1
|
Shah S, D'Souza GGM. Modeling Tumor Microenvironment Complexity In Vitro: Spheroids as Physiologically Relevant Tumor Models and Strategies for Their Analysis. Cells 2025; 14:732. [PMID: 40422235 DOI: 10.3390/cells14100732] [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: 04/18/2025] [Revised: 05/11/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
Drug delivery to solid tumors is challenged by multiple physiological barriers arising from the tumor microenvironment, including dense extracellular matrix, cellular heterogeneity, hypoxic gradients, and elevated interstitial fluid pressure. These features hinder the uniform distribution and accumulation of therapeutics, reducing treatment efficacy. Despite their widespread use, conventional two-dimensional monolayer cultures fail to reproduce these complexities, contributing to the poor translational predictability of many preclinical candidates. Three-dimensional multicellular tumor spheroids have emerged as more representative in vitro models that capture essential features of tumor architecture, stromal interactions, and microenvironmental resistance mechanisms. Spheroids exhibit spatially organized regions of proliferation, quiescence, and hypoxia, and can incorporate non-tumor cells to mimic tumor-stroma crosstalk. Advances in spheroid analysis now enable detailed evaluation of drug penetration, cellular migration, cytotoxic response, and molecular gradients using techniques such as optical and confocal imaging, large-particle flow cytometry, biochemical viability assays, and microfluidic integration. By combining physiological relevance with analytical accessibility, spheroid models support mechanistic studies of drug transport and efficacy under tumor-like conditions. Their adoption into routine preclinical workflows has the potential to improve translational accuracy while reducing reliance on animal models.
Collapse
Affiliation(s)
- Shrey Shah
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA
- Atom Bioworks Inc., Cary, NC 27513, USA
| | - Gerard G M D'Souza
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA
| |
Collapse
|
2
|
Zhang WX, Chen J, Guo Q, Lv QY, Song X, Cui HF. Reversal of doxorubicin-resistance of MCF-7/Adr cells via multiple regulations by glucose oxidase loaded AuNRs@MnO 2@SiO 2 nanocarriers. Colloids Surf B Biointerfaces 2025; 253:114748. [PMID: 40334474 DOI: 10.1016/j.colsurfb.2025.114748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/23/2025] [Accepted: 04/27/2025] [Indexed: 05/09/2025]
Abstract
Targeting to multiple MDR mechanisms is a desired strategy for efficient reversal of multidrug resistance (MDR). Herein, a multi-functional and hierarchical-structured AuNRs@MnO2@SiO2 (AMS) nanocarrier is reported for multiple regulations of MDR. The glucose oxidase (GOx) loaded AMS (AMS/G) showed efficient capabilities of hypoxia-relieving, O2-generation enhanced cancer starvation therapy (CST), and near-infrared (NIR) laser photothermal therapy (PTT) to MCF-7/Adr, a doxorubicin (Dox)-resistant breast cancer cell line. It was revealed that hypoxia inducible factor-1α and heat shock protein 90, can be significantly down-regulated by AMS/G. The Dox resistance and the adenosine triphosphate (ATP)-binding cassette (ABC) transporters: P-glycoprotein (P-gp), multidrug resistance-associated protein 1 (MRP1), and breast cancer resistance protein (BCRP), can be dramatically reversed by the AMS/G+NIR treatment. Specifically, the hypoxia-relieving function can down-regulate all the three ABC transporters. The enhanced CST decreases the expression of MRP1. The PTT diminishes the BCRP and MRP1. Assisted by the multiple and synergistic reversal mechanisms, the Dox co-loaded AMS/G (AMS/D/G) with NIR laser significantly inhibited the cell proliferation, migration, and drug efflux at both normoxia and hypoxia conditions. Cell apoptosis is greatly induced in a caspase-3 dependent manner. Tumor ATP depletion and Dox accumulation were confirmed in vivo. The tumor growth inhibition is greatly and synergistically enhanced, without inducing obvious side effects. Collectively, the nanostructured AMS/D/G can inhibit multiple ABC transporters and provide a promisingly platform for highly efficient reversal of tumor drug resistance.
Collapse
Affiliation(s)
- Wen-Xing Zhang
- School of Life Sciences, Zhengzhou University, Science Avenue 100#, Zhengzhou 450001, China
| | - Junyang Chen
- School of Life Sciences, Zhengzhou University, Science Avenue 100#, Zhengzhou 450001, China
| | - Qian Guo
- School of Life Sciences, Zhengzhou University, Science Avenue 100#, Zhengzhou 450001, China
| | - Qi-Yan Lv
- School of Life Sciences, Zhengzhou University, Science Avenue 100#, Zhengzhou 450001, China
| | - Xiaojie Song
- School of Life Sciences, Zhengzhou University, Science Avenue 100#, Zhengzhou 450001, China.
| | - Hui-Fang Cui
- School of Life Sciences, Zhengzhou University, Science Avenue 100#, Zhengzhou 450001, China.
| |
Collapse
|
3
|
Kanjanasirirat P, Jearawuttanakul K, Seemakhan S, Borwornpinyo S, Wongtrakoongate P, Hongeng S, Charoensutthivarakul S. High-throughput screening of FDA-approved drugs identifies colchicine as a potential therapeutic agent for atypical teratoid/rhabdoid tumors (AT/RTs). RSC Adv 2025; 15:12331-12341. [PMID: 40248220 PMCID: PMC12004362 DOI: 10.1039/d5ra01341k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/03/2025] [Indexed: 04/19/2025] Open
Abstract
Atypical teratoid/rhabdoid tumor (AT/RT) is a rare and aggressive tumor of the primary central nervous system primarily affecting children. It typically originates in the cerebellum and brain stem and is associated with a low survival rate. While standard chemotherapy has been used as a primary treatment for AT/RTs, its success rate is unsatisfactory, and patients often experience severe side effects. Therefore, there is an urgent need to develop new and effective treatment strategies. One promising approach for identifying new therapies is drug repurposing. Although many FDA-approved drugs have been repurposed for various cancers, there have been no reports of such applications for AT/RTs. In this study, a library of 2130 FDA-approved drugs was screened using a high-throughput screening system against 2D traditional cultures and 3D spheroid cultures of AT/RT cell lines (BT-12 and BT-16). From this screening, colchicine, a non-chemotherapeutic agent, was identified as a promising candidate. It exhibited IC50 values of 0.016 and 0.056 μM against 2D BT-12 and 2D BT-16 cells, respectively, and IC50 values of 0.004 and 0.023 μM against 3D BT-12 and BT-16 spheroid cultures. Additionally, the cytotoxic effects of colchicine on human brain endothelial cells and human astrocytes were evaluated, and CC50 > 20 μM was observed, which is over two orders of magnitude higher than its effective concentrations in AT/RT cells, indicating considerably lower toxicity to normal brain cells and brain endothelial cells. In conclusion, colchicine shows significant potential to be repurposed as a treatment for AT/RTs, providing a safer and more effective therapeutic option for this rare and challenging disease.
Collapse
Affiliation(s)
- Phongthon Kanjanasirirat
- School of Bioinnovation and Bio-based Product Intelligence, Faculty of Science, Mahidol University Bangkok 10400 Thailand +66-2-201-5899
- Department of Pathobiology, Faculty of Science, Mahidol University Bangkok 10400 Thailand
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University Bangkok 10400 Thailand
| | - Kedchin Jearawuttanakul
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University Bangkok 10400 Thailand
| | - Sawinee Seemakhan
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University Bangkok 10400 Thailand
| | - Suparerk Borwornpinyo
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University Bangkok 10400 Thailand
- Department of Biotechnology, Faculty of Science, Mahidol University Bangkok 10400 Thailand
| | - Patompon Wongtrakoongate
- Department of Biochemistry, Faculty of Science, Mahidol University Bangkok 10400 Thailand
- Center for Neuroscience, Faculty of Science, Mahidol University Bangkok 10400 Thailand
| | - Suradej Hongeng
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University Bangkok 10400 Thailand
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University Bangkok 10400 Thailand
| | - Sitthivut Charoensutthivarakul
- School of Bioinnovation and Bio-based Product Intelligence, Faculty of Science, Mahidol University Bangkok 10400 Thailand +66-2-201-5899
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University Bangkok 10400 Thailand
- Center for Neuroscience, Faculty of Science, Mahidol University Bangkok 10400 Thailand
| |
Collapse
|
4
|
An X, Sun L, Zheng H, Xiao Y, Sun W, Yu D. Mitochondria-associated non-coding RNAs and their impact on drug resistance. Front Pharmacol 2025; 16:1472804. [PMID: 40078288 PMCID: PMC11897306 DOI: 10.3389/fphar.2025.1472804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 02/07/2025] [Indexed: 03/14/2025] Open
Abstract
Drug resistance is a prevalent challenge in clinical disease treatment, often leading to disease relapse and poor prognosis. Therefore, it is crucial to gain a deeper understanding of the molecular mechanisms underlying drug resistance and to develop targeted strategies for its effective prevention and management. Mitochondria, as vital energy-producing organelles within cells, have been recognized as key regulators of drug sensitivity. Processes such as mitochondrial fission, fusion, mitophagy, changes in membrane potential, reactive oxygen species (ROS) accumulation, and oxidative phosphorylation (OXPHOS) are all linked to drug sensitivity. Non-coding RNAs (ncRNAs) enriched in mitochondria (mtncRNA), whether transcribed from mitochondrial DNA (mtDNA) or from the nucleus and transported to mitochondria, can regulate the transcription and translation of mtDNA, thus influencing mitochondrial function, including mitochondrial substance exchange and energy metabolism. This, in turn, directly or indirectly affects cellular sensitivity to drugs. This review summarizes the types of mtncRNAs associated with drug resistance and the molecular mechanisms regulating drug resistance. Our aim is to provide insights and strategies for overcoming drug resistance by modulating mtncRNAs.
Collapse
Affiliation(s)
- Xingna An
- Department of Core Facility, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Lina Sun
- Department of Hematology-Oncology, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Huan Zheng
- Department of Hematology-Oncology, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Yinghui Xiao
- Department of Core Facility, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Weixia Sun
- Department of Nephrology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Dehai Yu
- Department of Core Facility, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
5
|
Mughis H, Lye P, Imperio GE, Bloise E, Matthews SG. Hypoxia modulates P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) drug transporters in brain endothelial cells of the developing human blood-brain barrier. Heliyon 2024; 10:e30207. [PMID: 38737275 PMCID: PMC11088273 DOI: 10.1016/j.heliyon.2024.e30207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/14/2024] Open
Abstract
P-glycoprotein (P-gp) and Breast Cancer Resistance Protein (BCRP) multidrug resistance (MDR) transporters are localized at the luminal surface of the blood-brain barrier (BBB). They confer fetal brain protection against harmful compounds that may be circulating in the peripheral blood. The fetus develops in low oxygen levels; however, some obstetric pathologies such as pre-eclampsia, placenta accreta/previa may result in even greater fetal hypoxic states. We investigated how hypoxia impacts MDR transporters in human fetal brain endothelial cells (hfBECs) derived from early and mid-stages of pregnancy. Hypoxia decreased BCRP protein and activity in hfBECs derived in early pregnancy. In contrast, in hfBECs derived in mid-pregnancy there was an increase in P-gp and BCRP activity following hypoxia. Results suggest a hypoxia-induced reduction in fetal brain protection in early pregnancy, but a potential increase in transporter-mediated protection at the BBB during mid-gestation. This would modify accumulation of various key physiological and pharmacological substrates of P-gp and BCRP in the developing fetal brain and potentially contribute to the pathogenesis of neurodevelopmental disorders commonly associated with in utero hypoxia.
Collapse
Affiliation(s)
- Hafsah Mughis
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Sinai Health System, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
| | - Phetcharawan Lye
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Sinai Health System, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
| | - Guinever E. Imperio
- Sinai Health System, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
| | - Enrrico Bloise
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Departmento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Stephen G. Matthews
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Sinai Health System, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
- Department of Obstetrics & Gynaecology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Ibrahim AM, Nady S, Shafaa MW, Khalil MM. Radiation and chemotherapy variable response induced by tumor cell hypoxia: impact of radiation dose, anticancer drug, and type of cancer. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2022; 61:263-277. [PMID: 35396948 PMCID: PMC9021068 DOI: 10.1007/s00411-022-00974-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 01/21/2022] [Indexed: 06/14/2023]
Abstract
Hypoxia is a condition in which proliferating tumor cells are deprived of oxygen due to limited blood supply from abnormal tumor microvasculature. This study aimed to investigate the molecular changes that occur in tumor cell hypoxia with special emphasis placed on the efficacy of chemotherapeutic and radiation-related effects. Four commercially available chemotherapeutic agents: cisplatin, cyclophosphamide, doxorubicin, and 5-fluorouracil, were tested for their cytotoxic activity on the cancer cell lines PC3 (prostate), HepG2 (liver), and MCF-7 (breast). Tumor cell lines under hypoxia were treated with both IC50 concentrations of the different chemotherapeutic agents and irradiated with 5 and 10 Gy using a 137Cs gamma source. Hypoxia-inducible factor-1α (HIF-1α) protein levels were examined using an ELISA assay. Hypoxic cells showed a significant change in cell viability to all chemotherapeutic agents in comparison to normoxic controls. HepG2 cells were more resistant to the cytotoxic drug doxorubicin compared to other cancer cell lines. The flow cytometric analysis showed that hypoxic cells have lower levels of total apoptotic cell populations (early and late apoptosis) compared to normoxic cells suggesting decreased hypoxia-induced apoptosis in cancer cells. The highest reduction in HIF-1α level was observed in the MCF-7 cell line (95.5%) in response to the doxorubicin treatment combined with 10 Gy irradiation of cells. Chemoradiotherapy could result in minimal as well as a high reduction of HIF-1α based on cell type, type of chemotherapy, and amount of ionizing radiation. This study highlights future research work to optimize a combined chemoradiotherapeutic regime in individual cancer cell hypoxia.
Collapse
Affiliation(s)
- Ayman M Ibrahim
- Medical Biophysics, Department of Physics, Faculty of Science, Helwan University, Cairo, Egypt
| | - Soad Nady
- Immunology Laboratory, Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| | - Medhat W Shafaa
- Medical Biophysics, Department of Physics, Faculty of Science, Helwan University, Cairo, Egypt
| | - Magdy M Khalil
- Medical Biophysics, Department of Physics, Faculty of Science, Helwan University, Cairo, Egypt.
| |
Collapse
|
7
|
Guo J, Yang Z, Lu Y, Du C, Cao C, Wang B, Yue X, Zhang Z, Xu Y, Qin Z, Huang T, Wang W, Jiang W, Zhang J, Tang J. An antioxidant system through conjugating superoxide dismutase onto metal-organic framework for cardiac repair. Bioact Mater 2021; 10:56-67. [PMID: 34901529 PMCID: PMC8636922 DOI: 10.1016/j.bioactmat.2021.08.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 08/05/2021] [Accepted: 08/16/2021] [Indexed: 12/20/2022] Open
Abstract
Acute myocardial infarction (AMI) remains a dominant origin of morbidity, mortality and disability worldwide. Increases in reactive oxygen species (ROS) are key contributor to excessive cardiac injury after AMI. Here we developed an immobilized enzyme with Superoxide Dismutase (SOD) activity cross-link with Zr-based metal-organic framework (ZrMOF) (SOD-ZrMOF) for mitigate ROS-caused injury. In vitro and in vivo evidence indicates that SOD-ZrMOF exhibits excellent biocompatibility. By efficiently scavenging ROS and suppressing oxidative stress, SOD-ZrMOF can protect the function of mitochondria, reduce cell death and alleviate inflammation. More excitingly, long-term study using an animal model of AMI demonstrated that SOD-ZrMOF can reduce the infarct area, protect cardiac function, promote angiogenesis and inhibit pathological myocardial remodeling. Therefore, SOD-ZrMOF holds great potential as an efficacious and safe nanomaterial treatment for AMI.
Collapse
Affiliation(s)
- Jiacheng Guo
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Zhenzhen Yang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yongzheng Lu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Chunyan Du
- Laboratory Animal Center, School of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Chang Cao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Bo Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Xiaoting Yue
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Zenglei Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Yanyan Xu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Zhen Qin
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Tingting Huang
- Department of Polymer Science, College of Chemistry, Jilin University, Changchun, Jilin, 130012, China
| | - Wei Wang
- Henan Medical Association, Zhengzhou, Henan, 450052, China
| | - Wei Jiang
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Jinying Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Junnan Tang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| |
Collapse
|
8
|
Abdelgawad IY, Sadak KT, Lone DW, Dabour MS, Niedernhofer LJ, Zordoky BN. Molecular mechanisms and cardiovascular implications of cancer therapy-induced senescence. Pharmacol Ther 2021; 221:107751. [PMID: 33275998 PMCID: PMC8084867 DOI: 10.1016/j.pharmthera.2020.107751] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/16/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022]
Abstract
Cancer treatment has been associated with accelerated aging that can lead to early-onset health complications typically experienced by older populations. In particular, cancer survivors have an increased risk of developing premature cardiovascular complications. In the last two decades, cellular senescence has been proposed as an important mechanism of premature cardiovascular diseases. Cancer treatments, specifically anthracyclines and radiation, have been shown to induce senescence in different types of cardiovascular cells. Additionally, clinical studies identified increased systemic markers of senescence in cancer survivors. Preclinical research has demonstrated the potential of several approaches to mitigate cancer therapy-induced senescence. However, strategies to prevent and/or treat therapy-induced cardiovascular senescence have not yet been translated to the clinic. In this review, we will discuss how therapy-induced senescence can contribute to cardiovascular complications. Thereafter, we will summarize the current in vitro, in vivo, and clinical evidence regarding cancer therapy-induced cardiovascular senescence. Then, we will discuss interventional strategies that have the potential to protect against therapy-induced cardiovascular senescence. To conclude, we will highlight challenges and future research directions to mitigate therapy-induced cardiovascular senescence in cancer survivors.
Collapse
Affiliation(s)
- Ibrahim Y Abdelgawad
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA
| | - Karim T Sadak
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN 55455, USA; University of Minnesota Masonic Children's Hospital, Minneapolis, MN 55455, USA; University of Minnesota Masonic Cancer Center, Minneapolis, MN 55455, USA
| | - Diana W Lone
- University of Minnesota Masonic Children's Hospital, Minneapolis, MN 55455, USA
| | - Mohamed S Dabour
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism and Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Beshay N Zordoky
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA.
| |
Collapse
|
9
|
Tailor D, Resendez A, Garcia-Marques FJ, Pandrala M, Going CC, Bermudez A, Kumar V, Rafat M, Nambiar DK, Honkala A, Le QT, Sledge GW, Graves E, Pitteri SJ, Malhotra SV. Y box binding protein 1 inhibition as a targeted therapy for ovarian cancer. Cell Chem Biol 2021; 28:1206-1220.e6. [PMID: 33713600 DOI: 10.1016/j.chembiol.2021.02.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 12/29/2020] [Accepted: 02/17/2021] [Indexed: 12/15/2022]
Abstract
Y box binding protein 1 (YB-1) is a multifunctional protein associated with tumor progression and the emergence of treatment resistance (TR). Here, we report an azopodophyllotoxin small molecule, SU056, that potently inhibits tumor growth and progression via YB-1 inhibition. This YB-1 inhibitor inhibits cell proliferation, resistance to apoptosis in ovarian cancer (OC) cells, and arrests in the G1 phase. Inhibitor treatment leads to enrichment of proteins associated with apoptosis and RNA degradation pathways while downregulating spliceosome pathway. In vivo, SU056 independently restrains OC progression and exerts a synergistic effect with paclitaxel to further reduce disease progression with no observable liver toxicity. Moreover, in vitro mechanistic studies showed delayed disease progression via inhibition of drug efflux and multidrug resistance 1, and significantly lower neurotoxicity as compared with etoposide. These data suggest that YB-1 inhibition may be an effective strategy to reduce OC progression, antagonize TR, and decrease patient mortality.
Collapse
Affiliation(s)
- Dhanir Tailor
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA 94304, USA; Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA 94304, USA; Department of Cell, Development and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Angel Resendez
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Fernando Jose Garcia-Marques
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Mallesh Pandrala
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA 94304, USA; Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA 94304, USA; Department of Cell, Development and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Catherine C Going
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Abel Bermudez
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Vineet Kumar
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Marjan Rafat
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA 94304, USA; Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37212, USA
| | - Dhanya K Nambiar
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Alexander Honkala
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Quynh-Thu Le
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - George W Sledge
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Edward Graves
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA 94304, USA; Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Sharon J Pitteri
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Sanjay V Malhotra
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA 94304, USA; Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA 94304, USA; Department of Cell, Development and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA; Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA.
| |
Collapse
|
10
|
Fontana F, Raimondi M, Marzagalli M, Sommariva M, Gagliano N, Limonta P. Three-Dimensional Cell Cultures as an In Vitro Tool for Prostate Cancer Modeling and Drug Discovery. Int J Mol Sci 2020; 21:E6806. [PMID: 32948069 PMCID: PMC7554845 DOI: 10.3390/ijms21186806] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/13/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023] Open
Abstract
In the last decade, three-dimensional (3D) cell culture technology has gained a lot of interest due to its ability to better recapitulate the in vivo organization and microenvironment of in vitro cultured cancer cells. In particular, 3D tumor models have demonstrated several different characteristics compared with traditional two-dimensional (2D) cultures and have provided an interesting link between the latter and animal experiments. Indeed, 3D cell cultures represent a useful platform for the identification of the biological features of cancer cells as well as for the screening of novel antitumor agents. The present review is aimed at summarizing the most common 3D cell culture methods and applications, with a focus on prostate cancer modeling and drug discovery.
Collapse
MESH Headings
- Adenocarcinoma/drug therapy
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Androgens
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Cell Culture Techniques/instrumentation
- Cell Culture Techniques/methods
- Cell Hypoxia
- Drug Discovery/methods
- Drug Screening Assays, Antitumor/instrumentation
- Drug Screening Assays, Antitumor/methods
- Energy Metabolism
- Epithelial-Mesenchymal Transition
- Extracellular Matrix/metabolism
- Humans
- Inflammation
- Male
- Molecular Targeted Therapy
- Monitoring, Immunologic
- Neoplasm Metastasis
- Neoplasm Proteins/metabolism
- Neoplasms, Hormone-Dependent/drug therapy
- Neoplasms, Hormone-Dependent/metabolism
- Neoplasms, Hormone-Dependent/pathology
- Neoplastic Stem Cells/cytology
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neovascularization, Pathologic/drug therapy
- Oxidation-Reduction
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- Prostatic Neoplasms/therapy
- Spheroids, Cellular/drug effects
- Therapies, Investigational
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Fabrizio Fontana
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy; (M.R.); (M.M.); (P.L.)
| | - Michela Raimondi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy; (M.R.); (M.M.); (P.L.)
| | - Monica Marzagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy; (M.R.); (M.M.); (P.L.)
| | - Michele Sommariva
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, via Mangiagalli 31, 20133 Milan, Italy; (M.S.); (N.G.)
| | - Nicoletta Gagliano
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, via Mangiagalli 31, 20133 Milan, Italy; (M.S.); (N.G.)
| | - Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy; (M.R.); (M.M.); (P.L.)
| |
Collapse
|
11
|
Brüningk SC, Ziegenhein P, Rivens I, Oelfke U, Haar GT. A cellular automaton model for spheroid response to radiation and hyperthermia treatments. Sci Rep 2019; 9:17674. [PMID: 31776398 PMCID: PMC6881451 DOI: 10.1038/s41598-019-54117-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/03/2019] [Indexed: 01/09/2023] Open
Abstract
Thermo-radiosensitisation is a promising approach for treatment of radio-resistant tumours such as those containing hypoxic subregions. Response prediction and treatment planning should account for tumour response heterogeneity, e.g. due to microenvironmental factors, and quantification of the biological effects induced. 3D tumour spheroids provide a physiological in vitro model of tumour response and a systems oncology framework for simulating spheroid response to radiation and hyperthermia is presented. Using a cellular automaton model, 3D oxygen diffusion, delivery of radiation and/or hyperthermia were simulated for many ([Formula: see text]) individual cells forming a spheroid. The iterative oxygen diffusion model was compared to an analytical oxygenation model and simulations were calibrated and validated against experimental data for irradiated (0-10 Gy) and/or heated (0-240 CEM43) HCT116 spheroids. Despite comparable clonogenic survival, spheroid growth differed significantly following radiation or hyperthermia. This dynamic response was described well by the simulation ([Formula: see text] > 0.85). Heat-induced cell death was implemented as a fast, proliferation-independent process, allowing reoxygenation and repopulation, whereas radiation was modelled as proliferation-dependent mitotic catastrophe. This framework stands out both through its experimental validation and its novel ability to predict spheroid response to multimodality treatment. It provides a good description of response where biological dose-weighting based on clonogenic survival alone was insufficient.
Collapse
Affiliation(s)
- Sarah C Brüningk
- Joint Department of Physics at The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK.
| | - Peter Ziegenhein
- Joint Department of Physics at The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Ian Rivens
- Joint Department of Physics at The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Uwe Oelfke
- Joint Department of Physics at The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Gail Ter Haar
- Joint Department of Physics at The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| |
Collapse
|
12
|
Fernando W, Rupasinghe HPV, Hoskin DW. Dietary phytochemicals with anti-oxidant and pro-oxidant activities: A double-edged sword in relation to adjuvant chemotherapy and radiotherapy? Cancer Lett 2019; 452:168-177. [PMID: 30910593 DOI: 10.1016/j.canlet.2019.03.022] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 02/25/2019] [Accepted: 03/08/2019] [Indexed: 12/21/2022]
Abstract
Many advances have been made in the development and introduction of new anti-cancer drugs to the clinic. However, limited attention has been paid to improving the efficacy of currently available treatments through complementary phytochemical interventions that affect cellular reactive oxygen species (ROS) levels, which are important for the etiology of certain cancers and the effectiveness of radiotherapy and some chemotherapy. In this regard, the maintenance of redox homeostasis may be influenced by the intake of anti-oxidant and pro-oxidant compounds from dietary sources. Interestingly, certain dietary phytochemicals exhibit both anti-oxidant and pro-oxidant activities, depending on their concentration and cellular microenvironment. There is evidence that concurrent administration of some dietary phytochemicals enhances the efficacy of certain cancer treatments by increasing intracellular ROS accumulation. Paradoxically, consumption of the same dietary phytochemicals under conditions that result in the scavenging of ROS might also negatively affect the outcome of ROS-dependent cancer treatments. This review discusses the potential impact of consuming dietary phytochemicals with anti-oxidant and/or pro-oxidant activities on the effectiveness of concurrent chemotherapy and/or radiotherapy in cancer patients.
Collapse
Affiliation(s)
- Wasundara Fernando
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - H P Vasantha Rupasinghe
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada; Department of Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, NS, Canada.
| | - David W Hoskin
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada; Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada; Department of Surgery, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
13
|
Nagesetti A, Srinivasan S, McGoron AJ. Polyethylene glycol modified ORMOSIL theranostic nanoparticles for triggered doxorubicin release and deep drug delivery into ovarian cancer spheroids. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2017; 174:209-216. [PMID: 28800509 DOI: 10.1016/j.jphotobiol.2017.07.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 06/13/2017] [Accepted: 07/21/2017] [Indexed: 12/18/2022]
Abstract
A novel pegylated multifunctional probe of Ormosil nanoparticles (PEGCDSIR820) loaded with Near Infrared dye (NIR; IR820) and a chemotherapeutic drug, Doxorubicin (DOX) was developed for cancer theranostic applications. PEGCDSIR820 nanoparticles had an average diameter of 58.2±3.1nm, zeta potential of -6.9±0.1mV in cell culture media and stability against aggregation in physiological buffers. The encapsulation efficiency of DOX was 65.0±3.0%, and that of IR820 was 76.0±2.1%. PEGCDSIR820 showed no cytotoxicity in ovarian cancer cells (Skov-3). The cytotoxicity markedly increased when Skov-3 cells incubated with PEGCDSIR820 particles were exposed to 808nm laser due to the combination of adjuvant hyperthermia (43°C) and enhanced DOX release. Exposure to laser enhanced the release of DOX, 45% of DOX release was observed in 3h compared to 23% without laser exposure. Confocal imaging in Skov-3 cells showed that the combination of hyperthermia due to NIR exposure and release of DOX caused cell necrosis. Furthermore, in spheroids exposed to NIR laser penetration of DOX was deeper compared to the absence of laser exposure. Skov-3 spheroids incubated with pegylated nanoparticles for 24h and exposed to laser showed 94% reduction in cell viability. Encapsulation of IR820 in PEGCDSIR820 increased the in-vivo elimination half-life to 41.0±7.2h from 30.5±0.5h of free IR820.
Collapse
Affiliation(s)
- Abhignyan Nagesetti
- Biomedical Engineering Department, 10555 West Flagler Street, EC 2614, Florida International University, Miami, FL 33174, USA
| | - Supriya Srinivasan
- Biomedical Engineering Department, 10555 West Flagler Street, EC 2614, Florida International University, Miami, FL 33174, USA
| | - Anthony J McGoron
- Biomedical Engineering Department, 10555 West Flagler Street, EC 2614, Florida International University, Miami, FL 33174, USA.
| |
Collapse
|
14
|
3D tumor spheroids: an overview on the tools and techniques used for their analysis. Biotechnol Adv 2016; 34:1427-1441. [PMID: 27845258 DOI: 10.1016/j.biotechadv.2016.11.002] [Citation(s) in RCA: 564] [Impact Index Per Article: 62.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Revised: 11/03/2016] [Accepted: 11/10/2016] [Indexed: 12/14/2022]
Abstract
In comparison with 2D cell culture models, 3D spheroids are able to accurately mimic some features of solid tumors, such as their spatial architecture, physiological responses, secretion of soluble mediators, gene expression patterns and drug resistance mechanisms. These unique characteristics highlight the potential of 3D cellular aggregates to be used as in vitro models for screening new anticancer therapeutics, both at a small and large scale. Nevertheless, few reports have focused on describing the tools and techniques currently available to extract significant biological data from these models. Such information will be fundamental to drug and therapeutic discovery process using 3D cell culture models. The present review provides an overview of the techniques that can be employed to characterize and evaluate the efficacy of anticancer therapeutics in 3D tumor spheroids.
Collapse
|
15
|
The Use of a Liposomal Formulation Incorporating an Antimicrobial Peptide from Tilapia as a New Adjuvant to Epirubicin in Human Squamous Cell Carcinoma and Pluripotent Testicular Embryonic Carcinoma Cells. Int J Mol Sci 2015; 16:22711-34. [PMID: 26393585 PMCID: PMC4613332 DOI: 10.3390/ijms160922711] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Revised: 09/09/2015] [Accepted: 09/11/2015] [Indexed: 02/06/2023] Open
Abstract
This study aims to explore the effects and mechanisms of hepcidin, a potential antimicrobial peptide from Tilapia, and epirubicin (Epi), an antineoplastic agent, on the generation of reactive oxygen species (ROS) and link the ROS levels to the reversal mechanisms of multidrug resistance (MDR) by epirubicin and hepcidin in human squamous cell carcinoma SCC15 and human embryonal carcinoma NT2D1 cells. The cells, pretreated with hepcidin, epirubicin, or a combination of these compounds in PEGylated liposomes, were used to validate the molecular mechanisms involved in inhibiting efflux transporters and inducing apoptosis as evaluated by cytotoxicity, intracellular accumulation, mRNA levels, cell cycle distribution, and caspase activity of this combination. We found that hepcidin significantly enhanced the cytotoxicity of epirubicin in liposomes. The co-incubation of epirubicin with hepcidin in liposomes intensified the ROS production, including hydrogen peroxide and superoxide free radicals. Hepcidin significantly increased epirubicin intracellular uptake into NT2D1 and SCC15 cells, as supported by the diminished mRNA expressions of MDR1, MDR-associated protein (MRP) 1, and MRP2. Hepcidin and/or epirubicin in liposomes triggered apoptosis, as verified by the reduced mitochondrial membrane potential, increased sub-G1 phase of cell cycle, incremental populations of apoptosis using annexin V/PI assay, and chromatin condensation. As far as we know, this is the first example showing that PEGylated liposomal TH1-5 and epirubicin gives rise to cell death in human squamous carcinoma and testicular embryonic carcinoma cells through the reduced epirubicin efflux via ROS-mediated suppression of P-gp and MRPs and concomitant initiation of mitochondrial apoptosis pathway. Hence, hepcidin in PEGylated liposomes may function as an adjuvant to anticancer drugs, thus demonstrating a novel strategy for reversing MDR.
Collapse
|
16
|
Three-dimensional magnetic cell array for evaluation of anti-proliferative effects of chemo-thermo treatment on cancer spheroids. BIOTECHNOL BIOPROC E 2015. [DOI: 10.1007/s12257-014-0724-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
17
|
Wang J, Liu L, Cen J, Ji B. BME, a novel compound of anthraquinone, down regulated P-glycoprotein expression in doxorubicin-resistant human myelogenous leukemia (K562/DOX) cells via generation of reactive oxygen species. Chem Biol Interact 2015; 239:139-45. [PMID: 26169035 DOI: 10.1016/j.cbi.2015.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 06/11/2015] [Accepted: 07/06/2015] [Indexed: 12/09/2022]
Abstract
P-glycoprotein (P-gp)-mediated multidrug resistance (MDR) in tumor cells is still a main obstacle for the chemotherapeutic treatment of cancers. Thus, development of effective MDR reversing agents is an important approach in the clinic. The present study revealed that BME, a novel compound of anthraquinone, elevated intracellular accumulation of the P-gp substrates and reduced concentration resulting in 50% inhibition of cell growth (IC50) values for doxorubicin (DOX) in doxorubicin-resistant human myelogenous leukemia (K562/DOX) cells. Further more, BME was also reported to down regulated P-gp expression accompanying with generation of nontoxic low level of intracellular reactive oxygen species (iROS) and activation of extracellular signal-regulated kinase (ERK)1/2 as well as c-JUN N-terminal kinase (JNK). However, treatment with N-acetyl-cysteine (NAC), U0216 and SP600125 almost abolished actions of the BME mentioned above. These results indicated that the effect of the BME on the P-gp may be involved in generation of nontoxic low level of iROS and activation of ERK1/2 or JNK, which suggested valuable clues to screen and develop P-gp reversing agents.
Collapse
Affiliation(s)
- Jianhong Wang
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475001, China
| | - Lu Liu
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475001, China
| | - Juan Cen
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475001, China
| | - Biansheng Ji
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475001, China.
| |
Collapse
|
18
|
Suppressed expression of mitogen-activated protein kinases in hyperthermia induced defective neural tube. Neurosci Lett 2015; 594:6-11. [PMID: 25818329 DOI: 10.1016/j.neulet.2015.03.046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 03/05/2015] [Accepted: 03/23/2015] [Indexed: 11/23/2022]
Abstract
Neural tube defects (NTDs) are common congenital malformations. Mitogen-activated protein kinases (MAPKs) pathway is involved in many physiological processes. HMGB1 has been showed closely associated with neurulation and NTDs induced by hyperthermia and could activate MAPKs pathway. Since hyperthermia caused increased activation of MAPKs in many systems, the present study aims to investigate whether HMGB1 contributes to hyperthermia induced NTDs through MAPKs pathway. The mRNA levels of MAPKs and HMGB1 between embryonic day 8.5 and 10 (E8.5-10) in hyperthermia induced defective neural tube were detected by real-time quantitative polymerase chain reaction (qPCR). By immunofluorescence and western blotting, the expressions of HMGB1 and phosphorylated MAPKs (ERK1/2, JNK and p38) in neural tubes after hyperthermia were studied. The mRNA levels of MAPKs and HMGB1, as well as the expressions of HMGB1 along with phosphorylated JNK, p38 and ERK, were downregulated in NTDs groups induced by hyperthermia compared with control. The findings suggested that HMGB1 may contribute to hyperthermia induced NTDs formation through decreased cell proliferation due to inhibited phosphorylated ERK1/2 MAPK.
Collapse
|
19
|
Cerezo D, Cánovas M, García-Peñarrubia P, Martín-Orozco E. Collateral sensitivity to cold stress and differential BCL-2 family expression in new daunomycin-resistant lymphoblastoid cell lines. Exp Cell Res 2014; 331:11-20. [PMID: 25498972 DOI: 10.1016/j.yexcr.2014.11.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 11/12/2014] [Accepted: 11/24/2014] [Indexed: 01/02/2023]
Abstract
The acquisition of a multidrug-resistant (MDR) phenotype by tumor cells is one of the main causes of chemotherapy failure in cancer, and, usually, is due to the increased expression of P-glycoprotein (MDR-1, P-gp, ABCB1), a pump that expels chemotherapeutics from the cell and/or regulates apoptosis. Thus, it is fundamental to find drugs or stress stimuli with a capacity to induce apoptosis in such cells and to identify the mechanisms involved. We address this matter in human cells and establish new daunomycin (DNM)-resistant cell lines (IM-9R) by exposing the parental lymphoblastic cells (IM-9) to increasing doses of the anti-neoplastic drug, daunomycin. The resistance level of IM-9R cell lines, MDR-1 expression and functionality, collateral sensitivity and Bcl-2 and caspases protein expression are analyzed. As a result, we show for the first time that, unlike the parental cells, human lymphoblastic resistant cells exhibit collateral sensitivity to cold stress, confirming that this phenomenon is not exclusive to murine leukemic cells, but a broader one associated with the acquisition of drug resistance. Furthermore, the new resistant cell lines undergo a significant increase in active caspase-3 and -9 levels and drastic changes in Bcl-2 family protein expression during the process of MDR phenotype acquisition.
Collapse
Affiliation(s)
- David Cerezo
- Department of Biochemistry and Molecular Biology B and Immunology, University of Murcia, Murcia, Spain
| | - Manuel Cánovas
- Department of Biochemistry and Molecular Biology B and Immunology, University of Murcia, Murcia, Spain
| | - Pilar García-Peñarrubia
- Department of Biochemistry and Molecular Biology B and Immunology, University of Murcia, Murcia, Spain
| | - Elena Martín-Orozco
- Department of Biochemistry and Molecular Biology B and Immunology, University of Murcia, Murcia, Spain.
| |
Collapse
|
20
|
Budker VG, Monahan SD, Subbotin VM. Loco-regional cancer drug therapy: present approaches and rapidly reversible hydrophobization (RRH) of therapeutic agents as the future direction. Drug Discov Today 2014; 19:1855-70. [PMID: 25173702 DOI: 10.1016/j.drudis.2014.08.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 07/10/2014] [Accepted: 08/21/2014] [Indexed: 01/01/2023]
Abstract
Insufficient drug uptake by solid tumors remains the major problem for systemic chemotherapy. Many studies have demonstrated anticancer drug effects to be dose-dependent, although dose-escalation studies have resulted in limited survival benefit with increased systemic toxicities. One solution to this has been the idea of loco-regional drug treatments, which offer dramatically higher drug concentrations in tumor tissues while minimizing systemic toxicity. Although loco-regional delivery has been most prominent in cancers of the liver, soft tissues and serosal peritoneal malignancies, survival benefits are very far from desirable. This review discusses the evolution of loco-regional treatments, the present approaches and offers rapidly reversible hydrophobization of drugs as the new future direction.
Collapse
|
21
|
LEE SANGHAK, NAM JAEKOOK, PARK JONGKOOK, LEE JOOHO, MIN DOSIK, KUH HYOJEONG. Differential protein expression and novel biomarkers related to 5-FU resistance in a 3D colorectal adenocarcinoma model. Oncol Rep 2014; 32:1427-34. [DOI: 10.3892/or.2014.3337] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 06/04/2014] [Indexed: 11/05/2022] Open
|
22
|
Callaghan R, Luk F, Bebawy M. Inhibition of the multidrug resistance P-glycoprotein: time for a change of strategy? Drug Metab Dispos 2014; 42:623-31. [PMID: 24492893 PMCID: PMC3965902 DOI: 10.1124/dmd.113.056176] [Citation(s) in RCA: 310] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 01/31/2014] [Indexed: 01/30/2023] Open
Abstract
P-glycoprotein (P-gp) is a key player in the multidrug-resistant phenotype in cancer. The protein confers resistance by mediating the ATP-dependent efflux of an astonishing array of anticancer drugs. Its broad specificity has been the subject of numerous attempts to inhibit the protein and restore the efficacy of anticancer drugs. The general strategy has been to develop compounds that either compete with anticancer drugs for transport or act as direct inhibitors of P-gp. Despite considerable in vitro success, there are no compounds currently available to "block" P-gp-mediated resistance in the clinic. The failure may be attributed to toxicity, adverse drug interaction, and numerous pharmacokinetic issues. This review provides a description of several alternative approaches to overcome the activity of P-gp in drug-resistant cells. These include 1) drugs that specifically target resistant cells, 2) novel nanotechnologies to provide high-dose, targeted delivery of anticancer drugs, 3) compounds that interfere with nongenomic transfer of resistance, and 4) approaches to reduce the expression of P-gp within tumors. Such approaches have been developed through the pursuit of greater understanding of resistance mediators such as P-gp, and they show considerable potential for further application.
Collapse
Affiliation(s)
- Richard Callaghan
- Division of Biomedical Science & Biochemistry, Research School of Biology, College of Medicine, Biology & Environment, The Australian National University, Canberra, New South Wales, Australia (R.C.); and School of Pharmacy, Graduate School of Health, The University of Technology, Sydney, New South Wales, Australia (F.L., M.B.)
| | | | | |
Collapse
|
23
|
Lei T, Fernandez-Fernandez A, Manchanda R, Huang YC, McGoron AJ. Near-infrared dye loaded polymeric nanoparticles for cancer imaging and therapy and cellular response after laser-induced heating. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2014; 5:313-322. [PMID: 24778954 PMCID: PMC3999826 DOI: 10.3762/bjnano.5.35] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 02/21/2014] [Indexed: 05/31/2023]
Abstract
BACKGROUND In the past decade, researchers have focused on developing new biomaterials for cancer therapy that combine imaging and therapeutic agents. In our study, we use a new biocompatible and biodegradable polymer, termed poly(glycerol malate co-dodecanedioate) (PGMD), for the synthesis of nanoparticles (NPs) and loading of near-infrared (NIR) dyes. IR820 was chosen for the purpose of imaging and hyperthermia (HT). HT is currently used in clinical trials for cancer therapy in combination with radiotherapy and chemotherapy. One of the potential problems of HT is that it can up-regulate hypoxia-inducible factor-1 (HIF-1) expression and enhance vascular endothelial growth factor (VEGF) secretion. RESULTS We explored cellular response after rapid, short-term and low thermal dose laser-IR820-PGMD NPs (laser/NPs) induced-heating, and compared it to slow, long-term and high thermal dose heating by a cell incubator. The expression levels of the reactive oxygen species (ROS), HIF-1 and VEGF following the two different modes of heating. The cytotoxicity of NPs after laser/NP HT resulted in higher cell killing compared to incubator HT. The ROS level was highly elevated under incubator HT, but remained at the baseline level under the laser/NP HT. Our results show that elevated ROS expression inside the cells could result in the promotion of HIF-1 expression after incubator induced-HT. The VEGF secretion was also significantly enhanced compared to laser/NP HT, possibly due to the promotion of HIF-1. In vitro cell imaging and in vivo healthy mice imaging showed that IR820-PGMD NPs can be used for optical imaging. CONCLUSION IR820-PGMD NPs were developed and used for both imaging and therapy purposes. Rapid and short-term laser/NP HT, with a low thermal dose, does not up-regulate HIF-1 and VEGF expression, whereas slow and long term incubator HT, with a high thermal dose, enhances the expression of both transcription factors.
Collapse
Affiliation(s)
- Tingjun Lei
- Biomedical Engineering Department, Florida International University, 10555 West Flagler Street, Miami, FL 33174, USA
- Cirle, 1951 NW 7th Ave, Suite 13016, Miami, FL, 33136, USA
| | - Alicia Fernandez-Fernandez
- Biomedical Engineering Department, Florida International University, 10555 West Flagler Street, Miami, FL 33174, USA
- Physical Therapy Department, Nova Southeastern University, 3200 S. University Dr., Fort Lauderdale, FL 33328, USA
| | - Romila Manchanda
- Biomedical Engineering Department, Florida International University, 10555 West Flagler Street, Miami, FL 33174, USA
- Department of Basic and Applied Sciences, Galgotias University, UP, 201308, India
| | - Yen-Chih Huang
- Biomedical Engineering Department, Florida International University, 10555 West Flagler Street, Miami, FL 33174, USA
| | - Anthony J McGoron
- Biomedical Engineering Department, Florida International University, 10555 West Flagler Street, Miami, FL 33174, USA
| |
Collapse
|
24
|
Lee SM, Kim HJ, Kim SY, Kwon MK, Kim S, Cho A, Yun M, Shin JS, Yoo KH. Drug-loaded gold plasmonic nanoparticles for treatment of multidrug resistance in cancer. Biomaterials 2013; 35:2272-82. [PMID: 24342728 DOI: 10.1016/j.biomaterials.2013.11.068] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 11/21/2013] [Indexed: 11/16/2022]
Abstract
To investigate the possibility of treating multidrug-resistant tumors with targeted chemo-photothermal treatment, we conducted in vitro and in vivo studies using a doxorubicin (DOX)-resistant DLD-1 cell line (DLD-1/DOX) and nude mice with human xenograft tumors, respectively. The chemo-photothermal treatment consisted of DOX-loaded-poly(lactic-co-glycolic acid)-Au half-shell nanoparticles with targeting moieties of anti-death receptor-4 monoclonal antibody conjugated to the Au surface. The cells or xenografted tumors treated with nanoparticles were exposed to near infrared light for 10 min, which caused an increase in temperature to 45 °C. Chemo-photothermal treatment resulted in a large reduction in the rate of tumor xenograft growth on DLD-1/DOX tumor-bearing mice with a much smaller dose of DOX than conventional DOX chemotherapy. These results demonstrate that targeted chemo-photothermal treatment can provide high therapeutic efficacy and low toxicity in the treatment of multidrug-resistant tumors.
Collapse
Affiliation(s)
- Sun-Mi Lee
- Nanomedical Graduate Program, Yonsei University, Seoul 120-749, South Korea
| | - Hyung Joon Kim
- Nanomedical Graduate Program, Yonsei University, Seoul 120-749, South Korea
| | - Sook Young Kim
- Department of Microbiology, Severance Biomedical Science Institute, Institute for Immunology and Immunological Diseases, College of Medicine, Yonsei University, Seoul 120-752, South Korea
| | - Min-Kyung Kwon
- Department of Microbiology, Severance Biomedical Science Institute, Institute for Immunology and Immunological Diseases, College of Medicine, Yonsei University, Seoul 120-752, South Korea
| | - Sol Kim
- Department of Microbiology, Severance Biomedical Science Institute, Institute for Immunology and Immunological Diseases, College of Medicine, Yonsei University, Seoul 120-752, South Korea
| | - Arthur Cho
- Department of Nuclear Medicine, College of Medicine, Yonsei University, Seoul 120-752, South Korea
| | - Mijin Yun
- Department of Nuclear Medicine, College of Medicine, Yonsei University, Seoul 120-752, South Korea
| | - Jeon-Soo Shin
- Nanomedical Graduate Program, Yonsei University, Seoul 120-749, South Korea; Department of Microbiology, Severance Biomedical Science Institute, Institute for Immunology and Immunological Diseases, College of Medicine, Yonsei University, Seoul 120-752, South Korea.
| | - Kyung-Hwa Yoo
- Nanomedical Graduate Program, Yonsei University, Seoul 120-749, South Korea; Department of Physics, Yonsei University, Seoul 120-749, South Korea.
| |
Collapse
|
25
|
Li N, Bo CW, Zou CP, Ma WH, Zheng Z, An YH. Efficacy of continuous hyperthermic peritoneal perfusion chemotherapy for malignant seroperitoneum. Shijie Huaren Xiaohua Zazhi 2013; 21:3757-3761. [DOI: 10.11569/wcjd.v21.i33.3757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To compare the curative effect and adverse effects of continuous hyperthermic peritoneal perfusion chemotherapy versus simple intraperitoneal chemotherapy in the management of malignant seroperitoneum secondary to gastrointestinal tumors.
METHODS: Eighty-four inpatients with malignant seroperitoneum secondary to gastrointestinal tumors treated at our hospital between October 2010 and February 2013 were enrolled in the study. They were randomly and equally divided into either a study group to undergo continuous hyperthermic peritoneal perfusion chemotherapy or a control group to receive intraperitoneal chemotherapy. The differences in curative effect, quality of life and adverse effects were compared between the two groups.
RESULTS: The rate of ascites control was significantly higher in the study group than in the control group (88.33% vs 59.52%, P < 0.05). The rate of improvement of quality of life was also higher in the study group (88.1% vs 53.76%, P < 0.05). Adverse effects included mild nausea and vomiting, temporary abdominal pain, CTCAE grade Ⅰ-Ⅱ bone marrow depression (BMD), and no grade Ⅲ-Ⅳ BMD was identified. No significant differences in adverse effect were observed between the two groups.
CONCLUSION: Continuous hyperthermic peritoneal perfusion chemotherapy is superior to simple intraperitoneal chemotherapy in the management of malignant seroperitoneum in terms of ascite control, improvement of quality of life, and adverse effects.
Collapse
|
26
|
The role of mitochondria-derived reactive oxygen species in hyperthermia-induced platelet apoptosis. PLoS One 2013; 8:e75044. [PMID: 24023970 PMCID: PMC3762754 DOI: 10.1371/journal.pone.0075044] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 08/08/2013] [Indexed: 12/17/2022] Open
Abstract
A combination of hyperthermia with radiotherapy and chemotherapy for various solid tumors has been practiced clinically. However, hyperthermic therapy has side effects, such as thrombocytopenia. Up to now, the pathogenesis of hyperthermia-induced thrombocytopenia remains unclear. Previous studies have shown that hyperthermia induces platelet apoptosis. However, the signaling pathways and molecular mechanisms involved in hyperthermia-induced platelet apoptosis have not been determined. Here we show that hyperthermia induced intracellular reactive oxygen species (ROS) production and mitochondrial ROS generation in a time-dependent manner in platelets. The mitochondria-targeted ROS scavenger Mito-TEMPO blocked intracellular ROS and mitochondrial ROS generation. By contrast, inhibitors of reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, nitric oxide synthase, cyclooxygenase and lipoxygenase did not. Furthermore, Mito-TEMPO inhibited hyperthermia-induced malonyldialdehyde production and cardiolipin peroxidation. We also showed that hyperthermia-triggered platelet apoptosis was inhibited by Mito-TEMPO. Furthermore, Mito-TEMPO ameliorated hyperthermia-impaired platelet aggregation and adhesion function. Lastly, hyperthermia decreased platelet manganese superoxide dismutase (MnSOD) protein levels and enzyme activity. These data indicate that mitochondrial ROS play a pivotal role in hyperthermia-induced platelet apoptosis, and decreased of MnSOD activity might, at least partially account for the enhanced ROS levels in hyperthermia-treated platelets. Therefore, determining the role of mitochondrial ROS as contributory factors in platelet apoptosis, is critical in providing a rational design of novel drugs aimed at targeting mitochondrial ROS. Such therapeutic approaches would have potential clinical utility in platelet-associated disorders involving oxidative damage.
Collapse
|
27
|
Franke K, Kettering M, Lange K, Kaiser WA, Hilger I. The exposure of cancer cells to hyperthermia, iron oxide nanoparticles, and mitomycin C influences membrane multidrug resistance protein expression levels. Int J Nanomedicine 2013; 8:351-63. [PMID: 23378758 PMCID: PMC3554229 DOI: 10.2147/ijn.s37465] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Purpose The presence of multidrug resistance-associated protein (MRP) in cancer cells is known to be responsible for many therapeutic failures in current oncological treatments. Here, we show that the combination of different effectors like hyperthermia, iron oxide nanoparticles, and chemotherapeutics influences expression of MRP 1 and 3 in an adenocarcinoma cell line. Methods BT-474 cells were treated with magnetic nanoparticles (MNP; 1.5 to 150 μg Fe/cm2) or mitomycin C (up to 1.5 μg/cm2, 24 hours) in the presence or absence of hyperthermia (43°C, 15 to 120 minutes). Moreover, cells were also sequentially exposed to these effectors (MNP, hyperthermia, and mitomycin C). After cell harvesting, mRNA was extracted and analyzed via reverse transcription polymerase chain reaction. Additionally, membrane protein was isolated and analyzed via sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and immunoblotting. Results When cells were exposed to the effectors alone or to combinations thereof, no effects on MRP 1 and 3 mRNA expression were observed. In contrast, membrane protein expression was influenced in a selective manner. The effects on MRP 3 expression were less pronounced compared with MRP 1. Treatment with mitomycin C decreased MRP expression at high concentrations and hyperthermia intensified these effects. In contrast, the presence of MNP only increased MRP 1 and 3 expression, and hyperthermia reversed these effects. When combining hyperthermia, magnetic nanoparticles, and mitomycin C, no further suppression of MRP expression was observed in comparison with the respective dual treatment modalities. Discussion The different MRP 1 and 3 expression levels are not associated with de novo mRNA expression, but rather with an altered translocation of MRP 1 and 3 to the cell membrane as a result of reactive oxygen species production, and with shifting of intracellular MRP storage pools, changes in membrane fluidity, etc, at the protein level. Our results could be used to develop new treatment strategies by repressing mechanisms that actively export drugs from the target cell, thereby improving the therapeutic outcome in oncology.
Collapse
Affiliation(s)
- Karolin Franke
- Institute of Diagnostic and Interventional Radiology, Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology I, University Hospital Jena, Friedrich-Schiller Universität Jena, Jena, Germany
| | | | | | | | | |
Collapse
|
28
|
A potential daidzein derivative enhances cytotoxicity of epirubicin on human colon adenocarcinoma Caco-2 cells. Int J Mol Sci 2012; 14:158-76. [PMID: 23344026 PMCID: PMC3565256 DOI: 10.3390/ijms14010158] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 12/12/2012] [Accepted: 12/17/2012] [Indexed: 12/24/2022] Open
Abstract
In this study, we evaluated the effects of 8-hydroxydaidzein (8HD), an isoflavone isolated from fermented soy germ koji, and epirubicin (Epi), an antineoplastic agent, on the production of reactive oxygen species (ROS). We subsequently correlated the ROS levels to the anticancer mechanisms of Epi and 8HD in human colon adenocarcinoma Caco-2 cells. 8HD enhanced cytotoxicity of Epi and generated a synergistic effect. Epi and/or 8HD treatments increased the hydrogen peroxide and superoxide levels. Combined treatment markedly decreased mRNA expression levels of multidrug resistance protein 1 (MDR1), MDR-associated protein (MRP) 1, and MRP2. 8HD significantly intensified Epi intracellular accumulation in Caco-2 cells. 8HD and/or Epi-induced apoptosis, as indicated by the reduced mitochondrial membrane potential and increased sub-G1 phase in cell cycle. Moreover, 8HD and Epi significantly enhanced the mRNA expressions of Bax, p53, caspases-3, -8, and -9. To our best knowledge, this study verifies for the first time that 8HD effectively circumvents MDR in Caco-2 cells through the ROS-dependent inhibition of efflux transporters and p53-mediated activation of both death receptor and mitochondrial pathways of apoptosis. Our findings of 8HD shed light on the future search for potential biotransformed isoflavones to intensify the cytotoxicity of anticancer drugs through simultaneous reversal of pump and nonpump resistance.
Collapse
|
29
|
Thrall DE, Maccarini P, Stauffer P, Macfall J, Hauck M, Snyder S, Case B, Linder K, Lan L, McCall L, Dewhirst MW. Thermal dose fractionation affects tumour physiological response. Int J Hyperthermia 2012; 28:431-40. [PMID: 22804741 DOI: 10.3109/02656736.2012.689087] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PURPOSE It is unknown whether a thermal dose should be administered using a few large fractions with higher temperatures or a larger number of fractions with lower temperatures. To evaluate this we assessed the effect of administering the same total thermal dose, approximately 30 CEM43T(90), in one versus three to four fractions per week, over 5 weeks. MATERIALS AND METHODS Canine sarcomas were randomised to receive one of the hyperthermia fractionation schemes along with fractionated radiotherapy. Tumour response was based on changes in tumour volume, oxygenation, water diffusion quantified using MRI, and a panel of histological and immunohistochemical end points. RESULTS There was a greater reduction in tumour volume and water diffusion at the end of therapy in tumours receiving one hyperthermia fraction per week. There was a weak but significant association between improved tumour oxygenation 24 h after the first hyperthermia treatment and extent of volume reduction at the end of therapy. Finally, the direction of change of HIF-1α and CA-IX immunoreactivity after the first hyperthermia fraction was similar and there was an inverse relationship between temperature and the direction of change of CA-IX. There were no significant changes in interstitial fluid pressure, VEGF, vWF, apoptosis or necrosis as a function of treatment group or temperature. CONCLUSIONS We did not identify an advantage to a three to four per week hyperthermia prescription, and response data pointed to a one per week prescription being superior.
Collapse
Affiliation(s)
- Donald E Thrall
- College of Veterinary Medicine, North Carolina State University, Raleigh, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Cerezo D, Lencina M, Ruiz-Alcaraz AJ, Ferragut JA, Saceda M, Sanchez M, Cánovas M, García-Peñarrubia P, Martín-Orozco E. Acquisition of MDR phenotype by leukemic cells is associated with increased caspase-3 activity and a collateral sensitivity to cold stress. J Cell Biochem 2012; 113:1416-25. [PMID: 22173742 DOI: 10.1002/jcb.24016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The acquisition of a multidrug-resistant (MDR) phenotype by tumor cells that renders them unsusceptible to anti-neoplasic agents is one of the main causes of chemotherapy failure in human malignancies. The increased expression of P-glycoprotein (MDR1, P-gp, ABCB1) in tumor cells contributes to drug resistance by extruding chemotherapeutic agents or by regulating programmed cell death. In a study of MDR cell survival under cold stress conditions, it was found that resistant leukemic cells with P-gp over-expression, but not their sensitive counterparts, are hypersensitive to cold-induced cell death when exposed to temperatures below 4 °C. The transfection of parental cells with a P-gp-expressing plasmid makes these cells sensitive to cold stress, demonstrating an association between P-gp expression and cell death at low temperatures. Furthermore, we observed increased basal expression and activity of effector caspase-3 at physiological temperature (37 °C) in MDR cells compared with their parental cell line. Treatment with a caspase-3 inhibitor partially rescues MDR leukemic cells from cold-induced apoptosis, which suggests that the cell death mechanism may require caspase-3 activity. Taken together, these findings demonstrate that P-gp expression plays a role in MDR cell survival, and is accompanied by a collateral sensitivity to death induced by cold stress. These findings may assist in the design of specific therapeutic strategies to complement current chemotherapy treatment against cancer.
Collapse
Affiliation(s)
- David Cerezo
- Department of Biochemistry and Molecular Biology B and Immunology, University of Murcia, Murcia, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Ikemura K, Inoue K, Mizutani H, Oka H, Iwamoto T, Okuda M. An antioxidant Trolox restores decreased oral absorption of cyclosporine A after liver ischemia–reperfusion through distinct mechanisms between CYP3A and P-glycoprotein in the small intestine. Eur J Pharmacol 2012; 690:192-201. [DOI: 10.1016/j.ejphar.2012.06.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 06/20/2012] [Accepted: 06/20/2012] [Indexed: 10/28/2022]
|
32
|
Lo YL, Wang W, Ho CT. 7,3',4'-Trihydroxyisoflavone modulates multidrug resistance transporters and induces apoptosis via production of reactive oxygen species. Toxicology 2012; 302:221-32. [PMID: 22914566 DOI: 10.1016/j.tox.2012.08.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 07/31/2012] [Accepted: 08/02/2012] [Indexed: 11/16/2022]
Abstract
The development of multidrug resistance (MDR) to conventional chemoradiation therapy usually leads to failure in treating cervical cancer. This study aims to explore the effects and mechanisms of 7,3',4'-trihydroxyisoflavone (7,3',4'-THIF), one of the major metabolites of daidzein, on potentiating cytotoxicity of epirubicin (Epi), an anticancer drug in human cervical cancer HeLa cells. The cytotoxicity of Epi remarkably increased when it was combined with 7,3',4'-THIF. The cotreatment increased the reactive oxygen species (ROS) levels, including hydrogen peroxide and superoxide free radicals. 7,3',4'-THIF was shown to down-regulate the MDR1 promoter region composed of the elements of AP1, GC-box, and Y-box, as demonstrated by a luciferase assay. A negative regulation of hMDR1 gene with multiple transcription factors by this isoflavone may provide a novel molecular mechanism for MDR modulation. The mRNA expressions of MDR1, MDR-associated protein (MRP) 1, and MRP2 for the combined treatment were significantly lower than those of the Epi treatment. This result implies that MDR transporter-mediated Epi resistance is inhibited at various degrees by the addition of 7,3',4'-THIF. This isoflavone significantly enhanced intracellular Epi accumulation in HeLa cells. 7,3',4'-THIF and/or Epi triggered apoptosis through the upregulation of p53, Bax, and caspase-9. Apoptosis induction was also confirmed by the reduced mitochondrial membrane potential, increased sub-G1 and G2/M phases, nuclear DNA fragmentation, and chromatin condensation. Our findings demonstrate for the first time that 7,3',4'-THIF causes cell death in human cervical cancer cells through the ROS-dependent suppression of MDR transporters and p53-mediated activation of the intrinsic mitochondrial pathway of apoptosis. Thus, 7,3',4'-THIF has the potential to enhance the activity of a broad range of cancer chemotherapeutics in the MDR spectrum with the advantage of reducing adverse effects.
Collapse
Affiliation(s)
- Yu-Li Lo
- Department of Biological Sciences and Technology, National University of Tainan, Tainan, Taiwan.
| | | | | |
Collapse
|
33
|
Impact of environmental parameters on the activity of the P-glycoprotein. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 737:161-7. [PMID: 22259097 DOI: 10.1007/978-1-4614-1566-4_24] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
34
|
Hamilton TK, Hu N, Kolomitro K, Bell EN, Maurice DH, Graham CH, Siemens DR. Potential therapeutic applications of phosphodiesterase inhibition in prostate cancer. World J Urol 2012; 31:325-30. [DOI: 10.1007/s00345-012-0848-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 02/22/2012] [Indexed: 11/30/2022] Open
|
35
|
Huang C, Li Y, Cao P, Xie Z, Qin Z. Synergistic effect of hyperthermia and neferine on reverse multidrug resistance in adriamycin-resistant SGC7901/ADM gastric cancer cells. ACTA ACUST UNITED AC 2011; 31:488. [DOI: 10.1007/s11596-011-0478-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Indexed: 11/27/2022]
|
36
|
Deactivation of signal transducer and activator of transcription 3 reverses chemotherapeutics resistance of leukemia cells via down-regulating P-gp. PLoS One 2011; 6:e20965. [PMID: 21677772 PMCID: PMC3108986 DOI: 10.1371/journal.pone.0020965] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Accepted: 05/16/2011] [Indexed: 02/07/2023] Open
Abstract
Multidrug resistance (MDR) caused by overexpression of p-glycoprotein is a major obstacle in chemotherapy of malignant cancer, which usually is characterized by constitutive activation of signal transducer and activator of transcription 3 (STAT3), but their relation between MDR and STAT3 remains unclear. Here, we showed that STAT3 was overexpressed and highly activated in adriamycin-resistant K562/A02 cells compared with its parental K562 cells. Blockade of activation of STAT3 by STAT3 decoy oligodeoxynucleotide (ODN) promoted the accumulation and increased their sensitivity to adriamycin by down-regulating transcription of mdr1 and expression of P-gp, which were further confirmed by using STAT3-specific inhibitor JSI-124. Inhibition of STAT3 could also decrease mdr1 promoter mediated luciferase expression by using mdr1 promoter luciferase reporter construct. Otherwise, activation of STAT3 by STAT3C improved mdr1 transcription and P-gp expression. The ChIP results demonstrated that STAT3 could bind to the potential promoter region of mdr1, and STAT3 decoy depressed the binding. Further mutation assay show +64∼+72 region could be the STAT3 binding site. Our data demonstrate a role of STAT3 in regulation of mdr1 gene expression in myeloid leukemia and suggest that STAT3 may be a promising therapeutic target for overcoming MDR resistance in myeloid leukemia.
Collapse
|
37
|
Enhancement of drug efflux activity via MDR1 protein by spheroid culture of human hepatic cancer cells. J Biosci Bioeng 2011; 111:590-3. [PMID: 21354366 DOI: 10.1016/j.jbiosc.2011.01.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 12/29/2010] [Accepted: 01/04/2011] [Indexed: 11/20/2022]
Abstract
Human hepatic cancer cells, HepG2, formed spheroids on a poly-L-glutamic acid-coated dish. Doxorubicin (DOX) efflux activity of the cells in spheroid culture was higher than that in monolayer culture due to the higher expression of MDR1 protein of the cells in spheroids compared with those in monolayer. The amount of MDR1 per cell in spheroids was similar to that of hepatic tumor tissue in vivo. Consequently, it was suggested that the drug efflux activity of cells in spheroid culture reflected the activity of hepatic cancer cells. Furthermore, the IC(50) of DOX and epirubicin (EPI) in HepG2 cells, both of which are known to be exported by MDR1, were higher in spheroid compared with monolayer cells, while IC(50) of 5-fluorouracil (5-FU), which is not exported by MDR1 protein, was almost the same in both types of culture. The higher IC(50) of DOX and EPI in HepG2 cells in spheroid culture was associated with a higher efflux activity of the drugs in the spheroid-cultured cells, which appeared to reflect the IC(50) of DOX and EPI in cancer cells in vivo. Therefore, a spheroid culture of hepatic cancer cells seems to provide a promising cell-based in vitro assay system for examining the proper IC(50) values of anticancer agents that would reflect the drug resistance of cancer cells in vivo. In addition, the system would be useful in screening for inhibitors of MDR1 activity, which will help to overcome the multidrug resistance of cancer cells.
Collapse
|
38
|
El-Orabi NF, Rogers CB, Gray Edwards H, Schwartz DD. Heat-induced inhibition of superoxide dismutase and accumulation of reactive oxygen species leads to HT-22 neuronal cell death. J Therm Biol 2011. [DOI: 10.1016/j.jtherbio.2010.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
39
|
Moon EJ, Sonveaux P, Porporato PE, Danhier P, Gallez B, Batinic-Haberle I, Nien YC, Schroeder T, Dewhirst MW. NADPH oxidase-mediated reactive oxygen species production activates hypoxia-inducible factor-1 (HIF-1) via the ERK pathway after hyperthermia treatment. Proc Natl Acad Sci U S A 2010; 107:20477-82. [PMID: 21059928 PMCID: PMC2996638 DOI: 10.1073/pnas.1006646107] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Hyperthermia (HT) is a strong adjuvant treatment with radiotherapy and chemotherapy because it causes tumor reoxygenation. However, the detailed molecular mechanisms of how HT enhances tumor oxygenation have not been elucidated. Here we report that 1 h of HT activates hypoxia-inducible factor-1 (HIF-1) in tumors and its downstream targets, vascular endothelial growth factor (VEGF) and pyruvate dehydrogenase kinase 1 (PDK1). Consistent with HIF-1 activation and up-regulation of its downstream genes, HT also enhances tumor perfusion/vascularization and decreases oxygen consumption. As a result, tumor hypoxia is reduced after HT, suggesting that these physiological changes contribute to HT-induced tumor reoxygenation. Because HIF-1 is a potent regulator of tumor vascularization and metabolism, our findings suggest that HIF-1 plays a role in HT-induced tumor reoxygenation by transactivating its downstream targets. We demonstrate that NADPH oxidase-mediated reactive oxygen species production, as a mechanism, up-regulates HIF-1 after HT. Furthermore, we determine that this pathway is initiated by increased transcription of NADPH oxidase-1 through the ERK pathway. In conclusion, this study determines that, although HIF-1 is a good therapeutic target, the timing of its inhibition needs to be optimized to achieve the most beneficial outcome when it is combined with other treatments of HT, radiation, and chemotherapy.
Collapse
Affiliation(s)
| | | | - Paolo E. Porporato
- Unit of Pharmacology and Therapeutics and
- Unit of Biomedical Magnetic Resonance, University of Louvain Medical School, B-1200 Brussels, Belgium
| | - Pierre Danhier
- Unit of Pharmacology and Therapeutics and
- Unit of Biomedical Magnetic Resonance, University of Louvain Medical School, B-1200 Brussels, Belgium
| | - Bernard Gallez
- Unit of Biomedical Magnetic Resonance, University of Louvain Medical School, B-1200 Brussels, Belgium
| | | | | | - Thies Schroeder
- Radiation Oncology, Duke University Medical Center, Durham, NC 27710; and
| | - Mark W. Dewhirst
- Departments of Pathology and
- Radiation Oncology, Duke University Medical Center, Durham, NC 27710; and
| |
Collapse
|
40
|
Fang W, Geng X, Deng Y, Li Y, Shang E, Cen J, Lv P. Platelet activating factor induces blood brain barrier permeability alteration in vitro. J Neuroimmunol 2010; 230:42-7. [PMID: 20870297 DOI: 10.1016/j.jneuroim.2010.08.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Revised: 07/29/2010] [Accepted: 08/24/2010] [Indexed: 01/26/2023]
Abstract
The purposes of this article were to investigate whether blood brain barrier (BBB) permeability is altered after platelet activating factor (PAF) induced injury in vitro and elucidate the preliminary possible mechanisms of it. MTT method was used to observe cell damage after PAF incubation with rat brain microvessel endothelial cells (RBMECs). Intracellular concentrations of Nimodipine in normal and PAF injured RBMECs were estimated by LC-MS/MS analytical method to estimate BBB permeability. Accumulation of P-glycoprotein (P-gp) substrate rhodamine 123 in normal or PAF injured RBMECs was measured with Poly Immune Analysis System-1420 to evaluate the function of P-gp on RBMECs. Intercellular adhesion molecule-1 (ICAM-1) mRNA and protein expression levels in RBMECs were assayed by RT-PCR and flow cytometry respectively. Results showed that after RBMECs were incubated with 1 μM PAF for 24h, cell survival rate was decreased, and intracellular concentrations of Nimodipine were increased evidently. Rhodamine 123 accumulation between normal and PAF injured cells has no significant difference, but ICAM-1 mRNA and protein expression were increased remarkably in PAF injured cells, which could be inhibited by PAF antagonists. In conclusion, the present study demonstrated that BBB permeability was increased after PAF incubation, and which may be due to ICAM-1 up-regulating but not P-glycoprotein function alteration.
Collapse
Affiliation(s)
- Weirong Fang
- Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China
| | | | | | | | | | | | | |
Collapse
|
41
|
Liu L, Zhang H, Sun L, Gao Y, Jin H, Liang S, Wang Y, Dong M, Shi Y, Li Z, Fan D. ERK/MAPK activation involves hypoxia-induced MGr1-Ag/37LRP expression and contributes to apoptosis resistance in gastric cancer. Int J Cancer 2010; 127:820-829. [PMID: 19998339 DOI: 10.1002/ijc.25098] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We previously demonstrated that hypoxia increased the hypoxia-inducible factor (HIF-1)-dependent MGr1-Ag/37LRP expression, which enhanced adhesion of gastric cancer cells to laminin, inhibited drug-induced apoptosis and caused cell adhesion-mediated drug resistance (CAM-DR). Here, we investigated the role of extracellular-regulated kinase (ERK) 1/2 in the signaling mechanisms underlying these events. We found that hypoxia activated ERK activity in vitro and in vivo. Overexpression of mitogen-activated protein kinase (MAPK) kinase (MEK), which preferentially activated ERK, mimics, in a nonadditive way, hypoxia-induced activity of MGr1-Ag/37LRP promoter and expression of MGr1-Ag/37LRP. Furthermore, U0126, the MEK inhibitor, inhibited hypoxia- and MEK-induced MGr1-Ag/37LRP promoter activity in a dose-dependent manner. MEK inhibition also reversed hypoxia- and MEK-induced HIF-1 protein and its activity in a dose-dependent manner. We also investigated reactive oxygen species signaling this response. Exogenous addition of H(2)O(2) was sufficient to activate ERK in a dose-dependent profile. Reactive oxygen species scavengers of H(2)O(2) significantly inhibited hypoxia-induced ERK or HIF-1 activation and sequential expression of MGr1-Ag/37LRP. We also investigated the signaling in hypoxia-induced cell adhesion and apoptosis induced by vincristine. Hypoxia significantly enhanced adhesion of SGC7901 cells to laminin in a time-dependent manner, which might be inhibited by the MEK inhibitor U0126 and MGr1-Ag/37LRP siRNA. Consistent with results of adhesion assay, hypoxia-resistant apoptosis might be reversed by U0126 in a dose-dependent manner. Our results suggest that hypoxia-elicited MGr1-Ag/37LRP expression activated by HIF-1 depends on ERK activation. These events are dependent of reactive oxygen intermediates.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Apoptosis
- Blotting, Western
- Butadienes/pharmacology
- Cell Adhesion
- Cell Movement
- Dose-Response Relationship, Drug
- Enzyme Inhibitors/pharmacology
- Enzyme-Linked Immunosorbent Assay
- Humans
- Hydrogen Peroxide/pharmacology
- Hypoxia/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Mice
- Mitogen-Activated Protein Kinase 1/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 1/physiology
- Mitogen-Activated Protein Kinase 3/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 3/physiology
- Nitriles/pharmacology
- Oxidants/pharmacology
- Phosphorylation
- Promoter Regions, Genetic/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/pharmacology
- Reactive Oxygen Species/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Stomach Neoplasms/metabolism
- Stomach Neoplasms/pathology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Lili Liu
- Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Wartenberg M, Richter M, Datchev A, Günther S, Milosevic N, Bekhite MM, Figulla HR, Aran JM, Pétriz J, Sauer H. Glycolytic pyruvate regulates P-Glycoprotein expression in multicellular tumor spheroids via modulation of the intracellular redox state. J Cell Biochem 2010; 109:434-46. [PMID: 19950199 DOI: 10.1002/jcb.22422] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
UNLABELLED ABC transporters like P-glycoprotein (P-gp/ABCB1) are membrane proteins responsible for the transport of toxic compounds out of non-malignant cells and tumor tissue. AIM To investigate the effect of glycolysis and the tissue redox state on P-gp expression in multicellular tumor spheroids derived from prostate adenocarcinoma cells (DU-145), glioma cells (Gli36), and the human cervix carcinoma cell line KB-3-1 transfected with a P-gp-EGFP fusion gene that allows monitoring of P-gp expression in living cells. During cell culture of DU-145, Gli36, and KB-3-1 tumor spheroids P-gp expression was observed as well as increased lactate and decreased pyruvate levels and expression of glycolytic enzymes. Inhibition of glycolysis for 24 h by either iodoacetate (IA) or 2-deoxy-D-glucose (2-DDG) downregulated P-gp expression which was reversed upon coincubation with the radical scavenger ebselen as shown by semi-quantitative immunohistochemisty in DU-145 and Gli36 tumor spheroids, and by EGFP fluorescence in KB-3-1 tumor spheroids. Consequently endogenous ROS generation in DU-145 tumor spheroids was increased in the presence of either IA or 2-DDG, which was abolished upon coincubation with ebselen. Exogenous addition of pyruvate significantly reduced ROS generation, increased P-gp expression as well as efflux of the P-gp substrate doxorubicin. Doxorubicin transport was significantly blunted by 2-DDG and IA, indicating that inhibition of glycolysis reversed the multidrug resistance phenotype. In summary our data demonstrate that P-gp expression in tumor spheroids is closely related to the glycolytic metabolism of tumor cells and can be downregulated by glycolysis inhibitors via mechanisms that involve changes in the cellular redox state.
Collapse
Affiliation(s)
- Maria Wartenberg
- Department of Internal Medicine I, Cardiology Division, Friedrich Schiller University, Jena, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Thews O, Dillenburg W, Fellner M, Buchholz HG, Bausbacher N, Schreckenberger M, Rösch F. Activation of P-glycoprotein (Pgp)-mediated drug efflux by extracellular acidosis: in vivo imaging with 68Ga-labelled PET tracer. Eur J Nucl Med Mol Imaging 2010; 37:1935-42. [PMID: 20523987 DOI: 10.1007/s00259-010-1504-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 05/12/2010] [Indexed: 11/27/2022]
Abstract
PURPOSE In vitro it has been shown that the functional activity of P-glycoprotein (Pgp), an important drug transporter responsible for multidrug resistance, can be strongly increased by extracellular acidosis. Here mitogen-activated protein kinases (MAPK) (p38, ERK1/2) seem to play an important role for signal transduction. However, it is unclear whether these effects are also relevant in vivo. METHODS With the newly developed PET tracer Schiff base-based (68)Ga-MFL6.MZ the functional Pgp activity was visualized under acidic conditions and during inhibition of MAPKs non-invasively by means of microPET in rat tumours. Tumours were acidified either by inspiratory hypoxia (8% O(2)) or by injection of lactic acid. Inhibitors of the MAPK were injected intratumourally. RESULTS With increasing tumour volume the tumour pH changed from 7.0 to 6.7 and simultaneously the Pgp activity increased almost linearly. When the tumour was acidified by direct lactic acid injection the PET tracer uptake was reduced by 20% indicating a higher transport rate out of the cells. Changing the inspiratory O(2) fraction to 8% dynamically led to a reduction of extracellular pH and in parallel to a decrease of tracer concentration. While inhibition of the p38 pathway reduced the Pgp transport rate, inhibition of ERK1/2 had practically no impact. CONCLUSION An acidic extracellular environment significantly stimulates the Pgp activity. The p38 MAPK pathway plays an important role for Pgp regulation in vivo, whereas ERK1/2 is of minor importance. From these results new strategies for overcoming multidrug resistance (e.g. reducing tumour acidosis, inhibition of p38) may be developed.
Collapse
Affiliation(s)
- Oliver Thews
- Institute of Physiology and Pathophysiology, University Medicine Mainz, Duesbergweg 6, 55099 Mainz, Germany.
| | | | | | | | | | | | | |
Collapse
|
44
|
Rendic S, Guengerich FP. Update information on drug metabolism systems--2009, part II: summary of information on the effects of diseases and environmental factors on human cytochrome P450 (CYP) enzymes and transporters. Curr Drug Metab 2010; 11:4-84. [PMID: 20302566 PMCID: PMC4167379 DOI: 10.2174/138920010791110917] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Accepted: 02/22/2010] [Indexed: 12/14/2022]
Abstract
The present paper is an update of the data on the effects of diseases and environmental factors on the expression and/or activity of human cytochrome P450 (CYP) enzymes and transporters. The data are presented in tabular form (Tables 1 and 2) and are a continuation of previously published summaries on the effects of drugs and other chemicals on CYP enzymes (Rendic, S.; Di Carlo, F. Drug Metab. Rev., 1997, 29(1-2), 413-580., Rendic, S. Drug Metab. Rev., 2002, 34(1-2), 83-448.). The collected information presented here is as stated by the cited author(s), and in cases when several references are cited the latest published information is included. Inconsistent results and conclusions obtained by different authors are highlighted, followed by discussion of the major findings. The searchable database is available as an Excel file, for information about file availability contact the corresponding author.
Collapse
Affiliation(s)
- S Rendic
- University of Zagreb, Zagreb, Croatia.
| | | |
Collapse
|
45
|
Robertson SJ, Kania KD, Hladky SB, Barrand MA. P-glycoprotein expression in immortalised rat brain endothelial cells: comparisons following exogenously applied hydrogen peroxide and after hypoxia-reoxygenation. J Neurochem 2009; 111:132-41. [PMID: 19656260 DOI: 10.1111/j.1471-4159.2009.06306.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Levels of multidrug efflux transporter P-glycoprotein (P-gp) on endothelial cells lining brain blood vessels are important for limiting access of many compounds to the brain. In vivo studies have indicated that ischaemia-reperfusion that generates reactive oxygen species also increases P-gp levels in brain endothelial cells. To investigate possible mechanisms, in vitro studies were performed on immortalised (GPNT) and primary rat brain endothelial cells. Exposure to hydrogen peroxide (200 microM) resulted in intracellular oxidative stress as detected from higher levels of dichlorofluorescein fluorescence and raised levels of P-gp protein, mdr1a and mdr1b transcripts and, in GPNT cells, increased mdr1a and mdr1b promoter activity. The P-gp protein increases were abolished by pre-treatment with polyethylene glycol-catalase and were curtailed by co-culture with primary rat astrocytes. Exposure of GPNT cells to 6 h hypoxia followed by 24 h reoxygenation produced less intracellular oxidative stress as judged from smaller increments in dichlorofluorescein fluorescence but still resulted in raised levels of P-gp protein, an effect partially abolished by pre-treatment with polyethylene glycol-catalase. However, transcript levels and promoter activities were not significantly increased. These data suggest that hydrogen peroxide contributes to P-gp up-regulation following hypoxia-reoxygenation but the underlying mechanisms of its actions differ from those occurring after direct hydrogen peroxide application.
Collapse
|
46
|
Tang Y, McGoron AJ. Combined effects of laser-ICG photothermotherapy and doxorubicin chemotherapy on ovarian cancer cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2009; 97:138-44. [PMID: 19811928 DOI: 10.1016/j.jphotobiol.2009.09.001] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Revised: 08/31/2009] [Accepted: 09/03/2009] [Indexed: 12/31/2022]
Abstract
Doxorubicin (DOX) is an anthracycline antibiotic widely used in cancer chemotherapy. Its use is limited by cardiac toxicity and drug resistance. Hyperthermia can aid the functionality of DOX, but current hyperthermia delivery methods are hard to apply selectively and locally. The slow temperature increase associated with the external heating may lead to thermal tolerance in cancer cells. The FDA approved dye indocynine green (ICG) has been demonstrated to absorb near-infrared (NIR) light at 808 nm (ideal for tissue penetration) and emit the energy as heat, making it an ideal agent for localized hyperthermia with a rapid rate of temperature increase. The purpose of this study was to investigate the in vitro cytotoxic effect of combined chemotherapy and hyperthermia to a DOX resistant ovarian cancer cell line (SKOV-3). The effect of two different heating methods, ICG induced rapid rate heating and an incubator induced slow rate heating, were compared. All the experiments were conducted in 96-well plates. Cells were subjected to different concentrations of DOX and 60 min 43 degrees C incubation or 5 microM of ICG with 1 min 808 nm NIR laser. SRB assay was used to measure cell proliferation. ICG itself without laser irradiation was not toxic to SKOV-3 cells. The two types of hyperthermia individually produced similar cytotoxicity. DOX by itself was toxic with an IC(50) value of about 5 microM. Hyperthermia in combination with DOX achieved significantly greater cell killing/growth inhibition at all DOX concentrations compared to DOX alone. A subadditive cytotoxic effect was observed by combining DOX and 60 min 43 degrees C incubation which lead to a lowered DOX IC(50) value of about 1 microM. This value was even lower with 1 min laser-ICG photothermotherapy (0.1 microM) and, though not statistically significant, a synergistic effect may exist between DOX and laser-ICG photothermotherapy. The rate of heating may have an effect on chemotherapy-hyperthermia interaction. In conclusion, the combination of photothermal therapy and chemotherapy may provide a valuable tool for cancer treatment with minimized side effect.
Collapse
Affiliation(s)
- Yuan Tang
- Department of Biomedical Engineering, Florida International University, 10555 West Flagler Street, Miami, FL 33174, USA
| | | |
Collapse
|
47
|
Sauvant C, Nowak M, Wirth C, Schneider B, Riemann A, Gekle M, Thews O. Acidosis induces multi-drug resistance in rat prostate cancer cells (AT1) in vitro and in vivo by increasing the activity of the p-glycoprotein via activation of p38. Int J Cancer 2008; 123:2532-42. [PMID: 18729196 DOI: 10.1002/ijc.23818] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Because solid growing tumors often show hypoxia and pronounced extracellular acidosis, the aim of this study was to analyze the impact of an acidotic environment on the activity of the p-glycoprotein (pGP) and on the cellular content and cytotoxicity of the chemotherapeutic drug daunorubicin in the AT1 R-3327 Dunning prostate carcinoma cell line cultured in vitro and in vivo. In vitro, extracellular acidosis (pH 6.6) activated p38 and ERK1/2 and thereby induced daunorubicin resistance via a pronounced activation of pGP. De-novo protein synthesis was not necessary and analysis of transport kinetics indicated a fast and persistent pGP activation at pH 6.6 (when compared with 7.4). Intracellular acidification also induced daunorubicin resistance via activation of pGP, which was mediated by activation of p38 alone. In vivo, tumors were implanted subcutaneously, and the tumor pH was artificially lowered by forcing anaerobic metabolism. In vivo, the reduced extracellular pH of 6.6 was also able to induce daunorubicin resistance, which was abolished by inhibition of p38. These results suggest that pGP activity is dependent on extracellular pH in vitro and in vivo. Moreover, there is strong indication that this effect is mediated via activation of p38 in vivo. Activation of ERK is also suitable to induce pGP activity. Therefore, inhibition of p38 (and ERK) may be used to prevent acidosis induced increase in pGP activity and thereby attenuate multidrug resistance. In addition, supportive treatments reducing tumor acidosis may improve the cytotoxic effect of chemotherapeutic drugs.
Collapse
|
48
|
Yoshiba S, Ito D, Nagumo T, Shirota T, Hatori M, Shintani S. Hypoxia induces resistance to 5-fluorouracil in oral cancer cells via G(1) phase cell cycle arrest. Oral Oncol 2008; 45:109-15. [PMID: 18710819 DOI: 10.1016/j.oraloncology.2008.04.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2008] [Revised: 04/03/2008] [Accepted: 04/03/2008] [Indexed: 01/15/2023]
Abstract
Malignant tumors are exposed to various levels of hypoxic condition in vivo. It has been known that tumor cells under hypoxia are resistant to chemotherapies. To clarify the mechanism of the hypoxia-induced chemoresistance, we evaluated the effects of hypoxia on the resistance of oral squamous cell carcinoma (OSCC) cell lines to 5-fluorouracil (5-FU). OSCC cells were divided to two groups by the proliferation activity under hypoxic condition; hypoxia-resistant (HR) and hypoxia-sensitive (HS) cells. Growth of HS cells were inhibited by hypoxia and introduced to G(1) arrest in cell cycle. 5-FU effect on HS cell viability was markedly reduced in hypoxic condition without an induction of chemoresistant related protein, P-glycoprotein. However, proliferation, cell cycle, and 5-FU sensitivity of HR cells were not affected by hypoxia. Hypoxia-inducible factor (HIF)-1alpha was induced by hypoxia in all OSCC cell lines, but diminished in HS cells within 48h. Expression of p21 and p27 was strongly augmented and CyclinD expression was reduced by hypoxia in HS cells. However, the expression of these proteins was constitutive in HR cells during 48h hypoxic culture. Phosphorylation of mammalian target of rapamycin (mTOR) was reduced by hypoxia in HS cells. From these findings, we concluded that HS OSCC cells acquire 5-FU resistance under hypoxia by G(1)/S transition through an upregulation of cell cycle inhibitors.
Collapse
Affiliation(s)
- Sayaka Yoshiba
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, Kitasenzoku 2-1-1, Ohta-ku, Tokyo 145-8515, Japan.
| | | | | | | | | | | |
Collapse
|
49
|
Marignol L, Coffey M, Lawler M, Hollywood D. Hypoxia in prostate cancer: A powerful shield against tumour destruction? Cancer Treat Rev 2008; 34:313-27. [DOI: 10.1016/j.ctrv.2008.01.006] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2007] [Revised: 12/17/2007] [Accepted: 01/12/2008] [Indexed: 01/23/2023]
|
50
|
Xiao-Dong L, Zhi-Hong Y, Hui-Wen Y. Repetitive/temporal hypoxia increased P-glycoprotein expression in cultured rat brain microvascular endothelial cells in vitro. Neurosci Lett 2008; 432:184-7. [PMID: 18241990 DOI: 10.1016/j.neulet.2007.12.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2007] [Revised: 12/07/2007] [Accepted: 12/10/2007] [Indexed: 10/22/2022]
Abstract
The aim of the study was to investigate whether repetitive/temporal hypoxia up-regulated P-glycoprotein (P-gp) in cultured rat brain microvascular endothelial cells (rBMECs). Cultured rBMECs were used as in vitro blood brain barrier (BBB) model. Cells reached confluence were subjected to temporal hypoxic exposure. Under free-glucose cultured medium, the cells were covered by sterile paraffin oil for 15 min, inducing temporal hypoxic exposure. The hypoxic-exposure was carried out once every day up to 8 days, leading to the repetitive/temporal hypoxia in rBMECs. The cell viability was tested using CCK-8 kit, function and levels of P-gp in the cells were measured using rhodamine 123 uptake and western blot, respectively. It was found that 8-temporal hypoxic exposure induced 1.6-fold increase of P-gp level in cells, accompanied by decrease of cellular accumulation of rhodamine 123. Cellular accumulation of phenobarbital was also decreased. These findings indicated that repetitive/temporal hypoxia may be one of the factors resulting in P-gp overexpression in refractory epilepsy.
Collapse
Affiliation(s)
- Liu Xiao-Dong
- Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China.
| | | | | |
Collapse
|