1
|
Alsheleh T, Zraikat M, Daoud F, Alqudah DA, Abdelghany S, Abu Siniyeh A, Alshaer W. In vitro cellular interaction of drug-loaded liposomes with 2D and 3D cell culture of U87-MG cell line. PLoS One 2025; 20:e0320374. [PMID: 40131912 PMCID: PMC11936199 DOI: 10.1371/journal.pone.0320374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 02/18/2025] [Indexed: 03/27/2025] Open
Abstract
The distinctive physiological and physical properties of 3D cultures that mimic tumor microenvironments in vivo make them more suitable for assessing the efficacy of drugs and nanoparticles compared to 2D culture models. Therefore, this study aims to examine and contrast how liposomes interact with cell cultures in both 2D and 3D models. Hanging drop technique was used to generate 3D spheroids. Cellular toxicity of Doxorubicin and Doxil®-liposomes was tested using an MTT assay. Cellular uptake of Doxil®-liposomes was investigated in 3D and 2D cell culture models using flow cytometry and confocal microscopy. Finally, migration and invasion assays were used to investigate the Doxil®-liposomes interaction with the two models 2D model and 3D model, respectively. Our findings show that cells were able to form spheroid structures when a specific cell ratio was maintained. Flow cytometry analysis revealed that 2D cells exhibited higher Doxil®-liposome uptake than 3D cells. The data obtained from confocal and fluorescent microscopy supported the findings of the flow cytometry analysis. Furthermore, the MTT assay showed that Doxil®-liposomes induced less metabolic-disruption compared to free Doxorubicin. Our results also demonstrated that Doxil®-liposomes interacted more loosely with the 3D model than 2D cells, which was further confirmed by measurements of the total migration and invasion areas. Therefore, a 3D model replicating the in vivo conditions of tumor structure and extracellular matrix to assess the delivery of liposomal-nanoparticles to spheroids through a collagen matrix can be more informative and recapitulate the in vivo microenvironment than the 2D model.
Collapse
Affiliation(s)
- Tasneem Alsheleh
- Department of Biology, Faculty of Science, The University of Jordan, Amman, Jordan
| | - Manar Zraikat
- Department of Pharmacology, Faculty of Medicine, The University of Jordan, Amman, Jordan
| | - Fadwa Daoud
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Dana A. Alqudah
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Sharif Abdelghany
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The University of Jordan, Amman, Jordan
| | - Ahmed Abu Siniyeh
- Department of Clinical Laboratory Sciences, Faculty of Science, The University of Jordan, Amman, Jordan
| | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| |
Collapse
|
2
|
Scott NR, Kang S, Parekh SH. Mechanosensitive nuclear uptake of chemotherapy. SCIENCE ADVANCES 2024; 10:eadr5947. [PMID: 39693448 DOI: 10.1126/sciadv.adr5947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/14/2024] [Indexed: 12/20/2024]
Abstract
The nucleus is at the nexus of mechanotransduction and the final barrier for most first line chemotherapeutics. Here, we study the intersection between nuclear-cytoskeletal coupling and chemotherapy nuclear internalization. We find that chronic and acute modulation of intracellular filaments changes nuclear influx of doxorubicin (DOX). Rapid changes in cell strain by disruption of cytoskeletal and nuclear filaments sensitize nuclei to DOX, whereas chronic reduction of cell strain desensitize nuclei to DOX. Extracted nuclei from invasive cancer cells lines from different tissues have distinct nuclear permeability to DOX. Last, we show that mechano-priming of cells by paclitaxel markedly improves DOX nuclear internalization, rationalizing the observed drug synergies. Our findings reveal that nuclear uptake is a critical, previously unquantified aspect of drug resistance. With nuclear permeability to chemotherapy being tunable via modulation of nuclear mechanotransduction, mechano-priming may be useful to help overcome drug resistance in the future.
Collapse
Affiliation(s)
- Nicholas R Scott
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Sowon Kang
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Sapun H Parekh
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
3
|
Carretta A, Moscardini A, Signore G, Debellis D, Catalano F, Marotta R, Palmieri V, Tedeschi G, Scipioni L, Pozzi D, Caracciolo G, Beltram F, Cardarelli F. The supramolecular processing of liposomal doxorubicin hinders its therapeutic efficacy in cells. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200836. [PMID: 39050990 PMCID: PMC11268116 DOI: 10.1016/j.omton.2024.200836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/06/2024] [Accepted: 06/13/2024] [Indexed: 07/27/2024]
Abstract
The successful trajectory of liposome-encapsulated doxorubicin (e.g., Doxil, which has been approved by the U.S. Food and Drug Administration) as an anticancer nanodrug in clinical applications is contradicted by in vitro cell viability data that highlight its reduced efficacy in promoting cell death compared with non-encapsulated doxorubicin. No reports to date have provided a mechanistic explanation for this apparently discordant evidence. Taking advantage of doxorubicin intrinsic fluorescence and time-resolved optical microscopy, we analyze the uptake and intracellular processing of liposome-encapsulated doxorubicin (L-DOX) in several in vitro cellular models. Cell entry of L-DOX was found to lead to a rapid (seconds to minutes), energy- and temperature-independent release of crystallized doxorubicin nanorods into the cell cytoplasm, which then disassemble into a pool of fibril-shaped derivatives capable of crossing the cellular membrane while simultaneously releasing active drug monomers. Thus, a steady state is rapidly established in which the continuous supply of crystal nanorods from incoming liposomes is counteracted by a concentration-guided efflux in the extracellular medium of fibril-shaped derivatives and active drug monomers. These results demonstrate that liposome-mediated delivery is constitutively less efficient than isolated drug in establishing favorable conditions for drug retention in the cell. In addition to explaining previous contradictory evidence, present results impose careful rethinking of the synthetic identity of encapsulated anticancer drugs.
Collapse
Affiliation(s)
- Annalisa Carretta
- Scuola Normale Superiore, Laboratorio NEST, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Aldo Moscardini
- Scuola Normale Superiore, Laboratorio NEST, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Giovanni Signore
- Biochemistry Unit, Department of Biology, University of Pisa, via San Zeno 51, 56123 Pisa, Italy
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy
| | - Doriana Debellis
- Electron Microscopy Facility, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy
| | - Federico Catalano
- Electron Microscopy Facility, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy
| | - Roberto Marotta
- Electron Microscopy Facility, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy
| | - Valentina Palmieri
- Istituto dei Sistemi Complessi ISC CNR, Via dei Taurini 19, 00185 Rome, Italy
| | - Giulia Tedeschi
- Laboratory for Fluorescence Dynamics, Biomedical Engineering Department, University of California, Irvine, Irvine, CA, USA
| | - Lorenzo Scipioni
- Laboratory for Fluorescence Dynamics, Biomedical Engineering Department, University of California, Irvine, Irvine, CA, USA
| | - Daniela Pozzi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Giulio Caracciolo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Fabio Beltram
- Scuola Normale Superiore, Laboratorio NEST, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Francesco Cardarelli
- Scuola Normale Superiore, Laboratorio NEST, Piazza San Silvestro 12, 56127 Pisa, Italy
| |
Collapse
|
4
|
Kanno H, Hiramatsu K, Mikami H, Nakayashiki A, Yamashita S, Nagai A, Okabe K, Li F, Yin F, Tominaga K, Bicer OF, Noma R, Kiani B, Efa O, Büscher M, Wazawa T, Sonoshita M, Shintaku H, Nagai T, Braun S, Houston JP, Rashad S, Niizuma K, Goda K. High-throughput fluorescence lifetime imaging flow cytometry. Nat Commun 2024; 15:7376. [PMID: 39231964 PMCID: PMC11375057 DOI: 10.1038/s41467-024-51125-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 07/31/2024] [Indexed: 09/06/2024] Open
Abstract
Flow cytometry is a vital tool in biomedical research and laboratory medicine. However, its accuracy is often compromised by undesired fluctuations in fluorescence intensity. While fluorescence lifetime imaging microscopy (FLIM) bypasses this challenge as fluorescence lifetime remains unaffected by such fluctuations, the full integration of FLIM into flow cytometry has yet to be demonstrated due to speed limitations. Here we overcome the speed limitations in FLIM, thereby enabling high-throughput FLIM flow cytometry at a high rate of over 10,000 cells per second. This is made possible by using dual intensity-modulated continuous-wave beam arrays with complementary modulation frequency pairs for fluorophore excitation and acquiring fluorescence lifetime images of rapidly flowing cells. Moreover, our FLIM system distinguishes subpopulations in male rat glioma and captures dynamic changes in the cell nucleus induced by an anti-cancer drug. FLIM flow cytometry significantly enhances cellular analysis capabilities, providing detailed insights into cellular functions, interactions, and environments.
Collapse
Grants
- R35 GM152076 NIGMS NIH HHS
- This work was supported by JSPS Core-to-Core Program (K. G.), JSPS KAKENHI Grant Numbers 19H05633 and 20H00317 (K. G.), Ogasawara Foundation (K. G.), Nakatani Foundation (K. G.), Konica Minolta Foundation (K. G.), Philipp Franz von Siebold Award (K. G.), Humboldt Association of Japan (K. G.), Precise Measurement Technology Promotion Foundation (H. M.), JST PRESTO (JPMJPR1878) (K. H.), JST FOREST (21470594) (K. H.), JSPS Gran-in-Aid for Scientific Research (B) (22538379) (K. H.), JSPS Grant-in-Aid for Young Scientists (20K15227) (K. H.), Research Foundation for Opto-Science and Technology (K. H.), JSPS KAKENHI Grant Numbers 21J10600 and 24K18149 (H. K.), Konica Minolta Light Future Incentive Award (H. K.). We thank Mayu Sehara for her help with the cell sample preparation. The manuscript underwent editing with the assistance of a large language model (LLM).
Collapse
Affiliation(s)
- Hiroshi Kanno
- Department of Chemistry, The University of Tokyo, Tokyo, Japan.
- Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, Miyagi, Japan.
| | - Kotaro Hiramatsu
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
- Department of Chemistry, Kyushu University, Fukuoka, Japan
| | - Hideharu Mikami
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
- Research Institute for Electronic Science, Hokkaido University, Hokkaido, Japan
| | - Atsushi Nakayashiki
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Shota Yamashita
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Arata Nagai
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Kohki Okabe
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Fan Li
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | - Fei Yin
- Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Keita Tominaga
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | | | - Ryohei Noma
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, Osaka, Japan
| | - Bahareh Kiani
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Olga Efa
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Martin Büscher
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Tetsuichi Wazawa
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, Osaka, Japan
| | | | - Hirofumi Shintaku
- Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Takeharu Nagai
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, Osaka, Japan
| | - Sigurd Braun
- Institute for Genetics, Justus-Liebig-University Giessen, Giessen, Germany
| | - Jessica P Houston
- Department of Chemical and Materials Engineering, New Mexico State University, Las Cruces, NM, USA
| | - Sherif Rashad
- Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Department of Neurosurgical Engineering and Translational Neuroscience Graduate School of Biomedical Engineering, Tohoku University, Miyagi, Japan
| | - Kuniyasu Niizuma
- Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Department of Neurosurgical Engineering and Translational Neuroscience Graduate School of Biomedical Engineering, Tohoku University, Miyagi, Japan
| | - Keisuke Goda
- Department of Chemistry, The University of Tokyo, Tokyo, Japan.
- Institute of Technological Sciences, Wuhan University, Hubei, China.
- Department of Bioengineering, University of California, Los Angeles, CA, USA.
| |
Collapse
|
5
|
Januškevičienė I, Petrikaitė V. Exploring doxorubicin transport in 2D and 3D models of MDA-MB-231 sublines: impact of hypoxia and cellular heterogeneity on doxorubicin accumulation in cells. Am J Cancer Res 2024; 14:3584-3599. [PMID: 39113879 PMCID: PMC11301288 DOI: 10.62347/vnwh9165] [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: 05/31/2024] [Accepted: 07/12/2024] [Indexed: 08/10/2024] Open
Abstract
Triple-negative breast cancer (TNBC) treatment is challenging due to its aggressive nature and heterogeneity of this type of cancer, characterized by various subtypes and intratumoral diversity. Doxorubicin (DOX) plays a crucial role in TNBC chemotherapy reducing the tumor size and improving patient survival. However, decreased drug uptake and increased resistance in specific cell subpopulations reduce the effectiveness of the treatment. This study explored the differences in DOX transport in MDA-MB-231 phenotypic sublines in cell monolayer (2D model) and cell spheroids (3D cultures). Cell spheroids were formed using magnetic 3D Bioprinting method. DOX transport into cells and spheroids was evaluated using fluorescence microscopy after different incubation durations with DOX in normoxia and hypoxia. In hypoxia, DOX transport into cells was 2.5 to 5-fold lower than in normoxia. The subline F5 monolayer-cultured cells exhibited the highest DOX uptake, while subline H2 cells showed the lowest uptake in normoxia and hypoxia. In 3D cultures, DOX transport was up to 2-fold lower in spheroids formed from subline H2 cells. Spheroids from subline D8 and MDA-MB-231 parent cells had the highest DOX uptake. A correlation was observed between the characteristics of the cells and their resistance to anticancer drugs. The results indicate that different cancer cell subpopulations in tumours due to differences in drug uptake could significantly impact treatment efficacy.
Collapse
Affiliation(s)
- Indrė Januškevičienė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences Sukilėlių av., LT-50162 Kaunas, Lithuania
| | - Vilma Petrikaitė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences Sukilėlių av., LT-50162 Kaunas, Lithuania
| |
Collapse
|
6
|
Degerstedt O, O'Callaghan P, Clavero AL, Gråsjö J, Eriksson O, Sjögren E, Hansson P, Heindryckx F, Kreuger J, Lennernäs H. Quantitative imaging of doxorubicin diffusion and cellular uptake in biomimetic gels with human liver tumor cells. Drug Deliv Transl Res 2024; 14:970-983. [PMID: 37824040 DOI: 10.1007/s13346-023-01445-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2023] [Indexed: 10/13/2023]
Abstract
Novel tumor-on-a-chip approaches are increasingly used to investigate tumor progression and potential treatment options. To improve the effect of any cancer treatment it is important to have an in depth understanding of drug diffusion, penetration through the tumor extracellular matrix and cellular uptake. In this study, we have developed a miniaturized chip where drug diffusion and cellular uptake in different hydrogel environments can be quantified at high resolution using live imaging. Diffusion of doxorubicin was reduced in a biomimetic hydrogel mimicking tissue properties of cirrhotic liver and early stage hepatocellular carcinoma (373 ± 108 µm2/s) as compared to an agarose gel (501 ± 77 µm2/s, p = 0.019). The diffusion was further lowered to 256 ± 30 µm2/s (p = 0.028) by preparing the biomimetic gel in cell media instead of phosphate buffered saline. The addition of liver tumor cells (Huh7 or HepG2) to the gel, at two different densities, did not significantly influence drug diffusion. Clinically relevant and quantifiable doxorubicin concentration gradients (1-20 µM) were established in the chip within one hour. Intracellular increases in doxorubicin fluorescence correlated with decreasing fluorescence of the DNA-binding stain Hoechst 33342 and based on the quantified intracellular uptake of doxorubicin an apparent cell permeability (9.00 ± 0.74 × 10-4 µm/s for HepG2) was determined. Finally, the data derived from the in vitro model were applied to a spatio-temporal tissue concentration model to evaluate the potential clinical impact of a cirrhotic extracellular matrix on doxorubicin diffusion and tumor cell uptake.
Collapse
Affiliation(s)
- Oliver Degerstedt
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Paul O'Callaghan
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Ada Lerma Clavero
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Johan Gråsjö
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Olle Eriksson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Erik Sjögren
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Per Hansson
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Femke Heindryckx
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Johan Kreuger
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Hans Lennernäs
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
7
|
Sharma A, Verwilst P, Li M, Ma D, Singh N, Yoo J, Kim Y, Yang Y, Zhu JH, Huang H, Hu XL, He XP, Zeng L, James TD, Peng X, Sessler JL, Kim JS. Theranostic Fluorescent Probes. Chem Rev 2024; 124:2699-2804. [PMID: 38422393 PMCID: PMC11132561 DOI: 10.1021/acs.chemrev.3c00778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/31/2024] [Accepted: 02/08/2024] [Indexed: 03/02/2024]
Abstract
The ability to gain spatiotemporal information, and in some cases achieve spatiotemporal control, in the context of drug delivery makes theranostic fluorescent probes an attractive and intensely investigated research topic. This interest is reflected in the steep rise in publications on the topic that have appeared over the past decade. Theranostic fluorescent probes, in their various incarnations, generally comprise a fluorophore linked to a masked drug, in which the drug is released as the result of certain stimuli, with both intrinsic and extrinsic stimuli being reported. This release is then signaled by the emergence of a fluorescent signal. Importantly, the use of appropriate fluorophores has enabled not only this emerging fluorescence as a spatiotemporal marker for drug delivery but also has provided modalities useful in photodynamic, photothermal, and sonodynamic therapeutic applications. In this review we highlight recent work on theranostic fluorescent probes with a particular focus on probes that are activated in tumor microenvironments. We also summarize efforts to develop probes for other applications, such as neurodegenerative diseases and antibacterials. This review celebrates the diversity of designs reported to date, from discrete small-molecule systems to nanomaterials. Our aim is to provide insights into the potential clinical impact of this still-emerging research direction.
Collapse
Affiliation(s)
- Amit Sharma
- Amity
School of Chemical Sciences, Amity University
Punjab, Sector 82A, Mohali 140 306, India
| | - Peter Verwilst
- Rega
Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49, Box 1041, 3000 Leuven, Belgium
| | - Mingle Li
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
| | - Dandan Ma
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Nem Singh
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Jiyoung Yoo
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Yujin Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Ying Yang
- School of
Light Industry and Food Engineering, Guangxi
University, Nanning, Guangxi 530004, China
| | - Jing-Hui Zhu
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Haiqiao Huang
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Xi-Le Hu
- Key
Laboratory for Advanced Materials and Joint International Research
Laboratory of Precision Chemistry and Molecular Engineering, Feringa
Nobel Prize Scientist Joint Research Center, School of Chemistry and
Molecular Engineering, East China University
of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Xiao-Peng He
- Key
Laboratory for Advanced Materials and Joint International Research
Laboratory of Precision Chemistry and Molecular Engineering, Feringa
Nobel Prize Scientist Joint Research Center, School of Chemistry and
Molecular Engineering, East China University
of Science and Technology, 130 Meilong Road, Shanghai 200237, China
- National
Center for Liver Cancer, the International Cooperation Laboratory
on Signal Transduction, Eastern Hepatobiliary
Surgery Hospital, Shanghai 200438, China
| | - Lintao Zeng
- School of
Light Industry and Food Engineering, Guangxi
University, Nanning, Guangxi 530004, China
| | - Tony D. James
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
- School
of Chemistry and Chemical Engineering, Henan
Normal University, Xinxiang 453007, China
| | - Xiaojun Peng
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- State
Key Laboratory of Fine Chemicals, Dalian
University of Technology, Dalian 116024, China
| | - Jonathan L. Sessler
- Department
of Chemistry, The University of Texas at
Austin, Texas 78712-1224, United
States
| | - Jong Seung Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
- TheranoChem Incorporation, Seongbuk-gu, Seoul 02841, Korea
| |
Collapse
|
8
|
Al-Kenany SA, Al-Shawi NN. Protective effect of cafestol against doxorubicin-induced cardiotoxicity in rats by activating the Nrf2 pathway. Front Pharmacol 2023; 14:1206782. [PMID: 37377932 PMCID: PMC10291064 DOI: 10.3389/fphar.2023.1206782] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Doxorubicin (DOX) is an efficient antineoplastic agent with a broad antitumor spectrum; however, doxorubicin-associated cardiotoxic adverse effect through oxidative damage and apoptosis limits its clinical application. Cafestol (Caf) is a naturally occurring diterpene in unfiltered coffee with unique antioxidant, antimutagenic, and anti-inflammatory activities by activating the Nrf2 pathway. The present study aimed to investigate the potential chemoprotective effect of cafestol on DOX-induced cardiotoxicity in rats. Wistar albino rats of both sexes were administered cafestol (5 mg/kg/day) for 14 consecutive days by oral gavage alone or with doxorubicin which was injected as a single dose (15 mg/kg intraperitoneally at day 14) to induce toxicity. The result showed that Caf significantly improved cardiac injury induced by doxorubicin, decreased serum levels of CK-MB, LDH, ALP, and ALT, and improved histopathological changes. In addition, cafestol significantly inhibited DOX-induced cardiac oxidative stress as seen in the reduced level of MDA and increased GSH, SOD, CAT, and Gpx-1 cardiac tissue levels; cafestol significantly enhanced Nrf2 gene and protein expression and promoted the expression of downstream antioxidant genes HO-1 and NQO-1 and downregulated Keap1 and NF-κB genes' expression; in addition, Caf significantly reduced inflammatory mediators, TNF-α, and IL-1β levels and inhibited cardiac apoptosis by modulating Bax and Casp 3 tissue levels and reduced TUNEL-positive cardiomyocytes. In conclusion, the present study confirmed that cafestol improved the cardiotoxic effects induced by doxorubicin through the regulation of apoptosis and oxidative stress response through the Nrf2 pathway; this study suggests that cafestol may serve as a potential adjuvant in chemotherapy to alleviate DOX-induced toxicities.
Collapse
|
9
|
Li M, Wu J, Lin D, Yang J, Jiao N, Wang Y, Liu L. A diatom-based biohybrid microrobot with a high drug-loading capacity and pH-sensitive drug release for target therapy. Acta Biomater 2022; 154:443-453. [PMID: 36243369 DOI: 10.1016/j.actbio.2022.10.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/25/2022] [Accepted: 10/07/2022] [Indexed: 12/14/2022]
Abstract
Targeted delivery is a promising mean for various biomedical applications, and various micro/nano robots have been created for drug delivery. Mesoporous silica has been shown to be successful as a drug delivery carrier in numerous studies. However, mesoporous silica preparation usually requires expensive and toxic chemicals, which limits its biomedical applications. Diatoms, as the naturally porous silica structure, are promising substitutes for the artificial mesoporous silica preparation. However, the current studies utilizing intact diatom frustules as drug delivery packets lack flexible and controllable locomotion. Herein, we propose a biohybrid magnetic microrobot based on Thalassiosira weissflogii frustules (TWFs) as a cargo packet for targeted drug delivery using a simple preparation method. Biohybrid microrobots are fabricated in large quantities by attaching magnetic nanoparticles (Fe3O4) to the surface of diatoms via electrostatic adsorption. Biohybrid microrobots are agile and controllable under the influence of external magnetic fields. They could be precisely controlled to follow specific trajectories or to move as swarms. The cooperation of the two motion modes of the biohybrid microrobots increased microrobots' environmental adaptability. Microrobots have a high drug-loading capacity and pH-sensitive drug release. In vitro cancer cell experiments further demonstrated the controllability of diatom microrobots for targeted drug delivery. The biohybrid microrobots reported in this paper convert natural diatoms into cargo packets for biomedical applications, which possess active and controllable properties and show huge potential for targeted anticancer therapy. STATEMENT OF SIGNIFICANCE: In this study, diatoms with good biocompatibility were used to prepare biohybrid magnetic microrobots. Compared with the current diatom-based systems for drug delivery, the microrobots prepared in this study for targeted drug delivery have more flexible motion characteristics and exhibit certain swarming behaviors. Under the same magnetic field strength, by changing the magnetic field frequency, the movement state of the diatoms can be changed to pass through the narrow channel, so that it has better environmental adaptability.
Collapse
Affiliation(s)
- Mengyue Li
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China; Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junfeng Wu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China; Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Daojing Lin
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China; Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jia Yang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China; Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Niandong Jiao
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China; Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China.
| | - Yuechao Wang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China; Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China
| | - Lianqing Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China; Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China.
| |
Collapse
|
10
|
Druzhkova I, Nikonova E, Ignatova N, Koryakina I, Zyuzin M, Mozherov A, Kozlov D, Krylov D, Kuznetsova D, Lisitsa U, Shcheslavskiy V, Shirshin EA, Zagaynova E, Shirmanova M. Effect of Collagen Matrix on Doxorubicin Distribution and Cancer Cells' Response to Treatment in 3D Tumor Model. Cancers (Basel) 2022; 14:cancers14225487. [PMID: 36428580 PMCID: PMC9688511 DOI: 10.3390/cancers14225487] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/05/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
The extracellular matrix (ECM) plays an important role in regulation of many aspects of tumor growth and response to therapies. However, the specifics of the interaction of chemotherapeutic agents with cancer cells in the presence of collagen, the major component of ECM, is still poorly investigated. In this study, we explored distribution of doxorubicin (DOX) and its effects on cancer cells' metabolism in the presence of collagen with different structures in 3D models. For this, a combination of second harmonic generation imaging of collagen and multiphoton fluorescence microscopy of DOX, and metabolic cofactor NAD(P)H was used. It was found that collagen slowed down the diffusion of DOX and thus decreased the cellular drug uptake. Besides nuclei, DOX also targeted mitochondria leading to inhibition of oxidative phosphorylation, which was more pronounced in the cells growing in the absence of collagen. As a result, the cells in collagen displayed better viability upon treatment with DOX. Taken together, our data illustrate that tumor collagen contributes to heterogeneous and sub-optimal response to DOX and highlight the challenges in improving drug delivery and efficacy.
Collapse
Affiliation(s)
- Irina Druzhkova
- Research Institute of Experimental Oncology and Biotechnology, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia
- Correspondence:
| | - Elena Nikonova
- Lomonosov Moscow State University, 119991 Moscow, Russia
- Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia
| | - Nadezhda Ignatova
- Research Institute of Experimental Oncology and Biotechnology, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia
| | - Irina Koryakina
- School of Physics and Engineering, ITMO University, 9 Lomonosova St., 191002 St. Petersburg, Russia
| | - Mikhail Zyuzin
- School of Physics and Engineering, ITMO University, 9 Lomonosova St., 191002 St. Petersburg, Russia
| | - Artem Mozherov
- Research Institute of Experimental Oncology and Biotechnology, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia
| | - Dmitriy Kozlov
- Research Institute of Experimental Oncology and Biotechnology, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia
| | - Dmitry Krylov
- Research Institute of Experimental Oncology and Biotechnology, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia
| | - Daria Kuznetsova
- Research Institute of Experimental Oncology and Biotechnology, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia
- Lobachevsky State University of Nizhny Novgorod, 603022 Nizhny Novgorod, Russia
| | - Uliyana Lisitsa
- Research Institute of Experimental Oncology and Biotechnology, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia
| | - Vladislav Shcheslavskiy
- Research Institute of Experimental Oncology and Biotechnology, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia
| | - Evgeny A. Shirshin
- Lomonosov Moscow State University, 119991 Moscow, Russia
- Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia
| | - Elena Zagaynova
- Research Institute of Experimental Oncology and Biotechnology, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia
- Lobachevsky State University of Nizhny Novgorod, 603022 Nizhny Novgorod, Russia
| | - Marina Shirmanova
- Research Institute of Experimental Oncology and Biotechnology, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia
| |
Collapse
|
11
|
Krecsir A, Richter V, Wagner M, Schneckenburger H. Impact of Doxorubicin on Cell-Substrate Topology. Int J Mol Sci 2022; 23:ijms23116277. [PMID: 35682954 PMCID: PMC9181088 DOI: 10.3390/ijms23116277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/30/2022] [Accepted: 06/02/2022] [Indexed: 12/03/2022] Open
Abstract
Variable-Angle Total Internal Reflection Fluorescence Microscopy (VA-TIRFM) is applied in view of early detection of cellular responses to the cytostatic drug doxorubicin. Therefore, we determined cell-substrate topology of cultivated CHO cells transfected with a membrane-associated Green Fluorescent Protein (GFP) in the nanometer range prior to and subsequent to the application of doxorubicin. Cell-substrate distances increased up to a factor of 2 after 24 h of application. A reduction of these distances by again a factor 2 was observed upon cell aging, and an influence of the cultivation time is presently discussed. Applicability of VA-TIRFM was supported by measurements of MCF-7 breast cancer cells after membrane staining and incubation with doxorubicin, when cell-substrate distances increased again by a factor ≥ 2. So far, our method needs well-defined cell ages and staining of cell membranes or transfection with GFP or related molecules. Use of intrinsic fluorescence or even light-scattering methods to various cancer cell lines could make this method more universal in the future, e.g., in the context of early detection of apoptosis.
Collapse
|
12
|
Azimzadeh M, Aghili Z, Jannat B, Jafari S, Rafizadeh Tafti S, Nasirizadeh N. Nanocomposite of electrochemically reduced graphene oxide and gold nanourchins for electrochemical DNA detection. IET Nanobiotechnol 2022; 16:190-198. [PMID: 35442560 PMCID: PMC9178657 DOI: 10.1049/nbt2.12086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/24/2022] [Accepted: 04/02/2022] [Indexed: 12/19/2022] Open
Abstract
A nanocomposite of graphene oxide and gold nanourchins has been used here to modify the surface of a screen‐printed carbon electrode to enhance the sensitivity of the electrochemical DNA detection system. A specific single‐stranded DNA probe was designed based on the target DNA sequence and was thiolated to be self‐assembled on the surface of the gold nanourchins placed on the modified electrode. Doxorubicin was used as an electrochemical label to detect the DNA hybridisation using differential pulse voltammetry (DPV). The assembling process was confirmed using scanning electron microscopy (SEM) imaging, cyclic voltammetry (CV), and the EIS method. The high sensitivity of the proposed system led to a low detection limit of 0.16 fM and a wide linear range from 0.5 to 950.0 fM. The specificity of the DNA hybridisation and the signalling molecule (haematoxylin) caused very high selectivity towards the target DNA than other non‐specific sequences.
Collapse
Affiliation(s)
- Mostafa Azimzadeh
- Halal Research Center of IRI, MOH, Tehran, Iran.,Medical Nanotechnology & Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Department of Advanced Medical Sciences and Technologies, School of Paramedicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Zahra Aghili
- Food & Drug Control Reference Laboratories Center, FDA, MOH, Tehran, Iran
| | | | - Saeid Jafari
- Department of Textile and Polymer Engineering, Yazd Branch, Islamic Azad University, Yazd, Iran
| | - Saeed Rafizadeh Tafti
- Medical Nanotechnology & Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Navid Nasirizadeh
- Halal Research Center of IRI, MOH, Tehran, Iran.,Department of Textile and Polymer Engineering, Yazd Branch, Islamic Azad University, Yazd, Iran
| |
Collapse
|
13
|
Rao C, Sharma S, Garg R, Anjum F, Kaushik K, Nandi CK. Mapping the Time Dependent DNA Fragmentation caused by doxorubicin Loaded on PEGylated Carbogenic Nanodots using Fluorescence Lifetime Imaging and Super-resolution microscopy. Biomater Sci 2022; 10:4525-4537. [DOI: 10.1039/d2bm00641c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Doxorubicin is an anthracycline drug most commonly used in cancer therapy. It intercalates with the nuclear DNA and induces toxicity by causing DNA breaks and histone evictions. However, the kinetics...
Collapse
|
14
|
Gonsalves A, Tambe P, Le D, Thakore D, Wadajkar AS, Yang J, Nguyen KT, Menon JU. Synthesis and characterization of a novel pH-responsive drug-releasing nanocomposite hydrogel for skin cancer therapy and wound healing. J Mater Chem B 2021; 9:9533-9546. [PMID: 34757371 PMCID: PMC8725646 DOI: 10.1039/d1tb01934a] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Local skin cancer recurrence occurs in ∼12% of the patients post-surgery due to persistent growth of residual cancer cells. Wound infection is another significant complication following surgery. We report a novel in situ-forming nanocomposite hydrogel (NCH) containing PLGA-carboxymethyl chitosan nanoparticles (186 nm) for localized pH-responsive skin cancer therapy and wound healing. This injectable hydrogel, comprising of a citric acid-derived polymer backbone, gelled within 5 minutes, and demonstrated excellent swelling (283% of dry weight) and compressive strengths (∼5.34 MPa). Nanoparticle incorporation did not significantly affect hydrogel properties. The NCH effluents were cytocompatible with human dermal fibroblasts at 500 μg ml-1 concentration and demonstrated pH-dependent drug release and promising therapeutic efficacy against A431 and G361 skin cancer cells in vitro. Significant zones of inhibition were observed in S. aureus and E. coli cultures on NCH treatment, confirming its antibacterial properties. Our studies show that the pH-responsive NCH can be potentially used for adjuvant skin cancer treatment and wound healing.
Collapse
Affiliation(s)
- Andrea Gonsalves
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Road, Kingston, RI 02881, USA.
| | - Pranjali Tambe
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Duong Le
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Road, Kingston, RI 02881, USA.
| | - Dheeraj Thakore
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Aniket S Wadajkar
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jian Yang
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, USA
| | - Kytai T Nguyen
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jyothi U Menon
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Road, Kingston, RI 02881, USA.
- Department of Chemical Engineering, University of Rhode Island, Kingston, RI 02881, USA
| |
Collapse
|
15
|
Esawi E, Alshaer W, Mahmoud IS, Alqudah DA, Azab B, Awidi A. Aptamer-Aptamer Chimera for Targeted Delivery and ATP-Responsive Release of Doxorubicin into Cancer Cells. Int J Mol Sci 2021; 22:12940. [PMID: 34884745 PMCID: PMC8657665 DOI: 10.3390/ijms222312940] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 01/08/2023] Open
Abstract
Aptamers offer a great opportunity to develop innovative drug delivery systems that can deliver cargos specifically into targeted cells. In this study, a chimera consisting of two aptamers was developed to deliver doxorubicin into cancer cells and release the drug in cytoplasm in response to adenosine-5'-triphosphate (ATP) binding. The chimera was composed of the AS1411 anti-nucleolin aptamer for cancer cell targeting and the ATP aptamer for loading and triggering the release of doxorubicin in cells. The chimera was first produced by hybridizing the ATP aptamer with its complementary DNA sequence, which is linked with the AS1411 aptamer via a poly-thymine linker. Doxorubicin was then loaded inside the hybridized DNA region of the chimera. Our results show that the AS1411-ATP aptamer chimera was able to release loaded doxorubicin in cells in response to ATP. In addition, selective uptake of the chimera into cancer cells was demonstrated using flow cytometry. Furthermore, confocal laser scanning microscopy showed the successful delivery of the doxorubicin loaded in chimeras to the nuclei of targeted cells. Moreover, the doxorubicin-loaded chimeras effectively inhibited the growth of cancer cell lines and reduced the cytotoxic effect on the normal cells. Overall, the results of this study show that the AS1411-ATP aptamer chimera could be used as an innovative approach for the selective delivery of doxorubicin to cancer cells, which may improve the therapeutic potency and decrease the off-target cytotoxicity of doxorubicin.
Collapse
Affiliation(s)
- Ezaldeen Esawi
- Faculty of Medicine, The University of Jordan, Amman 11942, Jordan; (E.E.); (B.A.)
- Cell Therapy Centre, The University of Jordan, Amman 11942, Jordan;
| | - Walhan Alshaer
- Cell Therapy Centre, The University of Jordan, Amman 11942, Jordan;
| | - Ismail Sami Mahmoud
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa 13133, Jordan;
| | - Dana A. Alqudah
- Cell Therapy Centre, The University of Jordan, Amman 11942, Jordan;
| | - Bilal Azab
- Faculty of Medicine, The University of Jordan, Amman 11942, Jordan; (E.E.); (B.A.)
| | - Abdalla Awidi
- Faculty of Medicine, The University of Jordan, Amman 11942, Jordan; (E.E.); (B.A.)
- Cell Therapy Centre, The University of Jordan, Amman 11942, Jordan;
- Department of Hematology and Oncology, Jordan University Hospital, The University of Jordan, Amman 11942, Jordan
| |
Collapse
|
16
|
Blockade of TGF-βR improves the efficacy of doxorubicin by modulating the tumor cell motility and affecting the immune cells in a melanoma model. Naunyn Schmiedebergs Arch Pharmacol 2021; 394:2309-2322. [PMID: 34499199 DOI: 10.1007/s00210-021-02134-x] [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: 05/14/2021] [Accepted: 07/29/2021] [Indexed: 10/20/2022]
Abstract
TGF-β contributes to drug resistance and the invasiveness of tumor cells and weakens the anti-tumor immune responses. The present study aimed at examining the efficacy of the combination of SB431542, as a specific inhibitor of TGF-βR, and doxorubicin in controlling the melanoma tumor in mice. The impact of the combination of the doxorubicin and SB431542 on the cell growth, apoptosis, migration, and invasiveness of B16-F10 cells was examined. Besides, the B16-F10 tumor was induced in C57BL/6 mice, and the effects of the mentioned treatment on the tumor volume, survival, and the exhaustion state of T cells were evaluated. Although the combination of doxorubicin and SB431542 did not exhibit synergism in the inhibition of cell growth and apoptosis induction, it efficiently prohibited the migration and the epithelial to mesenchymal transition of B16-F10 cells, and the combination of doxorubicin and SB431542 caused an increase in mRNA levels of E-cadherin and, on the other hand, led to a decline in the expression of Vimentin. Tumor volume and the survival of tumor-bearing mice were efficiently controlled by the combination therapy. This treatment also eventuated in a decrease in the percentage of PD-L1+, TCD4+, and TCD8+ cells as indicators of exhausted T cells within the spleens of tumor-bearing mice. Blockade of TGF-βR also propelled the RAW 264.7 cells towards an anti-tumor M1 macrophage phenotype. The inhibition of TGF-βR demonstrated a potential to increase the efficacy of doxorubicin chemotherapy by the means of affecting cellular motility and restoring the anti-tumor immune responses.
Collapse
|
17
|
Wu J, Ma S, Li M, Hu X, Jiao N, Tung S, Liu L. Enzymatic/Magnetic Hybrid Micromotors for Synergistic Anticancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:31514-31526. [PMID: 34213305 DOI: 10.1021/acsami.1c07593] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Micro/nanomotors (MNMs), which propel by transforming various forms of energy into kinetic energy, have emerged as promising therapeutic nanosystems in biomedical applications. However, most MNMs used for anticancer treatment are only powered by one engine or provide a single therapeutic strategy. Although double-engined micromotors for synergistic anticancer therapy can achieve more flexible movement and efficient treatment efficacy, their design remains challenging. In this study, we used a facile preparation method to develop enzymatic/magnetic micromotors for synergetic cancer treatment via chemotherapy and starvation therapy (ST), and the size of micromotors can be easily regulated during the synthetic process. The enzymatic reaction of glucose oxidase, which served as the chemical engine, led to self-propulsion using glucose as a fuel and ST via a reduction in the energy available to cancer cells. Moreover, the incorporation of Fe3O4 nanoparticles as a magnetic engine enhanced the kinetic power and provided control over the direction of movement. Inherent pH-responsive drug release behavior was observed owing to the acidic decomposition of drug carriers in the intracellular microenvironment of cancer cells. This system displayed enhanced anticancer efficacy owing to the synergetic therapeutic strategies and increased cellular uptake in a targeted area because of the improved motion behavior provided by the double engines. Therefore, the demonstrated micromotors are promising candidates for anticancer biomedical microsystems.
Collapse
Affiliation(s)
- Junfeng Wu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuang Ma
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengyue Li
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xingyue Hu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Niandong Jiao
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China
| | - Steve Tung
- Department of Mechanical Engineering, University of Arkansas, Fayetteville, Arkansas 72701, United States
| | - Lianqing Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China
| |
Collapse
|
18
|
Español A, Salem A, Sanchez Y, Sales ME. Breast cancer: Muscarinic receptors as new targets for tumor therapy. World J Clin Oncol 2021; 12:404-428. [PMID: 34189066 PMCID: PMC8223712 DOI: 10.5306/wjco.v12.i6.404] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/26/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023] Open
Abstract
The development of breast cancer is a complex process that involves the participation of different factors. Several authors have demonstrated the overexpression of muscarinic acetylcholine receptors (mAChRs) in different tumor tissues and their role in the modulation of tumor biology, positioning them as therapeutic targets in cancer. The conventional treatment for breast cancer involves surgery, radiotherapy, and/or chemotherapy. The latter presents disadvantages such as limited specificity, the appearance of resistance to treatment and other side effects. To prevent these side effects, several schedules of drug administration, like metronomic therapy, have been developed. Metronomic therapy is a type of chemotherapy in which one or more drugs are administered at low concentrations repetitively. Recently, two chemotherapeutic agents usually used to treat breast cancer have been considered able to activate mAChRs. The combination of low concentrations of these chemotherapeutic agents with muscarinic agonists could be a useful option to be applied in breast cancer treatment, since this combination not only reduces tumor cell survival without affecting normal cells, but also decreases pathological neo-angiogenesis, the expression of drug extrusion proteins and the cancer stem cell fraction. In this review, we focus on the previous evidences that have positioned mAChRs as relevant therapeutic targets in breast cancer and analyze the effects of administering muscarinic agonists in combination with conventional chemotherapeutic agents in a metronomic schedule.
Collapse
Affiliation(s)
- Alejandro Español
- Laboratory of Immunopharmacology and Tumor Biology, CEFYBO CONICET University of Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Agustina Salem
- Laboratory of Immunopharmacology and Tumor Biology, CEFYBO CONICET University of Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Yamila Sanchez
- Laboratory of Immunopharmacology and Tumor Biology, CEFYBO CONICET University of Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - María Elena Sales
- Laboratory of Immunopharmacology and Tumor Biology, CEFYBO CONICET University of Buenos Aires, Buenos Aires C1121ABG, Argentina
| |
Collapse
|
19
|
Benassi JC, Barbosa FAR, Candiotto G, Grinevicius VMAS, Filho DW, Braga AL, Pedrosa RC. Docking and molecular dynamics predicted B-DNA and dihydropyrimidinone selenoesters interactions elucidating antiproliferative effects on breast adenocarcinoma cells. J Biomol Struct Dyn 2021; 40:8261-8273. [PMID: 33847252 DOI: 10.1080/07391102.2021.1910569] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Dihydropyrimidinones have demonstrated different biological activities including anticancer properties. Cytotoxic potential and antiproliferative potential of new dihydropyrimidinone-derived selenoesters (Se-DHPM) compounds were assessed in vitro against the breast adenocarcinoma cells (MCF-7). Among the eight Se-DHPM compounds tested just 49A and 49F were the most cytotoxic for MCF-7 and the most selective for the non-tumor strain (McCoy) and reduced cell viability in a time- and concentration-dependent manner. Compounds 49A and 49F increased the rate of cell death due to apoptosis and necrosis comparatively to the control, however only the 49F showed antiproliferative potential, reducing the number of colonies formed. In the molecular assay 49A interacts with CT-DNA and caused hyperchromism while 49F caused a hypochromic effect. The intercalation test revealed that the two compounds caused destabilization in the CT-DNA molecule. This effect was evidenced by the loss of fluorescence when the compounds competed and caused the displacement of propidium iodide. Simulations (docking and molecular dynamics) using B-DNA brought a greater understanding of ligand-B-DNA interactions. Furthermore, they predicted that the compounds act as minor groove ligands that are stabilized through hydrogen bonds and hydrophobic interactions. However, the form of interaction foreseen for 49A was more energetically favorable and had more stable hydrogen bonds during the simulation time. Despite some violations foreseen in the ADMET for 49F, the set of other results point to this Se-DHPM as a promising leader compound with anti-tumor potential for breast cancer.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Jean C Benassi
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Flavio A R Barbosa
- Department of Chemistry, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Graziâni Candiotto
- Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Danilo Wilhelm Filho
- Departament of Ecology and Zoology, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Antônio L Braga
- Department of Chemistry, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Rozangela C Pedrosa
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
20
|
Lyon PC, Suomi V, Jakeman P, Campo L, Coussios C, Carlisle R. Quantifying cell death induced by doxorubicin, hyperthermia or HIFU ablation with flow cytometry. Sci Rep 2021; 11:4404. [PMID: 33623089 PMCID: PMC7902827 DOI: 10.1038/s41598-021-83845-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/04/2021] [Indexed: 12/31/2022] Open
Abstract
Triggered release and targeted drug delivery of potent anti-cancer agents using hyperthermia-mediated focused-ultrasound (FUS) is gaining momentum in the clinical setting. In early phase studies, tissue biopsy samples may be harvested to assess drug delivery efficacy and demonstrate lack of instantaneous cell death due to FUS exposure. We present an optimised tissue cell recovery method and a cell viability assay, compatible with intra-cellular doxorubicin. Flow cytometry was used to determine levels of cell death with suspensions comprised of: (i) HT29 cell line exposed to hyperthermia (30 min at 47 °C) and/or doxorubicin, or ex-vivo bovine liver tissue exposed to (ii) hyperthermia (up to 2 h at 45 °C), or (iii) ablative high intensity FUS (HIFU). Flow cytometric analysis revealed maximal cell death in HT29 receiving both heat and doxorubicin insults and increases in both cell granularity (p < 0.01) and cell death (p < 0.01) in cells recovered from ex-vivo liver tissue exposed to hyperthermia and high pressures of HIFU (8.2 MPa peak-to-peak free-field at 1 MHz) relative to controls. Ex-vivo results were validated with microscopy using pan-cytokeratin stain. This rapid, sensitive and highly quantitative cell-viability method is applicable to the small masses of liver tissue typically recovered from a standard core biopsy (5-20 mg) and may be applied to tissues of other histological origins including immunostaining.
Collapse
Affiliation(s)
- Paul Christopher Lyon
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK.
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK.
| | - Visa Suomi
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Philip Jakeman
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Leticia Campo
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Constantin Coussios
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Robert Carlisle
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| |
Collapse
|
21
|
Anthracycline-induced cardiomyopathy: cellular and molecular mechanisms. Clin Sci (Lond) 2021; 134:1859-1885. [PMID: 32677679 DOI: 10.1042/cs20190653] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 02/06/2023]
Abstract
Despite the known risk of cardiotoxicity, anthracyclines are widely prescribed chemotherapeutic agents. They are broadly characterized as being a robust effector of cellular apoptosis in rapidly proliferating cells through its actions in the nucleus and formation of reactive oxygen species (ROS). And, despite the early use of dexrazoxane, no effective treatment strategy has emerged to prevent the development of cardiomyopathy, despite decades of study, suggesting that much more insight into the underlying mechanism of the development of cardiomyopathy is needed. In this review, we detail the specific intracellular activities of anthracyclines, from the cell membrane to the sarcoplasmic reticulum, and highlight potential therapeutic windows that represent the forefront of research into the underlying causes of anthracycline-induced cardiomyopathy.
Collapse
|
22
|
Tavener AM, Phelps MC, Daniels RL. Anthracycline-induced cytotoxicity in the GL261 glioma model system. Mol Biol Rep 2021; 48:1017-1023. [PMID: 33387196 PMCID: PMC7884566 DOI: 10.1007/s11033-020-06109-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 12/18/2020] [Indexed: 12/25/2022]
Abstract
Glioblastoma (GBM) is a lethal astrocyte-derived tumor that is currently treated with a multi-modal approach of surgical resection, radiotherapy, and temozolomide-based chemotherapy. Alternatives to current therapies are urgently needed as its prognosis remains poor. Anthracyclines are a class of compounds that show great potential as GBM chemotherapeutic agents and are widely used to treat solid tumors outside the central nervous system. Here we investigate the cytotoxic effects of doxorubicin and other anthracyclines on GL261 glioma tumor cells in anticipation of novel anthracycline-based CNS therapies. Three methods were used to quantify dose-dependent effects of anthracyclines on adherent GL261 tumor cells, a murine cell-based model of GBM. MTT assays quantified anthracycline effects on cell viability, comet assays examined doxorubicin genotoxicity, and flow cytometry with Annexin V/PI staining characterized doxorubicin-induced apoptosis and necrosis. Dose-dependent reductions in GL261 cell viability were found in cells treated with doxorubicin (EC50 = 4.9 μM), epirubicin (EC50 = 5.9 μM), and idarubicin (EC50 = 4.4 μM). Comet assays showed DNA damage following doxorubicin treatments, peaking at concentrations of 1.0 μM and declining after 25 μM. Lastly, flow cytometric analysis of doxorubicin-treated cells showed dose-dependent induction of apoptosis (EC50 = 5.2 μM). Together, these results characterized the cytotoxic effects of anthracyclines on GL261 glioma cells. We found dose-dependent apoptotic induction; however at high concentrations we find that cell death is likely necrotic. Our results support the continued exploration of anthracyclines as compounds with significant potential for improved GBM treatments.
Collapse
Affiliation(s)
- Amber M Tavener
- Department of Biology, The College of Idaho, Caldwell, ID, 83605, USA
| | - Megan C Phelps
- Department of Biology, The College of Idaho, Caldwell, ID, 83605, USA
| | - Richard L Daniels
- Department of Biology, The College of Idaho, Caldwell, ID, 83605, USA.
| |
Collapse
|
23
|
Asem H, Zheng W, Nilsson F, Zhang Y, Hedenqvist MS, Hassan M, Malmström E. Functional Nanocarriers for Drug Delivery by Surface Engineering of Polymeric Nanoparticle Post-Polymerization-Induced Self-Assembly. ACS APPLIED BIO MATERIALS 2020. [DOI: 10.1021/acsabm.0c01552] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Heba Asem
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Fibre and Polymer Technology, Division of Coating Technology, KTH Royal Institute of Technology, Stockholm SE-100 44, Sweden
| | - Wenyi Zheng
- Division of Experimental Cancer Medicine (ECM), Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm SE-141 86, Sweden
| | - Fritjof Nilsson
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Fibre and Polymer Technology, Division of Coating Technology, KTH Royal Institute of Technology, Stockholm SE-100 44, Sweden
- Division of Polymeric Materials, KTH Royal Institute of Technology, Stockholm SE-100 44, Sweden
| | - Yuning Zhang
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Fibre and Polymer Technology, Division of Coating Technology, KTH Royal Institute of Technology, Stockholm SE-100 44, Sweden
| | - Mikael S. Hedenqvist
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Fibre and Polymer Technology, Division of Coating Technology, KTH Royal Institute of Technology, Stockholm SE-100 44, Sweden
- Division of Polymeric Materials, KTH Royal Institute of Technology, Stockholm SE-100 44, Sweden
| | - Moustapha Hassan
- Division of Experimental Cancer Medicine (ECM), Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm SE-141 86, Sweden
- Clinical Research Centrum, Department of Stem Cell Transplantation (CAST), Karolinska University Hospital-Huddinge, Stockholm SE-141 86, Sweden
| | - Eva Malmström
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Fibre and Polymer Technology, Division of Coating Technology, KTH Royal Institute of Technology, Stockholm SE-100 44, Sweden
| |
Collapse
|
24
|
Kim D, Park S, Yoo H, Park S, Kim J, Yum K, Kim K, Kim H. Overcoming anticancer resistance by photodynamic therapy-related efflux pump deactivation and ultrasound-mediated improved drug delivery efficiency. NANO CONVERGENCE 2020; 7:30. [PMID: 32897469 PMCID: PMC7479087 DOI: 10.1186/s40580-020-00241-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/29/2020] [Indexed: 05/26/2023]
Abstract
One of the major obstacles to successful chemotherapy is multi-drug resistance (MDR). A multi-drug resistant cancerous cell abnormally overexpresses membrane transporters that pump anticancer drugs out of the cell, resulting in low anticancer drug delivery efficiency. To overcome the limitation, many attempts have been performed to inhibit the abilities of efflux receptors chemically or genetically or to increase the delivery efficiency of anticancer drugs. However, the results have not yet been satisfactory. In this study, we developed nanoparticle-microbubble complexes (DOX-NPs/Ce6-MBs) by conjugating doxorubicin loaded human serum albumin nanoparticles (DOX-NPs) onto the surface of Chlorin e6 encapsulated microbubbles (Ce6-MBs) in order to maximize anticancer efficiency by overcoming MDR. Under the ultrasound irradiation, DOX-NPs and Ce6 encapsulating self-assembled liposomes or micelles were effectively delivered into the cells due to the sonoporation effect caused by the microbubble cavitation. At the same time, reactive oxygen (ROS) generated from intracellularly delivered Ce6 by laser irradiation arrested the activity of ABCG2 efflux receptor overexpressed in doxorubicin-resistant breast cancer cells (MCF-7/ADR), resulting in increased the chemotherapy efficacy. In addition, the total number of side population cells that exhibit the properties of cancer stem-like cells were also reduced by the combination of photodynamic therapy and chemotherapy. In conclusion, DOX-NPs/Ce6-MBs will provide a platform for simultaneously overcoming MDR and increasing drug delivery and therefore, treatment efficiency, under ultrasound irradiation.
Collapse
Affiliation(s)
- Doyeon Kim
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Suhyun Park
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Hongkeun Yoo
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Suhyeon Park
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Jeewon Kim
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Kyuhee Yum
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Kwangmeyung Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hyuncheol Kim
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107, Republic of Korea.
- Department of Biomedical Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107, Republic of Korea.
| |
Collapse
|
25
|
Schleyer KA, Datko BD, Burnside B, Cui C, Ma X, Grey JK, Cui L. Responsive Fluorophore Aggregation Provides Spectral Contrast for Fluorescence Lifetime Imaging. Chembiochem 2020; 21:2196-2204. [PMID: 32180309 PMCID: PMC8247454 DOI: 10.1002/cbic.202000056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/03/2020] [Indexed: 11/06/2022]
Abstract
Fluorophores experience altered emission lifetimes when incorporated into and liberated from macromolecules or molecular aggregates; this trend suggests the potential for a fluorescent, responsive probe capable of undergoing self-assembly and aggregation and consequently altering the lifetime of its fluorescent moiety to provide contrast between the active and inactive probes. We developed a cyanobenzothioazole-fluorescein conjugate (1), and spectroscopically examined the lifetime changes caused by its reduction-induced aggregation in vitro. A decrease in lifetime was observed for compound 1 in a buffered system activated by the biological reducing agent glutathione, thus suggesting a possible approach for designing responsive self-aggregating lifetime imaging probes.
Collapse
Affiliation(s)
- Kelton A Schleyer
- Department of Chemistry and Chemical Biology, UNM Comprehensive Cancer Center, University of New Mexico, 300 Terrace St. NE, Albuquerque, NM 87131, USA
- Department of Medicinal Chemistry, College of Pharmacy, UF Health Science Center, UF Health Cancer Center University of Florida, 1345 Center Dr., Gainesville, FL 32610, USA
| | - Benjamin D Datko
- Department of Chemistry and Chemical Biology, UNM Comprehensive Cancer Center, University of New Mexico, 300 Terrace St. NE, Albuquerque, NM 87131, USA
- Center for High Technology Materials, University of New Mexico, MSC04 2710, 1313 Goddard St. SE, Albuquerque, NM 87106, USA
| | - Brandon Burnside
- Department of Chemistry and Chemical Biology, UNM Comprehensive Cancer Center, University of New Mexico, 300 Terrace St. NE, Albuquerque, NM 87131, USA
- Center for High Technology Materials, University of New Mexico, MSC04 2710, 1313 Goddard St. SE, Albuquerque, NM 87106, USA
| | - Chao Cui
- Department of Chemistry and Chemical Biology, UNM Comprehensive Cancer Center, University of New Mexico, 300 Terrace St. NE, Albuquerque, NM 87131, USA
- Department of Medicinal Chemistry, College of Pharmacy, UF Health Science Center, UF Health Cancer Center University of Florida, 1345 Center Dr., Gainesville, FL 32610, USA
| | - Xiaowei Ma
- Department of Chemistry and Chemical Biology, UNM Comprehensive Cancer Center, University of New Mexico, 300 Terrace St. NE, Albuquerque, NM 87131, USA
- Department of Medicinal Chemistry, College of Pharmacy, UF Health Science Center, UF Health Cancer Center University of Florida, 1345 Center Dr., Gainesville, FL 32610, USA
| | - John K Grey
- Department of Chemistry and Chemical Biology, UNM Comprehensive Cancer Center, University of New Mexico, 300 Terrace St. NE, Albuquerque, NM 87131, USA
- Center for High Technology Materials, University of New Mexico, MSC04 2710, 1313 Goddard St. SE, Albuquerque, NM 87106, USA
| | - Lina Cui
- Department of Chemistry and Chemical Biology, UNM Comprehensive Cancer Center, University of New Mexico, 300 Terrace St. NE, Albuquerque, NM 87131, USA
- Department of Medicinal Chemistry, College of Pharmacy, UF Health Science Center, UF Health Cancer Center University of Florida, 1345 Center Dr., Gainesville, FL 32610, USA
| |
Collapse
|
26
|
Wu SY, Chou HY, Tsai HC, Anbazhagan R, Yuh CH, Yang JM, Chang YH. Amino acid-modified PAMAM dendritic nanocarriers as effective chemotherapeutic drug vehicles in cancer treatment: a study using zebrafish as a cancer model. RSC Adv 2020; 10:20682-20690. [PMID: 35517745 PMCID: PMC9054295 DOI: 10.1039/d0ra01589j] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 04/27/2020] [Indexed: 12/22/2022] Open
Abstract
The use of nanomaterials for drug delivery offers many advantages including the targeted delivery of drugs and their controlled release. Nonetheless, entry into the target cells remains a challenge for many nanomaterials used for drug delivery. Moreover, cellular uptake limits the therapeutic efficiency of many anticancer drugs. An important goal is to increase the specific accumulation of these nanoparticles (NPs) at the desired cancerous tissues. Notably, cancer cells show a high demand for some amino acids and we have used this knowledge to develop novel carrier systems. In this study, drug carriers were produced by the conjugation of multiple amino acids such as l-histidine (H) and l-cysteine (C) or single amino acids such as only H with the G4.5 dendrimers (G) to produce GHC aggregates and GH NP carriers, respectively. Doxorubicin was loaded into the G4.5, GH, and GHC dendrimers (G/DOX, GH/DOX and GHC/DOX, respectively) and the release mechanism was demonstrated at pH 7.4 and pH 5.0. GH/DOX and GHC/DOX showed better stability under physiological conditions than the dendrimer alone (G/DOX). GH/DOX and GHC/DOX exhibited higher inhibition of HeLa cell proliferation in in vitro and in vivo studies in zebrafish, confirming the early release of DOX by disrupting the endosomal membrane and triggering the destabilization of carriers at a lower pH of 5.0.
Collapse
Affiliation(s)
- Szu-Yuan Wu
- Department of Food Nutrition and Health Biotechnology, College of Medical and Health Science, Asia University Taichung Taiwan
- Division of Radiation Oncology, Lo-Hsu Medical Foundation, LotungPoh-Ai Hospital Yilan Taiwan
- Big Data Center, Lo-Hsu Medical Foundation, LotungPoh-Ai Hospital Yilan 265 Taiwan
- Department of Healthcare Administration, College of Medical and Health Science, Asia University Taichung 41354 Taiwan
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University Taipei 110 Taiwan
| | - Hsiao-Ying Chou
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology Taipei Taiwan +886-2-27303625 +886-984252998
- Advanced Membrane Materials Center, National Taiwan University of Science and Technology Taipei Taiwan
| | - Hsieh-Chih Tsai
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology Taipei Taiwan +886-2-27303625 +886-984252998
- Advanced Membrane Materials Center, National Taiwan University of Science and Technology Taipei Taiwan
| | - Rajeshkumar Anbazhagan
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology Taipei Taiwan +886-2-27303625 +886-984252998
- Advanced Membrane Materials Center, National Taiwan University of Science and Technology Taipei Taiwan
| | - Chiou-Hwa Yuh
- Institute of Molecular and Genomic Medicine, National Health Research Institutes Zhunan Miaoli Taiwan
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University Hsinchu Taiwan
- Department of Biological Science and Technology, National Chiao Tung University Hsinchu Taiwan
| | - Jen Ming Yang
- Department of Chemical and Materials Engineering, Chang Gung University Tao-Yuan Taiwan +886-3-2118800-529
- Department of General Dentistry, Chang Gung Memorial Hospital Tao-Yuan, 333 Taiwan
| | - Yen-Hsiang Chang
- Department of General Dentistry, Chang Gung Memorial Hospital Tao-Yuan, 333 Taiwan
| |
Collapse
|
27
|
Peng Z, Nie K, Song Y, Liu H, Zhou Y, Yuan Y, Chen D, Peng X, Yan W, Song J, Qu J. Monitoring the Cellular Delivery of Doxorubicin-Cu Complexes in Cells by Fluorescence Lifetime Imaging Microscopy. J Phys Chem A 2020; 124:4235-4240. [PMID: 32364735 DOI: 10.1021/acs.jpca.0c00182] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the prodrug research field, information obtained from traditional end point biochemical assays in drug effect studies could provide neither the dynamic processes nor heterogeneous responses of individual cells. In situ imaging microscopy techniques, especially fluorescence lifetime imaging microscopy (FLIM), could fulfill these requirements. In this work, we used FLIM techniques to observe the entry and release of doxorubicin (Dox)-Cu complexes in live KYSE150 cells. The Dox-Cu complex has weaker fluorescence signals but similar lifetime values as compared to the raw Dox, whose fluorescence could be released by the addition of biothiol compound (such as glutathione). The cell viability results indicated that the Dox-Cu compound has a satisfactory killing effect on KYSE150 cells. The FLIM data showed that free doxorubicin was released from Dox-Cu complexes in cytoplasm of KYSE150 cells and then accumulated in the nucleus. After 90 min administration, the fluorescence lifetime signals reached 1.21 and 1.46 ns in the cytoplasm and nucleus, respectively, reflecting the transformation and transportation of Dox-Cu complexes. In conclusion, this work provides a satisfactory example for the research of prodrug monitored by FLIM techniques, expanding the useful applications of FLIM technique in drug development.
Collapse
Affiliation(s)
- Zheng Peng
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| | - Kaixuan Nie
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| | - Yiwan Song
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| | - Hao Liu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| | - Yingxin Zhou
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| | - Yufeng Yuan
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| | - Danni Chen
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| | - Xiao Peng
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| | - Wei Yan
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| | - Jun Song
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| |
Collapse
|
28
|
Engström J, Asem H, Brismar H, Zhang Y, Malkoch M, Malmström E. In Situ Encapsulation of Nile Red or Doxorubicin during RAFT‐Mediated Emulsion Polymerization via Polymerization‐Induced Self‐Assembly for Biomedical Applications. MACROMOL CHEM PHYS 2020. [DOI: 10.1002/macp.201900443] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Joakim Engström
- KTH Royal Institute of TechnologySchool of Engineering Sciences in ChemistryBiotechnology and HealthDepartment of Fibre and Polymer TechnologyDivision of Coating Technology Stockholm SE‐10044 Sweden
- Wallenberg Wood Science Centre Stockholm SE‐10044 Sweden
| | - Heba Asem
- KTH Royal Institute of TechnologySchool of Engineering Sciences in ChemistryBiotechnology and HealthDepartment of Fibre and Polymer TechnologyDivision of Coating Technology Stockholm SE‐10044 Sweden
| | - Hjalmar Brismar
- Department of Applied PhysicsScience for Life Laboratory Stockholm SE‐17121 Sweden
| | - Yuning Zhang
- KTH Royal Institute of TechnologySchool of Engineering Sciences in ChemistryBiotechnology and HealthDepartment of Fibre and Polymer TechnologyDivision of Coating Technology Stockholm SE‐10044 Sweden
| | - Michael Malkoch
- KTH Royal Institute of TechnologySchool of Engineering Sciences in ChemistryBiotechnology and HealthDepartment of Fibre and Polymer TechnologyDivision of Coating Technology Stockholm SE‐10044 Sweden
| | - Eva Malmström
- KTH Royal Institute of TechnologySchool of Engineering Sciences in ChemistryBiotechnology and HealthDepartment of Fibre and Polymer TechnologyDivision of Coating Technology Stockholm SE‐10044 Sweden
| |
Collapse
|
29
|
Rosqvist E, Niemelä E, Frisk J, Öblom H, Koppolu R, Abdelkader H, Soto Véliz D, Mennillo M, Venu AP, Ihalainen P, Aubert M, Sandler N, Wilén CE, Toivakka M, Eriksson JE, Österbacka R, Peltonen J. A low-cost paper-based platform for fast and reliable screening of cellular interactions with materials. J Mater Chem B 2020; 8:1146-1156. [PMID: 32011620 DOI: 10.1039/c9tb01958h] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
A paper-based platform was developed and tested for studies on basic cell culture, material biocompatibility, and activity of pharmaceuticals in order to provide a reliable, robust and low-cost cell study platform. It is based upon a paper or paperboard support, with a nanostructured latex coating to provide an enhanced cell growth and sufficient barrier properties. Wetting is limited to regions of interest using a flexographically printed hydrophobic polydimethylsiloxane layer with circular non-print areas. The nanostructured coating can be substituted for another coating of interest, or the regions of interest functionalized with a material to be studied. The platform is fully up-scalable, being produced with roll-to-roll rod coating, flexographic and inkjet printing methods. Results show that the platform efficiency is comparable to multi-well plates in colorimetric assays in three separate studies: a cell culture study, a biocompatibility study, and a drug screening study. The color intensity is quantified by using a common office scanner or an imaging device and the data is analyzed by a custom computer software without the need for expensive screening or analysis equipment.
Collapse
Affiliation(s)
- E Rosqvist
- Laboratory of Physical Chemistry, Center for Functional Materials, Åbo Akademi University, Porthansgatan 3-5, 20500 Åbo, Finland.
| | - E Niemelä
- Laboratory of Cell Biology, Center for Functional Materials, Åbo Akademi University, Bio City, Artillerigatan 6B, 20521 Åbo, Finland and Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Åbo, Finland
| | - J Frisk
- Laboratory of Physics, Center for Functional Materials, Åbo Akademi University, Porthansgatan 3-5, 20500 Åbo, Finland
| | - H Öblom
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Artillerigatan 6A, 20520 Åbo, Finland
| | - R Koppolu
- Laboratory of Paper Coating, Center for Functional Materials, Åbo Akademi University, Porthansgatan 3-5, 20500 Åbo, Finland
| | - H Abdelkader
- Laboratory of Cell Biology, Center for Functional Materials, Åbo Akademi University, Bio City, Artillerigatan 6B, 20521 Åbo, Finland and Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Åbo, Finland
| | - D Soto Véliz
- Laboratory of Paper Coating, Center for Functional Materials, Åbo Akademi University, Porthansgatan 3-5, 20500 Åbo, Finland
| | - M Mennillo
- Laboratory of Polymer Technology, Center for Functional Materials, Åbo Akademi University, Biskopsgatan 3-5, 20500 Åbo, Finland
| | - A P Venu
- Laboratory of Cell Biology, Center for Functional Materials, Åbo Akademi University, Bio City, Artillerigatan 6B, 20521 Åbo, Finland and Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Åbo, Finland
| | - P Ihalainen
- Laboratory of Physical Chemistry, Center for Functional Materials, Åbo Akademi University, Porthansgatan 3-5, 20500 Åbo, Finland.
| | - M Aubert
- Laboratory of Polymer Technology, Center for Functional Materials, Åbo Akademi University, Biskopsgatan 3-5, 20500 Åbo, Finland
| | - N Sandler
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Artillerigatan 6A, 20520 Åbo, Finland
| | - C-E Wilén
- Laboratory of Polymer Technology, Center for Functional Materials, Åbo Akademi University, Biskopsgatan 3-5, 20500 Åbo, Finland
| | - M Toivakka
- Laboratory of Paper Coating, Center for Functional Materials, Åbo Akademi University, Porthansgatan 3-5, 20500 Åbo, Finland
| | - J E Eriksson
- Laboratory of Cell Biology, Center for Functional Materials, Åbo Akademi University, Bio City, Artillerigatan 6B, 20521 Åbo, Finland and Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Åbo, Finland
| | - R Österbacka
- Laboratory of Physics, Center for Functional Materials, Åbo Akademi University, Porthansgatan 3-5, 20500 Åbo, Finland
| | - J Peltonen
- Laboratory of Physical Chemistry, Center for Functional Materials, Åbo Akademi University, Porthansgatan 3-5, 20500 Åbo, Finland.
| |
Collapse
|
30
|
Xie X, Chen Y, Chen Z, Feng Y, Wang J, Li T, Li S, Qin X, Wu C, Zheng C, Zhu J, You F, Liu Y, Yang H. Polymeric Hybrid Nanomicelles for Cancer Theranostics: An Efficient and Precise Anticancer Strategy for the Codelivery of Doxorubicin/miR-34a and Magnetic Resonance Imaging. ACS APPLIED MATERIALS & INTERFACES 2019; 11:43865-43878. [PMID: 31684723 DOI: 10.1021/acsami.9b14908] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
To realize precise tumor therapy, a versatile oncotherapy nanoplatform integrating both diagnostic and therapeutic functions is necessary. Herein, we fabricated a hybrid micelle (HM) utilizing two amphiphilic diblock copolymers, polyethylenimine-polycaprolactone (PEI-PCL) and diethylenetriaminepentaacetic acid gadolinium(III) (Gd-DTPA)-conjugated polyethyleneglycol-polycaprolactone (Gd-PEG-PCL), to codeliver the small-molecule chemotherapy drugs doxorubicin (Dox) and microRNA-34a (miR-34a), denoted as Gd-HM-Dox/34a. Conjugating Gd-DTPA on the surface of hybrid micelles, leading the relaxation rate of Gd-DTPA increased more than 1.4 times (13.6 mM-1 S-1). Furthermore, hybrid micelles enhanced the ability of miR-34a to escape from lysosomes/endosomes and Dox release to the nucleus. In addition, the released miR-34a subsequently downregulates Bcl-2, cyclin D1, CDK6, and Bax expression and inhibits proliferation and migration of MDA-MB-231 breast cancer cells. Moreover, the suitable micelle size improved the penetration of Dox into three-dimensional (3D) multicellular spheroids compared with Gd-HM-Dox and Free Dox, generating efficient cell killing in the 3D multicellular spheroids. Furthermore, the Gd-HM-Dox/34a exhibited augmented accumulation in the tumor tissue, which improved the magnetic resonance (MR) imaging contrast of solid tumors and enhanced the combined efficiency of chemotherapeutic drugs Dox and therapeutic gene miR-34a in suppressing tumor growth on MDA-MB-231 tumor-bearing mice. Therefore, we established a hybrid micelle to offer a promising theranostic approach that inhibits tumor growth and enhances MR imaging.
Collapse
Affiliation(s)
- Xiaoxue Xie
- Department of Biophysics, School of Life Science and Technology , University of Electronic Science and Technology of China , Chengdu 610054 , Sichuan , P.R. China
| | - Yu Chen
- Department of Biophysics, School of Life Science and Technology , University of Electronic Science and Technology of China , Chengdu 610054 , Sichuan , P.R. China
| | - Zhongyuan Chen
- Department of Biophysics, School of Life Science and Technology , University of Electronic Science and Technology of China , Chengdu 610054 , Sichuan , P.R. China
| | - Yi Feng
- Department of Biophysics, School of Life Science and Technology , University of Electronic Science and Technology of China , Chengdu 610054 , Sichuan , P.R. China
| | - Jing Wang
- Department of Biophysics, School of Life Science and Technology , University of Electronic Science and Technology of China , Chengdu 610054 , Sichuan , P.R. China
| | - Tingting Li
- Department of Biophysics, School of Life Science and Technology , University of Electronic Science and Technology of China , Chengdu 610054 , Sichuan , P.R. China
| | - Shun Li
- Department of Biophysics, School of Life Science and Technology , University of Electronic Science and Technology of China , Chengdu 610054 , Sichuan , P.R. China
- Center for Information in Biology , University of Electronic Science and Technology of China , Chengdu 610054 , Sichuan , P.R. China
| | - Xiang Qin
- Department of Biophysics, School of Life Science and Technology , University of Electronic Science and Technology of China , Chengdu 610054 , Sichuan , P.R. China
- Center for Information in Biology , University of Electronic Science and Technology of China , Chengdu 610054 , Sichuan , P.R. China
| | - Chunhui Wu
- Department of Biophysics, School of Life Science and Technology , University of Electronic Science and Technology of China , Chengdu 610054 , Sichuan , P.R. China
- Center for Information in Biology , University of Electronic Science and Technology of China , Chengdu 610054 , Sichuan , P.R. China
| | - Chuan Zheng
- Hospital of Chengdu University of Traditional Chinese Medicine , No. 39 Shi-er-qiao Road , Chengdu 610072 , Sichuan , P.R. China
| | - Jie Zhu
- Hospital of Chengdu University of Traditional Chinese Medicine , No. 39 Shi-er-qiao Road , Chengdu 610072 , Sichuan , P.R. China
| | - Fengming You
- Hospital of Chengdu University of Traditional Chinese Medicine , No. 39 Shi-er-qiao Road , Chengdu 610072 , Sichuan , P.R. China
| | - Yiyao Liu
- Department of Biophysics, School of Life Science and Technology , University of Electronic Science and Technology of China , Chengdu 610054 , Sichuan , P.R. China
- Center for Information in Biology , University of Electronic Science and Technology of China , Chengdu 610054 , Sichuan , P.R. China
- Hospital of Chengdu University of Traditional Chinese Medicine , No. 39 Shi-er-qiao Road , Chengdu 610072 , Sichuan , P.R. China
| | - Hong Yang
- Department of Biophysics, School of Life Science and Technology , University of Electronic Science and Technology of China , Chengdu 610054 , Sichuan , P.R. China
- Center for Information in Biology , University of Electronic Science and Technology of China , Chengdu 610054 , Sichuan , P.R. China
| |
Collapse
|
31
|
Tjandra KC, McCarthy N, Yang L, Laos AJ, Sharbeen G, Phillips PA, Forgham H, Sagnella SM, Whan RM, Kavallaris M, Thordarson P, McCarroll JA. Identification of Novel Medulloblastoma Cell-Targeting Peptides for Use in Selective Chemotherapy Drug Delivery. J Med Chem 2019; 63:2181-2193. [DOI: 10.1021/acs.jmedchem.9b00851] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Kristel C. Tjandra
- Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, UNSW Sydney, Sydney, NSW 2052, Australia
- School of Chemistry, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Nigel McCarthy
- Tumour Biology & Targeting Program, Children’s Cancer Institute, UNSW Sydney, Lowy Cancer Research Centre, Sydney, NSW 2031, Australia
| | - Lu Yang
- Tumour Biology & Targeting Program, Children’s Cancer Institute, UNSW Sydney, Lowy Cancer Research Centre, Sydney, NSW 2031, Australia
| | - Alistair J. Laos
- Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, UNSW Sydney, Sydney, NSW 2052, Australia
- School of Chemistry, UNSW Sydney, Sydney, NSW 2052, Australia
| | - George Sharbeen
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, and School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Phoebe A. Phillips
- Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, UNSW Sydney, Sydney, NSW 2052, Australia
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, and School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Helen Forgham
- Tumour Biology & Targeting Program, Children’s Cancer Institute, UNSW Sydney, Lowy Cancer Research Centre, Sydney, NSW 2031, Australia
- Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, UNSW Sydney, Sydney, NSW 2052, Australia
- School of Women’s and Children’s Health, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Sharon M. Sagnella
- Tumour Biology & Targeting Program, Children’s Cancer Institute, UNSW Sydney, Lowy Cancer Research Centre, Sydney, NSW 2031, Australia
| | - Renee M. Whan
- Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, UNSW Sydney, Sydney, NSW 2052, Australia
- Biomedical Imaging Facility Mark Wainwright Analytical Centre, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Maria Kavallaris
- Tumour Biology & Targeting Program, Children’s Cancer Institute, UNSW Sydney, Lowy Cancer Research Centre, Sydney, NSW 2031, Australia
- Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, UNSW Sydney, Sydney, NSW 2052, Australia
- School of Women’s and Children’s Health, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Pall Thordarson
- Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, UNSW Sydney, Sydney, NSW 2052, Australia
- School of Chemistry, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Joshua A. McCarroll
- Tumour Biology & Targeting Program, Children’s Cancer Institute, UNSW Sydney, Lowy Cancer Research Centre, Sydney, NSW 2031, Australia
- Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, UNSW Sydney, Sydney, NSW 2052, Australia
- School of Women’s and Children’s Health, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| |
Collapse
|
32
|
Caccuri F, Sommariva M, Marsico S, Giordano F, Zani A, Giacomini A, Fraefel C, Balsari A, Caruso A. Inhibition of DNA Repair Mechanisms and Induction of Apoptosis in Triple Negative Breast Cancer Cells Expressing the Human Herpesvirus 6 U94. Cancers (Basel) 2019; 11:cancers11071006. [PMID: 31323788 PMCID: PMC6679437 DOI: 10.3390/cancers11071006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/01/2019] [Accepted: 07/11/2019] [Indexed: 12/18/2022] Open
Abstract
Triple-negative breast cancer (TNBC) accounts for 15–20% of all breast cancers. In spite of initial good response to chemotherapy, the prognosis of TNBC remains poor and no effective specific targeted therapy is readily available. Recently, we demonstrated the ability of U94, the latency gene of human herpes virus 6 (HHV-6), to interfere with proliferation and with crucial steps of the metastatic cascade by using MDA-MB 231 TNBC breast cancer cell line. U94 expression was also associated with a partial mesenchymal-to-epithelial transition (MET) of cells, which displayed a less aggressive phenotype. In this study, we show the ability of U94 to exert its anticancer activity on three different TNBC cell lines by inhibiting DNA damage repair genes, cell cycle and eventually leading to cell death following activation of the intrinsic apoptotic pathway. Interestingly, we found that U94 acted synergistically with DNA-damaging drugs. Overall, we provide evidence that U94 is able to combat tumor cells with different mechanisms, thus attesting for the great potential of this molecule as a multi-target drug in cancer therapy.
Collapse
Affiliation(s)
- Francesca Caccuri
- Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Michele Sommariva
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan 20133, Italy
| | - Stefania Marsico
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza 87036, Italy
| | - Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza 87036, Italy
| | - Alberto Zani
- Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Arianna Giacomini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Cornel Fraefel
- Institute of Virology, University of Zurich, Zurich 8057, Switzerland
| | - Andrea Balsari
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan 20133, Italy
| | - Arnaldo Caruso
- Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy.
| |
Collapse
|
33
|
Liu S, Hu C, Liu Y, Zhao X, Pang M, Lin J. One‐Pot Synthesis of DOX@Covalent Organic Framework with Enhanced Chemotherapeutic Efficacy. Chemistry 2019; 25:4315-4319. [DOI: 10.1002/chem.201806242] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Indexed: 11/07/2022]
Affiliation(s)
- Sainan Liu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied ChemistryChinese Academy of Sciences Changchun 130022 P. R. China
- University of Science and Technology of China Hefei 230026 P. R. China
| | - Chunling Hu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied ChemistryChinese Academy of Sciences Changchun 130022 P. R. China
- University of Science and Technology of China Hefei 230026 P. R. China
| | - Ying Liu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied ChemistryChinese Academy of Sciences Changchun 130022 P. R. China
| | - Xueyan Zhao
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied ChemistryChinese Academy of Sciences Changchun 130022 P. R. China
| | - Maolin Pang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied ChemistryChinese Academy of Sciences Changchun 130022 P. R. China
- University of Science and Technology of China Hefei 230026 P. R. China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied ChemistryChinese Academy of Sciences Changchun 130022 P. R. China
- University of Science and Technology of China Hefei 230026 P. R. China
| |
Collapse
|
34
|
Paşcalău V, Pall E, Tertis M, Suciu M, Cristea C, Borodi G, Bodoki A, Topală T, Stiufiuc R, Moldovan A, Pavel C, Marinca T, Popa C. In vitro study of BSA gel/polyelectrolite complexes core shell microcapsules encapsulating doxorubicin for antitumoral targeted treatment. INT J POLYM MATER PO 2019. [DOI: 10.1080/00914037.2018.1525724] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Violeta Paşcalău
- Department of Materials Science and Engineering, Technical University of Cluj-Napoca, Cluj-Napoca, Romania
| | - Emoke Pall
- Clinical Department, University of Agricultural Sciences and Veterinary Medicine of Cluj-Napoca, Cluj-Napoca, Romania
| | - Mihaela Tertis
- Department of Analytical Chemistry, “Iuliu Haţieganu” University of Medicine and Pharmacy Cluj-Napoca, Cluj-Napoca, Romania
| | - Maria Suciu
- Electron Microscopy Integrated Laboratory Department, National Institute for Research and Development of Isotopic and Molecular Technologies, Cluj-Napoca, Romania
- Molecular Biology and Biotechnology Department, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Cecilia Cristea
- Department of Analytical Chemistry, “Iuliu Haţieganu” University of Medicine and Pharmacy Cluj-Napoca, Cluj-Napoca, Romania
| | - Gheorghe Borodi
- Molecular and Biomolecular Physics Department, National Institute for Research and Development of Isotopic and Molecular Technologies, Cluj-Napoca, Romania
| | - Andreea Bodoki
- General and Inorganic Chemistry Department, “Iuliu Haţieganu” University of Medicine and Pharmacy Cluj-Napoca, Cluj-Napoca, Romania
| | - Tamara Topală
- General and Inorganic Chemistry Department, “Iuliu Haţieganu” University of Medicine and Pharmacy Cluj-Napoca, Cluj-Napoca, Romania
| | - Rares Stiufiuc
- Nanobioscopy Department, MedFuture Research Center for Advanced Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy Cluj-Napoca, Cluj-Napoca, Romania
- Physics - Biophysics Department, “Iuliu Haţieganu” University of Medicine and Pharmacy Cluj-Napoca, Cluj-Napoca, Romania
| | - Alin Moldovan
- Nanobioscopy Department, MedFuture Research Center for Advanced Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy Cluj-Napoca, Cluj-Napoca, Romania
| | - Codruta Pavel
- Department of Materials Science and Engineering, Technical University of Cluj-Napoca, Cluj-Napoca, Romania
| | - Traian Marinca
- Department of Materials Science and Engineering, Technical University of Cluj-Napoca, Cluj-Napoca, Romania
| | - Catalin Popa
- Department of Materials Science and Engineering, Technical University of Cluj-Napoca, Cluj-Napoca, Romania
| |
Collapse
|
35
|
Fernandes E, Soares TB, Gonçalves H, Lúcio M. Spectroscopic Studies as a Toolbox for Biophysical and Chemical Characterization of Lipid-Based Nanotherapeutics. Front Chem 2018; 6:323. [PMID: 30109226 PMCID: PMC6080416 DOI: 10.3389/fchem.2018.00323] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/11/2018] [Indexed: 01/22/2023] Open
Abstract
The goal of this study is to provide tools to minimize trial-and-error in the development of novel lipid-based nanotherapeutics, in favor of a rational design process. For this purpose, we present case-study examples of biophysical assays that help addressing issues of lipid-based nanotherapeutics' profiling and assist in the design of lipid nanocarriers for therapeutic usage. The assays presented are rooted in spectroscopic methods (steady-state and time-resolved fluorescence; UV-Vis derivative spectroscopy; fluorescence anisotropy and fluorescence lifetime image microscopy) and allow accessing physical-chemical interactions between drugs and lipid nanocarriers, as well as studying interactions between lipid-based nanotherapeutics and membranes and/or proteins, as this is a key factor in predicting their therapeutic and off target effects. Derivative spectroscopy revealed Naproxen's high distribution (LogD ≈ 3) in different lipid-based nanocarriers (micelles and unilamellar or multilamellar vesicles) confirming the adequacy of such systems for encapsulating this anti-inflammatory drug. Fluorescence quenching studies revealed that the anti-inflammatory drugs Acemetacin and Indomethacin can reach an inner location at the lipid nanocarrier while being anchored with its carboxylic moiety at the polar headgroup. The least observed quenching effect suggested that Tolmetin is probably located at the polar headgroup region of the lipid nanocarriers and this superficial location may translate in a fast drug release from the nanocarriers. Fluorescent anisotropy measurements indicated that the drugs deeply buried within the lipid nanocarrier where the ones that had a greater fluidizing effect which can also translate in a faster drug release. The drug binding strength to serum albumin was also compared for a free drug (Clonixin) or for the same drug after encapsulation in a lipid nanocarrier DSPC:DODAP (2:1). Under both conditions there is a strong binding to serum albumin, at one binding site, suggesting the need to produce a stealth nanosystem. Finally the cellular uptake of lipid nanocarriers loaded with Daunorubicin was investigated in cancer cells using fluorescence lifetime imaging microscopy. From the images obtained it was possible to conclude that even at short incubation times (15 min) there was a distribution of the drug in the cytoplasm, whereas for longer incubation periods (4 h) the drug has reached the nucleus.
Collapse
Affiliation(s)
- Eduarda Fernandes
- Department of Physics, Centre of Physics of University of Minho and Porto, University of Minho, Braga, Portugal
| | - Telma B Soares
- Department of Physics, Centre of Physics of University of Minho and Porto, University of Minho, Braga, Portugal
| | - Hugo Gonçalves
- Department of Physics, Centre of Physics of University of Minho and Porto, University of Minho, Braga, Portugal
| | - Marlene Lúcio
- Department of Physics, Centre of Physics of University of Minho and Porto, University of Minho, Braga, Portugal
| |
Collapse
|
36
|
Sommer AK, Hermawan A, Ljepoja B, Fröhlich T, Arnold GJ, Wagner E, Roidl A. A proteomic analysis of chemoresistance development via sequential treatment with doxorubicin reveals novel players in MCF‑7 breast cancer cells. Int J Mol Med 2018; 42:1987-1997. [PMID: 30066829 PMCID: PMC6108857 DOI: 10.3892/ijmm.2018.3781] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 06/21/2018] [Indexed: 12/23/2022] Open
Abstract
Breast cancer exhibits the highest incidence of all cancer types and is the 2nd leading cause of cancer mortality in women. Up to 82% of breast cancer patients receive a chemotherapy-containing treatment regimen. However, numerous breast tumors recur within 10 years following an initial response and are frequently resistant to previous therapeutic agents. Thus, to analyze the crucial factors, and whether the development of resistance in tumor cells follows certain patterns, is of great importance. In the present study, the clinical treatment schedule of the frequently used chemotherapeutic drug doxorubicin was applied in an in vitro model, the Molecular Evolution Assay (MEA), leading to resistance formation. By investigating the alterations in protein expression in MCF-7 breast cancer cells with three biological replicates, it was observed that the development of resistance to doxorubicin is a multi-directed process. The number and composition of the differentially expressed proteins varied, in addition to the pathways involved in chemoresistance, leading to only a small number of proteins and pathways being commonly regulated in all the MEAs. The proteins 60S ribosomal export protein NMD3 and 4F2 cell-surface antigen heavy chain (SLC3A2) were identified to be the most promising differentially expressed targets; the gene ontology term 'apoptotic signaling pathway' was reduced and 'cell redox homeostasis' was upregulated. Based on the present findings in vitro, it may be hypothesized that the development of resistance in patients is an even more complex process, emphasizing the need for further investigations of resistance development in the clinic to eventually improve patient outcomes.
Collapse
Affiliation(s)
- Ann-Katrin Sommer
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig‑Maximilians‑Universität München, D‑81377 Munich, Germany
| | - Adam Hermawan
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig‑Maximilians‑Universität München, D‑81377 Munich, Germany
| | - Bojan Ljepoja
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig‑Maximilians‑Universität München, D‑81377 Munich, Germany
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig‑Maximilians‑Universität München, D‑81377 Munich, Germany
| | - Georg J Arnold
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig‑Maximilians‑Universität München, D‑81377 Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig‑Maximilians‑Universität München, D‑81377 Munich, Germany
| | - Andreas Roidl
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig‑Maximilians‑Universität München, D‑81377 Munich, Germany
| |
Collapse
|
37
|
Maji D, Lu J, Sarder P, Schmieder AH, Cui G, Yang X, Pan D, Lew MD, Achilefu S, Lanza GM. Cellular Trafficking of Sn-2 Phosphatidylcholine Prodrugs Studied with Fluorescence Lifetime Imaging and Super-resolution Microscopy. PRECISION NANOMEDICINE 2018; 1:128-145. [PMID: 31249994 PMCID: PMC6597004 DOI: 10.33218/prnano1(2).180724.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
While the in vivo efficacy of Sn-2 phosphatidylcholine prodrugs incorporated into targeted, non-pegylated lipid-encapsulated nanoparticles was demonstrated in prior preclinical studies, the microscopic details of cell prodrug internalization and trafficking events are unknown. Classic fluorescence microscopy, fluorescence lifetime imaging microscopy, and single-molecule super-resolution microscopy were used to investigate the cellular handling of doxorubicin-prodrug and AlexaFluor™-488-prodrug. Sn-2 phosphatidylcholine prodrugs delivered by hemifusion of nanoparticle and cell phospholipid membranes functioned as phosphatidylcholine mimics, circumventing the challenges of endosome sequestration and release. Phosphatidylcholine prodrugs in the outer cell membrane leaflet translocated to the inner membrane leaflet by ATP-dependent and ATP-independent mechanisms and distributed broadly within the cytosolic membranes over the next 12 h. A portion of the phosphatidylcholine prodrug populated vesicle membranes trafficked to the perinuclear Golgi/ER region, where the drug was enzymatically liberated and activated. Native doxorubicin entered the cells, passed rapidly to the nucleus, and bound to dsDNA, whereas DOX was first enzymatically liberated from DOX-prodrug within the cytosol, particularly in the perinuclear region, before binding nuclear dsDNA. Much of DOX-prodrug was initially retained within intracellular membranes. In vitro anti-proliferation effectiveness of the two drug delivery approaches was equivalent at 48 h, suggesting that residual intracellular DOX-prodrug may constitute a slow-release drug reservoir that enhances effectiveness. We have demonstrated that Sn-2 phosphatidylcholine prodrugs function as phosphatidylcholine mimics following reported pathways of phosphatidylcholine distribution and metabolism. Drug complexed to the Sn-2 fatty acid is enzymatically liberated and reactivated over many hours, which may enhance efficacy overtime.
Collapse
Affiliation(s)
- Dolonchampa Maji
- Optical Radiology Lab, Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | - Jin Lu
- Department of Electrical and Systems Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Pinaki Sarder
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine & Biomedical Sciences, University of Buffalo, Buffalo, NY 14203
| | - Anne H Schmieder
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Grace Cui
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaoxia Yang
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Dipanjan Pan
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Matthew D Lew
- Department of Electrical and Systems Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Samuel Achilefu
- Optical Radiology Lab, Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | - Gregory M Lanza
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA.,Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
38
|
Manara MC, Valente S, Cristalli C, Nicoletti G, Landuzzi L, Zwergel C, Mazzone R, Stazi G, Arimondo PB, Pasello M, Guerzoni C, Picci P, Nanni P, Lollini PL, Mai A, Scotlandi K. A Quinoline-Based DNA Methyltransferase Inhibitor as a Possible Adjuvant in Osteosarcoma Therapy. Mol Cancer Ther 2018; 17:1881-1892. [DOI: 10.1158/1535-7163.mct-17-0818] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 12/04/2017] [Accepted: 06/21/2018] [Indexed: 11/16/2022]
|
39
|
Martin JH, Bromfield EG, Aitken RJ, Lord T, Nixon B. Double Strand Break DNA Repair occurs via Non-Homologous End-Joining in Mouse MII Oocytes. Sci Rep 2018; 8:9685. [PMID: 29946146 PMCID: PMC6018751 DOI: 10.1038/s41598-018-27892-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 06/07/2018] [Indexed: 12/12/2022] Open
Abstract
The unique biology of the oocyte means that accepted paradigms for DNA repair and protection are not of direct relevance to the female gamete. Instead, preservation of the integrity of the maternal genome depends on endogenous protein stores and/or mRNA transcripts accumulated during oogenesis. The aim of this study was to determine whether mature (MII) oocytes have the capacity to detect DNA damage and subsequently mount effective repair. For this purpose, DNA double strand breaks (DSB) were elicited using the topoisomerase II inhibitor, etoposide (ETP). ETP challenge led to a rapid and significant increase in DSB (P = 0.0002) and the consequential incidence of metaphase plate abnormalities (P = 0.0031). Despite this, ETP-treated MII oocytes retained their ability to participate in in vitro fertilisation, though displayed reduced developmental competence beyond the 2-cell stage (P = 0.02). To account for these findings, we analysed the efficacy of DSB resolution, revealing a significant reduction in DSB lesions 4 h post-ETP treatment. Notably, this response was completely abrogated by pharmacological inhibition of key elements (DNA-PKcs and DNA ligase IV) of the canonical non-homologous end joining DNA repair pathway, thus providing the first evidence implicating this reparative cascade in the protection of the maternal genome.
Collapse
Affiliation(s)
- Jacinta H Martin
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia. .,Preganancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia.
| | - Elizabeth G Bromfield
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia.,Preganancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - R John Aitken
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia.,Preganancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Tessa Lord
- School of Molecular Biosciences, Centre for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164, USA
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia.,Preganancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| |
Collapse
|
40
|
Field LD, Walper SA, Susumu K, Lasarte-Aragones G, Oh E, Medintz IL, Delehanty JB. A Quantum Dot-Protein Bioconjugate That Provides for Extracellular Control of Intracellular Drug Release. Bioconjug Chem 2018; 29:2455-2467. [DOI: 10.1021/acs.bioconjchem.8b00357] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Lauren D. Field
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, D.C. 20375, United States
| | - Scott A. Walper
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, D.C. 20375, United States
| | - Kimihiro Susumu
- Optical Sciences Division, Code 5600, U.S. Naval Research Laboratory, Washington, D.C. 20375, United States
- KeyW Corporation, Hanover, Maryland 21076, United States
| | - Guillermo Lasarte-Aragones
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, D.C. 20375, United States
- George Mason University, College of Sciences, Fairfax, Virginia 22030 United States
| | - Eunkeu Oh
- Optical Sciences Division, Code 5600, U.S. Naval Research Laboratory, Washington, D.C. 20375, United States
- KeyW Corporation, Hanover, Maryland 21076, United States
| | - Igor L. Medintz
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, D.C. 20375, United States
| | - James B. Delehanty
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, D.C. 20375, United States
| |
Collapse
|
41
|
Huang H, Li Y, Wu M, Luo J, Nie J, Hou B, He Q, Diao Y, Qi L, Zhao Y, Liu Y, Yang D, Zhou L. Effects of ethanol on the anticancer function of doxorubicin in JJ012 cells. Future Oncol 2018; 14:1285-1297. [PMID: 29774752 DOI: 10.2217/fon-2017-0547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Chondrosarcoma is difficult to treat because of resistance to conventional chemotherapy and radiotherapy. This study evaluated the effects of ethanol in combination with doxorubicin in chondrosarcoma cells. MATERIALS & METHODS JJ012, was treated with doxorubicin alone or in combination with ethanol. Effects on cellular proliferation, migration, invasion, apoptosis, and the cell cycle were evaluated. RESULTS Treatment of JJ012 cells with 100 mM ethanol and doxorubicin resulted in reduced cell growth, invasion, and migration. In addition, doxorubicin uptake into the nucleus was enhanced and p53 mRNA expression was upregulated in JJ012 cells. CONCLUSION Ethanol combined with doxorubicin increased doxorubicin uptake in the nucleus and enhanced the effects of doxorubicin in JJ012 cells.
Collapse
Affiliation(s)
- Hui Huang
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, PR China.,Translational Medicine Center of Northern China, Harbin Medical University, Harbin, PR China.,Basic Medical Institute of Heilongjiang Medical Science Academy, Harbin, PR China.,Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, PR China
| | - Yanze Li
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, PR China.,Translational Medicine Center of Northern China, Harbin Medical University, Harbin, PR China.,Basic Medical Institute of Heilongjiang Medical Science Academy, Harbin, PR China.,Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, PR China
| | - Mingjuan Wu
- Heilongjiang Academy of Traditional Chinese Medicine, Harbin, PR China
| | - Jing Luo
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, PR China.,Translational Medicine Center of Northern China, Harbin Medical University, Harbin, PR China
| | - Junhui Nie
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, PR China.,Translational Medicine Center of Northern China, Harbin Medical University, Harbin, PR China
| | - Baoyu Hou
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, PR China.,Translational Medicine Center of Northern China, Harbin Medical University, Harbin, PR China
| | - Qi He
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, PR China.,Translational Medicine Center of Northern China, Harbin Medical University, Harbin, PR China
| | - Yan Diao
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, PR China.,Translational Medicine Center of Northern China, Harbin Medical University, Harbin, PR China
| | - Lin Qi
- Department of Radioimmunossay, Heilongjiang Province Hospital, Harbin, PR China
| | - Yuanyuan Zhao
- Department of Anesthesiology, Heilongjiang Province Hospital, Harbin, PR China
| | - Ying Liu
- Department of Gastroenterology, Heilongjiang Province Hospital, Harbin, PR China
| | - Dan Yang
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, PR China.,Translational Medicine Center of Northern China, Harbin Medical University, Harbin, PR China.,Basic Medical Institute of Heilongjiang Medical Science Academy, Harbin, PR China.,Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, PR China
| | - Lingyun Zhou
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, PR China.,Translational Medicine Center of Northern China, Harbin Medical University, Harbin, PR China.,Basic Medical Institute of Heilongjiang Medical Science Academy, Harbin, PR China.,Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, PR China
| |
Collapse
|
42
|
Baxter-Holland M, Dass CR. Doxorubicin, mesenchymal stem cell toxicity and antitumour activity: implications for clinical use. ACTA ACUST UNITED AC 2018; 70:320-327. [PMID: 29355940 DOI: 10.1111/jphp.12869] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 11/25/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVES The use of doxorubicin, an antineoplastic medication used for the treatment of cancers via mechanisms that prevent replication of cells or lead to their death, can result in damage to healthy cells as well as malignant. Among the affected cells are mesenchymal stem cells (MSCs), which are involved in the maintenance and repair of tissues in the body. This review explores the mechanisms of biological effects and damage attributed to doxorubicin on MSCs. The PubMed database was used as a source of literature for this review. KEY FINDINGS Doxorubicin has the potential to lead to significant and irreversible damage to the human bone marrow environment, including MSCs. The primary known mechanism of these changes is through free radical damage and activation of apoptotic pathways. The presence of MSCs in culture or in vivo appears to either suppress or promote tumour growth. Interactions between doxorubicin and MSCs have the potential to increase chemotherapy resistance. SUMMARY Doxorubicin-induced damage to MSCs is of concern clinically. However, MSCs also have been associated with resistance of tumour cells to drugs including doxorubicin. Further studies, particularly in vivo, are needed to provide consistent results of how the doxorubicin-induced changes to MSCs affect treatment and patient health.
Collapse
Affiliation(s)
- Mia Baxter-Holland
- School of Pharmacy and Biomedical Science, Curtin University, Perth, WA, Australia
| | - Crispin R Dass
- School of Pharmacy and Biomedical Science, Curtin University, Perth, WA, Australia.,Curtin Health Innovation Research Institute, Perth, WA, Australia
| |
Collapse
|
43
|
Hu G, Zhao X, Wang J, Lv L, Wang C, Feng L, Shen L, Ren W. miR-125b regulates the drug-resistance of breast cancer cells to doxorubicin by targeting HAX-1. Oncol Lett 2017; 15:1621-1629. [PMID: 29434858 PMCID: PMC5774474 DOI: 10.3892/ol.2017.7476] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 09/22/2017] [Indexed: 01/17/2023] Open
Abstract
MircroRNAs (miRNAs) are considered as essential regulators in the tumorigenesis and chemoresistance of multiple cancer types. In the present study, it was demonstrated that the expression levels of miR-125b were significantly downregulated in the tissues of patients with breast cancer (BC), as well as the BC cell lines in vitro. To study the association between chemoresistance and miR-125b in BC, doxorubicin (DOX)-resistant MCF-7 (MCF-7/R) cells were established, and gain- and loss-of-function experiments were performed. It was demonstrated that the overexpression of miR-125b increased the sensitivity of MCF-7/R cells to DOX. Furthermore, it was revealed that the sensitization of miR-125b mimics to DOX-induced cell death was regulated by the hematopoietic cell-specific protein 1-associated protein X-1 (HAX-1) vector and HAX-1 small interfering RNA. These results emphasized the notable function of miR-125b and its target of HAX-1 in regulating DOX-resistance. In addition, it was demonstrated that the miR-125b mimics promoted the loss of the mitochondrial membrane potential and the generation of reactive oxygen species induced by DOX treatment in MCF-7/R cells. These data suggest that the miR-125b-HAX-1-mitochondria pathway has a notable function in the treatment of DOX-resistant BC cells, which may provide a novel target for the chemotherapy of BC.
Collapse
Affiliation(s)
- Guinv Hu
- Department of Breast Surgery, Dongyang People's Hospital, Jinhua, Zhejiang 322100, P.R. China
| | - Xiaokang Zhao
- Department of Breast Surgery, Dongyang People's Hospital, Jinhua, Zhejiang 322100, P.R. China
| | - Jiang Wang
- Department of Breast Surgery, Dongyang People's Hospital, Jinhua, Zhejiang 322100, P.R. China
| | - Liting Lv
- Department of Breast Surgery, Dongyang People's Hospital, Jinhua, Zhejiang 322100, P.R. China
| | - Chaoqun Wang
- Department of Breast Surgery, Dongyang People's Hospital, Jinhua, Zhejiang 322100, P.R. China
| | - Liang Feng
- Department of Breast Surgery, Shaoxing Shangyu People's Hospital, Shaoxing, Zhejiang 312300, P.R. China
| | - Liangqiong Shen
- Department of Breast Surgery, Shaoxing Shangyu People's Hospital, Shaoxing, Zhejiang 312300, P.R. China
| | - Weili Ren
- Department of Breast Surgery, Shaoxing Shangyu People's Hospital, Shaoxing, Zhejiang 312300, P.R. China
| |
Collapse
|
44
|
Carlson M, Watson AL, Anderson L, Largaespada DA, Provenzano PP. Multiphoton fluorescence lifetime imaging of chemotherapy distribution in solid tumors. JOURNAL OF BIOMEDICAL OPTICS 2017; 22:1-9. [PMID: 29188660 PMCID: PMC5712660 DOI: 10.1117/1.jbo.22.11.116010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 11/08/2017] [Indexed: 05/22/2023]
Abstract
Doxorubicin is a commonly used chemotherapeutic employed to treat multiple human cancers, including numerous sarcomas and carcinomas. Furthermore, doxorubicin possesses strong fluorescent properties that make it an ideal reagent for modeling drug delivery by examining its distribution in cells and tissues. However, while doxorubicin fluorescence and lifetime have been imaged in live tissue, its behavior in archival samples that frequently result from drug and treatment studies in human and animal patients, and murine models of human cancer, has to date been largely unexplored. Here, we demonstrate imaging of doxorubicin intensity and lifetimes in archival formalin-fixed paraffin-embedded sections from mouse models of human cancer with multiphoton excitation and multiphoton fluorescence lifetime imaging microscopy (FLIM). Multiphoton excitation imaging reveals robust doxorubicin emission in tissue sections and captures spatial heterogeneity in cells and tissues. However, quantifying the amount of doxorubicin signal in distinct cell compartments, particularly the nucleus, often remains challenging due to strong signals in multiple compartments. The addition of FLIM analysis to display the spatial distribution of excited state lifetimes clearly distinguishes between signals in distinct compartments such as the cell nuclei versus cytoplasm and allows for quantification of doxorubicin signal in each compartment. Furthermore, we observed a shift in lifetime values in the nuclei of transformed cells versus nontransformed cells, suggesting a possible diagnostic role for doxorubicin lifetime imaging to distinguish normal versus transformed cells. Thus, data here demonstrate that multiphoton FLIM is a highly sensitive platform for imaging doxorubicin distribution in normal and diseased archival tissues.
Collapse
Affiliation(s)
- Marjorie Carlson
- University of Minnesota, Department of Biomedical Engineering, Minneapolis, Minnesota, United States
- University of Minnesota, Physical Sciences in Oncology Center, Minneapolis, Minnesota, United States
| | - Adrienne L. Watson
- University of Minnesota, Masonic Cancer Center, Minneapolis, Minnesota, United States
| | - Leah Anderson
- University of Minnesota, Masonic Cancer Center, Minneapolis, Minnesota, United States
| | - David A. Largaespada
- University of Minnesota, Masonic Cancer Center, Minneapolis, Minnesota, United States
| | - Paolo P. Provenzano
- University of Minnesota, Department of Biomedical Engineering, Minneapolis, Minnesota, United States
- University of Minnesota, Physical Sciences in Oncology Center, Minneapolis, Minnesota, United States
- University of Minnesota, Masonic Cancer Center, Minneapolis, Minnesota, United States
- University of Minnesota, Stem Cell Institute, Minneapolis, Minnesota, United States
- University of Minnesota, Institute for Engineering in Medicine, Minneapolis, Minnesota, United States
- Address all correspondence to: Paolo P. Provenzano, E-mail:
| |
Collapse
|
45
|
Li J, Hao D, Wang L, Wang H, Wang Y, Zhao Z, Li P, Deng C, Di LJ. Epigenetic targeting drugs potentiate chemotherapeutic effects in solid tumor therapy. Sci Rep 2017. [PMID: 28642588 PMCID: PMC5481380 DOI: 10.1038/s41598-017-04406-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Epigenetic therapy is a novel tumor therapeutic method and refers to the targeting of the aberrant epigenetic modifications presumably at cancer-related genes by chemicals which are epigenetic targeting drugs (ETDs). Not like in treating hematopoietic cancer, the clinical trials investigating the potential use of ETDs in the solid tumor is not encouraging. Instead, the curative effects of ETD delivered together with DNA targeting chemo drugs (DTDs) are quite promising according to our meta-analysis. To investigate the synergistic mechanism of ETD and DTD drug combination, the therapeutic effect was studied using both cell lines and mouse engrafted tumors. Mechanically we show that HDAC inhibitors and DNMT inhibitors are capable of increasing the chromatin accessibility to cisplatin (CP) and doxorubicin (Dox) through chromatin decompaction globally. Consequently, the combination of ETD and DTD enhances the DTD induced DNA damage and cell death. Engrafted tumors in SCID mice also show increased sensitivity to irradiation (IR) or CP when the tumors were pretreated by ETDs. Given the limited therapeutic effect of ETD alone, these results strongly suggest that the combination of DTD, including irradiation, and ETD treatment is a very promising choice in clinical solid tumor therapy.
Collapse
Affiliation(s)
- Jingjing Li
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau, China
| | - Dapeng Hao
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau, China
| | - Li Wang
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau, China.,Metabolomics Core, Faculty of Health Sciences, University of Macau, Macau, China
| | - Haitao Wang
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau, China
| | - Yuan Wang
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau, China
| | - Zhiqiang Zhao
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau, China
| | - Peipei Li
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau, China
| | - Chuxia Deng
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau, China
| | - Li-Jun Di
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau, China.
| |
Collapse
|
46
|
Muthiyan R, Nambikkairaj B, Mahanta N, Immanuel T, Mandal RS, Kumaran K, De AK. Antiproliferative and Proapoptotic Activities of Marine Sponge Hyrtios erectus Extract on Breast Carcinoma Cell Line (MCF-7). Pharmacogn Mag 2017; 13:S41-S47. [PMID: 28479725 PMCID: PMC5407115 DOI: 10.4103/0973-1296.203983] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 04/28/2016] [Indexed: 01/01/2023] Open
Abstract
Background: Marine sponge is a rich natural resource of many pharmacologically important compounds. Objective: Marine sponge Hyrtios erectus, collected from North Bay, South Andaman Sea, India, was screened for potential antiproliferative and proapoptotic properties on a breast adenocarcinoma cell line (MCF-7). Materials and Methods: 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay was used to test the antiproliferative and cytotoxicity effects of the sponge extract. Analysis of apoptosis and cell cycle stages were done by flow cytometry. The expression of several apoptotic-related proteins in MCF-7 cells treated by the extract was evaluated by Western blot analysis. Various analytical techniques including Fourier transform infrared spectroscopy, gas chromatography-mass spectrometry, and nuclear magnetic resonance were employed to determine the identity of the active compounds in the sponge extract. Results: N-Hexane extract of the sponge inhibited proliferation of the MCF-7 cell line in a dose- and time-dependent manner. Exposure of the sponge extract triggered apoptosis of the MCF-7 cells, induced DNA fragmentation, and arrested the cells in G2/M phase. Treatment of the sponge extract induced downregulation of antiapoptotic Bcl-2 protein and upregulation of Bax, caspase-3, caspase-9, and fragmented poly(ADP ribose)polymerase proteins in MCF-7 cells. Five bioactive compounds have been identified in the extract. Conclusion: The antiproliferative and proapoptotic activities of the tested extract suggested the pharmacologic potential of the identified compounds. Further characterization of the identified compounds are in progress. SUMMARY The N-hexane extract of the marine sponge Hyrtios erectus, collected from North Bay, South Andaman Sea, India, showed potential antiproliferative and proapoptotic properties against a breast adenocarcinoma cell line (MCF-7). The sponge extract retarded the growth of breast carcinoma cell line MCF-7 cells in a time- and dose-dependent manner. The sponge extract induced apoptosis of breast cancer cell line MCF-7 and arrested cells in G2/M phase. The sponge extract induced downregulation of Bcl-2 protein in MCF-7 cell line and upregulation of Bax, caspase-3, and cleaved PARP. Five bioactive compounds have been identified in the extract.
Abbreviations used: GC-MS: Gas chromatography-mass spectrometry; FT-IR: Fourier transform infrared spectroscopy; NMR: Nuclear magnetic resonance; MTT: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide.
Collapse
Affiliation(s)
| | - Balwin Nambikkairaj
- Department of Zoology, Voorhees College, Thiruvalluvar University, Vellore, India
| | - Nilkamal Mahanta
- Department of Chemistry, Institute for Genomic Biology, University of Illinois, Urbana-Champaign, Illinois, USA
| | - Titus Immanuel
- Division of Fisheries Sciences, Central Island Agricultural Research Institute, Port Blair, Andaman and Nicobar Islands, India
| | - Rahul Shubhra Mandal
- Biomedical Informatics Centre, National Institute of Cholera and Enteric Diseases, Beliaghata, Kolkata, India
| | | | - Arun Kumar De
- Department of Animal Sciences, Central Island Agricultural Research Institute, Port Blair, Andaman and Nicobar Islands, India.,Department of Animal Sciences, University of Illinois, Urbana-Champaign, Illinois, USA
| |
Collapse
|
47
|
Fluorescence properties of doxorubicin in PBS buffer and PVA films. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2017; 170:65-69. [PMID: 28390260 DOI: 10.1016/j.jphotobiol.2017.03.024] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 03/20/2017] [Accepted: 03/28/2017] [Indexed: 12/20/2022]
Abstract
We studied steady-state and time-resolved fluorescence properties of an anticancer drug Doxorubicin in a saline buffer and poly-vinyl alcohol (PVA) film. Absorption of Doxorubicin, located at blue-green spectral region, allows a convenient excitation with visible light emitting diodes or laser diodes. Emission of Doxorubicin with maximum near 600nm can be easily detected with photomultipliers and CCD cameras. Both, absorption and fluorescence spectra in polymeric matrix show more pronounced vibronic structures than in solution. Also, the steady-state anisotropy in the polymer film is significantly higher than in the saline solution. In PVA film the fluorescence anisotropy is about 0.30 whereas in the saline buffer only 0.07. Quantum efficiencies of Doxorubicin were compared to a known standard Rhodamine 101 which has fluorescence emission in a similar spectral region. The quantum yield of Doxorubicin in PVA film is more than 10% and about twice higher than in the saline solution. Similarly, the lifetime of doxorubicin in PVA film is about 2ns whereas in the saline solution only about 1ns. The fluorescence anisotropy decays show that Doxorubicin molecules are freely rotating in the saline buffer with a correlation time of about 290ps, and are almost completely immobilized in the PVA film. The spectroscopic investigations presented in this manuscript are important, as they provide answers to changes in molecular properties of Doxorubicin depending changes in the local environment, which is useful when synthesizing nanoparticles for Doxorubicin entrapment.
Collapse
|
48
|
Le DHT, Lee KL, Shukla S, Commandeur U, Steinmetz NF. Potato virus X, a filamentous plant viral nanoparticle for doxorubicin delivery in cancer therapy. NANOSCALE 2017; 9:2348-2357. [PMID: 28144662 PMCID: PMC5370163 DOI: 10.1039/c6nr09099k] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Plant viral nanoparticles (VNPs) are a novel class of nanocarriers with implications for drug delivery in cancer therapy. VNPs are characterized by their highly symmetrical nanoscale structures. Furthermore, plant VNPs are biocompatible, biodegradable, and non-infectious in mammals. VNPs provide a proteinaceous platform technology that can be readily engineered to carry contrast agents and therapies using chemical and genetic modifications. Of particular interest are high aspect ratio, elongated filaments such as the ones formed by potato virus X (PVX, measuring 515 × 13 nm). PVX has demonstrated enhanced tumor homing and penetration properties compared to spherical counterparts. Here, we sought to investigate the potential of PVX as a drug carrier delivering doxorubicin (DOX), a commonly used cancer chemotherapy. We synthesized therapeutic PVX nanoparticles using a simple in-solution mixing protocol; after 5 days of mixing of DOX and PVX and ultra-centrifugal purification, ∼1000 DOX per PVX were stably associated with the carrier, most likely based on hydrophobic interaction. Efficacy and drug activity of PVX-DOX were confirmed using a panel of cancer cell lines including ovarian cancer, breast cancer, and cervical cancer. Lastly, we demonstrated treatment of athymic mice bearing human MDA-MB-231 breast cancer xenografts: PVX-DOX treatment resulted in reduced tumor growth in this model. Our results open the door for further development of PVX and other high aspect ratio plant VNPs for applications in cancer therapy.
Collapse
Affiliation(s)
- Duc H T Le
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, 10900 Euclid Ave., Cleveland, OH 44106, USA.
| | - Karin L Lee
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, 10900 Euclid Ave., Cleveland, OH 44106, USA.
| | - Sourabh Shukla
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, 10900 Euclid Ave., Cleveland, OH 44106, USA.
| | - Ulrich Commandeur
- Department of Molecular Biotechnology, RWTH-Aachen University, 52064 Aachen, Germany
| | - Nicole F Steinmetz
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, 10900 Euclid Ave., Cleveland, OH 44106, USA. and Department of Radiology, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106, USA and Department of Materials Science and Engineering, Case Western Reserve University School of Engineering, 10900 Euclid Ave., Cleveland, OH 44106, USA and Department of Macromolecular Science and Engineering, Case Western Reserve University School of Engineering, 10900 Euclid Ave., Cleveland, OH 44106, USA and Division of General Medical Sciences-Oncology, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| |
Collapse
|
49
|
Suarasan S, Focsan M, Potara M, Soritau O, Florea A, Maniu D, Astilean S. Doxorubicin-Incorporated Nanotherapeutic Delivery System Based on Gelatin-Coated Gold Nanoparticles: Formulation, Drug Release, and Multimodal Imaging of Cellular Internalization. ACS APPLIED MATERIALS & INTERFACES 2016; 8:22900-13. [PMID: 27537061 DOI: 10.1021/acsami.6b07583] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
In this work, we developed a new pH- and temperature-responsive nanochemotherapeutic system based on Doxorubicin (DOX) noncovalently bound to biosynthesized gelatin-coated gold nanoparticles (DOX-AuNPs@gelatin). The real-time release profile of DOX was evaluated at different pH values (7.4, 5.3, and 4.6) and temperatures (22-45 °C) in aqueous solutions, and its therapeutic performance was examined in vitro against MCF-7 breast cancer cells. TEM, dark-field scattering, and wide-field fluorescence microscopy indicated the effective uptake of nanochemotherapeutics with the subsequent release and progressive accumulation of DOX in cell nuclei. MTT assays clearly showed the effectiveness of the treatment by inhibiting the growth of MCF-7 breast cancer cells for a loaded drug concentration of 5 μg/mL. The most informative data about the dynamic release and localization were provided by scanning confocal microscopy using time-resolved fluorescence and surface-enhanced Raman scattering (SERS) techniques. In particular, fluorescence-lifetime imaging (FLIM) recorded under 485 nm pulsed diode laser excitation revealed the bimodal distribution of DOX in cells. The shorter fluorescence lifetime of DOX localized in nuclei (1.52 ns) than in the cytoplasm (2.4 ns) is consistent with the cytotoxic mechanism induced by DOX-DNA intercalation. Remarkably, the few DOX molecules captured between nanoparticles ("electromagnetic hotspots") after most drug is released act as SERS reporters for the localization of plasmonic nanocarriers in MCF-7 cells. The high drug loading capacity and effective drug release under pH control combined with the advantage of multimodal visualization inside cells clearly indicate the high potential of our DOX-AuNPs@gelatin delivery system for implementation in nanomedicine.
Collapse
Affiliation(s)
- Sorina Suarasan
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute on Bio-Nano-Sciences Babes-Bolyai University , T. Laurian Str. 42, 400271 Cluj-Napoca, Romania
| | - Monica Focsan
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute on Bio-Nano-Sciences Babes-Bolyai University , T. Laurian Str. 42, 400271 Cluj-Napoca, Romania
| | - Monica Potara
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute on Bio-Nano-Sciences Babes-Bolyai University , T. Laurian Str. 42, 400271 Cluj-Napoca, Romania
| | - Olga Soritau
- Laboratory of Cell Biology and Radiobiology "Ion Chiricuta" Institute of Oncology , Republicii Str. 34-36, 400015 Cluj-Napoca, Romania
| | - Adrian Florea
- Department of Cell and Molecular Biology, Faculty of Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy , Louis Pasteur Str., 6, 400349 Cluj-Napoca, Romania
| | - Dana Maniu
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute on Bio-Nano-Sciences Babes-Bolyai University , T. Laurian Str. 42, 400271 Cluj-Napoca, Romania
- Department of Biomolecular Physics, Faculty of Physics, Babes-Bolyai University , M Kogalniceanu Str. 1, 400084 Cluj-Napoca, Romania
| | - Simion Astilean
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute on Bio-Nano-Sciences Babes-Bolyai University , T. Laurian Str. 42, 400271 Cluj-Napoca, Romania
- Department of Biomolecular Physics, Faculty of Physics, Babes-Bolyai University , M Kogalniceanu Str. 1, 400084 Cluj-Napoca, Romania
| |
Collapse
|
50
|
Kanehira Y, Togami K, Tada H, Chono S. Tumor distribution and anti-tumor effect of doxorubicin following intrapulmonary administration to mice with metastatic lung tumor. J Drug Deliv Sci Technol 2016. [DOI: 10.1016/j.jddst.2016.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|