1
|
Reyes Y, Larrey EK, Pathak R, Veisaga ML, Barbieri MA, Ward S, Kumar A, Sevilla MD, Adhikary A, Wnuk SF. Azido derivatives of sesquiterpene lactones: Synthesis, anticancer proliferation, and chemistry of nitrogen-centered radicals. RESULTS IN CHEMISTRY 2024; 9:101643. [PMID: 39498431 PMCID: PMC11533910 DOI: 10.1016/j.rechem.2024.101643] [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] [Indexed: 11/07/2024] Open
Abstract
Sesquiterpene lactones (SLs) such as parthenolide (PTL) and dehydroleucodine (DhL) selectively kill cancer cells without exerting normal tissue toxicity, potentially due to presence of α-methylene-γ-lactone (αMγL) fragment. We hypothesize that the addition of an azido group to the αMγL fragment of PTL or DhL further augments their anticancer properties as well as radiation sensitivity of cancer cells. Azido-SLs containing the azido group at the C14 methyl position of PTL (i.e., azido-melampomagnolide B, AzMMB) while preserving the mechanistically crucial exomethylene unit of αMγL fragment were also prepared. Sham-irradiated (i.e., unirradiated control) or irradiated human breast cancer cells (MCF7) were treated with different concentrations of azido-PTL (AzPTL) or azido-DhL (AzDhL) along with parental SLs. Proliferation rate of MCF7 cells were measured by MTT-assay, and their colony forming ability was determined by colony formation assay. Both AzPTL and AzDhL significantly suppress proliferation rate and colony forming ability of MCF-7 cells. AzPTL suppressed colony forming ability, not cellular proliferation, following irradiation to a greater extent than PTL at lower concentrations (5 and 10 μM). Electron spin resonance (ESR) studies were performed employing gamma-irradiated homogeneous supercooled aqueous solutions to investigate radical formation through addition of radiation-mediated prehydrated electrons to the azide group of AzPTL and AzDhL and to follow subsequent reactions of these radicals. In AzPTL, formation of a tertiary carbon-centered radical plausibly via a metastable aminyl radical was observed, whereas AzDhL produced both π-aminyl and α-azidoalkyl radicals. These radicals may contribute to the antitumor activities of AzPTL and AzDhL.
Collapse
Affiliation(s)
- Yahaira Reyes
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, USA
| | - Enoch K. Larrey
- Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas, 72205, USA
| | - Rupak Pathak
- Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas, 72205, USA
| | - Maria L. Veisaga
- Department of Biological Sciences, Florida International University, Miami, Florida 33199, USA
| | - Manuel A. Barbieri
- Department of Biological Sciences, Florida International University, Miami, Florida 33199, USA
| | - Samuel Ward
- Department of Chemistry, Oakland University, Rochester, Michigan 48309, USA
| | - Anil Kumar
- Department of Chemistry, Oakland University, Rochester, Michigan 48309, USA
| | - Michael D. Sevilla
- Department of Chemistry, Oakland University, Rochester, Michigan 48309, USA
| | - Amitava Adhikary
- Department of Chemistry, Oakland University, Rochester, Michigan 48309, USA
| | - Stanislaw F. Wnuk
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, USA
| |
Collapse
|
2
|
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
|
3
|
Parthenolide and Its Soluble Analogues: Multitasking Compounds with Antitumor Properties. Biomedicines 2022; 10:biomedicines10020514. [PMID: 35203723 PMCID: PMC8962426 DOI: 10.3390/biomedicines10020514] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/16/2022] [Accepted: 02/19/2022] [Indexed: 12/23/2022] Open
Abstract
Due to its chemical properties and multiple molecular effects on different tumor cell types, the sesquiterpene lactone parthenolide (PN) can be considered an effective drug with significant potential in cancer therapy. PN has been shown to induce either classic apoptosis or alternative caspase-independent forms of cell death in many tumor models. The therapeutical potential of PN has been increased by chemical design and synthesis of more soluble analogues including dimethylaminoparthenolide (DMAPT). This review focuses on the molecular mechanisms of both PN and analogues action in tumor models, highlighting their effects on gene expression, signal transduction and execution of different types of cell death. Recent findings indicate that these compounds not only inhibit prosurvival transcriptional factors such as NF-κB and STATs but can also determine the activation of specific death pathways, increasing intracellular reactive oxygen species (ROS) production and modifications of Bcl-2 family members. An intriguing property of these compounds is its specific targeting of cancer stem cells. The unusual actions of PN and its analogues make these agents good candidates for molecular targeted cancer therapy.
Collapse
|
4
|
Parthenolide as Cooperating Agent for Anti-Cancer Treatment of Various Malignancies. Pharmaceuticals (Basel) 2020; 13:ph13080194. [PMID: 32823992 PMCID: PMC7466132 DOI: 10.3390/ph13080194] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 12/18/2022] Open
Abstract
Primary and acquired resistance of cancer to therapy is often associated with activation of nuclear factor kappa B (NF-κB). Parthenolide (PN) has been shown to inhibit NF-κB signaling and other pro-survival signaling pathways, induce apoptosis and reduce a subpopulation of cancer stem-like cells in several cancers. Multimodal therapies that include PN or its derivatives seem to be promising approaches enhancing sensitivity of cancer cells to therapy and diminishing development of resistance. A number of studies have demonstrated that several drugs with various targets and mechanisms of action can cooperate with PN to eliminate cancer cells or inhibit their proliferation. This review summarizes the current state of knowledge on PN activity and its potential utility as complementary therapy against different cancers.
Collapse
|
5
|
Targets for improving tumor response to radiotherapy. Int Immunopharmacol 2019; 76:105847. [DOI: 10.1016/j.intimp.2019.105847] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/18/2019] [Accepted: 08/20/2019] [Indexed: 02/06/2023]
|
6
|
Morel KL, Ormsby RJ, Klebe S, Sweeney CJ, Sykes PJ. DMAPT is an Effective Radioprotector from Long-Term Radiation-Induced Damage to Normal Mouse Tissues In Vivo. Radiat Res 2019; 192:231-239. [PMID: 31095445 DOI: 10.1667/rr15404.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
While radiotherapy is widely used in cancer treatment, the benefits can be limited by radiation-induced damage to neighboring healthy tissues. We previously demonstrated in mice that the anti-inflammatory compound dimethylaminoparthenolide (DMAPT) selectively induces radiosensitivity in prostate tumor tissue from transgenic adenocarcinoma of mouse prostate (TRAMP) mice, while simultaneously protecting healthy tissues from 6 Gy whole-body radiation-induced apoptosis. Here, we examined the radioprotective effect of DMAPT on fibrosis in normal tissues after a partial-body fractionated radiation protocol that more closely mimics the image-guided fractionated radiotherapy protocols used clinically. Male C57BL/6J mice, 16 weeks old, received 20 Gy fractionated doses of X rays (2 Gy daily fractions, five days/week for two weeks) or sham irradiation to the lower abdomen, with or without a prior 20 mGy dose to mimic an image dose. In addition, mice received thrice weekly DMAPT (100 mg/kg by oral gavage) or vehicle control from 15 weeks of age until time of analysis at 6 weeks postirradiation. In the absence of exposure to radiation, there were no significant differences observed in the tissues of DMAPT and vehicle-treated mice (P > 0.05). DMAPT treatment significantly reduced radiation-induced testis weight loss by 60.9% (P < 0.0001), protected against a decrease in the seminiferous tubule diameter by 42.1% (P < 0.0001) and largely preserved testis morphology. Inclusion of the image dose had no significant effect on testis mass, seminiferous tubule diameter or testis morphology. DMAPT reduced radiation-induced fibrosis in the corpus cavernous region of the penis (98.1% reduction, P = 0.009) and in the muscle layer around the bladder (80.1% reduction, P = 0.0001). There was also a trend towards reduced collagen infiltration into the submucosal and muscle layers in the rectum. These results suggest that DMAPT could be useful in providing protection from the radiation-induced side effects of impotence and infertility, urinary incontinence and fecal urgency resulting from prostate cancer radiotherapy. DMAPT is a very well-tolerated drug and can conveniently be delivered orally without strict time windows relative to radiation exposure. Protection of normal tissues by DMAPT could potentially be useful in radiotherapy of other cancer types as well.
Collapse
Affiliation(s)
- Katherine L Morel
- a Flinders Centre for Innovation in Cancer, Flinders University and Medical Centre, Bedford Park, Adelaide, South Australia, Australia.,c Dana-Farber Cancer Institute, Harvard University, Boston, Massachusetts
| | - Rebecca J Ormsby
- a Flinders Centre for Innovation in Cancer, Flinders University and Medical Centre, Bedford Park, Adelaide, South Australia, Australia
| | - Sonja Klebe
- b Department of Anatomical Pathology, Flinders University and SA Pathology, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | | | - Pamela J Sykes
- a Flinders Centre for Innovation in Cancer, Flinders University and Medical Centre, Bedford Park, Adelaide, South Australia, Australia
| |
Collapse
|
7
|
Mortezaee K, Najafi M, Farhood B, Ahmadi A, Shabeeb D, Musa AE. NF‐κB targeting for overcoming tumor resistance and normal tissues toxicity. J Cell Physiol 2019; 234:17187-17204. [DOI: 10.1002/jcp.28504] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/22/2019] [Accepted: 03/05/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy School of Medicine, Kurdistan University of Medical Sciences Sanandaj Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department School of Paramedical Sciences, Kermanshah University of Medical Sciences Kermanshah Iran
| | - Bagher Farhood
- Departments of Medical Physics and Radiology Faculty of Paramedical Sciences, Kashan University of Medical Sciences Kashan Iran
| | - Amirhossein Ahmadi
- Pharmaceutical Sciences Research Center Faculty of Pharmacy, Mazandaran University of Medical Sciences Sari Iran
| | - Dheyauldeen Shabeeb
- Department of Physiology College of Medicine, University of Misan Misan Iraq
| | - Ahmed E. Musa
- Department of Medical Physics Tehran University of Medical Sciences (International Campus) Tehran Iran
| |
Collapse
|
8
|
Santos FTJ, Siqueira WN, Santos MLO, Silva HAMF, Sá JLF, Fernandes TS, Silva NH, França EJ, Silva EB, Melo AMMA. Radiosensitizer effect of usnic acid on Biomphalaria glabrata embryos. Int J Radiat Biol 2018; 94:838-843. [DOI: 10.1080/09553002.2018.1492757] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- F. T. J. Santos
- Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
- Departamento de Energia Nuclear, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - W. N. Siqueira
- Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
- Departamento de Energia Nuclear, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
- Serviço de Monitoração Ambiental, Centro Regional de Ciências Nucleares do Nordeste, Recife, Pernambuco, Brazil
| | - M. L. O. Santos
- Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
- Serviço de Monitoração Ambiental, Centro Regional de Ciências Nucleares do Nordeste, Recife, Pernambuco, Brazil
| | - H. A. M. F. Silva
- Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - J. L. F. Sá
- Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - T. S. Fernandes
- Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
- Departamento de Energia Nuclear, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - N. H. Silva
- Departamento de Bioquímica, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - E. J. França
- Serviço de Monitoração Ambiental, Centro Regional de Ciências Nucleares do Nordeste, Recife, Pernambuco, Brazil
| | - E. B. Silva
- Departamento de Energia Nuclear, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - A. M. M. A. Melo
- Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| |
Collapse
|
9
|
The Role of the Nuclear Factor κB Pathway in the Cellular Response to Low and High Linear Energy Transfer Radiation. Int J Mol Sci 2018; 19:ijms19082220. [PMID: 30061500 PMCID: PMC6121395 DOI: 10.3390/ijms19082220] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 07/24/2018] [Accepted: 07/24/2018] [Indexed: 12/19/2022] Open
Abstract
Astronauts are exposed to considerable doses of space radiation during long-term space missions. As complete shielding of the highly energetic particles is impracticable, the cellular response to space-relevant radiation qualities has to be understood in order to develop countermeasures and to reduce radiation risk uncertainties. The transcription factor Nuclear Factor κB (NF-κB) plays a fundamental role in the immune response and in the pathogenesis of many diseases. We have previously shown that heavy ions with a linear energy transfer (LET) of 100–300 keV/µm have a nine times higher potential to activate NF-κB compared to low-LET X-rays. Here, chemical inhibitor studies using human embryonic kidney cells (HEK) showed that the DNA damage sensor Ataxia telangiectasia mutated (ATM) and the proteasome were essential for NF-κB activation in response to X-rays and heavy ions. NF-κB’s role in cellular radiation response was determined by stable knock-down of the NF-κB subunit RelA. Transfection of a RelA short-hairpin RNA plasmid resulted in higher sensitivity towards X-rays, but not towards heavy ions. Reverse Transcriptase real-time quantitative PCR (RT-qPCR) showed that after exposure to X-rays and heavy ions, NF-κB predominantly upregulates genes involved in intercellular communication processes. This process is strictly NF-κB dependent as the response is completely absent in RelA knock-down cells. NF-κB’s role in the cellular radiation response depends on the radiation quality.
Collapse
|
10
|
Mendonca MS, Turchan WT, Alpuche ME, Watson CN, Estabrook NC, Chin-Sinex H, Shapiro JB, Imasuen-Williams IE, Rangel G, Gilley DP, Huda N, Crooks PA, Shapiro RH. DMAPT inhibits NF-κB activity and increases sensitivity of prostate cancer cells to X-rays in vitro and in tumor xenografts in vivo. Free Radic Biol Med 2017; 112:318-326. [PMID: 28782644 PMCID: PMC6322835 DOI: 10.1016/j.freeradbiomed.2017.08.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 07/20/2017] [Accepted: 08/01/2017] [Indexed: 01/22/2023]
Abstract
Constitutive activation of the pro-survival transcription factor NF-κB has been associated with resistance to both chemotherapy and radiation therapy in many human cancers, including prostate cancer. Our lab and others have demonstrated that the natural product parthenolide can inhibit NF-κB activity and sensitize PC-3 prostate cancers cells to X-rays in vitro; however, parthenolide has poor bioavailability in vivo and therefore has little clinical utility in this regard. We show here that treatment of PC-3 and DU145 human prostate cancer cells with dimethylaminoparthenolide (DMAPT), a parthenolide derivative with increased bioavailability, inhibits constitutive and radiation-induced NF-κB binding activity and slows prostate cancer cell growth. We also show that DMAPT increases single and fractionated X-ray-induced killing of prostate cancer cells through inhibition of DNA double strand break repair and also that DMAPT-induced radiosensitization is, at least partially, dependent upon the alteration of intracellular thiol reduction-oxidation chemistry. Finally, we demonstrate that the treatment of PC-3 prostate tumor xenografts with oral DMAPT in addition to radiation therapy significantly decreases tumor growth and results in significantly smaller tumor volumes compared to xenografts treated with either DMAPT or radiation therapy alone, suggesting that DMAPT might have a potential clinical role as a radiosensitizing agent in the treatment of prostate cancer.
Collapse
Affiliation(s)
- Marc S Mendonca
- Departments of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202 USA; Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA.
| | - William T Turchan
- Departments of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202 USA; Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Melanie E Alpuche
- Departments of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Christopher N Watson
- Departments of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202 USA; Richard L. Roudebush, VA Medical Center, Indianapolis, IN 46202 USA
| | - Neil C Estabrook
- Departments of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Helen Chin-Sinex
- Departments of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Jeremy B Shapiro
- Departments of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Imade E Imasuen-Williams
- Departments of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Gabriel Rangel
- Departments of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - David P Gilley
- Department of Chemistry and Applied Sciences, South Dakota School of Mines and Technology, Rapid City, SD 57701 USA
| | - Nazmul Huda
- Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Peter A Crooks
- College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Ronald H Shapiro
- Departments of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202 USA; Richard L. Roudebush, VA Medical Center, Indianapolis, IN 46202 USA
| |
Collapse
|
11
|
Börnigen D, Tyekucheva S, Wang X, Rider JR, Lee GS, Mucci LA, Sweeney C, Huttenhower C. Computational Reconstruction of NFκB Pathway Interaction Mechanisms during Prostate Cancer. PLoS Comput Biol 2016; 12:e1004820. [PMID: 27078000 PMCID: PMC4831844 DOI: 10.1371/journal.pcbi.1004820] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 02/19/2016] [Indexed: 12/21/2022] Open
Abstract
Molecular research in cancer is one of the largest areas of bioinformatic investigation, but it remains a challenge to understand biomolecular mechanisms in cancer-related pathways from high-throughput genomic data. This includes the Nuclear-factor-kappa-B (NFκB) pathway, which is central to the inflammatory response and cell proliferation in prostate cancer development and progression. Despite close scrutiny and a deep understanding of many of its members’ biomolecular activities, the current list of pathway members and a systems-level understanding of their interactions remains incomplete. Here, we provide the first steps toward computational reconstruction of interaction mechanisms of the NFκB pathway in prostate cancer. We identified novel roles for ATF3, CXCL2, DUSP5, JUNB, NEDD9, SELE, TRIB1, and ZFP36 in this pathway, in addition to new mechanistic interactions between these genes and 10 known NFκB pathway members. A newly predicted interaction between NEDD9 and ZFP36 in particular was validated by co-immunoprecipitation, as was NEDD9's potential biological role in prostate cancer cell growth regulation. We combined 651 gene expression datasets with 1.4M gene product interactions to predict the inclusion of 40 additional genes in the pathway. Molecular mechanisms of interaction among pathway members were inferred using recent advances in Bayesian data integration to simultaneously provide information specific to biological contexts and individual biomolecular activities, resulting in a total of 112 interactions in the fully reconstructed NFκB pathway: 13 (11%) previously known, 29 (26%) supported by existing literature, and 70 (63%) novel. This method is generalizable to other tissue types, cancers, and organisms, and this new information about the NFκB pathway will allow us to further understand prostate cancer and to develop more effective prevention and treatment strategies. In molecular research in cancer it remains challenging to uncover biomolecular mechanisms in cancer-related pathways from high-throughput genomic data, including the Nuclear-factor-kappa-B (NFκB) pathway. Despite close scrutiny and a deep understanding of many of the NFκB pathway members’ biomolecular activities, the current list of pathway members and a systems-level understanding of their interactions remains incomplete. In this study, we provide the first steps toward computational reconstruction of interaction mechanisms of the NFκB pathway in prostate cancer. We identified novel roles for 8 genes in this pathway and new mechanistic interactions between these genes and 10 known pathway members. We combined 651 gene expression datasets with 1.4M interactions to predict the inclusion of 40 additional genes in the pathway. Molecular mechanisms of interaction were inferred using recent advances in Bayesian data integration to simultaneously provide information specific to biological contexts and individual biomolecular activities, resulting in 112 interactions in the fully reconstructed NFκB pathway. This method is generalizable, and this new information about the NFκB pathway will allow us to further understand prostate cancer.
Collapse
Affiliation(s)
- Daniela Börnigen
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, United States of America.,The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Svitlana Tyekucheva
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Xiaodong Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jennifer R Rider
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, United States of America
| | - Gwo-Shu Lee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, United States of America
| | - Christopher Sweeney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, United States of America.,The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| |
Collapse
|
12
|
Gach K, Grądzka I, Wasyk I, Męczyńska-Wielgosz S, Iwaneńko T, Szymański J, Koszuk J, Janecki T, Kruszewski M, Janecka A. Anticancer activity and radiosensitization effect of methyleneisoxazolidin-5-ones in hepatocellular carcinoma HepG2 cells. Chem Biol Interact 2016; 248:68-73. [PMID: 26867810 DOI: 10.1016/j.cbi.2016.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 12/24/2015] [Accepted: 01/15/2016] [Indexed: 02/02/2023]
Abstract
Parthenolide (PTL), a well-known sesquiterpene lactone of natural origin with α,β-unsaturated carbonyl structure, has proven to show promising anti-cancer properties. In this report, anti-proliferative potential of two synthetic methyleneisoxazolidin-5-ones, MZ-6 and MZ-14, with the same structural motif, has been investigated in human hepatoma HepG2 cells. The effects on apoptosis induction and DNA damage were evaluated. All compounds decreased the number of live cells and increased the number of late apoptotic cells. However, only MZ-14 was able to induce DNA damage. Both synthetic compounds increased intracellular reactive oxygen species (ROS) generation and mitochondrial membrane potential changes at the same level as PTL. Additionally, cell survival was analyzed after a combined treatment, in which HepG2 cells were preincubated for 24 h with MZ-6, MZ-14 or PTL and irradiated with different doses of X-rays. The inhibition of cell survival was assessed by the clonogenic assay. We have shown that the clone formation was strongly inhibited by the combined treatment. The synergistic effect was observed for all three compounds but MZ-6 was significantly more effective. It is interesting to note that in HepG2 cells MZ-6 was the least cytotoxic of the tested compounds, did not induce DNA damage and was less active than the others in the clonogenic cell survival assay. It seems advantages from the point of view of the further in vivo studies that the compound with the lowest cytotoxic activity showed the strongest sensitizing effect.
Collapse
Affiliation(s)
- Katarzyna Gach
- Department of Biomolecular Chemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Iwona Grądzka
- Center for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Warsaw, Poland
| | - Iwona Wasyk
- Center for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Warsaw, Poland
| | - Sylwia Męczyńska-Wielgosz
- Center for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Warsaw, Poland
| | - Teresa Iwaneńko
- Center for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Warsaw, Poland
| | - Jacek Szymański
- Central Scientific Laboratory, Division of Public Health, Faculty of Health Sciences, Medical University of Lodz, Lodz, Poland
| | - Jacek Koszuk
- Institute of Organic Chemistry, Lodz University of Technology, Lodz, Poland
| | - Tomasz Janecki
- Institute of Organic Chemistry, Lodz University of Technology, Lodz, Poland
| | - Marcin Kruszewski
- Center for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Warsaw, Poland; Faculty of Medicine, University of Information Technology and Management, Rzeszów, Poland
| | - Anna Janecka
- Department of Biomolecular Chemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
13
|
Allen KT, Chin-Sinex H, DeLuca T, Pomerening JR, Sherer J, Watkins JB, Foley J, Jesseph JM, Mendonca MS. Dichloroacetate alters Warburg metabolism, inhibits cell growth, and increases the X-ray sensitivity of human A549 and H1299 NSC lung cancer cells. Free Radic Biol Med 2015; 89:263-73. [PMID: 26393423 DOI: 10.1016/j.freeradbiomed.2015.08.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 07/02/2015] [Accepted: 08/06/2015] [Indexed: 12/22/2022]
Abstract
We investigated whether altering Warburg metabolism (aerobic glycolysis) by treatment with the metabolic agent dichloroacetate (DCA) could increase the X-ray-induced cell killing of the radiation-resistant human non-small-cell lung cancer (NSCLC) cell lines A549 and H1299. Treatment with 50mM DCA decreased lactate production and glucose consumption in both A549 and H1299, clear indications of attenuated aerobic glycolysis. In addition, we found that DCA treatment also slowed cell growth, increased population-doubling time, and altered cell cycle distribution. Furthermore, we report that treatment with 50mM DCA significantly increased single and fractionated X-ray-induced cell killing of A549 and H1299 cells. Assay of DNA double-strand break repair by neutral comet assays demonstrated that DCA inhibited both the fast and the slow kinetics of X-ray-induced DSB repair in both A549 and H1299 NSCL cancer cells. Taken together the data suggest a correlation between an attenuated aerobic glycolysis and enhanced cytotoxicity and radiation-induced cell killing in radiation-resistant NSCLC cells.
Collapse
Affiliation(s)
- Kah Tan Allen
- Medical Sciences, Indiana University School of Medicine, Bloomington, IN 47405, USA
| | - Helen Chin-Sinex
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Thomas DeLuca
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | | | - Jeremy Sherer
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - John B Watkins
- Medical Sciences, Indiana University School of Medicine, Bloomington, IN 47405, USA
| | - John Foley
- Medical Sciences, Indiana University School of Medicine, Bloomington, IN 47405, USA; Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jerry M Jesseph
- Medical Sciences, Indiana University School of Medicine, Bloomington, IN 47405, USA
| | - Marc S Mendonca
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
14
|
Majdi M, Abdollahi MR, Maroufi A. Parthenolide accumulation and expression of genes related to parthenolide biosynthesis affected by exogenous application of methyl jasmonate and salicylic acid in Tanacetum parthenium. PLANT CELL REPORTS 2015; 34:1909-1918. [PMID: 26183953 DOI: 10.1007/s00299-015-1837-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 06/22/2015] [Accepted: 06/29/2015] [Indexed: 06/04/2023]
Abstract
Up-regulation of germacrene A synthase and down-regulation of parthenolide hydroxylase genes play key role in parthenolide accumulation of feverfew plants treated with methyl jasmonate and salicylic acid. Parthenolide is an important sesquiterpene lactone due to its anti-migraine and anti-cancer properties. Parthenolide amount was quantified by high-performance liquid chromatography after foliar application of methyl jasmonate (100 µM) or salicylic acid (1.0 mM) on feverfew leaves in time course experiment (3-96 h). Results indicate that exogenous application of methyl jasmonate or salicylic acid activated parthenolide biosynthesis. Parthenolide content reached its highest amount at 24 h after methyl jasmonate or salicylic acid treatments, which were 3.1- and 1.96-fold higher than control plants, respectively. Parthenolide transiently increased due to methyl jasmonate or salicylic acid treatments until 24 h, but did not show significant difference compared with control plants at 48 and 96 h time points in both treatments. Also, the transcript levels of early pathway (upstream) genes of terpene biosynthesis including 3-hydroxy-3-methylglutaryl-coenzyme A reductase, 1-deoxy-D-xylulose-5-phosphate reductoisomerase and hydroxy-2-methyl-2-(E)-butenyl 4-diphosphate reductase and the biosynthetic genes of parthenolide including germacrene A synthase, germacrene A oxidase, costunolide synthase and parthenolide synthase were increased by methyl jasmonate and salicylic acid treatments, but with different intensity. The transcriptional levels of these genes were higher in methyl jasmonate-treated plants than salicylic acid-treated plants. Parthenolide content measurements along with expression pattern analysis of the aforementioned genes and parthenolide hydroxylase as side branch gene of parthenolide suggest that the expression patterns of early pathway genes were not directly consistent with parthenolide accumulation pattern; hence, parthenolide accumulation is probably further modulated by the expression of its biosynthetic genes, especially germacrene A synthase and also its side branch gene, parthenolide hydroxylase.
Collapse
Affiliation(s)
- Mohammad Majdi
- Department of Agricultural Biotechnology, Faculty of Agriculture, University of Kurdistan, Sanandaj, Iran.
- Research Center for Medicinal Plant Breeding and Development, University of Kurdistan, Sanandaj, Iran.
| | - Mohammad Reza Abdollahi
- Department of Agronomy and Plant Breeding, Faculty of Agriculture, Bu-Ali Sina University, Hamedan, Iran
| | - Asad Maroufi
- Department of Agricultural Biotechnology, Faculty of Agriculture, University of Kurdistan, Sanandaj, Iran
| |
Collapse
|
15
|
Zhu HC, Qiu T, Dan C, Liu XH, Hu CH. Blockage of RelB expression by gene silencing enhances the radiosensitivity of androgen‑independent prostate cancer cells. Mol Med Rep 2014; 11:1167-73. [PMID: 25370388 DOI: 10.3892/mmr.2014.2857] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 08/29/2014] [Indexed: 11/05/2022] Open
Abstract
Levels of the nuclear factor‑kappa B (NF‑κB) alternative pathway member RelB have been shown to correlate with the effect of radiation therapy in prostate cancer. RelB expression was evaluated by immunohistochemistry in normal prostate, benign prostate hyperplasia and prostate cancer specimens. RM‑1 cells were pretreated with RelB siRNA prior to radiation therapy, and RelB expression in cytoplasmic and nuclear extracts was detected by real‑time polymerase chain reaction and western blot analysis. The apoptotic rates of experimental RM‑1 cell groups were assessed by flow cytometry. A clonogenic growth array was used to evaluate the radiosensitivity of RM‑1 cell groups. The NF‑κB family member RelB was expressed at a high level in prostate cancer specimens. Compared with irradiated control cells, RM‑1 cells transfected with RelB siRNA and treated with radiation therapy demonstrated a significant downregulation of RelB expression in the cytoplasm and nucleus. Notably, flow cytometry revealed that pretreatment of RM‑1 cells with RelB siRNA enhanced the apoptotic rate in response to radiation therapy compared with controls. Clonogenic growth assay results revealed enhanced radiosensitivity of RelB siRNA cells at various dosage points compared with control groups. Blockage of the alternative NF‑κB pathway via RelB silencing is a promising approach to enhance the radiosensitivity of prostate cancer.
Collapse
Affiliation(s)
- Heng-Cheng Zhu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Tao Qiu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Chao Dan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiu-Heng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Chun-Hai Hu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
16
|
Millimouno FM, Dong J, Yang L, Li J, Li X. Targeting apoptosis pathways in cancer and perspectives with natural compounds from mother nature. Cancer Prev Res (Phila) 2014; 7:1081-107. [PMID: 25161295 DOI: 10.1158/1940-6207.capr-14-0136] [Citation(s) in RCA: 180] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although the incidences are increasing day after day, scientists and researchers taken individually or by research group are trying to fight against cancer by several ways and also by different approaches and techniques. Sesquiterpenes, flavonoids, alkaloids, diterpenoids, and polyphenolic represent a large and diverse group of naturally occurring compounds found in a variety of fruits, vegetables, and medicinal plants with various anticancer properties. In this review, our aim is to give our perspective on the current status of the natural compounds belonging to these groups and discuss their natural sources, their anticancer activity, their molecular targets, and their mechanism of actions with specific emphasis on apoptosis pathways, which may help the further design and conduct of preclinical and clinical trials. Unlike pharmaceutical drugs, the selected natural compounds induce apoptosis by targeting multiple cellular signaling pathways including transcription factors, growth factors, tumor cell survival factors, inflammatory cytokines, protein kinases, and angiogenesis that are frequently deregulated in cancers and suggest that their simultaneous targeting by these compounds could result in efficacious and selective killing of cancer cells. This review suggests that they provide a novel opportunity for treatment of cancer, but clinical trials are still required to further validate them in cancer chemotherapy.
Collapse
Affiliation(s)
- Faya M Millimouno
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal University, Changchun, China. Dental Hospital, Jilin University, Changchun, China. Higher Institute of Science and Veterinary Medicine of Dalaba, Dalaba, Guinea
| | - Jia Dong
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| | - Liu Yang
- Dental Hospital, Jilin University, Changchun, China
| | - Jiang Li
- Dental Hospital, Jilin University, Changchun, China.
| | - Xiaomeng Li
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal University, Changchun, China.
| |
Collapse
|
17
|
Estabrook NC, Chin-Sinex H, Borgmann AJ, Dhaemers RM, Shapiro RH, Gilley D, Huda N, Crooks P, Sweeney C, Mendonca MS. Inhibition of NF-κB and DNA double-strand break repair by DMAPT sensitizes non-small-cell lung cancers to X-rays. Free Radic Biol Med 2011; 51:2249-58. [PMID: 22019440 DOI: 10.1016/j.freeradbiomed.2011.09.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Revised: 09/23/2011] [Accepted: 09/23/2011] [Indexed: 12/25/2022]
Abstract
We investigated the efficacy and mechanism of dimethylaminoparthenolide (DMAPT), an NF-κB inhibitor, to sensitize human lung cancer cells to X-ray killing in vitro and in vivo. We tested whether DMAPT increased the effectiveness of single and fractionated X-ray treatment through inhibition of NF-κB and/or DNA double-strand break (DSB) repair. Treatment with DMAPT decreased plating efficiency, inhibited constitutive and radiation-induced NF-κB binding activity, and enhanced radiation-induced cell killing by dose modification factors of 1.8 and 1.4 in vitro. X-ray fractionation demonstrated that DMAPT inhibited split-dose recovery/repair, and neutral DNA comet assays confirmed that DMAPT altered the fast and slow components of X-ray-induced DNA DSB repair. Knockdown of the NF-κB family member p65 by siRNA increased radiation sensitivity and completely inhibited split-dose recovery in a manner very similar to DMAPT treatment. The data suggest a link between inhibition of NF-κB and inhibition of DSB repair by DMAPT that leads to enhancement of X-ray-induced cell killing in vitro in non-small-cell lung cancer cells. Studies of A549 tumor xenografts in nude mice demonstrated that DMAPT enhanced X-ray-induced tumor growth delay in vivo.
Collapse
Affiliation(s)
- Neil C Estabrook
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Nambiar D, Rajamani P, Singh RP. Effects of phytochemicals on ionization radiation-mediated carcinogenesis and cancer therapy. Mutat Res 2011; 728:139-57. [PMID: 22030216 DOI: 10.1016/j.mrrev.2011.07.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 07/27/2011] [Accepted: 07/28/2011] [Indexed: 02/01/2023]
Abstract
Ionizing radiation (IR)-induced cellular damage is implicated in carcinogenesis as well as therapy of cancer. Advances in radiation therapy have led to the decrease in dosage and localizing the effects to the tumor; however, the development of radioresistance in cancer cells and radiation toxicity to normal tissues are still the major concerns. The development of radioresistance involves several mechanisms, including the activation of mitogenic and survival signaling, induction of DNA repair, and changes in redox signaling and epigenetic regulation. The current strategy of combining radiation with standard cytotoxic chemotherapeutic agents can potentially lead to unwanted side effects due to both agents. Thus agents are needed that could improve the efficacy of radiation killing of cancer cells and prevent the damage to normal cells and tissues caused by the direct and bystander effects of radiation, without have its own systemic toxicity. Chemopreventive phytochemicals, usually non-toxic agents with both cancer preventive and therapeutic activities, could rightly fit in this approach. In this regard, naturally occurring compounds, including curcumin, parthenolide, genistein, gossypol, ellagic acid, withaferin, plumbagin and resveratrol, have shown considerable potential. These agents suppress the radiation-induced activation of receptor tyrosine kinases and nuclear factor-κB signaling, can modify cell survival and DNA repair efficacy, and may potentiate ceramide signaling. These radiosensitizing and counter radioresistance mechanisms of phytochemicals in cancer cells are also associated with changes in epigenetic gene regulation. Because radioresistance involves multiple mechanisms, more studies are needed to discover novel phytochemicals having multiple mechanisms of radiosensitization and to overcome radioresistance of cancer cells. Pre-clinical studies are needed to address the appropriate dosage, timing, and duration of the application of phytochemicals with radiation to justify clinical trials. Nonetheless, some phytochemicals in combination with IR may play a significant role in enhancing the therapeutic index of cancer treatment.
Collapse
Affiliation(s)
- Dhanya Nambiar
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | | | | |
Collapse
|
19
|
Costunolide causes mitotic arrest and enhances radiosensitivity in human hepatocellular carcinoma cells. Radiat Oncol 2011; 6:56. [PMID: 21624128 PMCID: PMC3123574 DOI: 10.1186/1748-717x-6-56] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Accepted: 05/30/2011] [Indexed: 01/06/2023] Open
Abstract
PURPOSE This work aimed to investigate the effect of costunolide, a sesquiterpene lactone isolated from Michelia compressa, on cell cycle distribution and radiosensitivity of human hepatocellular carcinoma (HCC) cells. METHODS The assessment used in this study included: cell viability assay, cell cycle analysis by DNA histogram, expression of phosphorylated histone H3 (Ser 10) by flow cytometer, mitotic index by Liu's stain and morphological observation, mitotic spindle alignment by immunofluorescence of alpha-tubulin, expression of cell cycle-related proteins by Western blotting, and radiation survival by clonogenic assay. RESULTS Our results show that costunolide reduced the viability of HA22T/VGH cells. It caused a rapid G2/M arrest at 4 hours shown by DNA histogram. The increase in phosphorylated histone H3 (Ser 10)-positive cells and mitotic index indicates costunolide-treated cells are arrested at mitosis, not G2, phase. Immunofluorescence of alpha-tubulin for spindle formation further demonstrated these cells are halted at metaphase. Costunolide up-regulated the expression of phosphorylated Chk2 (Thr 68), phosphorylated Cdc25c (Ser 216), phosphorylated Cdk1 (Tyr 15) and cyclin B1 in HA22T/VGH cells. At optimal condition causing mitotic arrest, costunolide sensitized HA22T/VGH HCC cells to ionizing radiation with sensitizer enhancement ratio up to 1.9. CONCLUSIONS Costunolide could reduce the viability and arrest cell cycling at mitosis in hepatoma cells. Logical exploration of this mitosis-arresting activity for cancer therapeutics shows costunolide enhanced the killing effect of radiotherapy against human HCC cells.
Collapse
|
20
|
Shanmugam R, Kusumanchi P, Cheng L, Crooks P, Neelakantan S, Matthews W, Nakshatri H, Sweeney CJ. A water-soluble parthenolide analogue suppresses in vivo prostate cancer growth by targeting NFkappaB and generating reactive oxygen species. Prostate 2010; 70:1074-86. [PMID: 20209491 DOI: 10.1002/pros.21141] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND To characterize the molecular changes associated with DMAPT-induced prostate cancer cell death and its in vivo activity. METHODS CWR22Rv1 and PC-3 were subjected to flow cytometry, electrophoretic mobility shift assays, and Western blot studies to measure DMAPT's ability to generate reactive oxygen species (ROS), inhibit NFkappaB DNA binding, and cause changes in anti-apoptotic proteins. N-acetyl cysteine (NAC) and short hairpin RNA (shRNA) were used to determine the contribution of ROS and JNK2 activation, respectively. The BrdU incorporation assay was used to measure proliferation and trypan blue studies assessed cell viability after DMAPT treatment. The in vivo activity of DMAPT as a single agent and in combination with bicalutamide or docetaxel was assessed in a subcutaneous xenograft model with athymic nude female mice. RESULTS DMAPT generated ROS with subsequent JNK activation and inhibited NFkappaB DNA binding and expression of NFkappaB-regulated anti-apoptotic proteins. DMAPT increased necrotic and apoptotic cell death in a cell-type-dependent manner and both types of cell death were blocked by NAC. Additionally, shRNA JNK2 partially blocked the anti-proliferative activity of DMAPT. DMAPT inhibited CWR22Rv1 and PC-3 cellular proliferation by 100% with 10 and 20 microM respectively and in vivo, DMAPT was more effective at inhibiting growth than biclutamide (CWR22v1) and docetaxel (PC-3). CONCLUSIONS DMAPT promotes cell death by both generating ROS and inhibition of NFkappaB. Its in vivo activity supports the conduct of clinical trials in patients with castrate-resistant disease.
Collapse
|