101
|
Norouzi S, Yazdian Robati R, Ghandadi M, Abnous K, Behravan J, Mosaffa F. Comparative proteomics study of proteins involved in induction of higher rates of cell death in mitoxantrone-resistant breast cancer cells MCF-7/MX exposed to TNF-α. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:663-672. [PMID: 32742605 PMCID: PMC7374993 DOI: 10.22038/ijbms.2020.40029.9486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Objective(s): Resistance to medications is one of the main complications in chemotherapy of cancer. It has been shown that some multidrug resistant cancer cells indicate more sensitivity against cytotoxic effects of TNF-α compared to their parental cells. Our previous findings indicated vulnerability of the mitoxantrone-resistant breast cancer cells MCF-7/MX to cell death induced by TNF-α compared to the parent cells MCF-7. In this study, we performed a comparative proteomics analysis for identification of proteins involved in induction of higher susceptibility of MCF-7/MX cells to cytotoxic effect of TNF-α. Materials and Methods: Intensity of protein spots in 2D gel electrophoresis profiles of MCF-7 and MCF-7/MX cells were compared with Image Master Platinum 6.0 software. Selected differential protein-spots were identified with MALDI-TOF/TOF mass spectrometry and database searching. Pathway analyses of identified proteins were performed using PANTHER, KEGG PATHWAY, Gene MANIA and STRING databases. Western blot was performed for confirmation of the proteomics results. Results: Our results indicated that 48 hr exposure to TNF-α induced 87% death in MCF-7/MX cells compared to 19% death in MCF-7 cells. Forty landmarks per 2D gel electrophoresis were matched by Image Master Software. Six proteins were identified with mass spectrometry. Western blot showed that 14-3-3γ and p53 proteins were expressed higher in MCF-7/MX cells treated with TNF-α compared to MCF-7 cells treated with TNF-α. Conclusion: Our results showed that 14-3-3 γ, prohibitin, peroxiredoxin 2 and P53 proteins which were expressed differentially in MCF-7/MX cells treated with TNF-α may involve in the induction of higher rates of cell death in these cells compared to TNF-α-treated MCF-7 cells.
Collapse
Affiliation(s)
- Saeed Norouzi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Rezvan Yazdian Robati
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Morteza Ghandadi
- Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran.,Department of Pharmacognosy and Pharmaceutical Biotechnology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Behravan
- School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Mosaffa
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
102
|
Lee GY, Lee JS, Son CG, Lee NH. Combating Drug Resistance in Colorectal Cancer Using Herbal Medicines. Chin J Integr Med 2020; 27:551-560. [PMID: 32740824 DOI: 10.1007/s11655-020-3425-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2020] [Indexed: 12/11/2022]
Abstract
Colorectal cancer (CRC) is one of the most prevalent and lethal cancer types around the world. Most of the CRC patients are treated with chemotherapeutic drugs alone or combined. However, up to 90% of metastatic cancer patients experience the failure of treatment mostly because of the acquired drug resistance, which can be led to multidrug resistance (MDR). In this study, we reviewed the recent literature which studied potential CRC MDR reversal agents among herbal medicines (HMs). Among abundant HMs, 6 single herbs, Andrographis paniculata, Salvia miltiorrhiza, Hedyotis diffusa, Sophora flavescens, Curcuma longa, Bufo gargarizans, and 2 formulae, Pien Tze Huang and Zhi Zhen Fang, were found to overcome CRC MDR by two or more different mechanisms, which could be a promising candidate in the development of new drugs for adjuvant CRC chemotherapy.
Collapse
Affiliation(s)
- Ga-Young Lee
- Department of Clinical Oncology, Cheonan Korean Medicine Hospital of Daejeon University, Cheonan, 31099, Republic of Korea.,Liver & Immunology Research Center, Dunsan Korean Medicine Hospital of Daejeon University, Daejeon, 35235, Republic of Korea.,Department of Internal Medicine, Graduated School of Korean Medicine, University of Daejeon, Daejeon, 34520, Republic of Korea
| | - Jin-Seok Lee
- Liver & Immunology Research Center, Dunsan Korean Medicine Hospital of Daejeon University, Daejeon, 35235, Republic of Korea.,Department of Internal Medicine, Graduated School of Korean Medicine, University of Daejeon, Daejeon, 34520, Republic of Korea
| | - Chang-Gue Son
- Liver & Immunology Research Center, Dunsan Korean Medicine Hospital of Daejeon University, Daejeon, 35235, Republic of Korea.,Department of Internal Medicine, Graduated School of Korean Medicine, University of Daejeon, Daejeon, 34520, Republic of Korea
| | - Nam-Hun Lee
- Department of Clinical Oncology, Cheonan Korean Medicine Hospital of Daejeon University, Cheonan, 31099, Republic of Korea. .,Liver & Immunology Research Center, Dunsan Korean Medicine Hospital of Daejeon University, Daejeon, 35235, Republic of Korea. .,Department of Internal Medicine, Graduated School of Korean Medicine, University of Daejeon, Daejeon, 34520, Republic of Korea.
| |
Collapse
|
103
|
Bucevičius J, Kostiuk G, Gerasimaitė R, Gilat T, Lukinavičius G. Enhancing the biocompatibility of rhodamine fluorescent probes by a neighbouring group effect. Chem Sci 2020; 11:7313-7323. [PMID: 33777348 PMCID: PMC7983176 DOI: 10.1039/d0sc02154g] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/20/2020] [Indexed: 12/11/2022] Open
Abstract
Fluorescence microscopy is an essential tool for understanding dynamic processes in living cells and organisms. However, many fluorescent probes for labelling cellular structures suffer from unspecific interactions and low cell permeability. Herein, we demonstrate that the neighbouring group effect which results from positioning an amide group next to a carboxyl group in the benzene ring of rhodamines dramatically increases cell permeability of the rhodamine-based probes through stabilizing a fluorophore in a hydrophobic spirolactone state. Based on this principle, we create probes targeting tubulin, actin and DNA. Their superb staining intensity, tuned toxicity and specificity allows long-term 3D confocal and STED nanoscopy with sub-30 nm resolution. Due to their unrestricted cell permeability and efficient accumulation on the target, the new probes produce high contrast images at low nanomolar concentrations. Superior performance is exemplified by resolving the real microtubule diameter of 23 nm and selective staining of the centrosome inside living cells for the first time.
Collapse
Affiliation(s)
- Jonas Bucevičius
- Chromatin Labeling and Imaging Group , Department of NanoBiophotonics , Max Planck Institute for Biophysical Chemistry , Am Fassberg 11 , 37077 Göttingen , Germany .
| | - Georgij Kostiuk
- Chromatin Labeling and Imaging Group , Department of NanoBiophotonics , Max Planck Institute for Biophysical Chemistry , Am Fassberg 11 , 37077 Göttingen , Germany .
| | - Rūta Gerasimaitė
- Chromatin Labeling and Imaging Group , Department of NanoBiophotonics , Max Planck Institute for Biophysical Chemistry , Am Fassberg 11 , 37077 Göttingen , Germany .
| | - Tanja Gilat
- Department of NanoBiophotonics , Max Planck Institute for Biophysical Chemistry , Am Fassberg 11 , 37077 Göttingen , Germany
| | - Gražvydas Lukinavičius
- Chromatin Labeling and Imaging Group , Department of NanoBiophotonics , Max Planck Institute for Biophysical Chemistry , Am Fassberg 11 , 37077 Göttingen , Germany .
| |
Collapse
|
104
|
Chellan P, Sadler PJ. Enhancing the Activity of Drugs by Conjugation to Organometallic Fragments. Chemistry 2020; 26:8676-8688. [PMID: 32452579 PMCID: PMC7496994 DOI: 10.1002/chem.201904699] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/12/2020] [Indexed: 12/22/2022]
Abstract
Resistance to chemotherapy is a current clinical problem, especially in the treatment of microbial infections and cancer. One strategy to overcome this is to make new derivatives of existing drugs by conjugation to organometallic fragments, either by an appropriate linker, or by direct coordination of the drug to a metal. We illustrate this with examples of conjugated organometallic metallocene sandwich and half-sandwich complexes, RuII and OsII arene, and RhIII and IrIII cyclopentadienyl half-sandwich complexes. Ferrocene conjugates are particularly promising. The ferrocene-chloroquine conjugate ferroquine is in clinical trials for malaria treatment, and a ferrocene-tamoxifen derivative (a ferrocifen) seems likely to enter anticancer trails soon. Several other examples illustrate that organometallic conjugation can restore the activity of drugs to which resistance has developed.
Collapse
Affiliation(s)
- Prinessa Chellan
- Department of Chemistry and Polymer ScienceStellenbosch University7600Matieland, Western CapeSouth Africa
| | - Peter J. Sadler
- Department of ChemistryUniversity of WarwickCoventryCV4 7ALUK
| |
Collapse
|
105
|
Roy S, Kumaravel S, Sharma A, Duran CL, Bayless KJ, Chakraborty S. Hypoxic tumor microenvironment: Implications for cancer therapy. Exp Biol Med (Maywood) 2020; 245:1073-1086. [PMID: 32594767 DOI: 10.1177/1535370220934038] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
IMPACT STATEMENT Hypoxia contributes to tumor aggressiveness and promotes growth of many solid tumors that are often resistant to conventional therapies. In order to achieve successful therapeutic strategies targeting different cancer types, it is necessary to understand the molecular mechanisms and signaling pathways that are induced by hypoxia. Aberrant tumor vasculature and alterations in cellular metabolism and drug resistance due to hypoxia further confound this problem. This review focuses on the implications of hypoxia in an inflammatory TME and its impact on the signaling and metabolic pathways regulating growth and progression of cancer, along with changes in lymphangiogenic and angiogenic mechanisms. Finally, the overarching role of hypoxia in mediating therapeutic resistance in cancers is discussed.
Collapse
Affiliation(s)
- Sukanya Roy
- Department of Medical Physiology, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807, USA
| | - Subhashree Kumaravel
- Department of Medical Physiology, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807, USA
| | - Ankith Sharma
- Department of Medical Physiology, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807, USA
| | - Camille L Duran
- Department of Molecular & Cellular Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA
| | - Kayla J Bayless
- Department of Molecular & Cellular Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA
| | - Sanjukta Chakraborty
- Department of Medical Physiology, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807, USA
| |
Collapse
|
106
|
A Mechanistic, Enantioselective, Physiologically Based Pharmacokinetic Model of Verapamil and Norverapamil, Built and Evaluated for Drug-Drug Interaction Studies. Pharmaceutics 2020; 12:pharmaceutics12060556. [PMID: 32560124 PMCID: PMC7355632 DOI: 10.3390/pharmaceutics12060556] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 12/30/2022] Open
Abstract
The calcium channel blocker and antiarrhythmic agent verapamil is recommended by the FDA for drug–drug interaction (DDI) studies as a moderate clinical CYP3A4 index inhibitor and as a clinical Pgp inhibitor. The purpose of the presented work was to develop a mechanistic whole-body physiologically based pharmacokinetic (PBPK) model to investigate and predict DDIs with verapamil. The model was established in PK-Sim®, using 45 clinical studies (dosing range 0.1–250 mg), including literature as well as unpublished Boehringer Ingelheim data. The verapamil R- and S-enantiomers and their main metabolites R- and S-norverapamil are represented in the model. The processes implemented to describe the pharmacokinetics of verapamil and norverapamil include enantioselective plasma protein binding, enantioselective metabolism by CYP3A4, non-stereospecific Pgp transport, and passive glomerular filtration. To describe the auto-inhibitory and DDI potential, mechanism-based inactivation of CYP3A4 and non-competitive inhibition of Pgp by the verapamil and norverapamil enantiomers were incorporated based on in vitro literature. The resulting DDI performance was demonstrated by prediction of DDIs with midazolam, digoxin, rifampicin, and cimetidine, with 21/22 predicted DDI AUC ratios or Ctrough ratios within 1.5-fold of the observed values. The thoroughly built and qualified model will be freely available in the Open Systems Pharmacology model repository to support model-informed drug discovery and development.
Collapse
|
107
|
Vinod N, Hwang D, Azam SH, Van Swearingen AED, Wayne E, Fussell SC, Sokolsky-Papkov M, Pecot CV, Kabanov AV. High-capacity poly(2-oxazoline) formulation of TLR 7/8 agonist extends survival in a chemo-insensitive, metastatic model of lung adenocarcinoma. SCIENCE ADVANCES 2020; 6:eaba5542. [PMID: 32596460 PMCID: PMC7299629 DOI: 10.1126/sciadv.aba5542] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 05/06/2020] [Indexed: 05/03/2023]
Abstract
About 40% of patients with non-small cell lung cancer (NSCLC) have stage IV cancer at the time of diagnosis. The only viable treatment options for metastatic disease are systemic chemotherapy and immunotherapy. Nonetheless, chemoresistance remains a major cause of chemotherapy failure. New immunotherapeutic modalities such as anti-PD-1 immune checkpoint blockade have shown promise; however, response to such strategies is highly variable across patients. Here, we show that our unique poly(2-oxazoline)-based nanomicellar formulation (PM) of Resiquimod, an imidazoquinoline Toll-like receptor (TLR) 7/8 agonist, had a superior tumor inhibitory effect in a metastatic model of lung adenocarcinoma, relative to anti-PD-1 therapy or platinum-based chemotherapy. Investigation of the in vivo immune status following Resiquimod PM treatment showed that Resiquimod-based stimulation of antigen-presenting cells in the tumor microenvironment resulted in the mobilization of an antitumor CD8+ immune response. Our study demonstrates the promise of poly(2-oxazoline)-formulated Resiquimod for treating metastatic NSCLC.
Collapse
Affiliation(s)
- Natasha Vinod
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
- Joint UNC/NC State Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC 27599-7575, USA
| | - Duhyeong Hwang
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Salma H. Azam
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Amanda E. D. Van Swearingen
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Elizabeth Wayne
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Sloane Christian Fussell
- Department of Biology, Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Marina Sokolsky-Papkov
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Chad V. Pecot
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alexander V. Kabanov
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
- Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, M.V. Lomonosov Moscow State University, Moscow 119992, Russia
| |
Collapse
|
108
|
Kuroshima K, Yoshino H, Okamura S, Tsuruda M, Osako Y, Sakaguchi T, Sugita S, Tatarano S, Nakagawa M, Enokida H. Potential new therapy of Rapalink-1, a new generation mammalian target of rapamycin inhibitor, against sunitinib-resistant renal cell carcinoma. Cancer Sci 2020; 111:1607-1618. [PMID: 32232883 PMCID: PMC7226215 DOI: 10.1111/cas.14395] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 01/20/2023] Open
Abstract
Sunitinib, a multitargeted receptor tyrosine kinase inhibitor including vascular endothelial growth factor, has been widely used as a first-line treatment against metastatic renal cell carcinoma (mRCC). However, mRCC often acquires resistance to sunitinib, rendering it difficult to treat with this agent. Recently, Rapalink-1, a drug that links rapamycin and the mTOR kinase inhibitor MLN0128, has been developed with excellent therapeutic effects against breast cancer cells carrying mTOR resistance mutations. The aim of the present study was to evaluate the in vitro and in vivo therapeutic efficacy of Rapalink-1 against renal cell carcinoma (RCC) compared to temsirolimus, which is commonly used as a small molecule inhibitor of mTOR and is a derivative of rapamycin. In comparison with temsirolimus, Rapalink-1 showed significantly greater effects against proliferation, migration, invasion and cFolony formation in sunitinib-naïve RCC cells. Inhibition was achieved through suppression of the phosphorylation of substrates in the mTOR signal pathway, such as p70S6K, eukaryotic translation initiation factor 4E-binding protein 1 (4EBP1) and AKT. In addition, Rapalink-1 had greater tumor suppressive effects than temsirolimus against the sunitinib-resistant 786-o cell line (SU-R 786-o), which we had previously established, as well as 3 additional SU-R cell lines established here. RNA sequencing showed that Rapalink-1 suppressed not only the mTOR signaling pathway but also a part of the MAPK signaling pathway, the ErbB signaling pathway and ABC transporters that were associated with resistance to several drugs. Our study suggests the possibility of a new treatment option for patients with RCC that is either sunitinib-sensitive or sunitinib-resistant.
Collapse
Affiliation(s)
- Kazuki Kuroshima
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Hirofumi Yoshino
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Shunsuke Okamura
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Masafumi Tsuruda
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Yoichi Osako
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Takashi Sakaguchi
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Satoshi Sugita
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Shuichi Tatarano
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Masayuki Nakagawa
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Hideki Enokida
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
109
|
Xiao D, Zhou R. Advances in the Application of Liposomal Nanosystems in Anticancer Therapy. Curr Stem Cell Res Ther 2020; 16:14-22. [PMID: 32324519 DOI: 10.2174/1574888x15666200423093906] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/03/2020] [Accepted: 02/17/2020] [Indexed: 02/08/2023]
Abstract
Cancer is the disease with the highest mortality rate, which poses a great threat to people's lives. Cancer caused approximately 3.4 million death worldwide annually. Surgery, chemotherapy and radiotherapy are the main therapeutic methods in clinical practice. However, surgery is only suitable for patients with early-stage cancers, and chemotherapy as well as radiotherapy have various side effects, both of which limit the application of available therapeutic methods. In 1965, liposome was firstly developed to form new drug delivery systems given the unique properties of nanoparticles, such as enhanced permeability and retention effect. During the last 5 decades, liposome has been widely used for the purpose of anticancer drug delivery, and several advances have been made regarding liposomal technology, including long-circulating liposomes, active targeting liposomes and triggered release liposomes, while problems exist all along. This review introduced the advances as well as the problems during the development of liposomal nanosystems for cancer therapy in recent years.
Collapse
Affiliation(s)
- Dexuan Xiao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ronghui Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
110
|
Rudnik LAC, Farago PV, Manfron Budel J, Lyra A, Barboza FM, Klein T, Kanunfre CC, Nadal JM, Bandéca MC, Raman V, Novatski A, Loguércio AD, Zanin SMW. Co-Loaded Curcumin and Methotrexate Nanocapsules Enhance Cytotoxicity against Non-Small-Cell Lung Cancer Cells. Molecules 2020; 25:molecules25081913. [PMID: 32326159 PMCID: PMC7221560 DOI: 10.3390/molecules25081913] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/11/2020] [Accepted: 04/17/2020] [Indexed: 11/16/2022] Open
Abstract
Background: As part of the efforts to find natural alternatives for cancer treatment and to overcome the barriers of cellular resistance to chemotherapeutic agents, polymeric nanocapsules containing curcumin and/or methotrexate were prepared by an interfacial deposition of preformed polymer method. Methods: Physicochemical properties, drug release experiments and in vitro cytotoxicity of these nanocapsules were performed against the Calu-3 lung cancer cell line. Results: The colloidal suspensions of nanocapsules showed suitable size (287 to 325 nm), negative charge (-33 to -41 mV) and high encapsulation efficiency (82.4 to 99.4%). Spherical particles at nanoscale dimensions were observed by scanning electron microscopy. X-ray diffraction analysis indicated that nanocapsules exhibited a non-crystalline pattern with a remarkable decrease of crystalline peaks of the raw materials. Fourier-transform infrared spectra demonstrated no chemical bond between the drug(s) and polymers. Drug release experiments evidenced a controlled release pattern with no burst effect for nanocapsules containing curcumin and/or methotrexate. The nanoformulation containing curcumin and methotrexate (NCUR/MTX-2) statistically decreased the cell viability of Calu-3. The fluorescence and morphological analyses presented a predominance of early apoptosis and late apoptosis as the main death mechanisms for Calu-3. Conclusions: Curcumin and methotrexate co-loaded nanocapsules can be further used as a novel therapeutic strategy for treating non-small-cell lung cancer.
Collapse
Affiliation(s)
- Loanda Aparecida Cabral Rudnik
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmaceutical Sciences, State University of Ponta Grossa, 84030-900 Ponta Grossa, Brazil; (L.A.C.R.); (P.V.F.); (A.L.); (F.M.B.); (T.K.); (J.M.N.); (A.N.); (A.D.L.)
| | - Paulo Vitor Farago
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmaceutical Sciences, State University of Ponta Grossa, 84030-900 Ponta Grossa, Brazil; (L.A.C.R.); (P.V.F.); (A.L.); (F.M.B.); (T.K.); (J.M.N.); (A.N.); (A.D.L.)
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmacy, Federal University of Paraná, 81020-430 Curitiba, Brazil;
| | - Jane Manfron Budel
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmaceutical Sciences, State University of Ponta Grossa, 84030-900 Ponta Grossa, Brazil; (L.A.C.R.); (P.V.F.); (A.L.); (F.M.B.); (T.K.); (J.M.N.); (A.N.); (A.D.L.)
- Correspondence: ; Tel.: +55-42-3220-3124
| | - Amanda Lyra
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmaceutical Sciences, State University of Ponta Grossa, 84030-900 Ponta Grossa, Brazil; (L.A.C.R.); (P.V.F.); (A.L.); (F.M.B.); (T.K.); (J.M.N.); (A.N.); (A.D.L.)
| | - Fernanda Malaquias Barboza
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmaceutical Sciences, State University of Ponta Grossa, 84030-900 Ponta Grossa, Brazil; (L.A.C.R.); (P.V.F.); (A.L.); (F.M.B.); (T.K.); (J.M.N.); (A.N.); (A.D.L.)
| | - Traudi Klein
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmaceutical Sciences, State University of Ponta Grossa, 84030-900 Ponta Grossa, Brazil; (L.A.C.R.); (P.V.F.); (A.L.); (F.M.B.); (T.K.); (J.M.N.); (A.N.); (A.D.L.)
| | - Carla Cristine Kanunfre
- Postgraduate Program in Biomedical Science, Department of General Biology, State University of Ponta Grossa, 84030-900 Ponta Grossa, Brazil;
| | - Jessica Mendes Nadal
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmaceutical Sciences, State University of Ponta Grossa, 84030-900 Ponta Grossa, Brazil; (L.A.C.R.); (P.V.F.); (A.L.); (F.M.B.); (T.K.); (J.M.N.); (A.N.); (A.D.L.)
| | | | - Vijayasankar Raman
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, MS 38677, USA;
| | - Andressa Novatski
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmaceutical Sciences, State University of Ponta Grossa, 84030-900 Ponta Grossa, Brazil; (L.A.C.R.); (P.V.F.); (A.L.); (F.M.B.); (T.K.); (J.M.N.); (A.N.); (A.D.L.)
| | - Alessandro Dourado Loguércio
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmaceutical Sciences, State University of Ponta Grossa, 84030-900 Ponta Grossa, Brazil; (L.A.C.R.); (P.V.F.); (A.L.); (F.M.B.); (T.K.); (J.M.N.); (A.N.); (A.D.L.)
| | - Sandra Maria Warumby Zanin
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmacy, Federal University of Paraná, 81020-430 Curitiba, Brazil;
| |
Collapse
|
111
|
Paramasivan P, Kumar JD, Baskaran R, Weng CF, Padma VV. Reversal of doxorubicin resistance in lung cancer cells by neferine is explained by nuclear factor erythroid-derived 2-like 2 mediated lung resistance protein down regulation. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:647-665. [PMID: 35582448 PMCID: PMC8992493 DOI: 10.20517/cdr.2019.115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/27/2020] [Accepted: 03/16/2020] [Indexed: 05/27/2023]
Abstract
Aim: Development of multi drug resistance and dose limiting cardiotoxicity are hindering the use of Doxorubicin (Dox) in clinical settings. Augmented dox efflux induced by lung resistance protein (LRP) over expression has been related to multi drug resistance phenotype in various cancers. An alkaloid from lotus, Neferine (Nef) shows both anticancer and cardioprotective effects. Here, we have investigated the interconnection between nuclear factor erythroid-derived 2-like 2 (NRF2) and LRP in Dox resistance and how Nef can overcome Dox resistance in lung cancer cells by altering this signaling. Methods: Anti-proliferative and apoptotic-inducing effects of Nef and Dox combination in Parental and Dox resistant lung cancer cells were determined in monolayers and 3D spheroids. Intracellular Dox was analyzed using flow cytometry, siRNA knockdown and western blot analysis were used to elucidate NRF2-LRP crosstalk mechanism. Results: We observed that the Dox resistant lung cancer cells expressed higher levels of LRP, reduced glutathione (GSH) and NRF2. Combination of Dox and Nef induced apoptosis, leads to reactive oxygen species (ROS) generation, GSH depletion and reduction in LRP levels contributing to higher intracellular and intranuclear Dox accumulation. The use of N-acetylcysteine and knockdown studies confirmed an important role of ROS and NRF2 in LRP down regulation. Presence of NRF2 binding sites in LRP is support of direct interaction between LRP and NRF2. Conclusion: Nef sensitizes lung cancer cells to Dox by increasing intracellular and/or intra nuclear Dox accumulation via LRP down regulation. This is mediated by redox regulating NRF2. This decoded crosstalk mechanism reinforces the role of NRF2 and LRP in Dox resistance and as an important anticancer target.
Collapse
Affiliation(s)
- Poornima Paramasivan
- Department of Biotechnology, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
- Laboratory of Molecular Physiology, Institute of Biotechnology, Department of Life Sciences, National Dong Hwa University, Hualien 974, Taiwan
- Division of Science, School of Applied Sciences, University of Abertay Dundee, Dundee DD1 1HG, UK
| | - Jothi Dinesh Kumar
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool L3 5TR, UK
| | - Rathinasamy Baskaran
- Department of Biotechnology, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 41354, Taiwan
| | - Ching Feng Weng
- Laboratory of Molecular Physiology, Institute of Biotechnology, Department of Life Sciences, National Dong Hwa University, Hualien 974, Taiwan
| | - Viswanadha Vijaya Padma
- Department of Biotechnology, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
- Laboratory of Molecular Physiology, Institute of Biotechnology, Department of Life Sciences, National Dong Hwa University, Hualien 974, Taiwan
| |
Collapse
|
112
|
Martínez-Barriocanal Á, Arango D, Dopeso H. PVT1 Long Non-coding RNA in Gastrointestinal Cancer. Front Oncol 2020; 10:38. [PMID: 32083000 PMCID: PMC7005105 DOI: 10.3389/fonc.2020.00038] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 01/09/2020] [Indexed: 12/24/2022] Open
Abstract
Whole genome and transcriptome sequencing technologies have led to the identification of many long non-coding RNAs (lncRNAs) and stimulated the research of their role in health and disease. LncRNAs participate in the regulation of critical signaling pathways including cell growth, motility, apoptosis, and differentiation; and their expression has been found dysregulated in human tumors. Thus, lncRNAs have emerged as new players in the initiation, maintenance and progression of tumorigenesis. PVT1 (plasmacytoma variant translocation 1) lncRNA is located on chromosomal 8q24.21, a large locus frequently amplified in human cancers and predictive of increased cancer risk in genome-wide association studies (GWAS). Combined, colorectal and gastric adenocarcinomas are the most frequent tumor malignancies and also the leading cause of cancer-related deaths worldwide. PVT1 expression is elevated in gastrointestinal tumors and correlates with poor patient prognosis. In this review, we discuss the mechanisms of action underlying PVT1 oncogenic role in colorectal and gastric cancer such as MYC upregulation, miRNA production, competitive endogenous RNA (ceRNA) function, protein stabilization, and epigenetic regulation. We also illustrate the potential role of PVT1 as prognostic biomarker and its relationship with resistance to current chemotherapeutic treatments.
Collapse
Affiliation(s)
- Águeda Martínez-Barriocanal
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron University Hospital, Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.,Group of Molecular Oncology, IRB Lleida, Lleida, Spain
| | - Diego Arango
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron University Hospital, Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.,Group of Molecular Oncology, IRB Lleida, Lleida, Spain
| | - Higinio Dopeso
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron University Hospital, Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
113
|
Deficiency in Dipeptidyl Peptidase-4 Promotes Chemoresistance through the CXCL12/CXCR4/mTOR/TGFβ Signaling Pathway in Breast Cancer Cells. Int J Mol Sci 2020; 21:ijms21030805. [PMID: 31991851 PMCID: PMC7037814 DOI: 10.3390/ijms21030805] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/21/2020] [Accepted: 01/24/2020] [Indexed: 02/06/2023] Open
Abstract
Dipeptidyl peptidase (DPP)-4, a molecular target of DPP-4 inhibitors, which are type 2 diabetes drugs, is expressed in a variety of cell types, tissues and organs. DPP-4 has been shown to be involved in cancer biology, and we have recently shown that a DPP-4 inhibitor promoted the epithelial mesenchymal transition (EMT) in breast cancer cells. The EMT is known to associate with chemotherapy resistance via the induction of ATP-binding cassette (ABC) transporters in cancer cells. Here, we demonstrated that deficiency in DPP-4 promoted chemotherapy resistance via the CXCL12/CXCR4/mTOR axis, activating the TGFβ signaling pathway via the expression of ABC transporters. DPP-4 inhibition enhanced ABC transporters in vivo and in vitro. Doxorubicin (DOX) further induced ABC transporters in DPP-4-deficient 4T1 cells, and the induction of ABC transporters was suppressed by either the CXCR4 inhibitor AMD3100, the mTOR inhibitor rapamycin or a neutralizing TGFβ (1, 2 and 3) antibody(N-TGFβ). Knockdown of snail, an EMT-inducible transcription factor, suppressed ABC transporter levels in DOX-treated DPP-4-deficient 4T1 cells. In an allograft mouse model, however, the effects of DOX in either primary tumor or metastasis were not statistically different between control and DPP-4-kd 4T1. Taken together, our findings suggest that DPP-4 inhibitors potentiate chemotherapy resistance via the induction of ABC transporters by the CXCL12/CXCR4/mTOR/TGFβ signaling pathway in breast cancer cells.
Collapse
|
114
|
Hill C, Wang Y. The importance of epithelial-mesenchymal transition and autophagy in cancer drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:38-47. [PMID: 32226927 PMCID: PMC7100899 DOI: 10.20517/cdr.2019.75] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/13/2019] [Accepted: 12/13/2019] [Indexed: 12/18/2022]
Abstract
Epithelial-mesenchymal transition (EMT) and autophagy are both known to play an important role in the development of cancer. Subsequently, these processes are now being utilised as targets for therapy. Cancer is globally one of the leading causes of death, and despite many advances in treatment options, patients still face many challenges. Drug-resistance in cancer-therapy is a large problem, and both EMT and autophagy have been shown to contribute. However, given the context-dependent role of these processes and the complexity of the interactions between them, elucidating how they both act alone and interact together is important. In this review, we will provide an insight into the current landscape of the interactions of autophagy and EMT in the context of malignancy, and how this ultimately may affect drug-resistance in cancer-therapy.
Collapse
Affiliation(s)
- Charlotte Hill
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Yihua Wang
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| |
Collapse
|
115
|
Fu Z, Agudelo P, Wells CE. Detoxification-related gene expression accompanies anhydrobiosis in the foliar nematode ( Aphelenchoides fragariae). J Nematol 2020; 52:1-12. [PMID: 32449331 PMCID: PMC7266049 DOI: 10.21307/jofnem-2020-047] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Indexed: 12/19/2022] Open
Abstract
The foliar nematode (Aphelenchoides fragariae) is a quarantined pest that infects a broad range of herbaceous and woody plants. Previous work has demonstrated its remarkable ability to survive rapid and extreme desiccation, although the specific molecular mechanisms underlying its anhydrobiotic response have not been characterized. The authors used RNA sequencing and de novo transcriptome assembly to compare patterns of gene expression between hydrated and 24-hr desiccated nematodes. In total, 2,083 and 953 genes were significantly up- and downregulated, respectively, in desiccated nematodes. Of the 100 annotated genes with the largest positive fold-changes, more than one third encoded putative detoxification-related proteins. Genes encoding enzymes of Phase I and Phase II detoxification systems were among the most strongly upregulated in the transcriptome, including 35 cytochrome p450s, 23 short chain dehydrogenase/reductases, 5 glutathione-S-transferases, and 22 UDP-glucuronosyltransferases. Genes encoding heat shock proteins, unfolded protein response enzymes, and intrinsically disordered proteins were also upregulated. Anhydrobiosis in A. fragariae appears to involve both strategies to minimize protein misfolding and aggregation, and wholesale induction of the cellular detoxification machinery. These processes may be controlled in part through the activity of forkhead transcription factors similar to Caenorhabditis elegans’ daf-16, a number of which were differentially expressed under desiccation.
Collapse
Affiliation(s)
- Zhen Fu
- School of Agricultural, Forest, and Environmental Sciences , Clemson University , Clemson, SC, 29634 ; Department of Entomology , Washington State University , Pullman, WA, 99164
| | - Paula Agudelo
- School of Agricultural, Forest, and Environmental Sciences , Clemson University , Clemson, SC, 29634
| | - Christina E Wells
- Department of Biological Sciences , Clemson University , Clemson, SC, 29634
| |
Collapse
|
116
|
Old wine in new bottles: Drug repurposing in oncology. Eur J Pharmacol 2020; 866:172784. [DOI: 10.1016/j.ejphar.2019.172784] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 11/05/2019] [Accepted: 11/07/2019] [Indexed: 02/07/2023]
|
117
|
Valinezhad Sani F, Palizban A, Mosaffa F, Jamialahmadi K. Glucosamine reverses drug resistance in MRP2 overexpressing ovarian cancer cells. Eur J Pharmacol 2019; 868:172883. [PMID: 31866406 DOI: 10.1016/j.ejphar.2019.172883] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 12/13/2019] [Accepted: 12/17/2019] [Indexed: 01/08/2023]
Abstract
Glucosamine (GlcN), a natural amino sugar in human body, was reported to exhibit anticancer activity against some tumors. In the present study, we evaluated the cytotoxicity and multi-drug resistance (MDR) reversal activity of GlcN on resistant MRP2-overexpressing ovarian cancer A2780RCIS cells. The cytotoxicity and MDR reversal activity of GlcN on cancer cells were measured by MTT assay. The effects of GlcN on MRP1 and MRP2 mRNA expression and function were evaluated by qRT-PCR and flow cytometry, respectively. The cell migration capacity of ovarian cancer cells were assessed in the presence or absence of GlcN using wound healing migration assay. Furthermore, the effects of GlcN on the mRNA expression of E-cadherin, vimentin and α-smooth muscle actin as Epithelial-Mesenchymal Transition (EMT)-related markers were evaluated by qRT-PCR. Our results indicated that glucosamine reduced the proliferation of human ovarian cancer cell lines (A2780) and its cisplatin resistant variant (A2780RCIS) in a dose-dependent manner. The IC50 values for A2780RCIS cells treated with cisplatin in the presence of different concentrations of GlcN (0, 1, 2 and 3 mM) for 72 h were 44.463 ± 1.603, 35.17 ± 0.025, 22.25 ± 0.018, 17.78 ± 0.012 μM respectively. Also GlcN decreased the expression of MRP1 and MRP2 mRNA in ovarian cancer cells. Our results further demonstrated that although GlcN had no significant effects on the expression of studied EMT-related markers in invasive A2780RCIS cells, it was able to inhibit their migration in vitro. According to these findings, GlcN could effectively enhance cisplatin cytotoxicity in resistant A2780RCIS cells.
Collapse
Affiliation(s)
- Fatemeh Valinezhad Sani
- Department of Clinical Biochemistry, Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abbasali Palizban
- Department of Clinical Biochemistry, Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Mosaffa
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology(,) School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Khadijeh Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
118
|
Palmer AC, Chidley C, Sorger PK. A curative combination cancer therapy achieves high fractional cell killing through low cross-resistance and drug additivity. eLife 2019; 8:50036. [PMID: 31742555 PMCID: PMC6897534 DOI: 10.7554/elife.50036] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022] Open
Abstract
Curative cancer therapies are uncommon and nearly always involve multi-drug combinations developed by experimentation in humans; unfortunately, the mechanistic basis for the success of such combinations has rarely been investigated in detail, obscuring lessons learned. Here, we use isobologram analysis to score pharmacological interaction, and clone tracing and CRISPR screening to measure cross-resistance among the five drugs comprising R-CHOP, a combination therapy that frequently cures Diffuse Large B-Cell Lymphomas. We find that drugs in R-CHOP exhibit very low cross-resistance but not synergistic interaction: together they achieve a greater fractional kill according to the null hypothesis for both the Loewe dose-additivity model and the Bliss effect-independence model. These data provide direct evidence for the 50 year old hypothesis that a curative cancer therapy can be constructed on the basis of independently effective drugs having non-overlapping mechanisms of resistance, without synergistic interaction, which has immediate significance for the design of new drug combinations.
Collapse
Affiliation(s)
- Adam C Palmer
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, United States
| | - Christopher Chidley
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, United States
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, United States.,Department of Systems Biology, Harvard Medical School, Boston, United States
| |
Collapse
|
119
|
Chen W, Liu I, Tomiyasu H, Lee J, Cheng C, Liao AT, Liu B, Liu C, Lin C. Imatinib enhances the anti-tumour effect of doxorubicin in canine B-cell lymphoma cell line. Vet J 2019; 254:105398. [PMID: 31836165 DOI: 10.1016/j.tvjl.2019.105398] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 10/03/2019] [Accepted: 10/19/2019] [Indexed: 01/24/2023]
Abstract
Canine lymphoma is one of the most common malignant tumours occurring in dogs and has a high incidence worldwide. Despite advances in cancer prevention, the treatment of neoplastic diseases still requires improvement. Some cancer cells may resist the effect of chemotherapeutic agents by up-regulating drug transporters leading to increased drug efflux, resulting in intrinsic or acquired drug resistance, which is a mechanism commonly seen in doxorubicin-resistant tumour cells. In this study, canine B-cell lymphoma cell line CLBL1-8.0, a doxorubicin-resistant B cell lymphoma cell line derived from CLBL-1 by increasing the doxorubicin concentration during culturing, exhibited high expression of P-glycoprotein (P-gp, ATP-binding cassette sub-family B member 1 [ABCB1]). These proteins are commonly involved in cancer cell resistance to doxorubicin. Imatinib, a tyrosine kinase inhibitor significantly potentiated the sensitivity of doxorubicin in P-gp-overexpressing doxorubicin-resistant cells. Moreover, a combination of these two drugs may increase the retention of doxorubicin by decreasing the efflux of doxorubicin without affecting P-gp protein overexpression. In conclusion, imatinib reversed doxorubicin resistance by decreasing drug efflux in P-gp-overexpressing doxorubicin-resistant canine lymphoma cells. These results suggest that combining doxorubicin, one of the most widely used chemotherapeutic drugs in the treatment of canine lymphoma, with imatinib might potentially overcome doxorubicin resistance in a clinical setting.
Collapse
Affiliation(s)
- W Chen
- Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, 1 Sec 4 Roosevelt Road, Taipei, 10617, Taiwan
| | - I Liu
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, 1 Sec 4 Roosevelt Road, Taipei, 10617, Taiwan
| | - H Tomiyasu
- Department of Veterinary Internal Medicine, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - J Lee
- Graduate Institute of Veterinary Clinical Science, School of Veterinary Medicine, National Taiwan University, 1 Sec 4 Roosevelt Road, Taipei, 10617, Taiwan
| | - C Cheng
- Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, 1 Sec 4 Roosevelt Road, Taipei, 10617, Taiwan
| | - A T Liao
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, 1 Sec 4 Roosevelt Road, Taipei, 10617, Taiwan
| | - B Liu
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, 1 Sec 4 Roosevelt Road, Taipei, 10617, Taiwan
| | - C Liu
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, 1 Sec 4 Roosevelt Road, Taipei, 10617, Taiwan
| | - C Lin
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, 1 Sec 4 Roosevelt Road, Taipei, 10617, Taiwan.
| |
Collapse
|
120
|
Dante RAS, Ferrer RJE, Jacinto SD. Leaf Extracts from Dillenia philippinensis Rolfe Exhibit Cytotoxic Activity to both Drug-Sensitive and Multidrug-Resistant Cancer Cells. Asian Pac J Cancer Prev 2019; 20:3285-3290. [PMID: 31759350 PMCID: PMC7063009 DOI: 10.31557/apjcp.2019.20.11.3285] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Indexed: 01/10/2023] Open
Abstract
Background: Cancer is one of the leading causes of illness and death worldwide. Only palliative therapeutic options are available for many types of cancers, and most anticancer drugs in clinical use exhibit significant side effects. It is therefore important to develop new anticancer drugs that are more effective and less toxic. In this study, we evaluate the bioactivity of a Philippine endemic plant, “katmon” or Dillenia philippinensis, and its potential use in cancer therapy. Methods: The cytotoxicity of the crude leaf extract, partitions, and isocratic column chromatography fractions of Dillenia philippinensis was determined in vitro by MTT assay against drug-sensitive cancer cell lines MCF7 (human breast adenocarcinoma) and HCT 116 (human colorectal carcinoma), as well as against moderately multidrug resistant (MDR) cancer cell line HCT-15 (human colorectal carcinoma) and its highly MDR subline HCT-15/Dox. The selectivity of the extract to cancer cells was determined by computing for the selectivity index (SI) with respect to normal mouse embryonic fibroblasts (NIH/3T3) cell line. To check for a possible mechanism for overcoming cancer multiple drug resistance, Calcein-AM assay was performed to assess the activity of the extract against P-glycoprotein-activated efflux pump. Results: Dillenia philippinensis (DP1) fraction from the hexane partition exhibited cytotoxicity (IC50< 30 µg/ml) against MCF7, HCT 116, HCT-15, and HCT-15/Dox cells. DP1 also exhibited a moderate level of selectivity against cancer cells over normal cells as supported by the SI computed from the IC50 value obtained for the normal cell line. DP1 was able to inhibit P-glycoprotein (P-gp) activity in a dose-dependent manner, suggesting its possible role in targeting cancer cells with overexpressed P-gp. Conclusion: The present findings thus demonstrate the potential chemotherapeutic properties of D. philippinensis which can be promising for future drug development against cancer.
Collapse
Affiliation(s)
- Rachelle Anne S Dante
- Institute of Biology, University of the Philippines, Diliman, Quezon City, Philippines
| | - Regina Joyce E Ferrer
- Institute of Biology, University of the Philippines, Diliman, Quezon City, Philippines
| | - Sonia D Jacinto
- Institute of Biology, University of the Philippines, Diliman, Quezon City, Philippines
| |
Collapse
|
121
|
Sanchez-Carranza JN, González-Maya L, Razo-Hernández RS, Salas-Vidal E, Nolasco-Quintana NY, Clemente-Soto AF, García-Arizmendi L, Sánchez-Ramos M, Marquina S, Alvarez L. Achillin Increases Chemosensitivity to Paclitaxel, Overcoming Resistance and Enhancing Apoptosis in Human Hepatocellular Carcinoma Cell Line Resistant to Paclitaxel (Hep3B/PTX). Pharmaceutics 2019; 11:pharmaceutics11100512. [PMID: 31590262 PMCID: PMC6835644 DOI: 10.3390/pharmaceutics11100512] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/28/2019] [Accepted: 09/28/2019] [Indexed: 12/16/2022] Open
Abstract
Multidrug resistance (MDR) has become a major obstacle in the treatment of cancer, and is associated with mechanisms such as increased drug outflow, reduction of apoptosis, and/or altered drug metabolism. These problems can be mitigated by the coadministration of agents known as chemosensitizers, as they can reverse resistance to anticancer drugs and eventually resensitize cancer cells. We explore the chemosensitizing effect of Achillin, a guaianolide-type sesquiterpene lactone isolated from the Mexican medicinal plant Artemisia ludovisiana, to reverse MDR in Hep3B/PTX cells of hepatocellular carcinoma, which present resistance to paclitaxel (PTX). Achillin showed an important effect as chemosensitizer; indeed, the cytotoxic effect of PTX (25 nM) was enhanced, and the induction of G2/M phase cell cycle arrest and apoptosis were potentiated when combining with Achillin (100 μM). In addition, we observed that Achillin decreases P-gp levels and increases the intracellular retention of doxorubicin in Hep3B/PTX cells; in addition, homology structural modeling and molecular docking calculations predicted that Achillin interacts in two regions (M-site and R-site) of transporter drug efflux P-glycoprotein (P-gp). Our results suggest that the chemosensitizer effect demonstrated for Achillin could be associated with P-gp modulation. This work also provides useful information for the development of new therapeutic agents from guaianolide-type sesquiterpene lactones like Achillin.
Collapse
Affiliation(s)
- Jessica Nayelli Sanchez-Carranza
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa C.P. 62209, Cuernavaca, Mexico.
- Centro de Investigaciones Químicas-IICBA, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa C.P. 62209, Cuernavaca, Mexico.
| | - Leticia González-Maya
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa C.P. 62209, Cuernavaca, Mexico.
| | - Rodrigo Said Razo-Hernández
- Centro de Investigación en Dinámica Celular-IICBA, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Chamilpa, Cuernavaca 62209, Morelos, Mexico.
| | - Enrique Salas-Vidal
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca C.P. 62209, Morelos, Mexico.
| | - Ninfa Yaret Nolasco-Quintana
- Centro de Investigaciones Químicas-IICBA, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa C.P. 62209, Cuernavaca, Mexico.
- Centro de Investigación en Dinámica Celular-IICBA, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Chamilpa, Cuernavaca 62209, Morelos, Mexico.
| | - Aldo F Clemente-Soto
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa C.P. 62209, Cuernavaca, Mexico.
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa. Av. de las Américas y Blvd. Universitarios S/N, Culiacán 80010, Sinaloa, Mexico.
| | - Lucero García-Arizmendi
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa C.P. 62209, Cuernavaca, Mexico.
| | - Mariana Sánchez-Ramos
- Centro de Investigaciones Químicas-IICBA, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa C.P. 62209, Cuernavaca, Mexico.
| | - Silvia Marquina
- Centro de Investigaciones Químicas-IICBA, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa C.P. 62209, Cuernavaca, Mexico.
| | - Laura Alvarez
- Centro de Investigaciones Químicas-IICBA, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa C.P. 62209, Cuernavaca, Mexico.
| |
Collapse
|
122
|
Amawi H, Sim HM, Tiwari AK, Ambudkar SV, Shukla S. ABC Transporter-Mediated Multidrug-Resistant Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1141:549-580. [PMID: 31571174 DOI: 10.1007/978-981-13-7647-4_12] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
ATP-binding cassette (ABC) transporters are involved in active pumping of many diverse substrates through the cellular membrane. The transport mediated by these proteins modulates the pharmacokinetics of many drugs and xenobiotics. These transporters are involved in the pathogenesis of several human diseases. The overexpression of certain transporters by cancer cells has been identified as a key factor in the development of resistance to chemotherapeutic agents. In this chapter, the localization of ABC transporters in the human body, their physiological roles, and their roles in the development of multidrug resistance (MDR) are reviewed. Specifically, P-glycoprotein (P-GP), multidrug resistance-associated proteins (MRPs), and breast cancer resistance protein (BCRP/ABCG2) are described in more detail. The potential of ABC transporters as therapeutic targets to overcome MDR and strategies for this purpose are discussed as well as various explanations for the lack of efficacy of ABC drug transporter inhibitors to increase the efficiency of chemotherapy.
Collapse
Affiliation(s)
- Haneen Amawi
- Department of Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Hong-May Sim
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Suresh V Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Suneet Shukla
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
123
|
Basu R, Kopchick JJ. The effects of growth hormone on therapy resistance in cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:827-846. [PMID: 32382711 PMCID: PMC7204541 DOI: 10.20517/cdr.2019.27] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pituitary derived and peripherally produced growth hormone (GH) is a crucial mediator of longitudinal growth, organ development, metabolic regulation with tissue specific, sex specific, and age-dependent effects. GH and its cognate receptor (GHR) are expressed in several forms of cancer and have been validated as an anti-cancer target through a large body of in vitro, in vivo and epidemiological analyses. However, the underlying molecular mechanisms of GH action in cancer prognosis and therapeutic response had been sparse until recently. This review assimilates the critical details of GH-GHR mediated therapy resistance across different cancer types, distilling the therapeutic implications based on our current understanding of these effects.
Collapse
Affiliation(s)
- Reetobrata Basu
- Ohio University Heritage College of Osteopathic Medicine (OU-HCOM), Ohio University, Athens, OH 45701, USA.,Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| | - John J Kopchick
- Ohio University Heritage College of Osteopathic Medicine (OU-HCOM), Ohio University, Athens, OH 45701, USA.,Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
124
|
Duan H, Yang Z, Liang L, Zhou X. CA916798 gene expression is associated with multidrug resistance and predicts progression-free survival in patients with lung cancer. Oncol Lett 2019; 18:1171-1178. [PMID: 31423177 PMCID: PMC6607038 DOI: 10.3892/ol.2019.10436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 05/03/2019] [Indexed: 11/06/2022] Open
Abstract
CA916798 has been identified as a novel multidrug resistance gene in lung cancer cells. However, the expression patterns of CA916798 in tumor tissues prior and subsequent to chemotherapy remain unclear. In the present study, CA916798 expression levels in tumor tissues prior and subsequent to chemotherapy were detected by reverse transcription-quantitative polymerase chain reaction and immunohistochemistry analysis. The prognostic significance of CA916798 expression in tumor tissues was explored by Kaplan-Meier survival analysis and Cox proportional hazards regression analysis. The messenger RNA (mRNA) and protein expression levels of CA916798 in tumor tissues were downregulated post-chemotherapy in chemotherapy-sensitive patients with lung cancer, but not in chemotherapy-resistant patients. Downregulation of CA916798 mRNA and protein expression post-chemotherapy was significantly associated with improved progression-free survival time. The findings from the present study suggest that platinum-based chemotherapy may induce the expression of CA916798, and CA916798 may be a promising biomarker to predict chemotherapy resistance and improve therapies for patients with lung cancer.
Collapse
Affiliation(s)
- Hailing Duan
- Department of Respiratory Disease, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, P.R. China
| | - Zaixing Yang
- Department of Respiratory Disease, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, P.R. China
| | - Lan Liang
- Department of Respiratory Disease, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, P.R. China
| | - Xiangdong Zhou
- Department of Respiratory Disease, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, P.R. China
| |
Collapse
|
125
|
Aghamiri S, Mehrjardi KF, Shabani S, Keshavarz-Fathi M, Kargar S, Rezaei N. Nanoparticle-siRNA: a potential strategy for ovarian cancer therapy? Nanomedicine (Lond) 2019; 14:2083-2100. [PMID: 31368405 DOI: 10.2217/nnm-2018-0379] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Ovarian cancer is one of the most common causes of mortality throughout the world. Unfortunately, chemotherapy has failed to cure advanced cancers developing multidrug resistance (MDR). Moreover, it has critical side effects because of nonspecific toxicity. Thanks to specific silencing of oncogenes and MDR-associated genes, nano-siRNA drugs can be a great help address the limitations of chemotherapy. Here, we review the current advances in nanoparticle-mediated siRNA delivery strategies such as polymeric- and lipid-based systems, rigid nanoparticles and nanoparticles coupled to specific ligand systems. Nanoparticle-based codelivery of anticancer drugs and siRNA targeting various mechanisms of MDR is a cutting-edge strategy for ovarian cancer therapy, which is completely discussed in this review.
Collapse
Affiliation(s)
- Shahin Aghamiri
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technology in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 19839-63113, Iran
| | - Keyvan Fallah Mehrjardi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, 1416753955, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Tehran, 1419733151, Iran
| | - Sasan Shabani
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
| | - Mahsa Keshavarz-Fathi
- Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Tehran, 1419733151, Iran.,Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
| | - Saeed Kargar
- Department of Life Science Engineering, Faculty of New Sciences & Technologies, University of Tehran, Tehran, 1417466191, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, 1419733151, Iran
| |
Collapse
|
126
|
Singh H, Das S, Yadav J, Srivastava VK, Jyoti A, Kaushik S. In search of novel protein drug targets for treatment of Enterococcus faecalis infections. Chem Biol Drug Des 2019; 94:1721-1739. [PMID: 31260188 DOI: 10.1111/cbdd.13582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 06/08/2019] [Accepted: 06/17/2019] [Indexed: 12/27/2022]
Abstract
Enterococcus faecalis (Ef) is one of the major pathogens involved in hospital-acquired infections. It can cause nosocomial bacteremia, surgical wound infection, and urinary tract infection. It is important to mention here that Ef is developing resistance against many commonly occurring antibiotics. The occurrence of multidrug resistance (MDR) and extensive-drug resistance (XDR) is now posing a major challenge to the medical community. In this regard, to combat the infections caused by Ef, we have to look for an alternative. Rational structure-based drug design exploits the three-dimensional structure of the target protein, which can be unraveled by various techniques such as X-ray crystallography or nuclear magnetic resonance (NMR) spectroscopy. In this review, we have discussed the complete picture of Ef infections, the possible treatment available at present, and the alternative treatment options to be explored. This study will help in better understanding of novel biological targets against Ef and the compounds, which are likely to bind with these targets. Using these detailed structural informations, rational structure-based drug design is achievable and tight inhibitors against Ef can be prepared.
Collapse
Affiliation(s)
- Harpreet Singh
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | - Satyajeet Das
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | - Jyoti Yadav
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | | | - Anupam Jyoti
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | - Sanket Kaushik
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| |
Collapse
|
127
|
Kumar S, Kushwaha PP, Gupta S. Emerging targets in cancer drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:161-177. [PMID: 35582722 PMCID: PMC8992633 DOI: 10.20517/cdr.2018.27] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/08/2019] [Accepted: 03/14/2019] [Indexed: 02/05/2023]
Abstract
Drug resistance is a complex phenomenon that frequently develops as a failure to chemotherapy during cancer treatment. Malignant cells increasingly generate resistance to various chemotherapeutic drugs through distinct mechanisms and pathways. Understanding the molecular mechanisms involved in drug resistance remains an important area of research for identification of precise targets and drug discovery to improve therapeutic outcomes. This review highlights the role of some recent emerging targets and pathways which play critical role in driving drug resistance.
Collapse
Affiliation(s)
- Shashank Kumar
- School of Basic and Applied Sciences, Department of Biochemistry and Microbial Sciences, Central University of Punjab, Bathinda 151001, India
| | - Prem Prakash Kushwaha
- School of Basic and Applied Sciences, Department of Biochemistry and Microbial Sciences, Central University of Punjab, Bathinda 151001, India
| | - Sanjay Gupta
- Department of Urology, Case Western Reserve University, Cleveland, Ohio 44106, USA.,The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio 44106, USA.,Department of Nutrition, Case Western Reserve University, Cleveland, Ohio 44106, USA.,Divison of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, Ohio 44106, USA.,Department of Urology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio 44106, USA
| |
Collapse
|
128
|
Methanol Extract of Aerial Parts of Pavetta indica L. Enhances the Cytotoxic Effect of Doxorubicin and Induces Radiation Sensitization in MDA-MB-231 Triple-Negative Breast Cancer Cells. Molecules 2019; 24:molecules24122273. [PMID: 31216782 PMCID: PMC6631732 DOI: 10.3390/molecules24122273] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/13/2019] [Accepted: 06/18/2019] [Indexed: 12/13/2022] Open
Abstract
Pavetta indica L. is used in traditional medicine for the treatment of various diseases including hemorrhoids, headache, urinary conditions, ulcerated nose, and dropsy. However, no study has evaluated the anticancer effect of P. indica L. In this study, we found that a methanol extract of the leaves and branches of P. indica L. (MEPI) caused cellcycle arrest at the sub-G1 phase and induced apoptosis, as indicated by the activation of caspase-8, -3, -7, and c-PARP. Western blotting revealed that MEPI significantly reduced the levels of markers of the epithelial-mesenchymal transition, such as Vimentin, Snail, Slug, and matrix metallopeptidase 9. Notably, the expression of multidrug resistance-associated protein 1 in triple negative breast cancer (TNBC) was significantly decreased by MEPI. Moreover, the co-treatment with MEPI and doxorubicin resulted in a synergistic reduction in cell viability. MEPI also induced radiation sensitization of TNBC cells. Gas chromatography-mass spectrometry analysis revealed that 5,6-dehydrokawain (DK) is the major constituent of MEPI. Interestingly, DK exerted significant anti-invasive and anti-metastatic effects. Our results provide a strong rationale for investigating the molecular mechanisms of action of MEPI in TNBC.
Collapse
|
129
|
Reshma PL, Unnikrishnan BS, Preethi GU, Syama HP, Archana MG, Remya K, Shiji R, Sreekutty J, Sreelekha TT. Overcoming drug-resistance in lung cancer cells by paclitaxel loaded galactoxyloglucan nanoparticles. Int J Biol Macromol 2019; 136:266-274. [PMID: 31201909 DOI: 10.1016/j.ijbiomac.2019.06.075] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 06/04/2019] [Accepted: 06/11/2019] [Indexed: 12/16/2022]
Abstract
Paclitaxel, an effective chemotherapeutic drug, is insoluble in aqueous solvents and is usually administered with excipients which have side effects. The use of this drug is also limited due to multi-drug resistance. In this study polysaccharide nanoparticles are used in the delivery of chemotherapeutic drug while minimizing side-effects, solubility issues and drug resistance. The use of biopolymers like galactoxyloglucan to synthesize nanoparticle makes it more biocompatible. This study involves the synthesis of PST-PTX nanoparticles using tamarind seed polysaccharide and Paclitaxel by epichlorohydrin crosslinking. The particles were further characterized by Dynamic Light Scattering (DLS), High-resolution transmission electron microscopy (HR-TEM) Fourier Transform Infrared Spectroscopy (FTIR) and UV-Visible spectroscopy. The cytotoxicity of PST-PTX nanoparticles in cancer cell lines and resistant cancer cell lines were determined by MTT assay. The quantitative analysis of cell death was determined by Annexin V dead cell assay, Caspase 3/7 assay and expression of pro-apoptotic protein Bax. The ability of the nanoparticle to overcome multi-drug resistance was evaluated by the expression of multidrug-resistant proteins P-glycoprotein (P-gp) and Breast cancer resistant protein (BCRP) in lung adenocarcinoma resistant cells (A549R). The present study provides evidence for the ability of PST-PTX nanoparticle to overcome multi-drug resistance and cause apoptotic cell death. The particle was found to be more effective than Paclitaxel in causing cell death in resistant cancer cells. Moreover, the particles were found to downregulate the expression of multi-drug resistant proteins P-gp and BCRP in resistant cell lines suggesting the ability of PST-PTX nanoparticles to overcome multi-drug resistance.
Collapse
Affiliation(s)
- P L Reshma
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research, Regional Cancer Centre, Thiruvananthapuram 695011, India
| | - B S Unnikrishnan
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research, Regional Cancer Centre, Thiruvananthapuram 695011, India
| | - G U Preethi
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research, Regional Cancer Centre, Thiruvananthapuram 695011, India
| | - H P Syama
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research, Regional Cancer Centre, Thiruvananthapuram 695011, India
| | - M G Archana
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research, Regional Cancer Centre, Thiruvananthapuram 695011, India
| | - K Remya
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research, Regional Cancer Centre, Thiruvananthapuram 695011, India
| | - R Shiji
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research, Regional Cancer Centre, Thiruvananthapuram 695011, India
| | - J Sreekutty
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research, Regional Cancer Centre, Thiruvananthapuram 695011, India
| | - T T Sreelekha
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research, Regional Cancer Centre, Thiruvananthapuram 695011, India.
| |
Collapse
|
130
|
Hanna J, Hossain GS, Kocerha J. The Potential for microRNA Therapeutics and Clinical Research. Front Genet 2019; 10:478. [PMID: 31156715 PMCID: PMC6532434 DOI: 10.3389/fgene.2019.00478] [Citation(s) in RCA: 519] [Impact Index Per Article: 103.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 05/03/2019] [Indexed: 12/15/2022] Open
Abstract
As FDA-approved small RNA drugs start to enter clinical medicine, ongoing studies for the microRNA (miRNA) class of small RNAs expand its preclinical and clinical research applications. A growing number of reports suggest a significant utility of miRNAs as biomarkers for pathogenic conditions, modulators of drug resistance, and/or as drugs for medical intervention in almost all human health conditions. The pleiotropic nature of this class of nonprotein-coding RNAs makes them particularly attractive drug targets for diseases with a multifactorial origin and no current effective treatments. As candidate miRNAs begin to proceed toward initiation and completion of potential phase 3 and 4 trials in the future, the landscape of both diagnostic and interventional medicine will arguably continue to evolve. In this mini-review, we discuss miRNA drug discovery development and their current status in clinical trials.
Collapse
Affiliation(s)
- Johora Hanna
- Nova Clinical Research, LLC, Bradenton, FL, United States
| | - Gazi S Hossain
- Nova Clinical Research, LLC, Bradenton, FL, United States
| | - Jannet Kocerha
- Department of Chemistry and Biochemistry, Georgia Southern University, Statesboro, GA, United States
| |
Collapse
|
131
|
Carozzo A, Yaneff A, Gómez N, Di Siervi N, Sahores A, Diez F, Attorresi AI, Rodríguez-González Á, Monczor F, Fernández N, Abba M, Shayo C, Davio C. Identification of MRP4/ABCC4 as a Target for Reducing the Proliferation of Pancreatic Ductal Adenocarcinoma Cells by Modulating the cAMP Efflux. Mol Pharmacol 2019; 96:13-25. [PMID: 31043460 DOI: 10.1124/mol.118.115444] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 04/17/2019] [Indexed: 12/30/2022] Open
Abstract
Pancreatic cancer is one of the most lethal types of tumors with no effective therapy available; is currently the third leading cause of cancer in developed countries; and is predicted to become the second deadliest cancer in the United States by 2030. Due to the marginal benefits of current standard chemotherapy, the identification of new therapeutic targets is greatly required. Considering that cAMP pathway is commonly activated in pancreatic ductal adenocarcinoma (PDAC) and its premalignant lesions, we aim to investigate the multidrug resistance-associated protein 4 (MRP4)-dependent cAMP extrusion process as a cause of increased cell proliferation in human PDAC cell lines. Our results from in silico analysis indicate that MRP4 expression may influence PDAC patient outcome; thus, high MRP4 levels could be indicators of poor survival. In addition, we performed in vitro experiments and identified an association between higher MRP4 expression levels and more undifferentiated and malignant models of PDAC and cAMP extrusion capacity. We studied the antiproliferative effect and the overall cAMP response of three MRP4 inhibitors, probenecid, MK571, and ceefourin-1 in PDAC in vitro models. Moreover, MRP4-specific silencing in PANC-1 cells reduced cell proliferation (P < 0.05), whereas MRP4 overexpression in BxPC-3 cells significantly incremented their growth rate in culture (P < 0.05). MRP4 pharmacological inhibition or silencing abrogated cell proliferation through the activation of the cAMP/Epac/Rap1 signaling pathway. Also, extracellular cAMP reverted the antiproliferative effect of MRP4 blockade. Our data highlight the MRP4-dependent cAMP extrusion process as a key participant in cell proliferation, indicating that MRP4 could be an exploitable therapeutic target for PDAC. SIGNIFICANCE STATEMENT: ABCC4/MRP4 is the main transporter responsible for cAMP efflux. In this work, we show that MRP4 expression may influence PDAC patient outcome and identify an association between higher MRP4 expression levels and more undifferentiated and malignant in vitro models of PDAC. Findings prove the involvement of MRP4 in PDAC cell proliferation through a novel extracellular cAMP mitogenic pathway and further support MRP4 inhibition as a promising therapeutic strategy for PDAC treatment.
Collapse
Affiliation(s)
- Alejandro Carozzo
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina (A.C., A.Y., N.G., N.D.S., A.S., F.D., F.M., N.F., C.D.); IBioBA MPSP - Instituto de Investigaciones en Biomedicina de Buenos Aires, CONICET, Instituto Partner de la Sociedad Max Planck, Buenos Aires, Argentina (A.I.A.); Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina (Á.R.-G., C.S.); and Centro de Investigaciones Inmunológicas Básicas y Aplicadas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Buenos Aires, Argentina (M.A.)
| | - Agustín Yaneff
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina (A.C., A.Y., N.G., N.D.S., A.S., F.D., F.M., N.F., C.D.); IBioBA MPSP - Instituto de Investigaciones en Biomedicina de Buenos Aires, CONICET, Instituto Partner de la Sociedad Max Planck, Buenos Aires, Argentina (A.I.A.); Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina (Á.R.-G., C.S.); and Centro de Investigaciones Inmunológicas Básicas y Aplicadas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Buenos Aires, Argentina (M.A.)
| | - Natalia Gómez
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina (A.C., A.Y., N.G., N.D.S., A.S., F.D., F.M., N.F., C.D.); IBioBA MPSP - Instituto de Investigaciones en Biomedicina de Buenos Aires, CONICET, Instituto Partner de la Sociedad Max Planck, Buenos Aires, Argentina (A.I.A.); Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina (Á.R.-G., C.S.); and Centro de Investigaciones Inmunológicas Básicas y Aplicadas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Buenos Aires, Argentina (M.A.)
| | - Nicolás Di Siervi
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina (A.C., A.Y., N.G., N.D.S., A.S., F.D., F.M., N.F., C.D.); IBioBA MPSP - Instituto de Investigaciones en Biomedicina de Buenos Aires, CONICET, Instituto Partner de la Sociedad Max Planck, Buenos Aires, Argentina (A.I.A.); Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina (Á.R.-G., C.S.); and Centro de Investigaciones Inmunológicas Básicas y Aplicadas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Buenos Aires, Argentina (M.A.)
| | - Ana Sahores
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina (A.C., A.Y., N.G., N.D.S., A.S., F.D., F.M., N.F., C.D.); IBioBA MPSP - Instituto de Investigaciones en Biomedicina de Buenos Aires, CONICET, Instituto Partner de la Sociedad Max Planck, Buenos Aires, Argentina (A.I.A.); Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina (Á.R.-G., C.S.); and Centro de Investigaciones Inmunológicas Básicas y Aplicadas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Buenos Aires, Argentina (M.A.)
| | - Federico Diez
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina (A.C., A.Y., N.G., N.D.S., A.S., F.D., F.M., N.F., C.D.); IBioBA MPSP - Instituto de Investigaciones en Biomedicina de Buenos Aires, CONICET, Instituto Partner de la Sociedad Max Planck, Buenos Aires, Argentina (A.I.A.); Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina (Á.R.-G., C.S.); and Centro de Investigaciones Inmunológicas Básicas y Aplicadas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Buenos Aires, Argentina (M.A.)
| | - Alejandra I Attorresi
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina (A.C., A.Y., N.G., N.D.S., A.S., F.D., F.M., N.F., C.D.); IBioBA MPSP - Instituto de Investigaciones en Biomedicina de Buenos Aires, CONICET, Instituto Partner de la Sociedad Max Planck, Buenos Aires, Argentina (A.I.A.); Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina (Á.R.-G., C.S.); and Centro de Investigaciones Inmunológicas Básicas y Aplicadas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Buenos Aires, Argentina (M.A.)
| | - Ángela Rodríguez-González
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina (A.C., A.Y., N.G., N.D.S., A.S., F.D., F.M., N.F., C.D.); IBioBA MPSP - Instituto de Investigaciones en Biomedicina de Buenos Aires, CONICET, Instituto Partner de la Sociedad Max Planck, Buenos Aires, Argentina (A.I.A.); Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina (Á.R.-G., C.S.); and Centro de Investigaciones Inmunológicas Básicas y Aplicadas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Buenos Aires, Argentina (M.A.)
| | - Federico Monczor
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina (A.C., A.Y., N.G., N.D.S., A.S., F.D., F.M., N.F., C.D.); IBioBA MPSP - Instituto de Investigaciones en Biomedicina de Buenos Aires, CONICET, Instituto Partner de la Sociedad Max Planck, Buenos Aires, Argentina (A.I.A.); Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina (Á.R.-G., C.S.); and Centro de Investigaciones Inmunológicas Básicas y Aplicadas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Buenos Aires, Argentina (M.A.)
| | - Natalia Fernández
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina (A.C., A.Y., N.G., N.D.S., A.S., F.D., F.M., N.F., C.D.); IBioBA MPSP - Instituto de Investigaciones en Biomedicina de Buenos Aires, CONICET, Instituto Partner de la Sociedad Max Planck, Buenos Aires, Argentina (A.I.A.); Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina (Á.R.-G., C.S.); and Centro de Investigaciones Inmunológicas Básicas y Aplicadas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Buenos Aires, Argentina (M.A.)
| | - Martín Abba
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina (A.C., A.Y., N.G., N.D.S., A.S., F.D., F.M., N.F., C.D.); IBioBA MPSP - Instituto de Investigaciones en Biomedicina de Buenos Aires, CONICET, Instituto Partner de la Sociedad Max Planck, Buenos Aires, Argentina (A.I.A.); Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina (Á.R.-G., C.S.); and Centro de Investigaciones Inmunológicas Básicas y Aplicadas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Buenos Aires, Argentina (M.A.)
| | - Carina Shayo
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina (A.C., A.Y., N.G., N.D.S., A.S., F.D., F.M., N.F., C.D.); IBioBA MPSP - Instituto de Investigaciones en Biomedicina de Buenos Aires, CONICET, Instituto Partner de la Sociedad Max Planck, Buenos Aires, Argentina (A.I.A.); Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina (Á.R.-G., C.S.); and Centro de Investigaciones Inmunológicas Básicas y Aplicadas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Buenos Aires, Argentina (M.A.)
| | - Carlos Davio
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina (A.C., A.Y., N.G., N.D.S., A.S., F.D., F.M., N.F., C.D.); IBioBA MPSP - Instituto de Investigaciones en Biomedicina de Buenos Aires, CONICET, Instituto Partner de la Sociedad Max Planck, Buenos Aires, Argentina (A.I.A.); Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina (Á.R.-G., C.S.); and Centro de Investigaciones Inmunológicas Básicas y Aplicadas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Buenos Aires, Argentina (M.A.)
| |
Collapse
|
132
|
Hussain S, Singh A, Nazir SU, Tulsyan S, Khan A, Kumar R, Bashir N, Tanwar P, Mehrotra R. Cancer drug resistance: A fleet to conquer. J Cell Biochem 2019; 120:14213-14225. [PMID: 31037763 DOI: 10.1002/jcb.28782] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/08/2019] [Accepted: 01/14/2019] [Indexed: 12/18/2022]
Abstract
Cancer is a disease that claims millions of lives each year across the world. Despite advancement in technologies and therapeutics for treating the disease, these modes are often found to turn ineffective during the course of treatment. The resistance against drugs in cancer patients stems from multiple factors, which constitute genetic heterogeneity like gene mutations, tumor microenvironment, exosomes, miRNAs, high rate of drug efflux from cells, and so on. This review attempts to collate all such known and reported factors that influence cancer drug resistance and may help researchers with information that might be useful in developing better therapeutics in near future to enable better management of several cancers across the world.
Collapse
Affiliation(s)
- Showket Hussain
- Division of Cellular and Molecular Diagnostics, National Institute of Cancer Prevention and Research, Noida, India
| | - Ankita Singh
- Division of Cellular and Molecular Diagnostics, National Institute of Cancer Prevention and Research, Noida, India
| | - Sheeraz Un Nazir
- Division of Cellular and Molecular Diagnostics, National Institute of Cancer Prevention and Research, Noida, India
| | - Sonam Tulsyan
- Division of Preventive Oncology, National Institute of Cancer Prevention and Research, Noida, India
| | - Asiya Khan
- Department of Lab Oncology, AIIMS, New Delhi, India
| | - Ramesh Kumar
- Department of Biochemistry, Bundelkhand University, Jhansi, India
| | - Nasreena Bashir
- College of Applied Medicine, King Khalid University, Abha, Saudi Arabia
| | | | - Ravi Mehrotra
- Division of Preventive Oncology, National Institute of Cancer Prevention and Research, Noida, India
| |
Collapse
|
133
|
Speer JE, Gunasekara DB, Wang Y, Fallon JK, Attayek PJ, Smith PC, Sims CE, Allbritton NL. Molecular transport through primary human small intestinal monolayers by culture on a collagen scaffold with a gradient of chemical cross-linking. J Biol Eng 2019; 13:36. [PMID: 31061676 PMCID: PMC6487070 DOI: 10.1186/s13036-019-0165-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/08/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The luminal surface of the small intestine is composed of a monolayer of cells overlying a lamina propria comprised of extracellular matrix (ECM) proteins. The ECM provides a porous substrate critical for nutrient exchange and cellular adhesion. The enterocytes within the epithelial monolayer possess proteins such as transporters, carriers, pumps and channels that participate in the movement of drugs, metabolites, ions and amino acids and whose function can be regulated or altered by the properties of the ECM. Here, we characterized expression and function of proteins involved in transport across the human small intestinal epithelium grown on two different culture platforms. One strategy employs a conventional scaffolding method comprised of a thin ECM film overlaying a porous membrane while the other utilizes a thick ECM hydrogel placed on a porous membrane. The thick hydrogel possesses a gradient of chemical cross-linking along its length to provide a softer substrate than that of the ECM film-coated membrane while maintaining mechanical stability. RESULTS The monolayers on both platforms possessed goblet cells and abundant enterocytes and were impermeable to Lucifer yellow and fluorescein-dextran (70 kD) indicating high barrier integrity. Multiple transporter proteins were present in both primary-cell culture formats at levels similar to those present in freshly isolated crypts/villi; however, expression of breast cancer resistance protein (BCRP) and multidrug resistance protein 2 (MRP2) in the monolayers on the conventional scaffold was substantially less than that on the gradient cross-linked scaffold and freshly isolated crypts/villi. Monolayers on the conventional scaffold failed to transport the BCRP substrate prazosin while cells on the gradient cross-linked scaffold successfully transported this drug to better mimic the properties of in vivo small intestine. CONCLUSIONS The results of this comparison highlight the need to create in vitro intestinal transport platforms whose characteristics mimic the in vivo lamina propria in order to accurately recapitulate epithelial function. GRAPHICAL ABSTRACT
Collapse
Affiliation(s)
- Jennifer E. Speer
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599 USA
| | - Dulan B. Gunasekara
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27599 USA
| | - Yuli Wang
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599 USA
| | - John K. Fallon
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599 USA
| | - Peter J. Attayek
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27599 USA
| | - Philip C. Smith
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599 USA
| | - Christopher E. Sims
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599 USA
| | - Nancy L. Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599 USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27599 USA
| |
Collapse
|
134
|
ABCC3 is a novel target for the treatment of pancreatic cancer. Adv Biol Regul 2019; 73:100634. [PMID: 31053501 DOI: 10.1016/j.jbior.2019.04.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 04/23/2019] [Accepted: 04/23/2019] [Indexed: 02/06/2023]
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is a very aggressive disease, lacking effective therapeutic approaches and leaving PDAC patients with a poor prognosis. The life expectancy of PDAC patients has not experienced a significant change in the last few decades with a five-year survival rate of only 8%. To address this unmet need, novel pharmacological targets must be identified for clinical intervention. ATP Binding Cassette (ABC) transporters are frequently overexpressed in different cancer types and represent one of the major mechanisms responsible for chemoresistance. However, a more direct role for ABC transporters in tumorigenesis has not been widely investigated. Here, we show that ABCC3 (ABC Subfamily C Member 3; previously known as MRP3) is overexpressed in PDAC cell lines and also in clinical samples. We demonstrate that ABCC3 expression is regulated by mutant p53 via miR-34 and that the transporter drives PDAC progression via transport of the bioactive lipid lysophosphatidylinositol (LPI). Disruption of ABCC3 function either by genetic knockdown reduces pancreatic cancer cell growth in vitro and in vivo. Mechanistically, we demonstrate that knockdown of ABCC3 reduce cell proliferation by inhibition of STAT3 and HIF1α signalling pathways, previously been shown to be key regulators of PDAC progression. Collectively, our results identify ABCC3 as a novel and promising target in PDAC therapy.
Collapse
|
135
|
Emerging Roles of Aryl Hydrocarbon Receptors in the Altered Clearance of Drugs during Chronic Kidney Disease. Toxins (Basel) 2019; 11:toxins11040209. [PMID: 30959953 PMCID: PMC6521271 DOI: 10.3390/toxins11040209] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/12/2019] [Accepted: 04/03/2019] [Indexed: 12/11/2022] Open
Abstract
Chronic kidney disease (CKD) is a major public health problem, since 300,000,000 people in the world display a glomerular filtration rate (GFR) below 60 mL/min/1.73m². Patients with CKD have high rates of complications and comorbidities. Thus, they require the prescription of numerous medications, making the management of patients very complex. The prescription of numerous drugs associated with an altered renal- and non-renal clearance makes dose adjustment challenging in these patients, with frequent drug-related adverse events. However, the mechanisms involved in this abnormal drug clearance during CKD are not still well identified. We propose here that the transcription factor, aryl hydrocarbon receptor, which is the cellular receptor for indolic uremic toxins, could worsen the metabolism and the excretion of drugs in CKD patients.
Collapse
|
136
|
Zhou DR, Eid R, Boucher E, Miller KA, Mandato CA, Greenwood MT. Stress is an agonist for the induction of programmed cell death: A review. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:699-712. [DOI: 10.1016/j.bbamcr.2018.12.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/17/2018] [Accepted: 12/01/2018] [Indexed: 02/07/2023]
|
137
|
Bahar E, Kim JY, Yoon H. Chemotherapy Resistance Explained through Endoplasmic Reticulum Stress-Dependent Signaling. Cancers (Basel) 2019; 11:cancers11030338. [PMID: 30857233 PMCID: PMC6468910 DOI: 10.3390/cancers11030338] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/01/2019] [Accepted: 03/01/2019] [Indexed: 02/06/2023] Open
Abstract
Cancers cells have the ability to develop chemotherapy resistance, which is a persistent problem during cancer treatment. Chemotherapy resistance develops through different molecular mechanisms, which lead to modification of the cancer cells signals needed for cellular proliferation or for stimulating an immune response. The endoplasmic reticulum (ER) is an important organelle involved in protein quality control, by promoting the correct folding of protein and ER-mediated degradation of unfolded or misfolded protein, namely, ER-associated degradation. Disturbances of the normal ER functions causes an accumulation of unfolded or misfolded proteins in the ER lumen, resulting in a condition called “ER stress (ERS).” ERS triggers the unfolded protein response (UPR)—also called the ERS response (ERSR)—to restore homeostasis or activate cell death. Although the ERSR is one emerging potential target for chemotherapeutics to treat cancer, it is also critical for chemotherapeutics resistance, as well. However, the detailed molecular mechanism of the relationship between the ERSR and tumor survival or drug resistance remains to be fully understood. In this review, we aim to describe the most vital molecular mechanism of the relationship between the ERSR and chemotherapy resistance. Moreover, the review also discusses the molecular mechanism of ER stress-mediated apoptosis on cancer treatments.
Collapse
Affiliation(s)
- Entaz Bahar
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Gyeongnam, Korea.
| | - Ji-Ye Kim
- Department of Pathology, College of Medicine, Yonsei University, Seoul 03722, Korea.
- Department of Pathology, Ilsan Paik Hospital, Inje University, Goyang 10381, Gyeonggi-do, Korea.
- Department of Pathology, National Cancer Center, Goyang 10408, Gyeonggi-do, Korea.
| | - Hyonok Yoon
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Gyeongnam, Korea.
| |
Collapse
|
138
|
Di Giacomo S, Briz O, Monte MJ, Sanchez-Vicente L, Abete L, Lozano E, Mazzanti G, Di Sotto A, Marin JJG. Chemosensitization of hepatocellular carcinoma cells to sorafenib by β-caryophyllene oxide-induced inhibition of ABC export pumps. Arch Toxicol 2019; 93:623-634. [PMID: 30659321 DOI: 10.1007/s00204-019-02395-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 01/10/2019] [Indexed: 01/16/2023]
Abstract
Several ATP-binding cassette (ABC) proteins reduce intracellular concentrations of antitumor drugs and hence weaken the response of cancer cells to chemotherapy. Accordingly, the inhibition of these export pumps constitutes a promising strategy to chemosensitize highly chemoresistant tumors, such as hepatocellular carcinoma (HCC). Here, we have investigated the ability of β-caryophyllene oxide (CRYO), a naturally occurring sesquiterpene component of many essential oils, to inhibit, at non-toxic doses, ABC pumps and improve the response of HCC cells to sorafenib. First, we have obtained a clonal subline (Alexander/R) derived from human hepatoma cells with enhanced multidrug resistance (MDR) associated to up-regulation (mRNA and protein) of MRP1 and MRP2. Analysis of fluorescent substrates export (flow cytometry) revealed that CRYO did not affect the efflux of fluorescein (MRP3, MRP4 and MRP5) but inhibited that of rhodamine 123 (MDR1) and calcein (MRP1 and MRP2). This ability was higher for CRYO than for other sesquiterpenes assayed. CRYO also inhibited sorafenib efflux, increased its intracellular accumulation (HPLC-MS/MS) and enhanced its cytotoxic response (MTT). For comparison, the effect of known ABC pumps inhibitors was also determined. They induced strong (diclofenac on MRPs), modest (verapamil on MDR1) or null (fumitremorgin C on BCRP) effect on sorafenib efflux and cytotoxicity. In the mouse xenograft model, the response to sorafenib treatment of subcutaneous tumors generated by mouse hepatoma Hepa 1-6/R cells, with marked MDR phenotype, was significantly enhanced by CRYO co-administration. In conclusion, at non-toxic dose, CRYO is able to chemosensitizating liver cancer cells to sorafenib by favoring its intracellular accumulation.
Collapse
Affiliation(s)
- Silvia Di Giacomo
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy.
| | - Oscar Briz
- Laboratory of Experimental Hepatology and Drug Targeting (HEVEFARM), IBSAL, University of Salamanca, Salamanca, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Maria J Monte
- Laboratory of Experimental Hepatology and Drug Targeting (HEVEFARM), IBSAL, University of Salamanca, Salamanca, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Laura Sanchez-Vicente
- Laboratory of Experimental Hepatology and Drug Targeting (HEVEFARM), IBSAL, University of Salamanca, Salamanca, Spain
| | - Lorena Abete
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Elisa Lozano
- Laboratory of Experimental Hepatology and Drug Targeting (HEVEFARM), IBSAL, University of Salamanca, Salamanca, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Gabriela Mazzanti
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Antonella Di Sotto
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Jose J G Marin
- Laboratory of Experimental Hepatology and Drug Targeting (HEVEFARM), IBSAL, University of Salamanca, Salamanca, Spain.
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain.
- Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.
| |
Collapse
|
139
|
Bozeman EN, Yang L. Biological Events and Barriers to Effective Delivery of Cancer Therapeutics. Bioanalysis 2019. [DOI: 10.1007/978-3-030-01775-0_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
140
|
Li B, Cai M, Lin L, Sun W, Zhou Z, Wang S, Wang Y, Zhu K, Shuai X. MRI-visible and pH-sensitive micelles loaded with doxorubicin for hepatoma treatment. Biomater Sci 2019; 7:1529-1542. [DOI: 10.1039/c8bm01501e] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
A novel pH-sensitive micelle was constructed to efficiently co-deliver SPIONs and doxorubicin for cancer theranostic application.
Collapse
Affiliation(s)
- Bo Li
- Laboratory of Interventional Radiology
- Department of Minimally Invasive Interventional Radiology
- and Department of Radiology
- the Second Affiliated Hospital of Guangzhou Medical University
- Guangzhou
| | - Mingyue Cai
- Laboratory of Interventional Radiology
- Department of Minimally Invasive Interventional Radiology
- and Department of Radiology
- the Second Affiliated Hospital of Guangzhou Medical University
- Guangzhou
| | - Liteng Lin
- Laboratory of Interventional Radiology
- Department of Minimally Invasive Interventional Radiology
- and Department of Radiology
- the Second Affiliated Hospital of Guangzhou Medical University
- Guangzhou
| | - Weitong Sun
- Pharmaceutical college of Jiamusi University
- Jiamusi University
- Jiamusi
- China
| | - Zhimei Zhou
- Laboratory of Interventional Radiology
- Department of Minimally Invasive Interventional Radiology
- and Department of Radiology
- the Second Affiliated Hospital of Guangzhou Medical University
- Guangzhou
| | - Shiyin Wang
- PCFM Lab of Ministry of Education
- School of Materials Science and Engineering
- Sun Yat-sen University
- Guangzhou
- China
| | - Yong Wang
- PCFM Lab of Ministry of Education
- School of Materials Science and Engineering
- Sun Yat-sen University
- Guangzhou
- China
| | - Kangshun Zhu
- Laboratory of Interventional Radiology
- Department of Minimally Invasive Interventional Radiology
- and Department of Radiology
- the Second Affiliated Hospital of Guangzhou Medical University
- Guangzhou
| | - Xintao Shuai
- PCFM Lab of Ministry of Education
- School of Materials Science and Engineering
- Sun Yat-sen University
- Guangzhou
- China
| |
Collapse
|
141
|
Abstract
Drug resistance is a well-known phenomenon that occurs when initially responsive to chemotherapy cancer cells become tolerant and elude further effectiveness of anticancer drugs. Based on their mechanism of action, anticancer drugs can be divided into cytotoxic-based agents and target-based agents. An important role among the therapeutics of the second group is played by drugs targeting topoisomerases, nuclear enzymes critical to DNA function and cell survival. These enzymes are cellular targets of several groups of anticancer agents which generate DNA damage in rapidly proliferating cancer cells. Drugs targeting topoisomerase I are mostly analogs of camtothecin, a natural compound isolated from the bark of a tree growing in China. Drugs targeting topoisomerase II are divided into poisons, such as anthracycline antibiotics, whose action is based on intercalation between DNA bases, and catalytic inhibitors that block topoisomerase II at different stages of the catalytic cycle. Unfortunately, chemotherapy is often limited by the induction of drug resistance. Identifying mechanisms that promote drug resistance is critical for the improvement of patient prognosis. Cancer drug resistance is a complex phenomenon that may be influenced by many factors. Here we discuss various mechanisms by which cancer cells can develop resistance to topoisomerase-directed drugs, which include enhanced drug efflux, mutations in topoisomerase genes, hypophosphorylation of topoisomerase II catalytic domain, activation of NF-κB transcription factor and drug inactivation. All these events may lead to the ineffective induction of cancer cell death. Attempts at circumventing drug resistance through the inhibition of cellular efflux pumps, use of silencing RNAs or inhibition of some important mechanisms, which can allow cancer cells to survive therapy, are also presented.
Collapse
Affiliation(s)
- Karol Wtorek
- Department of Biomolecular Chemistry, Medical University of Łódź, Poland
| | - Angelika Długosz
- Department of Biomolecular Chemistry, Medical University of Łódź, Poland
| | - Anna Janecka
- Department of Biomolecular Chemistry, Medical University of Łódź, Poland
| |
Collapse
|
142
|
Ogino Y, Sato A, Uchiumi F, Tanuma SI. Genomic and tumor biological aspects of the anticancer nicotinamide phosphoribosyltransferase inhibitor FK866 in resistant human colorectal cancer cells. Genomics 2018; 111:1889-1895. [PMID: 30582964 DOI: 10.1016/j.ygeno.2018.12.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 12/17/2018] [Accepted: 12/20/2018] [Indexed: 01/22/2023]
Abstract
Cancer cells' resistance to drugs remains an important problem affecting cancer treatment strategies. We previously studied the nicotinamide phosphoribosyltransferase (NAMPT) inhibitor FK866's resistance mechanisms in the human colorectal cancer HCT116 cells. We established an acquired FK866-resistant cell line, HCT116RFK866. In this study, we investigated gene mutations in parental HCT116 and HCT116RFK866 cells using exome sequencing technology. The results indicated cluster genes related to NAD+ biosynthesis (including NAMPT), DNA repair, and ATP-binding cassette transporters were differentially altered in these cells. Interestingly, HCT116RFK866 cells, which are resistant to other class NAMPT inhibitors, were more sensitive to the anticancer 5-fluorouracil and cisplatin and γ-ray irradiation compared to parental HCT116 cells. This higher sensitivity appears to cause a genetic change in the identified gene clusters by resistance to the NAMPT inhibitor FK866. Collectively, these novel findings provide a better understanding of anticancer candidate NAMPT inhibitors with regard to resistance mechanisms and cancer chemotherapy strategies.
Collapse
Affiliation(s)
- Yoko Ogino
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan; Department of Gene Regulation, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Akira Sato
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan.
| | - Fumiaki Uchiumi
- Department of Gene Regulation, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Sei-Ichi Tanuma
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan; Department of Genomic Medicinal Science, Research Institute for Science and Technology, Organization for Research Advancement, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| |
Collapse
|
143
|
Side Population: Its Use in the Study of Cellular Heterogeneity and as a Potential Enrichment Tool for Rare Cell Populations. Stem Cells Int 2018; 2018:2472137. [PMID: 30627171 PMCID: PMC6304857 DOI: 10.1155/2018/2472137] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 11/07/2018] [Accepted: 11/14/2018] [Indexed: 12/16/2022] Open
Abstract
There is still much to learn about the cells used for cell- and gene-based therapies in the clinical setting. Stem cells are found in virtually all tissues in the human body. As a result, cells isolated from these tissues are a heterogeneous population consisting of various subpopulations including stem cells. Several strategies have been used to isolate and define the subpopulations that constitute these heterogeneous populations, one of which is the side population (SP) assay. SP cells are identified by their ability to efflux a fluorescent dye at a rate that is greater than the main cell population. This elevated rate of dye efflux has been attributed to the expression of members of the ATP-binding cassette (ABC) transporter protein family. SP cells have been identified in various tissues. In this review, we discuss the research to date on SP cells, focussing on SP cells identified in haematopoietic stem cells, adipose-derived stromal cells, and dental pulp.
Collapse
|
144
|
Utility of curcumin for the treatment of diabetes mellitus: Evidence from preclinical and clinical studies. JOURNAL OF NUTRITION & INTERMEDIARY METABOLISM 2018. [DOI: 10.1016/j.jnim.2018.05.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
145
|
Vehovszky Á, Farkas A, Csikós V, Székács A, Mörtl M, Győri J. Neonicotinoid insecticides are potential substrates of the multixenobiotic resistance (MXR) mechanism in the non-target invertebrate, Dreissena sp. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2018; 205:148-155. [PMID: 30384196 DOI: 10.1016/j.aquatox.2018.10.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/19/2018] [Accepted: 10/22/2018] [Indexed: 06/08/2023]
Abstract
Mussels are among the most frequently used invertebrate animals in aquatic toxicology to detect toxic exposure in the environment. The presence and activity of a cellular defence system, the multixenobiotic resistance (MXR) mechanism, was also established in these organisms. In isolated gill tissues of dreissenid mussels (D. bugensis) the MXR activity was assayed after treatment by commercially available insecticides (formulated products) which contain neonicotinoids as their active ingredients: Actara (thiamethoxam), Apacs (clothianidin), Calypso (thiacloprid) and Kohinor (imidacloprid), respectively. While applying the accumulation assay method, 0.5 μM rhodamine B was used as model substrate and 20 μM verapamil as model inhibitor of the MXR mechanism. In acute (in vitro) experiments when isolated gills were co-incubated in graded concentrations of insecticides and rhodamine B simultaneously, Calypso and Kohinor treatment resulted increasing rhodamine accumulation. Chemical analysis of gills in vitro incubated in insecticides demonstrated higher tissue concentrations of thiamethoxam, clothianidin and thiacloprid in the presence of verapamil suggesting that the active ingredients of Actara, Apacs and Calypso are potential substrates of the MXR mediated cellular efflux. In contrast, verapamil did significantly alter the accumulated imidacloprid concentrations in gills, suggesting that the active component of Kohinor is not transported by the MXR mechanism. Chronic (in vivo) exposures of the intact animals in lower, 1, 10 mg/L concentration of neonicotinoid products, resulted in a decreased level of both rhodamine accumulation and verapamil inhibition by the 12th-14th days of treatment. These results suggest an enhancement of MXR activity (chemostimulation), building up gradually in the animals exposed to Actara, Apacs and Kohinor, respectively. Neonicotinoid-type insecticides are generally considered as selective neurotoxins for insects, targeting the nicotinic type acetylcholine receptors (nAChRs) in their central nervous system. Our present results provide the first evidences that neonicotinoid insecticides are also able to alter the transmembrane transport mechanisms related to the MXR system.
Collapse
Affiliation(s)
- Ágnes Vehovszky
- MTA Centre for Ecological Research, Balaton Limnological Institute, Tihany, Hungary.
| | - Anna Farkas
- MTA Centre for Ecological Research, Balaton Limnological Institute, Tihany, Hungary
| | - Vivien Csikós
- MTA Centre for Ecological Research, Balaton Limnological Institute, Tihany, Hungary
| | - András Székács
- Agro-Environmental Research Institute, National Agricultural Research and Innovation Centre, Budapest, Hungary
| | - Mária Mörtl
- Agro-Environmental Research Institute, National Agricultural Research and Innovation Centre, Budapest, Hungary
| | - János Győri
- MTA Centre for Ecological Research, Balaton Limnological Institute, Tihany, Hungary
| |
Collapse
|
146
|
Li H, Krstin S, Wink M. Modulation of multidrug resistant in cancer cells by EGCG, tannic acid and curcumin. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 50:213-222. [PMID: 30466981 DOI: 10.1016/j.phymed.2018.09.169] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 08/10/2018] [Accepted: 09/17/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Cancer is one of the most common life-threatening diseases worldwide; many patients develop multidrug resistance after treatment with anticancer drugs. The main mechanism leading to multidrug resistance is the overexpression of ABC transporters in cancer cells. Chemosensitizers are needed to inhibit the activity of ABC transporters, resulting in higer intracellular concentration of anticancer drugs. Some secondary metabolites have been reported to be chemosensitizers by inhibiting ABC transporters. Epigallocatechin gallate (EGCG), tannic acid, and curcumin were employed in this study. Different assays were used to detect whether they have the ability to inhibit P-gp activity and overcome multidrug resistance in cancer cells overexpressing P-gp. Hypothesis/Purpose: CEM/ADR 5000 and Caco-2 cell lines, which overexpress P-gp, are multidrug resistant cell lines. We first detected whether the combination of polyphenols (EGCG, tannic acid, curcumin) and doxorubicin, an anticancer drug, is synergistic or not. To further understand the potential mechanism, EGCG, tannic acid, and curcumin were tested to check whether they have the ability to inhibit P-gp activity. When P-gp activity is inhibited, the intracellular concentration of doxorubicin is higher, resulting in enhanced cytotoxicity of doxorubicin. STUDY DESIGN The P-gp overexpressing human colon cancer cell line Caco-2 and human T-lymphoblastic leukemia cell line CEM/ADR 5000 were used in this study. Two-drug combinations (doxorubicin + polyphenol) and three-drug combinations (doxorubicin + polyphenol + digitonin) were tested to examine potential synergism. The potential mechanism leading to synergism would be the inhibition of P-gp activity. A Rhodamine 123 assay and Calcein-AM assay in Caco-2 and CEM/ADR 5000, respectively, were used to detect P-gp inhibition by EGCG, curcumin, and tannic acid. METHODS MTT assay was used to determine the cytotoxicity of doxorubicin, polyphenols and digitonin alone, and then their combinations. Furthermore, Rhodamine 123 and Calcein-AM were used to detect the effects of polyphenols on the activity of P-gp. RESULTS The results demonstrated that a combination of non-toxic concentrations of each polyphenol with doxorubicin synergistically sensitized Caco-2 and CEM/ADR 5000 cells. Furthermore, three-drug combinations (doxorubicin + polyphenol + digitonin) were much more effective. In addition, the activity of P-gp in Caco-2 and CEM/ADR 5000 cells was measured. Consistent with the combination results, tannic acid and curcumin decreased the activity of P-gp both in Caco-2 and CEM/ADR 5000. EGCG, which weakly affected the activity of P-gp in CEM/ADR 5000, only had an effect on P-gp under higher concentration in Caco-2 cells. CONCLUSION Our results show that EGCG, curcumin, and tannic acid, when combined with doxorubicin, can exert synergism, mediated by a reduced activity of P-gp. This study suggests that polyphenols, by modulating the activity of P-gp, may be used as chemosensitisers.
Collapse
Affiliation(s)
- Hanmei Li
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, D-69120, Heidelberg, Germany
| | - Sonja Krstin
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, D-69120, Heidelberg, Germany
| | - Michael Wink
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, D-69120, Heidelberg, Germany.
| |
Collapse
|
147
|
Cao Z, Li D, Liu L, Yang P. Effect of five novel 5-substituted tetrandrine derivatives on P-glycoprotein-mediated inhibition and transport in Caco-2 cells. Oncol Lett 2018; 16:6808-6814. [PMID: 30405825 DOI: 10.3892/ol.2018.9492] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 04/11/2018] [Indexed: 11/06/2022] Open
Abstract
Tetrandrine (Tet) is a potent inhibitor that reverses P-glycoprotein-mediated multidrug resistance (MDR). A number of novel 5-substituted tetrandrine derivatives were synthesized by the authors. The present study aimed at identifying potential P-gp inhibitor candidates, and intracellular uptake and efflux experiments and Caco-2 cell-based Transwell transport studies were performed. It was demonstrated that all five test compounds were able to inhibit efflux and increase intracellular uptake of the P-gp substrate, rhodamine-123 (Rho-123); the test compounds were P-gp inhibitors. The transepithelial transport experiment indicated that the secretory (basolateral-to-apical) of Rho-123 decreased, the absorption (apical-to-basolateral) increased and the transport efflux ratio (ER) reduced in the presence of the five compounds. Among the compounds, fluobenzene-Tet (TF) exhibited similar inhibitory effect as Tet. Although the other four test compounds exhibited weaker inhibitory effects than Tet and TF, the compounds exhibited stronger inhibitory effects compared with the reference compound verapamil. The study demonstrated that the five novel 5-substituted tetrandrine derivatives are able to act as inhibitors of P-gp to overcome P-gp-mediated drug resistance.
Collapse
Affiliation(s)
- Zhonglian Cao
- Instrumental Analysis Centre, School of Pharmacy, Fudan University, Shanghai 201203, P.R. China
| | - Dan Li
- Instrumental Analysis Centre, School of Pharmacy, Fudan University, Shanghai 201203, P.R. China.,Department of Pharmacology, Center for Pharmacological Evaluation and Research of SIPI, Shanghai Institute of Pharmaceutical Industry, Shanghai 200437, P.R. China
| | - Li Liu
- Department of Pharmacology, Center for Pharmacological Evaluation and Research of SIPI, Shanghai Institute of Pharmaceutical Industry, Shanghai 200437, P.R. China
| | - Ping Yang
- Instrumental Analysis Centre, School of Pharmacy, Fudan University, Shanghai 201203, P.R. China
| |
Collapse
|
148
|
Sterzyńska K, Klejewski A, Wojtowicz K, Świerczewska M, Nowicki M, Brązert J, Januchowski R. Myotilin, a New Topotecan Resistant Protein in Ovarian Cancer Cell Lines. J Cancer 2018; 9:4413-4421. [PMID: 30519347 PMCID: PMC6277650 DOI: 10.7150/jca.27342] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/24/2018] [Indexed: 11/17/2022] Open
Abstract
Background: Low effectiveness of chemotherapy in ovarian cancer results from development of drug resistance during treatment. Topotecan (TOP) is a chemotherapeutic drug used in second-line chemotherapy of this cancer. Unfortunately, during treatment cancer can develop diverse cellular and tissue specific mechanisms of resistance to cytotoxic drugs. Methods: We analyzed development of TOP resistance in ovarian cancer cell lines (A2780 and W1). On the base of our previous results where a set of “new genes” with different functions that can be related to TOP-resistance was described hereby we performed detailed analysis of MYOT expression. MYOT mRNA level (real time PCR analysis), protein expression in cell lysates and cell culture medium (western blot analysis) and protein expression in cancer cells (immunofluorescence analysis) were determined in this study. Results: We observed increased expression of MYOT in TOP resistant cell lines at both mRNA and protein level. MYOT, together with extracellular matrix molecules like COL1A2 and COL15A1 were also secreted to corresponding cell culture media. Conclusion: Our results suggest that upregulation of MYOT can be related to TOP resistance in ovarian cancer cell lines.
Collapse
Affiliation(s)
- Karolina Sterzyńska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznań, Poland
| | - Andrzej Klejewski
- Department of Nursing, Poznan University of Medical Sciences, Poznań, Poland.,Department of Obstetrics and Women's Diseases, Poznan University of Medical Sciences, Poznań, Poland
| | - Karolina Wojtowicz
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznań, Poland
| | - Monika Świerczewska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznań, Poland
| | - Michał Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznań, Poland
| | - Jacek Brązert
- Department of Obstetrics and Women's Diseases, Poznan University of Medical Sciences, Poznań, Poland
| | - Radosław Januchowski
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznań, Poland
| |
Collapse
|
149
|
Giarra S, Zappavigna S, Campani V, Abate M, Cossu AM, Leonetti C, Porru M, Mayol L, Caraglia M, De Rosa G. Chitosan-Based Polyelectrolyte Complexes for Doxorubicin and Zoledronic Acid Combined Therapy to Overcome Multidrug Resistance. Pharmaceutics 2018; 10:pharmaceutics10040180. [PMID: 30304840 PMCID: PMC6321278 DOI: 10.3390/pharmaceutics10040180] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/02/2018] [Accepted: 10/05/2018] [Indexed: 11/16/2022] Open
Abstract
This study aimed to develop nanovectors co-encapsulating doxorubicin (Doxo) and zoledronic acid (Zol) for a combined therapy against Doxo-resistant tumors. Chitosan (CHI)-based polyelectrolyte complexes (PECs) prepared by ionotropic gelation technique were proposed. The influence of some experimental parameters was evaluated in order to optimize the PECs in terms of size and polydispersity index (PI). PEC stability was studied by monitoring size and zeta potential over time. In vitro studies were carried out on wild-type and Doxo-resistant cell lines, to assess both the synergism between Doxo and Zol, as well as the restoring of Doxo sensitivity. Polymer concentration, incubation time, and use of a surfactant were found to be crucial to achieving small size and monodisperse PECs. Doxo and Zol, only when encapsulated in PECs, showed a synergistic antiproliferative effect in all the tested cell lines. Importantly, the incubation of Doxo-resistant cell lines with Doxo/Zol co-encapsulating PECs resulted in the restoration of Doxo sensitivity.
Collapse
Affiliation(s)
- Simona Giarra
- Department of Pharmacy, University of Naples Federico II, D. Montesano 49, 80131 Naples, Italy.
| | - Silvia Zappavigna
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, L. De Crecchio 7, 80138 Naples, Italy.
| | - Virginia Campani
- Department of Pharmacy, University of Naples Federico II, D. Montesano 49, 80131 Naples, Italy.
| | - Marianna Abate
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, L. De Crecchio 7, 80138 Naples, Italy.
| | - Alessia Maria Cossu
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, L. De Crecchio 7, 80138 Naples, Italy.
| | - Carlo Leonetti
- UOSD SAFU, IRCCS Regina Elena National Cancer Institute, E. Chianesi 53, 00144 Rome, Italy.
| | - Manuela Porru
- UOSD SAFU, IRCCS Regina Elena National Cancer Institute, E. Chianesi 53, 00144 Rome, Italy.
| | - Laura Mayol
- Department of Pharmacy, University of Naples Federico II, D. Montesano 49, 80131 Naples, Italy.
| | - Michele Caraglia
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, L. De Crecchio 7, 80138 Naples, Italy.
| | - Giuseppe De Rosa
- Department of Pharmacy, University of Naples Federico II, D. Montesano 49, 80131 Naples, Italy.
| |
Collapse
|
150
|
Zhou L, Qiu T, Lv F, Liu L, Ying J, Wang S. Self-Assembled Nanomedicines for Anticancer and Antibacterial Applications. Adv Healthc Mater 2018; 7:e1800670. [PMID: 30080319 DOI: 10.1002/adhm.201800670] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/03/2018] [Indexed: 01/28/2023]
Abstract
Self-assembly strategies have been widely applied in the nanomedicine field, which provide a convenient approach for building various structures for delivery carriers. When cooperating with biomolecules, self-assembly systems have significant influence on the cell activity and life process and could be used for regulating nanodrug activity. In this review, self-assembled nanomedicines are introduced, including materials, encapsulation, and releasing strategies, where self-assembly strategies are involved. Furthermore, as a promising and emerging area for nanomedicine, in situ self-assembly of anticancer drugs and supramolecular antibiotic switches is also discussed about how to regulate drug activity. Selective pericellular assembly can block mass transformation of cancer cells inducing cell apoptosis, and the intracellular assembly can either cause cell death or effectively avoid drug elimination from cytosol of cancer cells because of the assembly-induced retention (AIR) effect. Host-guest interactions of drug and competitive molecules offer reversible regulations of antibiotic activity, which can reduce drug-resistance and inhibit the generation of drug-resistant bacteria. Finally, the challenges and development trend in the field are discussed.
Collapse
Affiliation(s)
- Lingyun Zhou
- Beijing National Laboratory for Molecular Sciences; Key Laboratory of Organic Solids; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
- College of Chemistry; University of Chinese Academy of Sciences; Beijing 100049 P. R. China
| | - Tian Qiu
- Department of Pathology; National Cancer Center/National Clinical Research Center for; Cancer/Cancer Hospital; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing 100021 P. R. China
| | - Fengting Lv
- Beijing National Laboratory for Molecular Sciences; Key Laboratory of Organic Solids; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
| | - Libing Liu
- Beijing National Laboratory for Molecular Sciences; Key Laboratory of Organic Solids; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
| | - Jianming Ying
- Department of Pathology; National Cancer Center/National Clinical Research Center for; Cancer/Cancer Hospital; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing 100021 P. R. China
| | - Shu Wang
- Beijing National Laboratory for Molecular Sciences; Key Laboratory of Organic Solids; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
- College of Chemistry; University of Chinese Academy of Sciences; Beijing 100049 P. R. China
| |
Collapse
|