1
|
Al Amin M, Emran TB, Khan J, Zehravi M, Sharma I, Patil A, Gupta JK, Jeslin D, Krishnan K, Das R, Nainu F, Ahmad I, Wilairatana P. Research Progress of Indole Alkaloids: Targeting MAP Kinase Signaling Pathways in Cancer Treatment. Cancers (Basel) 2023; 15:5311. [PMID: 38001572 PMCID: PMC10670446 DOI: 10.3390/cancers15225311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 10/25/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer is the leading cause of morbidity and mortality in people throughout the world. There are many signaling pathways associated with cancerous diseases, from which the Mitogen-activated protein kinase (MAPK) pathway performs a significant role in this regard. Apoptosis and proliferation are correlated with MAPK signaling pathways. Plenty of experimental investigations were carried out to assess the role of indole alkaloids in MAPK-mediated cancerous diseases. Previous reports established that indole alkaloids, such as vincristine and evodiamine are useful small molecules in cancer treatment via the MAPK signaling system. Indole alkaloids have the anticancer potential through different pathways. Vincristine and evodiamine are naturally occurring indole alkaloids that have strong anticancer properties. Additionally, much research is ongoing or completed with molecules belonging to this group. The current review aims to evaluate how indole alkaloids affect the MAPK signaling pathway in cancer treatment. Additionally, we focused on the advancement in the role of indole alkaloids, with the intention of modifying the MAPK signaling pathways to investigate potential new anticancer small molecules. Furthermore, clinical trials with indole alkaloids in cancer treatment are also highlighted.
Collapse
Affiliation(s)
- Md. Al Amin
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh;
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh;
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, RI 02912, USA
| | - Jishan Khan
- Department of Pharmacy, International Islamic University Chittagong, Kumira, Chittagong 4318, Bangladesh
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry & Pharmacy, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia;
| | - Indu Sharma
- Department of Physics, Career Point University, Hamirpur 176041, Himachal Pradesh, India
| | - Anasuya Patil
- Department of Pharmaceutics, KLE College of Pharmacy, Bengaluru 560010, Karnataka, India
| | - Jeetendra Kumar Gupta
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura 281406, Uttar Pradesh, India;
| | - D. Jeslin
- Department of Pharmaceutics, Sree Balaji Medical College and Hospital Campus, Bharath Institute of Higher Education and Research, Chromepet, Chennai 600044, Tamil Nadu, India
| | - Karthickeyan Krishnan
- Department of Pharmacy Practice, School of Pharmaceutical Sciences, Vels Institute of Science, Technology and Advanced Studies (VISTAS), Pallavaram, Chennai 600117, Tamil Nadu, India;
| | - Rajib Das
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh
| | - Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Indonesia;
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 61411, Saudi Arabia
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
2
|
Muhammad N, Usmani D, Tarique M, Naz H, Ashraf M, Raliya R, Tabrez S, Zughaibi TA, Alsaieedi A, Hakeem IJ, Suhail M. The Role of Natural Products and Their Multitargeted Approach to Treat Solid Cancer. Cells 2022; 11:cells11142209. [PMID: 35883653 PMCID: PMC9318484 DOI: 10.3390/cells11142209] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/08/2022] [Accepted: 07/13/2022] [Indexed: 02/07/2023] Open
Abstract
Natural products play a critical role in the discovery and development of numerous drugs for the treatment of various types of cancer. These phytochemicals have demonstrated anti-carcinogenic properties by interfering with the initiation, development, and progression of cancer through altering various mechanisms such as cellular proliferation, differentiation, apoptosis, angiogenesis, and metastasis. Treating multifactorial diseases, such as cancer with agents targeting a single target, might lead to limited success and, in many cases, unsatisfactory outcomes. Various epidemiological studies have shown that the steady consumption of fruits and vegetables is intensely associated with a reduced risk of cancer. Since ancient period, plants, herbs, and other natural products have been used as healing agents. Likewise, most of the medicinal ingredients accessible today are originated from the natural resources. Regardless of achievements, developing bioactive compounds and drugs from natural products has remained challenging, in part because of the problem associated with large-scale sequestration and mechanistic understanding. With significant progress in the landscape of cancer therapy and the rising use of cutting-edge technologies, we may have come to a crossroads to review approaches to identify the potential natural products and investigate their therapeutic efficacy. In the present review, we summarize the recent developments in natural products-based cancer research and its application in generating novel systemic strategies with a focus on underlying molecular mechanisms in solid cancer.
Collapse
Affiliation(s)
- Naoshad Muhammad
- Department of Radiation Oncology, School of Medicine, Washington University, Saint Louis, MO 63130, USA;
| | | | - Mohammad Tarique
- Department of Child Health, University of Missouri, Columbia, MO 65211, USA;
| | - Huma Naz
- Department of Internal Medicine, University of Missouri, Columbia, MO 65211, USA;
| | - Mohammad Ashraf
- Department of Chemistry, Bundelkhand University Jhansi, Jhansi 284128, Uttar Pradesh, India;
| | - Ramesh Raliya
- IFFCO Nano Biotechnology Research Center, Kalol 382423, Gujarat, India;
| | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.T.); (T.A.Z.)
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Torki A. Zughaibi
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.T.); (T.A.Z.)
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Ahdab Alsaieedi
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Israa J. Hakeem
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah 21959, Saudi Arabia;
| | - Mohd Suhail
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.T.); (T.A.Z.)
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Correspondence:
| |
Collapse
|
3
|
Ben Sassi A, Ascrizzi R, Chiboub W, Cheikh Mhamed A, ElAyeb A, Skhiri F, Tounsi Saidani M, Mastouri M, Flamini G. Volatiles, phenolic compounds, antioxidant and antibacterial properties of kohlrabi leaves. Nat Prod Res 2021; 36:3143-3148. [PMID: 34154474 DOI: 10.1080/14786419.2021.1940177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
This work presents the volatile compounds and phenolic profile investigation of the leaves of Brassica oleracea L. growing in Tunisia, together with antioxidant and antibacterial properties. Volatile constituents were determined by HS-SPME coupled to GC/MS, and the results showed that α-pinene (31.6%) and limonene (16.9%) were the main volatiles. The phenolic profile was determined by HPLC analysis, the methanol extract revealed the presence of four hydroxycinnamic acids (chlorogenic, ferulic, p-coumaric and sinapic acids), two hydroxybenzoic acids (syringic and gallic acids), and four flavonoids (catechol, catechin hydrate, epigallocatechin and epicatechin 3-O-gallate). The methanol extract showed the best significantly antiradical activity by DPPH (2,2-diphenyl-1-picrylhydrazyl) and ABTS ((2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid)) antioxidant assays, with EC50 of 0.32 and 0.45 mg/mL, respectively. For antibacterial activity, the methanol extract inhibits all the tested strains. It can be concluded that kohlrabi leaves are rich in bioactive compounds and are a potential source of natural antioxidants and antibacterials.
Collapse
Affiliation(s)
- Ahlem Ben Sassi
- Laboratory of Transmissible Diseases and Biologically Active Substances, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | | | - Wiem Chiboub
- Laboratory of Genetics, Biodiversity and Valorisation of Bioresources, Higher Institute of Biotechnology, University of Monastir, Monastir, Tunisia
| | - Amina Cheikh Mhamed
- Laboratory of Transmissible Diseases and Biologically Active Substances, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Asma ElAyeb
- Laboratory of Genetics, Biodiversity and Valorisation of Bioresources, Higher Institute of Biotechnology, University of Monastir, Monastir, Tunisia
| | - Fethia Skhiri
- Laboratory of Genetics, Biodiversity and Valorisation of Bioresources, Higher Institute of Biotechnology, University of Monastir, Monastir, Tunisia
| | - Moufida Tounsi Saidani
- Biotechnology Center of Borj-cédria, Laboratory of Aromatic and medicinal plants, Monastir, Tunisia
| | - Maha Mastouri
- Laboratory of Transmissible Diseases and Biologically Active Substances, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Guido Flamini
- Department of Pharmacy, University of Pisa, Pisa, Italy
| |
Collapse
|
4
|
Mir RH, Mohi-ud-din R, Wani TU, Dar MO, Shah AJ, Lone B, Pooja C, Masoodi MH. Indole: A Privileged Heterocyclic Moiety in the Management of Cancer. CURR ORG CHEM 2021. [DOI: 10.2174/1385272825666210208142108] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Heterocyclic are a class of compounds that are intricately entwined into life processes.
Almost more than 90% of marketed drugs carry heterocycles. Synthetic chemistry, in
turn, allocates a cornucopia of heterocycles. Among the heterocycles, indole, a bicyclic structure
consisting of a six-membered benzene ring fused to a five-membered pyrrole ring with
numerous pharmacophores that generate a library of various lead molecules. Due to its profound
pharmacological profile, indole got wider attention around the globe to explore it fully
in the interest of mankind. The current review covers recent advancements on indole in the
design of various anti-cancer agents acting by targeting various enzymes or receptors, including
(HDACs), sirtuins, PIM kinases, DNA topoisomerases, and σ receptors.
Collapse
Affiliation(s)
- Reyaz Hassan Mir
- Pharmaceutical Chemistry Division, Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar-190006, Kashmir, India
| | - Roohi Mohi-ud-din
- Pharmacognosy Division, Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar, 190006, Kashmir, India
| | - Taha Umair Wani
- Pharmaceutics Lab, Department of Pharmaceutical Sciences, School of Applied Sciences and Technology, University of Kashmir, Hazratbal, Srinagar-190006, Kashmir, India
| | - Mohammad Ovais Dar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Mohali, Punjab, 160062, India
| | - Abdul Jaleel Shah
- Pharmaceutical Chemistry Division, Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar-190006, Kashmir, India
| | - Bashir Lone
- Natural Product Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-180001, India
| | - Chawla Pooja
- Department of Pharmaceutical Analysis, ISF College of Pharmacy, Moga-142001, India
| | - Mubashir Hussain Masoodi
- Pharmaceutical Chemistry Division, Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar-190006, Kashmir, India
| |
Collapse
|
5
|
Regulation of carcinogenesis and mediation through Wnt/β-catenin signaling by 3,3'-diindolylmethane in an enzalutamide-resistant prostate cancer cell line. Sci Rep 2021; 11:1239. [PMID: 33441906 PMCID: PMC7806813 DOI: 10.1038/s41598-020-80519-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 12/18/2020] [Indexed: 12/09/2022] Open
Abstract
Enzalutamide (ENZ) is an important drug used to treat castration-resistant prostate cancer (CRPC), which inhibits androgen receptor (AR) signaling. Previous study showed that 3,3′-diindolylmethane (DIM) is an AR antagonist that also inhibits Wnt signaling and epithelial-mesenchymal transition (EMT). To investigate whether combined treatment with ENZ and DIM can overcome ENZ resistance by regulating Wnt signaling to inhibit AR signaling and EMT in ENZ-resistant prostate cancer cells, 22Rv1 cells were cultured in normal medium and treated with ENZ, DIM, and DIM with ENZ. Exposure of ENZ-resistant cells to both DIM and ENZ significantly inhibited cell proliferation without cytotoxicity and invasion in comparison with the control. DIM significantly increased the E-cadherin expression and inhibited the expressions of Vimentin and Fibronectin, subsequently inhibiting EMT. Co-treatment with ENZ and DIM significantly increased the expressions of GSK3β and APC and decreased the β-catenin protein expression, causing inhibition of Wnt signaling and AR expression, it also significantly decreased the AR-v7 expression and down-regulated AR signaling. Via suppression of Wnt and AR signaling, co-treatment increased the E-cadherin and decreased the Vimentin and Fibronectin RNA and protein expressions, then inhibited EMT. Co-treatment with DIM and ENZ regulated Wnt signaling to reduce not only the AR expression, but also the AR-v7 expression, indicating suppression of EMT that inhibits cancer cell proliferation, invasion and migration to ameliorate ENZ resistance.
Collapse
|
6
|
Amare DE. Anti-Cancer and Other Biological Effects of a Dietary Compound 3,3ʹ-Diindolylmethane Supplementation: A Systematic Review of Human Clinical Trials . NUTRITION AND DIETARY SUPPLEMENTS 2020. [DOI: 10.2147/nds.s261577] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
7
|
Jiang Y, Fang Y, Ye Y, Xu X, Wang B, Gu J, Aschner M, Chen J, Lu R. Anti-Cancer Effects of 3, 3'-Diindolylmethane on Human Hepatocellular Carcinoma Cells Is Enhanced by Calcium Ionophore: The Role of Cytosolic Ca 2+ and p38 MAPK. Front Pharmacol 2019; 10:1167. [PMID: 31649538 PMCID: PMC6795059 DOI: 10.3389/fphar.2019.01167] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 09/11/2019] [Indexed: 12/19/2022] Open
Abstract
Purpose: 3,3'-Diindolylmethane (DIM), derived from indole-3-carbinol (I3C) in the Brassica species of cruciferous vegetables, has anticancer effects, but its exact underlying mechanism of action is unknown. We explored the roles of cytosolic free calcium ([Ca2+]i) and p38 MAPK in the anti-cancer effects of DIM in human hepatocellular carcinoma cells. Methods: Cell proliferation was measured with a Cell Counting Kit-8 (CCK-8) and the clonogenic formation assay. Cell apoptosis was examined by flow cytometric analysis and Hoechst dye staining. Cleaved-caspase3, cleaved-PARP, Bax, total, and phosphorylated p38 MAPK were assayed by western blotting. [Ca2+]i was measured with Fluo-3/AM by fluorescence microscopy. A23187, a calcium ionophore, was used to increase [Ca2+]i levels. Results: DIM inhibited cell proliferation in both SMMC-7721 and HepG2 cells in a concentration- and time-dependent manner. DIM also enhanced phosphorylation of p38 MAPK (p-p38), which was attenuated by SB203580. The proliferation inhibition and apoptosis induction by DIM were also blunted. In addition, DIM increased [Ca2+]i in HCC cells, and this effect was inhibited by the calcium chelator, BAPTA-AM, resulting in reduced p-p38 MAPK activation and apoptosis in DIM-treated cells, though the proliferation inhibition by DIM was unchanged. However, the DIM-induced cell proliferation inhibition and apoptosis were significantly enhanced by A23187, a selective calcium ionophore, which was attributed to exaggerated p-p38 MAPK. Conclusions: The calcium ionophore enhanced DIM-induced anti-cancer effects in hepatocellular carcinoma cells, secondary to [Ca2+]i-dependent activation of p38 MAPK. Treatment with a combination of DIM and calcium ionophore may offer a new approach to enhance the chemotherapeutic efficacy in liver cancer.
Collapse
Affiliation(s)
- Yuanyue Jiang
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China
- Department of Pathology, Kunshan Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Yanfei Fang
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China
- Department of Gastroenterology, The First People's Hospital of Taicang City, Taicang Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Ye
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xinming Xu
- Department of General Surgery, Affiliated Kunshan Hospital, Jiangsu University School of Medicine, Suzhou, China
| | - Bingfang Wang
- Department of Digestive Disease, Affiliated Kunshan Hospital, Jiangsu University School of Medicine, Suzhou, China
| | - Jie Gu
- Institute of Life Science, Jiangsu University, Zhenjiang, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Jian Chen
- Department of General Surgery, Affiliated Kunshan Hospital, Jiangsu University School of Medicine, Suzhou, China
| | - Rongzhu Lu
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China
- Center for Experimental Research, Affiliated Kunshan Hospital, Jiangsu University School of Medicine, Suzhou, China
| |
Collapse
|
8
|
Pal M, Hodge AM, Papa N, MacInnis RJ, Bassett JK, Bolton D, Davis ID, Millar J, English DR, Hopper JL, Severi G, Southey MC, Milne RL, Giles GG. Body size and dietary risk factors for aggressive prostate cancer: a case-control study. Cancer Causes Control 2019; 30:1301-1312. [PMID: 31552571 DOI: 10.1007/s10552-019-01234-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 09/14/2019] [Indexed: 12/24/2022]
Abstract
PURPOSE Diet and body size may affect the risk of aggressive prostate cancer (APC), but current evidence is inconclusive. METHODS A case-control study was conducted in men under 75 years of age recruited from urology practices in Victoria, Australia; 1,254 with APC and 818 controls for whom the presence of prostate cancer had been excluded by biopsy. Dietary intakes were assessed using a validated food frequency questionnaire. Multivariable unconditional logistic regression estimated odds ratios and confidence intervals for hypothesized risk factors, adjusting for age, family history of prostate cancer, country of birth, socioeconomic status, smoking, and other dietary factors. RESULTS Positive associations with APC (odds ratio, 95% confidence intervals, highest vs. lowest category or quintile) were observed for body mass index (1.34, 1.02-1.78, Ptrend = 0.04), and trouser size (1.54, 1.17-2.04, Ptrend = 0.001). Intakes of milk and all dairy products were inversely associated with APC risk (0.71, 9.53-0.96, Ptrend = 0.05, and 0.64, 0.48-0.87, Ptrend = 0.012, respectively), but there was little evidence of an association with other dietary variables (Ptrend > 0.05). CONCLUSIONS We confirmed previous evidence for a positive association between body size and risk of APC, and suggest that consumption of dairy products, and milk more specifically, is inversely associated with risk.
Collapse
Affiliation(s)
- Mikaela Pal
- Karolinska Institute, Stockholm, Sweden. .,Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia.
| | - Allison M Hodge
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC, Australia
| | - Nathan Papa
- Department of Surgery, Austin Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Robert J MacInnis
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC, Australia
| | - Julie K Bassett
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia
| | - Damien Bolton
- Department of Surgery, Austin Health, The University of Melbourne, Melbourne, VIC, Australia.,Olivia Newton-John Cancer and Wellness Centre, Austin Health, Heidelberg, VIC, Australia
| | - Ian D Davis
- Monash University, Melbourne, VIC, Australia.,Eastern Health, Box Hill, VIC, Australia
| | - Jeremy Millar
- Alfred Health Radiation Oncology, Alfred Hospital, Melbourne, VIC, Australia.,Department of Surgery, Central Clinical School, Monash University, The Alfred Centre, Melbourne, VIC, Australia
| | - Dallas R English
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC, Australia
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC, Australia
| | - Gianluca Severi
- Centre de Recherche en Épidémiologie et Santé des Populations (CESP, Inserm U1018), Facultés de Médecine, Université Paris-Saclay, UPS UVSQ, Gustave Roussy, Villejuif, France
| | - Melissa C Southey
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia.,Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia.,Genetic Epidemiology Laboratory, Department of Clinical Pathology, University of Melbourne, Parkville, VIC, Australia
| | - Roger L Milne
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC, Australia.,Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
| | - Graham G Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
9
|
Saitakis M, Dogniaux S, Goudot C, Bufi N, Asnacios S, Maurin M, Randriamampita C, Asnacios A, Hivroz C. Different TCR-induced T lymphocyte responses are potentiated by stiffness with variable sensitivity. eLife 2017; 6. [PMID: 28594327 PMCID: PMC5464771 DOI: 10.7554/elife.23190] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 05/07/2017] [Indexed: 12/26/2022] Open
Abstract
T cells are mechanosensitive but the effect of stiffness on their functions is still debated. We characterize herein how human primary CD4+ T cell functions are affected by stiffness within the physiological Young’s modulus range of 0.5 kPa to 100 kPa. Stiffness modulates T lymphocyte migration and morphological changes induced by TCR/CD3 triggering. Stiffness also increases TCR-induced immune system, metabolism and cell-cycle-related genes. Yet, upon TCR/CD3 stimulation, while cytokine production increases within a wide range of stiffness, from hundreds of Pa to hundreds of kPa, T cell metabolic properties and cell cycle progression are only increased by the highest stiffness tested (100 kPa). Finally, mechanical properties of adherent antigen-presenting cells modulate cytokine production by T cells. Together, these results reveal that T cells discriminate between the wide range of stiffness values found in the body and adapt their responses accordingly. DOI:http://dx.doi.org/10.7554/eLife.23190.001 Our immune system contains many cells that play various roles in defending the body against infection, cancer and other threats. For example, T cells constantly patrol the body ready to detect and respond to dangers. They do so by gathering cues from their surroundings, which can be specific chemical signals or physical properties such as the stiffness of tissues. Once the T cells are active they respond in several different ways including releasing hormones and dividing to produce more T cells. Tissue stiffness varies considerably between different organs. Furthermore, disease can lead to changes in tissue stiffness. For example, tissues become more rigid when they are inflamed. The stiffness and other physical properties of the surfaces that T cells interact with affect how the cells respond when they detect a threat, but few details are known about exactly how these cues tune T cell responses. Saitakis et al. studied how human T cells respond to artificial surfaces of varying stiffness that mimic the range found in the body. The experiments show that T cells that interact with stiff surfaces become more active than T cells that interact with softer surfaces. However, some responses are more sensitive to the stiffness of the surface than others. For example, the ability of the T cells to release hormones was affected by the whole range of stiffnesses tested in the experiments, whereas only very stiff surfaces stimulated the T cells to divide. These findings show that T cells can detect the stiffness of surfaces in the body and use this to adapt how they respond to threats. Future challenges will be to find out how T cells sense the physical properties of their surroundings and investigate whether cell and tissue stiffness affects immune responses in the body. This will help us to understand how T cells fight infections and other threats, and could be used to develop new ways of boosting these cells to fight cancer and other diseases. DOI:http://dx.doi.org/10.7554/eLife.23190.002
Collapse
Affiliation(s)
- Michael Saitakis
- Institut Curie Section Recherche, INSERM U932 & PSL Research University, Paris, France
| | - Stéphanie Dogniaux
- Institut Curie Section Recherche, INSERM U932 & PSL Research University, Paris, France
| | - Christel Goudot
- Institut Curie Section Recherche, INSERM U932 & PSL Research University, Paris, France
| | - Nathalie Bufi
- Laboratoire Matières et systèmes complexes, Université Paris-Diderot and CNRS, UMR 7057, Sorbonne Paris Cité, Paris, France
| | - Sophie Asnacios
- Laboratoire Matières et systèmes complexes, Université Paris-Diderot and CNRS, UMR 7057, Sorbonne Paris Cité, Paris, France.,Department of Physics, Sorbonne Universités, UPMC Université Paris, Paris, France
| | - Mathieu Maurin
- Institut Curie Section Recherche, INSERM U932 & PSL Research University, Paris, France
| | - Clotilde Randriamampita
- INSERM, U1016, Institut Cochin & UMR8104, CNRS & Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Atef Asnacios
- Laboratoire Matières et systèmes complexes, Université Paris-Diderot and CNRS, UMR 7057, Sorbonne Paris Cité, Paris, France
| | - Claire Hivroz
- Institut Curie Section Recherche, INSERM U932 & PSL Research University, Paris, France
| |
Collapse
|
10
|
Zhang X, Sukamporn P, Zhang S, Min KW, Baek SJ. 3,3'-diindolylmethane downregulates cyclin D1 through triggering endoplasmic reticulum stress in colorectal cancer cells. Oncol Rep 2017; 38:569-574. [PMID: 28586058 DOI: 10.3892/or.2017.5693] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 05/17/2017] [Indexed: 11/05/2022] Open
Abstract
As a major in vivo condensation product of indole-3-carbinol, which is mostly present in cruciferous vegetables, 3,3'-diindolylmethane (DIM) has been previously reported with anti-proliferative action in different types of cancer by our group and others. To further elucidate these underlying mechanisms, we examined the effect of DIM on cyclin D1, which was aberrantly overexpressed in various cancer cells and tumors. Herein, we found that DIM downregulated cyclin D1 expression in colorectal cancer cells (CRC), which was independent of PPARγ expression and protease activity. Furthermore, DIM did not affect cyclin D1 mRNA expression, suggesting DIM modulated cyclin D1 expression at the translational level. Subsequently, blocking eIF2α phosphorylation resulted from endoplasmic reticulum (ER) stress restored cyclin D1 in the presence of DIM. Thus, the present study demonstrates that DIM downregulates cyclin D1 through triggering ER stress in human colorectal cancer cells.
Collapse
Affiliation(s)
- Xiaobo Zhang
- Department of Basic Medical Science, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| | - Pakin Sukamporn
- Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | - Shiqiang Zhang
- Oncology Department, Shanghai TCM-Integrated Hospital, Shanghai 200082, P.R. China
| | - Kyung-Won Min
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Seung Joon Baek
- Laboratory of Signal Transduction, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
11
|
Wang W, Lv M, Wang Y, Zhang J. Development of novel application of 3,3'-diindolylmethane: sensitizing multidrug resistance human breast cancer cells to γ-irradiation. PHARMACEUTICAL BIOLOGY 2016; 54:3164-3168. [PMID: 27307186 DOI: 10.1080/13880209.2016.1192198] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 03/13/2016] [Accepted: 05/16/2016] [Indexed: 06/06/2023]
Abstract
CONTEXT Multidrug resistance (MDR) is known as a major obstacle to effective cancer therapy. The effects of irradiation on MDR in cancer cells had rarely been reported. OBJECTIVE The effect of 3,3'-diindolylmethane (DIM) sensitizing MDR human breast carcinoma to γ-irradiation was investigated. MATERIALS AND METHODS MCF-7/ADR cells were exposed to different concentrations of DIM (0-30 μM) for 48 or 2 h before IR (γ-Co60, 10 Gy, room temperature) then cultured for 48 h. Cell survival was determined by MTT assay. Intracellular reactive oxygen spices (ROS) induced by DIM (20 and 30 μM, 2 h before irradiation) was measured by flow cytometry. Propidium iodide staining assay was used for cell cycle distribution studies; cell apoptosis was measured by flow cytometry and confocal microscopy. RESULTS DIM (20 and 30 μM, 2 h before irradiation) sensitized MCF-7/ADR cells to IR with survival rates decreased from 100% to 79% and 63%, respectively. DIM combined with γ-radiation demonstrated that the activity of G2/M phase cell cycle arresting with percentages enhanced from 9% to 49% and 52%. DIM can increase intracellular ROS generation by 1.45- and 1.55-times compared to control group. Significantly enhanced radiation-induced apoptosis by DIM was also observed. DISCUSSION AND CONCLUSION These data provide a rationale for the use of DIM as a promising radio-sensitizer to MDR cancer cells.
Collapse
Affiliation(s)
- Wenjing Wang
- a Department of Blood Biopharmaceuticals and Viral Detection , Institute of Transfusion Medicine, The Academy of Military Medical Sciences , Beijing , P.R. China
| | - Maomin Lv
- a Department of Blood Biopharmaceuticals and Viral Detection , Institute of Transfusion Medicine, The Academy of Military Medical Sciences , Beijing , P.R. China
| | - Yanlin Wang
- a Department of Blood Biopharmaceuticals and Viral Detection , Institute of Transfusion Medicine, The Academy of Military Medical Sciences , Beijing , P.R. China
| | - Jingang Zhang
- a Department of Blood Biopharmaceuticals and Viral Detection , Institute of Transfusion Medicine, The Academy of Military Medical Sciences , Beijing , P.R. China
| |
Collapse
|
12
|
Kim SM. Cellular and Molecular Mechanisms of 3,3'-Diindolylmethane in Gastrointestinal Cancer. Int J Mol Sci 2016; 17:ijms17071155. [PMID: 27447608 PMCID: PMC4964527 DOI: 10.3390/ijms17071155] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 07/11/2016] [Accepted: 07/12/2016] [Indexed: 12/18/2022] Open
Abstract
Studies in humans have shown that 3,3′-diindolylmethane (DIM), which is found in cruciferous vegetables, such as cabbage and broccoli, is effective in the attenuation of gastrointestinal cancers. This review presents the latest findings on the use, targets, and modes of action of DIM for the treatment of human gastrointestinal cancers. DIM acts upon several cellular and molecular processes in gastrointestinal cancer cells, including apoptosis, autophagy, invasion, cell cycle regulation, metastasis, angiogenesis, and endoplasmic reticulum (ER) stress. In addition, DIM increases the efficacy of other drugs or therapeutic chemicals when used in combinatorial treatment for gastrointestinal cancer. The studies to date offer strong evidence to support the use of DIM as an anticancer and therapeutic agent for gastrointestinal cancer. Therefore, this review provides a comprehensive understanding of the preventive and therapeutic properties of DIM in addition to its different perspective on the safety of DIM in clinical applications for the treatment of gastrointestinal cancers.
Collapse
Affiliation(s)
- Soo Mi Kim
- Department of Physiology, Chonbuk National University Medical School, Jeonju 561-180, Korea.
| |
Collapse
|
13
|
Ye Y, Fang Y, Xu W, Wang Q, Zhou J, Lu R. 3,3'-Diindolylmethane induces anti-human gastric cancer cells by the miR-30e-ATG5 modulating autophagy. Biochem Pharmacol 2016; 115:77-84. [PMID: 27372603 DOI: 10.1016/j.bcp.2016.06.018] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 06/28/2016] [Indexed: 12/12/2022]
Abstract
3,3'-Diindolylmethane (DIM), a class of relatively non-toxic indole derivatives from cruciferous vegetables, has been reported as a promising anticancer phytochemical, but the underlying molecular mechanism is not completely elucidated. In the present study we report a novel regulation of autophagy by DIM in human gastric cancer cells. We found that DIM dose-dependently inhibited the growth of gastric cancer cells in vitro and in vivo. Moreover, ATG5 and LC3 were activated by DIM in gastric cancer cells. Furthermore, miR-30e was down-regulated by DIM and miR-30e targeted the 3'-UTR of ATG5 to inhibit its translation. Overall, these results suggest that DIM may through the miR-30e-ATG5 modulating autophagy inhibit the proliferation of gastric cancer cells.
Collapse
Affiliation(s)
- Yang Ye
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yanfei Fang
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China; Department of Digestive Diseases, Affiliated Kunshan Hospital, Jiangsu University, Kunshan, Suzhou, Jiangsu, China
| | - Wenxia Xu
- Labortaory of Cancer Biology, Key Laboratory of Biotherapy in Zhejiang, Sir Runrun Shaw Hospital, Medical School of Zhejiang University, Zhejiang, China
| | - Qiang Wang
- Department of Molecular Cell Biology and Toxicology, Cancer Center, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jianwei Zhou
- Department of Molecular Cell Biology and Toxicology, Cancer Center, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rongzhu Lu
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China; Center for Experimental Research, Affiliated Kunshan Hospital, Jiangsu University, Kunshan, Suzhou, Jiangsu, China.
| |
Collapse
|
14
|
Zhu Y, Zhang B, Gong A, Fu H, Zhang X, Shi H, Sun Y, Wu L, Pan Z, Mao F, Zhu W, Qian H, Xu W. Anti-cancer drug 3,3'-diindolylmethane activates Wnt4 signaling to enhance gastric cancer cell stemness and tumorigenesis. Oncotarget 2016; 7:16311-16324. [PMID: 26918831 PMCID: PMC4941316 DOI: 10.18632/oncotarget.7684] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 02/05/2016] [Indexed: 12/27/2022] Open
Abstract
As a natural health supplement, 3,3'-diindolylmethane (DIM) is proposed as a preventive and chemotherapeutic agent for cancer by inhibiting cell proliferation and inducing cell apoptosis. However, we found that in contrary to high level of DIM (30 μM), low level of DIM (1 μM and 10 μM) obviously promoted gastric cancer cell growth and migration. In addition, we found that low level of DIM increased the expression of stemness factors and enhanced the pluripotency of gastric cancer cells. Low level of DIM promoted gastric cancer progression by inducing the PORCN-dependent secretion of Wnt4 and the activation of β-catenin signaling. Wnt4 knockdown reversed the effects of low level of DIM on gastric cancer cells. The results of in vivo studies showed that gastric cancer cells treated with low level of DIM (1 μM) grew faster and expressed higher level of Wnt4 than control cells. Taken together, our findings indicate that low level of DIM activates autocrine Wnt4 signaling to enhance the progression of gastric cancer, which may suggest an adverse aspect of DIM in cancer therapy. Our findings will provide a new aspect for the safety of DIM in its clinical application.
Collapse
Affiliation(s)
- Yanhua Zhu
- Key Laboratory of Laboratory Medicine of Jiangsu Province, The Affiliated Hospital, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P. R. China
- Department of Clinical Laboratory, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan, P. R. China
| | - Bin Zhang
- Key Laboratory of Laboratory Medicine of Jiangsu Province, The Affiliated Hospital, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Aihua Gong
- Key Laboratory of Laboratory Medicine of Jiangsu Province, The Affiliated Hospital, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Hailong Fu
- Key Laboratory of Laboratory Medicine of Jiangsu Province, The Affiliated Hospital, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Xu Zhang
- Key Laboratory of Laboratory Medicine of Jiangsu Province, The Affiliated Hospital, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Hui Shi
- Key Laboratory of Laboratory Medicine of Jiangsu Province, The Affiliated Hospital, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Yaoxiang Sun
- Key Laboratory of Laboratory Medicine of Jiangsu Province, The Affiliated Hospital, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Lijun Wu
- Key Laboratory of Laboratory Medicine of Jiangsu Province, The Affiliated Hospital, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Zhaoji Pan
- Key Laboratory of Laboratory Medicine of Jiangsu Province, The Affiliated Hospital, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Fei Mao
- Key Laboratory of Laboratory Medicine of Jiangsu Province, The Affiliated Hospital, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Wei Zhu
- Key Laboratory of Laboratory Medicine of Jiangsu Province, The Affiliated Hospital, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Hui Qian
- Key Laboratory of Laboratory Medicine of Jiangsu Province, The Affiliated Hospital, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Wenrong Xu
- Key Laboratory of Laboratory Medicine of Jiangsu Province, The Affiliated Hospital, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| |
Collapse
|
15
|
Hwang C, Sethi S, Heilbrun LK, Gupta NS, Chitale DA, Sakr WA, Menon M, Peabody JO, Smith DW, Sarkar FH, Heath EI. Anti-androgenic activity of absorption-enhanced 3, 3'-diindolylmethane in prostatectomy patients. Am J Transl Res 2016; 8:166-176. [PMID: 27069550 PMCID: PMC4759426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 10/11/2015] [Indexed: 06/05/2023]
Abstract
Consumption of cruciferous vegetables is associated with a decreased risk of developing prostate cancer. Antineoplastic effects of cruciferous vegetables are attributable to bioactive indoles, most prominently, 3, 3'-diindolylmethane (DIM). In addition to effects on proliferation and apoptosis, DIM acts as an antiandrogen in prostate cancer cell lines. This study characterized the effects of prostatic DIM on the androgen receptor (AR) in patients with prostate cancer. Men with localized prostate cancer were treated with a specially formulated DIM capsule designed for enhanced bioavailability (BR-DIM) at a dose of 225 mg orally twice daily for a minimum of 14 days. DIM levels and AR activity were assessed at the time of prostatectomy. Out of 28 evaluable patients, 26 (93%) had detectable prostatic DIM levels, with a mean concentration of 14.2 ng/gm. The mean DIM plasma level on BR-DIM therapy was 9.0 ng/mL; levels were undetectable at baseline and in follow-up samples. AR localization in the prostate was assessed with immunohistochemistry. After BR-DIM therapy, 96% of patients exhibited exclusion of the AR from the cell nucleus. In contrast, in prostate biopsy samples obtained prior to BR-DIM therapy, no patient exhibited AR nuclear exclusion. Declines in PSA were observed in a majority of patients (71%). Compliance was excellent and toxicity was minimal. In summary, BR-DIM treatment resulted in reliable prostatic DIM levels and anti-androgenic biologic effects at well tolerated doses. These results support further investigation of BR-DIM as a chemopreventive and therapeutic agent in prostate cancer.
Collapse
Affiliation(s)
- Clara Hwang
- Department of Hematology/Oncology, Josephine Ford Cancer Institute, Henry Ford Health SystemDetroit, MI, USA
| | - Seema Sethi
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, MI, USA
| | - Lance K Heilbrun
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, MI, USA
| | - Nilesh S Gupta
- Department of Pathology, Josephine Ford Cancer Institute, Henry Ford Health SystemDetroit, MI, USA
| | - Dhananjay A Chitale
- Department of Pathology, Josephine Ford Cancer Institute, Henry Ford Health SystemDetroit, MI, USA
| | - Wael A Sakr
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, MI, USA
| | - Mani Menon
- Department of Vattikuti Institute of Urology, Josephine Ford Cancer Institute, Henry Ford Health SystemDetroit, MI, USA
| | - James O Peabody
- Department of Vattikuti Institute of Urology, Josephine Ford Cancer Institute, Henry Ford Health SystemDetroit, MI, USA
| | - Daryn W Smith
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, MI, USA
| | - Fazlul H Sarkar
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, MI, USA
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, MI, USA
| | - Elisabeth I Heath
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, MI, USA
| |
Collapse
|
16
|
Li F, Chen C, Chen SM, Xiao BK, Tao ZZ. ERK signaling mediates long-term low concentration 3,3'-diindolylmethane inhibited nasopharyngeal carcinoma growth and metastasis: An in vitro and in vivo study. Oncol Rep 2015; 35:955-61. [PMID: 26574660 DOI: 10.3892/or.2015.4428] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 10/13/2015] [Indexed: 11/05/2022] Open
Abstract
It is well known that crucifers have antitumor effects and 3,3'-diindolylmethane (DIM) is one of the major bioactive components, and the associated molecular mechanisms in a short-term high-dose manner are widely discussed. However, the antitumor effects of DIM in a long-term low-dose manner in nasopharyngeal carcinoma (NPC) has not been reported yet, as to the potential mechanisms in the human body. In the present study, NPC cells were induced by 20 µmol/l DIM for over a month, and the proliferation, apoptosis, migration and in vivo metastasis were investigated. The results showed that DIM significantly reduced the proliferation and migration; however, changes in apoptosis were not observed. In vivo study showed the metastasis was significantly reduced. Compared to the short-term high-dose manner, incomplete similar qualities were observed; next we explored the possible signal pathway revolved, the ERK signaling showed similar changes, while the PI3K/Akt, NF-κB, P38, JNK pathways were significantly altered in the short-term high-dose manner (our previous study) showed no obvious change, indicating the ERK signaling may be the main effector of DIM.
Collapse
Affiliation(s)
- Fen Li
- Otolaryngology-Head and Neck Surgery Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Chen Chen
- Otolaryngology-Head and Neck Surgery Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Shi-Ming Chen
- Otolaryngology-Head and Neck Surgery Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Bo-Kui Xiao
- Otolaryngology-Head and Neck Surgery Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Ze-Zhang Tao
- Otolaryngology-Head and Neck Surgery Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
17
|
3,3'-Diindolylmethane: A Promising Sensitizer of γ-Irradiation. BIOMED RESEARCH INTERNATIONAL 2015; 2015:465105. [PMID: 26579534 PMCID: PMC4633530 DOI: 10.1155/2015/465105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 10/08/2015] [Accepted: 10/11/2015] [Indexed: 11/23/2022]
Abstract
Purpose. Radiotherapy is an effective treatment modality in the clinical treatment of breast cancer. The present work investigated the effect of 3,3′-diindolylmethane (DIM) on γ-irradiation sensitizing human breast carcinoma. Methods. Cell survival, intracellular ROS levels, cell cycle distribution, cell apoptosis, and expression of proteins related to apoptosis were measured with MTT assays, flow cytometry, and Western blot analysis, respectively. Results. In vitro DIM plus γ-irradiation arrested the activity of G2/M phase cell cycle, increased intracellular ROS level, significantly suppressed PARP (poly ADP-ribose polymerase), and enhanced γ-irradiation-induced apoptosis, thereby inhibiting the proliferation of MCF-7 cells. Conclusion. These data provide a rationale for the use of DIM as a promising sensitizer of γ-irradiation.
Collapse
|
18
|
Popovics P, Frigo DE, Schally AV, Rick FG. Targeting the 5'-AMP-activated protein kinase and related metabolic pathways for the treatment of prostate cancer. Expert Opin Ther Targets 2015; 19:617-32. [PMID: 25600663 DOI: 10.1517/14728222.2015.1005603] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Increasing evidence suggests that prostate cancer cells undergo unique metabolic reprogramming during transformation. A master regulator of cellular homeostasis, 5'-AMP-activated protein kinase (AMPK), directs metabolic adaptation that supports the growth demands of rapidly dividing cancer cells. The utilization of AMPK as a therapeutic target may therefore provide an effective strategy in the treatment of prostate cancer. AREAS COVERED Our review describes the regulation of AMPK by androgens and upstream kinases including the calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2) in prostate cancer. Oncogenic, AMPK-regulated pathways that direct various metabolic processes are also addressed. Furthermore, we discuss the role of AMPK in growth arrest and autophagy as a potential survival pathway for cancer cells. In addition, by regulating non-metabolic pathways, AMPK may stimulate migration and mitosis. Finally, this review summarizes efforts to treat prostate cancer with pharmacological agents capable of modulating AMPK signaling. EXPERT OPINION Current research is primarily focused on developing drugs that activate AMPK as a treatment for prostate cancer. However, oncogenic aspects of AMPK signaling calls for caution about employing such therapies. We think that inhibitors of CaMKK2 or AMPK, or perhaps the modulation of downstream targets of AMPK, will gain importance in the clinical management of prostate cancer.
Collapse
Affiliation(s)
- Petra Popovics
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education , Research (151) 2A127, 1201 NW 16th St, Miami, FL 33125 , USA +1 305 5753477 ; +1 305 5753126 ;
| | | | | | | |
Collapse
|
19
|
Pondugula SR, Flannery PC, Abbott KL, Coleman ES, Mani S, Samuel T, Xie W. Diindolylmethane, a naturally occurring compound, induces CYP3A4 and MDR1 gene expression by activating human PXR. Toxicol Lett 2014; 232:580-9. [PMID: 25542144 DOI: 10.1016/j.toxlet.2014.12.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 12/08/2014] [Accepted: 12/20/2014] [Indexed: 11/24/2022]
Abstract
Activation of human pregnane X receptor (hPXR)-regulated expression of cytochrome P450 3A4 (CYP3A4) and multidrug resistance protein 1 (MDR1) plays an important role in mediating adverse drug interactions. Given the common use of natural products as part of adjunct human health behavior, there is a growing concern about natural products for their potential to induce undesired drug interactions through the activation of hPXR-regulated CYP3A4 and MDR1. Here, we studied whether 3,3'-diindolylmethane (DIM), a natural health supplement, could induce hPXR-mediated regulation of CYP3A4 and MDR1 in human hepatocytes and intestinal cells. DIM, at its physiologically relevant concentrations, not only induced hPXR transactivation of CYP3A4 promoter activity but also induced gene expression of CYP3A4 and MDR1. DIM decreased intracellular accumulation of MDR1 substrate rhodamine 123, suggesting that DIM induces the functional expression of MDR1. Pharmacologic inhibition or genetic knockdown of hPXR resulted in attenuation of DIM induced CYP3A4 and MDR1 gene expression, suggesting that DIM induces CYP3A4 and MDR1 in an hPXR-dependent manner. Together, these results support our conclusion that DIM induces hPXR-regulated CYP3A4 and MDR1 gene expression. The inductive effects of DIM on CYP3A4 and MDR1 expression caution the use of DIM in conjunction with other medications metabolized and transported via CYP3A4 and MDR1, respectively.
Collapse
Affiliation(s)
- Satyanarayana R Pondugula
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, AL, United States; Auburn University Research Initiative in Cancer, Auburn University, Auburn, AL, United States.
| | - Patrick C Flannery
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, AL, United States; Auburn University Research Initiative in Cancer, Auburn University, Auburn, AL, United States
| | - Kodye L Abbott
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, AL, United States; Auburn University Research Initiative in Cancer, Auburn University, Auburn, AL, United States
| | - Elaine S Coleman
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, AL, United States
| | - Sridhar Mani
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York, NY, United States
| | - Temesgen Samuel
- Department of Pathobiology, College of Veterinary Medicine, Nursing and Allied Health, Tuskegee University, AL, United States
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
20
|
Prabhu B, Padma R, Alwin D, Pazhanivel N, Balakrishnan D, Sundaresan S. Protective Effect of Diindolylmethane against N-Butyl-N-(4-hydroxybutyl) Nitrosamine-induced Bladder Carcinogenesis. ACTA ACUST UNITED AC 2014. [DOI: 10.1016/j.jecm.2014.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
21
|
Marques M, Laflamme L, Benassou I, Cissokho C, Guillemette B, Gaudreau L. Low levels of 3,3'-diindolylmethane activate estrogen receptor α and induce proliferation of breast cancer cells in the absence of estradiol. BMC Cancer 2014; 14:524. [PMID: 25048790 PMCID: PMC4223525 DOI: 10.1186/1471-2407-14-524] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 07/08/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND 3,3'-diindolylmethane (DIM) is an acid-catalyzed dimer of idole-3-carbinol (I3C), a phytochemical found in cruciferous vegetables that include broccoli, Brussels sprouts and cabbage. DIM is an aryl hydrocarbon receptor (AhR) ligand and a potential anticancer agent, namely for the treatment of breast cancer. It is also advertised as a compound that regulates sex hormone homeostasis. METHODS Here we make use of RNA expression assays coupled to Chromatin Immunoprecipitation (ChIP) in breast cancer cell lines to study the effect of DIM on estrogen signaling. We further make use of growth assays, as well as fluorescence-activated cell sorting (FACS) assays, to monitor cell growth. RESULTS In this study, we report that 'physiologically obtainable' concentrations of DIM (10 μM) activate the estrogen receptor α (ERα) signaling pathway in the human breast cancer cell lines MCF7 and T47D, in a 17β-estradiol (E2)-independent manner. Accordingly, we observe induction of ERα target genes such as GREB1 and TFF1, and an increase in cellular proliferation after treatment with 10 μM DIM in the absence of E2. By using an ERα specific inhibitor (ICI 182 780), we confirm that the transcriptional and proliferative effects of DIM treatment are mediated by ERα. We further show that the protein kinase A signaling pathway participates in DIM-mediated activation of ERα. In contrast, higher concentrations of DIM (e.g. 50 μM) have an opposite and expected effect on cells, which is to inhibit proliferation. CONCLUSIONS We document an unexpected effect of DIM on cell proliferation, which is to stimulate growth by inducing the ERα signaling pathway. Importantly, this proliferative effect of DIM happens with potentially physiological concentrations that can be provided by the diet or by taking caplet supplements.
Collapse
Affiliation(s)
| | | | | | | | | | - Luc Gaudreau
- Département de Biologie, Université de Sherbrooke, J1K 2R1 Sherbrooke, QC, Canada.
| |
Collapse
|
22
|
Zhang WW, Feng Z, Narod SA. Multiple therapeutic and preventive effects of 3,3'-diindolylmethane on cancers including prostate cancer and high grade prostatic intraepithelial neoplasia. J Biomed Res 2014; 28:339-48. [PMID: 25332705 PMCID: PMC4197384 DOI: 10.7555/jbr.28.20140008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 02/07/2014] [Accepted: 02/22/2014] [Indexed: 12/20/2022] Open
Abstract
Cruciferous vegetables belong to the plant family that has flowers with four equal-sized petals in the pattern of a crucifer cross. These vegetables are an abundant source of dietary phytochemicals, including glucosinolates and their hydrolysis products such as indole-3-carbinol (I3C) and 3,3′-diindolylmethane (DIM). By 2013, the total number of natural glucosinolates that have been documented is estimated to be 132. Recently, cruciferous vegetable intake has garnered great interest for its multiple health benefits such as anticancer, antiviral infections, human sex hormone regulation, and its therapeutic and preventive effects on prostate cancer and high grade prostatic intraepithelial neoplasia (HGPIN). DIM is a hydrolysis product of glucosinolates and has been used in various trials. This review is to provide an insight into the latest developments of DIM in treating or preventing both prostate cancer and HGPIN.
Collapse
Affiliation(s)
- William Weiben Zhang
- Division of Urology, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario M4N 3M5, Canada
| | - Zhenqing Feng
- Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Steven A Narod
- Department of Public Health Sciences, Women's College Hospital, Women's College Research Institute, University of Toronto, Toronto, Ontario M4N 3M5, Canada
| |
Collapse
|
23
|
Lee BH, Ryu PD, Lee SY. Serum starvation-induced voltage-gated potassium channel Kv7.5 expression and its regulation by Sp1 in canine osteosarcoma cells. Int J Mol Sci 2014; 15:977-93. [PMID: 24434641 PMCID: PMC3907850 DOI: 10.3390/ijms15010977] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 12/20/2013] [Accepted: 12/30/2013] [Indexed: 01/03/2023] Open
Abstract
The KCNQ gene family, whose members encode Kv7 channels, belongs to the voltage-gated potassium (Kv) channel group. The roles of this gene family have been widely investigated in nerve and muscle cells. In the present study, we investigated several characteristics of Kv7.5, which is strongly expressed in the canine osteosarcoma cell line, CCL-183. Serum starvation upregulated Kv7.5 expression, and the Kv7 channel opener, flupirtine, attenuated cell proliferation by arresting cells in the G0/G1 phase. We also showed that Kv7.5 knockdown helps CCL-183 cells to proliferate. In an effort to find an endogenous regulator of Kv7.5, we used mithramycin A to reduce the level of the transcription factor Sp1, and it strongly inhibited the induction of Kv7.5 in CCL-183 cells. These results suggest that the activation of Kv7.5 by flupirtine may exert an anti-proliferative effect in canine osteosarcoma. Therefore, Kv7.5 is a possible molecular target for canine osteosarcoma therapy.
Collapse
Affiliation(s)
- Bo Hyung Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea.
| | - Pan Dong Ryu
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea.
| | - So Yeong Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea.
| |
Collapse
|
24
|
Nicastro HL, Firestone GL, Bjeldanes LF. 3,3'-diindolylmethane rapidly and selectively inhibits hepatocyte growth factor/c-Met signaling in breast cancer cells. J Nutr Biochem 2013; 24:1882-8. [PMID: 23968581 DOI: 10.1016/j.jnutbio.2013.05.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 01/31/2013] [Accepted: 05/04/2013] [Indexed: 10/26/2022]
Abstract
3,3'-Diindolylmethane (DIM), an indole derivative from vegetables of the Brassica genus, has antiproliferative activity in breast cancer cells. Part of this activity is thought to be due to DIM inhibition of Akt signaling, but an upstream mechanism of DIM-induced Akt inhibition has not been described. The goals of this study were to investigate the kinetics of inhibition of Akt by physiologically relevant concentrations of DIM and to identify an upstream factor that mediates this effect. Here we report that DIM (5-25 μM) inhibited Akt activation from 30 min to 24h in tumorigenic MDA-MB-231 cells but did not inhibit Akt activation in non-tumorigenic preneoplastic MCF10AT cells. DIM inhibited hepatocyte growth factor (HGF)-induced Akt activation by up to 46%, cell migration by 66% and cell proliferation by up to 54%, but did not inhibit induction of Akt by epidermal growth factor or insulin-like growth factor-1. DIM decreased phosphorylation of the HGF receptor, c-Met, at tyrosines 1234 and 1235, indicating decreased activation of the receptor. This decrease was reversed by pretreatment with inhibitors of p38 or calcineurin. Our results demonstrate the important role of HGF and c-Met in DIM's anti-proliferative effect on breast cancer cells and suggest that DIM could have preventive or clinical value as an inhibitor of c-Met signaling.
Collapse
Affiliation(s)
- Holly L Nicastro
- Department of Nutritional Science & Toxicology, University of California Berkeley, Berkeley, CA 94720-3104.
| | | | | |
Collapse
|
25
|
W Watson G, M Beaver L, E Williams D, H Dashwood R, Ho E. Phytochemicals from cruciferous vegetables, epigenetics, and prostate cancer prevention. AAPS JOURNAL 2013; 15:951-61. [PMID: 23800833 DOI: 10.1208/s12248-013-9504-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 06/11/2013] [Indexed: 12/21/2022]
Abstract
Epidemiological evidence has demonstrated a reduced risk of prostate cancer associated with cruciferous vegetable intake. Follow-up studies have attributed this protective activity to the metabolic products of glucosinolates, a class of secondary metabolites produced by crucifers. The metabolic products of glucoraphanin and glucobrassicin, sulforaphane, and indole-3-carbinol respectively, have been the subject of intense investigation by cancer researchers. Sulforaphane and indole-3-carbinol inhibit prostate cancer by both blocking initiation and suppressing prostate cancer progression in vitro and in vivo. Research has largely focused on the anti-initiation and cytoprotective effects of sulforaphane and indole-3-carbinol through induction of phases I and II detoxification pathways. With regards to suppressive activity, research has focused on the ability of sulforaphane and indole-3-carbinol to antagonize cell signaling pathways known to be dysregulated in prostate cancer. Recent investigations have characterized the ability of sulforaphane and indole-3-carbinol derivatives to modulate the activity of enzymes controlling the epigenetic status of prostate cancer cells. In this review, we will summarize the well-established, "classic" non-epigenetic targets of sulforaphane and indole-3-carbinol, and highlight more recent evidence supporting these phytochemicals as epigenetic modulators for prostate cancer chemoprevention.
Collapse
Affiliation(s)
- Gregory W Watson
- Molecular and Cellular Biology, Oregon State University, Corvallis, Oregon, 97331, USA
| | | | | | | | | |
Collapse
|
26
|
Chen C, Chen SM, Xu B, Chen Z, Wang F, Ren J, Xu Y, Wang Y, Xiao BK, Tao ZZ. In vivo and in vitro study on the role of 3,3'-diindolylmethane in treatment and prevention of nasopharyngeal carcinoma. Carcinogenesis 2013; 34:1815-21. [DOI: 10.1093/carcin/bgt122] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
27
|
Gupta G, Chaudhari G, Tomar P, Gaikwad Y, Azad R, Pandya G, Waghulde G, Patil K. Synthesis of bis(indolyl)methanes using molten N-butylpyridinium bromide. ACTA ACUST UNITED AC 2012. [DOI: 10.5155/eurjchem.3.4.475-479.709] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
28
|
Liriodenine induces G1/S cell cycle arrest in human colon cancer cells via nitric oxide- and p53-mediated pathway. Process Biochem 2012. [DOI: 10.1016/j.procbio.2012.05.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
29
|
Tsai JY, Chou CT, Liu SI, Liang WZ, Kuo CC, Liao WC, Lin KL, Hsu SS, Lu YC, Huang JK, Jan CR. Effect of diindolylmethane on Ca2+ homeostasis and viability in PC3 human prostate cancer cells. J Recept Signal Transduct Res 2012; 32:271-8. [PMID: 22845469 DOI: 10.3109/10799893.2012.707212] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The effect of the natural product diindolylmethane on cytosolic Ca(2+) concentrations ([Ca(2+)](i)) and viability in PC3 human prostate cancer cells was explored. The Ca(2+)-sensitive fluorescent dye fura-2 was applied to measure [Ca(2+)](i). Diindolylmethane at concentrations of 20-50 µM induced [Ca(2+)](i) rise in a concentration-dependent manner. The response was reduced partly by removing Ca(2+). Diindolylmethane-evoked Ca(2+) entry was suppressed by nifedipine, econazole, SK&F96365, protein kinase C modulators and aristolochic acid. In the absence of extracellular Ca(2+), incubation with the endoplasmic reticulum Ca(2+) pump inhibitor thapsigargin or 2,5-di-tert-butylhydroquinone (BHQ) inhibited or abolished diindolylmethane-induced [Ca(2+)](i) rise. Incubation with diindolylmethane also inhibited thapsigargin or BHQ-induced [Ca(2+)](i) rise. Inhibition of phospholipase C with U73122 reduced diindolylmethane-induced [Ca(2+)](i) rise. At concentrations of 50-100 µM, diindolylmethane killed cells in a concentration-dependent manner. This cytotoxic effect was not altered by chelating cytosolic Ca(2+) with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA). Annexin V/PI staining data implicate that diindolylmethane (50 and 100 µM) induced apoptosis in a concentration-dependent manner. In conclusion, diindolylmethane induced a [Ca(2+)](i) rise in PC3 cells by evoking phospholipase C-dependent Ca(2+) release from the endoplasmic reticulum and Ca(2+) entry via phospholipase A(2)-sensitive store-operated Ca(2+) channels. Diindolylmethane caused cell death in which apoptosis may participate.
Collapse
Affiliation(s)
- Jeng-Yu Tsai
- Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Beaver LM, Yu TW, Sokolowski EI, Williams DE, Dashwood RH, Ho E. 3,3'-Diindolylmethane, but not indole-3-carbinol, inhibits histone deacetylase activity in prostate cancer cells. Toxicol Appl Pharmacol 2012; 263:345-51. [PMID: 22800507 DOI: 10.1016/j.taap.2012.07.007] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 06/22/2012] [Accepted: 07/07/2012] [Indexed: 12/21/2022]
Abstract
Increased consumption of cruciferous vegetables is associated with a reduced risk of developing prostate cancer. Indole-3-carbinol (I3C) and 3,3'-diindolylmethane (DIM) are phytochemicals derived from cruciferous vegetables that have shown promise in inhibiting prostate cancer in experimental models. Histone deacetylase (HDAC) inhibition is an emerging target for cancer prevention and therapy. We sought to examine the effects of I3C and DIM on HDACs in human prostate cancer cell lines: androgen insensitive PC-3 cells and androgen sensitive LNCaP cells. I3C modestly inhibited HDAC activity in LNCaP cells by 25% but no inhibition of HDAC activity was detected in PC-3 cells. In contrast, DIM significantly inhibited HDAC activity in both cell lines by as much as 66%. Decreases in HDAC activity correlated with increased expression of p21, a known target of HDAC inhibitors. DIM treatment caused a significant decrease in the expression of HDAC2 protein in both cancer cell lines but no significant change in the protein levels of HDAC1, HDAC3, HDAC4, HDAC6 or HDAC8 was detected. Taken together, these results show that inhibition of HDAC activity by DIM may contribute to the phytochemicals' anti-proliferative effects in the prostate. The ability of DIM to target aberrant epigenetic patterns, in addition to its effects on detoxification of carcinogens, may make it an effective chemopreventive agent by targeting multiple stages of prostate carcinogenesis.
Collapse
Affiliation(s)
- Laura M Beaver
- Linus Pauling Institute, Oregon State University, 307 Linus Pauling Science Center, Corvallis, OR 97331, USA.
| | | | | | | | | | | |
Collapse
|
31
|
KUO CHIENNENG, CHEN CHUNGYI, LAI CHIENHSIUNG, LAI LIJU, WU PEICHEN, HUNG CHIAHUI, CHEN CHINGHSEIN. Cell cycle regulation by bevacizumab in ARPE-19 human retinal pigment epithelial cells. Mol Med Rep 2012; 6:701-4. [DOI: 10.3892/mmr.2012.986] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 06/29/2012] [Indexed: 11/06/2022] Open
|
32
|
Yin XF, Chen J, Mao W, Wang YH, Chen MH. A selective aryl hydrocarbon receptor modulator 3,3'-Diindolylmethane inhibits gastric cancer cell growth. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2012; 31:46. [PMID: 22592002 PMCID: PMC3403951 DOI: 10.1186/1756-9966-31-46] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 05/16/2012] [Indexed: 12/20/2022]
Abstract
Background Aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor associated with gastric carcinogenesis. 3,3'-Diindolylmethane (DIM) is a relatively non-toxic selective AhR modulator. This study was to detect the effects of DIM on gastric cancer cell growth. Methods Gastric cancer cell SGC7901 was treated with DIM at different concentrations (0,10,20,30,40,50 μmol/L) with or without an AhR antagonist, resveratrol. The expression of AhR and Cytochrome P4501A1 (CYP1A1), a classic target gene of AhR pathway, were detected by RT-PCR and Western blot; cell viability was measured by MTT assay, and the changes in cell cycle and apoptosis were analyzed by flow cytometry. Results RT-PCR and western-blot showed that with the increase of the concentration of DIM, AhR protein gradually decreased and CYP1A1 expression increased, suggesting that DIM activated the AhR pathway and caused the translocation of AhR from cytoplasm to nucleus. MTT assay indicated that the viability of SGC7901 cells was significantly decreased in a concentration- and time-dependent manner after DIM treatment and this could be partially reversed by resveratrol. Flow cytometry analysis showed that DIM arrested cell cycle in G1 phase and induced cell apoptosis. Conclusion Selective aryl hydrocarbon receptor modulator 3,3'-Diindolylmethane inhibits SGC7901 cell proliferation by inducing apoptosis and delaying cell cycle progression. AhR may be a potential therapeutic target for gastric cancer treatment.
Collapse
Affiliation(s)
- Xiao-Fei Yin
- Department of Gastroenterology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
| | | | | | | | | |
Collapse
|
33
|
Déziel B, MacPhee J, Patel K, Catalli A, Kulka M, Neto C, Gottschall-Pass K, Hurta R. American cranberry (Vaccinium macrocarpon) extract affects human prostate cancer cell growth via cell cycle arrest by modulating expression of cell cycle regulators. Food Funct 2012; 3:556-64. [PMID: 22388548 DOI: 10.1039/c2fo10145a] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Prostate cancer is one of the most common cancers in the world, and its prevalence is expected to increase appreciably in the coming decades. As such, more research is necessary to understand the etiology, progression and possible preventative measures to delay or to stop the development of this disease. Recently, there has been interest in examining the effects of whole extracts from commonly harvested crops on the behaviour and progression of cancer. Here, we describe the effects of whole cranberry extract (WCE) on the behaviour of DU145 human prostate cancer cells in vitro. Following treatment of DU145 human prostate cancer cells with 10, 25 and 50 μg ml⁻¹ of WCE, respectively for 6 h, WCE significantly decreased the cellular viability of DU145 cells. WCE also decreased the proportion of cells in the G2-M phase of the cell cycle and increased the proportion of cells in the G1 phase of the cell cycle following treatment of cells with 25 and 50 μg ml⁻¹ treatment of WCE for 6 h. These alterations in cell cycle were associated with changes in cell cycle regulatory proteins and other cell cycle associated proteins. WCE decreased the expression of CDK4, cyclin A, cyclin B1, cyclin D1 and cyclin E, and increased the expression of p27. Changes in p16(INK4a) and pRBp107 protein expression levels also were evident, however, the changes noted in p16(INK4a) and pRBp107 protein expression levels were not statistically significant. These findings demonstrate that phytochemical extracts from the American cranberry (Vaccinium macrocarpon) can affect the behaviour of human prostate cancer cells in vitro and further support the potential health benefits associated with cranberries.
Collapse
Affiliation(s)
- Bob Déziel
- Department of Biology, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE C1A 4P3, Canada
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Weng JR, Bai LY, Chiu CF, Wang YC, Tsai MH. The dietary phytochemical 3,3'-diindolylmethane induces G2/M arrest and apoptosis in oral squamous cell carcinoma by modulating Akt-NF-κB, MAPK, and p53 signaling. Chem Biol Interact 2012; 195:224-30. [PMID: 22290291 DOI: 10.1016/j.cbi.2012.01.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 01/14/2012] [Accepted: 01/14/2012] [Indexed: 01/06/2023]
Abstract
In light of the growing incidence of oral cancer in Taiwan, this study is aimed at investigating the antitumor activity of 3,3'-diindolylmethane (DIM), an active metabolite of the phytochemical indole-3-carbinol (I3C), in oral squamous cell carcinoma (OSCC). DIM exhibited substantially higher antiproliferative potency than I3C in three OSCC cell lines with IC(50) values in SCC2095, SCC9, and SCC15 cells, respectively, of 22 versus 168μM, 25 versus 176μM, and 29versus 300μM. Flow cytometric analysis and Comet assay indicated that DIM suppressed the viability of SCC2095 cells by inducing apoptosis and G2/M arrest. Western blot analysis of various signaling markers revealed the ability of DIM to target pathways mediated by Akt, mitogen-activated protein (MAP) kinases, nuclear factor (NF)-κB, and p53, of which the concerted action underlined its antitumor efficacy. The concomitant inactivation of Akt and MAP kinases in response to DIM facilitated the dephosphorylation of the proapoptotic protein Bad at Ser-136 and Ser-112, respectively. Through endoplasmic reticulum (ER) stress, DIM stimulated the activation of p53 via Ser-15 phosphorylation, leading to increased expression of the BH3-only proapoptotic Bcl-2 members Puma and Noxa. Together, these changes decreased the mitochondrial threshold for apoptosis. G2/M arrest might be attributable to the suppressive effect of DIM on the expression of cyclin B1 and cdc25c. As many downstream effectors of the Akt-NF-κB pathway, including glycogen synthase kinase 3β, IκB kinase α, and cyclooxygenase-2, have been shown to promote oral tumorigenesis, the ability of DIM to inhibit this signaling axis underscores its chemopreventive potential in oral cancer.
Collapse
Affiliation(s)
- Jing-Ru Weng
- Department of Biological Science and Technology, China Medical University, 91 Hsueh-Shih Road, Taichung 40402, Taiwan.
| | | | | | | | | |
Collapse
|
35
|
Sharma DK, Rah B, Lambu MR, Hussain A, Yousuf SK, Tripathi AK, Singh B, Jamwal G, Ahmed Z, Chanauria N, Nargotra A, Goswami A, Mukherjee D. Design and synthesis of novel N,N′-glycoside derivatives of 3,3′-diindolylmethanes as potential antiproliferative agents. MEDCHEMCOMM 2012. [DOI: 10.1039/c2md20098h] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
36
|
The indolic diet-derivative, 3,3'-diindolylmethane, induced apoptosis in human colon cancer cells through upregulation of NDRG1. J Biomed Biotechnol 2011; 2012:256178. [PMID: 22187533 PMCID: PMC3228297 DOI: 10.1155/2012/256178] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Accepted: 08/29/2011] [Indexed: 11/17/2022] Open
Abstract
N-myc downstream regulated gene-1 participates in carcinogenesis, angiogenesis, metastases, and anticancer drug resistance. In the present study, we analyzed the expression pattern of N-myc downstream regulated gene-1 following treatment of human colonic cancer cell lines; HCT-116 (well differentiated with wild-type p53 gene) and Colo-320 (poorly differentiated with mutant p53 gene), with 3,3'-diindolylmethane, a well-established proapoptotic agent product derived from indole-3-carbinol. Treatment of Colo-320 and HCT-116 with 3,3'-diindolylmethane disclosed inhibition of cell viability in a dose-dependent manner, mediated through apoptosis induction. The increased expression of N-myc downstream regulated gene-1 was detected only in poorly differentiated colon cancer cells, Colo-320 cell line. Our results suggest that N-myc downstream regulated gene-1 expression is enhanced by 3,3'-diindolylmethane in poorly differentiated cells and followed by induction of apoptosis. 3,3'-diindolylmethane induced apoptosis may represent a new regulator of N-myc downstream regulated gene-1 in poorly differentiated colonic cancer cells.
Collapse
|
37
|
Banerjee S, Kong D, Wang Z, Bao B, Hillman GG, Sarkar FH. Attenuation of multi-targeted proliferation-linked signaling by 3,3'-diindolylmethane (DIM): from bench to clinic. Mutat Res 2011; 728:47-66. [PMID: 21703360 DOI: 10.1016/j.mrrev.2011.06.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 06/06/2011] [Accepted: 06/07/2011] [Indexed: 12/14/2022]
Abstract
Emerging evidence provide credible support in favor of the potential role of bioactive products derived from ingesting cruciferous vegetables such as broccoli, brussel sprouts, cauliflower and cabbage. Among many compounds, 3,3'-diindolylmethane (DIM) is generated in the acidic environment of the stomach following dimerization of indole-3-carbinol (I3C) monomers present in these classes of vegetables. Both I3C and DIM have been investigated for their use in preventing, inhibiting, and reversing the progression of cancer - as a chemopreventive agent. In this review, we summarize an updated, wide-ranging pleiotropic anti-tumor and biological effects elicited by DIM against tumor cells. It is unfeasible to point one single target as basis of cellular target of action of DIM. We emphasize key cellular and molecular events that are effectively modulated in the direction of inducing apoptosis and suppressing cell proliferation. Collectively, DIM orchestrates signaling through Ah receptor, NF-κB/Wnt/Akt/mTOR pathways impinging on cell cycle arrest, modulation of key cytochrome P450 enzymes, altering angiogenesis, invasion, metastasis and epigenetic behavior of cancer cells. The ability of DIM to selectively induce tumor cells to undergo apoptosis has been observed in preclinical models, and thus it has been speculated in improving the therapeutic efficacy of other anticancer agents that have diverse molecular targets. Consequently, DIM has moved through preclinical development into Phase I clinical trials, thereby suggesting that DIM could be a promising and novel agent either alone or as an adjunct to conventional therapeutics such as chemo-radio and targeted therapies. An important development has been the availability of DIM formulation with superior bioavailability for humans. Therefore, DIM appears to be a promising chemopreventive agent or chemo-radio-sensitizer for the prevention of tumor recurrence and/or for the treatment of human malignancies.
Collapse
Affiliation(s)
- Sanjeev Banerjee
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | - Dejuan Kong
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Zhiwei Wang
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Bin Bao
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Gilda G Hillman
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Fazlul H Sarkar
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
38
|
Wedel S, Hudak L, Seibel JM, Juengel E, Oppermann E, Haferkamp A, Blaheta RA. Critical analysis of simultaneous blockage of histone deacetylase and multiple receptor tyrosine kinase in the treatment of prostate cancer. Prostate 2011; 71:722-35. [PMID: 20954195 DOI: 10.1002/pros.21288] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Accepted: 09/07/2010] [Indexed: 11/09/2022]
Abstract
BACKGROUND The concept of molecular tumor targeting might be an innovative option to treat advanced prostate cancer. We analyzed the effect of combining the multiple receptor tyrosine kinase inhibitor AEE788 and the histone deacetylase (HDAC) inhibitor valproic acid (VPA) on adhesion and growth properties of prostate cancer cell lines. METHODS PC-3, DU-145, and LNCaP cells were treated with AEE788, VPA or with an AEE788-VPA combination, and cell cycle progression investigated. Furthermore, tumor cell adhesion to vascular endothelium or to immobilized extracellular matrix proteins was evaluated, and integrin α and β subtypes were analyzed. Finally, effects of drug treatment on cell signaling pathways were determined. RESULTS AEE788 moderately and VPA strongly reduced tumor cell adhesion and growth. VPA impaired cell cycle progression and altered the expression level of the cell cycle regulating proteins cdk1, cdk2, cdk4, cyclin B, D1, cyclin E, p21, and p27. VPA also acted on the membranous, cytoplasmic, and gene expression pattern of various integrin α and β subtypes. AEE788 acted likewise, but more moderately. Combining AEE788 and VPA did not result in an additive anti-tumor effect. Signaling analysis revealed that the EGFr downstream target Akt was similarly modified in the presence of VPA or the VPA-AEE788 combination, but not influenced by AEE788 alone. CONCLUSIONS The AEE788-VPA combination has no advantage over VPA monotreatment in vitro. The non-responsiveness of Akt
Collapse
Affiliation(s)
- Steffen Wedel
- Department of Urology, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | | | | | | | | | | | | |
Collapse
|
39
|
Combined targeting of the VEGFr/EGFr and the mammalian target of rapamycin (mTOR) signaling pathway delays cell cycle progression and alters adhesion behavior of prostate carcinoma cells. Cancer Lett 2011; 301:17-28. [DOI: 10.1016/j.canlet.2010.11.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Revised: 11/04/2010] [Accepted: 11/07/2010] [Indexed: 01/08/2023]
|
40
|
Cho HJ, Park SY, Kim EJ, Kim JK, Park JHY. 3,3′-diindolylmethane inhibits prostate cancer development in the transgenic adenocarcinoma mouse prostate model. Mol Carcinog 2010; 50:100-12. [DOI: 10.1002/mc.20698] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Revised: 09/30/2010] [Accepted: 10/01/2010] [Indexed: 11/12/2022]
|
41
|
Sun YW, Huang WJ, Hsiao CJ, Chen YC, Lu PH, Guh JH. Methoxychalcone induces cell-cycle arrest and apoptosis in human hormone-resistant prostate cancer cells through PI 3-kinase-independent inhibition of mTOR pathways. Prostate 2010; 70:1295-306. [PMID: 20623631 DOI: 10.1002/pros.21165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Chalcones are contained in fruits and vegetables, and have been suggested to display anticancer activities. In this study, the anticancer mechanism of WJ9708011 (a methoxychalcone derivative) was delineated in human prostate cancer cells. METHOD Cell proliferation was examined by sulforhodamine B and clonogenic assays. Cell-cycle progression and mitochondrial membrane potential (DeltaPsi(m)) were detected by flow cytometric analysis. Expressions of protein and mRNA were detected by Western blot and RT-PCR technique, respectively. The protein synthesis was examined by [(3)H]leucine incorporation assay. The overexpression or knockdown techniques for specific target protein were also used in this study. RESULTS WJ9708011 induced time- and concentration-dependent G1 arrest of the cell cycle and subsequent apoptosis in human prostate cancer cells. The G1-arrest effect was confirmed by down-regulated expressions of several G1-phase regulators, including cyclin D1, cyclin E, cyclin-dependent kinase (Cdk)-4, Cdk2, phospho-RB, E2F-1, and Cdc25A. The mRNA expressions of cyclin D1 and cyclin E were also inhibited through the suppression of NF-kappaB. WJ9708011 blocked the protein synthesis and inhibited mammalian target of rapamycin (mTOR) signaling pathways. The suppression of mTOR pathways were irrespective of Akt- and AMPK-activated protein kinase (AMPK), but were attributed to mitochondrial stress, in which the down-regulation of survivin protein level may play a crucial role. CONCLUSIONS The data suggest that WJ9708011 induces transcriptional and translational suppression of cell-cycle regulators that might be through Akt- and AMPK-independent loss of DeltaPsi(m) and inhibition of mTOR signaling pathway, leading to G1 arrest of the cell cycle and subsequent apoptotic cell death.
Collapse
Affiliation(s)
- Yu-Wei Sun
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
42
|
Dean JL, Thangavel C, McClendon AK, Reed CA, Knudsen ES. Therapeutic CDK4/6 inhibition in breast cancer: key mechanisms of response and failure. Oncogene 2010; 29:4018-32. [PMID: 20473330 DOI: 10.1038/onc.2010.154] [Citation(s) in RCA: 308] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A hallmark of cancer is the deregulation of cell-cycle machinery, ultimately facilitating aberrant proliferation that fuels tumorigenesis and disease progression. Particularly, in breast cancers, cyclin D1 has a crucial role in the development of disease. Recently, a highly specific inhibitor of CDK4/6 activity (PD-0332991) has been developed that may have efficacy in the treatment of breast cancer. To interrogate the utility of PD-0332991 in treating breast cancers, therapeutic response was evaluated on a panel of breast cancer cell lines. These analyses showed that the chronic loss of Rb is specifically associated with evolution to a CDK4/6-independent state and, ultimately, resistance to PD-0332991. However, to interrogate the functional consequence of Rb directly, knockdown experiments were performed in models that represent immortalized mammary epithelia and multiple subtypes of breast cancer. These studies showed a highly specific role for Rb in mediating the response to CDK4/6 inhibition that was dependent on transcriptional repression manifest through E2F, and the ability to attenuate CDK2 activity. Acquired resistance to PD-03322991 was specifically associated with attenuation of CDK2 inhibitors, indicating that redundancy in CDK functions represents a determinant of therapeutic failure. Despite these caveats, in specific models, PD-0332991 was a particularly effective therapy, which induced Rb-dependent cytostasis. Combined, these findings indicate the critical importance of fully understanding cell-cycle regulatory pathways in directing the utilization of CDK inhibitors in the clinic.
Collapse
Affiliation(s)
- J L Dean
- Kimmel Cancer Center, Philadelphia, PA, USA
| | | | | | | | | |
Collapse
|
43
|
Vivar OI, Zhao X, Saunier EF, Griffin C, Mayba OS, Tagliaferri M, Cohen I, Speed TP, Leitman DC. Estrogen receptor beta binds to and regulates three distinct classes of target genes. J Biol Chem 2010; 285:22059-66. [PMID: 20404318 DOI: 10.1074/jbc.m110.114116] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Estrogen receptor beta (ERbeta) has potent antiproliferative and anti-inflammatory properties, suggesting that ERbeta-selective agonists might be a new class of therapeutic and chemopreventive agents. To understand how ERbeta regulates genes, we identified genes regulated by the unliganded and liganded forms of ERalpha and ERbeta in U2OS cells. Microarray data demonstrated that virtually no gene regulation occurred with unliganded ERalpha, whereas many genes were regulated by estradiol (E(2)). These results demonstrated that ERalpha requires a ligand to regulate a single class of genes. In contrast, ERbeta regulated three classes of genes. Class I genes were regulated primarily by unliganded ERbeta. Class II genes were regulated only with E(2), whereas class III genes were regulated by both unliganded ERbeta and E(2). There were 453 class I genes, 258 class II genes, and 83 class III genes. To explore the mechanism whereby ERbeta regulates different classes of genes, chromatin immunoprecipitation-sequencing was performed to identify ERbeta binding sites and adjacent transcription factor motifs in regulated genes. AP1 binding sites were more enriched in class I genes, whereas ERE, NFkappaB1, and SP1 sites were more enriched in class II genes. ERbeta bound to all three classes of genes, demonstrating that ERbeta binding is not responsible for differential regulation of genes by unliganded and liganded ERbeta. The coactivator NCOA2 was differentially recruited to several target genes. Our findings indicate that the unliganded and liganded forms of ERbeta regulate three classes of genes by interacting with different transcription factors and coactivators.
Collapse
Affiliation(s)
- Omar I Vivar
- Department of Nutritional Science and Toxicology, University of California, Berkeley, California 94720, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Vivar OI, Saunier EF, Leitman DC, Firestone GL, Bjeldanes LF. Selective activation of estrogen receptor-beta target genes by 3,3'-diindolylmethane. Endocrinology 2010; 151:1662-7. [PMID: 20160136 PMCID: PMC2850231 DOI: 10.1210/en.2009-1028] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
3,3'-Diindolylmethane (DIM) is a natural compound found in cruciferous vegetables that has antiproliferative and estrogenic activity. However, it is not clear whether the estrogenic effects are mediated through estrogen receptor (ER)alpha, ERbeta, or both ER subtypes. We investigated whether DIM has ER subtype selectivity on gene transcription. DIM stimulated ERbeta but not ERalpha activation of an estrogen response element upstream of the luciferase reporter gene. DIM also selectively activated multiple endogenous genes through ERbeta. DIM did not bind to ERbeta, indicating that it activates genes by a ligand-independent mechanism. DIM causes ERbeta to bind regulatory elements and recruit the steroid receptor coactivator (SRC)-2 coactivator, which leads to the activation of ER target genes. Silencing of SRC-2 inhibited the activation of ER target genes, demonstrating that SRC-2 is required for transcriptional activation by DIM. Our results demonstrate that DIM is a new class of ERbeta-selective compounds, because it does not bind to ERbeta, but instead it selectively recruits ERbeta and coactivators to target genes.
Collapse
Affiliation(s)
- Omar I Vivar
- Department of Nutritional Science and Toxicology, University of California, Berkeley, Berkeley, California 94720-3104, USA
| | | | | | | | | |
Collapse
|
45
|
Rahimi M, Huang KL, Tang CK. 3,3'-Diindolylmethane (DIM) inhibits the growth and invasion of drug-resistant human cancer cells expressing EGFR mutants. Cancer Lett 2010; 295:59-68. [PMID: 20299148 DOI: 10.1016/j.canlet.2010.02.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 02/15/2010] [Accepted: 02/16/2010] [Indexed: 01/02/2023]
Abstract
Epidermal Growth Factor Receptor (EGFR) mutants are associated with resistance to chemotherapy, radiation, and targeted therapies. Here we found that the phytochemical 3,3'-Diindolylmethane (DIM) can inhibit the growth and also the invasion of breast cancer, glioma, and non-small cell lung cancer cells regardless of which EGFR mutant is expressed and the drug-resistant phenotype. DIM reduced an array of growth factor signaling pathways and altered cell cycle regulators and apoptotic proteins favoring cell cycle arrest and apoptosis. Therefore, DIM may be used in treatment regimens to inhibit cancer cell growth and invasion, and potentially overcome EGFR mutant-associated drug resistance.
Collapse
Affiliation(s)
- Massod Rahimi
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University Medical Center, Washington, DC 20057, United States
| | | | | |
Collapse
|
46
|
Levy N, Paruthiyil S, Zhao X, Vivar OI, Saunier EF, Griffin C, Tagliaferri M, Cohen I, Speed TP, Leitman DC. Unliganded estrogen receptor-beta regulation of genes is inhibited by tamoxifen. Mol Cell Endocrinol 2010; 315:201-7. [PMID: 19744542 DOI: 10.1016/j.mce.2009.08.030] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2009] [Revised: 08/19/2009] [Accepted: 08/31/2009] [Indexed: 12/29/2022]
Abstract
Tamoxifen can stimulate the growth of some breast tumors and others can become resistant to tamoxifen. We previously showed that unliganded ERbeta inhibits ERalpha-mediated proliferation of MCF-7 cells. We investigated if tamoxifen might have a potential negative effect on some breast cancer cells by blocking the effects of unliganded ERbeta on gene regulation. Gene expression profiles demonstrated that unliganded ERbeta upregulated 196 genes in MCF-7 cells. Tamoxifen significantly inhibited 73 of these genes by greater than 30%, including several growth-inhibitory genes. To explore the mechanism whereby unliganded ERbeta activates genes and how tamoxifen blocks this effect, we used doxycycline-inducible U2OS-ERbeta cells to produce unliganded ERbeta. Doxycycline produced a dose-dependent activation of the NKG2E, MSMB and TUB3A genes, which was abolished by tamoxifen. Unliganded ERbeta recruitment of SRC-2 to the NKG2E gene was blocked by tamoxifen. Our findings suggest that tamoxifen might exert a negative effect on ERbeta expressing tumors due to its antagonistic action on unliganded ERbeta.
Collapse
Affiliation(s)
- Nitzan Levy
- Department of Obstetrics, Gynecology and Reproductive , University of California, San Francisco, CA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|