1
|
Hu H, Li F, Zhu F, Li J, Wang S, He Z, Chen J, Cheng L, Zhong F. Indole-3-carbinol ameliorates ovarian damage in female old mice through Nrf2/HO-1 pathway activation. Biochem Pharmacol 2024; 223:116193. [PMID: 38582268 DOI: 10.1016/j.bcp.2024.116193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 03/22/2024] [Accepted: 04/04/2024] [Indexed: 04/08/2024]
Abstract
Ovarian aging leads to infertility and birth defects. We aimed to clarify the role of Indole-3-carbinol (I3C) in resistance to oxidative stress, apoptosis, and fibrosis in ovarian aging. I3C was administered via intraperitoneal injection for 3 weeks in young or old mice. Immunohistochemistry; Masson, Sirius red, and TUNEL staining; follicle counting; estrous cycle analysis; and Western blotting were used for validating the protective effect of I3C against ovarian senescence. Human granulosa-like tumor cell line and primary granulosa cells were used for in vitro assay. The results indicated that I3C inhibited ovarian fibrosis and apoptosis while increasing the number of primordial follicles. Mechanistic studies have shown that I3C promoted the nuclear translocation of nuclear factor-erythroid 2-related factor (Nrf2) and upregulated the expression of heme oxygenase 1 (HO-1). Additionally, I3C increased cell viability and decreased lactate dehydrogenase, malondialdehyde, reactive oxygen species and JC-1 levels. Furthermore, the antioxidant effect of I3C was found to be dependent on the activation of Nrf2 and HO-1, as demonstrated by the disappearance of the effect upon inhibition of Nrf2 expression. In conclusion, I3C can alleviate the ovarian damage caused by aging and may be a protective agent to delay ovarian aging.
Collapse
Affiliation(s)
- Huiqing Hu
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Fangfang Li
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Fengyu Zhu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Jun Li
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Siyuan Wang
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Zhuoying He
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Jiaqi Chen
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Linghui Cheng
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China.
| | - Fei Zhong
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China.
| |
Collapse
|
2
|
Shen J, Liu Y, Wang X, Bai J, Lin L, Luo F, Zhong H. A Comprehensive Review of Health-Benefiting Components in Rapeseed Oil. Nutrients 2023; 15:999. [PMID: 36839357 PMCID: PMC9962526 DOI: 10.3390/nu15040999] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/04/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
Rapeseed oil is the third most consumed culinary oil in the world. It is well-known for its high content of unsaturated fatty acids, especially polyunsaturated fatty acids, which make it of great nutritional value. There is increasing evidence that a diet rich in unsaturated fatty acids offers health benefits. Although the consumption of rapeseed oil cuts across many areas around the world, the nutritional elements of rapeseed oil and the exact efficacy of the nutrients remain unclear. In this review, we systematically summarized the latest studies on functional rapeseed components to ascertain which component of canola oil contributes to its function. Apart from unsaturated fatty acids, there are nine functional components in rapeseed oil that contribute to its anti-microbial, anti-inflammatory, anti-obesity, anti-diabetic, anti-cancer, neuroprotective, and cardioprotective, among others. These nine functional components are vitamin E, flavonoids, squalene, carotenoids, glucoraphanin, indole-3-Carbinol, sterols, phospholipids, and ferulic acid, which themselves or their derivatives have health-benefiting properties. This review sheds light on the health-benefiting effects of rapeseed oil in the hope of further development of functional foods from rapeseed.
Collapse
Affiliation(s)
- Junjun Shen
- National Engineering Laboratory for Deep Processing of Rice and Byproducts, Central South University of Forestry and Technology, Changsha 410004, China
- Faculty of Bioscience and Biotechnology, Central South University of Forestry and Technology, Changsha 410004, China
- The Research and Development Department, Hunan Jinjian Cereals Industry, Changde 415001, China
| | - Yejia Liu
- The Research and Development Department, Hunan Jinjian Cereals Industry, Changde 415001, China
- Faculty of Life and Environmental Sciences, Hunan University of Arts and Science, Changde 415006, China
| | - Xiaoling Wang
- Faculty of Bioscience and Biotechnology, Central South University of Forestry and Technology, Changsha 410004, China
| | - Jie Bai
- National Engineering Laboratory for Deep Processing of Rice and Byproducts, Central South University of Forestry and Technology, Changsha 410004, China
| | - Lizhong Lin
- National Engineering Laboratory for Deep Processing of Rice and Byproducts, Central South University of Forestry and Technology, Changsha 410004, China
- The Research and Development Department, Hunan Jinjian Cereals Industry, Changde 415001, China
| | - Feijun Luo
- National Engineering Laboratory for Deep Processing of Rice and Byproducts, Central South University of Forestry and Technology, Changsha 410004, China
| | - Haiyan Zhong
- National Engineering Laboratory for Deep Processing of Rice and Byproducts, Central South University of Forestry and Technology, Changsha 410004, China
| |
Collapse
|
3
|
Targeting Drug Chemo-Resistance in Cancer Using Natural Products. Biomedicines 2021; 9:biomedicines9101353. [PMID: 34680470 PMCID: PMC8533186 DOI: 10.3390/biomedicines9101353] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer is one of the leading causes of death globally. The development of drug resistance is the main contributor to cancer-related mortality. Cancer cells exploit multiple mechanisms to reduce the therapeutic effects of anticancer drugs, thereby causing chemotherapy failure. Natural products are accessible, inexpensive, and less toxic sources of chemotherapeutic agents. Additionally, they have multiple mechanisms of action to inhibit various targets involved in the development of drug resistance. In this review, we have summarized the basic research and clinical applications of natural products as possible inhibitors for drug resistance in cancer. The molecular targets and the mechanisms of action of each natural product are also explained. Diverse drug resistance biomarkers were sensitive to natural products. P-glycoprotein and breast cancer resistance protein can be targeted by a large number of natural products. On the other hand, protein kinase C and topoisomerases were less sensitive to most of the studied natural products. The studies discussed in this review will provide a solid ground for scientists to explore the possible use of natural products in combination anticancer therapies to overcome drug resistance by targeting multiple drug resistance mechanisms.
Collapse
|
4
|
Okagu IU, Ndefo JC, Aham EC, Udenigwe CC. Zanthoxylum Species: A Comprehensive Review of Traditional Uses, Phytochemistry, Pharmacological and Nutraceutical Applications. Molecules 2021; 26:molecules26134023. [PMID: 34209371 PMCID: PMC8272177 DOI: 10.3390/molecules26134023] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/22/2021] [Accepted: 06/25/2021] [Indexed: 02/07/2023] Open
Abstract
Zanthoxylum species (Syn. Fagara species) of the Rutaceae family are widely used in many countries as food and in trado-medicinal practice due to their wide geographical distribution and medicinal properties. Peer reviewed journal articles and ethnobotanical records that reported the traditional knowledge, phytoconstituents, biological activities and toxicological profiles of Z. species with a focus on metabolic and neuronal health were reviewed. It was observed that many of the plant species are used as food ingredients and in treating inflammation, pain, hypertension and brain diseases. Over 500 compounds have been isolated from Z. species, and the biological activities of both the plant extracts and their phytoconstituents, including their mechanisms of action, are discussed. The phytochemicals responsible for the biological activities of some of the species are yet to be identified. Similarly, biological activities of some isolated compounds remain unknown. Taken together, the Z. species extracts and compounds possess promising biological activities and should be further explored as potential sources of new nutraceuticals and drugs.
Collapse
Affiliation(s)
- Innocent Uzochukwu Okagu
- Department of Biochemistry, University of Nigeria, Nsukka 410001, Enugu State, Nigeria; (I.U.O.); (E.C.A.)
| | - Joseph Chinedu Ndefo
- Department of Science Laboratory Technology, University of Nigeria, Nsukka 410001, Enugu State, Nigeria
- Correspondence: (J.C.N.); (C.C.U.)
| | - Emmanuel Chigozie Aham
- Department of Biochemistry, University of Nigeria, Nsukka 410001, Enugu State, Nigeria; (I.U.O.); (E.C.A.)
- Natural Science Unit, School of General Studies, University of Nigeria, Nsukka 410001, Enugu State, Nigeria
| | - Chibuike C. Udenigwe
- School of Nutrition Sciences, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Correspondence: (J.C.N.); (C.C.U.)
| |
Collapse
|
5
|
Issinger OG, Guerra B. Phytochemicals in cancer and their effect on the PI3K/AKT-mediated cellular signalling. Biomed Pharmacother 2021; 139:111650. [PMID: 33945911 DOI: 10.1016/j.biopha.2021.111650] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 02/07/2023] Open
Abstract
Protein kinases belong to the largest family of enzymes controlling every aspect of cellular activity including gene expression, cell division, differentiation and metabolism. They are part of major intracellular signalling pathways. Hence, it is not surprising that they are involved in the development of major diseases such as cardiovascular disorders, diabetes, dementia and, most importantly, cancer when they undergo mutations, modifications and unbalanced expression. This review will explore the possibility to draw a connection between the application of natural phytochemicals and the treatment of cancer. We have chosen to focus on the PI3K/AKT cellular signalling pathway which has been shown to be a major target by natural compounds in cell cultures and animal models.
Collapse
Affiliation(s)
- Olaf-Georg Issinger
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark.
| | - Barbara Guerra
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark.
| |
Collapse
|
6
|
Maniewska J, Jeżewska D. Non-Steroidal Anti-Inflammatory Drugs in Colorectal Cancer Chemoprevention. Cancers (Basel) 2021; 13:cancers13040594. [PMID: 33546238 PMCID: PMC7913298 DOI: 10.3390/cancers13040594] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/29/2021] [Accepted: 01/30/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary There is growing evidence from epidemiologic, preclinical and clinical studies suggesting that non-steroidal anti-inflammatory drugs (NSAIDs) play a beneficial role in colorectal cancer chemoprevention. They reduce the risk of colorectal polyps, mostly by cyclooxygenase-2 inhibition. The aim of our work was to describe the current state of scientific knowledge on the potential added value of the use of NSAIDs (such as aspirin, sulindac, and celecoxib) as chemopreventive agents in patients at risk of colorectal cancer. The study confirmed that there is a link between the long-term use of the NSAIDs and a decrease in the risk of colorectal cancer. Abstract Since colorectal cancer is one of the world’s most common cancers, studies on its prevention and early diagnosis are an emerging area of clinical oncology these days. For this study, a review of randomized controlled, double-blind clinical trials of selected NSAIDs (aspirin, sulindac and celecoxib) in chemoprevention of colorectal cancer was conducted. The main molecular anticancer activity of NSAIDs is thought to be a suppression of prostaglandin E2 synthesis via cyclooxygenase-2 inhibition, which causes a decrease in tumor cell proliferation, angiogenesis, and increases apoptosis. The lower incidence of colorectal cancer in the NSAID patients suggests the long-lasting chemopreventive effect of drugs studied. This new approach to therapy of colorectal cancer may transform the disease from a terminal to a chronic one that can be taken under control.
Collapse
|
7
|
Biersack B. 3,3'-Diindolylmethane and its derivatives: nature-inspired strategies tackling drug resistant tumors by regulation of signal transduction, transcription factors and microRNAs. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:867-878. [PMID: 35582221 PMCID: PMC8992569 DOI: 10.20517/cdr.2020.53] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/31/2020] [Accepted: 09/22/2020] [Indexed: 11/14/2022]
Abstract
Indoles of cruciferous vegetables are promising anti-tumor agents. Studies with indole-3-carbinol and its dimeric product, 3,3'-diindolylmethane (DIM), suggest that these compounds have the ability to deregulate multiple cellular signaling pathways that are essential for tumor growth and spread. These natural compounds are also effective modulators of transcription factors and non-coding RNAs. These effects explain their ability to inhibit tumor spread and to overcome drug resistance. In this work, pertinent literature on the effects of DIM and its synthetic derivatives on resistant tumors and resistance mechanisms in tumors is highlighted.
Collapse
Affiliation(s)
- Bernhard Biersack
- Organic Chemistry 1, University of Bayreuth, Bayreuth 95440, Germany
| |
Collapse
|
8
|
Hosny S, Sahyon H, Youssef M, Negm A. Oleanolic Acid Suppressed DMBA-Induced Liver Carcinogenesis through Induction of Mitochondrial-Mediated Apoptosis and Autophagy. Nutr Cancer 2020; 73:968-982. [PMID: 32519911 DOI: 10.1080/01635581.2020.1776887] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Phytochemicals appeared as a rich source of efficient and safe agents against many diseases like cancer. Various herbal sources are rich in oleanolic acid (OA). The scope of this study was to assess the biochemical and molecular mechanisms implicated in the ameliorative potency of OA against DMBA-induced liver carcinogenesis. Forty-eight male albino mice were assigned randomly to five groups (eight mice each) as follows: control healthy group, olive oil group, OA group, DMBA group, and DMBA with OA. Apoptosis, autophagy, inflammation, proliferation, and angiogenesis were investigated in the tissue samples. Histopathological examination was carried out as well as liver enzymes activity and other hepatic antioxidant and inflammatory biomarkers. The treatment with OA effectively suppressed the DMBA-initiated liver carcinogenesis via modulation of antioxidant status, induction of apoptosis and autophagy through modulating the expression of Caspase-3, Bcl-2 and Beclin-1, inhibiting angiogenesis (VEGF), proliferation (PCNA), and improved liver function and histological picture with a reduction in AFP level. Additionally, OA applies its antitumor effects by inhibition of proinflammatory transcription factor NF-κB and inflammatory markers (TNF-α and Cox-2) associated with DMBA administration. The present study shows that OA treatment efficiently suppressed the DMBA-initiated liver carcinogenesis through induction of mitochondrial-mediated apoptosis and autophagy and modulating inflammation.
Collapse
Affiliation(s)
- Samar Hosny
- Chemistry Department, Biochemistry Division, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Heba Sahyon
- Chemistry Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Magdy Youssef
- Chemistry Department, Biochemistry Division, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Amr Negm
- Chemistry Department, Biochemistry Division, Faculty of Science, Mansoura University, Mansoura, Egypt.,Department of Chemistry, College of Science, King Faisal University, Al-Ahasa, Saudi Arabia
| |
Collapse
|
9
|
Extraction and Quantification of Sulforaphane and Indole-3-Carbinol from Rapeseed Tissues Using QuEChERS Coupled with UHPLC-MS/MS. Molecules 2020; 25:molecules25092149. [PMID: 32375365 PMCID: PMC7248958 DOI: 10.3390/molecules25092149] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 01/17/2023] Open
Abstract
Rapeseed (Brassica napus L.) is rich in phenols, vitamins, carotenoids, and mineral elements, such as selenium. Additionally, it contains the active ingredients sulforaphane and indole-3-carbinol, which have been demonstrated to have pharmacological effects. In this study, sulforaphane and indole-3-carbinol were extracted and quantified from rapeseeds using quick, easy, cheap, effective, rugged and safe (QuEChERS) method coupled with ultra high performance liquid chromarography tandem mass spectrometry (UHPLC-MS/MS). The major parameters for extraction and purification efficiency were optimized, including the hydrolysis reaction, extraction condition and type and amount of purification adsorbents. The limit of detection (LOD) and the limit of quantification (LOQ) for sulforaphane were 0.05 μg/kg and 0.15 μg/kg, and for indole-3-carbinol were 5 μg/kg and 15 μg/kg, respectively. The developed method was used to successfully analyze fifty rapeseed samples. The QuEChERS coupled with UHPLC-MS/MS simultaneously detect sulforaphane and indole-3-carbinol in vegetable matrix and evaluate the quality and nutrition of rapeseed samples.
Collapse
|
10
|
Sauter ER. Cancer prevention and treatment using combination therapy with natural compounds. Expert Rev Clin Pharmacol 2020; 13:265-285. [PMID: 32154753 DOI: 10.1080/17512433.2020.1738218] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: Naturally occurring compounds play an essential role in the prevention and treatment of various cancers. There are more than 100 plant and animal based natural compounds currently in clinical use.Areas covered: 1) The importance of natural products combinations in the prevention and treatment of cancer, 2) the need to maximize efficacy while minimizing side effects when using natural product combinations, and 3) specifics related to plant and animal derived natural products, as well as agents derived from natural products. Therapies using natural compounds that have been investigated, their rationale, mechanism of action and findings are reviewed. When the data warrant it, combined interventions that appear to increase efficacy (compared with monotherapy) while minimizing toxicity have been highlighted. Pubmed was used to search for relevant publications.Expert opinion: Combination therapy with natural compounds has the potential to be more effective than single agent therapy. Similar to pharmacologic agents, the goal is to maximize efficacy while mimimizing potential side effects. There is an increasing research focus on the development of agents derived from natural products, with notable successes already achieved from the effort.
Collapse
Affiliation(s)
- Edward R Sauter
- Division of Cancer Prevention, National Cancer Institute, Rockville, MD, USA
| |
Collapse
|
11
|
Lee YR, Chen M, Lee JD, Zhang J, Lin SY, Fu TM, Chen H, Ishikawa T, Chiang SY, Katon J, Zhang Y, Shulga YV, Bester AC, Fung J, Monteleone E, Wan L, Shen C, Hsu CH, Papa A, Clohessy JG, Teruya-Feldstein J, Jain S, Wu H, Matesic L, Chen RH, Wei W, Pandolfi PP. Reactivation of PTEN tumor suppressor for cancer treatment through inhibition of a MYC-WWP1 inhibitory pathway. Science 2019; 364:364/6441/eaau0159. [PMID: 31097636 DOI: 10.1126/science.aau0159] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 10/30/2018] [Accepted: 03/27/2019] [Indexed: 12/18/2022]
Abstract
Activation of tumor suppressors for the treatment of human cancer has been a long sought, yet elusive, strategy. PTEN is a critical tumor suppressive phosphatase that is active in its dimer configuration at the plasma membrane. Polyubiquitination by the ubiquitin E3 ligase WWP1 (WW domain-containing ubiquitin E3 ligase 1) suppressed the dimerization, membrane recruitment, and function of PTEN. Either genetic ablation or pharmacological inhibition of WWP1 triggered PTEN reactivation and unleashed tumor suppressive activity. WWP1 appears to be a direct MYC (MYC proto-oncogene) target gene and was critical for MYC-driven tumorigenesis. We identified indole-3-carbinol, a compound found in cruciferous vegetables, as a natural and potent WWP1 inhibitor. Thus, our findings unravel a potential therapeutic strategy for cancer prevention and treatment through PTEN reactivation.
Collapse
Affiliation(s)
- Yu-Ru Lee
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ming Chen
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jonathan D Lee
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jinfang Zhang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Shu-Yu Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Tian-Min Fu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
| | - Hao Chen
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA 02115, USA.,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Tomoki Ishikawa
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Shang-Yin Chiang
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan.,Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 106, Taiwan
| | - Jesse Katon
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Yang Zhang
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Yulia V Shulga
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Assaf C Bester
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jacqueline Fung
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Emanuele Monteleone
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Molecular Biotechnology and Health Sciences, and GenoBiToUS, Genomics and Bioinformatics Service, University of Turin, Turin, Italy
| | - Lixin Wan
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.,Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Chen Shen
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
| | - Chih-Hung Hsu
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA 02115, USA.,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.,Department of Public Health, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Antonella Papa
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria 3800, Australia
| | - John G Clohessy
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Preclinical Murine Pharmacogenetics Facility and Mouse Hospital, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Julie Teruya-Feldstein
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Suresh Jain
- Intonation Research Laboratories, Hyderabad, India
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
| | - Lydia Matesic
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Ruey-Hwa Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan.,Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 106, Taiwan
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Pier Paolo Pandolfi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215, USA. .,Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
12
|
Yan T, Zeng Q, Wang L, Wang N, Cao H, Xu X, Chen X. Harnessing the Power of Optical Microscopic and Macroscopic Imaging for Natural Products as Cancer Therapeutics. Front Pharmacol 2019; 10:1438. [PMID: 31849680 PMCID: PMC6892944 DOI: 10.3389/fphar.2019.01438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/11/2019] [Indexed: 01/03/2023] Open
Abstract
Natural products (NPs) are an important source for new drug discovery over the past decades, which have been demonstrated to be effectively used in cancer prevention, treatment, and adjuvant therapy. Many methods, such as the genomic and metabolomic approaches, immunochemistry, mass spectrometry, and chromatography, have been used to study the effects of NPs on cancer as well as themselves. Because of the advantages in specificity, sensitivity, high throughput, and cost-effectiveness, optical imaging (OI) approaches, including optical microscopic imaging and macroscopic imaging techniques have also been applied in the studies of NPs. Optical microscopic imaging can observe NPs as cancer therapeutics at the cellular level and analyze its cytotoxicity and mechanism of action. Optical macroscopic imaging observes the distribution, metabolic pathway, and target lesions of NPs in vivo, and evaluates NPs as cancer therapeutics at the whole-body level in small living animals. This review focuses on the recent advances in NPs as cancer therapeutics, with particular emphasis on the powerful use of optical microscopic and macroscopic imaging techniques, including the studies of observation of ingestion by cells, anticancer mechanism, and in vivo delivery. Finally, we prospect the wider application and future potential of OI approaches in NPs as cancer therapeutics.
Collapse
Affiliation(s)
- Tianyu Yan
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education and School of Life Science and Technology, Xidian University, Xi’an, China
| | - Qi Zeng
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education and School of Life Science and Technology, Xidian University, Xi’an, China
| | - Lin Wang
- School of Information Sciences and Technology, Northwest University, Xi’an, China
| | - Nan Wang
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education and School of Life Science and Technology, Xidian University, Xi’an, China
| | - Honghao Cao
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education and School of Life Science and Technology, Xidian University, Xi’an, China
| | - Xinyi Xu
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education and School of Life Science and Technology, Xidian University, Xi’an, China
| | - Xueli Chen
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education and School of Life Science and Technology, Xidian University, Xi’an, China
| |
Collapse
|
13
|
Kunnumakkara AB, Bordoloi D, Sailo BL, Roy NK, Thakur KK, Banik K, Shakibaei M, Gupta SC, Aggarwal BB. Cancer drug development: The missing links. Exp Biol Med (Maywood) 2019; 244:663-689. [PMID: 30961357 PMCID: PMC6552400 DOI: 10.1177/1535370219839163] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
IMPACT STATEMENT The success rate for cancer drugs which enter into phase 1 clinical trials is utterly less. Why the vast majority of drugs fail is not understood but suggests that pre-clinical studies are not adequate for human diseases. In 1975, as per the Tufts Center for the Study of Drug Development, pharmaceutical industries expended 100 million dollars for research and development of the average FDA approved drug. By 2005, this figure had more than quadrupled, to $1.3 billion. In order to recover their high and risky investment cost, pharmaceutical companies charge more for their products. However, there exists no correlation between drug development cost and actual sale of the drug. This high drug development cost could be due to the reason that all patients might not respond to the drug. Hence, a given drug has to be tested in large number of patients to show drug benefits and obtain significant results.
Collapse
Affiliation(s)
- Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Devivasha Bordoloi
- Cancer Biology Laboratory, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Bethsebie Lalduhsaki Sailo
- Cancer Biology Laboratory, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Nand Kishor Roy
- Cancer Biology Laboratory, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Krishan Kumar Thakur
- Cancer Biology Laboratory, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Kishore Banik
- Cancer Biology Laboratory, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Mehdi Shakibaei
- Faculty of Medicine, Institute of Anatomy, Ludwig Maximilian University of Munich, Munich D-80336, Germany
| | - Subash C Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | | |
Collapse
|
14
|
Karimabad MN, Mahmoodi M, Jafarzadeh A, Darekordi A, Hajizadeh MR, Hassanshahi G. Molecular Targets, Anti-cancer Properties and Potency of Synthetic Indole-3-carbinol Derivatives. Mini Rev Med Chem 2019; 19:540-554. [DOI: 10.2174/1389557518666181116120145] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 04/25/2017] [Accepted: 04/26/2017] [Indexed: 12/14/2022]
Abstract
The indole-3-carbinol (I3C) displays anti-cancer/proliferative activities against human cancer cells. Cellular proliferation is an event associated with the progress and its continuation. This manifest is described by variation in expression and/or functions of genes that are related with cell cycle relevant proteins. The constitutive activation of several signal transduction pathways stimulates cells proliferation as well. The immediate stages in cancer development are accompanied by a fibrogenic response and the progression of the hypoxic environment is in favor of survival and proliferatory functions of cancer stem cells. A main part for prevention of in cancer cells death may manifest through altering cell metabolism. Cellular proliferation and metastasis are reported to be supported with increased generation of responsible hormones (in hormone dependent malignancies), and further promotion the angiogenesis, with epithelial to mesenchymal transition. This may be facilitated by progression of autophagy phenomenon, as well as via taking cues from neighboring stromal cells. Several signaling pathways in association with various factors specific for cellular viability, including hypoxia inducible factor 1, NF-κB, insulin-like growth factor 1 (IGF-1) receptor, Human foreskin fibroblasts (HFF-1), phosphoinositide 3 kinase/Akt, Wnt, cell cycle related protein, with androgen and estrogen receptor signaling are reported to be inhibited by I3C. These evidences, in association with bioinformatics data represent very important information for describing signaling pathways in parallel with molecular targets that may serve as markers for early diagnosis and/or critical targets for designing and development of novel therapeutic regimes alone or combined with drugs, to prevent tumor formation and further progression. In particular, I3C and DIM have been extensively investigated for their importance against numbers human cancers both in vitro and in vivo. We aimed the present manuscript, current study, to review anticancer properties and the miscellaneous mechanisms underlying the antitumorigenicity in an in-depth study for broadening the I3C treating marvel.
Collapse
Affiliation(s)
- Mojgan Noroozi Karimabad
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mehdi Mahmoodi
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Abdolah Jafarzadeh
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Ali Darekordi
- Department of Chemistry, Faculty of Science, Vali-e-Asr University of Rafsanjan, Rafsanjan, Iran
| | - Mohamad Reza Hajizadeh
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Gholamhossein Hassanshahi
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
15
|
Dankhoff K, Ahmad A, Weber B, Biersack B, Schobert R. Anticancer properties of a new non-oxido vanadium(IV) complex with a catechol-modified 3,3'-diindolylmethane ligand. J Inorg Biochem 2019; 194:1-6. [PMID: 30784705 DOI: 10.1016/j.jinorgbio.2019.02.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/05/2019] [Accepted: 02/08/2019] [Indexed: 01/16/2023]
Abstract
In order to identify new active drug candidates against cancer diseases we investigated the tumor cell growth inhibition, formation of reactive oxygen species, mitochondrial membrane damage, cell cycle arrest and DNA binding activity of a new bis(triethylammonium) tris[1,1-bis(indol-3-yl)-1-(3,4-catecholate)methane]vanadate(IV) complex. It exhibited significant antiproliferative activity against various cancer cell lines, showed a stronger DNA binding than cisplatin and led to mitochondrial damage, a formation of reactive oxygen species, and a cell cycle arrest in the G2/M phase of cancer cells.
Collapse
Affiliation(s)
- Katja Dankhoff
- Inorganic Chemistry IV, University of Bayreuth, Universitaetsstrasse 30, 95447 Bayreuth, Germany
| | - Aamir Ahmad
- USA Mitchell Cancer Institute, 1660 Springhill Avenue, Mobile, AL 36604-1405, USA
| | - Birgit Weber
- Inorganic Chemistry IV, University of Bayreuth, Universitaetsstrasse 30, 95447 Bayreuth, Germany
| | - Bernhard Biersack
- Organic Chemistry Laboratory, University of Bayreuth, Universitaetsstrasse 30, 95447 Bayreuth, Germany.
| | - Rainer Schobert
- Organic Chemistry Laboratory, University of Bayreuth, Universitaetsstrasse 30, 95447 Bayreuth, Germany
| |
Collapse
|
16
|
Dutta S, Moses JA, Anandharamakrishnan C. Encapsulation of Nutraceutical Ingredients in Liposomes and Their Potential for Cancer Treatment. Nutr Cancer 2019; 70:1184-1198. [DOI: 10.1080/01635581.2018.1557212] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Sayantani Dutta
- Computational Modeling and Nano Scale Processing Unit, Indian Institute of Food Processing Technology (IIFPT), Ministry of Food Processing Industries, Government of India, Thanjavur, Tamil Nadu, India
| | - Jeyan Arthur Moses
- Computational Modeling and Nano Scale Processing Unit, Indian Institute of Food Processing Technology (IIFPT), Ministry of Food Processing Industries, Government of India, Thanjavur, Tamil Nadu, India
| | - C. Anandharamakrishnan
- Computational Modeling and Nano Scale Processing Unit, Indian Institute of Food Processing Technology (IIFPT), Ministry of Food Processing Industries, Government of India, Thanjavur, Tamil Nadu, India
| |
Collapse
|
17
|
Ahmad A, Dandawate P, Schruefer S, Padhye S, Sarkar FH, Schobert R, Biersack B. Pentafluorophenyl Substitution of Natural Di(indol-3-yl)methane Strongly Enhances Growth Inhibition and Apoptosis Induction in Various Cancer Cell Lines. Chem Biodivers 2019; 16:e1900028. [PMID: 30715794 DOI: 10.1002/cbdv.201900028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 02/04/2019] [Indexed: 11/11/2022]
Abstract
Di(indol-3-yl)methane (=3,3'-methanediyldi(1H-indole), DIM, 1) is a known weakly antitumoral compound formed by digestion of indole-3-carbinol (=1H-indol-3-ylmethanol), an ingredient of various Brassica vegetables. Out of a series of nine fluoroaryl derivatives of 1, three pentafluorophenyl derivatives 2c, 2h, and 2i were identified that exhibited a two to five times greater anti-proliferative effect and an increased apoptosis induction when compared with 1 in the following carcinoma cell lines: BxPC-3 pancreas, LNCaP prostate, C4-2B prostate, PC3 prostate and the triple-negative MDA-MB-231 breast carcinoma. Compound 2h was particularly efficacious against androgen-refractory C4-2B prostate cancer cells (IC50 =6.4 μm) and 2i against androgen-responsive LNCaP cells (IC50 =6.2 μm). In addition, 2c and 2h exhibited distinct activity in three cancer cell lines resistant to 1.
Collapse
Affiliation(s)
- Aamir Ahmad
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, 36604, USA
| | - Prasad Dandawate
- Department of Cancer Biology, School of Medicine, KU Medical Center, Kansas City, Kansas, 66160, USA.,ISTRA, Abeda Inamdar Senior College, Pune, 411001, India
| | - Sebastian Schruefer
- Organic Chemistry Laboratory, University of Bayreuth, 95447, Bayreuth, Germany
| | - Subhash Padhye
- Department of Cancer Biology, School of Medicine, KU Medical Center, Kansas City, Kansas, 66160, USA.,ISTRA, Abeda Inamdar Senior College, Pune, 411001, India
| | - Fazlul H Sarkar
- Retired as Distinguished Professor, Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Rainer Schobert
- Organic Chemistry Laboratory, University of Bayreuth, 95447, Bayreuth, Germany
| | - Bernhard Biersack
- Organic Chemistry Laboratory, University of Bayreuth, 95447, Bayreuth, Germany
| |
Collapse
|
18
|
Subramanian C, White PT, Kuai R, Kalidindi A, Castle VP, Moon JJ, Timmermann BN, Schwendeman A, Cohen MS. Synthetic high-density lipoprotein nanoconjugate targets neuroblastoma stem cells, blocking migration and self-renewal. Surgery 2018; 164:S0039-6060(18)30080-1. [PMID: 29753460 PMCID: PMC6814450 DOI: 10.1016/j.surg.2018.01.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 01/13/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND Pathways critical for neuroblastoma cancer stem cell function are targeted by 4,19,27-triacetyl withalongolide A (WGA-TA). Because neuroblastoma cells and their cancer stem cells highly overexpress the scavenger receptor class B type 1 receptor that binds to synthetic high-density lipoprotein, we hypothesized that a novel mimetic synthetic high-density lipoprotein nanoparticle would be an ideal carrier for the delivery of 4,19,27-triacetyl withalongolide to neuroblastoma and neuroblastoma cancer stem cells. METHODS Expression of scavenger receptor class B type 1 in validated human neuroblastoma cells was evaluated by quantitative polymerase chain reaction (qPCR) and Western blot. In vitro cellular uptake of synthetic high-density lipoprotein nanoparticles was observed with a fluorescence microscope. In vivo biodistribution of synthetic high-density lipoprotein nanoparticles was investigated with IVIS imaging. Self-renewal and migration/invasion were assessed by sphere formation and Boyden chamber assays, respectively. Viability was analyzed by CellTiter-Glo assay. Cancer stem cell markers were evaluated by flow cytometry. RESULTS qPCR and Western blot analysis revealed a higher level of scavenger receptor class B type 1 expression and drug uptake in N-myc amplified neuroblastoma cells. In vitro uptake of synthetic high-density lipoprotein was almost completely blocked by excess synthetic high-density lipoprotein. The synthetic high-density lipoprotein nanoparticles mainly accumulated in the tumor and liver, but not in other organs. Synthetic HDL-4,19,27-triacetyl withalongolide showed a 1,000-fold higher potency than the carrier (synthetic high-density lipoprotein) alone (P < .01) to kill neuroblastoma cells. Additionally, a dose-dependent decrease in sphere formation, invasion, migration, and cancer stem cell markers was observed after treatment of neuroblastoma cells with synthetic high-density lipoprotein-4,19,27-triacetyl withalongolide A. CONCLUSION Synthetic high-density lipoprotein is a promising platform to improve the delivery of anticancer drug 4,19,27-triacetyl withalongolide A to neuroblastomas and neuroblastoma cancer stem cells through SR-B1 targeting in vitro and in vivo.
Collapse
Affiliation(s)
| | - Peter T White
- Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Rui Kuai
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI; Biointerfaces Institute, University of Michigan, Ann Arbor, MI
| | | | | | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI; Biointerfaces Institute, University of Michigan, Ann Arbor, MI
| | | | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI; Biointerfaces Institute, University of Michigan, Ann Arbor, MI
| | - Mark S Cohen
- Department of Surgery, University of Michigan, Ann Arbor, MI; Department of Pharmacology, University of Michigan, Ann Arbor, MI.
| |
Collapse
|
19
|
Subramanian C, Grogan PT, Opipari VP, Timmermann BN, Cohen MS. Novel natural withanolides induce apoptosis and inhibit migration of neuroblastoma cells through down regulation of N-myc and suppression of Akt/mTOR/NF-κB activation. Oncotarget 2018; 9:14509-14523. [PMID: 29581860 PMCID: PMC5865686 DOI: 10.18632/oncotarget.24429] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 09/04/2017] [Indexed: 02/06/2023] Open
Abstract
Despite recent advances in intensive chemotherapy treatments, long-term success is achieved in less than 30% of children with high-risk neuroblastoma (NB). Key regulatory pathways including the PI3K/Akt, mTOR and NF-κB are implicated in the pathogenesis of NB. Although drugs targeting these individual pathways are in clinical trials, they are not effective due to the activation of compensatory mechanisms. We have previously reported that natural novel withanolides from Physalis longifolia can potently inhibit these key regulatory pathways simultaneously. In the present study, we examined the efficacy and mechanisms through which novel withanolides and their acetate derivatives (WGA-TA and WGB-DA) from P.longifolia kill NB cells. The results from the study demonstrated that our novel acetate derivatives are highly effective in inhibiting the proliferation, shifting the cell cycle and inducing apoptosis in a dose dependent manner. Analysis of oncogenic pathway proteins targeted by withanolides indicated induction of heat shock response due to oxidative stress. Dose dependent decrease in clients of HSP90 chaperone function due to suppression of Akt, mTOR, and NF-κB pathways led to decrease in the expressions of target genes such as cyclin D1, N-myc and Survivin. Additionally, there was a dose dependent attenuation of the migration and invasion of NB cells. Furthermore, the lead compound WGA-TA showed significant reduction in tumor growth of NB xenografts. Taken together, these results suggest that withanolides are an effective therapeutic option against NBs.
Collapse
Affiliation(s)
| | - Patrick T Grogan
- Department of Internal Medicine, University of Wisconsin, Madison, WI, USA
| | - Valerie P Opipari
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | | | - Mark S Cohen
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.,Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
20
|
Gorji N, Moeini R, Memariani Z. Almond, hazelnut and walnut, three nuts for neuroprotection in Alzheimer's disease: A neuropharmacological review of their bioactive constituents. Pharmacol Res 2017; 129:115-127. [PMID: 29208493 DOI: 10.1016/j.phrs.2017.12.003] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/01/2017] [Accepted: 12/01/2017] [Indexed: 01/13/2023]
Abstract
An increase in the prevalence of Alzheimer's disease (AD) as a multifactorial neurodegenerative disorder is an almost obvious issue in the world. Researches on natural products for finding effective drugs to prevent the disease are in progress. There is special attention to the three types of nuts including almond, hazelnut and walnut in manuscripts of traditional Persian medicine (PM) as the preventive agents against brainatrophy and memory loss. The purpose of this study is a pharmacological review of their bioactive constituents and introducing the value of these nuts as the effective supplements and natural medicinal foods in AD patients. Databases including PubMed and ScienceDirect were searched in title, abstract and keywords from year 2000 to present for AD-related researches on these tree nuts, their major phytochemicals and their mechanisms of action. As result, almond, hazelnut and walnut provide macronutrients, micronutrients, and phytochemicals which affect several pathways in AD pathogenesis such as amyloidogenesis, tau phosphorylation, oxidative stress, cholinergic pathways, and some non-target mechanisms including cholesterol lowering and anti-inflammatory properties, as well as effect on neurogenesis. These nuts are recommended in PM for their brain-protective activity and particularly reversing brain atrophy in case of hazelnut. The therapeutical statements of PM scholars mentioned in their books are based on their clinical observations with support of a long history of experiences. Beyond the molecular activities attributed to the phytochemicals, the use of these tree nuts could be more considered in scientific researches as the effective nutrients for prevention or even management of AD.
Collapse
Affiliation(s)
- Narjes Gorji
- Traditional Medicine & Medical History Research Center, Health Research Center, Department of Persian Medicine, Babol University of Medical Sciences, Babol, Islamic Republic of Iran
| | - Reihaneh Moeini
- Traditional Medicine & Medical History Research Center, Health Research Center, Department of Persian Medicine, Babol University of Medical Sciences, Babol, Islamic Republic of Iran
| | - Zahra Memariani
- Traditional Medicine & Medical History Research Center, Health Research Center, Department of Persian Medicine, Babol University of Medical Sciences, Babol, Islamic Republic of Iran.
| |
Collapse
|
21
|
Challa C, Ravindran J, Konai MM, Varughese S, Jacob J, Kumar BSD, Haldar J, Lankalapalli RS. Expedient Synthesis of Indolo[2,3- b]quinolines, Chromeno[2,3- b]indoles, and 3-Alkenyl-oxindoles from 3,3'-Diindolylmethanes and Evaluation of Their Antibiotic Activity against Methicillin-Resistant Staphylococcus aureus. ACS OMEGA 2017; 2:5187-5195. [PMID: 30023741 PMCID: PMC6044809 DOI: 10.1021/acsomega.7b00840] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/16/2017] [Indexed: 05/09/2023]
Abstract
Easily accessible 3,3'-diindolylmethanes (DIMs) were utilized to generate a focused library of indolo[2,3-b]quinolines (2), chromeno[2,3-b]indoles (3), and 3-alkenyl-oxindoles (4) under 2,3-Dichloro-5,6-dicyano-1,4-benzoquinone (DDQ)-mediated oxidative conditions. DIMs with ortho-NHTosyl (NHTs) phenyl group afforded indolo[2,3-b]quinolines (2), whereas DIMs with ortho-hydroxy phenyl groups yielded chromeno[2,3-b]indoles (3) and 3-alkenyl-oxindoles (4). The mild conditions and excellent yields of the products make this method a good choice to access a diverse library of bioactive molecules from a common starting material. Two optimized compounds 2a and 2n displayed excellent activity against clinical isolates of methicillin-resistant Staphylococcus aureus (MRSA). Compound 2a showed the minimum inhibitory concentration values in the concentration between 1 and 4 μg/mL, whereas compound 2n revealed the values of 1-2 μg/mL. Furthermore, both the compounds were highly bactericidal and capable to kill the MRSA completely within 360 min. Collectively, the results suggested that both compounds 2a and 2n possess enormous potential to be developed as anti-MRSA agents.
Collapse
Affiliation(s)
- Chandrasekhar Challa
- Chemical
Sciences and Technology Division and Academy of Scientific
and Innovative Research (AcSIR) and Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science
and Technology (CSIR-NIIST), Thiruvananthapuram 695019, Kerala, India
| | - Jaice Ravindran
- Chemical
Sciences and Technology Division and Academy of Scientific
and Innovative Research (AcSIR) and Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science
and Technology (CSIR-NIIST), Thiruvananthapuram 695019, Kerala, India
| | - Mohini Mohan Konai
- Antimicrobial
Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Sunil Varughese
- Chemical
Sciences and Technology Division and Academy of Scientific
and Innovative Research (AcSIR) and Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science
and Technology (CSIR-NIIST), Thiruvananthapuram 695019, Kerala, India
| | - Jubi Jacob
- Chemical
Sciences and Technology Division and Academy of Scientific
and Innovative Research (AcSIR) and Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science
and Technology (CSIR-NIIST), Thiruvananthapuram 695019, Kerala, India
| | - B. S. Dileep Kumar
- Chemical
Sciences and Technology Division and Academy of Scientific
and Innovative Research (AcSIR) and Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science
and Technology (CSIR-NIIST), Thiruvananthapuram 695019, Kerala, India
| | - Jayanta Haldar
- Antimicrobial
Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Ravi S. Lankalapalli
- Chemical
Sciences and Technology Division and Academy of Scientific
and Innovative Research (AcSIR) and Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science
and Technology (CSIR-NIIST), Thiruvananthapuram 695019, Kerala, India
- E-mail:
| |
Collapse
|
22
|
Wang Z, Li MY, Mi C, Wang KS, Ma J, Jin X. Mollugin Has an Anti-Cancer Therapeutic Effect by Inhibiting TNF-α-Induced NF-κB Activation. Int J Mol Sci 2017; 18:ijms18081619. [PMID: 28933726 PMCID: PMC5578011 DOI: 10.3390/ijms18081619] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 06/19/2017] [Accepted: 07/20/2017] [Indexed: 12/18/2022] Open
Abstract
The NF-κB signaling pathway plays a pivotal role in regulating the immune response and inflammation. However, it has been shown that NF-κB also has a major role in oncogenesis. Therefore, NF-κB inhibitors have been considered as potential drugs against cancer. Herein, we searched for NF-κB inhibitors from natural sources and identified mollugin from the roots of Rubia cordifolia L. as an inhibitor of NF-κB activation. We found that mollugin significantly inhibited the expression of an NF-κB reporter gene induced by tumor necrosis factor (TNF)-α in a dose-dependent manner. Moreover, mollugin inhibited TNF-α-induced phosphorylation and nuclear translocation of p65, phosphorylation and degradation of inhibitor of κB (IκBα), and IκB kinase (IKK) phosphorylation. Furthermore, we discovered that pretreatment of cells with mollugin prevented the TNF-α-induced expression of NF-κB target genes, such as genes related to proliferation (COX-2, Cyclin D1 and c-Myc), anti-apoptosis (Bcl-2, cIAP-1 and survivin), invasion (MMP-9 and ICAM-1), and angiogenesis (VEGF). We also demonstrated that mollugin potentiated TNF-α-induced apoptosis and inhibited proliferation of HeLa cells. We further demonstrated in vivo that mollugin suppressed the growth of tumor xenografts derived from HeLa cells. Taken together, mollugin may be a valuable candidate for cancer treatment by targeting NF-κB.
Collapse
Affiliation(s)
- Zhe Wang
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, China.
| | - Ming Yue Li
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, China.
| | - Chunliu Mi
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, China.
| | - Ke Si Wang
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, China.
| | - Juan Ma
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, China.
| | - Xuejun Jin
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, China.
| |
Collapse
|
23
|
Recent advances (2015-2016) in anticancer hybrids. Eur J Med Chem 2017; 142:179-212. [PMID: 28760313 DOI: 10.1016/j.ejmech.2017.07.033] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/13/2017] [Accepted: 07/18/2017] [Indexed: 12/26/2022]
Abstract
In spite of the development of a large number of novel anticancer drugs over the years, Cancer remains as a prominent cause of death, worldwide. Numerous drugs that are currently in clinical practice have developed multidrug resistance along with fatal side effects. Therefore, the utilization of single-target therapy is incapable of providing an effective control on the malignant process. Molecular hybridization, involving a combination of two or more pharmacophores of bioactive scaffolds to generate a single molecular architecture with improved affinity and activity, in comparison to their parent molecules, has emerged as a promising strategy in recent drug discovery research. Hybrid anticancer drugs are of great therapeutic interests since they can potentially overcome most of the pharmacokinetic drawbacks encountered with conventional anticancer drugs. Strategically, the design of anticancer drugs involved the blending or linking of an anticancer drug with another anticancer drug or a carrier molecule which can efficiently target cancer cells with improved biological potential. Major advantages of hybrid anticancer drugs involved increased specificity, better patient compliance, and lower side effects along with reduction in chemo-resistance. The successful utilization of this technique in design and synthesis of novel anticancer hybrids has been well illustrated and documented in the literature. The purpose of the present review article will be to provide an emphasis on the recent developments (2015-16) in anticancer hybrids with insights into their structure-activity relationship (SAR) and mechanism of action.
Collapse
|
24
|
Cebrián-Torrejón G, Doménech-Carbó A, Figadère B, Poupon E, Fournet A. Phytoelectrochemical analysis of Zanthoxylum chiloperone. PHYTOCHEMICAL ANALYSIS : PCA 2017; 28:171-175. [PMID: 27995663 DOI: 10.1002/pca.2657] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 09/27/2016] [Accepted: 09/28/2016] [Indexed: 06/06/2023]
Abstract
INTRODUCTION An innovative application of the voltammetry of microparticles methodology to characterize the phytochemical composition of extracts of different parts of Zanthoxylum chiloperone var. angustifolium Engl. is described. OBJECTIVE Characterize the phytochemical composition of extracts of different parts of plants by electrochemical methodologies. METHODS The voltammetry of microparticles methodology was applied to alcoholic extracts from leaves, seeds, fruits, roots and stem bark of Zanthoxylum chiloperone. RESULTS In contact with aqueous phosphate buffer, characteristic cathodic signals of its main natural products (canthin-6-one, 5-methoxycanthin-6-one and trans-avicennol) were recorded. The study of the voltammograns allows the estimation of the relative amounts of canthin-6-one, 5-methoxycanthin-6-one and trans-avicennol from the different parts of Zanthoxylum chiloperone. CONCLUSION The voltammetric responses of alcoholic extracts from different parts of Zanthoxylum chiloperone var. angustifolium allows their phytochemical characterization without need of sample pretreatment thus illustrating the capabilities of the voltammetry of microparticles methodology to increase the tools applied to phytochemical analysis. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Gerardo Cebrián-Torrejón
- Department de Química Analítica, Universitat de València, Dr. Moliner, 50, 46100 Burjasot (València), Spain
- BioCIS, Université Paris-Sud, CNRS, Université Paris-Saclay, 92290, Châtenay-Malabry, France
| | - Antonio Doménech-Carbó
- Department de Química Analítica, Universitat de València, Dr. Moliner, 50, 46100 Burjasot (València), Spain
| | - Bruno Figadère
- BioCIS, Université Paris-Sud, CNRS, Université Paris-Saclay, 92290, Châtenay-Malabry, France
| | - Erwan Poupon
- BioCIS, Université Paris-Sud, CNRS, Université Paris-Saclay, 92290, Châtenay-Malabry, France
| | - Alain Fournet
- IRD UMR 217, Laboratoire de Pharmacognosie, Faculté de Pharmacie, rue Jean-Baptiste Clément, 92296, Châtenay-Malabry, France
| |
Collapse
|
25
|
Sadeeshkumar V, Duraikannu A, Ravichandran S, Kodisundaram P, Fredrick WS, Gobalakrishnan R. Modulatory efficacy of dieckol on xenobiotic-metabolizing enzymes, cell proliferation, apoptosis, invasion and angiogenesis during NDEA-induced rat hepatocarcinogenesis. Mol Cell Biochem 2017; 433:195-204. [PMID: 28397013 DOI: 10.1007/s11010-017-3027-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/01/2017] [Indexed: 12/25/2022]
Abstract
Dieckol (DEK) is a major polyphenol of marine brown seaweed Ecklonia cava which is a potential candidate for cancer therapy. However, the underlying mechanism of DEK as an anticancer drug remains to be elucidated. In this study, we evaluated the molecular mechanisms involved in the chemopreventive efficacy of DEK in N-nitrosodiethylamine (NDEA)-induced hepatocarcinogenesis rats by analyzing markers of xenobiotic-metabolizing enzymes (XMEs), apoptosis, invasion, and angiogenesis. Rats administered NDEA developed hepatocarcinogenesis that displayed apoptosis avoidance coupled to upregulation of pro-inflammatory, invasion, and angiogenesis markers. Treatment of DEK effectively suppressed the NDEA-initiated hepatocarcinogenesis by modulation of XMEs, inducing of apoptosis via the mitochondrial pathway as revealed by modulating the Bcl-2 family proteins, cytochrome C, caspases, and inhibiting invasion, and angiogenesis as evidenced by changes in the activities of MMPs (MMP2/9) and the expression of VEGF. In addition, DEK exerts its anticancer effects via inhibition of pro-inflammatory transcription factor NF-κB (nuclear factor κB) and COX2 in NDEA-induced hepatocarcinogenesis. Taken together, this study demonstrates that DEK modulates the expression of key molecules that regulate apoptosis, inflammation, invasion, and angiogenesis. These results strongly indicate that DEK from E. cava is an attractive candidate for chemoprevention.
Collapse
Affiliation(s)
- Velayutham Sadeeshkumar
- Centre of Advanced Study in Marine Biology, Faculty of Marine Sciences, Annamalai University, Parangipettai, Chidambaram, Tamil Nadu, 608 502, India.
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Chidambaram, Tamil Nadu, 608 002, India.
| | - Arul Duraikannu
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Chidambaram, Tamil Nadu, 608 002, India
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, AB, T6G 2G3, Canada
| | - Samuthrapandian Ravichandran
- Centre of Advanced Study in Marine Biology, Faculty of Marine Sciences, Annamalai University, Parangipettai, Chidambaram, Tamil Nadu, 608 502, India
| | - Paulrasu Kodisundaram
- Department of Chemistry, Faculty of Science, Annamalai University, Chidambaram, Tamil Nadu, 608 002, India
| | - Wilson Sylvester Fredrick
- Centre of Advanced Study in Marine Biology, Faculty of Marine Sciences, Annamalai University, Parangipettai, Chidambaram, Tamil Nadu, 608 502, India
| | - Rajagopal Gobalakrishnan
- Centre of Advanced Study in Marine Biology, Faculty of Marine Sciences, Annamalai University, Parangipettai, Chidambaram, Tamil Nadu, 608 502, India
| |
Collapse
|
26
|
Guamán-Ortiz LM, Orellana MIR, Ratovitski EA. Natural Compounds As Modulators of Non-apoptotic Cell Death in Cancer Cells. Curr Genomics 2017; 18:132-155. [PMID: 28367073 PMCID: PMC5345338 DOI: 10.2174/1389202917666160803150639] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 11/24/2015] [Accepted: 11/28/2015] [Indexed: 02/07/2023] Open
Abstract
Cell death is an innate capability of cells to be removed from microenvironment, if and when they are damaged by multiple stresses. Cell death is often regulated by multiple molecular pathways and mechanism, including apoptosis, autophagy, and necroptosis. The molecular network underlying these processes is often intertwined and one pathway can dynamically shift to another one acquiring certain protein components, in particular upon treatment with various drugs. The strategy to treat human cancer ultimately relies on the ability of anticancer therapeutics to induce tumor-specific cell death, while leaving normal adjacent cells undamaged. However, tumor cells often develop the resistance to the drug-induced cell death, thus representing a great challenge for the anticancer approaches. Numerous compounds originated from the natural sources and biopharmaceutical industries are applied today in clinics showing advantageous results. However, some exhibit serious toxic side effects. Thus, novel effective therapeutic approaches in treating cancers are continued to be developed. Natural compounds with anticancer activity have gained a great interest among researchers and clinicians alike since they have shown more favorable safety and efficacy then the synthetic marketed drugs. Numerous studies in vitro and in vivo have found that several natural compounds display promising anticancer potentials. This review underlines certain information regarding the role of natural compounds from plants, microorganisms and sea life forms, which are able to induce non-apoptotic cell death in tumor cells, namely autophagy and necroptosis.
Collapse
Affiliation(s)
- Luis Miguel Guamán-Ortiz
- 1 Departamento de Ciencias de la Salud, Universidad Técnica Particular de Loja, Loja, Ecuador ; 2 Head and Neck Cancer Research Division, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Maria Isabel Ramirez Orellana
- 1 Departamento de Ciencias de la Salud, Universidad Técnica Particular de Loja, Loja, Ecuador ; 2 Head and Neck Cancer Research Division, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Edward A Ratovitski
- 1 Departamento de Ciencias de la Salud, Universidad Técnica Particular de Loja, Loja, Ecuador ; 2 Head and Neck Cancer Research Division, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
27
|
Wang LH, Jiang XR, Chen GL, Guo W, Zhang JY, Cui LJ, Li HH, Li M, Liu X, Yang JY, Wu CF. Anti-tumor activity of SL4 against breast cancer cells: induction of G 2/M arrest through modulation of the MAPK-dependent p21 signaling pathway. Sci Rep 2016; 6:36486. [PMID: 27819344 PMCID: PMC5098232 DOI: 10.1038/srep36486] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 10/05/2016] [Indexed: 02/06/2023] Open
Abstract
SL4, a chalcone-based compound, has been shown to retard tumor invasion and angiogenesis by suppressing HIF1 activity and to induce apoptosis by promoting ROS release. Here, we report that SL4 is able to inhibit the proliferation of different types of breast cancer cell in vitro and in vivo by inducing G2/M cell cycle arrest. Our results showed that SL4 exhibited strong anti-proliferative activity in several human breast cancer cell lines, with IC50 values lower than 1.3 μM. Further studies indicated that SL4 induced G2/M arrest in these cell lines. Mechanistically, SL4 reduces the expression of cyclin A2 and cdc25C and decreases the activity of the cdc2/cyclin B1 complex. Notably, SL4 treatment resulted in an obvious increase in p21 mRNA and protein levels through activation of MAPK signaling pathways, but not the TGF-β pathway. SP600125 and PD98059, specific inhibitors of JNK kinase and ERK kinase, significantly blocked the SL4-induced G2/M phase arrest and upregulation of p21. Furthermore, SL4 suppressed the growth of established breast tumors in nude mice through upregulation of p21 and downregulation of cdc25C, and displayed a good safety profile. Taken together, these findings demonstrate the potential value of SL4 as a novel multi-target anti-tumor drug candidate.
Collapse
Affiliation(s)
- Li-Hui Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Xiao-Rui Jiang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Guo-Liang Chen
- Key Laboratory of Structure-Based Drugs Design &Discovery of Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Wei Guo
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Jing-Yuan Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Li-Juan Cui
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Hua-Huan Li
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Meng Li
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Xing Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Jing-Yu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Chun-Fu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| |
Collapse
|
28
|
Biersack B. Non-coding RNA/microRNA-modulatory dietary factors and natural products for improved cancer therapy and prevention: Alkaloids, organosulfur compounds, aliphatic carboxylic acids and water-soluble vitamins. Noncoding RNA Res 2016; 1:51-63. [PMID: 30159411 PMCID: PMC6096427 DOI: 10.1016/j.ncrna.2016.09.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 09/01/2016] [Accepted: 09/01/2016] [Indexed: 02/06/2023] Open
Abstract
Non-coding small RNA molecules, the microRNAs (miRNAs), contribute decisively to the epigenetic regulation processes in cancer cells. Problematic pathogenic properties of cancer cells and the response of cancers towards anticancer drugs are highly influenced by miRNAs. Both increased drug activity and formation of tumor resistance are regulated by miRNAs. Further to this, the survival and proliferation of cancer cells and the formation of metastases is based on the modulated expression of certain miRNAs. In particular, drug-resistant cancer stem-like cells (CSCs) depend on the presence and absence of specific miRNAs. Fortunately, several small molecule natural compounds were discovered that target miRNAs involved in the modulation of tumor aggressiveness and drug resistance. This review gives an overview of the effects of a selection of naturally occurring small molecules (alkaloids, organosulfur compounds, aliphatic carboxylic acids and water-soluble vitamins) on miRNAs that are closely tangled with cancer diseases.
Collapse
Key Words
- AM, allyl mercaptan
- AOM, azoxymethane
- Aliphatic carboxylic acids
- Alkaloids
- Anticancer drugs
- CPT, camptothecin
- DADS, diallyl disulfide
- DHA, docosahexaenoic acid
- DIM, 3,3′-diindolylmethane
- EPA, eicosapentaenoic acid
- FA, folic acid
- GTC, green tea catechins
- I3C, indole-3-carbinol
- MiRNA
- NaB, sodium butyrate
- Organosulfur compounds
- PEITC, phenethylisothiocyanate
- PUFA, polyunsaturated fatty acid
- SAMC, S-allylmercaptocysteine
- SFN, sulforaphane
- TSA, trichostatin A
- Water-soluble vitamins
Collapse
|
29
|
Biersack B. Current state of phenolic and terpenoidal dietary factors and natural products as non-coding RNA/microRNA modulators for improved cancer therapy and prevention. Noncoding RNA Res 2016; 1:12-34. [PMID: 30159408 PMCID: PMC6096431 DOI: 10.1016/j.ncrna.2016.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 07/20/2016] [Accepted: 07/20/2016] [Indexed: 02/06/2023] Open
Abstract
The epigenetic regulation of cancer cells by small non-coding RNA molecules, the microRNAs (miRNAs), has raised particular interest in the field of oncology. These miRNAs play crucial roles concerning pathogenic properties of cancer cells and the sensitivity of cancer cells towards anticancer drugs. Certain miRNAs are responsible for an enhanced activity of drugs, while others lead to the formation of tumor resistance. In addition, miRNAs regulate survival and proliferation of cancer cells, in particular of cancer stem-like cells (CSCs), that are especially drug-resistant and, thus, cause tumor relapse in many cases. Various small molecule compounds were discovered that target miRNAs that are known to modulate tumor aggressiveness and drug resistance. This review comprises the effects of naturally occurring small molecules (phenolic compounds and terpenoids) on miRNAs involved in cancer diseases.
Collapse
Key Words
- 1,25-D, 1,25-dihydroxyvitamin D3
- 18-AGA, 18α-glycyrrhetinic acid
- 3,6-DHF, 3,6-dihydroxyflavone
- AKBA, 3-acetyl-11-keto-β-boswellic acid
- Anticancer drugs
- CAPE, caffeic acid phenethyl ester
- CDODA-Me, methyl 2-cyano-3,11-dioxo-18β-olean-1,12-dien-30-oate
- Dox, doxorubicin
- EGCG, (−)-epigallocatechin-3-O-gallate
- MicroRNA
- PEG, polyethylene glycol
- PPAP, polycyclic polyprenylated acylphloroglucinol
- Polyphenols
- RA, retinoic acid
- ROS, reactive oxygen species
- TQ, thymoquinone
- Terpenes
Collapse
Affiliation(s)
- Bernhard Biersack
- Organic Chemistry Laboratory, University of Bayreuth, Universitätsstraße 30, 95447 Bayreuth, Germany
| |
Collapse
|
30
|
Muenzner JK, Ahmad A, Rothemund M, Schrüfer S, Padhye S, Sarkar FH, Schobert R, Biersack B. Ferrocene-substituted 3,3′-diindolylmethanes with improved anticancer activity. Appl Organomet Chem 2016. [DOI: 10.1002/aoc.3452] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Julienne K. Muenzner
- Organic Chemistry Laboratory; University of Bayreuth; Universitaetsstrasse 30 95440 Bayreuth Germany
| | - Aamir Ahmad
- Karmanos Cancer Institute; Wayne State University School of Medicine; Detroit MI 48201 USA
| | - Matthias Rothemund
- Organic Chemistry Laboratory; University of Bayreuth; Universitaetsstrasse 30 95440 Bayreuth Germany
| | - Sebastian Schrüfer
- Organic Chemistry Laboratory; University of Bayreuth; Universitaetsstrasse 30 95440 Bayreuth Germany
| | - Subhash Padhye
- Abeda Inamdar Senior College; University of Pune; 2390 K. B. Hidayatullah Road , Azam Campus Pune 411 001 India
| | - Fazlul H. Sarkar
- Karmanos Cancer Institute; Wayne State University School of Medicine; Detroit MI 48201 USA
| | - Rainer Schobert
- Organic Chemistry Laboratory; University of Bayreuth; Universitaetsstrasse 30 95440 Bayreuth Germany
| | - Bernhard Biersack
- Organic Chemistry Laboratory; University of Bayreuth; Universitaetsstrasse 30 95440 Bayreuth Germany
| |
Collapse
|
31
|
Kundu A, Quirit JG, Khouri MG, Firestone GL. Inhibition of oncogenic BRAF activity by indole-3-carbinol disrupts microphthalmia-associated transcription factor expression and arrests melanoma cell proliferation. Mol Carcinog 2016; 56:49-61. [PMID: 26878440 DOI: 10.1002/mc.22472] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 01/28/2016] [Indexed: 12/18/2022]
Abstract
Indole-3-carbinol (I3C), an anti-cancer phytochemical derived from cruciferous vegetables, strongly inhibited proliferation and down-regulated protein levels of the melanocyte master regulator micropthalmia-associated transcription factor (MITF-M) in oncogenic BRAF-V600E expressing melanoma cells in culture as well as in vivo in tumor xenografted athymic nude mice. In contrast, wild type BRAF-expressing melanoma cells remained relatively insensitive to I3C anti-proliferative signaling. In BRAF-V600E-expressing melanoma cells, I3C treatment inhibited phosphorylation of MEK and ERK/MAPK, the down stream effectors of BRAF. The I3C anti-proliferative arrest was concomitant with the down-regulation of MITF-M transcripts and promoter activity, loss of endogenous BRN-2 binding to the MITF-M promoter, and was strongly attenuated by expression of exogenous MITF-M. Importantly, in vitro kinase assays using immunoprecipitated BRAF-V600E and wild type BRAF demonstrated that I3C selectively inhibited the enzymatic activity of the oncogenic BRAF-V600E but not of the wild type protein. In silico modeling predicted an I3C interaction site in the BRAF-V600E protomer distinct from where the clinically used BRAF-V600E inhibitor Vemurafenib binds to BRAF-V600E. Consistent with this prediction, combinations of I3C and Vemurafenib more potently inhibited melanoma cell proliferation and reduced MITF-M levels in BRAF-V600E expressing melanoma cells compared to the effects of each compound alone. Thus, our results demonstrate that oncogenic BRAF-V600E is a new cellular target of I3C that implicate this indolecarbinol compound as a potential candidate for novel single or combination therapies for melanoma. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Aishwarya Kundu
- Department of Molecular and Cell Biology and The Cancer Research Laboratory, University of California at Berkeley, Berkeley, California
| | - Jeanne G Quirit
- Department of Molecular and Cell Biology and The Cancer Research Laboratory, University of California at Berkeley, Berkeley, California
| | - Michelle G Khouri
- Department of Molecular and Cell Biology and The Cancer Research Laboratory, University of California at Berkeley, Berkeley, California
| | - Gary L Firestone
- Department of Molecular and Cell Biology and The Cancer Research Laboratory, University of California at Berkeley, Berkeley, California
| |
Collapse
|
32
|
Cebrián-Torrejón G, Doménech-Carbó A, Scotti M, Fournet A, Figadère B, Poupon E. Experimental and theoretical study of possible correlation between the electrochemistry of canthin-6-one and the anti-proliferative activity against human cancer stem cells. J Mol Struct 2015. [DOI: 10.1016/j.molstruc.2015.08.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
33
|
Sharma S, Gupta MK, Saxena AK, Bedi PMS. Triazole linked mono carbonyl curcumin-isatin bifunctional hybrids as novel anti tubulin agents: Design, synthesis, biological evaluation and molecular modeling studies. Bioorg Med Chem 2015; 23:7165-80. [PMID: 26515041 DOI: 10.1016/j.bmc.2015.10.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 10/06/2015] [Accepted: 10/11/2015] [Indexed: 12/13/2022]
Abstract
Keeping in view the limitations associated with currently available anticancer drugs, molecular hybrids of mono carbonyl curcumin and isatin tethered by triazole ring have been synthesized and evaluated for in vitro cytotoxicity against THP-1, COLO-205, HCT-116, A549, HeLa, CAKI-I, PC-3, MiaPaca-2 human cancer cell lines. The results revealed that the compounds SA-1 to SA-9, SB-2, SB-3, SB-4, SB-7 and SC-2 showed a good range of IC50 values against THP-1, COLO-205, HCT-116 and PC-3 cell lines, while the other four cell lines among these were found to be almost resistant. Structure activity relationship revealed that the nature of Ring X and substitution at position R influences the activity. Methoxy substituted phenyl ring as Ring X and H as R were found to be the ideal structural features. The most potent compounds (SA-2, SA-3, SA-4, SA-7) were further tested for tubulin inhibition. Compound SA-2 was found to significantly inhibit the tubulin polymerization (IC50=1.2 μM against HCT-116). Compound SA-2, moreover, lead to the disruption of microtubules as confirmed by immunofluorescence technique. The significant cytotoxicity and tubulin inhibition by SA-2 was streamlined by molecular modeling studies where it was docked at the curcumin binding site of tubulin.
Collapse
Affiliation(s)
- Sahil Sharma
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Manish K Gupta
- Lloyd Institute of Management and Technology, Greater Noida, UP, India
| | - Ajit K Saxena
- Indian Institute of Integrative Medicine, Jammu, India
| | - Preet Mohinder S Bedi
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| |
Collapse
|
34
|
Rothman D, Gao X, George E, Rasmusson T, Bhatia D, Alimov I, Wang L, Kamel A, Hatsis P, Feng Y, Tutter A, Michaud G, McDonald E, Venkatesan K, Farley D, Digan M, Ni Y, Harbinski F, Gunduz M, Wilson C, Buckler A, Labow M, Tallarico J, Myer V, Porter J, Wang S. Metabolic Enzyme Sulfotransferase 1A1 Is the Trigger for N-Benzyl Indole Carbinol Tumor Growth Suppression. ACTA ACUST UNITED AC 2015; 22:1228-37. [DOI: 10.1016/j.chembiol.2015.06.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 06/17/2015] [Accepted: 06/17/2015] [Indexed: 12/30/2022]
|
35
|
Fruit and vegetable consumption and risk of lung cancer: A dose–response meta-analysis of prospective cohort studies. Lung Cancer 2015; 88:124-30. [DOI: 10.1016/j.lungcan.2015.02.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 12/21/2014] [Accepted: 02/18/2015] [Indexed: 11/22/2022]
|
36
|
Molecular targets of naturopathy in cancer research: bridge to modern medicine. Nutrients 2015; 7:321-34. [PMID: 25569626 PMCID: PMC4303842 DOI: 10.3390/nu7010321] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 12/23/2014] [Indexed: 01/10/2023] Open
Abstract
The relevance of naturopathy (defined as the practice of medicine for the treatment of human diseases with natural agents) in human cancer is beginning to be appreciated, as documented by renewed interest in nutraceutical research, the natural anticancer agents of dietary origin. Because of their pleiotropic effects and the ability to modulate multiple signaling pathways, which is a good attribute of natural agents, nutraceuticals have frequently been demonstrated to re-sensitize drug-resistant cancers. The effectiveness of nutraceuticals can be further enhanced if the tools for the relative assessment of their molecular targets are readily available. Such information can be critical for determining their most effective uses. Here, we discuss the anticancer potential of nutraceuticals and the associated challenges that have interfered with their translational potential as a naturopathic approach for the management of cancers. In the years to come, an efficient screening and assessment of molecular targets will be the key to make rapid progress in the area of drug design and discovery, especially focusing on evidence-based development of naturopathy for the treatment of human malignancies.
Collapse
|
37
|
Kandi SK, Manohar S, Vélez Gerena CE, Zayas B, Malhotra SV, Rawat DS. C5-curcuminoid-4-aminoquinoline based molecular hybrids: design, synthesis and mechanistic investigation of anticancer activity. NEW J CHEM 2015. [DOI: 10.1039/c4nj00936c] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Novel hybrids exhibiting excellent anticancer activity against most of the NCI 60 cell lines through apoptotic pathways are reported herein.
Collapse
Affiliation(s)
| | - Sunny Manohar
- Department of Chemistry
- University of Delhi
- Delhi-110007
- India
| | | | - Beatriz Zayas
- School of Environmental Affairs
- Universidad Metropolitana
- San Juan
- USA
| | - Sanjay V. Malhotra
- Laboratory of Synthetic Chemistry
- Leidos Biomedical Research Inc
- Frederick National Laboratory for Cancer Research
- Frederick
- USA
| | - Diwan S. Rawat
- Department of Chemistry
- University of Delhi
- Delhi-110007
- India
| |
Collapse
|
38
|
Abstract
In spite of the expensive preclinical testing, the consistent failure to translate many promising targeted drugs from the laboratory bench to the clinic raises the question of whether the single-pathway drug-discovery strategies offer the correct perspective. As revealed by network biology, cancers harbor robust biological networks that are inherently resistant to changes, such as those induced by drugs with very narrow mechanisms of action. Therefore, network pharmacology strategies, the treatment of cancer by modulating more than one target, are needed. Different promiscuous approaches targeting multiple avenues within cancer-associated networks, such as the pleiotropic natural products, are emerging. Nevertheless, there is a long way before such 'proof-of-concept strategies' can be successfully applied in the clinical setting. This article provides a perspective on the current challenges in drug discovery, the reasons for high failure rates and how network pharmacology can aid the successful design of agents against cancer.
Collapse
|
39
|
Azmi AS, Bao B, Sarkar FH. Exosomes in cancer development, metastasis, and drug resistance: a comprehensive review. Cancer Metastasis Rev 2014; 32:623-42. [PMID: 23709120 DOI: 10.1007/s10555-013-9441-9] [Citation(s) in RCA: 864] [Impact Index Per Article: 86.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Trafficking of biological material across membranes is an evolutionary conserved mechanism and is part of any normal cell homeostasis. Such transport is composed of active, passive, export through microparticles, and vesicular transport (exosomes) that collectively maintain proper compartmentalization of important micro- and macromolecules. In pathological states, such as cancer, aberrant activity of the export machinery results in expulsion of a number of key proteins and microRNAs resulting in their misexpression. Exosome-mediated expulsion of intracellular drugs could be another barrier in the proper action of most of the commonly used therapeutics, targeted agents, and their intracellular metabolites. Over the last decade, a number of studies have revealed that exosomes cross-talk and/or influence major tumor-related pathways, such as hypoxia-driven epithelial-to-mesenchymal transition, cancer stemness, angiogenesis, and metastasis involving many cell types within the tumor microenvironment. Emerging evidence suggests that exosome-secreted proteins can also propel fibroblast growth, resulting in desmoplastic reaction, a major barrier in effective cancer drug delivery. This comprehensive review highlights the advancements in the understanding of the biology of exosomes secretions and the consequence on cancer drug resistance. We propose that the successful combination of cancer treatments to tackle exosome-mediated drug resistance requires an interdisciplinary understanding of these cellular exclusion mechanisms, and how secreted biomolecules are involved in cellular cross-talk within the tumor microenvironment.
Collapse
Affiliation(s)
- Asfar S Azmi
- Department of Pathology, Wayne State University School of Medicine, 4100 John R, HWCRC 740, Detroit, MI, 48201, USA,
| | | | | |
Collapse
|
40
|
Tin AS, Park AH, Sundar SN, Firestone GL. Essential role of the cancer stem/progenitor cell marker nucleostemin for indole-3-carbinol anti-proliferative responsiveness in human breast cancer cells. BMC Biol 2014; 12:72. [PMID: 25209720 PMCID: PMC4180847 DOI: 10.1186/s12915-014-0072-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Nucleostemin is a nucleolus residing GTPase that is considered to be an important cancer stem/progenitor cell marker protein due to its high expression levels in breast cancer stem cells and its role in tumor-initiation of human mammary tumor cells. It has been proposed that nucleostemin may represent a valuable therapeutic target for breast cancer; however, to date evidence supporting the cellular mechanism has not been elucidated. RESULTS Expression of exogenous HER2, a member of the EGF receptor gene family, in the human MCF-10AT preneoplastic mammary epithelial cell line formed a new breast cancer cell line, 10AT-Her2, which is highly enriched in cells with stem/progenitor cell-like character. 10AT-Her2 cells display a CD44+/CD24-/low phenotype with high levels of the cancer stem/progenitor cell marker proteins nucleostemin, and active aldehyde dehydrogenase-1. The overall expression pattern of HER2 protein and the stem/progenitor cell marker proteins in the 10AT-Her2 cell population is similar to that of the luminal HER2+ SKBR3 human breast cancer cell line, whereas, both MCF-7 and MDA-MB-231 cells display reduced levels of nucleostemin and no detectable expression of ALDH-1. Importantly, in contrast to the other well-established human breast cancer cell lines, 10AT-Her2 cells efficiently form tumorspheres in suspension cultures and initiate tumor xenograft formation in athymic mice at low cell numbers. Furthermore, 10AT-Her2 cells are highly sensitive to the anti-proliferative apoptotic effects of indole-3-carbinol (I3C), a natural anti-cancer indolecarbinol from cruciferous vegetables of the Brassica genus such as broccoli and cabbage. I3C promotes the interaction of nucleostemin with MDM2 (Murine Double Mutant 2), an inhibitor of the p53 tumor suppressor, and disrupts the MDM2 interaction with p53. I3C also induced nucleostemin to sequester MDM2 in a nucleolus compartment, thereby freeing p53 to mediate its apoptotic activity. siRNA knockdown of nucleostemin functionally documented that nucleostemin is required for I3C to trigger its cellular anti-proliferative responses, inhibit tumorsphere formation, and disrupt MDM2-p53 protein-protein interactions. Furthermore, expression of an I3C-resistant form of elastase, the only known target protein for I3C, prevented I3C anti-proliferative responses in cells and in tumor xenografts in vivo, as well as disrupt the I3C stimulated nucleostemin-MDM2 interactions. CONCLUSIONS Our results provide the first evidence that a natural anti-cancer compound mediates its cellular and in vivo tumor anti-proliferative responses by selectively stimulating cellular interactions of the stem/progenitor cell marker nucleostemin with MDM2, which frees p53 to trigger its apoptotic response. Furthermore, our study provides a new mechanistic template that can be potentially exploited for the development of cancer stem/progenitor cell targeted therapeutic strategies.
Collapse
|
41
|
Aronchik I, Kundu A, Quirit JG, Firestone GL. The antiproliferative response of indole-3-carbinol in human melanoma cells is triggered by an interaction with NEDD4-1 and disruption of wild-type PTEN degradation. Mol Cancer Res 2014; 12:1621-1634. [PMID: 25009292 DOI: 10.1158/1541-7786.mcr-14-0018] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
UNLABELLED Human melanoma cells displaying distinct PTEN genotypes were used to assess the cellular role of this important tumor-suppressor protein in the antiproliferative response induced by the chemopreventative agent indole-3-carbinol (I3C), a natural indolecarbinol compound derived from the breakdown of glucobrassicin produced in cruciferous vegetables such as broccoli and Brussels sprouts. I3C induced a G1-phase cell-cycle arrest and apoptosis by stabilization of PTEN in human melanoma cells that express wild-type PTEN, but not in cells with mutant or null PTEN genotypes. Importantly, normal human epidermal melanocytes were unaffected by I3C treatment. In wild-type PTEN-expressing melanoma xenografts, formed in athymic mice, I3C inhibited the in vivo tumor growth rate and increased PTEN protein levels in the residual tumors. Mechanistically, I3C disrupted the ubiquitination of PTEN by NEDD4-1 (NEDD4), which prevented the proteasome-mediated degradation of PTEN without altering its transcript levels. RNAi-mediated knockdown of PTEN prevented the I3C-induced apoptotic response, whereas knockdown of NEDD4-1 mimicked the I3C apoptotic response, stabilized PTEN protein levels, and downregulated phosphorylated AKT-1 levels. Co-knockdown of PTEN and NEDD4-1 revealed that I3C-regulated apoptotic signaling through NEDD4-1 requires the presence of the wild-type PTEN protein. Finally, in silico structural modeling, in combination with isothermal titration calorimetry analysis, demonstrated that I3C directly interacts with purified NEDD4-1 protein. IMPLICATIONS This study identifies NEDD4-1 as a new I3C target protein, and that the I3C disruption of NEDD4-1 ubiquitination activity triggers the stabilization of the wild-type PTEN tumor suppressor to induce an antiproliferative response in melanoma. Mol Cancer Res; 12(11); 1621-34. ©2014 AACR.
Collapse
Affiliation(s)
- Ida Aronchik
- Department of Molecular and Cell Biology and The Cancer Research Laboratory, Univ. of California at Berkeley, Berkeley, CA 94720-3200
| | - Aishwarya Kundu
- Department of Molecular and Cell Biology and The Cancer Research Laboratory, Univ. of California at Berkeley, Berkeley, CA 94720-3200
| | - Jeanne G Quirit
- Department of Molecular and Cell Biology and The Cancer Research Laboratory, Univ. of California at Berkeley, Berkeley, CA 94720-3200
| | - Gary L Firestone
- Department of Molecular and Cell Biology and The Cancer Research Laboratory, Univ. of California at Berkeley, Berkeley, CA 94720-3200
| |
Collapse
|
42
|
Guo J, Chen XF, Liu J, Lin HY, Han HW, Liu HC, Huang SC, Shahla BK, Kulek A, Qi JL, Wang XM, Ling LJ, Yang YH. Novel Shikonin Derivatives Targeting Tubulin as Anticancer Agents. Chem Biol Drug Des 2014; 84:603-15. [DOI: 10.1111/cbdd.12353] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 03/20/2014] [Accepted: 04/22/2014] [Indexed: 12/15/2022]
Affiliation(s)
- Jing Guo
- State Key Laboratory of Pharmaceutical Biotechnology; School of Life Sciences; Nanjing University; Nanjing 210093 China
| | - Xiao-Feng Chen
- Department of oncology; the First Affiliated Hospital of Nanjing Medical University; Nanjing 210029 China
| | - Jing Liu
- State Key Laboratory of Pharmaceutical Biotechnology; School of Life Sciences; Nanjing University; Nanjing 210093 China
| | - Hong-Yan Lin
- State Key Laboratory of Pharmaceutical Biotechnology; School of Life Sciences; Nanjing University; Nanjing 210093 China
| | - Hong-Wei Han
- State Key Laboratory of Pharmaceutical Biotechnology; School of Life Sciences; Nanjing University; Nanjing 210093 China
| | - Hong-Chang Liu
- State Key Laboratory of Pharmaceutical Biotechnology; School of Life Sciences; Nanjing University; Nanjing 210093 China
| | - Shou-Cheng Huang
- State Key Laboratory of Pharmaceutical Biotechnology; School of Life Sciences; Nanjing University; Nanjing 210093 China
| | - Baloch K. Shahla
- State Key Laboratory of Pharmaceutical Biotechnology; School of Life Sciences; Nanjing University; Nanjing 210093 China
| | - Andrew Kulek
- Department of Emergency Medicine; Wayne State University School of Medicine; Detroit MI 48201 USA
| | - Jin-Liang Qi
- State Key Laboratory of Pharmaceutical Biotechnology; School of Life Sciences; Nanjing University; Nanjing 210093 China
| | - Xiao-Ming Wang
- State Key Laboratory of Pharmaceutical Biotechnology; School of Life Sciences; Nanjing University; Nanjing 210093 China
| | - Li-Jun Ling
- Department of Breast Surgery; the First Affiliated Hospital of Nanjing Medical University; Nanjing 210029 China
| | - Yong-Hua Yang
- State Key Laboratory of Pharmaceutical Biotechnology; School of Life Sciences; Nanjing University; Nanjing 210093 China
| |
Collapse
|
43
|
Subramanian C, Zhang H, Gallagher R, Hammer G, Timmermann B, Cohen M. Withanolides are Potent Novel Targeted Therapeutic Agents Against Adrenocortical Carcinomas. World J Surg 2014; 38:1343-52. [DOI: 10.1007/s00268-014-2532-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
44
|
Yu SY, Liao CH, Chien MH, Tsai TY, Lin JK, Weng MS. Induction of p21(Waf1/Cip1) by garcinol via downregulation of p38-MAPK signaling in p53-independent H1299 lung cancer. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:2085-2095. [PMID: 24533688 DOI: 10.1021/jf4037722] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Garcinol, a polyisoprenylated benzophenone, from Garcinia indica fruit rind has possessed anti-inflammatory, antioxidant, antiproliferation, and anticancer activities. However, the anticancer mechanisms of garcinol in lung cancer were still unclear. Therefore, we examine the effects of garcinol on antiproliferation in human lung cancer cells. Treatments with garcinol for 24 h exhibited morphological changes and inhibited the proliferation of H460 (p53-wild type) and H1299 (p53-null) cells in dose- and time-dependent manners. Furthermore, a significant G1 cell cycle arrest was observed in a dose-dependent treatment after H1299 cells were exposed in garcinol, whereas garcinol induced apoptosis rather than cell cycle arrest in H460 cells. Moreover, cyclin-dependent kinase 2 (CDK2), cyclin-dependent kinase 4 (CDK4), cyclin D1, and cyclin D3 were decreased, although cyclin E and cyclin-dependent kinase 6 (CDK6) were increased in garcinol-treated H1299 cells. Meanwhile, the protein levels of CDK inhibitors p21(Waf1/Cip1) and p27(KIP1) also exhibited upregulation after garcinol treatments. The enhanced protein-associated level between p21(Waf1/Cip1) and CDK4/2 rather than p27(KIP1) and CDK4/2 was demonstrated in garcinol-treated cells. Additionally, knock-down p21(Waf1/Cip1) by specific siRNA competently prevented garcinol-induced G1 arrest. Besides, garcinol also inhibited ERK and p38-MAPK activations in time-dependent mode. The pretreatment with p38-MAPK inhibitor but not ERK inhibitor raised garcinol-induced G1 population cells. Co-treatment with p38-MAPK inhibitor and garcinol synergistically elevated cyclin E, p21(Waf1/Cip1), and p27(Kip1) expressions. Meanwhile, overexpression dominant negative p38-MAPK also enhanced garcinol-induced p21(Waf1/Cip1) expression in H1299 cells. Accordingly, our data suggested that garcinol induced G1 cell cycle arrest and apoptosis in lung cancer cells under different p53 statuses. The p53-independent G1 cell cycle arrest induced by garcinol might be through upregulation of p21(Waf1/Cip1) triggered from p38-MAPK signaling inactivation.
Collapse
Affiliation(s)
- Sheng-Yung Yu
- Department of Nutritional Science, Fu Jen Catholic University , New Taipei City 24205, Taiwan
| | | | | | | | | | | |
Collapse
|
45
|
Hong M, Ren M, Silva J, Kennedy T, Choi J, Cowell JK, Hao Z. Sepantronium is a DNA damaging agent that synergizes with PLK1 inhibitor volasertib. Am J Cancer Res 2014; 4:135-147. [PMID: 24660103 PMCID: PMC3960451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 02/06/2014] [Indexed: 06/03/2023] Open
Abstract
In a search for novel agents that boost the anti-neoplastic effects of polo-like kinase 1 (PLK1) inhibitor volasertib, we found that a sepantronium and volasertib combination at the nano mole concentration potently inhibited growth of various non-small cell lung cancer (NSCLC) cell lines than either drug alone in vitro. Combination use of volasertib with sepantronium inhibited adaptation of cells to polo arrest. Addition of sepantronium to volasertib prevented accumulation of survivin and cyclin B protein at a concentration causing no appreciable survivin down regulation. Sepantronium induced cell cycle arrest from G1 or G2/M phase. Further studies demonstrated DNA damage of cancer cells when they are treated with sepantronium, which is evidenced by induction of phospho-γH2AX. In line with induction of a DNA damage response in cancer cells, known DNA damage response sensors and transducers ATM, ATR, CHK1, CHK2, p53 are phosphorylated following drug treatment. Meanwhile, expression of CDKN1A, BAX and XRCC5 are induced at the mRNA level as determined by quantitative real time PCR. A single cell electrophoresis assay (Comet assay) of cells treated with sepantronium revealed severe DNA strand breaks. M-phase arrest does not increase the lethality of DNA damage by sepantronium as compared to G1 phase arrest. Knock down of survivin did not cause DNA damage. Hence, sepantronium is a DNA damaging agent that synergizes with volasertib and down-regulation of survivin is likely the consequence of DNA damage induced by sepantronium.
Collapse
Affiliation(s)
- Mei Hong
- Cancer Center, Department of Internal Medicine, Medical College of Georgia, Georgia Regents UniversityAugusta, Georgia 30912
| | - Mingqiang Ren
- Cancer Center, Department of Internal Medicine, Medical College of Georgia, Georgia Regents UniversityAugusta, Georgia 30912
| | - Jeane Silva
- Cancer Center, Department of Internal Medicine, Medical College of Georgia, Georgia Regents UniversityAugusta, Georgia 30912
| | - Thomas Kennedy
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Medical College of Georgia, Georgia Regents UniversityAugusta, Georgia 30912
| | - Justin Choi
- Cancer Center, Department of Internal Medicine, Medical College of Georgia, Georgia Regents UniversityAugusta, Georgia 30912
| | - John K Cowell
- Cancer Center, Department of Internal Medicine, Medical College of Georgia, Georgia Regents UniversityAugusta, Georgia 30912
| | - Zhonglin Hao
- Cancer Center, Department of Internal Medicine, Medical College of Georgia, Georgia Regents UniversityAugusta, Georgia 30912
- Division of Hematology and Oncology, Department of Internal Medicine, Medical College of Georgia, Georgia Regents UniversityAugusta, Georgia 30912
| |
Collapse
|
46
|
Uzoigwe J, Sauter ER. Cancer prevention and treatment using combination therapy with plant- and animal-derived compounds. Expert Rev Clin Pharmacol 2014; 5:701-9. [DOI: 10.1586/ecp.12.62] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
47
|
Bao B, Azmi AS, Li Y, Ahmad A, Ali S, Banerjee S, Kong D, Sarkar FH. Targeting CSCs in tumor microenvironment: the potential role of ROS-associated miRNAs in tumor aggressiveness. Curr Stem Cell Res Ther 2014; 9:22-35. [PMID: 23957937 PMCID: PMC4493722 DOI: 10.2174/1574888x113089990053] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 08/01/2013] [Accepted: 08/06/2013] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) have been widely considered as critical cellular signaling molecules involving in various biological processes such as cell growth, differentiation, proliferation, apoptosis, and angiogenesis. The homeostasis of ROS is critical to maintain normal biological processes. Increased production of ROS, namely oxidative stress, due to either endogenous or exogenous sources causes irreversible damage of bio-molecules such as DNA, proteins, lipids, and sugars, leading to genomic instability, genetic mutation, and altered gene expression, eventually contributing to tumorigenesis. A great amount of experimental studies in vitro and in vivo have produced solid evidence supporting that oxidative stress is strongly associated with increased tumor cell growth, treatment resistance, and metastasis, and all of which contribute to tumor aggressiveness. More recently, the data have indicated that altered production of ROS is also associated with cancer stem cells (CSCs), epithelial-to-mesenchymal transition (EMT), and hypoxia, the most common features or phenomena in tumorigenesis and tumor progression. However, the exact mechanism by which ROS is involved in the regulation of CSC and EMT characteristics as well as hypoxia- and, especially, HIF-mediated pathways is not well known. Emerging evidence suggests the role of miRNAs in tumorigenesis and progression of human tumors. Recently, the data have indicated that altered productions of ROS are associated with deregulated expression of miRNAs, suggesting their potential roles in the regulation of ROS production. Therefore, targeting ROS mediated through the deregulation of miRNAs by novel approaches or by naturally occurring anti-oxidant agents such as genistein could provide a new therapeutic approach for the prevention and/or treatment of human malignancies. In this article, we will discuss the potential role of miRNAs in the regulation of ROS production during tumorigenesis. Finally, we will discuss the role of genistein, as a potent anti-tumor agent in the regulation of ROS production during tumorigenesis and tumor development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Fazlul H Sarkar
- Departments of Pathology and Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, 740 HWCRC, 4100 John R Street, Detroit, MI 48201, USA.
| |
Collapse
|
48
|
Jiang Y, Liang M, Svejkar D, Hart-Smith G, Lu H, Scarano W, Stenzel MH. Albumin-micelles via a one-pot technology platform for the delivery of drugs. Chem Commun (Camb) 2014; 50:6394-7. [PMID: 24811583 DOI: 10.1039/c4cc00616j] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Yanyan Jiang
- Centre for Advanced Macromolecular Design (CAMD), School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia.
| | | | | | | | | | | | | |
Collapse
|
49
|
YU MEILING, TONG XUHUI, QI BENQUAN, QU HONGDANG, DONG SHUYING, YU BINBIN, ZHANG NAIJU, TANG NAN, WANG LINGZHI, ZHANG CUILING. Berberine enhances chemosensitivity to irinotecan in colon cancer via inhibition of NF-κB. Mol Med Rep 2013; 9:249-54. [DOI: 10.3892/mmr.2013.1762] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 10/17/2013] [Indexed: 11/05/2022] Open
|
50
|
Bao B, Li Y, Ahmad A, Azmi AS, Bao G, Ali S, Banerjee S, Kong D, Sarkar FH. Targeting CSC-related miRNAs for cancer therapy by natural agents. Curr Drug Targets 2013; 13:1858-68. [PMID: 23140295 DOI: 10.2174/138945012804545515] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 10/09/2012] [Accepted: 11/03/2012] [Indexed: 12/22/2022]
Abstract
The theory of cancer stem cells (CSCs) has provided evidence on fundamental clinical implications because of the involvement of CSCs in cell migration, invasion, metastasis, and treatment resistance, which leads to the poor clinical outcome of cancer patients. Therefore, targeting CSCs will provide a novel therapeutic strategy for the treatment and/or prevention of tumors. However, the regulation of CSCs and its signaling pathways during tumorigenesis are not well understood. MicroRNAs (miRNAs) have been proved to act as key regulators of the post-transcriptional regulation of genes, which involve in a wide array of biological processes including tumorigenesis. The altered expressions of miRNAs are associated with poor clinical outcome of patients diagnosed with a variety of tumors. Therefore, emerging evidence strongly suggest that miRMAs play critical roles in tumor development and progression. Emerging evidence also suggest that miRNAs participate in the regulation of tumor cell growth, migration, invasion, angiogenesis, drug resistance, and metastasis. Moreover, miRNAs such as let-7, miR-21, miR-22, miR-34, miR-101, miR-146a, and miR-200 have been found to be associated with CSC phenotype and function mediated through targeting oncogenic signaling pathways. In this article, we will discuss the role of miRNAs in the regulation of CSC phenotype and function during tumor development and progression. We will also discuss the potential role of naturally occurring agents (nutraceuticals) as potent anti-tumor agents that are believed to function by targeting CSC-related miRNAs.
Collapse
Affiliation(s)
- Bin Bao
- Departments of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| | | | | | | | | | | | | | | | | |
Collapse
|