1
|
Zhang X, Tian H, Chen Y, Liang B, Nice EC, Huang C, Xie N, Zheng S. A metal-organic nanoframework for efficient colorectal cancer immunotherapy by the cGAS-STING pathway activation and immune checkpoint blockade. J Nanobiotechnology 2024; 22:592. [PMID: 39343911 PMCID: PMC11441132 DOI: 10.1186/s12951-024-02836-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/04/2024] [Indexed: 10/01/2024] Open
Abstract
Immunotherapy has shown marked progress in promoting systemic anti-colorectal cancer (CRC) clinical effects. For further effectively sensitizing CRC to immunotherapy, we have engineered a pH-sensitive zeolitic imidazolate framework-8 (CS/NPs), capable of efficient cGAS-STING pathway activation and immune checkpoint blockade, by encapsulating the chemotherapeutic mitoxantrone (MTX) and immunomodulator thymus pentapeptide (TP5) and tailoring with tumor-targeting chondroitin sulfate (CS). In this nanoframework, CS endows CS/NPs with specific tumor-targeting activity and reduced systemic toxicity. Of note, the coordinated Zn2+ disrupts glycolytic processes and downregulates the expression of glucose transporter type 1 (GLUT1), thus depriving the cancer cells of their energy. Zn2+ further initiates the adenosine 5'-monophosphate activated protein kinase (AMPK) pathway, which leads to PD-L1 protein degradation and sensitizes CRC cells to immunotherapy. Moreover, the damaged double-stranded DNA during MTX treatment activates the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway, which works together with TP5 induced the proliferation and differentiation of T lymphocytes and dendritic cells to further enhance the anti-CRC immune response. Therefore, CS/NPs efficiently sensitize cells to chemotherapy and stimulate systemic antitumor immune responses both in vitro and in vivo, representing a promising strategy to increase the feasibility of CRC immunotherapy.
Collapse
Affiliation(s)
- Xiaodian Zhang
- Key Laboratory of Emergency and Trauma of Ministry of Education, Engineering Research Center for Hainan Biological Sample Resources of Major Diseases, The Hainan Branch of National Clinical Research Center for Cancer, the First Clinical College & the First Affiliated Hospital, Hainan Medical University, Haikou, 570102, China
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, 571199, China
| | - Hailong Tian
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yang Chen
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Baichuan Liang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Canhua Huang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Na Xie
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Shaojiang Zheng
- Key Laboratory of Emergency and Trauma of Ministry of Education, Engineering Research Center for Hainan Biological Sample Resources of Major Diseases, The Hainan Branch of National Clinical Research Center for Cancer, the First Clinical College & the First Affiliated Hospital, Hainan Medical University, Haikou, 570102, China.
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, 571199, China.
| |
Collapse
|
2
|
Ooi YJ, Wen Y, Zhu J, Song X, Li J. Codelivery of Doxorubicin and p53 Gene by β-Cyclodextrin-Based Supramolecular Nanoparticles Formed via Host-Guest Complexation and Electrostatic Interaction. Biomacromolecules 2024; 25:2980-2989. [PMID: 38587905 DOI: 10.1021/acs.biomac.4c00123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
We developed a supramolecular system for codelivery of doxorubicin (Dox) and p53 gene based on a β-CD-containing star-shaped cationic polymer. First, a star-shaped cationic polymer consisting of a β-CD core and 3 arms of oligoethylenimine (OEI), named CD-OEI, was used to form a supramolecular inclusion complex with hydrophobic Dox. The CD-OEI/Dox complex was subsequently used to condense plasmid DNA via electrostatic interactions to form CD-OEI/Dox/DNA polyplex nanoparticles with positive surface charges that enhanced the cellular uptake of both Dox and DNA. This supramolecular drug and gene codelivery system showed high gene transfection efficiency and effective protein expression in cancer cells. The codelivery of Dox and DNA encoding the p53 gene resulted in reduced cell viability and enhanced antitumor effects at low Dox concentrations. With its enhanced cellular uptake and anticancer efficacy, the system holds promise as a delivery carrier for potential combination cancer therapies.
Collapse
Affiliation(s)
- Ying Jie Ooi
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Yuting Wen
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Jingling Zhu
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
- NUS Environmental Research Institute (NERI), National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Xia Song
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Jun Li
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
- NUS Environmental Research Institute (NERI), National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| |
Collapse
|
3
|
Abalymov AA, Anisimov RA, Demina PA, Kildisheva VA, Kalinova AE, Serdobintsev AA, Novikova NG, Petrenko DB, Sadovnikov AV, Voronin DV, Lomova MV. Time-Delayed Anticancer Effect of an Extremely Low Frequency Alternating Magnetic Field and Multimodal Protein-Tannin-Mitoxantrone Carriers with Brillouin Microspectroscopy Visualization In Vitro. Biomedicines 2024; 12:443. [PMID: 38398045 PMCID: PMC10887239 DOI: 10.3390/biomedicines12020443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/09/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
The effect of an extremely low frequency alternating magnetic field (ELF AMF) at frequencies of 17, 48, and 95 Hz at 100 mT on free and internalized 4T1 breast cancer cell submicron magnetic mineral carriers with an anticancer drug, mitoxantrone, was shown. The alternating magnetic field (100 mT; 17, 48, 95 Hz; time of treatment-10.5 min with a 30 s delay) does not lead to the significant destruction of carrier shells and release of mitoxantrone or bovine serum albumin from them according to the data of spectrophotometry, or the heating of carriers in the process of exposure to magnetic fields. The most optimal set of factors that would lead to the suppression of proliferation and survival of cells with anticancer drug carriers on the third day (in comparison with the control and first day) is exposure to an alternating magnetic field of 100 mT in a pulsed mode with a frequency of 95 Hz. The presence of magnetic nanocarriers in cell lines was carried out by a direct label-free method, space-resolved Brillouin light scattering (BLS) spectrometry, which was realized for the first time. The analysis of the series of integrated BLS spectra showed an increase in the magnetic phase in cells with a growth in the number of particles per cell (from 10 to 100) after their internalization. The safety of magnetic carriers in the release of their constituent ions has been evaluated using atomic absorption spectrometry.
Collapse
Affiliation(s)
- Anatolii A. Abalymov
- Science Medical Centre, Saratov State University, 83 Astrakhanskayast, Saratov 410012, Russia
| | - Roman A. Anisimov
- Science Medical Centre, Saratov State University, 83 Astrakhanskayast, Saratov 410012, Russia
| | - Polina A. Demina
- Science Medical Centre, Saratov State University, 83 Astrakhanskayast, Saratov 410012, Russia
- Institute of Chemistry, Saratov State University, 83 Astrakhanskayast, Saratov 410012, Russia
| | - Veronika A. Kildisheva
- Science Medical Centre, Saratov State University, 83 Astrakhanskayast, Saratov 410012, Russia
| | - Alexandra E. Kalinova
- Institute of Physics, Saratov State University, 83 Astrakhanskayast, Saratov 410012, Russia
| | - Alexey A. Serdobintsev
- Institute of Physics, Saratov State University, 83 Astrakhanskayast, Saratov 410012, Russia
| | - Nadezhda G. Novikova
- Institute of Comprehensive Exploitation, Mineral Resources Russian Academy of Sciences, Moscow 111020, Russia
- The Core Shared Research Facility “Industrial Biotechnologies”, Aleksei Nikolayevich Bach Institute of Biochemistry, Russian Academy of Sciences, Moscow 119071, Russia
| | - Dmitry B. Petrenko
- Geological Institute, Russian Academy of Sciences, Moscow 119017, Russia
- Faculty of Natural Sciences, Department of Theoretical and Applied Chemistry, Federal State University of Education, Mytischi 141014, Russia
| | - Alexandr V. Sadovnikov
- Institute of Physics, Saratov State University, 83 Astrakhanskayast, Saratov 410012, Russia
| | - Denis V. Voronin
- Department of Physical and Colloid Chemistry, National University of Oil and Gas “Gubkin University”, Moscow 119991, Russia
| | - Maria V. Lomova
- Science Medical Centre, Saratov State University, 83 Astrakhanskayast, Saratov 410012, Russia
| |
Collapse
|
4
|
Yurttaş L, Evren AE, AlChaib H, Temel HE, Akalin Çiftçi G. Synthesis, molecular docking, and molecular dynamic simulation studies of new 1,3,4-thiadiazole derivatives as potential apoptosis inducers in A549 lung cancer cell line. J Biomol Struct Dyn 2024:1-16. [PMID: 38166545 DOI: 10.1080/07391102.2023.2300125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 12/22/2023] [Indexed: 01/04/2024]
Abstract
1,3,4-Thiadiazoles are structures that are bioisosteres of 1,3,4-oxadiazole and pyrimidine ring, which are found in the structure of many drugs and anticancer active newly studied derivatives. In the past, high effect profiles have been observed in many molecules created, based on the anticancer effects of the 2-amino-1,3,4-thiadiazole (NSC 4728) molecule and acetazolamide molecules. Focusing on these molecules and evaluating them in terms of mechanistic effects, twelve new N-[5-((3,5-dichlorophenoxy) methyl]-1,3,4-thiadiazole derivatives (3a-3i) were synthesized and their biological activities were investigated in lung cancer cells. The anticancer effects of the compounds were evaluated on the A549 and L929 cell lines. Compound 3f, namely 2-[(5-chlorobenzotiyazol-2-yl)thio]-N-[5-[(3,5-dichlorophenoxy)methyl]-1,3,4-thiadiazol-2-yl]acetamide, showed better activity than cisplatin, exhibiting high inhibitory potency (IC50: <0.98 μg/mL) and selectivity against A549 cell line even at the lowest concentration tested. Compounds 3c, 3f, and 3h with the lowest IC50 values of the compounds exhibited an excellent percentage of apoptosis between 72.48 and 91.95% compared to cisplatin. The caspase-3 activation and mitochondrial membrane potential change of the aforementioned three compounds were also studied. Moreover, matrix metalloproteinase-9 (MMP-9) inhibition potential of all final compounds was also investigated and IC50 values for compounds 3b and 3g were identified as 154.23 and 107.28 µM. Molecular docking and molecular dynamic simulation studies for MMP-9 enzyme inhibition were realized on these compounds and the nitrogen atoms of amide and thiadiazole moieties' ascertained that they play a key role in chelating with Zn metal, at the same time, (thio)ether moieties allow conformational change resulting in the ligand can make more stable contacts.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Leyla Yurttaş
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Asaf Evrim Evren
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
- Vocational School of Health Services, Şeyh Edebali University, Bilecik, Turkey
| | - Huda AlChaib
- Department of Medical Biochemistry, Faculty of Health Sciences, Eskişehir Osmangazi University, Eskişehir, Turkey
| | - Halide Edip Temel
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Gülşen Akalin Çiftçi
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| |
Collapse
|
5
|
Kowalczyk A, Piotrowicz M, Gapińska M, Trzybiński D, Woźniak K, Golding TM, Stringer T, Smith GS, Czerwieniec R, Kowalski K. Chemistry of glycol nucleic acid (GNA): Synthesis, photophysical characterization and insight into the biological activity of phenanthrenyl GNA constituents. Bioorg Chem 2022; 125:105847. [DOI: 10.1016/j.bioorg.2022.105847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 11/02/2022]
|
6
|
Molecular encapsulation by eosin yellow-β-cyclodextrin conjugate: Differential binding to quadruplex and duplex DNA. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
7
|
The Achilles' heel of cancer: targeting tumors via lysosome-induced immunogenic cell death. Cell Death Dis 2022; 13:509. [PMID: 35637197 PMCID: PMC9151667 DOI: 10.1038/s41419-022-04912-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 03/10/2022] [Accepted: 05/04/2022] [Indexed: 12/14/2022]
Abstract
Interest in the lysosome's potential role in anticancer therapies has recently been appreciated in the field of immuno-oncology. Targeting lysosomes triggers apoptotic pathways, inhibits cytoprotective autophagy, and activates a unique form of apoptosis known as immunogenic cell death (ICD). This mechanism stimulates a local and systemic immune response against dead-cell antigens. Stressors that can lead to ICD include an abundance of ROS which induce lysosome membrane permeability (LMP). Dying cells express markers that activate immune cells. Dendritic cells engulf the dying cell and then present the cell's neoantigens to T cells. The discovery of ICD-inducing agents is important due to their potential to trigger autoimmunity. In this review, we discuss the various mechanisms of activating lysosome-induced cell death in cancer cells specifically and the strategies that current laboratories are using to selectively promote LMP in tumors.
Collapse
|
8
|
Vincent MP, Navidzadeh JO, Bobbala S, Scott EA. Leveraging self-assembled nanobiomaterials for improved cancer immunotherapy. Cancer Cell 2022; 40:255-276. [PMID: 35148814 PMCID: PMC8930620 DOI: 10.1016/j.ccell.2022.01.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/22/2021] [Accepted: 01/18/2022] [Indexed: 12/12/2022]
Abstract
Nanomaterials and targeted drug delivery vehicles improve the therapeutic index of drugs and permit greater control over their pharmacokinetics, biodistribution, and bioavailability. Here, nanotechnologies applied to cancer immunotherapy are discussed with a focus on current and next generation self-assembling drug delivery systems composed of lipids and/or polymers. Topics covered include the fundamental design, suitability, and inherent properties of nanomaterials that induce anti-tumor immune responses and support anti-cancer vaccination. Established active and passive targeting strategies as well as newer "indirect" methods are presented together with insights into how nanocarrier structure and surface chemistry can be leveraged for controlled delivery to the tumor microenvironment while minimizing off-target effects.
Collapse
Affiliation(s)
- Michael P Vincent
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Justin O Navidzadeh
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Sharan Bobbala
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| | - Evan A Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA; Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL 60208, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA; Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
9
|
Ahmed OM, Galaly SR, Mostafa MAMA, Eed EM, Ali TM, Fahmy AM, Zaky MY. Thyme Oil and Thymol Counter Doxorubicin-Induced Hepatotoxicity via Modulation of Inflammation, Apoptosis, and Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6702773. [PMID: 35178158 PMCID: PMC8844103 DOI: 10.1155/2022/6702773] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/26/2021] [Accepted: 01/02/2022] [Indexed: 12/14/2022]
Abstract
Doxorubicin (DOX) is an effective anticancer agent with a wide spectrum of activities. However, it has many adverse effects on various organs especially on the liver. Thymol, one of the major components of thyme oil, has biological properties that include anti-inflammatory and antioxidant activities. Thus, this study was designed to examine thyme oil and thymol for their ability to prevent doxorubicin-induced hepatotoxicity in Wistar rats. Hepatotoxicity was induced by an intraperitoneal injection of doxorubicin, at a dose of 2 mg/kg bw/week, for seven weeks. Doxorubicin-injected rats were supplemented with thyme oil and thymol at doses 250 and 100 mg/kg bw, respectively, four times/week by oral gavage for the same period. Treatment of rats with thyme oil and thymol reversed the high serum activities of AST, ALT, and ALP and total bilirubin, AFP, and CA19.9 levels, caused by doxorubicin. Thyme oil and thymol also reduced the high levels of TNF-α and the decreased levels of both albumin and IL-4. These agents ameliorated doxorubicin-induced elevation in hepatic lipid peroxidation and associated reduction in GSH content and GST and GPx activities. Further, the supplementation with thyme oil and thymol significantly augmented mRNA expression of the level of antiapoptotic protein Bcl-2 and significantly downregulated nuclear and cytoplasmic levels of the hepatic apoptotic mediator p53. Thus, thyme oil and thymol successfully counteracted doxorubicin-induced experimental hepatotoxicity via their anti-inflammatory, antioxidant, and antiapoptotic properties.
Collapse
Affiliation(s)
- Osama M. Ahmed
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Sanaa R. Galaly
- Cell Biology and Histology Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Mennah-Allah M. A. Mostafa
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Emad M. Eed
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Tarek M. Ali
- Department of Physiology, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Alzhraa M. Fahmy
- Tropical Medicine and Infectious Diseases Department, Beni-Suef University Faculty of Medicine, Beni-Suef, Egypt
| | - Mohamed Y. Zaky
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
- Department of Medical Oncology Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
| |
Collapse
|
10
|
Soundarapandian S, Alexander A, Sumohan Pillai A, Enoch IVMV, Yousuf S. Molecular encapsulation of berberine and ethidium bromide in anthraquinonecarboxamido-β-cyclodextrin conjugate: supramolecular association with DNA duplex and G-quadruplexes. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2021; 40:542-558. [PMID: 33823737 DOI: 10.1080/15257770.2021.1907591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 02/22/2021] [Accepted: 03/20/2021] [Indexed: 10/21/2022]
Abstract
G-quadruplex DNA in recognized as a potential target for anti-cancer drugs. In this work, an anthraquinonecarboxamido derivative of β-cyclodextrin (AQCC) is synthesized as a novel DNA binder that further can deliver an additional molecule at the target, carrying it in the cavity of modified cyclodextrin. The binding of AQCC with ethidium bromide (EtBr), berberine (Ber), duplex calf-thymus DNA (CT-DNA), quadruplexes (G4) viz., kit22, myc22, and telo24 are studied. The compound acts as a host molecule for the encapsulation of DNA binders viz., EtBr, Ber and enhances their fluorescence due to the encapsulation in its AQCC's cyclodextrin cavity. The binding constant of the host: guest complex of EtBr and Ber with AQCC's cavity are 6.4 × 105 and 3.3 × 106 mol-1 dm3, respectively. The proximity of the protons of the guest and host molecules is confirmed by two-dimensional rotating-frame Overhauser effect spectroscopy (2D ROESY). The conjugate displays a quenching of fluorescence selectively on the association with CT-DNA and quadruplex kit22 that is contrast to the spectral behavior with quadruplex myc22 and telo24. CT-DNA exhibits dissimilar fluorescence spectra in free- and EtBr-bound forms. In addition, kit22 exhibit dissimilar emission profile when AQCC encapsulates Ber. Therefore, the Ber-loaded complexes and the AQCC molecule bind to different G-quadruplexes with different binding strengths. In addition, the effect of Ber in binding to the target DNAs is pronounces since the Ber molecule has more affinity to bind to quadruplexes than the duplex.
Collapse
Affiliation(s)
- Suganthi Soundarapandian
- Department of Chemistry, Karunya Institute of Technology and Sciences (Deemed-to-be University), Coimbatore, Tamil Nadu, India
| | - Aleyamma Alexander
- Centre for Nanoscience & Genomics, Karunya Institute of Technology and Sciences (Deemed-to-be University), Coimbatore, Tamil Nadu, India
| | - Archana Sumohan Pillai
- Centre for Nanoscience & Genomics, Karunya Institute of Technology and Sciences (Deemed-to-be University), Coimbatore, Tamil Nadu, India
| | - Israel V M V Enoch
- Centre for Nanoscience & Genomics, Karunya Institute of Technology and Sciences (Deemed-to-be University), Coimbatore, Tamil Nadu, India
| | - Sameena Yousuf
- Department of Chemistry, Sri Shakthi Institute of Engineering and Technology, Coimbatore, Tamil Nadu, India
| |
Collapse
|
11
|
Jin X, Zeng Q, Zheng J, Xing D, Zhang T. Aptamer-Functionalized Upconverting Nanoformulations for Light-Switching Cancer-Specific Recognition and In Situ Photodynamic-Chemo Sequential Theranostics. ACS APPLIED MATERIALS & INTERFACES 2021; 13:9316-9328. [PMID: 33089995 DOI: 10.1021/acsami.0c14730] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Biomarker-activatable theranostic formulations offer the potential for removing specific tumors with a high diagnostic accuracy and a significant pharmacological effect. Herein, we developed a novel activatable theranostic nanoformulation UAS-PD [upconversion nanophosphor (UCNP)-aptamer/ssDNA-pyropheophorbide-a (PPA)-doxyrubicin (DOX)], which can recognize specific cancer cells with sensitivity and trigger the localized photodynamic destruction and enhanced chemotherapy. UAS-PD was constructed by the conjugation of UCNPs and aptamer probes containing the photosensitizer PPA and the chemotherapeutic drug DOX. When cancer cells are present, the UAS-PD specifically binds to PTK7, an overexpressed protein present on the surface of cancer cells, through conformational recombination of the aptamer structure and switches its upconversion luminescence from 655 to 540 nm. This long-lived ratiometric optical signal provides an ultrasensitive detection limit as low as 3.9 nM for PTK7. Changes in the conformation of UAS-PD can also induce PPA to approach UCNPs, which can produce cytotoxic singlet oxygens under near-infrared excitation to destroy the cell membrane and enhance its permeability for the simultaneously released DOX that targets cellular DNA degradation, which results in a highly effective tumor-killing effect by synergistic extra-intracellular sequential damage.
Collapse
Affiliation(s)
- Xudong Jin
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
| | - Qin Zeng
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
| | - Judun Zheng
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
| | - Da Xing
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
| | - Tao Zhang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
| |
Collapse
|
12
|
Mitochondrion targeting peptide-modified magnetic graphene oxide delivering mitoxantrone for impairment of tumor mitochondrial functions. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2020.09.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
13
|
Egorova KS, Posvyatenko AV, Fakhrutdinov AN, Galushko AS, Seitkalieva MM, Ananikov VP. Synergistic/antagonistic cytotoxic effects in mixtures of ionic liquids with doxorubicin or mitoxantrone. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2020.114870] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
14
|
Czepas J, Matczak K, Koceva-Chyła A, Grobelski B, Jóźwiak Z, Gwoździński K. Doxyl Nitroxide Spin Probes Can Modify Toxicity of Doxorubicin towards Fibroblast Cells. Molecules 2020; 25:E5138. [PMID: 33158261 PMCID: PMC7663118 DOI: 10.3390/molecules25215138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 11/16/2022] Open
Abstract
The biological properties of doxyl stearate nitroxides (DSs): 5-DS, Met-12-DS, and 16-DS, commonly used as spin probes, have not been explored in much detail so far. Furthermore, the influence of DSs on the cellular changes induced by the anticancer drug doxorubicin (DOX) has not yet been investigated. Therefore, we examined the cytotoxicity of DSs and their ability to induce cell death and to influence on fluidity and lipid peroxidation (LPO) in the plasma membrane of immortalised B14 fibroblasts, used as a model neoplastic cells, susceptible to DOX-induced changes. The influence of DSs on DOX toxicity was also investigated and compared with that of a natural reference antioxidant α-Tocopherol. By employing the trypan blue exclusion test and double fluorescent staining, we found a significant level of cytotoxicity for DSs and showed that their ability to induce apoptosis and modify plasma membrane fluidity (measured fluorimetrically) is more potent than for α-Tocopherol. The most cytotoxic nitroxide was 5-DS. The electron paramagnetic resonance (EPR) measurements revealed that 5-DS was reduced in B14 cells at the fastest and Met-12-DS at the slowest rate. In the presence of DOX, DSs were reduced slower than alone. The investigated compounds, administered with DOX, enhanced DOX-induced cell death and demonstrated concentration-dependent biphasic influence on membrane fluidity. A-Tocopherol showed weaker effects than DSs, regardless the mode of its application-alone or with DOX. High concentrations of α-Tocopherol and DSs decreased DOX-induced LPO. Substantial cytotoxicity of the DSs suggests that they should be used more carefully in the investigations performed on sensitive cells. Enhancement of DOX toxicity by DSs showed their potential to act as chemosensitizers of cancer cells to anthracycline chemotherapy.
Collapse
Affiliation(s)
- Jan Czepas
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Łódź, 141/143 Pomorska st., 90-236 Łódź, Poland; (B.G.); (K.G.)
| | - Karolina Matczak
- Department of Medical Biophysics, Faculty of Biology and Environmental Protection, University of Łódź, 141/143 Pomorska st., 90-236 Łódź, Poland; (K.M.); (A.K.-C.); (Z.J.)
| | - Aneta Koceva-Chyła
- Department of Medical Biophysics, Faculty of Biology and Environmental Protection, University of Łódź, 141/143 Pomorska st., 90-236 Łódź, Poland; (K.M.); (A.K.-C.); (Z.J.)
| | - Bartłomiej Grobelski
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Łódź, 141/143 Pomorska st., 90-236 Łódź, Poland; (B.G.); (K.G.)
| | - Zofia Jóźwiak
- Department of Medical Biophysics, Faculty of Biology and Environmental Protection, University of Łódź, 141/143 Pomorska st., 90-236 Łódź, Poland; (K.M.); (A.K.-C.); (Z.J.)
| | - Krzysztof Gwoździński
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Łódź, 141/143 Pomorska st., 90-236 Łódź, Poland; (B.G.); (K.G.)
| |
Collapse
|
15
|
Lee SJ, Kwon OS. O-GlcNAc Transferase Inhibitor Synergistically Enhances Doxorubicin-Induced Apoptosis in HepG2 Cells. Cancers (Basel) 2020; 12:cancers12113154. [PMID: 33121131 PMCID: PMC7693581 DOI: 10.3390/cancers12113154] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 10/19/2020] [Accepted: 10/22/2020] [Indexed: 01/31/2023] Open
Abstract
Simple Summary We found that the combination treatment of doxorubicin (DOX) and O-GlcNAc transferase (OGT) inhibitor OSMI-1 has synergic therapeutic efficacy in the treatment of liver cancer. Our data show that DOX displayed cytotoxicity via the activation of p53 and the inflammatory NF-κB signaling pathway, while OSMI-1 evoked the ER stress response and inhibited NF-κB signaling. Therefore, DOX in combination with the OSMI-1 group showed a 20-fold reduction of tumor formation, whereas the DOX alone group reduced by 1.8-fold compared with control in a HepG2 cell xenograft model. Abstract The combination of chemotherapy with chemosensitizing agents is a common approach to enhance anticancer activity while reducing the dose-dependent adverse side effects of cancer treatment. Herein, we investigated doxorubicin (DOX) and O-GlcNAc transferase (OGT) inhibitor OSMI-1 combination treatment, which significantly enhanced apoptosis in hepatocellular carcinoma cells (HepG2) as a result of synergistic drug action in disparate stress signaling pathways. Treatment with a low dose of DOX or a suboptimal dose of OSMI-1 alone did not induce apoptotic cell death in HepG2 cells. However, the combination of DOX with OSMI-1 in HepG2 cells synergistically increased apoptotic cell death through the activation of both the p53 and mitochondrial Bcl2 pathways compared to DOX alone. We also demonstrated that the combination of DOX and OSMI-1 stimulated cell death, dramatically reducing cell proliferation and tumor growth in vivo using a HepG2 xenograft mouse model. These findings indicate that OSMI-1 acts as a potential chemosensitizer by enhancing DOX-induced cell death. This study provides insight into a possible mechanism of chemotherapy resistance, identifies potential novel drug targets, and suggests that OGT inhibition could be utilized in clinical applications to treat hepatocellular carcinoma as well as other cancer types.
Collapse
|
16
|
Costa VM, Capela JP, Sousa JR, Eleutério RP, Rodrigues PRS, Dores-Sousa JL, Carvalho RA, Lourdes Bastos M, Duarte JA, Remião F, Almeida MG, Varner KJ, Carvalho F. Mitoxantrone impairs proteasome activity and prompts early energetic and proteomic changes in HL-1 cardiomyocytes at clinically relevant concentrations. Arch Toxicol 2020; 94:4067-4084. [DOI: 10.1007/s00204-020-02874-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/12/2020] [Indexed: 11/24/2022]
|
17
|
Alfarouk KO, Ahmed SBM, Elliott RL, Benoit A, Alqahtani SS, Ibrahim ME, Bashir AHH, Alhoufie STS, Elhassan GO, Wales CC, Schwartz LH, Ali HS, Ahmed A, Forde PF, Devesa J, Cardone RA, Fais S, Harguindey S, Reshkin SJ. The Pentose Phosphate Pathway Dynamics in Cancer and Its Dependency on Intracellular pH. Metabolites 2020; 10:E285. [PMID: 32664469 PMCID: PMC7407102 DOI: 10.3390/metabo10070285] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 12/21/2022] Open
Abstract
The Pentose Phosphate Pathway (PPP) is one of the key metabolic pathways occurring in living cells to produce energy and maintain cellular homeostasis. Cancer cells have higher cytoplasmic utilization of glucose (glycolysis), even in the presence of oxygen; this is known as the "Warburg Effect". However, cytoplasmic glucose utilization can also occur in cancer through the PPP. This pathway contributes to cancer cells by operating in many different ways: (i) as a defense mechanism via the reduced form of nicotinamide adenine dinucleotide phosphate (NADPH) to prevent apoptosis, (ii) as a provision for the maintenance of energy by intermediate glycolysis, (iii) by increasing genomic material to the cellular pool of nucleic acid bases, (iv) by promoting survival through increasing glycolysis, and so increasing acid production, and (v) by inducing cellular proliferation by the synthesis of nucleic acid, fatty acid, and amino acid. Each step of the PPP can be upregulated in some types of cancer but not in others. An interesting aspect of this metabolic pathway is the shared regulation of the glycolytic and PPP pathways by intracellular pH (pHi). Indeed, as with glycolysis, the optimum activity of the enzymes driving the PPP occurs at an alkaline pHi, which is compatible with the cytoplasmic pH of cancer cells. Here, we outline each step of the PPP and discuss its possible correlation with cancer.
Collapse
Affiliation(s)
- Khalid O. Alfarouk
- Alfarouk Biomedical Research LLC, Temple Terrace, FL 33617, USA
- American Biosciences Inc., New York, NY 10913, USA;
- Al-Ghad International College for Applied Medical Sciences, Al-Madinah Al-Munawarah 42316, Saudi Arabia
| | | | - Robert L. Elliott
- The Elliott-Elliott-Baucom Breast Cancer Research and Treatment Center, Baton Rouge, LA 70806, USA;
- The Sallie A. Burdine Breast Foundation, Baton Rouge, LA 70806, USA;
| | - Amanda Benoit
- The Sallie A. Burdine Breast Foundation, Baton Rouge, LA 70806, USA;
| | - Saad S. Alqahtani
- Clinical Pharmacy Department, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
| | - Muntaser E. Ibrahim
- Institute of Endemic Diseases, University of Khartoum, Khartoum 11111, Sudan; (M.E.I.); (A.H.H.B.)
| | - Adil H. H. Bashir
- Institute of Endemic Diseases, University of Khartoum, Khartoum 11111, Sudan; (M.E.I.); (A.H.H.B.)
| | - Sari T. S. Alhoufie
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Taibah University, Al-Madinah Al-Munwarah 42353, Saudi Arabia;
| | - Gamal O. Elhassan
- Unaizah College of Pharmacy, Qassim University, Unaizah 56264, Saudi Arabia;
| | | | | | - Heyam S. Ali
- Department of Pharmaceutics, Faculty of Pharmacy, University of Khartoum, Khartoum 11111, Sudan;
| | - Ahmed Ahmed
- Department of Oesphogastric and General Surgery, University Hospitals of Leicester, Leicester LE5 4PW, UK;
| | - Patrick F. Forde
- CancerResearch@UCC, Western Gateway Building, University College Cork, Cork T12 XF62, Ireland;
| | - Jesus Devesa
- Scientific Direction, Foltra Medical Centre, Travesía de Montouto 24, 15886 Teo, Spain;
| | - Rosa A. Cardone
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, 90126 Bari, Italy; (R.A.C.); (S.J.R.)
| | - Stefano Fais
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| | - Salvador Harguindey
- Department of Oncology, Institute for Clinical Biology and Metabolism, 01004 Vitoria, Spain;
| | - Stephan J. Reshkin
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, 90126 Bari, Italy; (R.A.C.); (S.J.R.)
| |
Collapse
|
18
|
Murray M, Roseblade A, Chen Y, Bourget K, Rawling T. Carbon Chain Length Modulates MDA‐MB‐231 Breast Cancer Cell Killing Mechanisms by Mitochondrially Targeted Aryl−Urea Fatty Acids. ChemMedChem 2020; 15:247-255. [DOI: 10.1002/cmdc.201900577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Indexed: 01/06/2023]
Affiliation(s)
- Michael Murray
- Discipline of Pharmacology School of Medical Sciences Faculty of Medicine and HealthUniversity of Sydney Camperdown, New South Wales 2006 Australia
| | - Ariane Roseblade
- School of Mathematical and Physical Sciences Faculty of ScienceUniversity of Technology Sydney Ultimo, New South Wales 2007 Australia
| | - Yongjuan Chen
- Discipline of Pharmacology School of Medical Sciences Faculty of Medicine and HealthUniversity of Sydney Camperdown, New South Wales 2006 Australia
| | - Kirsi Bourget
- Discipline of Pharmacology School of Medical Sciences Faculty of Medicine and HealthUniversity of Sydney Camperdown, New South Wales 2006 Australia
| | - Tristan Rawling
- School of Mathematical and Physical Sciences Faculty of ScienceUniversity of Technology Sydney Ultimo, New South Wales 2007 Australia
| |
Collapse
|
19
|
Feilcke R, Arnouk G, Raphane B, Richard K, Tietjen I, Andrae-Marobela K, Erdmann F, Schipper S, Becker K, Arnold N, Frolov A, Reiling N, Imming P, Fobofou SAT. Biological activity and stability analyses of knipholone anthrone, a phenyl anthraquinone derivative isolated from Kniphofia foliosa Hochst. J Pharm Biomed Anal 2019; 174:277-285. [PMID: 31185339 DOI: 10.1016/j.jpba.2019.05.065] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 05/13/2019] [Accepted: 05/29/2019] [Indexed: 12/20/2022]
Abstract
Knipholone (1) and knipholone anthrone (2), isolated from the Ethiopian medicinal plant Kniphofia foliosa Hochst. are two phenyl anthraquinone derivatives, a compound class known for biological activity. In the present study, we describe the activity of both 1 and 2 in several biological assays including cytotoxicity against four human cell lines (Jurkat, HEK293, SH-SY5Y and HT-29), antiplasmodial activity against Plasmodium falciparum 3D7 strain, anthelmintic activity against the model organism Caenorhabditis elegans, antibacterial activity against Aliivibrio fischeri and Mycobacterium tuberculosis and anti-HIV-1 activity in peripheral blood mononuclear cells (PBMCs) infected with HIV-1c. In parallel, we investigated the stability of knipholone (2) in solution and in culture media. Compound 1 displays strong cytotoxicity against Jurkat, HEK293 and SH-SY5Y cells with growth inhibition ranging from approximately 62-95% when added to cells at 50 μM, whereas KA (2) exhibits weak to strong activity with 26, 48 and 70% inhibition of cell growth, respectively. Both 1 and 2 possess significant antiplasmodial activity against Plasmodium falciparum 3D7 strain with IC50 values of 1.9 and 0.7 μM, respectively. These results complement previously reported data on the cytotoxicity and antiplasmodial activity of 1 and 2. Furthermore, compound 2 showed HIV-1c replication inhibition (growth inhibition higher than 60% at tested concentrations 0.5, 5, 15 and 50 μg/ml and an EC50 value of 4.3 μM) associated with cytotoxicity against uninfected PBMCs. The stability study based on preincubation, HPLC and APCI-MS (atmospheric-pressure chemical ionization mass spectrometry) analysis indicates that compound 2 is unstable in culture media and readily oxidizes to form compound 1. Therefore, the biological activity attributed to 2 might be influenced by its degradation products in media including 1 and other possible dimers. Hence, bioactivity results previously reported from this compound should be taken with caution and checked if they differ from those of its degradation products. To the best of our knowledge, this is the first report on the anti-HIV activity and stability analysis of compound 2.
Collapse
Affiliation(s)
- Ruth Feilcke
- Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle (Saale), Germany
| | - Georgette Arnouk
- Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle (Saale), Germany
| | - Boingotlo Raphane
- Department of Biological Sciences, Faculty of Science, University of Botswana, Block 235, Private Bag, 0022 Gaborone, Botswana
| | - Khumoekae Richard
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Ian Tietjen
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Kerstin Andrae-Marobela
- Department of Biological Sciences, Faculty of Science, University of Botswana, Block 235, Private Bag, 0022 Gaborone, Botswana
| | - Frank Erdmann
- Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle (Saale), Germany
| | - Susanne Schipper
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University Giessen, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany
| | - Katja Becker
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University Giessen, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany
| | - Norbert Arnold
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120 Halle (Saale), Germany
| | - Andrej Frolov
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120 Halle (Saale), Germany; Department of Biochemistry, St. Petersburg State University, 199904 St. Petersburg, Russia
| | - Norbert Reiling
- Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel, 23845 Borstel, Germany
| | - Peter Imming
- Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle (Saale), Germany
| | - Serge A T Fobofou
- Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle (Saale), Germany; Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120 Halle (Saale), Germany.
| |
Collapse
|
20
|
Janockova J, Korabecny J, Plsikova J, Babkova K, Konkolova E, Kucerova D, Vargova J, Koval J, Jendzelovsky R, Fedorocko P, Kasparkova J, Brabec V, Rosocha J, Soukup O, Hamulakova S, Kuca K, Kozurkova M. In vitro investigating of anticancer activity of new 7-MEOTA-tacrine heterodimers. J Enzyme Inhib Med Chem 2019; 34:877-897. [PMID: 30938202 PMCID: PMC6450562 DOI: 10.1080/14756366.2019.1593159] [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] [Indexed: 10/31/2022] Open
Abstract
A combination of biochemical, biophysical and biological techniques was used to study calf thymus DNA interaction with newly synthesized 7-MEOTA-tacrine thiourea 12-17 and urea heterodimers 18-22, and to measure interference with type I and II topoisomerases. Their biological profile was also inspected in vitro on the HL-60 cell line using different flow cytometric techniques (cell cycle distribution, detection of mitochondrial membrane potential dissipation, and analysis of metabolic activity/viability). The compounds exhibited a profound inhibitory effect on topoisomerase activity (e.g. compound 22 inhibited type I topoisomerase at 1 µM concentration). The treatment of HL-60 cells with the studied compounds showed inhibition of cell growth especially with hybrids containing thiourea (14-17) and urea moieties (21 and 22). Moreover, treatment of human dermal fibroblasts with the studied compounds did not indicate significant cytotoxicity. The observed results suggest beneficial selectivity of the heterodimers as potential drugs to target cancer cells.
Collapse
Affiliation(s)
- Jana Janockova
- a Department of Biochemistry, Institute of Chemistry, Faculty of Science , P. J. Šafárik University , Kosice , Slovak Republic.,b Biomedical Research Center , University Hospital Hradec Kralove , Hradec Kralove , Czech Republic
| | - Jan Korabecny
- b Biomedical Research Center , University Hospital Hradec Kralove , Hradec Kralove , Czech Republic.,c Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences , University of Defence , Hradec Kralove , Czech Republic
| | - Jana Plsikova
- a Department of Biochemistry, Institute of Chemistry, Faculty of Science , P. J. Šafárik University , Kosice , Slovak Republic.,d Associated Tissue Bank, Faculty of Medicine , P.J. Šafárik University , Kosice , Slovak Republic
| | - Katerina Babkova
- b Biomedical Research Center , University Hospital Hradec Kralove , Hradec Kralove , Czech Republic.,c Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences , University of Defence , Hradec Kralove , Czech Republic
| | - Eva Konkolova
- a Department of Biochemistry, Institute of Chemistry, Faculty of Science , P. J. Šafárik University , Kosice , Slovak Republic
| | - Dana Kucerova
- e Department of Cellular Biology, Institute of Biology and Ecology, Faculty of Science , P. J. Šafárik University , Kosice , Slovak Republic
| | - Jana Vargova
- e Department of Cellular Biology, Institute of Biology and Ecology, Faculty of Science , P. J. Šafárik University , Kosice , Slovak Republic
| | - Jan Koval
- e Department of Cellular Biology, Institute of Biology and Ecology, Faculty of Science , P. J. Šafárik University , Kosice , Slovak Republic
| | - Rastislav Jendzelovsky
- e Department of Cellular Biology, Institute of Biology and Ecology, Faculty of Science , P. J. Šafárik University , Kosice , Slovak Republic
| | - Peter Fedorocko
- e Department of Cellular Biology, Institute of Biology and Ecology, Faculty of Science , P. J. Šafárik University , Kosice , Slovak Republic
| | - Jana Kasparkova
- f Department of Biophysics, Faculty of Science , Palacke University , Olomouc , Czech Republic
| | - Viktor Brabec
- f Department of Biophysics, Faculty of Science , Palacke University , Olomouc , Czech Republic
| | - Jan Rosocha
- d Associated Tissue Bank, Faculty of Medicine , P.J. Šafárik University , Kosice , Slovak Republic
| | - Ondrej Soukup
- b Biomedical Research Center , University Hospital Hradec Kralove , Hradec Kralove , Czech Republic.,c Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences , University of Defence , Hradec Kralove , Czech Republic
| | - Slavka Hamulakova
- g Department of Organic Chemistry, Institute of Chemistry, Faculty of Science , P. J. Šafárik University , Kosice , Slovak Republic
| | - Kamil Kuca
- b Biomedical Research Center , University Hospital Hradec Kralove , Hradec Kralove , Czech Republic
| | - Maria Kozurkova
- a Department of Biochemistry, Institute of Chemistry, Faculty of Science , P. J. Šafárik University , Kosice , Slovak Republic.,b Biomedical Research Center , University Hospital Hradec Kralove , Hradec Kralove , Czech Republic
| |
Collapse
|
21
|
Tian W, Li J, Su Z, Lan F, Li Z, Liang D, Wang C, Li D, Hou H. Novel Anthraquinone Compounds Induce Cancer Cell Death through Paraptosis. ACS Med Chem Lett 2019; 10:732-736. [PMID: 31097991 DOI: 10.1021/acsmedchemlett.8b00624] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/25/2019] [Indexed: 12/14/2022] Open
Abstract
Novel anthraquinone compounds that induce ER stress and paraptosis-like cell death were designed and synthesized. Compound 4a is the first organic micromolecule to kill tumor cells by only paraptosis, and its mechanism of action has been further explored. Paraptosis does not appear to involve either phosphatidylserine translocation associated with apoptosis or cell cycle arrest. The bisbenzyloxy and N-(2-hydroxyethyl)formamide structures may be two critical pharmacophores for paraptosis. Bisbenzyloxy can induce ER stress, and the N-(2-hydroxyethyl)formamide structure can increase the ratio of LC3II/I and cytoplasmic vacuolization and facilitates paraptosis. Some antitumor drugs fail to eradicate malignant cell lines with impaired apoptotic pathways; paraptosis may be a route to kill such cells and provides a new potential strategy for cancer chemotherapy.
Collapse
Affiliation(s)
- Wei Tian
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Junying Li
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Zhengying Su
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Fu Lan
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Zhaoquan Li
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Dandan Liang
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Chunmiao Wang
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Danrong Li
- Life Sciences Institute, Guangxi Medical University, Nanning 530021, China
| | - Huaxin Hou
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
22
|
Gajek A, Rogalska A, Koceva-Chyła A. Aclarubicin in subtoxic doses reduces doxorubicin cytotoxicity in human non-small cell lung adenocarcinoma (A549) and human hepatocellular carcinoma (HepG2) cells by decreasing DNA damage. Toxicol In Vitro 2019; 55:140-150. [DOI: 10.1016/j.tiv.2018.12.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/16/2018] [Accepted: 12/19/2018] [Indexed: 10/27/2022]
|
23
|
Awasthi P, Vatsal M, Sharma A. Structural and biological study of synthesized anthraquinone series of compounds with sulfonamide feature. J Biomol Struct Dyn 2019; 37:4465-4480. [DOI: 10.1080/07391102.2018.1552198] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Pamita Awasthi
- Department of Chemistry, National Institute of Technology, Hamirpur, Himachal Pradesh, India
| | - Manu Vatsal
- Department of Chemistry, National Institute of Technology, Hamirpur, Himachal Pradesh, India
| | - Anjali Sharma
- Department of Chemistry, National Institute of Technology, Hamirpur, Himachal Pradesh, India
| |
Collapse
|
24
|
Li S, Zhao J, Shang D, Kass DJ, Zhao Y. Ubiquitination and deubiquitination emerge as players in idiopathic pulmonary fibrosis pathogenesis and treatment. JCI Insight 2018; 3:120362. [PMID: 29769446 DOI: 10.1172/jci.insight.120362] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal fibrotic lung disease that is associated with aberrant activation of TGF-β, myofibroblast differentiation, and abnormal extracellular matrix (ECM) production. Proper regulation of protein stability is important for maintenance of intracellular protein homeostasis and signaling. Ubiquitin E3 ligases mediate protein ubiquitination, and deubiquitinating enzymes (DUBs) reverse the process. The role of ubiquitin E3 ligases and DUBs in the pathogenesis of IPF is relatively unexplored. In this review, we provide an overview of how ubiquitin E3 ligases and DUBs modulate pulmonary fibrosis through regulation of both TGF-β-dependent and -independent pathways. We also summarize currently available small-molecule inhibitors of ubiquitin E3 ligases and DUBs as potential therapeutic strategies for the treatment of IPF.
Collapse
Affiliation(s)
- Shuang Li
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of General Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jing Zhao
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Acute Lung Injury Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Dong Shang
- Department of General Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Daniel J Kass
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yutong Zhao
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Acute Lung Injury Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
25
|
Almeida D, Pinho R, Correia V, Soares J, Bastos MDL, Carvalho F, Capela JP, Costa VM. Mitoxantrone is More Toxic than Doxorubicin in SH-SY5Y Human Cells: A 'Chemobrain' In Vitro Study. Pharmaceuticals (Basel) 2018; 11:ph11020041. [PMID: 29734752 PMCID: PMC6027466 DOI: 10.3390/ph11020041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/28/2018] [Accepted: 04/29/2018] [Indexed: 12/20/2022] Open
Abstract
The potential neurotoxic effects of anticancer drugs, like doxorubicin (DOX) and mitoxantrone (MTX; also used in multiple sclerosis), are presently important reasons for concern, following epidemiological data indicating that cancer survivors submitted to chemotherapy may suffer cognitive deficits. We evaluated the in vitro neurotoxicity of two commonly used chemotherapeutic drugs, DOX and MTX, and study their underlying mechanisms in the SH-SY5Y human neuronal cell model. Undifferentiated human SH-SY5Y cells were exposed to DOX or MTX (0.13, 0.2 and 0.5 μM) for 48 h and two cytotoxicity assays were performed, the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium (MTT) reduction and the neutral red (NR) incorporation assays. Phase contrast microphotographs, Hoechst, and acridine orange/ethidium bromide stains were performed. Mitochondrial membrane potential was also assessed. Moreover, putative protective drugs, namely the antioxidants N-acetyl-l-cysteine (NAC; 1 mM) and 100 μM tiron, the inhibitor of caspase-3/7, Ac-DEVD-CHO (100 μM), and a protein synthesis inhibitor, cycloheximide (CHX; 10 nM), were tested to prevent DOX- or MTX-induced toxicity. The MTT reduction assay was also done in differentiated SH-SY5Y cells following exposure to 0.2 μM DOX or MTX. MTX was more toxic than DOX in both cytotoxicity assays and according to the morphological analyses. MTX also evoked a higher number of apoptotic nuclei than DOX. Both drugs, at the 0.13 μM concentration, caused mitochondrial membrane potential depolarization after a 48-h exposure. Regarding the putative neuroprotectors, 1 mM NAC was not able to prevent the cytotoxicity caused by either drug. Notwithstanding, 100 μM tiron was capable of partially reverting MTX-induced cytotoxicity in the NR uptake assay. One hundred μM Ac-DEVD-CHO and 10 nM cycloheximide (CHX) also partially prevented the toxicity induced by DOX in the NR uptake assay. MTX was more toxic than DOX in differentiated SH-SY5Y cells, while MTX had similar toxicity in differentiated and undifferentiated SH-SY5Y cells. In fact, MTX was the most neurotoxic drug tested and the mechanisms involved seem dissimilar among drugs. Thus, its toxicity mechanisms need to be further investigated as to determine the putative neurotoxicity for multiple sclerosis and cancer patients.
Collapse
Affiliation(s)
- Daniela Almeida
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Rita Pinho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Verónica Correia
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Jorge Soares
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Maria de Lourdes Bastos
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Félix Carvalho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - João Paulo Capela
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
- FP-ENAS (Unidade de Investigação UFP em Energia, Ambiente e Saúde), CEBIMED (Centro de Estudos em Biomedicina), Faculdade de Ciências da Saúde, Universidade Fernando Pessoa, 4249-004 Porto, Portugal.
| | - Vera Marisa Costa
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
26
|
Jabłoński A, Matczak K, Koceva-Chyła A, Durka K, Steverding D, Jakubiec-Krześniak K, Solecka J, Trzybiński D, Woźniak K, Andreu V, Mendoza G, Arruebo M, Kochel K, Krawczyk B, Szczukocki D, Kowalski K. Cymantrenyl-Nucleobases: Synthesis, Anticancer, Antitrypanosomal and Antimicrobial Activity Studies. Molecules 2017; 22:E2220. [PMID: 29240697 PMCID: PMC6149849 DOI: 10.3390/molecules22122220] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 12/11/2017] [Accepted: 12/12/2017] [Indexed: 11/17/2022] Open
Abstract
The synthesis of four cymantrene-5-fluorouracil derivatives (1-4) and two cymantrene-adenine derivatives (5 and 6) is reported. All of the compounds were characterized by spectroscopic methods and the crystal structure of two derivatives (1 and 6), together with the previously described cymantrene-adenine compound C was determined by X-ray crystallography. While the compounds 1 and 6 crystallized in the triclinic P-1 space group, compound C crystallized in the monoclinic P2₁/m space group. The newly synthesized compounds 1-6 were tested together with the two previously described cymantrene derivatives B and C for their in vitro antiproliferative activity against seven cancer cell lines (MCF-7, MCF-7/DX, MDA-MB-231, SKOV-3, A549, HepG2m and U-87-MG), five bacterial strains Staphylococcus aureus (methicillin-sensitive, methicillin-resistant and vancomycin-intermediate strains), Staphylococcus epidermidis, and Escherichia coli, including clinical isolates of S. aureus and S. epidermidis, as well as against the protozoan parasite Trypanosoma brucei. The most cytotoxic compounds were derivatives 2 and C for A549 and SKOV-3 cancer cell lines, respectively, with 50% growth inhibition (IC50) values of about 7 µM. The anticancer activity of the cymantrene compounds was determined to be due to their ability to induce oxidative stress and to trigger apoptosis and autophagy in cancer cells. Three derivatives (1, 4 and 5) displayed promising antitrypanosomal activity, with GI50 values in the low micromolar range (3-4 µM). The introduction of the 5-fluorouracil moiety in 1 enhanced the trypanocidal activity when compared to the activity previously reported for the corresponding uracil derivative. The antibacterial activity of cymantrene compounds 1 and C was within the range of 8-64 µg/mL and seemed to be the result of induced cell shrinking.
Collapse
Affiliation(s)
- Artur Jabłoński
- Department of Organic Chemistry, Faculty of Chemistry, University of Łódź, Tamka 12, 91-403 Łódź, Poland;
| | - Karolina Matczak
- Department of Medical Biophysics, Faculty of Biology and Environmental Protection, University of Łódź, Pomorska 141/143, 90-236 Łódź, Poland; (K.M.); (A.K.-C.); (K.D.); (Kr.K.)
| | - Aneta Koceva-Chyła
- Department of Medical Biophysics, Faculty of Biology and Environmental Protection, University of Łódź, Pomorska 141/143, 90-236 Łódź, Poland; (K.M.); (A.K.-C.); (K.D.); (Kr.K.)
| | - Kamil Durka
- Department of Medical Biophysics, Faculty of Biology and Environmental Protection, University of Łódź, Pomorska 141/143, 90-236 Łódź, Poland; (K.M.); (A.K.-C.); (K.D.); (Kr.K.)
| | - Dietmar Steverding
- Bob Champion Research & Education Building, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK;
| | - Katarzyna Jakubiec-Krześniak
- National Institute of Public Health-National Institute of Hygiene, Chocimska 24, 00-791 Warszawa, Poland; (K.J.-K.); (J.S.)
| | - Jolanta Solecka
- National Institute of Public Health-National Institute of Hygiene, Chocimska 24, 00-791 Warszawa, Poland; (K.J.-K.); (J.S.)
| | - Damian Trzybiński
- Biological and Chemical Research Centre, Department of Chemistry, University of Warsaw, Żwirki and Wigury 101, 02-089 Warszawa, Poland; (D.T.); (K.W.)
| | - Krzysztof Woźniak
- Biological and Chemical Research Centre, Department of Chemistry, University of Warsaw, Żwirki and Wigury 101, 02-089 Warszawa, Poland; (D.T.); (K.W.)
| | - Vanesa Andreu
- Department of Chemical Engineering. Aragon Institute of Nanoscience (INA), University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain; (V.A.); (G.M.); (M.A.)
| | - Gracia Mendoza
- Department of Chemical Engineering. Aragon Institute of Nanoscience (INA), University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain; (V.A.); (G.M.); (M.A.)
| | - Manuel Arruebo
- Department of Chemical Engineering. Aragon Institute of Nanoscience (INA), University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain; (V.A.); (G.M.); (M.A.)
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, 28029 Madrid, Spain
| | - Krzysztof Kochel
- Department of Medical Biophysics, Faculty of Biology and Environmental Protection, University of Łódź, Pomorska 141/143, 90-236 Łódź, Poland; (K.M.); (A.K.-C.); (K.D.); (Kr.K.)
| | - Barbara Krawczyk
- Faculty of Chemistry, Department of Inorganic and Analytical Chemistry, University of Łódź, Tamka 12, 91-403 Łódź, Poland; (B.K.); (D.Sz.)
| | - Dominik Szczukocki
- Faculty of Chemistry, Department of Inorganic and Analytical Chemistry, University of Łódź, Tamka 12, 91-403 Łódź, Poland; (B.K.); (D.Sz.)
| | - Konrad Kowalski
- Department of Organic Chemistry, Faculty of Chemistry, University of Łódź, Tamka 12, 91-403 Łódź, Poland;
| |
Collapse
|
27
|
Buranrat B, Mairuae N, Kanchanarach W. Cytotoxic and antimigratory effects of Cratoxy formosum extract against HepG2 liver cancer cells. Biomed Rep 2017; 6:441-448. [PMID: 28413643 DOI: 10.3892/br.2017.871] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/20/2017] [Indexed: 12/24/2022] Open
Abstract
The aim of the present study was to investigate the molecular mechanisms underlying Cratoxylum formosum (CF) Dyer-induced cancer cell death and antimigratory effects in HepG2 liver cancer cells. The cytotoxic, antiproliferative and antimigratory effects of CF leaf extract on human liver cancer HepG2 cell lines were evaluated using sulforhodamine B, colony formation, and wound healing assays. In addition, apoptosis induction mechanisms were investigated via reactive oxygen species (ROS) formation, caspase 3 activities, and mitochondrial membrane potential (ΔΨm) disruption. Gene expression and apoptosis-associated protein levels were measured by reverse transcription-quantitative polymerase chain reaction and western blotting. CF induced HepG2 cell death in a time- and dose-dependent manner with half maximal inhibitory concentration values of 219.03±9.96 and 124.90±6.86 µg/ml at 24 and 48 h, respectively. Treatment with CF caused a significant and dose-dependent decrease in colony forming ability and cell migration. Furthermore, the present study demonstrated that CF induced ROS formation, increased caspase 3 activities, decreased the ΔΨm, and caused HepG2 apoptosis. CF marginally decreased the expression level of the cell cycle regulatory protein, ras-related C3 botulinum toxin substrate 1 (rho family, small GTP binding protein Rac1) and the downstream protein, cyclin dependent kinase 6. Additionally, CF significantly enhanced p21 levels, reduced cyclin D1 protein levels and triggered cancer cell death. CF leaf extracts induced cell death, stimulated apoptosis and inhibited migration in HepG2 cells. Thus, CF may be useful for developing an anticancer drug candidate for the treatment of liver cancer.
Collapse
Affiliation(s)
- Benjaporn Buranrat
- Faculty of Medicine, Mahasarakham University, Muang, Maha Sarakham 44000, Thailand
| | - Nootchanat Mairuae
- Faculty of Medicine, Mahasarakham University, Muang, Maha Sarakham 44000, Thailand
| | - Watchara Kanchanarach
- Microbiology and Applied Microbiology Research Unit, Department of Biology, Faculty of Science, Mahasarakham University, Maha Sarakham 44150, Thailand
| |
Collapse
|
28
|
Jeong H, Lee SH, Hwang Y, Yoo H, Jung H, Kim SH, Mok H. Multivalent Aptamer-RNA Conjugates for Simple and Efficient Delivery of Doxorubicin/siRNA into Multidrug-Resistant Cells. Macromol Biosci 2016; 17. [PMID: 27863037 DOI: 10.1002/mabi.201600343] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/07/2016] [Indexed: 01/08/2023]
Abstract
Multivalent aptamer-siRNA conjugates containing multiple mucin-1 aptamers and BCL2-specific siRNA are synthesized, and doxorubicin, an anthracycline anticancer drug, is loaded into these conjugates through intercalation with nucleic acids. These doxorubicin-incorporated multivalent aptamer-siRNA conjugates are transfected to mucin-1 overexpressing MCF-7 breast cancer cells and their multidrug-resistant cell lines. Doxorubicin-incorporated multivalent aptamer-siRNA conjugates exert promising anticancer effects, such as activation of caspase-3/7 and decrease of cell viability, on multidrug-resistant cancer cells because of their high intracellular uptake efficiency. Thus, this delivery system is an efficient tool for combination oncotherapy with chemotherapeutics and nucleic acid drugs to overcome multidrug resistance.
Collapse
Affiliation(s)
- Hyosook Jeong
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Soo Hyeon Lee
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology Zurich (ETHZ), 8093 Zurich, Switzerland
| | - Yeonju Hwang
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Hyundong Yoo
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Heesun Jung
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Sun Hwa Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 136-791, Republic of Korea
| | - Hyejung Mok
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| |
Collapse
|
29
|
Koceva-Chyła A, Matczak K, Hikisz MP, Durka MK, Kochel MK, Süss-Fink G, Furrer J, Kowalski K. Insights into the in vitro Anticancer Effects of Diruthenium-1. ChemMedChem 2016; 11:2171-2187. [DOI: 10.1002/cmdc.201600315] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Indexed: 01/22/2023]
Affiliation(s)
- Aneta Koceva-Chyła
- Department of Medical Biophysics; Faculty of Biology and Environmental Protection; University of Łódź; Pomorska St. 141/143 90236 Łódź Poland
| | - Karolina Matczak
- Department of Medical Biophysics; Faculty of Biology and Environmental Protection; University of Łódź; Pomorska St. 141/143 90236 Łódź Poland
| | - Msc. Paweł Hikisz
- Department of Medical Biophysics; Faculty of Biology and Environmental Protection; University of Łódź; Pomorska St. 141/143 90236 Łódź Poland
| | - Msc. Kamil Durka
- Department of Medical Biophysics; Faculty of Biology and Environmental Protection; University of Łódź; Pomorska St. 141/143 90236 Łódź Poland
| | - Msc. Krzysztof Kochel
- Department of Medical Biophysics; Faculty of Biology and Environmental Protection; University of Łódź; Pomorska St. 141/143 90236 Łódź Poland
| | - Georg Süss-Fink
- Institut de Chimie; Université de Neuchâtel; Avenue de Bellevaux 51 2000 Neuchâtel Switzerland
| | - Julien Furrer
- Department für Chemie und Biochemie; Universität Bern; Freiestrasse 3 3012 Bern Switzerland
| | - Konrad Kowalski
- Department of Organic Chemistry; Faculty of Chemistry; University of Łódź; Tamka St. 12 91403 Łódź Poland
| |
Collapse
|
30
|
Influence of nucleotide excision repair on mitoxantrone cytotoxicity. DNA Repair (Amst) 2016; 42:33-43. [DOI: 10.1016/j.dnarep.2016.04.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 01/08/2016] [Indexed: 01/25/2023]
|
31
|
Cai Y, Wang S, Wu M, Tsosie JK, Xie X, Wan J, He C, Tian H, Chen X, Chen M. PCL–F68–PCL/PLGA–PEG–PLGA mixed micelles mediated delivery of mitoxantrone for reversing multidrug resistant in breast cancer. RSC Adv 2016. [DOI: 10.1039/c5ra27648a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Mitoxantrone-loaded PCL–F68–PCL/PLGA–PEG–PLGA mixed micelles for reversing multidrug resistant in breast cancer.
Collapse
Affiliation(s)
- Yuee Cai
- State Key Laboratory of Quality Research in Chinese Medicine
- Institute of Chinese Medical Sciences
- University of Macau
- China
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine
- Institute of Chinese Medical Sciences
- University of Macau
- China
| | - Minghui Wu
- Department of Cell Biology and Anatomy
- College of Medicine
- University of Florida
- Gainesville
- USA
| | | | - Xi Xie
- School of Chemistry and Chemical Engineering
- Sun Yat-Sen University
- Guangzhou 510275
- China
| | - Jianbo Wan
- State Key Laboratory of Quality Research in Chinese Medicine
- Institute of Chinese Medical Sciences
- University of Macau
- China
| | - Chengwei He
- State Key Laboratory of Quality Research in Chinese Medicine
- Institute of Chinese Medical Sciences
- University of Macau
- China
| | - Huayu Tian
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine
- Institute of Chinese Medical Sciences
- University of Macau
- China
| |
Collapse
|
32
|
Sarkar S, Cohen N, Sabhachandani P, Konry T. Phenotypic drug profiling in droplet microfluidics for better targeting of drug-resistant tumors. LAB ON A CHIP 2015; 15:4441-50. [PMID: 26456240 PMCID: PMC4666301 DOI: 10.1039/c5lc00923e] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Acquired drug resistance is a key factor in the failure of chemotherapy. Due to intratumoral heterogeneity, cancer cells depict variations in intracellular drug uptake and efflux at the single cell level, which may not be detectable in bulk assays. In this study we present a droplet microfluidics-based approach to assess the dynamics of drug uptake, efflux and cytotoxicity in drug-sensitive and drug-resistant breast cancer cells. An integrated droplet generation and docking microarray was utilized to encapsulate single cells as well as homotypic cell aggregates. Drug-sensitive cells showed greater death in the presence or absence of Doxorubicin (Dox) compared to the drug-resistant cells. We observed heterogeneous Dox uptake in individual drug-sensitive cells while the drug-resistant cells showed uniformly low uptake and retention. Dox-resistant cells were classified into distinct subsets based on their efflux properties. Cells that showed longer retention of extracellular reagents also demonstrated maximal death. We further observed homotypic fusion of both cell types in droplets, which resulted in increased cell survival in the presence of high doses of Dox. Our results establish the applicability of this microfluidic platform for quantitative drug screening in single cells and multicellular interactions.
Collapse
Affiliation(s)
- S Sarkar
- Department of Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, Boston, 02115 MA, USA.
| | - N Cohen
- Department of Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, Boston, 02115 MA, USA.
| | - P Sabhachandani
- Department of Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, Boston, 02115 MA, USA.
| | - T Konry
- Department of Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, Boston, 02115 MA, USA.
| |
Collapse
|
33
|
Szwed M, Wrona D, Kania KD, Koceva-Chyla A, Marczak A. Doxorubicin-transferrin conjugate triggers pro-oxidative disorders in solid tumor cells. Toxicol In Vitro 2015; 31:60-71. [PMID: 26607004 DOI: 10.1016/j.tiv.2015.11.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 11/03/2015] [Accepted: 11/16/2015] [Indexed: 12/26/2022]
Abstract
The formation of reactive oxygen species (ROS) is a widely accepted mechanism of doxorubicin (DOX) toxicity toward cancer cells. However, little is known about the potential of new systems, designed for more efficient and targeted doxorubicin delivery (i.e. protein conjugates, polymeric micelles, liposomes, monoclonal antibodies), to induce oxidative stress (OS) in tumors and hematological malignancies. Therefore, the objective of our study was to determine the relation between the toxicity of doxorubicin-transferring (DOX-TRF) conjugate and its capability to generate oxidative/nitrosative stress in solid tumor cells. Our research proves that DOX-TRF conjugate displays higher cytotoxicity towards lung adenocarcinoma epithelial (A549) and hepatocellular carcinoma (HepG2) cell lines than the reference free drug (DOX) and induces more extensive OS, characterized by a significant decrease in the total cellular antioxidant capacity, glutathione level and amount of -SH groups and an increase in hydroperoxide content. The intracellular redox imbalance was accompanied by changes in the transcription of genes encoding key antioxidant enzymes engaged in the sustaining of cellular redox homeostasis: superoxide dismutase (SOD), catalase (CAT), glutathione transferase (GST) and glutathione peroxidase (GP).
Collapse
Affiliation(s)
- Marzena Szwed
- Department of Thermobiology, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St. 141/143, 90-236 Lodz, Poland.
| | - Dominika Wrona
- Department of Thermobiology, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St. 141/143, 90-236 Lodz, Poland.
| | - Katarzyna D Kania
- Laboratory of Transcriptional Regulation, Institute of Medical Biology, PAS, Lodowa St. 106, 93-232 Lodz, Poland.
| | - Aneta Koceva-Chyla
- Department of Thermobiology, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St. 141/143, 90-236 Lodz, Poland.
| | - Agnieszka Marczak
- Department of Thermobiology, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St. 141/143, 90-236 Lodz, Poland.
| |
Collapse
|
34
|
Hikisz P, Szczupak Ł, Koceva-Chyła A, Guśpiel A, Oehninger L, Ott I, Therrien B, Solecka J, Kowalski K. Anticancer and Antibacterial Activity Studies of Gold(I)-Alkynyl Chromones. Molecules 2015; 20:19699-718. [PMID: 26528965 PMCID: PMC6331995 DOI: 10.3390/molecules201119647] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/18/2015] [Accepted: 10/23/2015] [Indexed: 12/11/2022] Open
Abstract
Three gold(I) complexes of alkynyl chromones were synthesized and characterized. The single-crystal X-ray structure analysis of a dinuclear compound and of a flavone derivative exhibit a typical d10 gold(I)-alkynyl linear arrangement. All complexes were evaluated as anticancer and antibacterial agents against four human cancer cell lines and four pathogenic bacterial strains. All compounds show antiproliferative activity at lower micromolar range concentrations. Complex 4 showed a broad activity profile, being more active than the reference drug auranofin against HepG2, MCF-7 and CCRF-CEM cancer cells. The cellular uptake into MCF-7 cells of the investigated complexes was measured by atomic absorption spectroscopy (AAS). These measurements showed a positive correlation between an increased cellular gold content and the incubation time of the complexes. Unexpectedly an opposite effect was observed for the most active compound. Biological assays revealed various molecular mechanisms for these compounds, comprising: (i) thioredoxin reductase (TrxR) inhibition, (ii) caspases-9 and -3 activation; (iii) DNA damaging activity and (iv) cell cycle disturbance. The gold(I) complexes were also bactericidal against Gram-positive methicillin-sensitive Staphylococcus aureus (MSSA) and methicillin-resistant S. aureus (MRSA) bacterial strains, while showing no activity against the Gram-negative Escherichia coli bacterial strain.
Collapse
Affiliation(s)
- Paweł Hikisz
- Department of Thermobiology, Faculty of Biology and Environmental Protection, University of Łódź, Pomorska 141/143, Łódź PL-90236, Poland; (P.H.); (A.K.-C.)
| | - Łukasz Szczupak
- Department of Organic Chemistry, Faculty of Chemistry, University of Łódź, Tamka 12, Łódź PL-91403, Poland;
| | - Aneta Koceva-Chyła
- Department of Thermobiology, Faculty of Biology and Environmental Protection, University of Łódź, Pomorska 141/143, Łódź PL-90236, Poland; (P.H.); (A.K.-C.)
| | - Adam Guśpiel
- Laboratory of Biologically Active Compounds, National Institute of Public Health-National Institute of Hygiene, Chocimska 24, Warsaw PL-00791, Poland; (A.G.); (J.S.)
| | - Luciano Oehninger
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstr. 55, Braunschweig D-38106, Germany; l.oehninger@tu-braunschweig (L.O.); (I.O.)
| | - Ingo Ott
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstr. 55, Braunschweig D-38106, Germany; l.oehninger@tu-braunschweig (L.O.); (I.O.)
| | - Bruno Therrien
- Institute of Chemistry, Faculty of Science, University of Neuchatel, Avenue de Bellevaux 51, Neuchatel CH-2000, Switzerland;
| | - Jolanta Solecka
- Laboratory of Biologically Active Compounds, National Institute of Public Health-National Institute of Hygiene, Chocimska 24, Warsaw PL-00791, Poland; (A.G.); (J.S.)
| | - Konrad Kowalski
- Department of Organic Chemistry, Faculty of Chemistry, University of Łódź, Tamka 12, Łódź PL-91403, Poland;
- Correspondence: ; Tel.: +48-42-635-5759 (ext. 123); Fax: +48-42-665-5258
| |
Collapse
|
35
|
Szwed M, Kania KD, Jozwiak Z. Assessment of pro-apoptotic activity of doxorubicin-transferrin conjugate in cells derived from human solid tumors. Int J Biochem Cell Biol 2015; 70:57-67. [PMID: 26520467 DOI: 10.1016/j.biocel.2015.10.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 09/11/2015] [Accepted: 10/18/2015] [Indexed: 02/09/2023]
Abstract
Conjugates of anthracyclines are a new possibility for anticancer agent delivery, which seems to be a very promising alternative to the currently used cancer treatment strategies. In our study, we investigated the ability of a doxorubicin-transferrin (DOX-TRF) conjugate to induce cell death in two solid tumor cell lines: non-small cell lung cancer (A549) and hepatocellular liver carcinoma (HepG2). The observed effects of the DOX-TRF conjugate on these cell cultures were compared with those of free doxorubicin (DOX), a widely used antineoplastic therapeutic agent. Our results provided direct evidence that the investigated conjugate is considerably more cytotoxic to the examined human cancer cell lines than is DOX alone. Moreover, we confirmed that the antitumor efficacy of DOX-TRF conjugate is related to its apoptosis-inducing ability, which was shown during measurements of typical features of programmed cell death. In solid tumor cell lines, the DOX-TRF conjugate induced changes in cellular morphology, mitochondrial membrane potential and caspases-3 and -9 activities. Furthermore, all of the analyzed hallmarks of apoptosis were confirmed by the oligonucleosomal DNA fragmentation assay and by a real-time PCR quantitative study, which displayed the superiority of the conjugate-induced programmed cell death over free drug-triggered cell death.
Collapse
Affiliation(s)
- Marzena Szwed
- Department of Thermobiology, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143 Street, 90-236 Lodz, Poland.
| | - Katarzyna Dominika Kania
- Laboratory of Transcriptional Regulation, Institute for Medical Biology, PAS, Lodowa 106 Street, 93-232 Lodz, Poland.
| | - Zofia Jozwiak
- Department of Thermobiology, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143 Street, 90-236 Lodz, Poland.
| |
Collapse
|
36
|
Mohammad N, Singh SV, Malvi P, Chaube B, Athavale D, Vanuopadath M, Nair SS, Nair B, Bhat MK. Strategy to enhance efficacy of doxorubicin in solid tumor cells by methyl-β-cyclodextrin: Involvement of p53 and Fas receptor ligand complex. Sci Rep 2015; 5:11853. [PMID: 26149967 PMCID: PMC4493576 DOI: 10.1038/srep11853] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/20/2015] [Indexed: 12/25/2022] Open
Abstract
Doxorubicin (DOX) is one of the preferred drugs for treating breast and liver cancers. However, its clinical application is limited due to severe side effects and the accompanying drug resistance. In this context, we investigated the effect on therapeutic efficacy of DOX by cholesterol depleting agent methyl-β-cyclodextrin (MCD), and explored the involvement of p53. MCD sensitizes MCF-7 and Hepa1–6 cells to DOX, Combination of MCD and marginal dose of DOX reduces the cell viability, and promoted apoptosis through induction of pro-apoptotic protein, Bax, activation of caspase-8 and caspase-7, down regulation of anti-apoptotic protein Bcl-2 and finally promoting PARP cleavage. Mechanistically, sensitization to DOX by MCD was due to the induction of FasR/FasL pathway through p53 activation. Furthermore, inhibition of p53 by pharmacological inhibitor pifithrin-α (PFT-α) or its specific siRNA attenuated p53 function and down-regulated FasR/FasL, thereby preventing cell death. Animal experiments were performed using C57BL/6J mouse isografted with Hepa1–6 cells. Tumor growth was retarded and survival increased in mice administered MCD together with DOX to as compared to either agent alone. Collectively, these results suggest that MCD enhances the sensitivity to DOX for which wild type p53 is an important determinant.
Collapse
Affiliation(s)
- Naoshad Mohammad
- National Centre for Cell Science, Pune University Campus, Ganeshkhind, Pune- 411007, India
| | - Shivendra Vikram Singh
- National Centre for Cell Science, Pune University Campus, Ganeshkhind, Pune- 411007, India
| | - Parmanand Malvi
- National Centre for Cell Science, Pune University Campus, Ganeshkhind, Pune- 411007, India
| | - Balkrishna Chaube
- National Centre for Cell Science, Pune University Campus, Ganeshkhind, Pune- 411007, India
| | - Dipti Athavale
- National Centre for Cell Science, Pune University Campus, Ganeshkhind, Pune- 411007, India
| | | | | | - Bipin Nair
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham University, Kollam-690525, India
| | - Manoj Kumar Bhat
- National Centre for Cell Science, Pune University Campus, Ganeshkhind, Pune- 411007, India
| |
Collapse
|
37
|
MicroRNA-208a Silencing Attenuates Doxorubicin Induced Myocyte Apoptosis and Cardiac Dysfunction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:597032. [PMID: 26137188 PMCID: PMC4475537 DOI: 10.1155/2015/597032] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 05/31/2015] [Indexed: 12/17/2022]
Abstract
AIMS GATA4 depletion is a distinct mechanism by which doxorubicin leads to cardiomyocyte apoptosis, and preservation of GATA4 mitigates doxorubicin induced myocyte apoptosis and cardiac dysfunction. We investigated a novel approach of attenuating doxorubicin induced cardiac toxicity by silencing miR-208a, a heart specific microRNA known to target GATA4. METHODS AND RESULTS Eight-week-old female Balb/C mice were randomly assigned to sham, antagomir, and control groups. Antagomir group were pretreated with miR-208a antagomir 4 days before doxorubicin administration. At day 0, control and antagomir groups received 20 mg/kg of doxorubicin, while sham mice received phosphate buffered solution. Echocardiography was done at day 7, after which animals were sacrificed and hearts harvested and assessed for apoptosis and expression of miR-208a, GATA4, and BCL-2. Doxorubicin significantly upregulated miR-208a, downregulated GATA4, and increased myocyte apoptosis, with resulting decrease in cardiac function. In contrast, therapeutic silencing of miR-208a salvaged GATA4 and BCL-2 and decreased apoptosis, with improvement in cardiac function. CONCLUSION Doxorubicin upregulates miR-208a and promotes cardiomyocyte apoptosis, while therapeutic silencing of miR-208a attenuates doxorubicin induced myocyte apoptosis with subsequent improvement in cardiac function. These novel results highlight the therapeutic potential of targeting miR-208a to prevent doxorubicin cardiotoxicity.
Collapse
|
38
|
Self-assembled betulinic acid protects doxorubicin induced apoptosis followed by reduction of ROS–TNF-α–caspase-3 activity. Biomed Pharmacother 2015; 72:144-57. [DOI: 10.1016/j.biopha.2015.04.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 04/04/2015] [Accepted: 04/15/2015] [Indexed: 01/11/2023] Open
|
39
|
Baviskar AT, Amrutkar SM, Trivedi N, Chaudhary V, Nayak A, Guchhait SK, Banerjee UC, Bharatam PV, Kundu CN. Switch in Site of Inhibition: A Strategy for Structure-Based Discovery of Human Topoisomerase IIα Catalytic Inhibitors. ACS Med Chem Lett 2015; 6:481-5. [PMID: 25941559 DOI: 10.1021/acsmedchemlett.5b00040] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 02/23/2015] [Indexed: 11/29/2022] Open
Abstract
A study of structure-based modulation of known ligands of hTopoIIα, an important enzyme involved in DNA processes, coupled with synthesis and in vitro assays led to the establishment of a strategy of rational switch in mode of inhibition of the enzyme's catalytic cycle. 6-Arylated derivatives of known imidazopyridine ligands were found to be selective inhibitors of hTopoIIα, while not showing TopoI inhibition and DNA binding. Interestingly, while the parent imidazopyridines acted as ATP-competitive inhibitors, arylated derivatives inhibited DNA cleavage similar to merbarone, indicating a switch in mode of inhibition from ATP-hydrolysis to the DNA-cleavage stage of catalytic cycle of the enzyme. The 6-aryl-imidazopyridines were relatively more cytotoxic than etoposide in cancer cells and less toxic to normal cells. Such unprecedented strategy will encourage research on "choice-based change" in target-specific mode of action for rapid drug discovery.
Collapse
Affiliation(s)
| | | | | | | | - Anmada Nayak
- School
of Biotechnology, KIIT University, Campus-11, Patia, Bhubaneswar, Orissa 751024, India
| | | | | | | | - Chanakya N. Kundu
- School
of Biotechnology, KIIT University, Campus-11, Patia, Bhubaneswar, Orissa 751024, India
| |
Collapse
|
40
|
Kucinska M, Skupin-Mrugalska P, Szczolko W, Sobotta L, Sciepura M, Tykarska E, Wierzchowski M, Teubert A, Fedoruk-Wyszomirska A, Wyszko E, Gdaniec M, Kaczmarek M, Goslinski T, Mielcarek J, Murias M. Phthalocyanine derivatives possessing 2-(morpholin-4-yl)ethoxy groups as potential agents for photodynamic therapy. J Med Chem 2015; 58:2240-55. [PMID: 25700089 DOI: 10.1021/acs.jmedchem.5b00052] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Three 2-(morpholin-4-yl)ethoxy substituted phthalocyanines were synthesized and characterized. Phthalocyanine derivatives revealed moderate to high quantum yields of singlet oxygen production depending on the solvent applied (e.g., in DMF ranging from 0.25 to 0.53). Their photosensitizing potential for photodynamic therapy was investigated in an in vitro model using cancer cell lines. Biological test results were found particularly encouraging for the zinc(II) phthalocyanine derivative possessing two 2-(morpholin-4-yl)ethoxy substituents in nonperipheral positions. Cells irradiated for 20 min at 2 mW/cm(2) revealed the lowest IC50 value at 0.25 μM for prostate cell line (PC3), whereas 1.47 μM was observed for human malignant melanoma (A375) cells. The cytotoxic activity in nonirradiated cells of novel phthalocyanine was found to be very low. Moreover, the cellular uptake, localization, cell cycle, apoptosis through an ELISA assay, and immunochemistry method were investigated in LNCaP cells. Our results showed that the tested photosensitizer possesses very interesting biological activity, depending on experimental conditions.
Collapse
Affiliation(s)
- Malgorzata Kucinska
- Department of Toxicology, Poznan University of Medical Sciences , Dojazd 30, 60-631 Poznan, Poland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Truong WT, Su Y, Gloria D, Braet F, Thordarson P. Dissolution and degradation of Fmoc-diphenylalanine self-assembled gels results in necrosis at high concentrations in vitro. Biomater Sci 2014. [PMID: 26218120 DOI: 10.1039/c4bm00244j] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Herein we report an approach to assess in vitro cellular responses to the dissolution or degradation products from Fmoc-diphenylalanine (Fmoc-FF) self-assembled hydrogels. Three cell lines were used in these studies and two-way ANOVA was used to assess (i) the age of gel dissolution and degradation products and (ii) exposure time on cell fate and state, using viability assays in conjunction with time-lapse fluorescence and high-resolution scanning electron microscopy investigation. The studies show that leaching time but not the exposure time affects the overall cell viability. The cytotoxic effect was only observed once the gel is completely dissolved. Further analysis revealed that the principal mechanism of cell death is necrosis. In addition, the effect of chemotherapeutics (5-fluorouracil and paclitaxel) released from the Fmoc-FF gel (with addition before and after gelation) on colorectal cancer cells were investigated using this methodology, demonstrating enhanced activity of these drugs compared to bulk control. This enhanced activity, however, appears to be a combination of the apoptosis caused by the cancer drugs and necrosis caused by gel dissolution and degradation products. Given that in vivo studies by others on Fmoc-peptides that this material is not harmful to animals, our work highlights that conventional in vitro cellular assays may yield conflicting messages when used for the evaluation of cytotoxicity and drug release from self-assembled gels such as Fmoc-FF and that better in vitro models, (e.g. 3D cell culture systems) need to be developed to evaluate these materials for biomedical applications.
Collapse
Affiliation(s)
- Warren Ty Truong
- School of Chemistry, the Australian Centre for Nanomedicine and the ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of New South Wales, Sydney, NSW 2052, Australia.
| | | | | | | | | |
Collapse
|
42
|
Kowalski K, Hikisz P, Szczupak Ł, Therrien B, Koceva-Chyła A. Ferrocenyl and dicobalt hexacarbonyl chromones – New organometallics inducing oxidative stress and arresting human cancer cells in G2/M phase. Eur J Med Chem 2014; 81:289-300. [DOI: 10.1016/j.ejmech.2014.05.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 04/09/2014] [Accepted: 05/06/2014] [Indexed: 10/25/2022]
|
43
|
Cratoxylum formosum (Jack) Dyer ssp. pruniflorum (Kurz) Gogel. (Hóng yá mù) extract induces apoptosis in human hepatocellular carcinoma HepG2 cells through caspase-dependent pathways. Chin Med 2014; 9:12. [PMID: 24708784 PMCID: PMC3985586 DOI: 10.1186/1749-8546-9-12] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Accepted: 04/04/2014] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Cratoxylum formosum (Jack) Dyer ssp. pruniflorum (Kurz) Gogel. (Hóng yá mù) (CF) has been used for treatment of fever, cough, and peptic ulcer. Previously, a 50% ethanol-water extract from twigs of CF was shown highly selective in cytotoxicity against cancer cells. This study aims to investigate the molecular mechanisms underlying the apoptosis-inducing effect of CF. METHODS The cytotoxicity of CF was evaluated in the human hepatocellular carcinoma (HCC) HepG2 cell line in comparison with a non-cancerous African green monkey kidney epithelial cell line (Vero) by a neutral red assay. The apoptosis induction mechanisms were investigated through nuclear morphological changes, DNA fragmentation, mitochondrial membrane potential alterations, and caspase enzyme activities. RESULTS CF selectively induced HepG2 cell death compared with non-cancerous Vero cells. A 1.5-fold higher apoptotic effect compared with melphalan was induced by 120 μg/mL of the 50% ethanol-water extract of CF. The apoptotic cell death in HepG2 cells occurred via extrinsic and intrinsic caspase-dependent pathways in dose- and time-dependent manners by significantly increasing the activities of caspase 3/7, 8, and 9, decreasing the mitochondrial membrane potential, and causing apoptotic body formation and DNA fragmentation. CONCLUSIONS CF extract induced a caspase-dependent apoptosis in HepG2 cells.
Collapse
|
44
|
Marczak A, Bukowska B, Rogalska A. WP 631 and Epo B synergize in SKOV-3 human ovarian cancer cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2014; 37:256-266. [PMID: 24374386 DOI: 10.1016/j.etap.2013.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Revised: 11/27/2013] [Accepted: 12/02/2013] [Indexed: 06/03/2023]
Abstract
Combined therapy is one of the basic methods of treatment different types of cancer. It allows to reduce the side effects of each component while maximizing the therapeutic action. The aim of this study was to evaluate the impact of two new drugs: WP 631 (bisanthracycline) and epothilone B (Epo B), added in combination on the SKOV-3 human ovarian cancer cells. To assess the type of interaction between WP 631 and Epo B isobolografic analysis was applied based on the cytotoxicity of drugs determined by the MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolinum bromide) assay. Apoptotic and necrotic cell levels were measured by double staining with Hoechst 33258 and propidium iodide, Annexin V-FITC staining and by using TUNEL assay. The combination of WP 631 and Epo B is more potent than drugs added alone. The quantitative analysis indicated that the major mode of cell death induced by the combination after 72 h treatment was early apoptosis, whereas drugs administered alone generated less intensive apoptosis. The present report demonstrates for the first time that WP 631 and Epo B co-administered synergize in SKOV-3 cell line (Z(ex)/Z(th)<1).
Collapse
Affiliation(s)
- Agnieszka Marczak
- Department of Thermobiology, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland.
| | - Barbara Bukowska
- Department of Thermobiology, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| | - Aneta Rogalska
- Department of Thermobiology, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| |
Collapse
|
45
|
Ahmad K, Shah AH, Adhikari B, Rana UA, uddin SN, Vijayaratnam C, Muhammad N, Shujah S, Rauf A, Hussain H, Badshah A, Qureshi R, Kraatz HB, Shah A. pH-dependent redox mechanism and evaluation of kinetic and thermodynamic parameters of a novel anthraquinone. RSC Adv 2014. [DOI: 10.1039/c4ra04462b] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The pH-dependent oxidation of a novel anthraquinone was investigated and several important kinetic and thermodynamic parameters were determined.
Collapse
Affiliation(s)
- Khurshid Ahmad
- Department of Chemistry
- Quaid-i-Azam University
- Islamabad, Pakistan
| | | | - Bimalendu Adhikari
- Department of Physical and Environmental Sciences
- University of Toronto Scarborough
- Toronto, Canada
| | - Usman Ali Rana
- Deanship of Scientific Research
- College of Engineering
- King Saud University
- Riyadh 11421, Saudi Arabia
| | - Syed Noman uddin
- Department of Chemistry
- Quaid-i-Azam University
- Islamabad, Pakistan
| | - Chandrika Vijayaratnam
- Department of Physical and Environmental Sciences
- University of Toronto Scarborough
- Toronto, Canada
| | - Niaz Muhammad
- Department of Chemistry
- Abdul Wali Khan University Mardan
- Pakistan
| | - Shaukat Shujah
- Department of Chemistry
- Kohat University of Science & Technology
- Kohat, Pakistan
| | - Abdur Rauf
- Department of Chemistry
- Quaid-i-Azam University
- Islamabad, Pakistan
| | - Hidayat Hussain
- UoN Chair of Oman’s Medicinal Plants and Marine Natural Products
- University of Nizwa
- Nizwa 616, Sultanate of Oman
| | - Amin Badshah
- Department of Chemistry
- Quaid-i-Azam University
- Islamabad, Pakistan
| | - Rumana Qureshi
- Department of Chemistry
- Quaid-i-Azam University
- Islamabad, Pakistan
| | - Heinz-Bernhard Kraatz
- Department of Physical and Environmental Sciences
- University of Toronto Scarborough
- Toronto, Canada
| | - Afzal Shah
- Department of Chemistry
- Quaid-i-Azam University
- Islamabad, Pakistan
- Department of Physical and Environmental Sciences
- University of Toronto Scarborough
| |
Collapse
|
46
|
Kostrzewa-Nowak D, Tarasiuk J. Bioreductive activation of mitoxantrone by NADPH cytochrome P450 reductase does not change its apoptotic stimuli properties in regard to sensitive and multidrug resistant leukaemia HL60 cells. Eur J Pharmacol 2013; 721:141-50. [DOI: 10.1016/j.ejphar.2013.09.041] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 09/10/2013] [Accepted: 09/19/2013] [Indexed: 11/25/2022]
|
47
|
Kontek R, Jakubczak M, Matlawska-Wasowska K. The antioxidants, vitamin A and E but not vitamin C and melatonin enhance the proapoptotic effects of irinotecan in cancer cells in vitro. Toxicol In Vitro 2013; 28:282-91. [PMID: 24287113 DOI: 10.1016/j.tiv.2013.11.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 11/13/2013] [Accepted: 11/15/2013] [Indexed: 12/17/2022]
Abstract
Irinotecan is one of the camptothecin analog which has been shown to have a broad spectrum of antitumor activities against various malignancies. The aim of this study was to evaluate the effect of vitamin A, C, E and melatonin on proapoptotic activity of irinotecan in human cancer cells in vitro. We observed that irinotecan induced apoptosis in all types of analyzed cell lines when used as a single agent. Combination of selected antioxidants with various doses of irinotecan (7.5-60μM) resulted in significant increase in apoptotic cell death in A549 and HT29 cancer cell lines. The highest killing efficiency was observed after co-incubation of the cells with irinotecan and vitamin A (10μM), or vitamin E (25μM), respectively. The addition of vitamin C and melatonin to irinotecan treatment did not promote increase in killing of cancer cells. Our results indicate that some antioxidants can enhance the proapoptoic activity (properties) of irinotecan in human cancer cells in vitro. These findings may be supportive for the optimization of therapeutic efficacy of irinotecan treatment.
Collapse
Affiliation(s)
- Renata Kontek
- University of Lodz, Department of General Genetics, Molecular Biology and Biotechnology, Laboratory of Cytogenetics, Lodz, Poland.
| | - Marta Jakubczak
- University of Lodz, Department of General Genetics, Molecular Biology and Biotechnology, Laboratory of Cytogenetics, Lodz, Poland
| | - Ksenia Matlawska-Wasowska
- University of New Mexico Health Sciences Center, Department of Pediatrics, Division of Hematology and Oncology, Albuquerque, NM, USA
| |
Collapse
|
48
|
Kim TH, Shin YJ, Won AJ, Lee BM, Choi WS, Jung JH, Chung HY, Kim HS. Resveratrol enhances chemosensitivity of doxorubicin in multidrug-resistant human breast cancer cells via increased cellular influx of doxorubicin. Biochim Biophys Acta Gen Subj 2013; 1840:615-25. [PMID: 24161697 DOI: 10.1016/j.bbagen.2013.10.023] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 10/13/2013] [Accepted: 10/15/2013] [Indexed: 12/21/2022]
Abstract
BACKGROUND Multidrug resistance is a major problem in the treatment of breast cancer, and a number of studies have attempted to find an efficient strategy with which to overcome it. In this study, we investigate the synergistic anticancer effects of resveratrol (RSV) and doxorubicin (Dox) against human breast cancer cell lines. METHODS The synergistic effects of RSV on chemosensitivity were examined in Dox-resistant breast cancer (MCF-7/adr) and MDA-MB-231 cells. In vivo experiments were performed using a nude mouse xenograft model to investigate the combined sensitization effect of RSV and Dox. RESULTS AND CONCLUSION RSV markedly enhanced Dox-induced cytotoxicity in MCF-7/adr and MDA-MB-231 cells. Treatment with a combination of RSV and Dox significantly increased the cellular accumulation of Dox by down-regulating the expression levels of ATP-binding cassette (ABC) transporter genes, MDR1, and MRP1. Further in vivo experiments in the xenograft model revealed that treatment with a combination of RSV and Dox significantly inhibited tumor volume by 60%, relative to the control group. GENERAL SIGNIFICANCE These results suggest that treatment with a combination of RSV and Dox would be a helpful strategy for increasing the efficacy of Dox by promoting an intracellular accumulation of Dox and decreasing multi-drug resistance in human breast cancer cells.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Apoptosis/drug effects
- Blotting, Western
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Proliferation/drug effects
- Doxorubicin/administration & dosage
- Drug Resistance, Multiple/drug effects
- Drug Resistance, Neoplasm/drug effects
- Drug Synergism
- Female
- Humans
- Mice
- Mice, Inbred BALB C
- Multidrug Resistance-Associated Proteins/genetics
- Multidrug Resistance-Associated Proteins/metabolism
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Resveratrol
- Reverse Transcriptase Polymerase Chain Reaction
- Stilbenes/administration & dosage
- Tissue Distribution
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Tae Hyung Kim
- Laboratory of Molecular Toxicology, College of Pharmacy, Pusan National University, San 30, Jangjeon-dong, Geumjeung-gu, Busan 609-735, Republic of Korea; Division of Toxicology, College of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 440-746, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
49
|
|
50
|
In vitro and in vivo evaluation of paclitaxel-loaded mesoporous silica nanoparticles with three pore sizes. Int J Pharm 2013; 445:12-9. [DOI: 10.1016/j.ijpharm.2013.01.058] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 12/19/2012] [Accepted: 01/24/2013] [Indexed: 11/17/2022]
|