1
|
Patnam S, Singh AD, Ali MS, Thakur BK, Rengan AK, Manda SV. Development and In Vitro Characterization of Milk-Derived Extracellular Vesicle-Mithramycin Formulations for Potential Glioma Therapy. Mol Pharm 2025. [PMID: 40138182 DOI: 10.1021/acs.molpharmaceut.4c01189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Glioblastoma (GBM) is a highly aggressive brain tumor with resistance to conventional therapies. Mithramycin (Mit-A), a potent antitumor agent, has shown promise in several tumor types including, GBM. However, its clinical application is limited by toxicity. To address this, we explored the use of milk-derived extracellular vesicles (mEVs) as a delivery system to enhance the therapeutic efficacy of Mit-A. In this study, mEVs were isolated using a 3000 PEG precipitation method and confirmed their size, morphology, and stability through dynamic light scattering (DLS), transmission electron microscopy (TEM), and atomic force microscopy (AFM). The isolated vesicles with a size of 125.6 ± 2.78 nm, a polydispersity index (PDI) of 0.083 ± 0.02, and a ζ-potential of 15 ± 0.57 mV. The presence of typical EV markers such as TSG101, HSP70, and CD63 confirmed their purity. Encapsulation of Mit-A within mEVs led to a slight increase in size to 131.8 ± 6.9 nm, a PDI of 0.081 ± 0.006, and a decrease in ζ-potential to -17 ± 2.0 mV, with an encapsulation efficiency of 58% by the freeze-thaw method. The in vitro transepithelial transport assay revealed that mEV(Mit-A) transported Mit-A more effectively than free Mit-A. The mEV(Mit-A) formulation demonstrated excellent stability in simulated salivary and gastrointestinal fluids, with a sustained release of Mit-A observed over 24 h in vitro in PBS (pH 6.8). Furthermore, mEV(Mit-A) formulations significantly inhibited glioma cell growth, and migration, and induced apoptosis, showing a 2-fold lower IC50 than free Mit-A, indicating superior efficacy. These findings suggest that mEVs represent a promising delivery vehicle for Mit-A, enhancing its potential as an effective treatment for glioblastoma.
Collapse
Affiliation(s)
- Sreekanth Patnam
- Apollo Hospitals Educational and Research Foundation (AHERF), Hyderabad, Telangana 500096, India
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502284, India
- Exomed Therapeutics Private Ltd., Hyderabad, Telangana 500096, India
| | - Anula Divyash Singh
- Apollo Hospitals Educational and Research Foundation (AHERF), Hyderabad, Telangana 500096, India
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502284, India
| | - Mohammad Sadik Ali
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502284, India
| | - Basant Kumar Thakur
- Department of Pediatrics III, University Hospital Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502284, India
| | - Sasidhar Venkata Manda
- Apollo Hospitals Educational and Research Foundation (AHERF), Hyderabad, Telangana 500096, India
- UrvogelBio Private Ltd., Hyderabad, Telangana 500096, India
| |
Collapse
|
2
|
Lin MY, Damron TA, Horton JA. Cell cycle arrest and apoptosis are early events in radiosensitization of EWS::FLI1 + Ewing sarcoma cells by Mithramycin A. Int J Radiat Biol 2023; 99:1570-1583. [PMID: 36913323 DOI: 10.1080/09553002.2023.2188930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 02/23/2023] [Indexed: 03/14/2023]
Abstract
PURPOSE The oncogenic fusion protein EWS::FLI1 is an attractive therapeutic target in Ewing sarcoma (ES). Mithramycin A (MithA) is a potent and specific inhibitor of EWS::FLI1 that can selectively radiosensitize ES cells through transcriptional inhibition of DNA double-strand break (DSB) repair. Here, we evaluate temporal changes in cell cycle progression and apoptosis in ES cells treated with MithA and/or ionizing radiation (RTx), testing the hypothesis that combining MithA with ionizing radiation would synergistically impair cell cycle progression and enhance apoptotic elimination to a greater extent than either agent alone. MATERIALS AND METHODS Four EWS::FLI1+ ES cell lines TC-71, RD-ES, SK-ES-1, and A673, and one EWS::ERG cell line (CHLA-25) were exposed to 10nM MithA or vehicle and followed 24 h later by exposure to 2 Gy x-radiation or sham irradiation. Reactive oxygen species (ROS) activity was evaluated by cytometric assay, and assay of antioxidant gene expression by RT-qPCR. Cell cycle changes were evaluated by flow cytometry of nuclei stained with propidium iodide. Apoptosis was assessed by cytometric assessment of Caspase-3/7 activity and by immunoblotting of PARP-1 cleavage. Radiosensitization was evaluated by clonogenic survival assay. Proliferation (EdU) and apoptosis (TUNEL) were evaluated in SK-ES-1 xenograft tumors following pretreatment with 1 mg/kg MithA, followed 24 h later by a single 4 Gy fraction of x-radiation. RESULTS MithA-treated cells showed reduced levels of ROS, and were associated with increased expression of antioxidant genes SOD1, SOD2, and CAT. It nonetheless induced persistent G0/G1 arrest and a progressive increase of the sub-G1 fraction, suggesting apoptotic degeneration. In vitro assays of Caspase-3/7 activity and immunoblotting of Caspase-3/7 dependent cleavage of PARP-1 indicated that apoptosis began as early as 24 h after MithA exposure, reducing clonogenic survival. Tumors from xenograft mice treated with either radiation alone, or in combination with MithA showed a significant reduction of tumor cell proliferation, while apoptosis was significantly increased in the group receiving the combination of MithA and RTx. CONCLUSIONS Taken together, our data show that the anti-proliferative and cytotoxic effects of MithA are the prominent components of radiosensitization of EWS::FLI1+ ES, rather than the result of acutely enhanced ROS levels.
Collapse
Affiliation(s)
- Mei Yun Lin
- Department of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
- Cell & Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Timothy A Damron
- Department of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
- Cell & Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, USA
- Department of Radiation Oncology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Jason A Horton
- Department of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
- Cell & Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, USA
- Department of Radiation Oncology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
3
|
Kaur M, Kaur M, Bandopadhyay T, Sharma A, Priya A, Singh A, Banerjee B. Naturally occurring, natural product inspired and synthetic heterocyclic anti-cancer drugs. PHYSICAL SCIENCES REVIEWS 2022. [DOI: 10.1515/psr-2022-0003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Abstract
This chapter describes the importance and activity of a huge number of commercially available naturally occurring, natural product derived or synthetic heterocyclic anti-cancer drugs.
Collapse
Affiliation(s)
- Manmeet Kaur
- Department of Chemistry , Akal University , Talwandi Sabo , Bathinda , Punjab 151302 , India
| | - Mandeep Kaur
- Department of Chemistry , Akal University , Talwandi Sabo , Bathinda , Punjab 151302 , India
| | - Tania Bandopadhyay
- Completed MBBS from North Bengal Medical College and Hospital , Darjeeling , West Bengal , Pin-734432 , India
| | - Aditi Sharma
- Department of Chemistry , Akal University , Talwandi Sabo , Bathinda , Punjab 151302 , India
| | - Anu Priya
- Department of Chemistry , Akal University , Talwandi Sabo , Bathinda , Punjab 151302 , India
| | - Arvind Singh
- Department of Chemistry , Akal University , Talwandi Sabo , Bathinda , Punjab 151302 , India
| | - Bubun Banerjee
- Department of Chemistry , Akal University , Talwandi Sabo , Bathinda , Punjab 151302 , India
| |
Collapse
|
4
|
Dutta R, Khalil R, Mayilsamy K, Green R, Howell M, Bharadwaj S, Mohapatra SS, Mohapatra S. Combination Therapy of Mithramycin A and Immune Checkpoint Inhibitor for the Treatment of Colorectal Cancer in an Orthotopic Murine Model. Front Immunol 2021; 12:706133. [PMID: 34381456 PMCID: PMC8350740 DOI: 10.3389/fimmu.2021.706133] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/25/2021] [Indexed: 01/10/2023] Open
Abstract
The axis of Programmed cell death-1 receptor (PD-1) with its ligand (PD-L1) plays a critical role in colorectal cancer (CRC) in escaping immune surveillance, and blocking this axis has been found to be effective in a subset of patients. Although blocking PD-L1 has been shown to be effective in 5-10% of patients, the majority of the cohorts show resistance to this checkpoint blockade (CB) therapy. Multiple factors assist in the growth of resistance to CB, among which T cell exhaustion and immunosuppressive effects of immune cells in the tumor microenvironment (TME) play a critical role along with other tumor intrinsic factors. We have previously shown the polyketide antibiotic, Mithramycin-A (Mit-A), an effective agent in killing cancer stem cells (CSCs) in vitro and in vivo in a subcutaneous murine model. Since TME plays a pivotal role in CB therapy, we tested the immunomodulatory efficacy of Mit-A with anti-PD-L1 mAb (αPD-L1) combination therapy in an immunocompetent MC38 syngeneic orthotopic CRC mouse model. Tumors and spleens were analyzed by flow cytometry for the distinct immune cell populations affected by the treatment, in addition to RT-PCR for tumor samples. We demonstrated the combination treatment decreases tumor growth, thus increasing the effectiveness of the CB. Mit-A in the presence of αPD-L1 significantly increased CD8+ T cell infiltration and decreased immunosuppressive granulocytic myeloid-derived suppressor cells and anti-inflammatory macrophages in the TME. Our results revealed Mit-A in combination with αPD-L1 has the potential for augmented CB therapy by turning an immunologically "cold" into "hot" TME in CRC.
Collapse
Affiliation(s)
- Rinku Dutta
- James A. Haley Veterans’ Hospital, Tampa, FL, United States
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Center for Research and Education in Nano-Bioengineering, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Roukiah Khalil
- James A. Haley Veterans’ Hospital, Tampa, FL, United States
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Center for Research and Education in Nano-Bioengineering, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Karthick Mayilsamy
- James A. Haley Veterans’ Hospital, Tampa, FL, United States
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Center for Research and Education in Nano-Bioengineering, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Ryan Green
- James A. Haley Veterans’ Hospital, Tampa, FL, United States
- Center for Research and Education in Nano-Bioengineering, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Mark Howell
- James A. Haley Veterans’ Hospital, Tampa, FL, United States
- Center for Research and Education in Nano-Bioengineering, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Srinivas Bharadwaj
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Shyam S. Mohapatra
- James A. Haley Veterans’ Hospital, Tampa, FL, United States
- Center for Research and Education in Nano-Bioengineering, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Subhra Mohapatra
- James A. Haley Veterans’ Hospital, Tampa, FL, United States
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Center for Research and Education in Nano-Bioengineering, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
5
|
Mohan CD, Rangappa S, Nayak SC, Jadimurthy R, Wang L, Sethi G, Garg M, Rangappa KS. Bacteria as a treasure house of secondary metabolites with anticancer potential. Semin Cancer Biol 2021; 86:998-1013. [PMID: 33979675 DOI: 10.1016/j.semcancer.2021.05.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/03/2021] [Accepted: 05/03/2021] [Indexed: 12/27/2022]
Abstract
Cancer stands in the frontline among leading killers worldwide and the annual mortality rate is expected to reach 16.4 million by 2040. Humans suffer from about 200 different types of cancers and many of them have a small number of approved therapeutic agents. Moreover, several types of major cancers are diagnosed at advanced stages as a result of which the existing therapies have limited efficacy against them and contribute to a dismal prognosis. Therefore, it is essential to develop novel potent anticancer agents to counteract cancer-driven lethality. Natural sources such as bacteria, plants, fungi, and marine microorganisms have been serving as an inexhaustible source of anticancer agents. Notably, over 13,000 natural compounds endowed with different pharmacological properties have been isolated from different bacterial sources. In the present article, we have discussed about the importance of natural products, with special emphasis on bacterial metabolites for cancer therapy. Subsequently, we have comprehensively discussed the various sources, mechanisms of action, toxicity issues, and off-target effects of clinically used anticancer drugs (such as actinomycin D, bleomycin, carfilzomib, doxorubicin, ixabepilone, mitomycin C, pentostatin, rapalogs, and romidepsin) that have been derived from different bacteria. Furthermore, we have also discussed some of the major secondary metabolites (antimycins, chartreusin, elsamicins, geldanamycin, monensin, plicamycin, prodigiosin, rebeccamycin, salinomycin, and salinosporamide) that are currently in the clinical trials or which have demonstrated potent anticancer activity in preclinical models. Besides, we have elaborated on the application of metagenomics in drug discovery and briefly described about anticancer agents (bryostatin 1 and ET-743) identified through the metagenomics approach.
Collapse
Affiliation(s)
| | - Shobith Rangappa
- Adichunchanagiri Institute for Molecular Medicine, Adichunchanagiri University, BG Nagara, 571448, Nagamangala Taluk, India
| | - S Chandra Nayak
- Department of Studies in Biotechnology, University of Mysore, Manasagangotri, Mysore, 570006, India
| | - Ragi Jadimurthy
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore, 570006, India
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Uttar Pradesh, Noida, 201313, India
| | | |
Collapse
|
6
|
Geng H, Su Y, Huang R, Fan M, Li X, Lu X, Sheng H. Specific protein 1 inhibitor mithramycin A protects cardiomyocytes from myocardial infarction via interacting with PARP. In Vitro Cell Dev Biol Anim 2021; 57:315-323. [PMID: 33580416 DOI: 10.1007/s11626-021-00543-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/05/2021] [Indexed: 10/22/2022]
Abstract
Specific protein 1 (SP1) might act as a critical transcription regulator in myocardial infarction (MI), but little evidence about its function in regulating cardiac apoptosis, a major cause of MI development, has been revealed. This study tried to investigate the role of SP1 in MI and its interaction with poly-ADP-ribose polymerase (PARP)-1 by using SP1 inhibitor, mithramycin A (mithA). Primary mouse cardiomyocytes and commercial mouse cardiomyocytes were subjected to mithA treatment under hypoxia conditions, while cell viability, Nix promoter activity, and its expression were detected correspondingly. PARP overexpression and knockdown were conducted, respectively, in mithA-treated and SP1-overexpressing cells. Co-immunoprecipitation was used to verify the interaction between PARP and SP1. For in vivo experiments, mithA administration was performed after the injections of adenovirus for PARP overexpression, and then, MI introduction was carried out. Infarct size and lactate dehydrogenase level were measured to assess MI injury. SP1 inhibitor mithA attenuated hypoxia-induced decrease of cell viability and Nix transcriptional activation, which could be inhibited by PARP overexpression. Knockdown of PARP prevented SP1-induced transcription of Nix and cell viability change, and PARP showed direct interaction with SP1. Furthermore, mithA administration reduced MI injuries, while PARP overexpression could suppress the improvement. The cardioprotective role of SP1 inhibitor mithA was demonstrated here expanding the role of SP1 in MI development involving hypoxia-induced cardiac apoptosis. Moreover, PARP acted as a transcriptional coactivator in Nix transcription involving its interaction with SP1.
Collapse
Affiliation(s)
- Haihua Geng
- Department of Cardiology, Affiliated Hospital of Nantong University, No. 20 Xisi Rd, Nantong, 226001, Jiangsu, China
| | - Yamin Su
- Department of Cardiology, Affiliated Hospital of Nantong University, No. 20 Xisi Rd, Nantong, 226001, Jiangsu, China
| | - Rong Huang
- Department of Cardiology, Affiliated Hospital of Nantong University, No. 20 Xisi Rd, Nantong, 226001, Jiangsu, China
| | - Mengkang Fan
- Department of Cardiology, Affiliated Hospital of Nantong University, No. 20 Xisi Rd, Nantong, 226001, Jiangsu, China
| | - Xiaofei Li
- Department of Cardiology, Affiliated Hospital of Nantong University, No. 20 Xisi Rd, Nantong, 226001, Jiangsu, China
| | - Xiaochen Lu
- Department of Cardiology, Affiliated Hospital of Nantong University, No. 20 Xisi Rd, Nantong, 226001, Jiangsu, China
| | - Hongzhuan Sheng
- Department of Cardiology, Affiliated Hospital of Nantong University, No. 20 Xisi Rd, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
7
|
Beug ST, Cheung HH, Sanda T, St-Jean M, Beauregard CE, Mamady H, Baird SD, LaCasse EC, Korneluk RG. The transcription factor SP3 drives TNF-α expression in response to Smac mimetics. Sci Signal 2019; 12:12/566/eaat9563. [PMID: 30696705 DOI: 10.1126/scisignal.aat9563] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The controlled production and downstream signaling of the inflammatory cytokine tumor necrosis factor-α (TNF-α) are important for immunity and its anticancer effects. Although chronic stimulation with TNF-α is detrimental to the health of the host in several autoimmune and inflammatory disorders, TNF-α-contrary to what its name implies-leads to cancer formation by promoting cell proliferation and survival. Smac mimetic compounds (SMCs), small-molecule antagonists of inhibitor of apoptosis proteins (IAPs), switch the TNF-α signal from promoting survival to promoting death in cancer cells. Using a genome-wide siRNA screen to identify factors required for SMC-to-TNF-α-mediated cancer cell death, we identified the transcription factor SP3 as a critical molecule in both basal and SMC-induced production of TNF-α by engaging the nuclear factor κB (NF-κB) transcriptional pathway. Moreover, the promotion of TNF-α expression by SP3 activity confers differential sensitivity of cancer versus normal cells to SMC treatment. The key role of SP3 in TNF-α production and signaling will help us further understand TNF-α biology and provide insight into mechanisms relevant to cancer and inflammatory disease.
Collapse
Affiliation(s)
- Shawn T Beug
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, Ontario K1H 8L1, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Herman H Cheung
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, Ontario K1H 8L1, Canada
| | - Tarun Sanda
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, Ontario K1H 8L1, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Martine St-Jean
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, Ontario K1H 8L1, Canada
| | - Caroline E Beauregard
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, Ontario K1H 8L1, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Hapsatou Mamady
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, Ontario K1H 8L1, Canada
| | - Stephen D Baird
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, Ontario K1H 8L1, Canada
| | - Eric C LaCasse
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, Ontario K1H 8L1, Canada.
| | - Robert G Korneluk
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, Ontario K1H 8L1, Canada. .,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
8
|
Choi ES, Nam JS, Jung JY, Cho NP, Cho SD. Modulation of specificity protein 1 by mithramycin A as a novel therapeutic strategy for cervical cancer. Sci Rep 2014; 4:7162. [PMID: 25418289 PMCID: PMC4241519 DOI: 10.1038/srep07162] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 11/03/2014] [Indexed: 11/09/2022] Open
Abstract
Cervical cancer is the third most common cancer and the third leading cause of death among women. However, the standard treatment for cervical cancer includes cisplatin, which can cause side effects such as hematological damage or renal toxicity. New innovations in cervical cancer treatment focus on developing more effective and better-tolerated therapies such as Sp1-targeting drugs. Previous studies suggested that mithramycin A (Mith) inhibits the growth of various cancers by decreasing Sp1 protein. However, how Sp1 protein is decreased by Mith is not clear. Few studies have investigated the regulation of Sp1 protein by proteasome-dependent degradation as a possible control mechanism for the regulation of Sp1 in cancer cells. Here, we show that Mith decreased Sp1 protein by inducing proteasome-dependent degradation, thereby suppressing cervical cancer growth through a DR5/caspase-8/Bid signaling pathway. We found that prolonged Mith treatment was well tolerated after systemic administration to mice carrying cervical cancer cells. Reduction of body weight was minimal, indicating that Mith was a good therapeutic candidate for treatment of cancers in which Sp1 is involved in promoting and developing disease.
Collapse
Affiliation(s)
- Eun-Sun Choi
- Division of High-risk Pathogen Research, Korea Centers for Disease Control and Prevention, Osong, Republic of Korea
| | - Jeong-Seok Nam
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Graduate School of Medicine, Incheon 406-840, Korea
| | - Ji-Youn Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan 314-701, Republic of Korea
| | - Nam-Pyo Cho
- Department of Oral Pathology, School of Dentistry and Institute of Biodegradable Material, Institute of Oral Bioscience, Brain Korea 21 Project, Chonbuk National University, Jeon-ju 561-756, Republic of Korea
| | - Sung-Dae Cho
- Department of Oral Pathology, School of Dentistry and Institute of Biodegradable Material, Institute of Oral Bioscience, Brain Korea 21 Project, Chonbuk National University, Jeon-ju 561-756, Republic of Korea
| |
Collapse
|
9
|
Abstract
The role of specificity protein 1 (Sp1) in controlling gene expression in lung tumor development and metastasis is not well understood. In this study, we showed that the Sp1 level was highly increased and required for lung tumor growth in transgenic mice bearing Kras-induced lung tumors under the control of doxycycline. Furthermore, the Sp1 level was highly upregulated in lung adenocarcinoma cells with low invasiveness and in patients with stage I lung cancer. We also demonstrated that Sp1 was downregulated in lung adenocarcinoma cells with high invasiveness and in patients with stage IV lung adenocarcinoma. Moreover, Sp1 inversely regulated migration, invasion and metastasis of lung adenocarcinoma cells in vivo. In addition, a decrease in the Sp1 level in highly invasive lung adenocarcinoma cells resulted from instability of the Sp1 protein. Furthermore, overexpression of Sp1 in highly invasive lung adenocarcinoma cells increased expression of E-cadherin, a suppressor of metastasis, and attenuated the translocation of β-catenin into the cellular nucleus that leads to tumor malignancy. Taken together, Sp1 level accumulated strongly in early stage and then declined in late stage, which is important for lung cancer cell proliferation and metastasis during tumorigenesis.
Collapse
|
10
|
Gao Y, Jia Z, Kong X, Li Q, Chang DZ, Wei D, Le X, Huang S, Wang L, Xie K. Combining betulinic acid and mithramycin a effectively suppresses pancreatic cancer by inhibiting proliferation, invasion, and angiogenesis. Cancer Res 2011; 71:5182-93. [PMID: 21673052 PMCID: PMC3245664 DOI: 10.1158/0008-5472.can-10-2016] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Both betulinic acid (BA) and mithramycin A (MIT) exhibit potent antitumor activity through distinct mechanisms of Sp1 inhibition. However, it is unknown whether a combination of these two compounds results in a synergistic inhibitory effect on pancreatic cancer growth and/or has a therapeutic advantage over gemcitabine. In xenograft mouse models of human pancreatic cancer, treatment with either BA or MIT alone showed dose-dependent antitumor activity but led to systemic side effects as measured by overall weight loss. Treatment with a nontoxic dose of either compound alone had only marginal antitumor effects. Importantly, combination treatment with nontoxic doses of BA and MIT produced synergistic antitumor activity, including inhibitory effects on cell proliferation, invasion, and angiogenesis. The treatment combination also produced less discernible side effects than therapeutic doses of gemcitabine. Moreover, combined treatment of BA and MIT resulted in drastic inhibition of Sp1 recruitment onto Sp1 and VEGF promoters, leading to transcriptional inhibition of both Sp1 and VEGF and downregulation of Sp1 and VEGF protein expression. Ectopic overexpression of Sp1 rendered tumor cells resistant to BA, MIT, and the combination of the two. Overall, our findings argue that Sp1 is an important target of BA and MIT and that their combination can produce an enhanced therapeutic response in human pancreatic cancer.
Collapse
Affiliation(s)
- Yong Gao
- Department of Oncology, Shanghai Tongji University Affiliated East Hospital, Shanghai, People’s Republic of China
- Department of Cardiothoracic Surgery, Second Military Medical University Affiliated Changhai Hospital, Shanghai, People’s Republic of China
| | - Zhiliang Jia
- Department of Gastrointestinal Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Xiangyu Kong
- Department of Gastroenterology, Second Military Medical University Affiliated Changhai Hospital, Shanghai, People’s Republic of China
- Department of Gastrointestinal Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Qiang Li
- Department of Gastrointestinal Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - David Z. Chang
- Department of Gastrointestinal Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Daoyan Wei
- Department of Gastrointestinal Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Xiangdong Le
- Department of Gastrointestinal Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Shengdong Huang
- Department of Cardiothoracic Surgery, Second Military Medical University Affiliated Changhai Hospital, Shanghai, People’s Republic of China
| | - Liwei Wang
- Shanghai Key Laboratory of Pancreatic Diseases Research and Department of Oncology, Shanghai Jiaotong University Affiliated First People’s Hospital, Shanghai, People’s Republic of China
| | - Keping Xie
- Department of Gastrointestinal Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| |
Collapse
|
11
|
Basha R, Ingersoll SB, Sankpal UT, Ahmad S, Baker CH, Edwards JR, Holloway RW, Kaja S, Abdelrahim M. Tolfenamic acid inhibits ovarian cancer cell growth and decreases the expression of c-Met and survivin through suppressing specificity protein transcription factors. Gynecol Oncol 2011; 122:163-70. [DOI: 10.1016/j.ygyno.2011.03.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 03/11/2011] [Accepted: 03/17/2011] [Indexed: 11/29/2022]
|
12
|
Jia Z, Gao Y, Wang L, Li Q, Zhang J, Le X, Wei D, Yao JC, Chang DZ, Huang S, Xie K. Combined treatment of pancreatic cancer with mithramycin A and tolfenamic acid promotes Sp1 degradation and synergistic antitumor activity. Cancer Res 2010; 70:1111-9. [PMID: 20086170 DOI: 10.1158/0008-5472.can-09-3282] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Mithramycin (MIT) and tolfenamic acid (TA) inhibit the activity of the transcription factor Sp1. In the present study, we investigated whether pancreatic cancer treatment with a combination of these compounds has a synergistic effect on Sp1 activity, tumor growth, and their underlying response mechanisms. Treatment of pancreatic tumor xenografts with MIT and TA produced dose-dependent antitumor activity, and significant antitumor activity of either compound alone was directly associated with systemic side effects. Combination treatment with nontoxic doses of both compounds produced synergistic antitumor activity, whereas treatment with a nontoxic dose of either compound alone lacked a discernible antitumor effect. Synergistic therapeutic effects correlated directly with synergistic antiproliferation and antiangiogenesis in vitro. Moreover, combination treatment resulted in Sp1 protein degradation, drastically downregulating expression of Sp1 and vascular endothelial growth factor. Our findings established that Sp1 is a critical target of TA and MIT in human pancreatic cancer therapy, rationalizing clinical studies to determine the effect of existing pancreatic cancer therapy regimens on Sp1 signaling in tumors and normal pancreatic tissue, and the ability of Sp1-targeting strategies to modify cancer responses.
Collapse
Affiliation(s)
- Zhiliang Jia
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Abstract
Tumor necrosis factor (TNF) is a multifunctional cytokine that plays important roles in diverse cellular events such as cell survival, proliferation, differentiation, and death. As a pro-inflammatory cytokine, TNF is secreted by inflammatory cells, which may be involved in inflammation-associated carcinogenesis. TNF exerts its biological functions through activating distinct signaling pathways such as nuclear factor-kappaB (NF-kappaB) and c-Jun N-terminal kinase (JNK). NF-kappaB is a major cell survival signal that is anti-apoptotic, whereas sustained JNK activation contributes to cell death. The crosstalk between the NF-kappaB and JNK is involved in determining cellular outcomes in response to TNF. In regard to cancer, TNF is a double-dealer. On one hand, TNF could be an endogenous tumor promoter, because TNF stimulates the growth, proliferation, invasion and metastasis, and tumor angiogenesis of cancer cells. On the other hand, TNF could be a cancer killer. The property of TNF in inducing cancer cell death renders it a potential cancer therapeutic, although much work is needed to reduce its toxicity for systematic TNF administration. Recent studies have focused on sensitizing cancer cells to TNF-induced apoptosis through inhibiting survival signals such as NF-kappaB, by combined therapy. In this article we provide an overview of the roles of TNF-induced signaling pathways in cancer biology with specific emphasis on carcinogenesis and cancer therapy.
Collapse
Affiliation(s)
- Xia WANG
- Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yong LIN
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute 2425 Ridgecrest DR., SE, Albuquerque, NM 87108, USA
| |
Collapse
|
14
|
Liu D, Wei Y, Zhou F, Ge Y, Xu J, Chen H, Zhang W, Yun X, Jiang J. E1AF promotes mithramycin A-induced Huh-7 cell apoptosis depending on its DNA-binding domain. Arch Biochem Biophys 2008; 477:20-6. [PMID: 18510939 DOI: 10.1016/j.abb.2008.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Revised: 04/11/2008] [Accepted: 05/07/2008] [Indexed: 11/28/2022]
Abstract
Transcription factor E1AF is widely known to play critical roles in tumor metastasis via directly binding to the promoters of genes involved in tumor migration and invasion. Here, we reported for the first time the pro-apoptotic role of E1AF in tumor cells. The expression of E1AF at protein level was obviously increased during Huh-7 and Hep3B cells apoptosis induced by the anticancer agent mithramycin A. E1AF overexpression markedly enhanced mithramycin A-induced Huh-7 cell apoptosis and the expression of pro-apoptotic protein Bax depending on its DNA-binding domain. And, reduction of E1AF inhibited mithramycin A-induced Huh-7 cell apoptosis. Furthermore, reducing the expression of Bax significantly inhibited E1AF-increased Huh-7 cell apoptosis induced by mithramycin A. Taken together, E1AF increases mithramycin A-induced Huh-7 cells apoptosis and Bax expression depending on its DNA-binding domain, indicating that E1AF might contribute to the therapeutic efficiency of mithramycin A for hepatoma.
Collapse
Affiliation(s)
- Dan Liu
- Key Laboratory of Glycoconjugates Research, Ministry of Public Health & Gene Research Center, Shanghai Medical College of Fudan University, Dongan Road 130, Shanghai 200032, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Development and application of a stable HeLa cell line capable of site-specific transgenesis using the Cre-lox system: establishment and application of a stable TNFRI knockdown cell line to cytotoxicity assay. Toxicol In Vitro 2008; 22:1077-87. [PMID: 18356016 DOI: 10.1016/j.tiv.2008.01.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2007] [Revised: 09/03/2007] [Accepted: 01/22/2008] [Indexed: 10/22/2022]
Abstract
Mammalian cell models for gene knock-out/knock-in experiments are important for functional analysis of genes and have a potential of useful tool for toxicological studies. However, uncontrolled insertion of transgenes has raised significant concerns over unwanted side effects. To address this issue, we established a stable HeLa55 cell line capable of site-specific transgenesis by means of Cre-mediated cassette exchange at a site on the long arm of human chromosome 9 containing no constitutive transcripts. We applied HeLa55 to transgenesis of the green fluorescent protein (GFP) gene based on recombinase-mediated cassette exchange. The transformants stably expressed GFP transgenes, even after cryopreservation, without compromising physiological properties. We produced an RNA interference (RNAi)-inducible knockdown stable cell line against human tumor necrosis factor (TNF) receptor I, and one cloned stable cell line (TNFRIKD cells) exhibited long-term gene silencing with significant reduction (ca. 85%) and markedly resisted cytotoxicity induced by TNFalpha. Furthermore, xenobiotics were exposed to stable TNFRIKD cells and different cytotoxicity was exhibited based on various toxicological properties. Thus, we showed the feasibility of RNAi-based stable knockdown cells for xenobiotics-induced cytotoxicity, and HeLa55 has wide application for the generation of stable knock-in and knock-down cells mediated by RNAi.
Collapse
|
16
|
Yuan P, Wang L, Wei D, Zhang J, Jia Z, Li Q, Le X, Wang H, Yao J, Xie K. Therapeutic inhibition of Sp1 expression in growing tumors by mithramycin a correlates directly with potent antiangiogenic effects on human pancreatic cancer. Cancer 2008; 110:2682-90. [PMID: 17973266 DOI: 10.1002/cncr.23092] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND Human pancreatic cancer over expresses the transcription factor Sp1. However, the role of Sp1 in pancreatic cancer angiogenesis and its use as target for antiangiogenic therapy remain unexplored. METHODS Archived human pancreatic cancer specimens were used to assess gene expression and microvessel density (MVD) status by immunohistochemistry: Small-interfering RNA (siRNA) was used to determine the impact of altered Sp1 expression on tumor growth and angiogenesis, and mithramycin A (MIT) was used to evaluate Sp1-targeted antiangiogenic treatment of human pancreatic cancer in animal models. RESULTS The expression level of Sp1 was correlated directly with the MVD status (P < .001) and the expression level of vascular endothelial growth factor (VEGF) (P < .05). Knockdown of Sp1 expression did not affect the growth of pancreatic cancer cells in vitro but inhibited their growth and metastasis in mouse models. This antitumor activity was consistent with the in vitro and in vivo antiangiogenic activity resulting from Sp1 knockdown. Subcutaneous and intraperitoneal injection of MIT significantly suppressed the growth of human pancreatic cancer in mouse models. This tumor suppression was correlated with the suppression of Sp1 expression in growing tumors but not in normal tissues. Moreover, treatment with MIT reduced tumor MVD, which was consistent with the down-regulation of VEGF, platelet-derived growth factor, and epidermal growth factor receptor. CONCLUSIONS Both clinical and experimental evidence indicated that Sp1 is a critical regulator of human pancreatic cancer angiogenesis and the antitumor activity of MIT is a result, at least in part, of the suppression of Sp1 expression and consequent down-regulation the downstream targets of Sp1 that are key to angiogenesis.
Collapse
Affiliation(s)
- Ping Yuan
- Department of Pathology, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Amrán D, Sánchez Y, Fernández C, Ramos AM, de Blas E, Bréard J, Calle C, Aller P. Arsenic trioxide sensitizes promonocytic leukemia cells to TNFα-induced apoptosis via p38-MAPK-regulated activation of both receptor-mediated and mitochondrial pathways. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1773:1653-63. [PMID: 17673311 DOI: 10.1016/j.bbamcr.2007.06.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2007] [Revised: 06/14/2007] [Accepted: 06/15/2007] [Indexed: 10/23/2022]
Abstract
Treatment with the anti-leukemic drug arsenic trioxide (As(2)O(3), 1-4 microM) sensitizes U937 promonocytes and other human myeloid leukemia cell lines (HL60, NB4) to apoptosis induction by TNFalpha. As(2)O(3) plus TNFalpha increases TNF receptor type 1 (TNF-R1) expression, decreases c-FLIP(L) expression, and causes caspase-8 and Bid activation, and apoptosis is reduced by anti-TNF-R1 neutralizing antibody and caspase-8 inhibitor. The treatment also causes Bax translocation to mitochondria, cytochrome c and Omi/HtrA2 release from mitochondria, XIAP down-regulation, and caspase-9 and caspase-3 activation. Bcl-2 over-expression inhibits cytochrome c release and apoptosis, and also prevents c-FLIP(L) down-regulation and caspase-8 activation, but not TNF-R1 over-expression. As(2)O(3) does not affect Akt phosphorylation/activation or intracellular GSH content, nor prevents the TNFalpha-provoked stimulation of p65-NF-kappaB translocation to the nucleus and the increase in NF-kappaB binding activity. Treatments with TNFalpha alone or with As(2)O(3) plus TNFalpha cause TNF-R1-mediated p38-MAPK phosphorylation/activation. P38-MAPK-specific inhibitors attenuate the As(2)O(3) plus TNFalpha-provoked activation of caspase-8/Bid, Bax translocation, cytochrome c release, and apoptosis induction. In conclusion, the sensitization by As(2)O(3) to TNFalpha-induced apoptosis in promonocytic leukemia cells is an Akt/NF-kappaB-independent, p38-MAPK-regulated process, which involves the interplay of both the receptor-mediated and mitochondrial executioner pathways.
Collapse
Affiliation(s)
- Donna Amrán
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Lin RK, Hsu CH, Wang YC. Mithramycin A inhibits DNA methyltransferase and metastasis potential of lung cancer cells. Anticancer Drugs 2007; 18:1157-64. [PMID: 17893516 DOI: 10.1097/cad.0b013e3282a215e9] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Abnormal CpG island hypermethylation of multiple tumor-suppressor genes (TSGs) can lead to the initiation and progression of human cancer. The cytosine of the CpG island on the promoter region is methylated by 5'-cytosine-methyltransferases (DNMTs). Pharmacologic inhibitors of CpG island methylation provide a rational approach to reactivate the TSGs in tumor cells and to restore the critical cellular pathways in cancer cells. Mithramycin A (MMA) is known to be a GC- and CG-rich DNA-binding agent. We sought to determine whether MMA could inhibit CpG island methylation and DNMT expression in lung cancer cells. We found that MMA reduced the CpG island methylation of antimetastasis TSGs, including SLIT2 and TIMP-3 genes, and was associated with the prevention of metastasis. When highly metastatic CL1-5 lung cancer cells were treated with low doses (10 nmol/l) of MMA for 14 days, they reexpressed mRNA levels for these genes. MMA also inhibited the invasion phenotypes of CL1-5 cells as indicated by its inhibition of cancer cell migration using wound-healing and transwell assays. Molecular docking of MMA onto the DNMT1 catalytic domain revealed that MMA might interact with the catalytic pocket of DNMT1. Western blots showed that DNMT1 protein levels were depleted after MMA. These data support the idea that MMA has demethylation and antimetastasis effects on lung cancer cells. This mechanism might be mediated by the interaction of MMA and DNMT1, leading to the depletion of the DNMT1 protein and the reversal of the metastasis phenotype in lung cancer cells.
Collapse
Affiliation(s)
- Ruo-Kai Lin
- Department of Life Sciences, National Taiwan Normal University, Taipei, Taiwan, ROC
| | | | | |
Collapse
|
19
|
Hsieh AC, Small EJ, Ryan CJ. Androgen-response elements in hormone-refractory prostate cancer: implications for treatment development. Lancet Oncol 2007; 8:933-9. [PMID: 17913662 DOI: 10.1016/s1470-2045(07)70316-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Many attempts have been made to derive genetic signatures for progressive prostate cancer for both prognostic and therapeutic purposes. These investigations have resulted in the discovery of many pathways, but the signatures exhibit heterogeneity and restricted reproducibility. A thorough and disciplined analysis of genes with androgen-response elements that are expressed in progressive, castration-resistant prostate cancer is an integral step towards the development of new therapeutic or diagnostic targets. We discuss the effects of bona-fide downstream targets of the androgen receptor on cellular proliferation, evasion of apoptosis, and angiogenesis, and consider the clinical potential of these targets.
Collapse
Affiliation(s)
- Andrew C Hsieh
- Department of Medicine, and UCSF Comprehensive Cancer Center, University of California-San Francisco, 94143, USA.
| | | | | |
Collapse
|
20
|
Jia Z, Zhang J, Wei D, Wang L, Yuan P, Le X, Li Q, Yao J, Xie K. Molecular basis of the synergistic antiangiogenic activity of bevacizumab and mithramycin A. Cancer Res 2007; 67:4878-85. [PMID: 17510417 DOI: 10.1158/0008-5472.can-06-3494] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The impact of antiangiogenic therapy on the Sp1/vascular endothelial growth factor (VEGF) pathway and that of alteration of Sp1 signaling on the efficacy of antiangiogenic therapy is unclear, yet understanding their interactions has significant clinical implications. Treatment with bevacizumab, a neutralizing antibody against VEGF, suppressed human pancreatic cancer growth in nude mice. Gene expression analyses revealed that this treatment substantially up-regulated the expression of Sp1 and its downstream target genes, including VEGF and epidermal growth factor receptor, in tumor tissues, whereas it did not have this effect on pancreatic cancer cells in culture. Treatment with mithramycin A, an Sp1 inhibitor, suppressed the expression of Sp1 and its downstream target genes in both cell culture and tumors growing in nude mice. Combined treatment with bevacizumab and mithramycin A produced synergistic tumor suppression, which was consistent with suppression of the expression of Sp1 and its downstream target genes. Thus, treatment with bevacizumab may block VEGF function but activate the pathway of its expression via positive feedback. Given the fact that Sp1 is an important regulator of the expression of multiple angiogenic factors, bevacizumab-initiated up-regulation of Sp1 and subsequent overexpression of its downstream target genes may profoundly affect the potential angiogenic phenotype and effectiveness of antiangiogenic strategies for human pancreatic cancer. Therefore, this study is the first to show the significance and clinical implications of alteration of Sp1 signaling in antiangiogenic therapy for pancreatic cancer and other cancers.
Collapse
MESH Headings
- Adenocarcinoma/blood supply
- Adenocarcinoma/drug therapy
- Adenocarcinoma/genetics
- Adenocarcinoma/metabolism
- Angiogenesis Inhibitors/pharmacology
- Animals
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized
- Bevacizumab
- Cell Line, Tumor
- Drug Synergism
- Female
- Gene Expression/drug effects
- Humans
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Pancreatic Neoplasms/blood supply
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Plicamycin/analogs & derivatives
- Plicamycin/pharmacology
- Promoter Regions, Genetic
- Sp1 Transcription Factor/biosynthesis
- Sp1 Transcription Factor/genetics
- Sp1 Transcription Factor/metabolism
- Up-Regulation/drug effects
- Vascular Endothelial Growth Factor A/biosynthesis
- Vascular Endothelial Growth Factor A/genetics
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Zhiliang Jia
- Department of Gastrointestinal Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Lee TJ, Jung EM, Lee JT, Kim S, Park JW, Choi KS, Kwon TK. Mithramycin A sensitizes cancer cells to TRAIL-mediated apoptosis by down-regulation of XIAP gene promoter through Sp1 sites. Mol Cancer Ther 2007; 5:2737-46. [PMID: 17121920 DOI: 10.1158/1535-7163.mct-06-0426] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mithramycin A is a DNA-binding antitumor agent, which has been clinically used in the therapies of several types of cancer and Paget's disease. In this study, we investigated the combined effect of mithramycin A and tumor necrosis factor-alpha-related apoptosis-inducing ligand (TRAIL) on apoptosis of cancer cells. In Caki renal cancer cells, which are resistant to TRAIL, cotreatment with subtoxic doses of mithramycin A and TRAIL resulted in a marked increase in apoptosis. This combined treatment was also cytotoxic to Caki cells overexpressing Bcl-2 but not to normal mesengial cells. Moreover, apoptosis by the combined treatment with mithramycin A and TRAIL was dramatically induced in various cancer cell types, thus offering an attractive strategy for safely treating malignant tumors. Mithramycin A-stimulated TRAIL-induced apoptosis was blocked by pretreatment with the broad caspase inhibitor zVAD-fmk or Crm-A overexpression, showing its dependence on caspases. We found that mithramycin A selectively down-regulated XIAP protein levels in various cancer cells. Luciferase reporter assay and the chromatin immunoprecipitation assay using the XIAP promoter constructs show that mithramycin A down-regulates the transcription of XIAP gene through inhibition of Sp1 binding to its promoter. Although XIAP overexpression significantly attenuated apoptosis induced by mithramycin A plus TRAIL, suppression of XIAP expression by transfection with its small interfering RNA prominently enhanced TRAIL-induced apoptosis. We present here for the first time that mithramycin A-induced suppression of XIAP transcription plays a critical role in the recovery of TRAIL sensitivity in various cancer cells.
Collapse
Affiliation(s)
- Tae-Jin Lee
- Department of Immunology, School of Medicine, Keimyung University, 194 DongSan-Dong Jung-Gu, Taegu 700-712, South Korea
| | | | | | | | | | | | | |
Collapse
|
22
|
Albertini V, Jain A, Vignati S, Napoli S, Rinaldi A, Kwee I, Nur-e-Alam M, Bergant J, Bertoni F, Carbone GM, Rohr J, Catapano CV. Novel GC-rich DNA-binding compound produced by a genetically engineered mutant of the mithramycin producer Streptomyces argillaceus exhibits improved transcriptional repressor activity: implications for cancer therapy. Nucleic Acids Res 2006; 34:1721-34. [PMID: 16571899 PMCID: PMC1420802 DOI: 10.1093/nar/gkl063] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The aureolic acid antibiotic mithramycin (MTM) binds selectively to GC-rich DNA sequences and blocks preferentially binding of proteins, like Sp1 transcription factors, to GC-rich elements in gene promoters. Genetic approaches can be applied to alter the MTM biosynthetic pathway in the producing microorganism and obtain new products with improved pharmacological properties. Here, we report on a new analog, MTM SDK, obtained by targeted gene inactivation of the ketoreductase MtmW catalyzing the last step in MTM biosynthesis. SDK exhibited greater activity as transcriptional inhibitor compared to MTM. SDK was a potent inhibitor of Sp1-dependent reporter activity and interfered minimally with reporters of other transcription factors, indicating that it retained a high degree of selectivity toward GC-rich DNA-binding transcription factors. RT-PCR and microarray analysis showed that SDK repressed transcription of multiple genes implicated in critical aspects of cancer development and progression, including cell cycle, apoptosis, migration, invasion and angiogenesis, consistent with the pleiotropic role of Sp1 family transcription factors. SDK inhibited proliferation and was a potent inducer of apoptosis in ovarian cancer cells while it had minimal effects on viability of normal cells. The new MTM derivative SDK could be an effective agent for treatment of cancer and other diseases with abnormal expression or activity of GC-rich DNA-binding transcription factors.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mohammad Nur-e-Alam
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky725 Rose Street, Lexington, KY 40536-0082, USA
| | - Julia Bergant
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky725 Rose Street, Lexington, KY 40536-0082, USA
| | | | | | - Jürgen Rohr
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky725 Rose Street, Lexington, KY 40536-0082, USA
- To whom correspondence should be addressed. Tel: +41 91 820 0365; Fax: +41 91 820 0397;
| | - Carlo V. Catapano
- To whom correspondence should be addressed. Tel: +41 91 820 0365; Fax: +41 91 820 0397;
| |
Collapse
|
23
|
Leroy I, Laurent G, Quillet-Mary A. Mithramycin A activates Fas death pathway in leukemic cell lines. Apoptosis 2006; 11:113-9. [PMID: 16374547 DOI: 10.1007/s10495-005-3089-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Mithramycin A (MMA, trade name Plicamycin) can facilitate TNFalpha- (Tumor Necrosis Factor) and Fas ligand-induced apoptosis. Besides, several drugs play their anticancer effect through Fas apoptotic pathway. So we investigated the effect of MMA on Fas signaling. In this study we show that MMA induces apoptosis in Fas sensitive Jurkat cells and Fas resistant KG1a cells. This effect involves Fas apoptotic pathway: cell exposure to MMA leads to Fas clustering at the cell surface, DISC (Death Inducing Signaling Complex) formation and caspase cleavage. This phenomenon is independent of Fas ligand/Fas interaction and blockade of Fas death pathway partially inhibits MMA-induced apoptosis. Moreover the activation of Fas apoptotic pathway by MMA is correlated to the modulation of c-Flip(L) expression. Finally, pre-treatment with sub-lethal doses of MMA sensitizes KG1a cells to chemotherapeutic agents. Thus all these results may have important implications to improve clinical treatments.
Collapse
Affiliation(s)
- I Leroy
- INSERM U563/CPTP, Toulouse, France
| | | | | |
Collapse
|