1
|
Chattopadhyay M, Nath N, Kodela R, Metkar S, Soyemi SA, Kashfi K. NOSH-aspirin (NBS-1120) inhibits estrogen receptor-negative breast cancer in vitro and in vivo by modulating redox-sensitive signaling pathways. J Pharmacol Exp Ther 2025; 392:100019. [PMID: 39892987 DOI: 10.1124/jpet.124.002240] [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: 03/28/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/29/2024] Open
Abstract
Estrogen receptor (ER)-negative breast cancers are known to be aggressive and unresponsive to antiestrogen therapy, and triple-negative breast cancers are associated with poor prognosis and metastasis. Thus, new targeted therapies are needed. Forkhead box M1 (FOXM1) is abundantly expressed in human cancers and implicated in protecting tumor cells from oxidative stress by reducing the levels of intracellular reactive oxygen species (ROS). Aspirin, a prototypical anticancer agent with deleterious side effects that has been modified to release nitric oxide and hydrogen sulfide is called nitric oxide-hydrogen sulfide-releasing aspirin (NOSH-aspirin, NOSH-ASA), generating a "safer" class of new anti-inflammatory agents. We evaluated NOSH-ASA against ER-negative breast cancer using cell lines and a xenograft mouse model. NOSH-ASA strongly inhibited growth of MDA-MB-231 and SKBR3 breast cancer cells with low IC50s of 90 ± 5 and 82 ± 5 nM, respectively, with marginal effects on a normal breast epithelial cell line. NOSH-ASA inhibited cell proliferation, caused G0/G1 phase arrest, increased apoptosis, and was associated with increases in ROS. In MDA-MB-231 cell xenografts, NOSH-ASA reduced tumor size markedly, which was associated with reduced proliferation (decreased proliferating cell nuclear antigen expression), induction of apoptosis (increased terminal deoxynucleotidyl transferase dUTP nick end labeling-positive cells), and increased ROS, whereas nuclear factor κ-light-chain-enhancer of activated B cells and FoxM1 that were high in untreated xenografts were significantly reduced. mRNA data for FoxM1, p21, and cyclin D1 corroborated with the respective protein expressions and arrest of cells. Taken together, these molecular events contribute to NOSH-ASA-mediated growth inhibition and apoptotic death of ER-negative breast cells in vitro and in vivo. Additionally, as a ROS inducer and FOXM1 inhibitor, NOSH-ASA has potential as a targeted therapy. SIGNIFICANCE STATEMENT: We examined the cellular effects and xenograft tumor inhibitory potential of NOSH-aspirin, a nitric oxide- and hydrogen sulfide-donating hybrid, against estrogen receptor-negative breast cancer, which currently lacks effective therapeutic options. Inducing reactive oxygen species and downregulating forkhead box M1 are plausible mechanisms contributing to decreased cell proliferation and increased apoptosis. NOSH-aspirin reduced tumor size by 90% without inducing any observable gross toxicity, underscoring its promising translational potential.
Collapse
Affiliation(s)
- Mitali Chattopadhyay
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, New York
| | - Niharika Nath
- Department of Biological and Chemical Sciences, New York Institute of Technology, New York, New York
| | - Ravinder Kodela
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, New York
| | - Shalaka Metkar
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, New York
| | - Sarin A Soyemi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, New York
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, New York; Graduate Program in Biology, City University of New York Graduate Center, New York, New York.
| |
Collapse
|
2
|
Chen M, Wang Q, Wang Y, Xuan Y, Shen M, Hu X, Li Y, Guo Y, Wang J, Tan F. Thiostrepton induces oxidative stress, mitochondrial dysfunction and ferroptosis in HaCaT cells. Cell Signal 2024; 121:111285. [PMID: 38969192 DOI: 10.1016/j.cellsig.2024.111285] [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: 02/27/2024] [Revised: 06/15/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
TST has been mainly studied for its anti-tumor proliferation and antimicrobial effects, but not widely used in dermatological diseases. The mechanism of cellular damage by TST in response to H2O2-mediated oxidative stress was investigated in human skin immortalized keratinocytes (HaCaT) as an in vitro model. The findings reveal that TST treatment leads to increased oxidative stress in the cells by reducing levels of superoxide dismutase (SOD), glutathione (GSH), and catalase (CAT). This effect is further supported by an upsurge in the expression of malondialdehyde (MDA, a pivotal marker of lipid peroxidation). Additionally, dysregulation of FoxM1 at both gene and protein levels corroborates its involvement TST associated effects. Analysis of ferroptosis-related genes confirms dysregulation following TST treatment in HaCaT cells. Furthermore, TST treatment exhibits effects on mitochondrial morphology and function, affirming its induction of apoptosis in the cells through heightened oxidative stress due to mitochondrial damage and dysregulation of mitochondrial membrane potential.
Collapse
Affiliation(s)
- MeiYu Chen
- Shanghai Skin Disease Clinical College, The Fifth Clinical Medical College, Anhui Medical University, Shanghai Skin Disease Hospital, Shanghai 200443, People's Republic of China
| | - QiXia Wang
- Department of General Practice, Xi'an Central Hospital, Xi'an, Shaanxi 710000, China
| | - YaoQun Wang
- Shanghai Skin Disease Clinical College, The Fifth Clinical Medical College, Anhui Medical University, Shanghai Skin Disease Hospital, Shanghai 200443, People's Republic of China
| | - Yuan Xuan
- Shanghai Skin Disease Clinical College, The Fifth Clinical Medical College, Anhui Medical University, Shanghai Skin Disease Hospital, Shanghai 200443, People's Republic of China
| | - MengYuan Shen
- Shanghai Skin Disease Clinical College, The Fifth Clinical Medical College, Anhui Medical University, Shanghai Skin Disease Hospital, Shanghai 200443, People's Republic of China
| | - XiaoPing Hu
- Shanghai Skin Disease Clinical College, The Fifth Clinical Medical College, Anhui Medical University, Shanghai Skin Disease Hospital, Shanghai 200443, People's Republic of China
| | - YunJin Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Yi Guo
- SICU, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences, Kunming, Yunnan 650102, China
| | - Juan Wang
- Department of Ophthalmology of Shanghai Tongji Hospital, Laboratory of Clinical Visual Science of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China; Department of Medical Genetics, School of Medicine, Tongji University, Shanghai, China.
| | - Fei Tan
- Shanghai Skin Disease Clinical College, The Fifth Clinical Medical College, Anhui Medical University, Shanghai Skin Disease Hospital, Shanghai 200443, People's Republic of China; Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China.
| |
Collapse
|
3
|
Xu G, Wang J, Mao X, Xu M. 17β-estradiol Inhibits Oxidative Stress-Induced Apoptosis in Endometrial Cancer Cells by Promoting FOXM1 Expression. Cell Biochem Biophys 2024; 82:1243-1251. [PMID: 38724756 DOI: 10.1007/s12013-024-01277-x] [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] [Accepted: 04/08/2024] [Indexed: 08/25/2024]
Abstract
The steroid hormone 17β-estradiol (E2) has a significant impact on the development and progression of tumors. E2 stimulates tumor cell growth and metabolism, leading to an increase in reactive oxygen species (ROS) production. However, the rise in ROS levels is not sufficient to cause severe harm to cancer cells. and the mechanisms that regulate ROS are not well understood. Since FOXM1 plays a crucial role in the production of ROS, we aimed to investigate the impact of E2 on oxidative stress and the involvement of FOXM1 in the Ishikawa endometrial cancer cell line. Our research revealed that E2 controls the levels of ROS inside cells and safeguards them from apoptosis by promoting the expression of FOXM1. We observed a decrease in the expression of FOXM1 alongside an increase in oxidative damage. Moreover, cells demonstrated elevated levels of FOXM1 and ERα upon E2 treatment. Overall, our findings suggest that E2 prevents apoptosis induced by oxidative stress in endometrial cancer cells by encouraging the expression of FOXM1, potentially affecting ERα.
Collapse
Affiliation(s)
- Ge Xu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu, China.
| | - Jiao Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu, China.
| | - Xiaojie Mao
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu, China.
| | - Maohong Xu
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu, China
| |
Collapse
|
4
|
Merjaneh N, Hajjar M, Lan YW, Kalinichenko VV, Kalin TV. The Promise of Combination Therapies with FOXM1 Inhibitors for Cancer Treatment. Cancers (Basel) 2024; 16:756. [PMID: 38398147 PMCID: PMC10886945 DOI: 10.3390/cancers16040756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/21/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Forkhead box M1 (FOXM1) is a transcription factor in the forkhead (FOX) family, which is required for cellular proliferation in normal and neoplastic cells. FOXM1 is highly expressed in many different cancers, and its expression is associated with a higher tumor stage and worse patient-related outcomes. Abnormally high expression of FOXM1 in cancers compared to normal tissue makes FOXM1 an attractive target for pharmacological inhibition. FOXM1-inhibiting agents and specific FOXM1-targeted small-molecule inhibitors have been developed in the lab and some of them have shown promising efficacy and safety profiles in mouse models. While the future goal is to translate FOXM1 inhibitors to clinical trials, potential synergistic drug combinations can maximize anti-tumor efficacy while minimizing off-target side effects. Hence, we discuss the rationale and efficacy of all previously studied drug combinations with FOXM1 inhibitors for cancer therapies.
Collapse
Affiliation(s)
- Nawal Merjaneh
- Center for Cancer and Blood Disorders, Phoenix Children’s Hospital, Phoenix, AZ 85016, USA
- Department of Child Health, Division of Hematology and Oncology, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
| | - Mona Hajjar
- The Columbian College of Arts and Sciences, George Washington University, Washington, DC 20052, USA;
| | - Ying-Wei Lan
- Phoenix Children’s Research Institute, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA; (Y.-W.L.)
| | - Vladimir V. Kalinichenko
- Phoenix Children’s Research Institute, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA; (Y.-W.L.)
- Division of Neonatology, Phoenix Children’s Hospital, Phoenix, AZ 85016, USA
| | - Tanya V. Kalin
- Center for Cancer and Blood Disorders, Phoenix Children’s Hospital, Phoenix, AZ 85016, USA
- Department of Child Health, Division of Hematology and Oncology, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
- Phoenix Children’s Research Institute, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA; (Y.-W.L.)
| |
Collapse
|
5
|
Miller ZA, Mueller A, Kim T, Jolivert JF, Ma RZ, Muthuswami S, Park A, McMahon DB, Nead KT, Carey RM, Lee RJ. Lidocaine induces apoptosis in head and neck squamous cell carcinoma through activation of bitter taste receptor T2R14. Cell Rep 2023; 42:113437. [PMID: 37995679 PMCID: PMC10842818 DOI: 10.1016/j.celrep.2023.113437] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 09/22/2023] [Accepted: 10/29/2023] [Indexed: 11/25/2023] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) have high mortality and significant treatment-related morbidity. It is vital to discover effective, minimally invasive therapies that improve survival and quality of life. Bitter taste receptors (T2Rs) are expressed in HNSCCs, and T2R activation can induce apoptosis. Lidocaine is a local anesthetic that also activates bitter taste receptor 14 (T2R14). Lidocaine has some anti-cancer effects, but the mechanisms are unclear. Here, we find that lidocaine causes intracellular Ca2+ mobilization through activation of T2R14 in HNSCC cells. T2R14 activation with lidocaine depolarizes mitochondria, inhibits proliferation, and induces apoptosis. Concomitant with mitochondrial Ca2+ influx, ROS production causes T2R14-dependent accumulation of poly-ubiquitinated proteins, suggesting that proteasome inhibition contributes to T2R14-induced apoptosis. Lidocaine may have therapeutic potential in HNSCCs as a topical gel or intratumor injection. In addition, we find that HPV-associated (HPV+) HNSCCs are associated with increased TAS2R14 expression. Lidocaine treatment may benefit these patients, warranting future clinical studies.
Collapse
Affiliation(s)
- Zoey A Miller
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Pharmacology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Arielle Mueller
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - TaeBeom Kim
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer F Jolivert
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ray Z Ma
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Sahil Muthuswami
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - April Park
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Derek B McMahon
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kevin T Nead
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ryan M Carey
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - Robert J Lee
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
6
|
Cheng W, Cai C, Xu Y, Xiao X, Shi T, Liao Y, Wang X, Chen S, Zhou M, Liao Z. The TRIM21-FOXD1-BCL-2 axis underlies hyperglycaemic cell death and diabetic tissue damage. Cell Death Dis 2023; 14:825. [PMID: 38092733 PMCID: PMC10719266 DOI: 10.1038/s41419-023-06355-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 11/26/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023]
Abstract
Chronic hyperglycaemia is a devastating factor that causes diabetes-induced damage to the retina and kidney. However, the precise mechanism by which hyperglycaemia drives apoptotic cell death is incompletely known. Herein, we found that FOXD1, a FOX family transcription factor specifically expressed in the retina and kidney, regulated the transcription of BCL-2, a master regulator of cell survival. Intriguingly, the protein level of FOXD1, which responded negatively to hyperglycaemic conditions, was controlled by the TRIM21-mediated K48-linked polyubiquitination and subsequent proteasomal degradation. The TRIM21-FOXD1-BCL-2 signalling axis was notably active during diabetes-induced damage to murine retinal and renal tissues. Furthermore, we found that tartary buckwheat flavonoids effectively reversed the downregulation of FOXD1 protein expression and thus restored BCL-2 expression and facilitated the survival of retinal and renal tissues. In summary, we identified a transcription factor responsible for BCL-2 expression, a signalling axis (TRM21-FOXD1-BCL-2) underlying hyperglycaemia-triggered apoptosis, and a potential treatment for deleterious diabetic complications.
Collapse
Affiliation(s)
- Wenwen Cheng
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Cifeng Cai
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Yifan Xu
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Xueqi Xiao
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Tiantian Shi
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Yueling Liao
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Xiaoyi Wang
- First Affiliated Hospital of Huzhou University, Huzhou, 313000, China
| | - Shasha Chen
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China.
| | - Meiliang Zhou
- Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100081, China.
| | - Zhiyong Liao
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China.
| |
Collapse
|
7
|
Kuthethur R, Adiga D, Kandettu A, Jerome MS, Mallya S, Mumbrekar KD, Kabekkodu SP, Chakrabarty S. MiR-4521 perturbs FOXM1-mediated DNA damage response in breast cancer. Front Mol Biosci 2023; 10:1131433. [PMID: 37025658 PMCID: PMC10070856 DOI: 10.3389/fmolb.2023.1131433] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 03/06/2023] [Indexed: 04/08/2023] Open
Abstract
Introduction: Forkhead (FOX) transcription factors are involved in cell cycle control, cellular differentiation, maintenance of tissues, and aging. Mutation or aberrant expression of FOX proteins is associated with developmental disorders and cancers. FOXM1, an oncogenic transcription factor, is a promoter of cell proliferation and accelerated development of breast adenocarcinomas, squamous carcinoma of the head, neck, and cervix, and nasopharyngeal carcinoma. High FOXM1 expression is correlated with chemoresistance in patients treated with doxorubicin and Epirubicin by enhancing the DNA repair in breast cancer cells. Method: miRNA-seq identified downregulation of miR-4521 in breast cancer cell lines. Stable miR-4521 overexpressing breast cancer cell lines (MCF-7, MDA-MB-468) were developed to identify miR-4521 target gene and function in breast cancer. Results: Here, we showed that FOXM1 is a direct target of miR-4521 in breast cancer. Overexpression of miR-4521 significantly downregulated FOXM1 expression in breast cancer cells. FOXM1 regulates cell cycle progression and DNA damage response in breast cancer. We showed that miR-4521 expression leads to increased ROS levels and DNA damage in breast cancer cells. FOXM1 plays a critical role in ROS scavenging and promotes stemness which contributes to drug resistance in breast cancer. We observed that breast cancer cells stably expressing miR-4521 lead to cell cycle arrest, impaired FOXM1 mediated DNA damage response leading to increased cell death in breast cancer cells. Additionally, miR-4521-mediated FOXM1 downregulation perturbs cell proliferation, invasion, cell cycle progression, and epithelial-to-mesenchymal progression (EMT) in breast cancer. Discussion: High FOXM1 expression has been associated with radio and chemoresistance contributing to poor patient survival in multiple cancers, including breast cancer. Our study showed that FOXM1 mediated DNA damage response could be targeted using miR-4521 mimics as a novel therapeutic for breast cancer.
Collapse
Affiliation(s)
- Raviprasad Kuthethur
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Amoolya Kandettu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Maria Sona Jerome
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sandeep Mallya
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Kamalesh Dattaram Mumbrekar
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
- Center for DNA Repair and Genome Stability (CDRGS), Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sanjiban Chakrabarty
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
- Center for DNA Repair and Genome Stability (CDRGS), Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
8
|
Kaynak A, N’Guessan KF, Patel PH, Lee JH, Kogan AB, Narmoneva DA, Qi X. Electric Fields Regulate In Vitro Surface Phosphatidylserine Exposure of Cancer Cells via a Calcium-Dependent Pathway. Biomedicines 2023; 11:biomedicines11020466. [PMID: 36831002 PMCID: PMC9953458 DOI: 10.3390/biomedicines11020466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/23/2023] [Accepted: 01/30/2023] [Indexed: 02/09/2023] Open
Abstract
Cancer is the second leading cause of death worldwide after heart disease. The current treatment options to fight cancer are limited, and there is a critical need for better treatment strategies. During the last several decades, several electric field (EF)-based approaches for anti-cancer therapies have been introduced, such as electroporation and tumor-treating fields; still, they are far from optimal due to their invasive nature, limited efficacy and significant side effects. In this study, we developed a non-contact EF stimulation system to investigate the in vitro effects of a novel EF modality on cancer biomarkers in normal (human astrocytes, human pancreatic ductal epithelial -HDPE-cells) and cancer cell lines (glioblastoma U87-GBM, human pancreatic cancer cfPac-1, and MiaPaCa-2). Our results demonstrate that this EF modality can successfully modulate an important cancer cell biomarker-cell surface phosphatidylserine (PS). Our results further suggest that moderate, but not low, amplitude EF induces p38 mitogen-activated protein kinase (MAPK), actin polymerization, and cell cycle arrest in cancer cell lines. Based on our results, we propose a mechanism for EF-mediated PS exposure in cancer cells, where the magnitude of induced EF on the cell surface can differentially regulate intracellular calcium (Ca2+) levels, thereby modulating surface PS exposure.
Collapse
Affiliation(s)
- Ahmet Kaynak
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Kombo F. N’Guessan
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Priyankaben H. Patel
- Department of Biomedical Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Jing-Huei Lee
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Andrei B. Kogan
- Department of Physics, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Daria A. Narmoneva
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Xiaoyang Qi
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Correspondence: ; Tel.: +1-513-558-4025
| |
Collapse
|
9
|
Poudel S, Martins G, Cancela ML, Gavaia PJ. Resveratrol-Mediated Reversal of Doxorubicin-Induced Osteoclast Differentiation. Int J Mol Sci 2022; 23:ijms232315160. [PMID: 36499492 PMCID: PMC9738652 DOI: 10.3390/ijms232315160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/16/2022] [Accepted: 11/29/2022] [Indexed: 12/05/2022] Open
Abstract
Secondary osteoporosis has been associated with cancer patients undertaking Doxorubicin (DOX) chemotherapy. However, the molecular mechanisms behind DOX-induced bone loss have not been elucidated. Molecules that can protect against the adverse effects of DOX are still a challenge in chemotherapeutic treatments. We investigated the effect and mechanism of DOX in osteoclast differentiation and used the Sirt 1 activator resveratrol (RES) to counteract DOX-induced effects. RAW 264.7 cells were differentiated into osteoclasts under cotreatment with DOX and RES, alone or combined. RES treatment inhibited DOX-induced osteoclast differentiation, reduced the expression of osteoclast fusion marker Oc-stamp and osteoclast differentiation markers Rank, Trap, Ctsk and Nfatc1. Conversely, RES induced the upregulation of antioxidant genes Sod 1 and Nrf 2 while DOX significantly reduced the FoxM1 expression, resulting in oxidative stress. Treatment with the antioxidant MitoTEMPO did not influence DOX-induced osteoclast differentiation. DOX-induced osteoclastogenesis was studied using the cathepsin-K zebrafish reporter line (Tg[ctsk:DsRed]). DOX significantly increased ctsk signal, while RES cotreatment resulted in a significant reduction in ctsk positive cells. RES significantly rescued DOX-induced mucositis in this model. Additionally, DOX-exposed zebrafish displayed altered locomotor behavior and locomotory patterns, while RES significantly reversed these effects. Our research shows that RES prevents DOX-induced osteoclast fusion and activation in vitro and in vivo and reduces DOX-induced mucositis, while improving locomotion parameters.
Collapse
Affiliation(s)
- Sunil Poudel
- Centre of Marine Sciences, University of Algarve, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, 8005-139 Faro, Portugal
- PhD Program in Biomedical Sciences, FMCB, University of Algarve, 8005-139 Faro, Portugal
| | - Gil Martins
- Centre of Marine Sciences, University of Algarve, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, 8005-139 Faro, Portugal
- PhD Program in Biomedical Sciences, FMCB, University of Algarve, 8005-139 Faro, Portugal
| | - M. Leonor Cancela
- Centre of Marine Sciences, University of Algarve, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, 8005-139 Faro, Portugal
- Algarve Biomedical Center, University of Algarve, 8005-139 Faro, Portugal
| | - Paulo J. Gavaia
- Centre of Marine Sciences, University of Algarve, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, 8005-139 Faro, Portugal
- Correspondence: ; Tel.: +351-289-800057 or +351-289-800900 (ext. 7057)
| |
Collapse
|
10
|
Liu SX, Zhou Y, Zhao L, Zhou LS, Sun J, Liu GJ, Du YS, Zhou YN. Thiostrepton confers protection against reactive oxygen species-related apoptosis by restraining FOXM1-triggerred development of gastric cancer. Free Radic Biol Med 2022; 193:385-404. [PMID: 36152915 DOI: 10.1016/j.freeradbiomed.2022.09.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 09/06/2022] [Accepted: 09/17/2022] [Indexed: 12/01/2022]
Abstract
Gastric cancer is a leading cause of tumor-associated death worldwide. Metastasis and chemoresistance are crucial barriers for gastric cancer treatment. The Forkhead Box M1 (FOXM1) transcription factor has been reported as a promising treatment target for various types of tumors, but its effects on gastric cancer progression are not fully understood. In the present study, we found that FOXM1 expression levels were significantly up-regulated in human gastric cancer cell lines and tissues, and its expression was much higher in patients with metastasis. We then found that suppressing FOXM1 with its inhibitor thiostrepton (THIO) significantly reduced the proliferation of gastric cancer cells, while induced G0/G1 and apoptosis. Moreover, reactive oxygen species (ROS) production, mitochondrial impair and autophagy were remarkably provoked in gastric cancer cells treated with THIO, which were required for the regulation of apoptotic cell death. Furthermore, THIO exposure considerably suppressed the migration, invasion and angiogenesis in gastric cancer cells. The inhibitory effects of THIO on tumor growth and metastasis were confirmed in an established gastric cancer xenograft mouse model without detectable toxicity. Intriguingly, our in vitro studies showed that the anti-cancer effects of THIO on gastric cancer were almost abolished upon FOXM1 over-expression, indicating the necessity of FOXM1 suppression in THIO-inhibited tumor growth. In addition, higher FOXM1 expression was detected in gastric cancer cells with chemoresistance. Both in vitro and in vivo studies illustrated that THIO strongly promoted the drug-resistant gastric cancer cells to chemotherapies, proved by the considerably decreased cell proliferation and epithelial-mesenchymal transition (EMT) process. Together, these findings revealed that FOXM1 was a promising therapeutic target for gastric cancer treatment, and THIO exerted potential as an therapeutic agent for the disease.
Collapse
Affiliation(s)
- Shi-Xiong Liu
- Department of Geriatrics (II), The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Yun Zhou
- Department of Geriatrics (II), The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Li Zhao
- Department of Medical Ultrasound, The Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, 730020, China
| | - Ling-Shan Zhou
- Department of Geriatrics (II), The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Jie Sun
- Department of Geriatrics (I), The First Hospital of Lanzhou University, Lanzho, 730000, China
| | - Ge-Jing Liu
- Department of Geriatrics (I), The First Hospital of Lanzhou University, Lanzho, 730000, China
| | - Ying-Shi Du
- Department of Geriatrics (I), The First Hospital of Lanzhou University, Lanzho, 730000, China
| | - Yong-Ning Zhou
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
11
|
Wei WS, Wang N, Deng MH, Dong P, Liu JY, Xiang Z, Li XD, Li ZY, Liu ZH, Peng YL, Li Z, Jiang LJ, Yao K, Ye YL, Lu WH, Zhang ZL, Zhou FJ, Liu ZW, Xie D, Yu CP. LRPPRC regulates redox homeostasis via the circANKHD1/FOXM1 axis to enhance bladder urothelial carcinoma tumorigenesis. Redox Biol 2021; 48:102201. [PMID: 34864630 PMCID: PMC8645923 DOI: 10.1016/j.redox.2021.102201] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 01/03/2023] Open
Abstract
Reactive oxygen species (ROS) which are continuously generated mainly by mitochondria, have been proved to play an important role in the stress signaling of cancer cells. Moreover, pentatricopeptide repeat (PPR) proteins have been suggested to take part in mitochondrial metabolism. However, the mechanisms integrating the actions of these distinct networks in urothelial carcinoma of the bladder (UCB) pathogenesis are elusive. In this study, we found that leucine rich pentatricopeptide repeat containing (LRPPRC) was frequently upregulated in UCB and that it was an independent prognostic factor in UCB. We further revealed that LRPPRC promoted UCB tumorigenesis by regulating the intracellular ROS homeostasis. Mechanistically, LRPPRC modulates ROS balance and protects UCB cells from oxidative stress via mt-mRNA metabolism and the circANKHD1/FOXM1 axis. In addition, the SRA stem-loop interacting RNA binding protein (SLIRP) directly interacted with LRPPRC to protect it from ubiquitination and proteasomal degradation. Notably, we showed that LRPPRC modulated the tumorigenesis of UCB cells in a circANKHD1-FOXM1-dependent manner. In conclusion, LRPPRC exerts critical roles in regulating UCB redox homeostasis and tumorigenesis, and is a prognostic factor for UCB; suggesting that LRPPRC may serve as an exploitable therapeutic target in UCB.
Collapse
Affiliation(s)
- Wen-Su Wei
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Ning Wang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Min-Hua Deng
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Pei Dong
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Jian-Ye Liu
- Department of Urology, Xiangya Third Hospital, No. 106, 2nd Zhongshan Road, Changsha, PR China
| | - Zhen Xiang
- Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China
| | - Xiang-Dong Li
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Zhi-Yong Li
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Zhen-Hua Liu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Yu-Lu Peng
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Zhen Li
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Li-Juan Jiang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Kai Yao
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Yun-Lin Ye
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Wen-Hua Lu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Zhi-Ling Zhang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Fang-Jian Zhou
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China
| | - Zhuo-Wei Liu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China.
| | - Dan Xie
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, PR China.
| | - Chun-Ping Yu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, PR China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, PR China.
| |
Collapse
|
12
|
Liu C, Barger CJ, Karpf AR. FOXM1: A Multifunctional Oncoprotein and Emerging Therapeutic Target in Ovarian Cancer. Cancers (Basel) 2021; 13:3065. [PMID: 34205406 PMCID: PMC8235333 DOI: 10.3390/cancers13123065] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 02/08/2023] Open
Abstract
Forkhead box M1 (FOXM1) is a member of the conserved forkhead box (FOX) transcription factor family. Over the last two decades, FOXM1 has emerged as a multifunctional oncoprotein and a robust biomarker of poor prognosis in many human malignancies. In this review article, we address the current knowledge regarding the mechanisms of regulation and oncogenic functions of FOXM1, particularly in the context of ovarian cancer. FOXM1 and its associated oncogenic transcriptional signature are enriched in >85% of ovarian cancer cases and FOXM1 expression and activity can be enhanced by a plethora of genomic, transcriptional, post-transcriptional, and post-translational mechanisms. As a master transcriptional regulator, FOXM1 promotes critical oncogenic phenotypes in ovarian cancer, including: (1) cell proliferation, (2) invasion and metastasis, (3) chemotherapy resistance, (4) cancer stem cell (CSC) properties, (5) genomic instability, and (6) altered cellular metabolism. We additionally discuss the evidence for FOXM1 as a cancer biomarker, describe the rationale for FOXM1 as a cancer therapeutic target, and provide an overview of therapeutic strategies used to target FOXM1 for cancer treatment.
Collapse
Affiliation(s)
| | | | - Adam R. Karpf
- Eppley Institute and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68918-6805, USA; (C.L.); (C.J.B.)
| |
Collapse
|
13
|
Tabatabaei Dakhili SA, Pérez DJ, Gopal K, Haque M, Ussher JR, Kashfi K, Velázquez-Martínez CA. SP1-independent inhibition of FOXM1 by modified thiazolidinediones. Eur J Med Chem 2020; 209:112902. [PMID: 33069434 DOI: 10.1016/j.ejmech.2020.112902] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/16/2020] [Accepted: 09/28/2020] [Indexed: 11/25/2022]
Abstract
This research article describes an approach to modify the thiazolidinedione scaffold to produce test drugs capable of binding to, and inhibit, the in vitro transcriptional activity of the oncogenic protein FOXM1. This approach allowed us to obtain FOXM1 inhibitors that bind directly to the FOXM1-DNA binding domain without targeting the expression levels of Sp1, an upstream transcription factor protein known to activate the expression of FOXM1. Briefly, we modified the chemical structure of the thiazolidinedione scaffold present in anti-diabetic medications such as pioglitazone, rosiglitazone and the former anti-diabetic drug troglitazone, because these drugs have been reported to exert inhibition of FOXM1 but hit other targets as well. After the chemical synthesis of 11 derivatives possessing a modified thiazolidinedione moiety, we screened all test compounds using in vitro protocols to measure their ability to (a) dissociate a FOXM1-DNA complex (EMSA assay); (b) decrease the expression of FOXM1 in triple negative-breast cancer cells (WB assay); (c) downregulate the expression of FOXM1 downstream targets (luciferase reporter assays and qPCR); and inhibit the formation of colonies of MDA-MB-231 cancer cells (colony formation assay). We also identified a potential binding mode associated with these compounds in which compound TFI-10, one of the most active molecules, exerts binding interactions with Arg289, Trp308, and His287. Unlike the parent drug, troglitazone, compound TFI-10 does not target the in vitro expression of Sp1, suggesting that it is possible to design FOXM1 inhibitors with a better selectivity profile.
Collapse
Affiliation(s)
| | - David J Pérez
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada; Unidad Radiofarmacia-Ciclotrón, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Keshav Gopal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Moinul Haque
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada; Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Khosrow Kashfi
- Department of Molecular, Cellular, & Biomedical Sciences, City University of New York School of Medicine, New York, USA; Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, USA
| | | |
Collapse
|
14
|
FoxM1-dependent and fatty acid oxidation-mediated ROS modulation is a cell-intrinsic drug resistance mechanism in cancer stem-like cells. Redox Biol 2020; 36:101589. [PMID: 32521504 PMCID: PMC7286985 DOI: 10.1016/j.redox.2020.101589] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/01/2020] [Accepted: 05/17/2020] [Indexed: 12/12/2022] Open
Abstract
Increased oxidative phosphorylation (OXPHOS) and reactive oxygen species (ROS) levels are inherently linked. ROS are essential signaling molecules, with detrimental effects when produced in excess during chemotherapy, leading to cell death. Cancer stem-like cells (CSCs) are a subpopulation of tumor cells resistant to chemotherapy, highly invasive and metastagenic, driving malignant cancer behavior. In this study, we demonstrated that CSCs exhibit increased OXPHOS but paradoxically low ROS levels. Considering the detrimental effects of large amounts of ROS, CSCs have developed potential mechanisms for quenching excess ROS to maintain redox homeostasis. We aimed to investigate the distinct metabolic features and mechanisms of ROS regulation in gastric CSCs and explore potential therapeutic strategies targeting CSCs. Human gastric cancer cell lines, AGS and MKN1, were subjected to liquid chromatography/mass spectrometry-based metabolomic and microarray analyses. Mitochondrial properties such as mitochondrial mass, membrane potential, and ROS were assessed by flow cytometric analysis. CSCs with increased OXPHOS levels maintained low ROS levels by coupling FoxM1-dependent Prx3 expression and fatty acid oxidation-mediated NADPH regeneration. Thus, interventions targeting ROS homeostasis in CSCs may be a useful strategy for targeting this drug-resistant tumor cell subpopulation.
Collapse
|
15
|
Lee HA, Chu KB, Moon EK, Kim SS, Quan FS. Sensitization to oxidative stress and G2/M cell cycle arrest by histone deacetylase inhibition in hepatocellular carcinoma cells. Free Radic Biol Med 2020; 147:129-138. [PMID: 31870798 DOI: 10.1016/j.freeradbiomed.2019.12.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 12/13/2019] [Accepted: 12/18/2019] [Indexed: 12/13/2022]
Abstract
Oxidative stress resistance in cancer cells has contributed to multi-drug resistance, which poses a serious challenge to cancer therapy. To surmount this, combinatorial treatment involving anticancer drugs and histone deacetylase inhibitors (HDACi) have emerged as a chemotherapeutic option. Yet, HDACi's role in redox states of cancer cells still requires elucidation. In the present study, we hypothesized that HDACi sensitizes cancer cells to oxidative stress and results in G2/M cell cycle arrest. Cell viability and cell cycle were analyzed using Cell Counting Kit 8 (CCK8) and fluorescent activated cell sorting (FACS), respectively. The transcriptomes of cells were investigated by massive analysis of cDNA end (MACE). Expression of mRNA and proteins were analyzed by quantitative real-time PCR (qPCR) and Western blot, respectively. Intracellular oxidative stress induced by tert-Butyl hydroperoxide (tBHP) reduced cell viability and resulted in G2/M cell cycle arrest in a dose-dependent manner in hepatocellular carcinoma (HCC) cells. The effects of sorafenib on cell cycle arrest and HCC viability were enhanced through HDACi treatment. MACE revealed that genes related to progression of G2/M cell cycle including Foxm1, Aurka, Plk1, and Ccnb1 were significantly down-regulated in tBHP and HDACi-treated HepG2 cells. Inhibition of FOXM1 with thiostrepton also resulted in reduced cell viability and expression of FOXM1 target genes such as Aurka, Plk1, and Ccnb1. These results indicate that HDACi sensitizes HepG2 cells to oxidative stress and results in G2/M cell cycle arrest via down-regulation of FOXM1, which plays a key role in progression of G2/M cell cycle.
Collapse
Affiliation(s)
- Hae-Ahm Lee
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ki-Back Chu
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Eun-Kyung Moon
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Sung Soo Kim
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Biochemistry and Molecular Biology, Kyung Hee University School of Medicine, Seoul, 02447, Republic of Korea
| | - Fu-Shi Quan
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
16
|
Ma J, Liu B, Yu D, Zuo Y, Cai R, Yang J, Cheng J. SIRT3 deacetylase activity confers chemoresistance in AML via regulation of mitochondrial oxidative phosphorylation. Br J Haematol 2019; 187:49-64. [PMID: 31236919 PMCID: PMC6790595 DOI: 10.1111/bjh.16044] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/15/2019] [Accepted: 04/16/2019] [Indexed: 12/14/2022]
Abstract
Acute myeloid leukaemia (AML) cells possess metabolism profiles, such as higher rates of oxidative phosphorylation and dependence on fatty acid oxidation for survival, and are dependent on the sophisticated regulation of reactive oxygen species (ROS) generation for survival, drug resistance and stemness maintenance. We found that sensitivity of primary AML cells to cytarabine correlated with SOD2 acetylation and the ability of the drug to induce mitochondrial ROS. The SOD2 deacetylase, SIRT3, protected AML cells from chemotherapy as shown by inhibited apoptosis via inhibited drug-induced production of mitochondrial ROS. SIRT3 significantly decreased nicotinamide adenine dinucleotide phosphate (NADP)/reduced NADP ratio and increased reduced glutathione/oxidized glutathione ratio. Furthermore, SIRT3 enhanced oxidative phosphorylation (OxPhos) in AML cells under both basic and cytarabine-treated conditions. A xenograft mouse model showed that SIRT3 overexpressing AML cells and patient-derived xenograft mice bearing high SIRT3 deacetylase activity were more resistant to chemotherapy in vivo. SIRT3 inhibitor displayed synergy with cytarabine to ablate AML cells in vitro and in mouse models. Taken together, our study showed that SIRT3 is capable of reprograming mitochondrial metabolism towards OxPhos and downregulating ROS generation, which contribute to the chemoresistance of AML cells. SIRT3 can be utilized as a potential therapeutic target to improve the anti-leukaemic efficacy of standard chemotherapeutic agents for AML.
Collapse
MESH Headings
- Acetylation
- Adult
- Aged
- Animals
- Antimetabolites, Antineoplastic/pharmacology
- Antimetabolites, Antineoplastic/therapeutic use
- Apoptosis/drug effects
- Apoptosis/physiology
- Cytarabine/pharmacology
- Cytarabine/therapeutic use
- Drug Resistance, Neoplasm/physiology
- Female
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/enzymology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Male
- Mice, SCID
- Middle Aged
- Mitochondria/metabolism
- Oxidative Phosphorylation
- Reactive Oxygen Species/metabolism
- Sirtuin 3/physiology
- Superoxide Dismutase/metabolism
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Jiao Ma
- Department of Biochemistry and Molecular Cell BiologyShanghai Jiaotong University School of MedicineShanghaiChina
| | - Bin Liu
- Department of HaematologyChanghai Hospital of the Second Military Medical UniversityShanghaiChina
- Department of Respiratory MedicineThe People's Liberation Army General HospitalShanghaiChina
| | - Dan Yu
- Department of Biochemistry and Molecular Cell BiologyShanghai Jiaotong University School of MedicineShanghaiChina
| | - Yong Zuo
- Department of Biochemistry and Molecular Cell BiologyShanghai Jiaotong University School of MedicineShanghaiChina
| | - Rong Cai
- Department of Biochemistry and Molecular Cell BiologyShanghai Jiaotong University School of MedicineShanghaiChina
| | - Jianmin Yang
- Department of HaematologyChanghai Hospital of the Second Military Medical UniversityShanghaiChina
| | - Jinke Cheng
- Department of Biochemistry and Molecular Cell BiologyShanghai Jiaotong University School of MedicineShanghaiChina
| |
Collapse
|
17
|
High impact of miRNA-4521 on FOXM1 expression in medulloblastoma. Cell Death Dis 2019; 10:696. [PMID: 31541075 PMCID: PMC6754377 DOI: 10.1038/s41419-019-1926-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 08/06/2019] [Accepted: 08/27/2019] [Indexed: 12/11/2022]
Abstract
Medulloblastoma, an embryonal tumor of the cerebellum/fourth ventricle, is one of the most frequent malignant brain tumors in children. Although genetic variants are increasingly used in treatment stratification, survival of high-risk patients, characterized by leptomeningeal dissemination, TP53 mutation or MYC amplification, is still poor. FOXM1, a proliferation-specific oncogenic transcription factor, is deregulated in various solid tumors, including medulloblastoma, and triggers cellular proliferation, migration and genomic instability. In tissue samples obtained from medulloblastoma patients, the significant upregulation of FOXM1 was associated with a loss of its putative regulating microRNA, miR-4521. To understand the underlying mechanism, we investigated the effect of miR-4521 on the expression of the transcription factor FOXM1 in medulloblastoma cell lines. Transfection of this microRNA reduced proliferation and invasion of several medulloblastoma cell lines and induced programmed cell death through activation of caspase 3/7. Further, downstream targets of FOXM1 such as PLK1 and cyclin B1 were significantly reduced thus affecting the cell cycle progression in medulloblastoma cell lines. In conclusion, a restoration of miRNA-4521 may selectively suppress the pathophysiological effect of aberrant FOXM1 expression and serve as a targeted approach for medulloblastoma therapy.
Collapse
|
18
|
Mascarenhas S, Mutnuri S, Ganguly A. Silica - A trace geogenic element with emerging nephrotoxic potential. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 645:297-317. [PMID: 30029111 DOI: 10.1016/j.scitotenv.2018.07.075] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 06/14/2018] [Accepted: 07/06/2018] [Indexed: 06/08/2023]
Abstract
Silica is a trace-geogenic compound with limited-bioavailability. It inflicts health-perils like pulmonary-silicosis and chronic kidney disease (CKD), when available via anthropogenic-disturbances. Amidst silica-imposed pathologies, pulmonary toxicological-mechanisms are well-described, ignoring the renal-pathophysiological mechanisms. Hence, the present-study aimed to elucidate cellular-cum-molecular toxicological-mechanisms underlying silica-induced renal-pathology in-vitro. Various toxicity-assessments were used to study effects of silica on the physiological-functions of HK-cells (human-kidney proximal-tubular cells - the toxin's prime target) on chronic (1-7 days) sub-toxic (80 mg/L) and toxic (100-120 mg/L) dosing. Results depicted that silica triggered dose-cum-time dependent cytotoxicity/cell-death (MTT-assay) that significantly increased on long-term dosing with ≥100 mg/L silica; establishing the nephrotoxic-potential of this dose. Contrarily, insignificant cell-death on sub-toxic (80 mg/L) dosing was attributed to rapid intracellular toxin-clearance at lower-doses preventing toxic-effects. The proximal-tubular (HK-cells) cytotoxicity was found to be primarily mediated by silica-triggered incessant oxidative-stress (elevated ROS).·This enhanced ROS inflicted severe inflammation and subsequent fibrosis, evident from increased pro-inflammatory-cum-fibrogenic cytokines generation (IL-1β, IL-2, IL-6, TNF-α and TGF-β). Simultaneously, ROS induced persistent DNA-damage (Comet-assay) that stimulated G2/M arrest for p53-mediated damage-repair, aided by checkpoint-promoter (Chk1) activation and mitotic-inducers (i.e. Cdc-25, Cdk1, cyclinB1) inhibition. However, DNA-injuries surpassed the cellular-repair, which provoked the p53-gene to induce mitochondrial-mediated apoptotic cell-death via activation of Bax, cytochrome-c and caspase-cascade (9/3). This persistent apoptotic cell-death and simultaneous incessant inflammation culminated in the development of tubular-atrophy and fibrosis, the major pathological-manifestations of CKD. These findings provided novel-insights into the pathological-mechanisms (cellular and molecular) of silica-induced CKD, inflicted on chronic toxic-dosing (≥100 mg/L).Thereby, encouraging the development of therapeutic-strategies (e.g. anti-oxidant treatment) for specific molecular-targets (e.g. ROS) to retard silica-induced CKD-progression, for reduction in the global-CKD burden.
Collapse
Affiliation(s)
- Starlaine Mascarenhas
- Department of Biological Sciences, BITS Pilani, K K Birla Goa Campus, NH 17 B, Zuarinagar, Goa 403 726, India.
| | - Srikanth Mutnuri
- Department of Biological Sciences, BITS Pilani, K K Birla Goa Campus, NH 17 B, Zuarinagar, Goa 403 726, India.
| | - Anasuya Ganguly
- Department of Biological Sciences, BITS Pilani, K K Birla Goa Campus, NH 17 B, Zuarinagar, Goa 403 726, India.
| |
Collapse
|
19
|
Lee VS, McRobb LS, Moutrie V, Santos ED, Siu TL. Effects of FOXM1 inhibition and ionizing radiation on melanoma cells. Oncol Lett 2018; 16:6822-6830. [PMID: 30405826 DOI: 10.3892/ol.2018.9482] [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: 02/06/2018] [Accepted: 07/26/2018] [Indexed: 12/18/2022] Open
Abstract
Metastatic melanoma can be highly refractory to conventional radiotherapy and chemotherapy but combinatorial-targeted therapeutics are showing greater promise on improving treatment efficacy. Previous studies have shown that knockdown of Forkhead box M1 (FOXM1) can sensitize various tumor types to radiation-induced cell death. The effect of combining radiation with a small molecule FOXM1 inhibitor, Siomycin A, on growth, death and migration of a metastatic melanoma cell line (SK-MEL-28) that overexpresses this pleiotropic cell cycle regulator was investigated. Siomycin A (SIOA) was found to be a strong inducer of apoptosis, and inhibitor of proliferation and migration in a scratch wound assay in this cell line. Induction of apoptosis occurred at concentrations >1 µM in association with reductions in the constitutive FOXM1 and anti-apoptotic B-cell lymphoma 2 protein levels found in these cells. Single doses of ionizing radiation (0-40 Gy) delivered by linear accelerator caused inhibition of growth and migration without significant induction of cell death. Pretreatment with SIOA did not increase the sensitivity of this melanoma cell line to radiation as observed in other tumor types. These data confirm that as a single agent, SIOA is an effective inducer of cell death and inhibitor of migration in metastatic melanoma cells expressing constitutive FOXM1. In combination with radiation, SIOA pre-treatment, however, may not be of added benefit.
Collapse
Affiliation(s)
- Vivienne S Lee
- Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Lucinda S McRobb
- Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Vaughan Moutrie
- Genesis Cancer Care, Macquarie University Hospital, Sydney, New South Wales 2109, Australia
| | - Estavam D Santos
- Genesis Cancer Care, Macquarie University Hospital, Sydney, New South Wales 2109, Australia
| | - Timothy L Siu
- Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| |
Collapse
|
20
|
Gwangwa MV, Joubert AM, Visagie MH. Crosstalk between the Warburg effect, redox regulation and autophagy induction in tumourigenesis. Cell Mol Biol Lett 2018; 23:20. [PMID: 29760743 PMCID: PMC5935986 DOI: 10.1186/s11658-018-0088-y] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/27/2018] [Indexed: 02/07/2023] Open
Abstract
Tumourigenic tissue uses modified metabolic signalling pathways in order to support hyperproliferation and survival. Cancer-associated aerobic glycolysis resulting in lactic acid production was described nearly 100 years ago. Furthermore, increased reactive oxygen species (ROS) and lactate quantities increase metabolic, survival and proliferation signalling, resulting in increased tumourigenesis. In order to maintain redox balance, the cell possesses innate antioxidant defence systems such as superoxide dismutase, catalase and glutathione. Several stimuli including cells deprived of nutrients or failure of antioxidant systems result in oxidative stress and cell death induction. Among the cell death machinery is autophagy, a compensatory mechanism whereby energy is produced from damaged and/or redundant organelles and proteins, which prevents the accumulation of waste products, thereby maintaining homeostasis. Furthermore, autophagy is maintained by several pathways including phosphoinositol 3 kinases, the mitogen-activated protein kinase family, hypoxia-inducible factor, avian myelocytomatosis viral oncogene homolog and protein kinase receptor-like endoplasmic reticulum kinase. The persistent potential of cancer metabolism, redox regulation and the crosstalk with autophagy in scientific investigation pertains to its ability to uncover essential aspects of tumourigenic transformation. This may result in clinical translational possibilities to exploit tumourigenic oxidative status and autophagy to advance our capabilities to diagnose, monitor and treat cancer.
Collapse
Affiliation(s)
- Mokgadi Violet Gwangwa
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Arcadia, 0007 South Africa
| | - Anna Margaretha Joubert
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Arcadia, 0007 South Africa
| | - Michelle Helen Visagie
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Arcadia, 0007 South Africa
| |
Collapse
|
21
|
Oxidative Stress Gene Expression Profile Correlates with Cancer Patient Poor Prognosis: Identification of Crucial Pathways Might Select Novel Therapeutic Approaches. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:2597581. [PMID: 28770020 PMCID: PMC5523271 DOI: 10.1155/2017/2597581] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 05/30/2017] [Indexed: 12/17/2022]
Abstract
The role of altered redox status and high reactive oxygen species (ROS) is still controversial in cancer development and progression. Intracellular levels of ROS are elevated in cancer cells suggesting a role in cancer initiation and progression; on the contrary, ROS elevated levels may induce programmed cell death and have been associated with cancer suppression. Thus, it is crucial to consider the double-face of ROS, for novel therapeutic strategies targeting redox regulatory mechanisms. In this review, in order to derive cancer-type specific oxidative stress genes' profile and their potential prognostic role, we integrated a publicly available oxidative stress gene signature with patient survival data from the Cancer Genome Atlas database. Overall, we found several genes statistically significant associated with poor prognosis in the examined six tumor types. Among them, FoxM1 and thioredoxin reductase1 expression showed the same pattern in four out of six cancers, suggesting their specific critical role in cancer-related oxidative stress adaptation. Our analysis also unveiled an enriched cellular network, highlighting specific pathways, in which many genes are strictly correlated. Finally, we discussed novel findings on the correlation between oxidative stress and cancer stem cells in order to define those pathways to be prioritized in drug development.
Collapse
|
22
|
Mascarenhas S, Mutnuri S, Ganguly A. Deleterious role of trace elements - Silica and lead in the development of chronic kidney disease. CHEMOSPHERE 2017; 177:239-249. [PMID: 28292724 DOI: 10.1016/j.chemosphere.2017.02.155] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 02/27/2017] [Accepted: 02/28/2017] [Indexed: 06/06/2023]
Abstract
Chronic-Kidney-Disease of Unknown-etiology (CKDu) has been reported in developing-countries like Sri-Lanka, India and Central-America without sparing the Indian sub-district (namely Canacona) located in south-Goa. The disease etiology is unlinked to common causes of diabetes and hypertension and assumed to be environmentally induced due to its asymptomatic-nature and occurrence in groundwater relying communities. This study aimed to understand environmental risk-factors underlying CKDu-etiology using Indian sub-district (Canacona) as case-study. Biochemical-analysis of CKDu-affected and non-affected individual's blood and detailed hydro-geochemical analyses of CKDu-affected and non-affected region's groundwater (drinking-water)were conducted. Trace geogenic-element-silica was highly dominant in affected-region's groundwater, thus its nephrotoxic-potential was analysed via in-vitro cytotoxicity-assays on human-kidney-cell-lines. All CKDu-affected-subjects showed increased-levels of serum-urea (52.85 mM),creatinine (941.5 μM),uric-acid (1384.5 μM), normal blood-glucose (4.65 mM), being distinct biomarkers of environmentally-induced CKD-'chronic-tubulo-interstitial-nephritis'. Affected-subjects reported high blood-lead levels (1.48 μM)suggesting direct-nephrotoxicity resulting in impaired blood-clearance and also exhibits indirect-nephrotoxicity by disrupting calcium-homeostasis causing skeletal-disorders and prolonged-consumption of NSAID's (pain-alleviation), indirectly causing renal-damage. Affected-region's groundwater was acidic (pH-5.6), resulting in borderline-lead (9.98 μgL-1) and high-silica (115.5 mgL-1)contamination. Silica's bio-availability (determining its nephrotoxicity) was enhanced at groundwater's acidic-pH and Ca-Mg-deficient-composition (since these cations complex with silica reducing bioavailability). Silica exhibited renal-proximal-tubular-cytotoxicity on long-term exposure comparable with affected-region's groundwater silica-levels, by apoptosis-mediated-cell-death resulting in tubular-atrophy, interstitial-fibrosis and irreversible renal-damage (CKD). Thus this study provides novel-insights into nephrotoxic-potential of trace-geogenic-element-silica in CKDu causation. It highlights direct-indirect nephrotoxicity exhibited by lead at low-levels due to its bio-accumulative-capacity. Silica's nephrotoxic-potential can be considered when deciphering etiology of CKDu-problem in developing-countries (relying on groundwater).
Collapse
Affiliation(s)
- Starlaine Mascarenhas
- Department of Biological Sciences, BITS Pilani, K K Birla Goa Campus, NH 17 B, Zuarinagar, Goa, 403 726, India.
| | - Srikanth Mutnuri
- Department of Biological Sciences, BITS Pilani, K K Birla Goa Campus, NH 17 B, Zuarinagar, Goa, 403 726, India.
| | - Anasuya Ganguly
- Department of Biological Sciences, BITS Pilani, K K Birla Goa Campus, NH 17 B, Zuarinagar, Goa, 403 726, India.
| |
Collapse
|
23
|
Lee HY, Parkinson EI, Granchi C, Paterni I, Panigrahy D, Seth P, Minutolo F, Hergenrother PJ. Reactive Oxygen Species Synergize To Potently and Selectively Induce Cancer Cell Death. ACS Chem Biol 2017; 12:1416-1424. [PMID: 28345875 DOI: 10.1021/acschembio.7b00015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A distinctive feature of cancer cells is their elevated levels of reactive oxygen species (ROS), a trait that can cause cancer cells to be more sensitive to ROS-inducing agents than normal cells. ROS take several forms, each with different reactivity and downstream consequence. Here we show that simultaneous generation of superoxide and hydrogen peroxide within cancer cells results in significant synergy, potently and selectively causing cancer cell death. In these experiments superoxide is generated using the NAD(P)H quinone oxidoreductase 1 (NQO1) substrate deoxynyboquinone (DNQ), and hydrogen peroxide is generated using the lactate dehydrogenase A (LDH-A) inhibitor NHI-Glc-2. This combination reduces tumor burden and prolongs survival in a mouse model of lung cancer. These data suggest that simultaneous induction of superoxide and hydrogen peroxide can be a powerful and selective anticancer strategy.
Collapse
Affiliation(s)
- Hyang Yeon Lee
- Department
of Chemistry, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Elizabeth I. Parkinson
- Department
of Chemistry, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Carlotta Granchi
- Dipartimento
di Farmacia, Università di Pisa, Via Bonanno 33, 56126 Pisa, Italy
| | - Ilaria Paterni
- Dipartimento
di Farmacia, Università di Pisa, Via Bonanno 33, 56126 Pisa, Italy
| | | | | | - Filippo Minutolo
- Dipartimento
di Farmacia, Università di Pisa, Via Bonanno 33, 56126 Pisa, Italy
| | - Paul J. Hergenrother
- Department
of Chemistry, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
24
|
Nachat A, Turoff-Ortmeyer S, Liu C, Mcculloch M. PEITC in End-Stage B-Cell Prolymphocytic Leukemia: Case Report of Possible Sensitization to Salvage R-CHOP. Perm J 2017; 20:74-80. [PMID: 27168399 DOI: 10.7812/tpp/15-153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
INTRODUCTION B-cell prolymphocytic leukemia (B-PLL) is a rare, aggressive leukemia distinct from chronic lymphocytic leukemia, with median survival of only 3 years. B-PLL is resistant to most chemotherapy and newer targeted therapies such as alemtuzumab and thalidomide. Phenylethyl isothiocyanate (PEITC) is a natural compound from horseradish with evidence for therapeutic potential in multiple leukemia types. CASE PRESENTATION Here we present a case report of a 53-year-old man whose chronic lymphocytic leukemia transformed to end-stage B-PLL, disqualifying him for allogenic stem cell transplantation. He was treated with PEITC followed by salvage R-CHOP (Rituximab, Cyclophosphamide, Hydroxydaunorubicin [doxorubicin hydrochloride], Oncovin [vincristine sulfate], Prednisone or Prednisolone) chemotherapy, which led to normalized white blood cell count and disease stabilization that requalified him for allogenic peripheral stem-cell transplant therapy. We conducted a systematic review to analyze and interpret the potential contribution of PEITC to his unexpectedly favorable R-CHOP response. Following sequential 8 weeks of PEITC/pentostatin and 6 cycles of R-CHOP, the patient received allogenic peripheral blood stem cell transplant on an outpatient basis and remains well at the time of this publication, with no evidence of CD20+ small B-cells. DISCUSSION Given the limited data for R-CHOP in B-PLL, this patient's recovery suggests presensitization of B-PLL cells toward R-CHOP, potentially justifying further investigation.
Collapse
Affiliation(s)
- Arian Nachat
- Physician Lead for Integrative Medicine at Walnut Creek Hospital in CA.
| | | | - Chunnan Liu
- Medical Oncologist at Walnut Creek Hospital in CA.
| | - Michael Mcculloch
- Chief of Research for Integrative Medicine at the Pine Street Foundation in San Anselmo and at Walnut Creek Hospital in CA.
| |
Collapse
|
25
|
Tahmasbpour E, Ghanei M, Panahi Y. Two Lung Cancer Development-Related Genes, Forkhead Box M1 ( FOXM1) and Apolipoprotein E ( APOE), are overexpressed in Bronchial of Patients after Long-Term Exposure to Sulfur Mustard. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2017; 16:1487-1494. [PMID: 29552057 PMCID: PMC5843310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Sulfur mustard (SM) is a strong alkylating and mutagenic compound that targets human airway system. We considered the expression of Forkhead box M1 (FOXM1) and apolipoprotein E (APOE) genes, which are responsible for cell proliferation, differentiation, tumorigenesis, and increased risk of lung cancer, in the lung bronchial tissue of patients exposed to SM. After performing pulmonary functional tests (PFTs), 11 human subjects (five controls and six SM-exposed patients) were entered in this study. Total RNA were extracted from all biopsy samples and then cDNA was synthesised for each specimen using RT-PCR. Changes in gene expression were measured using a RT2 Profiler ™PCR Array. PFTs have demonstrated more obstructive and restrictive spirometric patterns among patients compared to the controls. A higher expression was recorded for both examined genes in bronchial of SM-exposed patients. Expression of FOXM1 and APOE genes in bronchial of the patients was significantly (p < 0.001) overexpressed by 14.8316 and 3.9504-folds, respectively. Mustard lungs were associated with increased expression of FOXM1 and APOE genes, which suggests an increased risk of lung cancer among these patients. Since FOXM1 and APOE are considered as oxidative stress responsive genes, we speculate that increased expression of these genes is more likely linked to overproduction of reactive oxygen species (ROS) and oxidative stress (OS) in mustard lungs. Further studies are required at protein level among SM-exposed patients with lung cancer to use these genes as lung cancer biomarkers among these patients.
Collapse
|
26
|
Abstract
Forkhead box (Fox) transcription factors are evolutionarily conserved in organisms ranging from yeast to humans. They regulate diverse biological processes both during development and throughout adult life. Mutations in many Fox genes are associated with human disease and, as such, various animal models have been generated to study the function of these transcription factors in mechanistic detail. In many cases, the absence of even a single Fox transcription factor is lethal. In this Primer, we provide an overview of the Fox family, highlighting several key Fox transcription factor families that are important for mammalian development.
Collapse
Affiliation(s)
- Maria L Golson
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
27
|
Chen H, Wang J, Yang H, Chen D, Li P. Association between FOXM1 and hedgehog signaling pathway in human cervical carcinoma by tissue microarray analysis. Oncol Lett 2016; 12:2664-2673. [PMID: 27698840 DOI: 10.3892/ol.2016.4932] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 02/19/2016] [Indexed: 12/22/2022] Open
Abstract
Forkhead box M1 (FOXM1) and hedgehog (Hh) signaling pathway are implicated in the formation and development of human tumors, including cervical cancer. Previous studies have indicated that FOXM1 may be a downstream target gene of the Hh signaling pathway, but their association in cervical cancer is largely unknown. In the present study, the expression of FOXM1 and Hh signaling molecules was evaluated by immunohistochemical analysis in a tissue microarray that contained 70 cervical cancer tissues and 10 normal cervical tissues. In addition, the association of these molecules with clinicopathological parameters, and the association between FOXM1 and various molecules involved in the Hh signaling pathway was investigated. The results indicated that FOXM1 and Hh signaling molecules were overexpressed in cervical cancer tissues. The protein expression levels of FOXM1, glioma-associated oncogene 1 (GLI1) and smoothened (SMO) correlated with the clinical stage of the tumors, while the protein expression levels of Sonic Hh (SHh), patched 1 (PTCH1) and GLI1 correlated with the pathological grade of the tumors. The expression levels of GLI1 were lower in tissues without lymph node metastasis than in tissues with lymph node metastasis. In addition, FOXM1 expression correlated with GLI1, SHh and PTCH1 expression in cancer tissues. These findings confirmed the participation of FOXM1 and the Hh signaling pathway in cervical cancer. Furthermore, the finding that FOXM1 may be a downstream target gene of the Hh signaling pathway in cervical cancer provides a potential novel diagnostic and therapeutic target for cervical cancer.
Collapse
Affiliation(s)
- Hong Chen
- Department of Gynecology and Obstetrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Jingjing Wang
- Department of Gynecology and Obstetrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Hong Yang
- Department of Gynecology and Obstetrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Dan Chen
- Department of Gynecology and Obstetrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Panpan Li
- Department of Gynecology and Obstetrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
28
|
Kwok CTD, Leung MH, Qin J, Qin Y, Wang J, Lee YL, Yao KM. The Forkhead box transcription factor FOXM1 is required for the maintenance of cell proliferation and protection against oxidative stress in human embryonic stem cells. Stem Cell Res 2016; 16:651-661. [PMID: 27062359 DOI: 10.1016/j.scr.2016.03.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 03/05/2016] [Accepted: 03/17/2016] [Indexed: 12/30/2022] Open
Abstract
Human embryonic stem cells (hESCs) exhibit unique cell cycle structure, self-renewal and pluripotency. The Forkhead box transcription factor M1 (FOXM1) is critically required for the maintenance of pluripotency in mouse embryonic stem cells and mouse embryonal carcinoma cells, but its role in hESCs remains unclear. Here, we show that FOXM1 expression was enriched in undifferentiated hESCs and was regulated in a cell cycle-dependent manner with peak levels detected at the G2/M phase. Expression of FOXM1 did not correlate with OCT4 and NANOG during in vitro differentiation of hESCs. Importantly, knockdown of FOXM1 expression led to aberrant cell cycle distribution with impairment in mitotic progression but showed no profound effect on the undifferentiated state. Interestingly, FOXM1 depletion sensitized hESCs to oxidative stress. Moreover, genome-wide analysis of FOXM1 targets by ChIP-seq identified genes important for M phase including CCNB1 and CDK1, which were subsequently confirmed by ChIP and RNA interference analyses. Further peak set comparison against a differentiating hESC line and a cancer cell line revealed a substantial difference in the genomic binding profile of FOXM1 in hESCs. Taken together, our findings provide the first evidence to support FOXM1 as an important regulator of cell cycle progression and defense against oxidative stress in hESCs.
Collapse
Affiliation(s)
- C T D Kwok
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - M H Leung
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - J Qin
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Y Qin
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - J Wang
- Centre for Genomic Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Y L Lee
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - K-M Yao
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
29
|
Tahmasbpour E, Ghanei M, Qazvini A, Vahedi E, Panahi Y. Gene expression profile of oxidative stress and antioxidant defense in lung tissue of patients exposed to sulfur mustard. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2016; 800-801:12-21. [PMID: 27085470 DOI: 10.1016/j.mrgentox.2016.03.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 02/07/2016] [Accepted: 03/10/2016] [Indexed: 01/13/2023]
Abstract
Sulfur mustard (SM) is a potent alkylating agent that targets several organs, especially lung tissue. Although pathological effects of SM on mustard lung have been widely considered, molecular and cellular mechanisms for these pathologies are poorly understood. We investigated changes in expression of genes related to oxidative stress (OS) and antioxidant defense caused by SM in lung tissue of patients. We performed gene expression profiling of OS and antioxidant defense in lung tissue samples from healthy controls (n=5) and SM-exposed patients (n=6). Changes in gene expression were measured using a 96-well RT(2) Profiler ™PCR Array: Human Oxidative Stress and Antioxidant Defense, which arrayed 84 genes functionally involved in cellular OS response. 47 (55.95%) genes were found to be significantly upregulated in patients with mustard lung compared with controls (p<0.05), whereas 7 (8.33%) genes were significantly downregulated (p<0.05). Among the most upregulated genes were OS responsive-1 (OXSR1), forkhead box M1 (FOXM1), and glutathione peroxidase-2 (GPX2), while metallothionein-3 (MT3) and glutathione reductase (GSR) were the most downregulated genes. Expression of hypoxia-induced genes (CYGB and MB), antioxidants and reactive oxygen species (ROS)-producing genes were significantly altered, suggesting an increased oxidative damage in mustard lungs. Mustard lungs were characterized by hypoxia, massive production of ROS, OS, disruption of epithelial cells, surfactant dysfunction, as well as increased risk of lung cancer and pulmonary fibrosis. Oxidative stress induced by ROS is the major mechanism for direct effect of SM exposure on respiratory system. Antioxidant treatment may improve the main features of mustard lungs.
Collapse
Affiliation(s)
- Eisa Tahmasbpour
- Chemical Injuries Research Center, Baqiatallah University of Medical Sciences, Tehran, Iran
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Baqiatallah University of Medical Sciences, Tehran, Iran
| | - Ali Qazvini
- Chemical Injuries Research Center, Baqiatallah University of Medical Sciences, Tehran, Iran; Department of Pulmonology, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ensieh Vahedi
- Chemical Injuries Research Center, Baqiatallah University of Medical Sciences, Tehran, Iran; Department of Pulmonology, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Yunes Panahi
- Chemical Injuries Research Center, Baqiatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
30
|
Guo CH, Hsia S, Hsiung DY, Chen PC. Supplementation with Selenium yeast on the prooxidant-antioxidant activities and anti-tumor effects in breast tumor xenograft-bearing mice. J Nutr Biochem 2015; 26:1568-79. [PMID: 26344777 DOI: 10.1016/j.jnutbio.2015.07.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 07/26/2015] [Accepted: 07/29/2015] [Indexed: 12/29/2022]
Abstract
Selenium (Se) is essential for antioxidant activity involved in immune function and anti-carcinogenic action, whereas at higher concentrations, Se may have pro-oxidant properties. The present study was aimed at determining the effects of Se supplementation, as Se yeast, on oxidative stress in non-tumor/tumor tissues, as well as regulation of the apoptotic process, and immune responses in mice-bearing breast tumor xenografts. Female BALB/cByJNarl mice were divided into control (CNL and CNL-con), Se-supplemented control (CNL-HS, given as a single oral dose of 912 ng Se daily), breast tumor-bearing (TB and TB-con), TB-LS (228 ng Se), TB-MS (456 ng Se) and TB-HS (912 ng Se) groups. All mice were treated with/without Se for 14 days. A number of variables were further measured. Compared with the TB groups, tumor bearing mice with Se supplement had increased plasma Se concentrations, reduced erythrocyte Se-dependent glutathione peroxidase (GPx) activity and malondialdehyde (MDA) products and inhibited tumor growth. They have also higher Se concentrations in non-tumor and tumor tissues. Significantly elevated concentrations of MDA and reduced GPx activities, as well as increased anti-apoptotic bcl-2 and tumor suppressor p53 concentrations in tumor tissues were observed as Se accumulated in tumor, whereas lower MDA products were found in various non-tumor tissues than did the corresponding values. Further, there were elevated concentrations of Th1-derived cytokines and decreased Th2-type interleukin (IL)-4 in tumor-bearing mice with the treatment of Se. In conclusion, accumulation of Se in tumors may induce oxidative stress and p53-dependent pro-oxidative apoptosis, thus inhibiting the growth of breast tumor.
Collapse
Affiliation(s)
- Chih-Hung Guo
- Institute of Biomedical Nutrition, Hung-Kuang University, Taichung, 433, Taiwan; Department of Medical Research, China Medical University Hospital, Taichung, 404, Taiwan; Department of Health and Nutrition Biotechnology, Asia University, Taichung, 413, Taiwan; Taiwan Nutraceutical Association, Taipei 115, Taiwan.
| | - Simon Hsia
- Institute of Biomedical Nutrition, Hung-Kuang University, Taichung, 433, Taiwan; Taiwan Nutraceutical Association, Taipei 115, Taiwan
| | - Der-Yun Hsiung
- Department of Nursing, Hung-Kuang University, Taichung, 404, Taiwan; School of Nursing, China Medical University, Taichung, 404, Taiwan
| | - Pei-Chung Chen
- Taiwan Nutraceutical Association, Taipei 115, Taiwan; College of Life Sciences, National Chung Hsing University, Taichung, 402, Taiwan; College of Engineering, National Chiao Tung University, Hsinchu, 300, Taiwan.
| |
Collapse
|
31
|
Yuan F, Wang W. MicroRNA-802 suppresses breast cancer proliferation through downregulation of FoxM1. Mol Med Rep 2015; 12:4647-4651. [PMID: 26080894 DOI: 10.3892/mmr.2015.3921] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 11/19/2014] [Indexed: 11/05/2022] Open
Abstract
An increasing number of studies have shown that microRNAs (miRNAs) are critical in tumor cell proliferation, as they modulate key gene transcripts. In the present study, the expression and roles of miRNA (miR)‑802 were analyzed by quantitative polymerase chain reaction in breast cancer cells. The results showed that expression levels of miR‑802 were significantly reduced in breast cancer tissues and cells compared with those of normal tissue and normal breast epithelial cells. In vitro and in vivo experiments demonstrated that miR‑802 overexpression inhibited cell proliferation in MCF‑7 breast cancer cells and tumor growth in nude mice, respectively. Furthermore, mechanistic investigation with western blotting and luciferase reporter assays revealed that miR‑802 overexpression downregulated protein expression levels of Forkhead box protein M1 (FoxM1). Therefore, the results of the present study provided evidence for a previously undetermined miR‑802/FoxM1 molecular network, which was involved in the regulation of breast cancer cell proliferation.
Collapse
Affiliation(s)
- Feng Yuan
- Department of Breast Cancer, Hubei Cancer Hospital, Wuhan, Hubei 430079, P.R. China
| | - Wei Wang
- Department of Breast Cancer, Hubei Cancer Hospital, Wuhan, Hubei 430079, P.R. China
| |
Collapse
|
32
|
Gupta P, Wright SE, Srivastava SK. PEITC treatment suppresses myeloid derived tumor suppressor cells to inhibit breast tumor growth. Oncoimmunology 2015; 4:e981449. [PMID: 25949878 PMCID: PMC4404818 DOI: 10.4161/2162402x.2014.981449] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 10/20/2014] [Accepted: 10/22/2014] [Indexed: 01/26/2023] Open
Abstract
Breast tumors are heterogeneous with a complex etiology. The immune system plays a crucial role in the development of tumors and can facilitate tumor growth pleiotropically. Myeloid derived suppressor cells (MDSCs) generate reactive oxygen species (ROS) and cytokines to suppress T cells, dendritic cells and natural killer (NK) cells. Hence, the inhibition of MDSCs could be an important strategy for anticancer therapeutics. Phenethyl isothiocyanate (PEITC), a bioactive compound present in cruciferous vegetables, is known to have anticancer properties. However, the effects of PEITC administration on the immune system have not been previously reported. In the current study, we evaluated the effects of administering PEITC to immunocompromised NOD-SCID IL2Rγ-/- (SCID/NSG) host mice bearing MDA-MB-231 xenografts on MDSCs in the peripheral blood. Our results reveal that oral administration of 12 μmol PEITC attenuated tumor growth by 76%. This was marked tumor-inhibitory phenotype was associated with a significant reduction in the levels of MDSCs bearing the surface markers CD33, CD34 and CD11b in PEITC treated mice, indicating that overall tumor growth suppression by PEITC correlates with inhibition of MDSCs. To the best of our knowledge, this is the first study showing effects of PEITC on MDSCs.
Collapse
Affiliation(s)
- Parul Gupta
- Department of Biomedical Sciences and Cancer Biology Center; Texas Tech University Health Sciences Center; Amarillo, TX USA
| | - Stephen E. Wright
- Department of Biomedical Sciences and Cancer Biology Center; Texas Tech University Health Sciences Center; Amarillo, TX USA
- Department of Internal Medicine; Texas Tech University Health Sciences Center; Amarillo, TX USA
- Harrington Cancer Center; Amarillo, TX USA
| | - Sanjay K. Srivastava
- Department of Biomedical Sciences and Cancer Biology Center; Texas Tech University Health Sciences Center; Amarillo, TX USA
- Cancer Preventive Material Development Research Center; College of Korean Medicine; Department of Pathology; Kyunghee University; Dongdaemun-ku, Seoul, South Korea
| |
Collapse
|
33
|
Hu CJ, Wang B, Tang B, Chen BJ, Xiao YF, Qin Y, Yong X, Luo G, Zhang JW, Zhang D, Li S, He F, Yang SM. The FOXM1-induced resistance to oxaliplatin is partially mediated by its novel target gene Mcl-1 in gastric cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:290-9. [PMID: 25482013 DOI: 10.1016/j.bbagrm.2014.11.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 11/24/2014] [Accepted: 11/26/2014] [Indexed: 02/07/2023]
Abstract
Myeloid cell leukemia-1 (Mcl-1) is an anti-apoptotic protein that belongs to the Bcl-2 family. The aberrant expression of Mcl-1 is important for sensitivity to chemotherapy drugs in gastric cancer. However, the regulatory mechanism of Mcl-1 in gastric cancer cells remains unclear. In this study, we first found that Forkhead box M1 (FOXM1) and Mcl-1 expression levels were positively correlated in human gastric cancer specimens and that both are associated with poor prognosis of patients treated with oxaliplatin. Second, we demonstrated that the expression level of Mcl-1 was correlated with FOXM1 expression in gastric cancer cells. Third, reporter assays showed that FOXM1 upregulated the promoter activity of the Mcl-1 gene. Electrophoretic mobility shift assays (EMSA) and chromatin immunoprecipitation (ChIP) assays further demonstrated that FOXM1 could bind to a particular site (-635acaaacaa-628) in the promoter region of the Mcl-1 gene. Moreover, CCK-8 assays and analyses of apoptosis revealed that the suppression of the FOXM1/Mcl-1 pathway induced apoptosis and thus increased sensitivity to oxaliplatin in gastric cancer cells, whereas the enhancement of the FOXM1/Mcl-1 pathway inhibited apoptosis and decreased sensitivity to oxaliplatin in gastric cancer cells. Taken together, this study is the first to not only show that Mcl-1 is a novel target gene of FOXM1 but also suggest that targeting FOXM1/Mcl-1 may be a novel strategy to enhance sensitivity to oxaliplatin in gastric cancer.
Collapse
Affiliation(s)
- Chang-Jiang Hu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Bin Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Bo Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Bai-jun Chen
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Yu-Feng Xiao
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Yong Qin
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Xin Yong
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Gang Luo
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Jian-Wei Zhang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Dan Zhang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Song Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Fengtian He
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China.
| | - Shi-Ming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| |
Collapse
|
34
|
Rotblat B, Grunewald TGP, Leprivier G, Melino G, Knight RA. Anti-oxidative stress response genes: bioinformatic analysis of their expression and relevance in multiple cancers. Oncotarget 2014; 4:2577-90. [PMID: 24342878 PMCID: PMC3926850 DOI: 10.18632/oncotarget.1658] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cells mount a transcriptional anti-oxidative stress (AOS) response program to scavenge reactive oxygen species (ROS) that arise from chemical, physical, and metabolic challenges. This protective program has been shown to reduce carcinogenesis triggered by chemical and physical insults. However, it is also hijacked by established cancers to thrive and proliferate within the hostile tumor microenvironment and to gain resistance against chemo- and radiotherapies. Therefore, targeting the AOS response proteins that are exploited by cancer cells is an attractive therapeutic strategy. In order to identify the AOS genes that are suspected to support cancer progression and resistance, we analyzed the expression patterns of 285 genes annotated for being involved in oxidative stress in 994 tumors and 353 normal tissues. Thereby we identified a signature of 116 genes that are highly overexpressed in multiple carcinomas while being only minimally expressed in normal tissues. To establish which of these genes are more likely to functionally drive cancer resistance and progression, we further identified those whose overexpression correlates with negative patient outcome in breast and lung carcinoma. Gene-set enrichment, GO, network, and pathway analyses revealed that members of the thioredoxin and glutathione pathways are prominent components of this oncogenic signature and that activation of these pathways is common feature of many cancer entities. Interestingly, a large fraction of these AOS genes are downstream targets of the transcription factors NRF2, NF-kappaB and FOXM1, and relay on NADPH for their enzymatic activities highlighting promising drug targets. We discuss these findings and propose therapeutic strategies that may be applied to overcome cancer resistance.
Collapse
Affiliation(s)
- Barak Rotblat
- Medical Research Council, Toxicology Unit, Leicester University, Leicester, UK
| | | | | | | | | |
Collapse
|
35
|
Gartel AL. Suppression of the Oncogenic Transcription Factor FOXM1 by Proteasome Inhibitors. SCIENTIFICA 2014; 2014:596528. [PMID: 25093142 PMCID: PMC4095980 DOI: 10.1155/2014/596528] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Accepted: 06/14/2014] [Indexed: 06/03/2023]
Abstract
The oncogenic transcription factor FOXM1 is one of the key regulators of tumorigenesis. We found that FOXM1 upregulates its own transcription and its protein stability depends on its interaction with the chaperone nucleophosmin. We also determined that FOXM1 is negatively regulated by the tumor suppressor p53. We identified the thiazole antibiotics Siomycin A and thiostrepton as inhibitors of transcriptional activity and FOXM1 expression via proteasome inhibition. In addition, we found that all tested proteasome inhibitors target FOXM1. We showed synergy between thiostrepton and bortezomib in different human cancer cell lines and in vivo. We generated isogenic human cancer cell lines of different origin with wild-type p53 or p53 knockdown and we demonstrated that proteasome inhibitors induce p53-independent apoptosis in these cells. Using RNA-interference or proteasome inhibitors to inhibit FOXM1 we found that suppression of FOXM1 sensitized human cancer cells to apoptosis induced by DNA-damaging agents or oxidative stress. We encapsulated thiostrepton into micelle-nanoparticles and after injection we detected accumulation of nanoparticles in tumors and in the livers of treated mice. This treatment led to inhibition of human xenograft tumor growth in nude mice. Our data indicate that targeting FOXM1 increases apoptosis and inhibits tumor growth.
Collapse
Affiliation(s)
- Andrei L. Gartel
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 606012, USA
| |
Collapse
|