuclear factor kappa B (NF-κB) pathway. Down-regulation of hypoxia inducing factor 1-alpha (HIF-1α) in MDA-MB-231-BCRP resistant cells may be a second mechanism, since it is a target gene of NF-κB. Moreover, PT also showed epigenetic effect by inhibition of HDAC activity as shown using both molecular docking and HDAC activity assay. Based on COMPARE and hierarchical cluster analyses, we found gene expression profiles that predicted sensitivity or resistance of 47 tumor cell lines toward PT. Interestingly, pathway analyses of gene expression profiles revealed NF-κB and HIF signaling as top networks of these genes, cellular functions and canonical pathways influencing the activity of PT against tumor cells. In conclusion, PT exerted profound cytotoxic activity against various cancer cell lines mainly against BCRP-overexpressing tumor cells, suggesting PT as novel candidate for cancer treatment.
Collapse
Affiliation(s)
- Mona Dawood
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Edna Ooko
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
15
|
Li R, Shi Y, Zhao S, Shi T, Zhang G. NF-κB signaling and integrin-β1 inhibition attenuates osteosarcoma metastasis via increased cell apoptosis. Int J Biol Macromol 2019; 123:1035-1043. [DOI: 10.1016/j.ijbiomac.2018.11.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 10/13/2018] [Accepted: 11/01/2018] [Indexed: 12/31/2022]
|
16
|
Marino S, Bishop RT, Mollat P, Idris AI. Pharmacological Inhibition of the Skeletal IKKβ Reduces Breast Cancer-Induced Osteolysis. Calcif Tissue Int 2018; 103:206-216. [PMID: 29455416 PMCID: PMC6061461 DOI: 10.1007/s00223-018-0406-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 02/12/2018] [Indexed: 11/30/2022]
Abstract
IKKβ has previously been implicated in breast cancer bone metastasis and bone remodelling. However, the contribution of IKKβ expressed by bone cells of the tumour microenvironment to breast cancer-induced osteolysis has yet to be investigated. Here, we studied the effects of the verified selective IKKβ inhibitors IKKβIII or IKKβV on osteoclast formation and osteoblast differentiation in vitro and in vivo, human and mouse breast cancer cells' support for osteoclast formation and signalling in vitro and osteolysis ex vivo and in immunocompetent mice after supracalvarial injection of human MDA-MB-231 conditioned medium or intra-cardiac injection of syngeneic 4T1 breast cancer cells. Pre-treatment with IKKβIII or IKKβV prior to exposure to tumour-derived factors from human and mouse breast cancer cell lines protected against breast cancer-induced osteolysis in two independent immunocompetent mouse models of osteolysis and the ex vivo calvarial bone organ system. Detailed functional and mechanistic studies showed that direct inhibition of IKKβ kinase activity in osteoblasts and osteoclasts was associated with significant reduction of osteoclast formation, enhanced osteoclast apoptosis and reduced the ability of osteoblasts to support osteoclastogenesis in vitro. When combined with previous findings that suggest NFκB inhibition reduces breast cancer tumorigenesis and metastasis our present findings have an important clinical implication on raising the possibility that IKKβ inhibitors, as bone anabolics, osteoclast inhibitors as well as anti-metastatic agents, may have advantages over anti-osteoclasts agents in the treatment of both skeletal and non-skeletal complications associated with metastatic breast cancer.
Collapse
Affiliation(s)
- Silvia Marino
- Department of Oncology and Metabolism, University of Sheffield, Medical School, 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, University of Sheffield, Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Patrick Mollat
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230, Romainville, France
| | - Aymen I Idris
- Department of Oncology and Metabolism, University of Sheffield, Medical School, 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
|
Li H, Lu H, Lv M, Wang Q, Sun Y. Parthenolide facilitates apoptosis and reverses drug-resistance of human gastric carcinoma cells by inhibiting the STAT3 signaling pathway. Oncol Lett 2018; 15:3572-3579. [PMID: 29467878 PMCID: PMC5796286 DOI: 10.3892/ol.2018.7739] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 12/06/2017] [Indexed: 01/22/2023] Open
Abstract
In the present study, SGC-7901/DDP cells were treated with different concentrations of parthenolide (PN) (2.5–15 µmol/l), cisplatin (DDP) (1.25–15 µg/ml) and PN+DDP. The proliferation inhibition rates were measured using an MTT assay, and the synergies of PN and DDP were analyzed. The effect of PN and DDP on SGC-7901/DDP cell proliferation demonstrated a time- and concentration-dependent association, and a synergy between PN and DDP was identified. DAPI staining and flow cytometry results indicated that 15 µmol/l PN significantly induced SGC-7901/DDP apoptosis and G1 phase arrest compared with the untreated control group. Western blotting analysis results indicated that among the apoptosis-associated proteins, there were dose-dependent increases in the protein expression of apoptosis regulator BAX, cellular tumor antigen p53, cleaved caspase-3 and cleaved capase-9, and decreases in apoptosis regulator Bcl-2 and Bcl-xL protein expression levels. Among the cell cycle-associated proteins, cyclin D1 expression was significantly decreased, cyclin-dependent kinase inhibitor 1 expression was significantly increased, and signal transducer and activator of transcription 3 (STAT3) activation was inhibited. Scratch and Transwell assay results revealed that PN significantly inhibited SGC-7901/DDP cell migration, and invasion. The present study demonstrated that PN induces SGC-7901/DDP apoptosis, inhibits SGC-7901/DDP proliferation, migration and invasion, and enhances the drug sensitivity of the cells to DDP. The underlying mechanisms may be associated with inhibition of the STAT3 signaling pathway and regulation of the downstream apoptotic protein and cyclin expression levels.
Collapse
Affiliation(s)
- Hua Li
- Department of Oncology, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China.,Department of Oncology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Hang Lu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Meng Lv
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Qingsheng Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Yuping Sun
- Department of Oncology, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
| |
Collapse
|
18
|
Kim SL, Park YR, Lee ST, Kim SW. Parthenolide suppresses hypoxia-inducible factor-1α signaling and hypoxia induced epithelial-mesenchymal transition in colorectal cancer. Int J Oncol 2017; 51:1809-1820. [PMID: 29075793 DOI: 10.3892/ijo.2017.4166] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 09/29/2017] [Indexed: 11/06/2022] Open
Abstract
Activation of hypoxia-inducible factor 1α (HIF‑1α) is frequently observed in solid tumors and it has been associated with various pathophysiological processes, including epithelial‑mesenchymal transition (EMT). Previously, we reported that parthenolide (PT), an inhibitor of nuclear factor-κB (NF-κB), is a promising anticancer agent because it promotes apoptosis of human colorectal cancer (CRC). Here, we investigated a new molecular mechanism by which PT acts on HIF‑1α and hypoxia contributing to EMT by NF‑κB inhibition. Cell viability, DNA binding activity, vascular cell tube formation and cell motility were studied after treatment of PT in hypoxic or normoxic condition. Moreover, effects of PT on hypoxia signaling and hypoxia-induced EMT signaling were investigated. We also examined the inhibitory effect of PT on CRC progression in xenografts. We demonstrated that PT markedly inhibits hypoxia dependent HIF‑1α activity and angiogenesis by preventing NF-κB activation. We also report that PT decreases the level of proteins associated with glucose metabolism, angiogenesis, development and survival that are regulated by HIF‑1α. Furthermore, we verified that PT protects the morphological change from epithelial to mesenchymal state, inhibits matrix metalloproteinase (MMP) enzyme activity and decreases cell motility involved in the -regulation of the hypoxia-induced EMT markers. In addition, PT inhibits growth in CRC xenograft models and regulates NF‑κB, HIF‑1α and EMT specific marker in tissue specimens. Our data demonstrated that PT can inhibit HIF‑1α signaling and hypoxia-induced EMT, suggesting a novel molecular mechanism for HIF‑1α mediated cancer progression and metastasis.
Collapse
Affiliation(s)
- Se Lim Kim
- Department of Internal Medicine, Research Institute of Clinical Medicine of Chonbuk National University, Jeonju, Republic of Korea
| | - Young Ran Park
- Department of Internal Medicine, Research Institute of Clinical Medicine of Chonbuk National University, Jeonju, Republic of Korea
| | - Soo Teik Lee
- Department of Internal Medicine, Research Institute of Clinical Medicine of Chonbuk National University, Jeonju, Republic of Korea
| | - Sang-Wook Kim
- Department of Internal Medicine, Research Institute of Clinical Medicine of Chonbuk National University, Jeonju, Republic of Korea
| |
Collapse
|
19
|
Li M, Guan H. Noncoding RNAs Regulating NF-κB Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 927:317-36. [PMID: 27376741 DOI: 10.1007/978-981-10-1498-7_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
As transcription factors that regulate expression of a variety of genes essential for diverse physiological and pathological processes, nuclear factor kappa B (NF-κB) family molecules play important roles in the development and progression of malignant tumor, and constitutive activation of NF-κB has been evidenced in various types of tumor tissues. Underlying its pathologic role, deregulated expression and/or transactivating activity of NF-κB usually involves multiple layers of molecular mechanisms. Noncoding RNAs, including microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), are known to modulate expression and biological functions of regulatory proteins in a variety of cancer contexts. In this chapter, the regulatory role of miRNAs and lncRNAs in NF-κB signaling in malignant diseases will be discussed.
Collapse
Affiliation(s)
- Mengfeng Li
- Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan Road II, Guangzhou, China.
| | - Hongyu Guan
- Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan Road II, Guangzhou, China
| |
Collapse
|
20
|
Liu YC, Kim SL, Park YR, Lee ST, Kim SW. Parthenolide promotes apoptotic cell death and inhibits the migration and invasion of SW620 cells. Intest Res 2017; 15:174-181. [PMID: 28522946 PMCID: PMC5430008 DOI: 10.5217/ir.2017.15.2.174] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 06/14/2016] [Accepted: 06/14/2016] [Indexed: 12/25/2022] Open
Abstract
Background/Aims Parthenolide (PT), a principle component derived from feverfew (Tanacetum parthenium), is a promising anticancer agent and has been shown to promote apoptotic cell death in various cancer cells. In this study, we focused on its functional role in apoptosis, migration, and invasion of human colorectal cancer (CRC) cells. Methods SW620 cells were employed as representative human CRC cells. We performed the MTT assay and cell cycle analysis to measure apoptotic cell death. The wound healing, Transwell migration, and Matrigel invasion assays were performed to investigate the effect of PT on cell migration/invasion. Western blotting was used to establish the signaling pathway of apoptosis and cell migration/invasion. Results PT exerts antiproliferative effect and induces apoptotic cell death of SW620 cells. In addition, PT prevents cell migration and invasion in a dose-dependent manner. Moreover, PT markedly suppressed migration/invasion-related protein expression, including E-cadherin, β-catenin, vimentin, Snail, cyclooxygenase-2, matrix metalloproteinase-2 (MMP-2), and MMP-9 in SW620 cells. PT also inhibited the expression of antiapoptotic proteins (Bcl-2 and Bcl-xL) and activated apoptosis terminal factor (caspase-3) in a dose-dependent manner. Conclusions Our results suggest that PT is a potential novel therapeutic agent for aggressive CRC treatment.
Collapse
Affiliation(s)
- Yu Chuan Liu
- Department of Internal Medicine, Research Institute of Clinical Medicine of Chonbuk National University Hospital, Jeonju, Korea.,Biomedical Research Institute of Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju, Korea
| | - Se Lim Kim
- Department of Internal Medicine, Research Institute of Clinical Medicine of Chonbuk National University Hospital, Jeonju, Korea.,Biomedical Research Institute of Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju, Korea
| | - Young Ran Park
- Department of Internal Medicine, Research Institute of Clinical Medicine of Chonbuk National University Hospital, Jeonju, Korea.,Biomedical Research Institute of Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju, Korea
| | - Soo-Teik Lee
- Department of Internal Medicine, Research Institute of Clinical Medicine of Chonbuk National University Hospital, Jeonju, Korea.,Biomedical Research Institute of Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju, Korea
| | - Sang Wook Kim
- Department of Internal Medicine, Research Institute of Clinical Medicine of Chonbuk National University Hospital, Jeonju, Korea.,Biomedical Research Institute of Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju, Korea
| |
Collapse
|
21
|
Ren Y, Yu J, Kinghorn AD. Development of Anticancer Agents from Plant-Derived Sesquiterpene Lactones. Curr Med Chem 2017; 23:2397-420. [PMID: 27160533 DOI: 10.2174/0929867323666160510123255] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 04/26/2016] [Accepted: 05/09/2016] [Indexed: 12/24/2022]
Abstract
Sesquiterpene lactones are of considerable interest due to their potent bioactivities, including cancer cell cytotoxicity and antineoplastic efficacy in in vivo studies. Among these compounds, artesunate, dimethylaminoparthenolide, and L12ADT peptide prodrug, a derivative of thapsigargin, are being evaluated in the current cancer clinical or preclinical trials. Based on the structures of several antitumor sesquiterpene lactones, a number of analogues showing greater potency have been either isolated as natural products or partially synthesized, and some potential anticancer agents that have emerged from this group of lead compounds have been investigated extensively. The present review focuses on artemisinin, parthenolide, thapsigargin, and their naturally occurring or synthetic analogues showing potential anticancer activity. This provides an overview of the advances in the development of these types of sesquiterpene lactones as potential anticancer agents, including their structural characterization, synthesis and synthetic modification, and antitumor potential, with the mechanism of action and structure-activity relationships also discussed. It is hoped that this will be helpful in stimulating the further interest in developing sesquiterpene lactones and their derivatives as new anticancer agents.
Collapse
Affiliation(s)
| | | | - A Douglas Kinghorn
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| |
Collapse
|
22
|
Perez DR, Smagley Y, Garcia M, Carter MB, Evangelisti A, Matlawska-Wasowska K, Winter SS, Sklar LA, Chigaev A. Cyclic AMP efflux inhibitors as potential therapeutic agents for leukemia. Oncotarget 2016; 7:33960-82. [PMID: 27129155 PMCID: PMC5085131 DOI: 10.18632/oncotarget.8986] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 04/16/2016] [Indexed: 12/24/2022] Open
Abstract
Apoptotic evasion is a hallmark of cancer. We propose that some cancers may evade cell death by regulating 3'-5'-cyclic adenosine monophosphate (cAMP), which is associated with pro-apoptotic signaling. We hypothesize that leukemic cells possess mechanisms that efflux cAMP from the cytoplasm, thus protecting them from apoptosis. Accordingly, cAMP efflux inhibition should result in: cAMP accumulation, activation of cAMP-dependent downstream signaling, viability loss, and apoptosis. We developed a novel assay to assess cAMP efflux and performed screens to identify inhibitors. In an acute myeloid leukemia (AML) model, several identified compounds reduced cAMP efflux, appropriately modulated pathways that are responsive to cAMP elevation (cAMP-responsive element-binding protein phosphorylation, and deactivation of Very Late Antigen-4 integrin), and induced mitochondrial depolarization and caspase activation. Blocking adenylyl cyclase activity was sufficient to reduce effects of the most potent compounds. These compounds also decreased cAMP efflux and viability of B-lineage acute lymphoblastic leukemia (B-ALL) cell lines and primary patient samples, but not of normal primary peripheral blood mononuclear cells. Our data suggest that cAMP efflux is a functional feature that could be therapeutically targeted in leukemia. Furthermore, because some of the identified drugs are currently used for treating other illnesses, this work creates an opportunity for repurposing.
Collapse
Affiliation(s)
- Dominique R. Perez
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
- University of New Mexico Center for Molecular Discovery, Albuquerque, NM, USA
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Yelena Smagley
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
- University of New Mexico Center for Molecular Discovery, Albuquerque, NM, USA
| | - Matthew Garcia
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
- University of New Mexico Center for Molecular Discovery, Albuquerque, NM, USA
| | - Mark B. Carter
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
- University of New Mexico Center for Molecular Discovery, Albuquerque, NM, USA
| | - Annette Evangelisti
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
- University of New Mexico Center for Molecular Discovery, Albuquerque, NM, USA
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Ksenia Matlawska-Wasowska
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
- Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Stuart S. Winter
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
- Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Larry A. Sklar
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
- University of New Mexico Center for Molecular Discovery, Albuquerque, NM, USA
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Alexandre Chigaev
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
- University of New Mexico Center for Molecular Discovery, Albuquerque, NM, USA
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| |
Collapse
|
23
|
Rana S, Blowers EC, Tebbe C, Contreras JI, Radhakrishnan P, Kizhake S, Zhou T, Rajule RN, Arnst JL, Munkarah AR, Rattan R, Natarajan A. Isatin Derived Spirocyclic Analogues with α-Methylene-γ-butyrolactone as Anticancer Agents: A Structure-Activity Relationship Study. J Med Chem 2016; 59:5121-7. [PMID: 27077228 DOI: 10.1021/acs.jmedchem.6b00400] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Design, synthesis, and evaluation of α-methylene-γ-butyrolactone analogues and their evaluation as anticancer agents is described. SAR identified a spirocyclic analogue 19 that inhibited TNFα-induced NF-κB activity, cancer cell growth and tumor growth in an ovarian cancer model. A second iteration of synthesis and screening identified 29 which inhibited cancer cell growth with low-μM potency. Our data suggest that an isatin-derived spirocyclic α-methylene-γ-butyrolactone is a suitable core for optimization to identify novel anticancer agents.
Collapse
Affiliation(s)
| | | | - Calvin Tebbe
- Division of Gynecology Oncology, Department of Women's Health and Josephine Ford Cancer Center, Henry Ford Hospital , Detroit, Michigan 48202, United States
| | | | | | | | | | | | | | - Adnan R Munkarah
- Division of Gynecology Oncology, Department of Women's Health and Josephine Ford Cancer Center, Henry Ford Hospital , Detroit, Michigan 48202, United States
| | - Ramandeep Rattan
- Division of Gynecology Oncology, Department of Women's Health and Josephine Ford Cancer Center, Henry Ford Hospital , Detroit, Michigan 48202, United States
| | | |
Collapse
|
24
|
Natural compounds for pediatric cancer treatment. Naunyn Schmiedebergs Arch Pharmacol 2015; 389:131-49. [DOI: 10.1007/s00210-015-1191-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 11/08/2015] [Indexed: 12/13/2022]
|
25
|
Anti-Inflammatory and Cytostatic Activities of a Parthenolide-Like Sesquiterpene Lactone from Cota palaestina subsp. syriaca. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:474597. [PMID: 26089941 PMCID: PMC4452324 DOI: 10.1155/2015/474597] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 04/14/2015] [Indexed: 11/17/2022]
Abstract
A sesquiterpene lactone 1-β,10-Epoxy-6-hydroxy-1,10H-inunolide (K100) was isolated through "bioassay-guided fractionation" from Cota palaestina subsp. syriaca, an Eastern Mediterranean endemic plant. K100 inhibited endotoxin- (ET-) induced proinflammatory markers: IL-6, MMP-9, and NO in normal mouse mammary SCp2 Cells. Molecular docking in silico suggested that K100, having highly analogous structure as parthenolide (PTL), an anticancer compound, could bind PTL target proteins at similar positions and with comparable binding affinities. Both compounds, K100 and PTL, inhibited the proliferation and prolonged the S-phase of the cell cycle of breast adenocarcinoma MDA-MB-231 cells grown in 2D cultures. Noncytotoxic concentrations of K100 and PTL decreased the proliferation rate of MDA-MB-231 and shifted their morphology from stellate to spherical colonies in 3D cultures. This was accompanied with a significant increase in the amount of small colonies and a decrease in the amount of large colonies. Moreover, K100 and PTL decreased cellular motility and invasiveness of MDA-MB-231 cells. In summary, these results suggest that K100 exhibits PTL-analogous anti-inflammatory, cytostatic, and antimetastatic effects.
Collapse
|
26
|
Modulatory effect of celastrol on Th1/Th2 cytokines profile, TLR2 and CD3+ T-lymphocyte expression in a relapsing-remitting model of multiple sclerosis in rats. Eur J Pharmacol 2014; 742:102-12. [PMID: 25218987 DOI: 10.1016/j.ejphar.2014.09.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 08/29/2014] [Accepted: 09/01/2014] [Indexed: 02/05/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune inflammatory demyelinating disease of brain and spinal cord that has an increasing incidence worldwide and classically presents in a relapsing-remitting form. This study was designed to induce a relapsing-remitting model of experimental autoimmune encephalomyelitis (EAE) to investigate the possible modulatory effect of celastrol on Th1/Th2 cytokines profile, immunohistochemical expression of TLR2, and CD3+T-lymphocytic count. Eighteen female Sprague Dawley rats were divided into 3 groups; where group I served as normal control, group II as EAE+vehicle, and group III as EAE treated by celastrol (1mg/kg/day, i.p.) started at 10th day till 42nd day post-immunization. The clinical score of rats in group II (EAE+vehicle) was relapsed after the re-challenge at the 35th day post-immunization and exhibited significant positive association with serum TNF-α, NF-κB expression and nitrites levels in brain and spinal cord, and CD3+ T-lymphocytic count in brain tissues while serum IL-10 showed significant negative association. Treatment of EAE by celastrol caused amelioration of the clinical score and inhibited the relapse. It caused significant shift in cytokines profile from Th1 by decrease in TNF-α towards Th2 pattern by increase in IL-10. Moreover, celastrol treatment resulted in significant reduction in NF-κB expression, nitrites levels, as well as immunohistochemical expression of TLR2 and CD3+ T-lymphocytic count. The beneficial effect of celastrol was further confirmed histopathologically by reduction in H&E score. Collectively, these results provide a promising pre-clinical evidence and conclusion about use of celastrol in treatment of multiple sclerosis that must be accessed in further clinical studies.
Collapse
|
27
|
Liu Q, Manzano D, Tanić N, Pesic M, Bankovic J, Pateraki I, Ricard L, Ferrer A, de Vos R, van de Krol S, Bouwmeester H. Elucidation and in planta reconstitution of the parthenolide biosynthetic pathway. Metab Eng 2014; 23:145-53. [PMID: 24704560 DOI: 10.1016/j.ymben.2014.03.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 03/04/2014] [Accepted: 03/25/2014] [Indexed: 01/08/2023]
Abstract
Parthenolide, the main bioactive compound of the medicinal plant feverfew (Tanacetum parthenium), is a promising anti-cancer drug. However, the biosynthetic pathway of parthenolide has not been elucidated yet. Here we report on the isolation and characterization of all the genes from feverfew that are required for the biosynthesis of parthenolide, using a combination of 454 sequencing of a feverfew glandular trichome cDNA library, co-expression analysis and metabolomics. When parthenolide biosynthesis was reconstituted by transient co-expression of all pathway genes in Nicotiana benthamiana, up to 1.4μgg(-1) parthenolide was produced, mostly present as cysteine and glutathione conjugates. These relatively polar conjugates were highly active against colon cancer cells, with only slightly lower activity than free parthenolide. In addition to these biosynthetic genes, another gene encoding a costunolide and parthenolide 3β-hydroxylase was identified opening up further options to improve the water solubility of parthenolide and therefore its potential as a drug.
Collapse
Affiliation(s)
- Qing Liu
- Laboratory of Plant Physiology, Wageningen University, Wageningen, The Netherlands
| | - David Manzano
- Department of Molecular Genetics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB Bellaterra, E-08193 Barcelona, Spain; Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain
| | - Nikola Tanić
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Serbia
| | - Milica Pesic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Serbia
| | - Jasna Bankovic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Serbia
| | - Irini Pateraki
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain; Department of Plant and Environmental Sciences, Faculty of Science, University of Copenhagen, Thorvaldsensvej 40, Frederiksberg C, Copenhagen, Denmark
| | - Lea Ricard
- Laboratory of Plant Physiology, Wageningen University, Wageningen, The Netherlands
| | - Albert Ferrer
- Department of Molecular Genetics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB Bellaterra, E-08193 Barcelona, Spain; Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain
| | - Ric de Vos
- Plant Research International, Wageningen, The Netherlands; Centre for BioSystems Genomics, Wageningen, The Netherlands; Netherlands Metabolomics Centre, Leiden, The Netherlands
| | - Sander van de Krol
- Laboratory of Plant Physiology, Wageningen University, Wageningen, The Netherlands
| | - Harro Bouwmeester
- Laboratory of Plant Physiology, Wageningen University, Wageningen, The Netherlands.
| |
Collapse
|
28
|
Amorim MHR, Gil da Costa RM, Lopes C, Bastos MMSM. Sesquiterpene lactones: adverse health effects and toxicity mechanisms. Crit Rev Toxicol 2014; 43:559-79. [PMID: 23875764 DOI: 10.3109/10408444.2013.813905] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Sesquiterpene lactones (STLs) present a wide range of biological activities, mostly based on their alkylating capabilities, which underlie their therapeutic potential. These compounds are the active constituents of a variety of plants, frequently used as herbal remedies. STLs such as artemisinin and its derivatives are in use as first-line antimalarials while others, such as parthenolide, have recently reached cancer clinical trials. However, the toxicological profile of these compounds must be thoroughly characterized, since the same properties that make STL useful medicines can also cause severe toxicity. STL-containing plants have long been known to induce a contact dermatitis in exposed farm workers, and also to cause several toxic syndromes in farm animals. More recently, concerns are been raised regarding the genotoxic potential of these compounds and the embryotoxicity of artemisinins. A growing number of STLs are being reported to be mutagenic in different in vitro and in vivo assays. As yet no systematic studies have been published, but the genotoxicity of STLs seems to depend not so much on direct DNA alkylation as on oxidative DNA damage and other partially elucidated mechanisms. As the medicinal use of these compounds increases, further studies of their toxic potential are needed, especially those focusing on the structural determinants of genotoxicity and embryotoxicity.
Collapse
Affiliation(s)
- M Helena R Amorim
- Chemical Engineering Department, Faculty of Engineering, University of Porto, Portugal
| | | | | | | |
Collapse
|
29
|
Brabletz T, Lyden D, Steeg PS, Werb Z. Roadblocks to translational advances on metastasis research. Nat Med 2013; 19:1104-9. [PMID: 24013756 DOI: 10.1038/nm.3327] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Promising advances in cancer therapy stemming from an increasing understanding of the molecular and genetic underpinnings of the tumorigenic process have been fueled by a strong, determined scientific community, influential patient advocacy groups and committed funding bodies. Despite these efforts, the development of effective drugs to prevent systemic dissemination of cancer cells or to eliminate overt metastasis in secondary organs remains a challenge to both researchers and physicians. In an attempt to tackle the most relevant and timely translational issues, a meeting held in 2012 as a result of a successful partnership between the Volkswagen Foundation and Nature Medicine brought together a group of metastasis research experts to identify the most important hurdles and help create a framework for potential clinical and translational strategies.
Collapse
Affiliation(s)
- Thomas Brabletz
- 1] Department of General and Visceral Surgery, Comprehensive Cancer Center and BIOSS Centre for Biological Signalling Studies, University of Freiburg Medical Center, Freiburg, Germany. [2]
| | | | | | | |
Collapse
|
30
|
Banerjee S, Thayanithy V, Sangwan V, Mackenzie TN, Saluja AK, Subramanian S. Minnelide reduces tumor burden in preclinical models of osteosarcoma. Cancer Lett 2013; 335:412-20. [PMID: 23499892 PMCID: PMC4386634 DOI: 10.1016/j.canlet.2013.02.050] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 02/22/2013] [Accepted: 02/26/2013] [Indexed: 01/10/2023]
Abstract
Osteosarcoma is the most common bone cancer in children and adolescents with a 5-year survival rate of about 70%. In this study, we have evaluated the preclinical therapeutic efficacy of the novel synthetic drug, Minnelide, a prodrug of triptolide on osteosarcoma. Triptolide was effective in significantly inducing apoptosis in all osteosarcoma cell lines tested but had no significant effect on the human osteoblast cells. Notably, Minnelide treatment significantly reduced tumor burden and lung metastasis in the orthotopic and lung colonization models. Triptolide/Minnelide effectively downregulated the levels of pro-survival proteins such as heat shock proteins, cMYC, survivin and targets the NF-κB pathway.
Collapse
Affiliation(s)
- Sulagna Banerjee
- Division of Basic and Translational Research, Department of Surgery, University of Minnesota
| | - Venugopal Thayanithy
- Division of Basic and Translational Research, Department of Surgery, University of Minnesota
| | - Veena Sangwan
- Division of Basic and Translational Research, Department of Surgery, University of Minnesota
| | - Tiffany N. Mackenzie
- Division of Basic and Translational Research, Department of Surgery, University of Minnesota
| | - Ashok K. Saluja
- Division of Basic and Translational Research, Department of Surgery, University of Minnesota
- Masonic Cancer Center, University of Minnesota
| | - Subbaya Subramanian
- Division of Basic and Translational Research, Department of Surgery, University of Minnesota
- Masonic Cancer Center, University of Minnesota
| |
Collapse
|
31
|
Parthenolide reverses doxorubicin resistance in human lung carcinoma A549 cells by attenuating NF-κB activation and HSP70 up-regulation. Toxicol Lett 2013; 221:73-82. [PMID: 23792430 DOI: 10.1016/j.toxlet.2013.06.215] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 06/10/2013] [Accepted: 06/12/2013] [Indexed: 11/23/2022]
Abstract
Chemotherapy resistance represents a major problem for the treatment of patients with lung carcinomas. Parthenolide (PN), a naturally occurring small molecule found in herb feverfew, has been used in clinical treatment. Although its importance in treating the chemotherapy resistance has been shown, the pharmacological benefits of PN for lung cancer with multidrug resistance are underappreciated. Using human lung epithelial carcinoma A549 and A549 derived DOX-resistant A549/DOX cell lines, we found that PN enhanced the apoptotic cytotoxicity of DOX in A549/DOX cells. PN inhibited P-glycoprotein (P-gp) up-regulation and promoted the intracellular accumulation of DOX in A549/DOX cells. PN also exhibited inhibitory effect on NF-κB activation in A549/DOX cells, suggesting that inhibition of NF-κB was involved in attenuating P-gp expression by PN. Moreover, we found that PN could also effectively inhibit the HSP70 up-regulation in A549/DOX cells. Further studies revealed a positive correlation between HSP70 and P-gp expression. Overexpression of HSP70 upregulated P-gp expression independently of NF-κB activation in A549 cells, and knockdown of HSP70 caused a reduced expression of P-gp in A549/DOX cells. RT-PCR experiments showed that HSP70 modulated the P-gp expression mainly at transcription level. Taken together, PN can reverse DOX resistance through suppressing P-gp expression by mechanisms involving attenuation of NF-κB activation and HSP70 up-regulation. Our results not only provide insight into potential use of PN in reversing P-gp mediated MDR to facilitate lung cancer chemotherapy, but also highlight a potential role of HSP70 in the development of drug resistance.
Collapse
|
32
|
Sung B, Prasad S, Yadav VR, Gupta SC, Reuter S, Yamamoto N, Murakami A, Aggarwal BB. RANKL signaling and osteoclastogenesis is negatively regulated by cardamonin. PLoS One 2013; 8:e64118. [PMID: 23691159 PMCID: PMC3656934 DOI: 10.1371/journal.pone.0064118] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 04/09/2013] [Indexed: 01/02/2023] Open
Abstract
Bone loss/resorption or osteoporosis is a disease that is accelerated with aging and age-associated chronic diseases such as cancer. Bone loss has been linked with human multiple myeloma, breast cancer, and prostate cancer and is usually treated with bisphosphonates, and recently approved denosumab, an antibody against receptor activator of NF-κB ligand (RANKL). Because of the numerous side effects of the currently available drugs, the search continues for safe and effective therapies for bone loss. RANKL, a member of the TNF superfamily, has emerged as a major mediator of bone loss via activation of osteoclastogenesis. We have identified cardamonin, a chalcone isolated from Alpinia katsumadai Hayata that can affect osteoclastogenesis through modulation of RANKL. We found that treatment of monocytes with cardamonin suppressed RANKL-induced NF-κB activation and this suppression correlated with inhibition of IκBα kinase and of phosphorylation and degradation of IκBα, an inhibitor of NF-κB. Furthermore, cardamonin also downregulated RANKL-induced phosphorylation of MAPK including ERK and p38 MAPK. Cardamonin suppressed the RANKL-induced differentiation of monocytes to osteoclasts in a dose-dependent and time-dependent manner. We also found that an inhibitor of NF-κB essential modulator (NEMO) blocked RANKL-induced osteoclastogenesis, indicating a direct link with NF-κB. Finally, osteoclastogenesis induced by human breast cancer cells or human multiple myeloma cells were completely suppressed by cardamonin. Collectively, our results indicate that cardamonin suppresses osteoclastogenesis induced by RANKL and tumor cells by suppressing activation of the NF-κB and MAPK pathway.
Collapse
Affiliation(s)
- Bokyung Sung
- Cytokine Research Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Sahdeo Prasad
- Cytokine Research Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Vivek R. Yadav
- Cytokine Research Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Subash C. Gupta
- Cytokine Research Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Simone Reuter
- Cytokine Research Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Norio Yamamoto
- Food Science Research Center, House Wellness Foods Corporation, Itami, Japan
| | - Akira Murakami
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Bharat B. Aggarwal
- Cytokine Research Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
33
|
Ghantous A, Sinjab A, Herceg Z, Darwiche N. Parthenolide: from plant shoots to cancer roots. Drug Discov Today 2013; 18:894-905. [PMID: 23688583 DOI: 10.1016/j.drudis.2013.05.005] [Citation(s) in RCA: 211] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 05/07/2013] [Accepted: 05/09/2013] [Indexed: 02/07/2023]
Abstract
Parthenolide (PTL), a sesquiterpene lactone (SL) originally purified from the shoots of feverfew (Tanacetum parthenium), has shown potent anticancer and anti-inflammatory activities. It is currently being tested in cancer clinical trials. Structure-activity relationship (SAR) studies of parthenolide revealed key chemical properties required for biological activities and epigenetic mechanisms, and led to the derivatization of an orally bioavailable analog, dimethylamino-parthenolide (DMAPT). Parthenolide is the first small molecule found to be selective against cancer stem cells (CSC), which it achieves by targeting specific signaling pathways and killing cancer from its roots. In this review, we highlight the exciting journey of parthenolide, from plant shoots to cancer roots.
Collapse
Affiliation(s)
- Akram Ghantous
- International Agency for Research on Cancer, Lyon, France
| | | | | | | |
Collapse
|
34
|
Xu Y, Fang F, Miriyala S, Crooks PA, Oberley TD, Chaiswing L, Noel T, Holley AK, Zhao Y, Kiningham KK, Clair DKS, Clair WHS. KEAP1 is a redox sensitive target that arbitrates the opposing radiosensitive effects of parthenolide in normal and cancer cells. Cancer Res 2013; 73:4406-17. [PMID: 23674500 DOI: 10.1158/0008-5472.can-12-4297] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Elevated oxidative stress is observed more frequently in cancer cells than in normal cells. It is therefore expected that additional exposure to a low level of reactive oxygen species (ROS) will push cancer cells toward death, whereas normal cells might maintain redox homeostasis through adaptive antioxidant responses. We previously showed that parthenolide enhances ROS production in prostate cancer cells through activation of NADPH oxidase. The present study identifies KEAP1 as the downstream redox target that contributes to parthenolide's radiosensitization effect in prostate cancer cells. In vivo, parthenolide increases radiosensitivity of mouse xenograft tumors but protects normal prostate and bladder tissues against radiation-induced injury. Mechanistically, parthenolide increases the level of cellular ROS and causes oxidation of thioredoxin (TrX) in prostate cancer cells, leading to a TrX-dependent increase in a reduced state of KEAP1, which in turn leads to KEAP1-mediated PGAM5 and Bcl-xL (BCL2L1) degradation. In contrast, parthenolide increases oxidation of KEAP1 in normal prostate epithelial cells, leading to increased Nrf2 (NFE2L2) levels and subsequent Nrf2-dependent expression of antioxidant enzymes. These results reveal a novel redox-mediated modification of KEAP1 in controlling the differential effect of parthenolide on tumor and normal cell radiosensitivity. Furthermore, they show it is possible to develop a tumor-specific radiosensitizing agent with radioprotective properties in normal cells.
Collapse
Affiliation(s)
- Yong Xu
- Graduate Center for Toxicology, Department of Radiation Medicine, University of Kentucky, Lexington, Kentucky 40536, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Sesquiterpene lactones as drugs with multiple targets in cancer treatment: focus on parthenolide. Anticancer Drugs 2013; 23:883-96. [PMID: 22797176 DOI: 10.1097/cad.0b013e328356cad9] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Sesquiterpene lactones (SLs) constitute a large and diverse group of biologically active plant compounds that possess anti-inflammatory and antitumor activity. The subclass germacranolides is one of the major groups of SLs. It includes parthenolide, a highly cytotoxic SL that is being tested in clinical trials as an anti-cancer agent. In this review, we focus on SL antitumor activity related to cell-cycle arrest, differentiation, apoptosis induction through the intrinsic pathway, and sensitization of the extrinsic pathway. We also address the regression of tumors in response to cotreatment with conventional chemotherapeutics. We review the nuclear factor-κB-targeted anti-inflammatory activity in vitro and in vivo and relate it to the SL structural features involved in the molecular mechanisms. It is obvious that SLs are emerging as promising anticancer agents, but more investigations are required to fully understand the molecular mechanisms of known SLs in different cell death modalities and how these mechanisms contribute toward the potent antitumor and anti-inflammatory activities of SLs.
Collapse
|
36
|
Acuña UM, Shen Q, Ren Y, Lantvit DD, Wittwer JA, Kinghorn AD, Swanson SM, de Blanco EJC. Goyazensolide Induces Apoptosis in Cancer Cells in vitro and in vivo. INTERNATIONAL JOURNAL OF CANCER RESEARCH 2013; 9:36-53. [PMID: 25621077 PMCID: PMC4303185 DOI: 10.3923/ijcr.2013.36.53] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
As part of the screening program for anticancer agents from natural sources, the sesquiterpene lactone goyazensolide (GZL) was identified as a potent NF-κB inhibitor. The hollow-fiber assay was used to evaluate the anti-tumor efficacy of GZL in vivo. The mechanistic effects of GZL were evaluated in the HT-29 colonic cell line to reveal the pathway through which GZL exerts its effects. NF-κB subunits p65 and p50 were inhibited, and the upstream mediator IκB kinase (IKKβ) was downregulated in a dose-dependent manner. Apoptosis was mediated by caspase-3, and cell cycle arrest was detected in G1-phase. Consequently, 96% of the cell population was in sub G1-phase after treatment with GZL (10 μM).The antitumor effect of GZL was observed at a dose of 12.5 mg/kg. Cell adhesion was affected as a result of NF-κB inhibition. GZL appears to selectively target the transcription factor NF-κB. In summary, GZL sensitizes HT-29 colon cancer cells to apoptosis and cell death in a dose-dependent manner both in vivo and in vitro, through NF-κB inhibition (IC50 = 3.8 μM). Thus, it is a new potent lead compound for further development into a new effective chemotherapeutic agent.
Collapse
Affiliation(s)
- Ulyana Muñoz Acuña
- Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210
| | - Qi Shen
- Program for Collaborative Research in the Pharmaceutical Sciences and Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Yulin Ren
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210
| | - Daniel D. Lantvit
- Program for Collaborative Research in the Pharmaceutical Sciences and Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Jennifer A Wittwer
- Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210
| | - A. Douglas Kinghorn
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210
| | - Steven M. Swanson
- Program for Collaborative Research in the Pharmaceutical Sciences and Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Esperanza J. Carcache de Blanco
- Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
37
|
Li C, Guo S, Shi T. Role of NF-κB activation in matrix metalloproteinase 9, vascular endothelial growth factor and interleukin 8 expression and secretion in human breast cancer cells. Cell Biochem Funct 2012; 31:263-8. [PMID: 23086737 DOI: 10.1002/cbf.2899] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 08/25/2012] [Accepted: 08/30/2012] [Indexed: 11/09/2022]
Abstract
The aims of this study were to assess the effects and potential mechanisms of parthenolide on the expression of vascular endothelial growth factor (VEGF), interleukin 8 (IL-8) and matrix metalloproteinase 9 (MMP-9) in human breast cancer cell line MDA-MB-231. After incubation with different concentrations of parthenolide for 24 h, MDA-MB-231 cells were collected, and the expressions of VEGF, IL-8 and MMP-9 were measured by real-time PCR and Western blot. The secretions of VEGF, IL-8 and MMP-9 in culture supernatant of MDA-MB-231 cells were then measured with ELISA assays. The NF-κB DNA-binding activity of breast cancer cells treated with parthenolide was analyzed using electrophoretic mobility assays. The real-time PCR and Western blot data showed that the expressions of VEGF, IL-8 and MMP-9 were significantly inhibited by parthenolide at both transcription level and protein level in MDA-MB-231 cells. ELISA results also confirmed these effects at a secretion level. The electrophoretic mobility assay results demonstrated that parthenolide can inhibit NF-κB DNA-binding activity of the breast cancer cells. Hence, the expression of VEGF, IL-8 and MMP-9 may be suppressed by parthenolide through the inhibition of NF-κB DNA-binding activity in MDA-MB-231 cells.
Collapse
Affiliation(s)
- Caijuan Li
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | | | | |
Collapse
|
38
|
Zuch D, Giang AH, Shapovalov Y, Schwarz E, Rosier R, O'Keefe R, Eliseev RA. Targeting radioresistant osteosarcoma cells with parthenolide. J Cell Biochem 2012; 113:1282-91. [PMID: 22109788 DOI: 10.1002/jcb.24002] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Osteosarcoma is a devastating tumor of bone, primarily affecting adolescents. Osteosarcoma tumors are notoriously radioresistant. Radioresistant cancers, including osteosarcoma, typically exhibit a considerable potential for relapse and development of metastases following treatment. Relapse and metastatic potential can, in part, be due to a specific radioresistant subpopulation of cells with stem-like characteristics, cancer stem cells, which maintain the capacity to regenerate entire tumors. In the current study, we have investigated whether in vitro treatments with parthenolide, a naturally occurring small molecule that interferes with NF-κB signaling and has various other effects, will re-sensitize cancer stem cells and the entire cell population to radiotherapy in osteosarcoma. Our results indicate that parthenolide and ionizing radiation synergistically induce cell death in LM7 osteosarcoma cells. Importantly, the combination treatment results in a significant reduction in the viability of both the overall population of osteosarcoma cells and the cancer stem cell subpopulation. This effect is dependent on the ability of parthenolide to induce oxidative stress. Therefore, as a supplement to current multimodal therapy, parthenolide may sensitize osteosarcoma tumors to radiation and greatly reduce the prevalence of relapse and metastatic progression.
Collapse
Affiliation(s)
- Daniel Zuch
- Center for Musculoskeletal Research, University of Rochester School of Medicine & Dentistry, 575 Elmwood Ave., Rochester, New York 14642, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Mixed micelles of PEG2000-DSPE and vitamin-E TPGS for concurrent delivery of paclitaxel and parthenolide: Enhanced chemosenstization and antitumor efficacy against non-small cell lung cancer (NSCLC) cell lines. Eur J Pharm Sci 2012; 46:64-71. [DOI: 10.1016/j.ejps.2012.02.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Revised: 01/20/2012] [Accepted: 02/12/2012] [Indexed: 11/19/2022]
|
40
|
Tang QL, Xie XB, Wang J, Chen Q, Han AJ, Zou CY, Yin JQ, Liu DW, Liang Y, Zhao ZQ, Yong BC, Zhang RH, Feng QS, Deng WG, Zhu XF, Zhou BP, Zeng YX, Shen JN, Kang T. Glycogen synthase kinase-3β, NF-κB signaling, and tumorigenesis of human osteosarcoma. J Natl Cancer Inst 2012; 104:749-63. [PMID: 22534782 PMCID: PMC3352834 DOI: 10.1093/jnci/djs210] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Background Glycogen synthase kinase-3β (GSK-3β), a serine/threonine protein kinase, may function as a tumor suppressor or an oncogene, depending on the tumor type. We sought to determine the biological function of GSK-3β in osteosarcoma, a rare pediatric cancer for which the identification of new therapeutic targets is urgent. Methods We used cell viability assays, colony formation assays, and apoptosis assays to analyze the effects of altered GSK-3β expression in U2OS, MG63, SAOS2, U2OS/MTX300, and ZOS osteosarcoma cell lines. Nude mice (n = 5–8 mice per group) were injected with U2OS/MTX300, and ZOS cells to assess the role of GSK-3β in osteosarcoma growth in vivo and to evaluate the effects of inhibitors and/or anticancer drugs on tumor growth. We used an antibody array, polymerase chain reaction, western blotting, and a luciferase reporter assay to establish the effect of GSK-3β inhibition on the nuclear factor-κB (NF-κB) pathway. Immunochemistry was performed on primary tumor specimens from osteosarcoma patients (n = 74) to determine the relationship of GSK-3β activity with overall survival. Results Osteosarcoma cells with low levels of inactive p-Ser9-GSK-3β formed colonies in vitro and tumors in vivo more readily than cells with higher levels and cells in which GSK-3β had been silenced formed fewer colonies and smaller tumors than parental cells. Silencing or pharmacological inhibition of GSK-3β resulted in apoptosis of osteosarcoma cells. Inhibition of GSK-3β resulted in inhibition of the NF-κB pathway and reduction of NF-κB-mediated transcription. Combination treatments with GSK-3β inhibitors, NF-κB inhibitors, and chemotherapy drugs increased the effectiveness of chemotherapy drugs in vitro and in vivo. Patients whose osteosarcoma specimens had hyperactive GSK-3β, and nuclear NF-κB had a shorter median overall survival time (49.2 months) compared with patients whose tumors had inactive GSK-3β and NF-κB (109.2 months). Conclusion GSK-3β activity may promote osteosarcoma tumor growth, and therapeutic targeting of the GSK-3β and/or NF-κB pathways may be an effective way to enhance the therapeutic activity of anticancer drugs against osteosarcoma.
Collapse
Affiliation(s)
- Qing-Lian Tang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Janecka A, Wyrębska A, Gach K, Fichna J, Janecki T. Natural and synthetic α-methylenelactones and α-methylenelactams with anticancer potential. Drug Discov Today 2012; 17:561-72. [PMID: 22309965 DOI: 10.1016/j.drudis.2012.01.013] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 09/12/2011] [Accepted: 01/18/2012] [Indexed: 01/31/2023]
Abstract
α-Methylene-γ- and δ-lactones, as well as α-methylene-γ- and δ-lactams, are plant-derived compounds often used in traditional medicine for the treatment of inflammatory diseases. In recent years, the anticancer properties of these compounds and the molecular mechanisms of their action have been studied extensively. In the search for modern anticancer drugs, various synthetic analogs of α-methylene-γ- and δ-lactones and lactams have been synthesized and tested for their cytotoxic activity. In this review, we give a brief description of the occurrence and biological activity of such compounds isolated from plants and their diverse synthetic analogs.
Collapse
Affiliation(s)
- Anna Janecka
- Department of Biomolecular Chemistry, Medical University of Lodz, Poland.
| | | | | | | | | |
Collapse
|
42
|
López-Camarillo C, Ocampo EA, Casamichana ML, Pérez-Plasencia C, Álvarez-Sánchez E, Marchat LA. Protein kinases and transcription factors activation in response to UV-radiation of skin: implications for carcinogenesis. Int J Mol Sci 2011; 13:142-72. [PMID: 22312244 PMCID: PMC3269678 DOI: 10.3390/ijms13010142] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2011] [Revised: 12/14/2011] [Accepted: 12/16/2011] [Indexed: 12/18/2022] Open
Abstract
Solar ultraviolet (UV) radiation is an important environmental factor that leads to immune suppression, inflammation, photoaging, and skin carcinogenesis. Here, we reviewed the specific signal transduction pathways and transcription factors involved in the cellular response to UV-irradiation. Increasing experimental data supporting a role for p38, MAPK, JNK, ERK1/2, and ATM kinases in the response network to UV exposure is discussed. We also reviewed the participation of NF-κB, AP-1, and NRF2 transcription factors in the control of gene expression after UV-irradiation. In addition, we discussed the promising chemotherapeutic intervention of transcription factors signaling by natural compounds. Finally, we focused on the review of data emerging from the use of DNA microarray technology to determine changes in global gene expression in keratinocytes and melanocytes in response to UV treatment. Efforts to obtain a comprehensive portrait of the transcriptional events regulating photodamage of intact human epidermis after UV exposure reveals the existence of novel factors participating in UV-induced cell death. Progress in understanding the multitude of mechanisms induced by UV-irradiation could lead to the potential use of protein kinases and novel proteins as specific targets for the prevention and control of skin cancer.
Collapse
Affiliation(s)
- César López-Camarillo
- Genomics Sciences Program, Oncogenomics and Cancer Proteomics Laboratory, University Autonomous of Mexico City, Av. San Lorenzo 290, 03100, Mexico; E-Mails: (M.L.-C.); (E.Á.-S.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +55-5488-6661 ext. 15307
| | - Elena Aréchaga Ocampo
- Carcinogenesis Laboratory, National Institute of Cancerology, Av. Saint Fernando 22, 14080, México; E-Mail:
| | - Mavil López Casamichana
- Genomics Sciences Program, Oncogenomics and Cancer Proteomics Laboratory, University Autonomous of Mexico City, Av. San Lorenzo 290, 03100, Mexico; E-Mails: (M.L.-C.); (E.Á.-S.)
| | - Carlos Pérez-Plasencia
- Massive Sequencing Unit, National Institute of Cancerology, Av. Saint Fernando 22, 14080, México; E-Mail:
- Genomics Laboratory, FES-I, UBIMED, National Autonomous University of Mexico, Av. De los Barrios 1, 54090, México
| | - Elizbeth Álvarez-Sánchez
- Genomics Sciences Program, Oncogenomics and Cancer Proteomics Laboratory, University Autonomous of Mexico City, Av. San Lorenzo 290, 03100, Mexico; E-Mails: (M.L.-C.); (E.Á.-S.)
| | - Laurence A. Marchat
- Biotechnology Program, Institutional Program of Molecular Biomedicine, National School of Medicine and Homeopathy of the National Polytechnic Institute, Guillermo Massieu Helguera 239, 07320, Mexico; E-Mail:
| |
Collapse
|
43
|
Yadav VR, Prasad S, Reuter S, Sung B, Yamamoto N, Murakami A, Aggarwal BB. WITHDRAWN: Cardamonin Inhibits Osteoclastogenesis Induced by Tumor Cells Through Interruption of the Signaling Pathway Activated by Receptor Activator of NF-κB Ligand. Cancer Lett 2011:S0304-3835(11)00758-0. [PMID: 22182452 PMCID: PMC3769506 DOI: 10.1016/j.canlet.2011.12.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 11/04/2011] [Accepted: 12/07/2011] [Indexed: 11/18/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Vivek R Yadav
- Cytokine Research Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | | | | | | | | | | |
Collapse
|
44
|
Maeda Y, Tamura T, Fukutomi Y, Mukai T, Kai M, Makino M. A lipopeptide facilitate induction of Mycobacterium leprae killing in host cells. PLoS Negl Trop Dis 2011; 5:e1401. [PMID: 22132248 PMCID: PMC3222628 DOI: 10.1371/journal.pntd.0001401] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 10/06/2011] [Indexed: 12/24/2022] Open
Abstract
Little is known of the direct microbicidal activity of T cells in leprosy, so a lipopeptide consisting of the N-terminal 13 amino acids lipopeptide (LipoK) of a 33-kD lipoprotein of Mycobacterium leprae, was synthesized. LipoK activated M. leprae infected human dendritic cells (DCs) to induce the production of IL-12. These activated DCs stimulated autologous CD4+ or CD8+ T cells towards type 1 immune response by inducing interferon-gamma secretion. T cell proliferation was also evident from the CFSE labeling of target CD4+ or CD8+ T cells. The direct microbicidal activity of T cells in the control of M. leprae multiplication is not well understood. The present study showed significant production of granulysin, granzyme B and perforin from these activated CD4+ and CD8+ T cells when stimulated with LipoK activated, M. leprae infected DCs. Assessment of the viability of M. leprae in DCs indicated LipoK mediated T cell-dependent killing of M. leprae. Remarkably, granulysin as well as granzyme B could directly kill M. leprae in vitro. Our results provide evidence that LipoK could facilitate M. leprae killing through the production of effector molecules granulysin and granzyme B in T cells.
Collapse
Affiliation(s)
- Yumi Maeda
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
45
|
Steinmann P, Walters DK, Arlt MJE, Banke IJ, Ziegler U, Langsam B, Arbiser J, Muff R, Born W, Fuchs B. Antimetastatic activity of honokiol in osteosarcoma. Cancer 2011; 118:2117-27. [PMID: 21935912 DOI: 10.1002/cncr.26434] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Revised: 06/15/2011] [Accepted: 06/16/2011] [Indexed: 11/06/2022]
Abstract
BACKGROUND Metastasizing osteosarcoma has a mean 5-year survival rate of only 20% to 30%. Therefore, novel chemotherapeutics for more effective treatment of this disease are required. METHODS The antineoplastic activity of honokiol, which was demonstrated previously in numerous malignancies, was studied in vivo in C3H mice subcutaneously injected with syngeneic β-galactosidase bacterial gene (lacZ)-expressing LM8 osteosarcoma (LM8-lacZ) cells. In vitro cytotoxic effects of honokiol were investigated in 8 human and 2 murine osteosarcoma cell lines with different in vivo metastatic potential. RESULTS Seven days after subcutaneous flank injection of LM8-lacZ cells, daily intraperitoneal treatment of mice with 150 mg/kg honokiol reduced the number of micrometastases in the lung by 41% and reduced the number of macrometastases in the lung and liver by 69% and 80%, respectively, compared with control. Primary tumor growth was not inhibited. In osteosarcoma cell lines, honokiol inhibited the metabolic activity with a half-maximal concentration (IC(50) ) between 8.0 μg/mL and 16 μg/mL. Cyclosporin A partially reversed the inhibition of metabolic activity in LM8-lacZ cells. Cell proliferation and wound healing migration of LM8-lacZ cells were inhibited by honokiol with an IC(50) between 5.0 μg/mL and 10 μg/mL. Higher concentrations caused rapid cell death, which was distinct from necrosis, apoptosis, or autophagy but was associated with swelling of the endoplasmic reticulum, cytoplasmic vacuolation, and morphologically altered mitochondria. CONCLUSIONS Honokiol exhibited prominent antimetastatic activity in experimental osteosarcoma and caused rapid cell death in vitro that was unrelated to necrosis, apoptosis, or autophagy. The authors concluded that honokiol has considerable potential for the treatment of metastasizing osteosarcoma.
Collapse
Affiliation(s)
- Patrick Steinmann
- Laboratory for Orthopedic Research, Department of Orthopedics, University of Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Arlt MJE, Banke IJ, Walters DK, Puskas GJ, Steinmann P, Muff R, Born W, Fuchs B. LacZ transgene expression in the subcutaneous Dunn/LM8 osteosarcoma mouse model allows for the identification of micrometastasis. J Orthop Res 2011; 29:938-46. [PMID: 21284029 DOI: 10.1002/jor.21304] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Accepted: 10/18/2010] [Indexed: 02/04/2023]
Abstract
More effective treatment of patients with metastasizing osteosarcoma (OS) with a mean 5-year survival rate of <20% requires more detailed knowledge on the complex mechanisms of metastasis for the design of new drugs, which selectively target metastasizing cells. Moreover, novel diagnostic imaging technology for early detection of metastases is needed. Mouse models, which reproduce human metastasizing OS and allow visualization of single metastatic cells are instrumental for preclinical testing of new pharmaceuticals and diagnostic instruments. Here, the low metastatic Dunn cell line and its highly metastatic LM8 subline, both equipped with a constitutively expressed lacZ gene, were used to improve the well-established OS models in syngeneic C3H mice to achieve ex vivo visualization of single metastatic cells in affected organs by X-gal staining. These models, combined with a technique for in situ high quality lung tissue-maintaining perfusion revealed, as a novel finding, single metastasizing Dunn cells in lung and liver. Importantly, constitutive lacZ gene expression did not affect in vitro and in vivo tumorigenic and metastatic properties of Dunn and LM8 cells. Thus, these improved Dunn and LM8 OS mouse models will in the future serve as a benchmark for the development of new metastasis-targeting drugs and metastasis-imaging technology.
Collapse
Affiliation(s)
- Matthias J E Arlt
- Department of Orthopedics, Balgrist University Hospital, University of Zürich, Zürich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Sugiyasu K, Nanno K, Tamai N, Hashimoto N, Kishida Y, Yoshikawa H, Myoui A. Radio-sensitization of the murine osteosarcoma cell line LM8 with parthenolide, a natural inhibitor of NF-κB. Oncol Lett 2011; 2:407-412. [PMID: 22866095 DOI: 10.3892/ol.2011.277] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 02/24/2011] [Indexed: 11/06/2022] Open
Abstract
Nuclear factor (NF)-κB has been shown to be associated with cancer resistance to radiotherapy (RT), and is constitutively active in the murine osteosarcoma cell line, LM8. Parthenolide has been reported to show antitumor activity through inhibition of the NF-κB pathway. In this study, we investigated the radio-sensitizing activity of parthenolide. We established Luc-LM8, a stable transfectant reporter construct of NF-κB transcriptional activity into LM8. Luc-LM8 maintained the malignancy observed with LM8. In vitro, Luc-LM8 cells were cultured with or without parthenolide treatment, irradiated, and subjected to cell viability and apoptosis assays. In vivo, to investigate whether parthenolide enhances radio-sensitivity of tumors, a tumor growth assay was conducted. Parthenolide enhanced the growth inhibitory effect of RT and induced the apoptosis of Luc-LM8 cells with RT in vitro. The in vivo tumor growth was significantly suppressed in the mice treated with parthenolide and RT. The present study suggests that parthenolide sensitizes Luc-LM8 cells to irradiation. Thus, parthenolide is a potential candidate for use as a potent radio-sensitizing drug for use in cancer RT.
Collapse
Affiliation(s)
- Kenjiro Sugiyasu
- Department of Orthopaedics, Osaka University Graduate School of Medicine, Osaka University Hospital, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | |
Collapse
|
48
|
Sung B, Cho SG, Liu M, Aggarwal BB. Butein, a tetrahydroxychalcone, suppresses cancer-induced osteoclastogenesis through inhibition of receptor activator of nuclear factor-kappaB ligand signaling. Int J Cancer 2011; 129:2062-72. [PMID: 21170936 DOI: 10.1002/ijc.25868] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Revised: 12/06/2010] [Accepted: 12/08/2010] [Indexed: 01/02/2023]
Abstract
Osteoclastogenesis is associated with aging and various age-related inflammatory chronic diseases, including cancer. Receptor activator of nuclear factor-kappaB (NF-κB) ligand (RANKL), a member of the tumor necrosis factor superfamily, has been implicated as a major mediator of bone resorption, suggesting that agents that can suppress RANKL signaling might inhibit osteoclastogenesis, a process closely linked to bone resorption. We therefore investigated whether butein, a tetrahydroxychalcone, could inhibit RANKL signaling and suppress osteoclastogenesis induced by RANKL or tumor cells. We found that human multiple myeloma cells (MM.1S and U266), breast tumor cells (MDA-MB-231) and prostate tumor cells (PC-3) induced differentiation of macrophages to osteoclasts, as indicated by tartrate-resistant acid phosphatase (TRAP)-positive cells, and that butein suppressed this process. The chalcone also suppressed the expression of RANKL by the tumor cells. We further found that butein suppressed RANKL-induced NF-κB activation and that this suppression correlated with the inhibition of IκBα kinase and suppression of phosphorylation and degradation of IκBα, an inhibitor of NF-κB. Finally, butein also suppressed the RANKL-induced differentiation of macrophages to osteoclasts in a dose-dependent and time-dependent manner. Collectively, our results indicate that butein suppresses the osteoclastogenesis induced by tumor cells and by RANKL, by suppression of the NF-κB activation pathway.
Collapse
Affiliation(s)
- Bokyung Sung
- Department of Experimental Therapeutics, Cytokine Research Laboratory, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | |
Collapse
|
49
|
Ghanta KS, Pakala SB, Reddy SDN, Li DQ, Nair SS, Kumar R. MTA1 coregulation of transglutaminase 2 expression and function during inflammatory response. J Biol Chem 2011; 286:7132-8. [PMID: 21156794 PMCID: PMC3044970 DOI: 10.1074/jbc.m110.199273] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2010] [Revised: 12/05/2010] [Indexed: 12/26/2022] Open
Abstract
Although both metastatic tumor antigen 1 (MTA1), a master chromatin modifier, and transglutaminase 2 (TG2), a multifunctional enzyme, are known to be activated during inflammation, it remains unknown whether these molecules regulate inflammatory response in a coordinated manner. Here we investigated the role of MTA1 in the regulation of TG2 expression in bacterial lipopolysaccharide (LPS)-stimulated mammalian cells. While studying the impact of MTA1 status on global gene expression, we unexpectedly discovered that MTA1 depletion impairs the basal as well as the LPS-induced expression of TG2 in multiple experimental systems. We found that TG2 is a chromatin target of MTA1 and of NF-κB signaling in LPS-stimulated cells. In addition, LPS-mediated stimulation of TG2 expression is accompanied by the enhanced recruitment of MTA1, p65RelA, and RNA polymerase II to the NF-κB consensus sites in the TG2 promoter. Interestingly, both the recruitment of p65 and TG2 expression are effectively blocked by a pharmacological inhibitor of the NF-κB pathway. These findings reveal an obligatory coregulatory role of MTA1 in the regulation of TG2 expression and of the MTA1-TG2 pathway, at least in part, in LPS modulation of the NF-κB signaling in stimulated macrophages.
Collapse
Affiliation(s)
- Krishna Sumanth Ghanta
- From the Department of Biochemistry and Molecular Biology and Institute of Coregulator Biology, The George Washington University Medical Center, Washington, D. C. 20037
| | - Suresh B. Pakala
- From the Department of Biochemistry and Molecular Biology and Institute of Coregulator Biology, The George Washington University Medical Center, Washington, D. C. 20037
| | - Sirigiri Divijendra Natha Reddy
- From the Department of Biochemistry and Molecular Biology and Institute of Coregulator Biology, The George Washington University Medical Center, Washington, D. C. 20037
| | - Da-Qiang Li
- From the Department of Biochemistry and Molecular Biology and Institute of Coregulator Biology, The George Washington University Medical Center, Washington, D. C. 20037
| | - Sujit S. Nair
- From the Department of Biochemistry and Molecular Biology and Institute of Coregulator Biology, The George Washington University Medical Center, Washington, D. C. 20037
| | - Rakesh Kumar
- From the Department of Biochemistry and Molecular Biology and Institute of Coregulator Biology, The George Washington University Medical Center, Washington, D. C. 20037
| |
Collapse
|
50
|
Li W, Vogel CFA, Wu D, Matsumura F. Non-genomic action of TCDD to induce inflammatory responses in HepG2 human hepatoma cells and in liver of C57BL/6J mice. Biol Chem 2011; 391:1205-19. [PMID: 20707612 DOI: 10.1515/bc.2010.126] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
To assess the significance of the non-genomic signaling of TCDD (=dioxin) on liver of C57BL/6 mice and HepG2 human hepatoma cells, we first determined the group of markers that are susceptible to inhibition by parthenolide, a compound known to specifically suppress NF-κB-mediated inflammation. Of those, the most consistent marker turned out to be SOCS3 (a suppressor of cytokine signaling) known to respond to inflammation. An early diagnostic test on the action of TCDD on HepG2 cells in vitro within 3-6 h indicated that Cox-2 and SOCS3 are mainly induced via a non-genomic route, whereas PAI-2 appears to be induced through the classical action route. More detailed diagnostic tests at later stages of action of TCDD in HepG2 cells revealed that induction of IL-1β, BAFF, and iNOS are largely mediated by the protein kinase-dependent non-genomic route. An in vivo study on the 7 day action of TCDD on liver of AhR(NLS) mice showed that several early markers (e.g., Cox-2, MCP-1 and SOCS3) are induced, but not late markers such as IL-1β. Together, these results show that the non-genomic pathway contributes significantly to the early stress response reactions to TCDD that includes inflammation in hepatoma cells as well as in the liver.
Collapse
Affiliation(s)
- Wen Li
- Department of Environmental Toxicology and Center for Health and the Environment, University of California, One Shields Avenue, Davis, CA 95616, USA
| | | | | | | |
Collapse
|