1
|
Hsu CY, Rajabi S, Hamzeloo-Moghadam M, Kumar A, Maresca M, Ghildiyal P. Sesquiterpene lactones as emerging biomolecules to cease cancer by targeting apoptosis. Front Pharmacol 2024; 15:1371002. [PMID: 38529189 PMCID: PMC10961375 DOI: 10.3389/fphar.2024.1371002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 02/26/2024] [Indexed: 03/27/2024] Open
Abstract
Apoptosis is a programmed cell death comprising two signaling cascades including the intrinsic and extrinsic pathways. This process has been shown to be involved in the therapy response of different cancer types, making it an effective target for treating cancer. Cancer has been considered a challenging issue in global health. Cancer cells possess six biological characteristics during their developmental process known as cancer hallmarks. Hallmarks of cancer include continuous growth signals, unlimited proliferation, resistance to proliferation inhibitors, apoptosis escaping, active angiogenesis, and metastasis. Sesquiterpene lactones are one of the large and diverse groups of planet-derived phytochemicals that can be used as sources for a variety of drugs. Some sesquiterpene lactones possess many biological activities such as anti-inflammatory, anti-viral, anti-microbial, anti-malarial, anticancer, anti-diabetic, and analgesic. This review article briefly overviews the intrinsic and extrinsic pathways of apoptosis and the interactions between the modulators of both pathways. Also, the present review summarizes the potential effects of sesquiterpene lactones on different modulators of the intrinsic and extrinsic pathways of apoptosis in a variety of cancer cell lines and animal models. The main purpose of the present review is to give a clear picture of the current knowledge about the pro-apoptotic effects of sesquiterpene lactones on various cancers to provide future direction in cancer therapeutics.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Sadegh Rajabi
- Traditional Medicine and Materia Medica Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Hamzeloo-Moghadam
- Traditional Medicine and Materia Medica Research Center and Department of Traditional Pharmacy, School of Traditional Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abhinav Kumar
- Department of Nuclear and Renewable Energy, Ural Federal University Named After the First President of Russia Boris Yeltsin, Ekaterinburg, Russia
| | - Marc Maresca
- Aix Marseille Univ, CNRS, Centrale Marseille, iSm2, Marseille, France
| | - Pallavi Ghildiyal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| |
Collapse
|
2
|
Denda Y, Matsuo Y, Sugita S, Eguchi Y, Nonoyama K, Murase H, Kato T, Imafuji H, Saito K, Morimoto M, Ogawa R, Takahashi H, Mitsui A, Kimura M, Takiguchi S. The Natural Product Parthenolide Inhibits Both Angiogenesis and Invasiveness and Improves Gemcitabine Resistance by Suppressing Nuclear Factor κB Activation in Pancreatic Cancer Cell Lines. Nutrients 2024; 16:705. [PMID: 38474833 DOI: 10.3390/nu16050705] [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: 01/24/2024] [Revised: 02/18/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
We previously established pancreatic cancer (PaCa) cell lines resistant to gemcitabine and found that the activity of nuclear factor κB (NF-κB) was enhanced upon the acquisition of gemcitabine resistance. Parthenolide, the main active ingredient in feverfew, has been reported to exhibit antitumor activity by suppressing the NF-κB signaling pathway in several types of cancers. However, the antitumor effect of parthenolide on gemcitabine-resistant PaCa has not been elucidated. Here, we confirmed that parthenolide significantly inhibits the proliferation of both gemcitabine-resistant and normal PaCa cells at concentrations of 10 µM and higher, and that the NF-κB activity is significantly inhibited, even by 1 µM parthenolide. In Matrigel invasion assays and angiogenesis assays, the invasive and angiogenic potentials were higher in gemcitabine-resistant than normal PaCa cells and were inhibited by a low concentration of parthenolide. Furthermore, Western blotting showed suppressed MRP1 expression in gemcitabine-resistant PaCa treated with a low parthenolide concentration. In a colony formation assay, the addition of 1 µM parthenolide improved the sensitivity of gemcitabine-resistant PaCa cell lines to gemcitabine. These results suggest that parthenolide may be used as a novel therapeutic agent for the treatment of gemcitabine-resistant PaCa.
Collapse
Affiliation(s)
- Yuki Denda
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Yoichi Matsuo
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Saburo Sugita
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Yuki Eguchi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Keisuke Nonoyama
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Hiromichi Murase
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Tomokatsu Kato
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Hiroyuki Imafuji
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Kenta Saito
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Mamoru Morimoto
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Ryo Ogawa
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Hiroki Takahashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Akira Mitsui
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Masahiro Kimura
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| |
Collapse
|
3
|
LIU X, WANG X. Recent advances on the structural modification of parthenolide and its derivatives as anticancer agents. Chin J Nat Med 2022; 20:814-829. [DOI: 10.1016/s1875-5364(22)60238-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Indexed: 11/23/2022]
|
4
|
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
|
5
|
Karam L, Abou Staiteieh S, Chaaban R, Hayar B, Ismail B, Neipel F, Darwiche N, Abou Merhi R. Anticancer activities of parthenolide in primary effusion lymphoma preclinical models. Mol Carcinog 2021; 60:567-581. [PMID: 34101920 DOI: 10.1002/mc.23324] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 12/31/2022]
Abstract
The sesquiterpene lactone parthenolide is a major component of the feverfew medicinal plant, Tanacetum parthenium. Parthenolide has been extensively studied for its anti-inflammatory and anticancer properties in several tumor models. Parthenolide's antitumor activities depend on several mechanisms but it is mainly known as an inhibitor of the nuclear factor-κB (NF-κB) pathway. This pathway is constitutively activated and induces cell survival in primary effusion lymphoma (PEL), a rare aggressive AIDS-related lymphoproliferative disorder that is commonly caused by the human herpesvirus 8 (HHV-8) infection. The aim of this study is to evaluate the targeted effect of Parthenolide both in vitro and in vivo. Herein, parthenolide significantly inhibited cell growth, induced G0 /G1 cell cycle arrest, and induced massive apoptosis in PEL cells and ascites. In addition, parthenolide inhibited the NF-ĸB pathway suppressing IĸB phosphorylation and p65 nuclear translocation. It also reduced the expression of the DNA methylase inhibitor (DNMT1). Parthenolide induced HHV-8 lytic gene expression without inhibiting latent viral gene expression. Importantly, DMAPT, the more soluble parthenolide prodrug, promoted delay in ascites development and prolonged the survival of PEL xenograft mice. This study supports the therapeutic use of parthenolide in PEL and encourages its further clinical development.
Collapse
Affiliation(s)
- Louna Karam
- Faculty of Sciences, GSBT Laboratory, R. Hariri Campus, Lebanese University, Hadath, Lebanon.,Department of Natural Sciences, School of Arts & Sciences, Lebanese American University, Beirut, Lebanon
| | - Soumaiah Abou Staiteieh
- Faculty of Sciences, GSBT Laboratory, R. Hariri Campus, Lebanese University, Hadath, Lebanon
| | - Rady Chaaban
- Faculty of Sciences, GSBT Laboratory, R. Hariri Campus, Lebanese University, Hadath, Lebanon
| | - Berthe Hayar
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Bassel Ismail
- Faculty of Sciences, GSBT Laboratory, R. Hariri Campus, Lebanese University, Hadath, Lebanon
| | - Frank Neipel
- Virologisches Institut, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Nadine Darwiche
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Raghida Abou Merhi
- Faculty of Sciences, GSBT Laboratory, R. Hariri Campus, Lebanese University, Hadath, Lebanon
| |
Collapse
|
6
|
Talib WH, Alsalahat I, Daoud S, Abutayeh RF, Mahmod AI. Plant-Derived Natural Products in Cancer Research: Extraction, Mechanism of Action, and Drug Formulation. Molecules 2020; 25:E5319. [PMID: 33202681 PMCID: PMC7696819 DOI: 10.3390/molecules25225319] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer is one of the main causes of death globally and considered as a major challenge for the public health system. The high toxicity and the lack of selectivity of conventional anticancer therapies make the search for alternative treatments a priority. In this review, we describe the main plant-derived natural products used as anticancer agents. Natural sources, extraction methods, anticancer mechanisms, clinical studies, and pharmaceutical formulation are discussed in this review. Studies covered by this review should provide a solid foundation for researchers and physicians to enhance basic and clinical research on developing alternative anticancer therapies.
Collapse
Affiliation(s)
- Wamidh H. Talib
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan;
| | - Izzeddin Alsalahat
- Department of Pharmaceutical Chemistry and Pharmacognosy, Applied Science Private University, Amman 11931, Jordan; (I.A.); (S.D.); (R.F.A.)
| | - Safa Daoud
- Department of Pharmaceutical Chemistry and Pharmacognosy, Applied Science Private University, Amman 11931, Jordan; (I.A.); (S.D.); (R.F.A.)
| | - Reem Fawaz Abutayeh
- Department of Pharmaceutical Chemistry and Pharmacognosy, Applied Science Private University, Amman 11931, Jordan; (I.A.); (S.D.); (R.F.A.)
| | - Asma Ismail Mahmod
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan;
| |
Collapse
|
7
|
Hamarsheh S, Zeiser R. NLRP3 Inflammasome Activation in Cancer: A Double-Edged Sword. Front Immunol 2020; 11:1444. [PMID: 32733479 PMCID: PMC7360837 DOI: 10.3389/fimmu.2020.01444] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/03/2020] [Indexed: 12/21/2022] Open
Abstract
Inflammation is involved in tumor development and progression as well as antitumor response to therapy. In the past decade, the crosstalk between inflammation, immunity, and cancer has been investigated extensively, which led to the identification of several underlying mechanisms and cells involved. The formation of inflammasome complexes leads to the activation of caspase-1, production of interleukin (IL)-1β, and IL-18 and pyroptosis. Multiple studies have shown the involvement of NLRP3 inflammasome in tumorigenesis. Conversely, other reports have indicated a protective role in certain cancers. In this review, we summarize these contradictory roles of NLRP3 inflammasome in cancer, shed the light on oncogenic signaling leading to NLRP3 activation and IL-1β production and outline the current knowledge on therapeutic approaches.
Collapse
Affiliation(s)
- Shaima'a Hamarsheh
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Center Freiburg (CCCF), University of Freiburg, Freiburg, Germany.,Center for Biological Signalling Studies (BIOSS) and Center for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany
| |
Collapse
|
8
|
Applications of Sesquiterpene Lactones: A Review of Some Potential Success Cases. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10093001] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sesquiterpene lactones, a vast range of terpenoids isolated from Asteraceae species, exhibit a broad spectrum of biological effects and several of them are already commercially available, such as artemisinin. Here the most recent and impactful results of in vivo, preclinical and clinical studies involving a selection of ten sesquiterpene lactones (alantolactone, arglabin, costunolide, cynaropicrin, helenalin, inuviscolide, lactucin, parthenolide, thapsigargin and tomentosin) are presented and discussed, along with some of their derivatives. In the authors’ opinion, these compounds have been neglected compared to others, although they could be of great use in developing important new pharmaceutical products. The selected sesquiterpenes show promising anticancer and anti-inflammatory effects, acting on various targets. Moreover, they exhibit antifungal, anxiolytic, analgesic, and antitrypanosomal activities. Several studies discussed here clearly show the potential that some of them have in combination therapy, as sensitizing agents to facilitate and enhance the action of drugs in clinical use. The derivatives show greater pharmacological value since they have better pharmacokinetics, stability, potency, and/or selectivity. All these natural terpenoids and their derivatives exhibit properties that invite further research by the scientific community.
Collapse
|
9
|
Freund RRA, Gobrecht P, Fischer D, Arndt HD. Advances in chemistry and bioactivity of parthenolide. Nat Prod Rep 2020; 37:541-565. [DOI: 10.1039/c9np00049f] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
(−)-Parthenolide is a germacrane sesquiterpene lactone, available in ample amounts from the traditional medical plant feverfew (Tanacetum parthenium).
Collapse
Affiliation(s)
- Robert R. A. Freund
- Institut für Organische Chemie und Makromolekulare Chemie
- Friedrich-Schiller-Universität
- D-07743 Jena
- Germany
| | - Philipp Gobrecht
- Lehrstuhl für Zellphysiologie
- Ruhr-Universität Bochum
- D-44780 Bochum
- Germany
| | - Dietmar Fischer
- Lehrstuhl für Zellphysiologie
- Ruhr-Universität Bochum
- D-44780 Bochum
- Germany
| | - Hans-Dieter Arndt
- Institut für Organische Chemie und Makromolekulare Chemie
- Friedrich-Schiller-Universität
- D-07743 Jena
- Germany
| |
Collapse
|
10
|
Xu S, Li X, Liu Y, Xia Y, Chang R, Zhang C. Inflammasome inhibitors: promising therapeutic approaches against cancer. J Hematol Oncol 2019; 12:64. [PMID: 31242947 PMCID: PMC6595574 DOI: 10.1186/s13045-019-0755-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 06/14/2019] [Indexed: 12/21/2022] Open
Abstract
Inflammation has long been accepted as a key component of carcinogenesis. During inflammation, inflammasomes are potent contributors to the activation of inflammatory cytokines that lead to an inflammatory cascade. Considering the contributing role of inflammasomes in cancer progression, inflammasome inhibitors seem to have a promising future in cancer treatment and prevention. Here, we summarize the structures and signaling pathways of inflammasomes and detail some inflammasome inhibitors used to treat various forms of cancer, which we expect to be used in novel anticancer approaches. However, the practical application of inflammasome inhibitors is limited in regard to specific types of cancer, and the associated clinical trials have not yet been completed. Therefore, additional studies are required to explore more innovative and effective medicines for future clinical treatment of cancer.
Collapse
Affiliation(s)
- Shengchao Xu
- Department of Thoracic Surgery, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Xizhe Li
- Department of Thoracic Surgery, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Yuanqi Liu
- Department of Thoracic Surgery, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Yu Xia
- Department of Thoracic Surgery, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Ruimin Chang
- Department of Thoracic Surgery, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, People's Republic of China.
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|
11
|
Berdan CA, Ho R, Lehtola HS, To M, Hu X, Huffman TR, Petri Y, Altobelli CR, Demeulenaere SG, Olzmann JA, Maimone TJ, Nomura DK. Parthenolide Covalently Targets and Inhibits Focal Adhesion Kinase in Breast Cancer Cells. Cell Chem Biol 2019; 26:1027-1035.e22. [PMID: 31080076 DOI: 10.1016/j.chembiol.2019.03.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 02/14/2019] [Accepted: 03/27/2019] [Indexed: 02/07/2023]
Abstract
Parthenolide, a natural product from the feverfew plant and member of the large family of sesquiterpene lactones, exerts multiple biological and therapeutic activities including anti-inflammatory and anti-cancer effects. Here, we further study the parthenolide mechanism of action using activity-based protein profiling-based chemoproteomic platforms to map additional covalent targets engaged by parthenolide in human breast cancer cells. We find that parthenolide, as well as other related exocyclic methylene lactone-containing sesquiterpenes, covalently modify cysteine 427 of focal adhesion kinase 1 (FAK1), leading to impairment of FAK1-dependent signaling pathways and breast cancer cell proliferation, survival, and motility. These studies reveal a functional target exploited by members of a large family of anti-cancer natural products.
Collapse
Affiliation(s)
- Charles A Berdan
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Novartis-Berkeley Center for Proteomics and Chemistry Technologies, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Raymond Ho
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Haley S Lehtola
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Milton To
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Xirui Hu
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Tucker R Huffman
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Yana Petri
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA; Novartis-Berkeley Center for Proteomics and Chemistry Technologies, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Chad R Altobelli
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sasha G Demeulenaere
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA
| | - James A Olzmann
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Thomas J Maimone
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA; Novartis-Berkeley Center for Proteomics and Chemistry Technologies, University of California, Berkeley, Berkeley, CA 94720, USA.
| | - Daniel K Nomura
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Novartis-Berkeley Center for Proteomics and Chemistry Technologies, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
12
|
Wang J, Li M, Cui X, Lv D, Jin L, Khan M, Ma T. Brevilin A promotes oxidative stress and induces mitochondrial apoptosis in U87 glioblastoma cells. Onco Targets Ther 2018; 11:7031-7040. [PMID: 30410360 PMCID: PMC6198872 DOI: 10.2147/ott.s179730] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Sesquiterpene lactones are plant-derived, natural, bioactive molecules often used against inflammatory diseases in traditional Chinese medicines. Recently, sesquiterpene lactones have been reported to exhibit potent anticancer activity. In the present study, we have investigated the anticancer activity of Brevilin A, a sesquiterpene lactone component of Centipeda minima, against U87 glioblastoma cells. Materials and methods The cell proliferation was determined by MTT assay. Cell morphological changes were observed by phase-contrast microscopy. Flow cytometry was used to measure apoptosis. Glutathione (GSH), ROS generation, and mitochondrial membrane potential were measured using commercially available kits. The expression of proteins was measured by Western blotting analysis. Results Brevilin A inhibited the proliferation of, and induced severe morphological changes and apoptotic cell death in, U87 glioblastoma cells in a dose-dependent manner. Further mechanistic study revealed that Brevilin A induces oxidative stress, as evident from ROS generation, GSH depletion, and increased phosphorylation of stress-activated proteins p38 and JNK. Furthermore, Brevilin A bcl-xl/bak ratio, decreased mitochondrial membrane potential and induced cytochrome c release from mitochondria into cytosol in a dose-dependent manner. Finally, Brevilin A decreased the expression of Xiap and increased the expression of cleaved forms of caspase-9 and -3 and PARP in a dose-dependent manner. Conclusion Collective findings demonstrated that Brevilin A is a potent, anticancer, bioactive molecule and it effectively induces apoptosis in U87 glioblastoma cells, which is associated with induction of oxidative stress and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jie Wang
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China, .,Department of Neurology, The China-Japan Union Hospital of Jilin University, Changchun, China
| | - Miao Li
- Department of Neurology, The China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiaolin Cui
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China,
| | - Dongyue Lv
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China,
| | - Lingling Jin
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China,
| | - Muhammad Khan
- Department of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore, Pakistan
| | - Tonghui Ma
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China,
| |
Collapse
|
13
|
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
|
14
|
Szadvari I, Krizanova O, Babula P. Athymic nude mice as an experimental model for cancer treatment. Physiol Res 2017; 65:S441-S453. [PMID: 28006926 DOI: 10.33549/physiolres.933526] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Athymic nude mice, a murine strain bearing spontaneous deletion in the Foxn1 gene that causes deteriorated or absent thymus (which results in inhibited immune system with reduction of number of T cells), represent a widely used model in cancer research having long lasting history as a tool for preclinical testing of drugs. The review describes three models of athymic mice that utilize cancer cell lines to induce tumors. In addition, various methods that can be applied in order to evaluate activity of anticancer agents in these models are shown and discussed. Although each model has certain disadvantages, they are still considered as inevitable instruments in many fields of cancer research, particularly in finding new drugs that would more effectively combat the cancer disease or enhance the use of current chemotherapy. Finally, the review summarizes strengths and weaknesses as well as future perspectives of the athymic nude mice model in cancer research.
Collapse
Affiliation(s)
- I Szadvari
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| | | | | |
Collapse
|
15
|
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
|
16
|
Shi B, Wang LF, Meng WS, Chen L, Meng ZL. Carnosic acid and fisetin combination therapy enhances inhibition of lung cancer through apoptosis induction. Int J Oncol 2017; 50:2123-2135. [DOI: 10.3892/ijo.2017.3970] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 01/09/2017] [Indexed: 11/05/2022] Open
|
17
|
Seca AM, Silva AM, Pinto DC. Parthenolide and Parthenolide-Like Sesquiterpene Lactones as Multiple Targets Drugs. STUDIES IN NATURAL PRODUCTS CHEMISTRY 2017. [DOI: 10.1016/b978-0-444-63931-8.00009-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
18
|
Kim SL, Kim SH, Park YR, Liu YC, Kim EM, Jeong HJ, Kim YN, Seo SY, Kim IH, Lee SO, Lee ST, Kim SW. Combined Parthenolide and Balsalazide Have Enhanced Antitumor Efficacy Through Blockade of NF-κB Activation. Mol Cancer Res 2016; 15:141-151. [PMID: 28108625 DOI: 10.1158/1541-7786.mcr-16-0101] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 10/08/2016] [Accepted: 10/10/2016] [Indexed: 11/16/2022]
Abstract
Balsalazide is a colon-specific prodrug of 5-aminosalicylate that is associated with a reduced risk of colon cancer in patients with ulcerative colitis. Parthenolide, a strong NF-κB inhibitor, has recently been demonstrated to be a promising therapeutic agent, promoting apoptosis of cancer cells. In the current study, the antitumor effect of balsalazide combined with parthenolide in human colorectal cancer cells and colitis-associated colon cancers (CAC) was investigated. The results demonstrate that the combination of balsalazide and parthenolide markedly suppress proliferation, nuclear translocation of NF-κB, IκB-α phosphorylation, NF-κB DNA binding, and expression of NF-κB targets. Apoptosis via NF-κB signaling was confirmed by detecting expression of caspases, p53 and PARP. Moreover, treatment of a CAC murine model with parthenolide and balsalazide together resulted in significant recovery of body weight and improvement in histologic severity. Administration of parthenolide and balsalazide to CAC mice also suppressed carcinogenesis as demonstrated by uptake of 18F-fluoro-2-deoxy-D-glucose (FDG) using micro-PET/CT scans. These results demonstrate that parthenolide potentiates the efficacy of balsalazide through synergistic inhibition of NF-κB activation and the combination of dual agents prevents colon carcinogenesis from chronic inflammation. IMPLICATIONS This study represents the first evidence that combination therapy with balsalazide and parthenolide could be a new regimen for colorectal cancer treatment. Mol Cancer Res; 15(2); 141-51. ©2016 AACR.
Collapse
Affiliation(s)
- Se-Lim Kim
- Department of Internal Medicine, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Seong Hun Kim
- Department of Internal Medicine, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Young Ran Park
- Department of Internal Medicine, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Yu-Chuan Liu
- Department of Internal Medicine, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Eun-Mi Kim
- Department of Nuclear Medicine, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Hwan-Jeong Jeong
- Department of Nuclear Medicine, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Yo Na Kim
- Department of Pathology, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Seung Young Seo
- Department of Internal Medicine, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - In Hee Kim
- Department of Internal Medicine, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Seung Ok Lee
- Department of Internal Medicine, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Soo Teik Lee
- Department of Internal Medicine, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Sang-Wook Kim
- Department of Internal Medicine, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea.
| |
Collapse
|
19
|
Chen CH, Fabian C, Hursting S, deGraffenried LA. Breast Cancer Genetic and Molecular Subtype Impacts Response to Omega-3 Fatty Acid Ethyl Esters. Nutr Cancer 2016; 68:1021-33. [PMID: 27367296 DOI: 10.1080/01635581.2016.1192199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Epidemiological studies have correlated frequent omega-3 (n-3) fatty acid consumption with a lower risk for breast cancer; however, recent prospective studies have been less conclusive. Efforts in the preventive setting have focused on the use of n-3 fatty acids, and the pharmaceutical ethyl esters (EE) of these natural compounds, for high-risk patient populations. Limited understanding of specific mechanisms by which these agents function has hampered identification of the cancer subtype(s) that would gain the greatest therapeutic benefit. In this study, we investigated the in vitro effects of n-3 EEs in four distinct breast cancer subtypes and explored how they affect not only breast cancer cell survival but also modulate the nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB) and peroxisome proliferator-activated receptor gamma signaling pathways. Similar to the high variance in response observed in human studies, we found that the effectiveness of n-3 EEs depends on the molecular characteristics of the MCF-7, CAMA-1, MDA-MB-231, and SKBR3 breast cancer cell lines and is closely associated with the suppression of NF-κB. These data strongly suggest that the use of n-3 fatty acids and their pharmaceutical ether esters in the prevention and therapeutic setting should be guided by specific tumor characteristics.
Collapse
Affiliation(s)
- Ching Hui Chen
- a Department of Nutritional Sciences , The University of Texas at Austin , Texas , USA
| | - Carol Fabian
- b Department of Internal Medicine , University of Kansas Medical Center , Kansas City , Kansas , USA
| | - Stephen Hursting
- c Department of Nutrition , University of North Carolina at Chapel Hill , Chapel Hill , North Carolina , USA
| | - Linda A deGraffenried
- a Department of Nutritional Sciences , The University of Texas at Austin , Texas , USA
| |
Collapse
|
20
|
Tsai TY, Chan P, Gong CL, Wong KL, Su TH, Shen PC, Leung YM, Liu ZM. Parthenolide-Induced Cytotoxicity in H9c2 Cardiomyoblasts Involves Oxidative Stress. ACTA CARDIOLOGICA SINICA 2016; 31:33-41. [PMID: 27122844 DOI: 10.6515/acs20140422b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Cardiac cellular injury as a consequence of ischemia and reperfusion involves nuclear factor-κB (NF-κ B), amongst other factors, and NF-κ B inhibitors could substantially reduce myocardial infarct size. Parthenolide, a sesquiterpene lactone compound which could inhibit NF-κ B, has been shown to ameliorate myocardial reperfusion injury but may also produce toxic effects in cardiomyocytes at high concentrations. The aim of this study was to examine the cytotoxic effects of this drug on H9c2 cardiomyoblasts, which are precursor cells of cardiomyocytes. METHODS Cell viability and apoptosis were examined by MTT and TUNEL assay, respectively, and protein expression was analyzed by western blot. Reactive oxygen species (ROS) production was measured using DCFH-DA as dye. Cytosolic Ca(2+) concentration and mitochondrial membrane potential were measured microfluorimetrically using, respectively, fura 2 and rhodamine 123 as dyes. RESULTS Parthenolide caused apoptosis at 30 μ M, as judged by TUNEL assay and Bax and cytochrome c translocation. It also caused collapse of mitochondrial membrane potential and endoplasmic reticulum stress. Parthenolide triggered ROS formation, and vitamin C (antioxidant) partially alleviated parthenolide-induced cell death. CONCLUSIONS The results suggested that parthenolide at high concentrations caused cytotoxicity in cardiomyoblasts in part by inducing oxidative stress, and demonstrated the imperative for cautious and appropriate use of this agent in cardioprotection. KEY WORDS Cardiomyoblast; Endoplasmic reticulum stress; Oxidative stress; Parthenolide; Reperfusion injury.
Collapse
Affiliation(s)
- Tien-Yao Tsai
- Cardiovascular Division, Lotung Poh-Ai Hospital, Luodong; ; Department of Biomedical Engineering, Chung Yuan Christian University, Chungli
| | - Paul Chan
- Division of Cardiology, Department of Medicine, Taipei Medical University Wan Fan Hospital, Taipei
| | - Chi-Li Gong
- Department of Physiology, China Medical University
| | - Kar-Lok Wong
- Department of Anesthesiology, China Medical University Hospital, Taichung, Taiwan
| | - Tzu-Hui Su
- Department of Anesthesiology, China Medical University Hospital, Taichung, Taiwan
| | - Pei-Chen Shen
- Department of Cardiology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Yuk-Man Leung
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, Taichung, Taiwan
| | - Zhong-Min Liu
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University, Shanghai, China
| |
Collapse
|
21
|
Kim HY, Kim SL, Park YR, Liu YC, Seo SY, Kim SH, Kim IH, Lee SO, Lee ST, Kim SW. Balsalazide Potentiates Parthenolide-Mediated Inhibition of Nuclear Factor-κB Signaling in HCT116 Human Colorectal Cancer Cells. Intest Res 2015; 13:233-41. [PMID: 26130998 PMCID: PMC4479738 DOI: 10.5217/ir.2015.13.3.233] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 12/16/2014] [Accepted: 12/29/2014] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND/AIMS Balsalazide is an anti-inflammatory drug used in the treatment of inflammatory bowel disease. Balsalazide can reduce inflammatory responses via several mechanisms, including inhibition of nuclear factor-κB (NF-κB) activity. Parthenolide (PT) inhibits NF-κB and exerts promising anticancer effects by promoting apoptosis. The present investigated the antitumor effects of balsalazide, combined with PT, on NF-κB in a representative human colorectal carcinoma cell line, HCT116. METHODS We counted cells and conducted annexin-V assays and cell cycle analysis to measure apoptotic cell death. Western blotting was used investigate the levels of proteins involved in apoptosis. RESULTS PT and balsalazide produced synergistic anti-proliferative effects and induced apoptotic cell death. The combination of balsalazide and PT markedly suppressed nuclear translocation of the NF-κB p65 subunit and the phosphorylation of inhibitor of NF-κB. Moreover, PT and balsalazide dramatically enhanced NF-κB p65 phosphorylation. Apoptosis, through the mitochondrial pathway, was confirmed by detecting effects on Bcl-2 family members, cytochrome c release, and activation of caspase-3 and -8. CONCLUSIONS Combination treatment with PT and balsalazide may offer an effective strategy for the induction of apoptosis in HCT116 cells.
Collapse
Affiliation(s)
- Hyun-Young Kim
- Department of Internal Medicine, Medical School of Chonbuk National University, Jeonju, Korea
| | - Se-Lim Kim
- Department of Internal Medicine, Medical School of Chonbuk National University, Jeonju, Korea. ; Colon Carcinogenesis and Inflammation Laboratory, Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Young-Ran Park
- Department of Internal Medicine, Medical School of Chonbuk National University, Jeonju, Korea. ; Colon Carcinogenesis and Inflammation Laboratory, Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Yu-Chuan Liu
- Department of Internal Medicine, Medical School of Chonbuk National University, Jeonju, Korea. ; Colon Carcinogenesis and Inflammation Laboratory, Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Seung Young Seo
- Department of Internal Medicine, Medical School of Chonbuk National University, Jeonju, Korea
| | - Seong Hun Kim
- Department of Internal Medicine, Medical School of Chonbuk National University, Jeonju, Korea
| | - In Hee Kim
- Department of Internal Medicine, Medical School of Chonbuk National University, Jeonju, Korea
| | - Seung Ok Lee
- Department of Internal Medicine, Medical School of Chonbuk National University, Jeonju, Korea
| | - Soo Teik Lee
- Department of Internal Medicine, Medical School of Chonbuk National University, Jeonju, Korea. ; Colon Carcinogenesis and Inflammation Laboratory, Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Sang Wook Kim
- Department of Internal Medicine, Medical School of Chonbuk National University, Jeonju, Korea. ; Colon Carcinogenesis and Inflammation Laboratory, Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| |
Collapse
|
22
|
Induction of apoptosis by parthenolide in human oral cancer cell lines and tumor xenografts. Oral Oncol 2015; 51:602-9. [DOI: 10.1016/j.oraloncology.2015.03.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 02/26/2015] [Accepted: 03/10/2015] [Indexed: 11/20/2022]
|
23
|
Kim SL, Liu YC, Seo SY, Kim SH, Kim IH, Lee SO, Lee ST, Kim DG, Kim SW. Parthenolide induces apoptosis in colitis-associated colon cancer, inhibiting NF-κB signaling. Oncol Lett 2015; 9:2135-2142. [PMID: 26137027 DOI: 10.3892/ol.2015.3017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 01/13/2015] [Indexed: 12/15/2022] Open
Abstract
Recently, the nuclear factor (NF)-κB inhibitor parthenolide (PT) was identified as a promising anticancer agent for the promotion of cancer cell apoptosis. Additionally, our previous study demonstrated that PT administration suppresses tumor growth in a xenograft model of colorectal cancer cells via regulation of the B-cell lymphoma-2 (Bcl-2) family. However, the role of PT in the development of colitis-associated colon cancer (CAC) is poorly understood. Therefore, the aim of the present study was to investigate the effects of PT administration on CAC using a murine model. Azoxymethane (AOM) and dextran sulfate sodium (DSS) were administered to induce experimental CAC in the following three groups of treated mice: i) AOM and DSS plus vehicle; ii) AOM, DSS and 2 mg/kg PT; and iii) AOM, DSS and 4 mg/kg PT. It was demonstrated that the histological acuteness of AOM/DSS-induced CAC was significantly reduced following the administration of PT, resulting in decreased NF-κB p65 expression levels via a blockade of phosphorylation and subsequent degradation of inhibitor of κB-α (IκBα). Furthermore, PT administration appeared to enhance the process of carcinogenesis via the downregulation of the antiapoptotic proteins Bcl-2 and Bcl-extra large, mediated by inhibition of NF-κB activation. Apoptosis and caspase-3 expression were markedly increased in the PT-treated group. These findings indicate that PT inhibits IκBα phosphorylation and NF-κB activation, resulting in the initiation of apoptosis and the eventual suppression of CAC development. The beneficial effects of PT treatment observed in the experimental CAC model indicate the potential chemopreventive and therapeutic role of PT in CAC.
Collapse
Affiliation(s)
- Se Lim Kim
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea ; Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
| | - Yu Chuan Liu
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea ; Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
| | - Seung Young Seo
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea ; Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
| | - Seong Hun Kim
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea ; Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
| | - In Hee Kim
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea ; Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
| | - Seung Ok Lee
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea ; Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
| | - Soo Teik Lee
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea ; Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
| | - Dae-Ghon Kim
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea ; Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
| | - Sang Wook Kim
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea ; Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
| |
Collapse
|
24
|
Lu C, Wang W, Jia Y, Liu X, Tong Z, Li B. Inhibition of AMPK/autophagy potentiates parthenolide-induced apoptosis in human breast cancer cells. J Cell Biochem 2015; 115:1458-66. [PMID: 24619908 DOI: 10.1002/jcb.24808] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 03/10/2014] [Indexed: 12/25/2022]
Abstract
Parthenolide is the main bioactive component in feverfew, a common used herbal medicine, and has been extensively studied in relation to its anti-cancer properties. However there have been very few in-depth studies of the activities of this compound at the molecular level. Here, we showed that parthenolide increased reactive oxygen species (ROS), induced cell death, activated AMPK and autophagy, and led to M phase cell cycle arrest in breast cancer cells. Removal of ROS inhibited all parthenolide-associated events, such as cell death, AMPK activation, autophagy induction, and cell cycle arrest. Blockade of autophagy relieved cell cycle arrest, whereas inhibition of AMPK activity significantly repressed the induction of both autophagy and cell cycle arrest. These observations clearly showed that parthenolide-driven ROS activated AMPK-autophagy pathway. Furthermore, inhibition of either AMPK or autophagy significantly potentiated parthenolide-induced apoptosis. Therefore, our results show that parthenolide activates both apoptosis pathway and AMPK-autophagy survival pathway through the generation of ROS, and that suppression of AMPK or autophagy can potentially enhance the anti-cancer effect of parthenolide on breast cancer cells.
Collapse
Affiliation(s)
- Can Lu
- Laboratory of Cancer Cell Biology, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, P.R. China; Department of Breast Oncology, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, P.R. China
| | | | | | | | | | | |
Collapse
|
25
|
KIM SELIM, LIU YUCHUAN, PARK YOUNGRAN, SEO SEUNGYOUNG, KIM SEONGHUN, KIM INHEE, LEE SEUNGOK, LEE SOOTEIK, KIM DAEGHON, KIM SANGWOOK. Parthenolide enhances sensitivity of colorectal cancer cells to TRAIL by inducing death receptor 5 and promotes TRAIL-induced apoptosis. Int J Oncol 2014; 46:1121-30. [PMID: 25502339 DOI: 10.3892/ijo.2014.2795] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 11/26/2014] [Indexed: 11/05/2022] Open
|
26
|
KIM SELIM, LEE SOOTEIK, TRANG KIEUTHITHU, KIM SEONGHUN, KIM INHEE, LEE SEUNGOK, KIM DAEGHON, KIM SANGWOOK. Parthenolide exerts inhibitory effects on angiogenesis through the downregulation of VEGF/VEGFRs in colorectal cancer. Int J Mol Med 2014; 33:1261-7. [DOI: 10.3892/ijmm.2014.1669] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 02/19/2014] [Indexed: 11/06/2022] Open
|
27
|
Trang KTT, Kim SL, Park SB, Seo SY, Choi CH, Park JK, Moon JC, Lee ST, Kim SW. Parthenolide Sensitizes Human Colorectal Cancer Cells to Tumor Necrosis Factor-related Apoptosis-inducing Ligand through Mitochondrial and Caspase Dependent Pathway. Intest Res 2014; 12:34-41. [PMID: 25349561 PMCID: PMC4204686 DOI: 10.5217/ir.2014.12.1.34] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 10/22/2013] [Accepted: 10/22/2013] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND/AIMS Combination therapy utilizing tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) in conjunction with other anticancer agents, is a promising strategy to overcome TRAIL resistance in malignant cells. Recently, parthenolide (PT) has proved to be a promising anticancer agent, and several studies have explored its use in combination therapy. Here, we investigated the molecular mechanisms by which PT sensitizes colorectal cancer (CRC) cells to TRAIL-induced apoptosis. METHODS HT-29 cells (TRAIL-resistant) were treated with PT and/or TRAIL for 24 hours. The inhibitory effect on proliferation was detected using the 3-(4, 5-dimethylthiazol-2yl)-2, 5-diphenyltetrazolium bromide (MTT) assay. Annexin V staining, cell cycle analysis, and Hoechst 33258 staining were used to assess apoptotic cell death. Activation of an apoptotic pathway was confirmed by Western blot. RESULTS Treatment with TRAIL alone inhibited the proliferation of HCT 116 cells in a dose-dependent manner, whereas proliferation was not affected in HT-29 cells. Combination PT and TRAIL treatment significantly inhibited cell growth and induced apoptosis of HT-29 cells. We observed that the synergistic effect was associated with misregulation of B-cell lymphoma 2 (Bcl-2) family members, release of cytochrome C to the cytosol, activation of caspases, and increased levels of p53. CONCLUSION Combination therapy using PT and TRAIL might offer an effetive strategy to overcome TRAIL resistance in certain CRC cells.
Collapse
Affiliation(s)
- Kieu Thi Thu Trang
- Department of Internal Medicine, Biomedical Research Institute, Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju, Korea. ; Research Institute of Clinical Medicine, Biomedical Research Institute, Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju, Korea
| | - Se-Lim Kim
- Department of Internal Medicine, Biomedical Research Institute, Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju, Korea. ; Research Institute of Clinical Medicine, Biomedical Research Institute, Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju, Korea
| | - Sang-Bae Park
- Department of Internal Medicine, Biomedical Research Institute, Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju, Korea
| | - Seung-Young Seo
- Department of Internal Medicine, Biomedical Research Institute, Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju, Korea
| | - Chung-Hwan Choi
- Department of Internal Medicine, Biomedical Research Institute, Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju, Korea
| | - Jin-Kyoung Park
- Department of Internal Medicine, Biomedical Research Institute, Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju, Korea
| | - Jin-Chang Moon
- Department of Internal Medicine, Biomedical Research Institute, Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju, Korea
| | - Soo-Teik Lee
- Department of Internal Medicine, Biomedical Research Institute, Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju, Korea. ; Research Institute of Clinical Medicine, Biomedical Research Institute, Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju, Korea
| | - Sang-Wook Kim
- Department of Internal Medicine, Biomedical Research Institute, Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju, Korea. ; Research Institute of Clinical Medicine, Biomedical Research Institute, Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju, Korea
| |
Collapse
|
28
|
Phytoagents for cancer management: regulation of nucleic acid oxidation, ROS, and related mechanisms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:925804. [PMID: 24454991 PMCID: PMC3886269 DOI: 10.1155/2013/925804] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 09/27/2013] [Accepted: 10/05/2013] [Indexed: 12/28/2022]
Abstract
Accumulation of oxidized nucleic acids causes genomic instability leading to senescence, apoptosis, and tumorigenesis. Phytoagents are known to reduce the risk of cancer development; whether such effects are through regulating the extent of nucleic acid oxidation remains unclear. Here, we outlined the role of reactive oxygen species in nucleic acid oxidation as a driving force in cancer progression. The consequential relationship between genome instability and cancer progression highlights the importance of modulation of cellular redox level in cancer management. Current epidemiological and experimental evidence demonstrate the effects and modes of action of phytoagents in nucleic acid oxidation and provide rationales for the use of phytoagents as chemopreventive or therapeutic agents. Vitamins and various phytoagents antagonize carcinogen-triggered oxidative stress by scavenging free radicals and/or activating endogenous defence systems such as Nrf2-regulated antioxidant genes or pathways. Moreover, metal ion chelation by phytoagents helps to attenuate oxidative DNA damage caused by transition metal ions. Besides, the prooxidant effects of some phytoagents pose selective cytotoxicity on cancer cells and shed light on a new strategy of cancer therapy. The “double-edged sword” role of phytoagents as redox regulators in nucleic acid oxidation and their possible roles in cancer prevention or therapy are discussed in this review.
Collapse
|
29
|
D’ANNEO A, CARLISI D, EMANUELE S, BUTTITTA G, DI FIORE R, VENTO R, TESORIERE G, LAURICELLA M. Parthenolide induces superoxide anion production by stimulating EGF receptor in MDA-MB-231 breast cancer cells. Int J Oncol 2013; 43:1895-900. [DOI: 10.3892/ijo.2013.2137] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 09/10/2013] [Indexed: 11/06/2022] Open
|
30
|
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
|
31
|
Sun Y, Zang Z, Zhong L, Wu M, Su Q, Gao X, Zan W, Lin D, Zhao Y, Zhang Z. Identification of adiponectin receptor agonist utilizing a fluorescence polarization based high throughput assay. PLoS One 2013; 8:e63354. [PMID: 23691032 PMCID: PMC3653934 DOI: 10.1371/journal.pone.0063354] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 03/30/2013] [Indexed: 01/07/2023] Open
Abstract
Adiponectin, the adipose-derived hormone, plays an important role in the suppression of metabolic disorders that can result in type 2 diabetes, obesity, and atherosclerosis. It has been shown that up-regulation of adiponectin or adiponectin receptor has a number of therapeutic benefits. Given that it is hard to convert the full size adiponectin protein into a viable drug, adiponectin receptor agonists could be designed or identified using high-throughput screening. Here, we report on the development of a two-step screening process to identify adiponectin agonists. First step, we developed a high throughput screening assay based on fluorescence polarization to identify adiponectin ligands. The fluorescence polarization assay reported here could be adapted to screening against larger small molecular compound libraries. A natural product library containing 10,000 compounds was screened and 9 hits were selected for validation. These compounds have been taken for the second-step in vitro tests to confirm their agonistic activity. The most active adiponectin receptor 1 agonists are matairesinol, arctiin, (-)-arctigenin and gramine. The most active adiponectin receptor 2 agonists are parthenolide, taxifoliol, deoxyschizandrin, and syringin. These compounds may be useful drug candidates for hypoadiponectin related diseases.
Collapse
Affiliation(s)
- Yiyi Sun
- Chengdu Medical College, Pharmacy School, Chengdu, Sichuan Province, China
| | - Zhihe Zang
- Chengdu Medical College, Pharmacy School, Chengdu, Sichuan Province, China
| | - Ling Zhong
- Chengdu Medical College, Pharmacy School, Chengdu, Sichuan Province, China
| | - Min Wu
- Chengdu Medical College, Pharmacy School, Chengdu, Sichuan Province, China
| | - Qing Su
- Chengdu Medical College, Pharmacy School, Chengdu, Sichuan Province, China
| | - Xiurong Gao
- Chengdu Medical College, Pharmacy School, Chengdu, Sichuan Province, China
| | - Wang Zan
- Chengdu Medical College, Pharmacy School, Chengdu, Sichuan Province, China
| | - Dong Lin
- Chengdu Lang-Guan Technology Co., Ltd., Chengdu, Sichuan Province, China
| | - Yan Zhao
- Chengdu Medical College, Pharmacy School, Chengdu, Sichuan Province, China
| | - Zhonglin Zhang
- Chengdu Medical College, Pharmacy School, Chengdu, Sichuan Province, China
- * E-mail:
| |
Collapse
|
32
|
Kim SL, Kim SH, Trang KTT, Kim IH, Lee SO, Lee ST, Kim DG, Kang SB, Kim SW. Synergistic antitumor effect of 5-fluorouracil in combination with parthenolide in human colorectal cancer. Cancer Lett 2013; 335:479-86. [PMID: 23507557 DOI: 10.1016/j.canlet.2013.03.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 03/08/2013] [Accepted: 03/08/2013] [Indexed: 02/08/2023]
Abstract
Parthenolide (PT), a NF-κB inhibitor, has recently been demonstrated as a promising anticancer agent that promotes apoptosis of cancer cells. 5-fluorouracil (5-FU) has been a drug of choice for treatment of colorectal cancer (CRC). Unfortunately, many of the therapies that use 5-FU alone or in combination with other agents are likely to become ineffective due to drug resistance. In the present study, we investigated the antitumor effect of PT combined with 5-FU on a human CRC cell line, SW620. The results demonstrated that combination of PT and 5-FU induced apoptosis which was determined using MTT, cell cycle analysis, annexin-V assay, and Hoechst 33258 staining. Apoptosis through the mitochondrial pathway was confirmed by detecting regulation of Bcl-2 family members, cytochrome C release, and activation of caspase 3 and 9. Moreover, intra-peritoneal injection of PT and 5-FU showed significant inhibition of tumor growth in the xenograft model. These results demonstrate that PT exhibits anticancer activity in human colorectal cancer in vitro and in vivo. These findings provide an efficacious strategy to overcome 5-FU resistance in certain CRC.
Collapse
Affiliation(s)
- Se-Lim Kim
- Department of Internal Medicine of Chonbuk National University Hospital, Chonbuk National University, Jeonju 561-712, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
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
|