1
|
Alaouna M, Molefi T, Khanyile R, Chauke-Malinga N, Chatziioannou A, Luvhengo TE, Raletsena M, Penny C, Hull R, Dlamini Z. The potential of the South African plant Tulbaghia Violacea Harv for the treatment of triple negative breast cancer. Sci Rep 2025; 15:5737. [PMID: 39962120 PMCID: PMC11832780 DOI: 10.1038/s41598-025-88417-2] [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: 07/03/2024] [Accepted: 01/28/2025] [Indexed: 02/20/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is difficult to treat and has a low five-year survival rate. In South Africa, a large percentage of the population still relies on traditional plant-based medicine. To establish the utility of both methanol and water-soluble extracts from the leaves of Tulbaghia violacea, cytotoxicity assays were carried out to establish the IC50 values against a TNBC cell line. Cell cycle and apoptosis assays were carried out using the extracts. To identify the molecular compounds, present in water-soluble leaf extracts, NMR spectroscopy was performed. Compounds of interest were then used in computational docking studies with the anti-apoptotic protein COX-2. The IC50 values for the water- and methanol-soluble extracts were determined to be 400 and 820 µg/mL, respectively. The water-soluble extract induced apoptosis in the TNBC cell line to a greater extent than in the normal cell line. RNAseq indicated that there was an increase in the transcription of pro-apoptotic genes in the TNBC cell line. The crude extract also caused these cells to stall in the S phase. Of the 61 compounds identified in this extract, five demonstrated a high binding affinity for COX-2. Based on these findings, the compounds within the extract show significant potential for further investigation as candidates for the development of cancer therapeutics, particularly for TNBC.
Collapse
Affiliation(s)
- Mohammed Alaouna
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Thulo Molefi
- Department of Medical Oncology, Steve Biko Academic Hospital, University of Pretoria, Pretoria, 0001, South Africa
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- SA-MRC Precision Oncology Research Unit (PORU), DSTI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria Hatfield, Pretoria, 0028, South Africa
| | - Richard Khanyile
- Department of Medical Oncology, Steve Biko Academic Hospital, University of Pretoria, Pretoria, 0001, South Africa
- SA-MRC Precision Oncology Research Unit (PORU), DSTI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria Hatfield, Pretoria, 0028, South Africa
| | - Nkhensani Chauke-Malinga
- Papillon Aesthetics, Suite 302b Netcare Linksfield Hospital, 24 12th Ave, Linksfield West, Johannesburg, 2192, South Africa
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- SA-MRC Precision Oncology Research Unit (PORU), DSTI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria Hatfield, Pretoria, 0028, South Africa
| | - Aristotelis Chatziioannou
- Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens, Athens, 11527, Greece
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- SA-MRC Precision Oncology Research Unit (PORU), DSTI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria Hatfield, Pretoria, 0028, South Africa
| | - Thifhelimbilu Emmanuel Luvhengo
- Department of Surgery, Charlotte Maxeke Johannesburg Academic Hospital, University of the Witwatersrand, Parktown, Johannesburg, 2193, South Africa
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Maropeng Raletsena
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Chemistry, University of South Africa, Florida Campus, Johannesburg, South Africa
| | - Clement Penny
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Rodney Hull
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
- SA-MRC Precision Oncology Research Unit (PORU), DSTI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria Hatfield, Pretoria, 0028, South Africa.
| | - Zodwa Dlamini
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
- SA-MRC Precision Oncology Research Unit (PORU), DSTI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria Hatfield, Pretoria, 0028, South Africa.
| |
Collapse
|
2
|
Pricop A, Negrea A, Pascu B, Nemeş NS, Ciopec M, Negrea P, Ianăşi C, Svera P, Muntean D, Ivan A, Cristea IM. Copper Nanoparticles Synthesized by Chemical Reduction with Medical Applications. Int J Mol Sci 2025; 26:1628. [PMID: 40004090 PMCID: PMC11855514 DOI: 10.3390/ijms26041628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Copper nanoparticles (CuNPs) have attracted attention due to their low cost and high specific surface area. In this work, a simple and inexpensive two-step synthesis method was proposed to prepare highly stable and well-dispersed spherical CuNPs in solution with a particle size of approximately 37 nm. Synthesis of CuNPs was carried on in the presence of complexing agent trisodium citrate (TSC), while for the chemical reduction step, sodium borohydride (NaBH4) was used. Taking into account the potential of this type of nanoparticles, their synthesis and characterization represent a current and relevant topic in the field. The ability to control the size, shape and properties of CuNPs by adjusting the synthesis parameters (pH, precursor:stabilizer:reductant ratio, homogenization time, temperature) offers extraordinary flexibility in the development of these materials. The combination of characterization techniques such as SEM, EDX, UV-Vis, Raman, FT-IR and AFM provides a thorough understanding of the structure and properties of CuNPs, allowing the modulation of the properties of the obtained nanoparticles in the desired direction. Based on the studies, the copper reduction mechanism was proposed. For the theoretical verification of the size of the experimentally obtained spherical CuNPs, Mie theory was applied. A stability study of the synthesized CuNPs in optimal conditions was performed using UV-Vis analysis at specific time intervals (1, 3, 30 and 60 days), the sample being kept in the dark, inside a drawer at 25 °C. The CuNPs obtained after setting the optimal synthesis parameters (Cu(II):TSC:BH4+ = 1:1:0.2, pH = 5, homogenization time 60 min and temperature 25 °C) were then tested to highlight their antibacterial effect on some reference bacterial strains. The obtained CuNPs demonstrated very good antimicrobial efficacy compared to traditional antimicrobials, for both Gram-negative and Gram-positive bacteria. This may reduce the development of antimicrobial resistance, an urgent medical issue. After evaluating the cytotoxic effects of CuNPs on the SKBR3 cancer cell line, a significant decrease in cell proliferation was observed at the 0.5 mg/mL concentration, with a reduction of 89% after 60 h of cultivation. Higher concentrations of CuNPs induced a more rapid cytotoxic effect, leading to an accelerated decline in cell viability.
Collapse
Affiliation(s)
- Alexandra Pricop
- Faculty of Chemical Engineering, Biotechnologies and Environmental Protection, Politehnica University Timişoara, Victoriei Square, no. 2, 300006 Timişoara, Romania; (A.P.); (A.N.); (M.C.); (P.N.)
| | - Adina Negrea
- Faculty of Chemical Engineering, Biotechnologies and Environmental Protection, Politehnica University Timişoara, Victoriei Square, no. 2, 300006 Timişoara, Romania; (A.P.); (A.N.); (M.C.); (P.N.)
| | - Bogdan Pascu
- Research Institute for Renewable Energies—ICER, Politehnica University Timişoara, Gavril Musicescu Street, no. 138, 300774 Timisoara, Romania
| | - Nicoleta Sorina Nemeş
- Research Institute for Renewable Energies—ICER, Politehnica University Timişoara, Gavril Musicescu Street, no. 138, 300774 Timisoara, Romania
| | - Mihaela Ciopec
- Faculty of Chemical Engineering, Biotechnologies and Environmental Protection, Politehnica University Timişoara, Victoriei Square, no. 2, 300006 Timişoara, Romania; (A.P.); (A.N.); (M.C.); (P.N.)
| | - Petru Negrea
- Faculty of Chemical Engineering, Biotechnologies and Environmental Protection, Politehnica University Timişoara, Victoriei Square, no. 2, 300006 Timişoara, Romania; (A.P.); (A.N.); (M.C.); (P.N.)
| | - Cătălin Ianăşi
- Coriolan Drăgulescu’ Institute of Chemistry, Bv. Mihai Viteazul, No. 24, 300223 Timisoara, Romania;
| | - Paula Svera
- INCEMC—National Institute for Research and Development in Electrochemistry and Condensed Matter-Timisoara, No. 144 Dr. A. Paunescu Podeanu Street, 300569 Timisoara, Romania;
| | - Delia Muntean
- Multidisciplinary Research Centre on Antimicrobial Resistance, Department of Microbiology, University of Medicine and Pharmacy “Victor Babes”, Eftimie Murgu Sq. No. 2, 300041 Timişoara, Romania;
| | - Alexandra Ivan
- Center of Immuno-Physiology and Biotechnologies (CIFBIOTEH), University of Medicine and Pharmacy “Victor Babes”, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania;
- OncoGen Centre, Clinical County Hospital “Pius Branzeu”, Blvd. Liviu Rebreanu 156, 300723 Timisoara, Romania;
| | - Iustina Mirabela Cristea
- OncoGen Centre, Clinical County Hospital “Pius Branzeu”, Blvd. Liviu Rebreanu 156, 300723 Timisoara, Romania;
| |
Collapse
|
3
|
Gheorghiu M, Polonschii C, Popescu O, Gheorghiu E. Advanced Optogenetic-Based Biosensing and Related Biomaterials. MATERIALS (BASEL, SWITZERLAND) 2021; 14:4151. [PMID: 34361345 PMCID: PMC8347019 DOI: 10.3390/ma14154151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/12/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023]
Abstract
The ability to stimulate mammalian cells with light, brought along by optogenetic control, has significantly broadened our understanding of electrically excitable tissues. Backed by advanced (bio)materials, it has recently paved the way towards novel biosensing concepts supporting bio-analytics applications transversal to the main biomedical stream. The advancements concerning enabling biomaterials and related novel biosensing concepts involving optogenetics are reviewed with particular focus on the use of engineered cells for cell-based sensing platforms and the available toolbox (from mere actuators and reporters to novel multifunctional opto-chemogenetic tools) for optogenetic-enabled real-time cellular diagnostics and biosensor development. The key advantages of these modified cell-based biosensors concern both significantly faster (minutes instead of hours) and higher sensitivity detection of low concentrations of bioactive/toxic analytes (below the threshold concentrations in classical cellular sensors) as well as improved standardization as warranted by unified analytic platforms. These novel multimodal functional electro-optical label-free assays are reviewed among the key elements for optogenetic-based biosensing standardization. This focused review is a potential guide for materials researchers interested in biosensing based on light-responsive biomaterials and related analytic tools.
Collapse
Affiliation(s)
- Mihaela Gheorghiu
- International Centre of Biodynamics, 1B Intrarea Portocalelor, 060101 Bucharest, Romania;
| | - Cristina Polonschii
- International Centre of Biodynamics, 1B Intrarea Portocalelor, 060101 Bucharest, Romania;
| | - Octavian Popescu
- Molecular Biology Center, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai-University, 400084 Cluj-Napoca, Romania;
- Institute of Biology Bucharest, Romanian Academy, 296 Splaiul Independentei, 060031 Bucharest, Romania
| | - Eugen Gheorghiu
- International Centre of Biodynamics, 1B Intrarea Portocalelor, 060101 Bucharest, Romania;
| |
Collapse
|
4
|
Zanetti TA, Biazi BI, Coatti GC, Baranoski A, Marques LA, Corveloni AC, Mantovani MS. Dimethoxycurcumin reduces proliferation and induces apoptosis in renal tumor cells more efficiently than demethoxycurcumin and curcumin. Chem Biol Interact 2021; 338:109410. [PMID: 33582110 DOI: 10.1016/j.cbi.2021.109410] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/22/2021] [Accepted: 02/08/2021] [Indexed: 12/21/2022]
Abstract
Curcumin (Cur), is a pigment with antiproliferative activity but has some pharmacokinetic limitations, which led researchers to look for more effective structure analogs. This work investigated the effects of Cur and compared them with the two analogs, demethoxycurcumin (DeMC) and dimethoxycurcumin (DiMC), to elucidate their mechanisms of action. The cytotoxic, antiproliferative, and genotoxic effects these compounds were correlated based on gene expression analysis in the human renal adenocarcinoma cells (786-O). Cur decreased CYP2D6 expression and exhibited cytotoxic effects, such as inducing monopolar spindle formation and mitotic arrest mediated by the increase in CDKN1A (p21) mRNA. This dysregulation induced cell death through a caspase-independent pathway but was mediated by decrease in MTOR and BCL2 mRNA expression, suggesting that apoptosis occurred by autophagy. DeMC and DiMC had similar effects in that they induced monopolar spindle and mitotic arrest, were genotoxic, and activated GADD45A, an important molecule in repair mechanisms, and CDKN1A. However, the induction of apoptosis by DeMC was delayed and regulated by the decrease of antiapoptotic mRNA BCL.XL and subsequent activation of caspase 9 and caspase 3/7. DiMC treatment increased the expression of CYP1A2, CYP2C19, and CYP3A4 and exhibited higher cytotoxicity compared with other compounds. It induced apoptosis by increasing mRNA expression of BBC3, MYC, and CASP7 and activation of caspase 9 and caspase 3/7. These data revealed that different gene regulation processes are involved in cell death induced by Cur, DeMC, and DiMC. All three can be considered as promising chemotherapy candidates, with DiMC showing the greatest potency.
Collapse
Affiliation(s)
- Thalita Alves Zanetti
- Department of General Biology, Center of Biological Sciences, Londrina State University - UEL, Rodovia Celso Garcia Cid, Pr 445 Km 380, Londrina, Paraná, Brazil.
| | - Bruna Isabela Biazi
- Department of General Biology, Center of Biological Sciences, Londrina State University - UEL, Rodovia Celso Garcia Cid, Pr 445 Km 380, Londrina, Paraná, Brazil
| | | | - Adrivanio Baranoski
- Department of General Biology, Center of Biological Sciences, Londrina State University - UEL, Rodovia Celso Garcia Cid, Pr 445 Km 380, Londrina, Paraná, Brazil
| | - Lilian Areal Marques
- Department of General Biology, Center of Biological Sciences, Londrina State University - UEL, Rodovia Celso Garcia Cid, Pr 445 Km 380, Londrina, Paraná, Brazil
| | - Amanda Cristina Corveloni
- Department of General Biology, Center of Biological Sciences, Londrina State University - UEL, Rodovia Celso Garcia Cid, Pr 445 Km 380, Londrina, Paraná, Brazil
| | - Mario Sergio Mantovani
- Department of General Biology, Center of Biological Sciences, Londrina State University - UEL, Rodovia Celso Garcia Cid, Pr 445 Km 380, Londrina, Paraná, Brazil
| |
Collapse
|
5
|
Gheorghiu M, Stănică L, Ghinia Tegla MG, Polonschii C, Bratu D, Popescu O, Badea T, Gheorghiu E. Cellular sensing platform with enhanced sensitivity based on optogenetic modulation of cell homeostasis. Biosens Bioelectron 2020; 154:112003. [PMID: 32056953 PMCID: PMC7685521 DOI: 10.1016/j.bios.2019.112003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 12/26/2019] [Accepted: 12/28/2019] [Indexed: 12/29/2022]
Abstract
We demonstrate a new biosensing concept with impact on the development of rapid, point of need cell based sensing with boosted sensitivity and wide relevance for bioanalysis. It involves optogenetic stimulation of cells stably transfected to express light sensitive protein channels for optical control of membrane potential and of ion homeostasis. Time-lapse impedance measurements are used to reveal cell dynamics changes encompassing cellular responses to bioactive stimuli and optically induced homeostasis disturbances. We prove that light driven perturbations of cell membrane potential induce homeostatic reactions and modulate transduction mechanisms that amplify cellular response to bioactive compounds. This allows cell based biosensors to respond more rapidly and sensitively to low concentrations of bioactive/toxic analytes: statistically relevant impedance changes are recorded in less than 30 min, in comparison with >8 h in the best alternative reported tests for the same low concentration (e.g. a concentration of 25 μM CdCl2, lower than the threshold concentration in classical cellular sensors). Comparative analysis of model bioactive/toxic compounds (ouabain and CdCl2) demonstrates that cellular reactivity can be boosted by light driven perturbations of cellular homeostasis and that this biosensing concept is able to discriminate analytes with different modes of action (i.e. CdCl2 toxicity versus ion pump inhibition by ouabain), a significant advance against state of the art cell based sensors.
Collapse
Affiliation(s)
- Mihaela Gheorghiu
- International Centre of Biodynamics, Intr. Portocalelor 1 B, 060101, Bucharest, Romania; Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, Bucharest, 050095, Romania.
| | - Luciana Stănică
- International Centre of Biodynamics, Intr. Portocalelor 1 B, 060101, Bucharest, Romania; Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, Bucharest, 050095, Romania
| | - Miruna G Ghinia Tegla
- Institute for Interdisciplinary Research in Bio-Nano-Sciences, Molecular Biology Center, Babes-Bolyai-University, 400084, Cluj-Napoca, Romania; Retinal Circuit Development & Genetics Unit N-NRL/NEI/NIH 6 Center Drive Bethesda, 20892, Maryland, United States
| | - Cristina Polonschii
- International Centre of Biodynamics, Intr. Portocalelor 1 B, 060101, Bucharest, Romania.
| | - Dumitru Bratu
- International Centre of Biodynamics, Intr. Portocalelor 1 B, 060101, Bucharest, Romania.
| | - Octavian Popescu
- Institute for Interdisciplinary Research in Bio-Nano-Sciences, Molecular Biology Center, Babes-Bolyai-University, 400084, Cluj-Napoca, Romania; Institute of Biology, Romanian Academy, 296 Splaiul Independentei, 060031, Bucharest, Romania
| | - Tudor Badea
- Retinal Circuit Development & Genetics Unit N-NRL/NEI/NIH 6 Center Drive Bethesda, 20892, Maryland, United States.
| | - Eugen Gheorghiu
- International Centre of Biodynamics, Intr. Portocalelor 1 B, 060101, Bucharest, Romania; Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, Bucharest, 050095, Romania.
| |
Collapse
|
6
|
Brown WGA, Needham K, Begeng JM, Thompson AC, Nayagam BA, Kameneva T, Stoddart PR. Thermal damage threshold of neurons during infrared stimulation. BIOMEDICAL OPTICS EXPRESS 2020; 11:2224-2234. [PMID: 32341879 PMCID: PMC7173919 DOI: 10.1364/boe.383165] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/11/2020] [Accepted: 03/23/2020] [Indexed: 05/25/2023]
Abstract
In infrared neural stimulation (INS), laser-evoked thermal transients are used to generate small depolarising currents in neurons. The laser exposure poses a moderate risk of thermal damage to the target neuron. Indeed, exogenous methods of neural stimulation often place the target neurons under stressful non-physiological conditions, which can hinder ordinary neuronal function and hasten cell death. Therefore, quantifying the exposure-dependent probability of neuronal damage is essential for identifying safe operating limits of INS and other interventions for therapeutic and prosthetic use. Using patch-clamp recordings in isolated spiral ganglion neurons, we describe a method for determining the dose-dependent damage probabilities of individual neurons in response to both acute and cumulative infrared exposure parameters based on changes in injection current. The results identify a local thermal damage threshold at approximately 60 °C, which is in keeping with previous literature and supports the claim that damage during INS is a purely thermal phenomenon. In principle this method can be applied to any potentially injurious stimuli, allowing for the calculation of a wide range of dose-dependent neural damage probabilities. Unlike histological analyses, the technique is well-suited to quantifying gradual neuronal damage, and critical threshold behaviour is not required.
Collapse
Affiliation(s)
- William G. A. Brown
- Faculty of Science, Engineering and Technology, Swinburne University of Technology, John Street, Hawthorn, VIC 3122, Australia
| | - Karina Needham
- Department of Surgery (Otolaryngology), University of Melbourne, Royal Victoria Eye & Ear Hospital, 32 Gisborne St, East Melbourne, VIC 3002, Australia
| | - James M. Begeng
- Faculty of Science, Engineering and Technology, Swinburne University of Technology, John Street, Hawthorn, VIC 3122, Australia
| | | | - Bryony A. Nayagam
- Department of Audiology and Speech Pathology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Tatiana Kameneva
- Faculty of Science, Engineering and Technology, Swinburne University of Technology, John Street, Hawthorn, VIC 3122, Australia
| | - Paul R. Stoddart
- Faculty of Science, Engineering and Technology, Swinburne University of Technology, John Street, Hawthorn, VIC 3122, Australia
| |
Collapse
|
7
|
Fasbender F, Obholzer M, Metzler S, Stöber R, Hengstler JG, Watzl C. Enhanced activation of human NK cells by drug-exposed hepatocytes. Arch Toxicol 2020; 94:439-448. [PMID: 32060585 DOI: 10.1007/s00204-020-02668-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/06/2020] [Indexed: 01/06/2023]
Abstract
Drug-induced liver injury (DILI) represents one of the major causes why drugs have to be withdrawn from the market. In this study, we describe a new interaction between drug-exposed hepatocytes and natural killer (NK) cells. In a previous genome-wide expression analysis of primary human hepatocytes that had been exposed to clinically relevant concentrations of 148 drugs, we found that several activating ligands for NK cell receptors were regulated by various drugs (e.g., valproic acid, ketoconazole, promethazine, isoniazid). Especially expression of the activating NKG2D ligands (MICA, MICB and ULBPs) and the NKp30 ligand B7-H6 were upregulated in primary human hepatocytes upon exposure to many different drugs. Using the human hepatocyte cell lines Huh7 and HepG2, we confirmed that protein levels of activating NK cell ligands were elevated after drug exposure. Hepatocyte cell lines or primary human hepatocytes co-cultivated with NK cells caused enhanced NK cell activation after pretreatment with drugs at in vivo relevant concentrations compared to solvent controls. Enhanced NK cell activation was evident by increased cytotoxicity against hepatocytes and interferon (IFN)-γ production. NK cell activation could be blocked by specific antibodies against activating NK cell receptors. These data support the hypothesis that NK cells can modulate drug-induced liver injury by direct interaction with hepatocytes resulting in cytotoxicity and IFN-γ production.
Collapse
Affiliation(s)
- Frank Fasbender
- Department of Immunology, Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Martin Obholzer
- Department of Immunology, Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Sarah Metzler
- Department of Immunology, Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Regina Stöber
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Jan G Hengstler
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Carsten Watzl
- Department of Immunology, Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany.
| |
Collapse
|
8
|
Morgan K, Gamal W, Samuel K, Morley SD, Hayes PC, Bagnaninchi P, Plevris JN. Application of Impedance-Based Techniques in Hepatology Research. J Clin Med 2019; 9:jcm9010050. [PMID: 31878354 PMCID: PMC7019217 DOI: 10.3390/jcm9010050] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 12/22/2022] Open
Abstract
There are a variety of end-point assays and techniques available to monitor hepatic cell cultures and study toxicity within in vitro models. These commonly focus on one aspect of cell metabolism and are often destructive to cells. Impedance-based cellular assays (IBCAs) assess biological functions of cell populations in real-time by measuring electrical impedance, which is the resistance to alternating current caused by the dielectric properties of proliferating of cells. While the uses of IBCA have been widely reported for a number of tissues, specific uses in the study of hepatic cell cultures have not been reported to date. IBCA monitors cellular behaviour throughout experimentation non-invasively without labelling or damage to cell cultures. The data extrapolated from IBCA can be correlated to biological events happening within the cell and therefore may inform drug toxicity studies or other applications within hepatic research. Because tight junctions comprise the blood/biliary barrier in hepatocytes, there are major consequences when these junctions are disrupted, as many pathologies centre around the bile canaliculi and flow of bile out of the liver. The application of IBCA in hepatology provides a unique opportunity to assess cellular polarity and patency of tight junctions, vital to maintaining normal hepatic function. Here, we describe how IBCAs have been applied to measuring the effect of viral infection, drug toxicity /IC50, cholangiopathies, cancer metastasis and monitoring of the gut-liver axis. We also highlight key areas of research where IBCAs could be used in future applications within the field of hepatology.
Collapse
Affiliation(s)
- Katie Morgan
- The University of Edinburgh Hepatology Laboratory, Division of Heath Sciences, University of Edinburgh Medical School, Chancellor’s Building, Edinburgh BioQuarter, 49 Little France Crescent, Edinburgh EH16 4SB, UK; (S.D.M.); (P.C.H.); (J.N.P.)
- Correspondence:
| | - Wesam Gamal
- James Nasmyth Building, Institute of Mechanical, Process and Energy Engineering, Heriot-Watt University School of Engineering and Physical Sciences, Edinburgh EH14 4AS, UK;
| | - Kay Samuel
- The Jack Copland Centre, Advanced Therapeutics, Scottish National Blood Transfusion Service, 52 Research Avenue North, Edinburgh EH14 4BE, UK;
| | - Steven D. Morley
- The University of Edinburgh Hepatology Laboratory, Division of Heath Sciences, University of Edinburgh Medical School, Chancellor’s Building, Edinburgh BioQuarter, 49 Little France Crescent, Edinburgh EH16 4SB, UK; (S.D.M.); (P.C.H.); (J.N.P.)
| | - Peter C. Hayes
- The University of Edinburgh Hepatology Laboratory, Division of Heath Sciences, University of Edinburgh Medical School, Chancellor’s Building, Edinburgh BioQuarter, 49 Little France Crescent, Edinburgh EH16 4SB, UK; (S.D.M.); (P.C.H.); (J.N.P.)
| | - Pierre Bagnaninchi
- MRC Centre for Regenerative Medicine 5 Little France Drive, Edinburgh EH16 4UU, UK;
| | - John N. Plevris
- The University of Edinburgh Hepatology Laboratory, Division of Heath Sciences, University of Edinburgh Medical School, Chancellor’s Building, Edinburgh BioQuarter, 49 Little France Crescent, Edinburgh EH16 4SB, UK; (S.D.M.); (P.C.H.); (J.N.P.)
| |
Collapse
|
9
|
Zanetti TA, Biazi BI, Coatti GC, Baranoski A, Marques LA, Corveloni AC, Mantovani MS. Mitotic spindle defects and DNA damage induced by dimethoxycurcumin lead to an intrinsic apoptosis pathway in HepG2/C3A cells. Toxicol In Vitro 2019; 61:104643. [DOI: 10.1016/j.tiv.2019.104643] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/30/2019] [Accepted: 09/06/2019] [Indexed: 01/30/2023]
|
10
|
Synergistic Cytotoxicity of Renieramycin M and Doxorubicin in MCF-7 Breast Cancer Cells. Mar Drugs 2019; 17:md17090536. [PMID: 31527453 PMCID: PMC6780817 DOI: 10.3390/md17090536] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/05/2019] [Accepted: 08/07/2019] [Indexed: 12/17/2022] Open
Abstract
Renieramycin M (RM) is a KCN-stabilized tetrahydroisoquinoline purified from the blue sponge Xestospongia sp., with nanomolar IC50s against several cancer cell lines. Our goal is to evaluate its combination effects with doxorubicin (DOX) in estrogen receptor positive MCF-7 breast cancer cells. MCF-7 cells were treated simultaneously or sequentially with various combination ratios of RM and DOX for 72 h. Cell viability was determined using the MTT assay. Synergism or antagonism was determined using curve-shift analysis, combination index method and isobologram analysis. Synergism was observed with pharmacologically achievable concentrations of DOX when administered simultaneously, but not sequentially. The IC95 values of RM and DOX after combination were reduced by up to four-fold and eight-fold, respectively. To gain insights on the mechanism of synergy, real-time profiling, cell cycle analysis, apoptosis assays, and transcriptome analysis were conducted. The combination treatment displayed a similar profile with DNA-damaging agents and induced a greater and faster cell killing. The combination treatment also showed an increase in apoptosis. DOX induced S and G2/M arrest while RM did not induce significant changes in the cell cycle. DNA replication and repair genes were downregulated commonly by RM and DOX. p53 signaling and cell cycle checkpoints were regulated by DOX while ErbB/PI3K-Akt, integrin and focal adhesion signaling were regulated by RM upon combination. Genes involved in cytochrome C release and interferon gamma signaling were regulated specifically in the combination treatment. This study serves as a basis for in vivo studies and provides a rationale for using RM in combination with other anticancer drugs.
Collapse
|
11
|
Chiu CH, Lei KF, Chan YS, Ueng SWN, Chen ACY. Real-time detection of antibiotics cytotoxicity in rabbit periosteal cells using microfluidic devices with comparison to conventional culture assays. BMC Musculoskelet Disord 2019; 20:339. [PMID: 31349830 PMCID: PMC6659314 DOI: 10.1186/s12891-019-2705-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 07/04/2019] [Indexed: 02/13/2023] Open
Abstract
BACKGROUND Local antibiotic application has been widely used in orthopedic surgery. The dose-related toxicity of antibiotics towards periosteal tissues and resulting effects on osteogenic expression are yet to be studied. METHODS Periosteal cells harvested from the medial tibia of New Zealand White rabbits were used. A seeding density of 5 × 103 cells/cm2 was determined to be optimal for testing in the pilot study; the cells were cultured in xCELLigence 96-well plates. Microfluidic impedance analyzers were used to monitor cellular proliferation in microfluidic culture systems with exposure to three different concentrations (10 μg/mL, 100 μg/mL, and 1000 μg/mL) of cefazolin, ciprofloxacin, and vancomycin, respectively. The correlation of cell index at day 7 with optical density values from WST-1 assays using conventional cultures was evaluated by calculating the Pearson's coefficient. RNA analysis was performed to investigate the expression of osteogenic markers in the cultured cells, including core-binding factor alpha 1 (Cbfa1), osteopontin (OPN), and osteopontin promoter (OPNp), relative to glyceraldehyde-3-phosphate dehydrogenase (GAPDH) as the endogenous control. RESULTS A significant dose-related inhibition of cell index was found for all the 3 antibiotics, whereas the WST-1 assays showed a significant dose-related inhibition of cellular proliferation only at a high dose of cefazolin (1000 μg/mL) and medium-to-high dose of ciprofloxacin (100 μg/mL and 1000 μg/mL). Pearson's coefficient analysis indicated a high correlation between the cell index and optical density values of WST-1 assays only for medium and high doses of ciprofloxacin (100 μg/mL and 1000 μg/mL); a moderate correlation was seen for cefazolin, and a low dose of ciprofloxacin (10 μg/mL). RNA analysis confirmed significant dose-related inhibition of cfba1, OPN, and OPNp expression by all three antibiotics. CONCLUSION With optimal seeding amounts, rabbit periosteal cells can be dynamically monitored in the xCELLigence microfluidic system. Dose-related inhibition of cellular proliferation and osteogenic expression was found after exposure to cefazolin and ciprofloxacin. By providing real-time detection and exhibiting comparable correlation, microfluidic impedance-based analyzer is a feasible alternative to the conventional WST-1 assays.
Collapse
Affiliation(s)
- Chih-Hao Chiu
- Bone and Joint Research Center, Department of Orthopedic Surgery, Chang Gung Memorial Hospital-Linkou and University College of Medicine, 5th, Fu-Shin Street, Kweishan Dist, Taoyuan, 333 Taiwan, Republic of China
- Graduate Institute of Medical Mechatronics, Chang Gung University, Taiwan, Republic of China
| | - Kin Fong Lei
- Graduate Institute of Medical Mechatronics, Chang Gung University, Taiwan, Republic of China
| | - Yi-Sheng Chan
- Bone and Joint Research Center, Department of Orthopedic Surgery, Chang Gung Memorial Hospital-Linkou and University College of Medicine, 5th, Fu-Shin Street, Kweishan Dist, Taoyuan, 333 Taiwan, Republic of China
| | - Steve W. N. Ueng
- Bone and Joint Research Center, Department of Orthopedic Surgery, Chang Gung Memorial Hospital-Linkou and University College of Medicine, 5th, Fu-Shin Street, Kweishan Dist, Taoyuan, 333 Taiwan, Republic of China
| | - Alvin Chao-Yu Chen
- Bone and Joint Research Center, Department of Orthopedic Surgery, Chang Gung Memorial Hospital-Linkou and University College of Medicine, 5th, Fu-Shin Street, Kweishan Dist, Taoyuan, 333 Taiwan, Republic of China
| |
Collapse
|
12
|
Gamal W, Wu H, Underwood I, Jia J, Smith S, Bagnaninchi PO. Impedance-based cellular assays for regenerative medicine. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0226. [PMID: 29786561 DOI: 10.1098/rstb.2017.0226] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2018] [Indexed: 12/22/2022] Open
Abstract
Therapies based on regenerative techniques have the potential to radically improve healthcare in the coming years. As a result, there is an emerging need for non-destructive and label-free technologies to assess the quality of engineered tissues and cell-based products prior to their use in the clinic. In parallel, the emerging regenerative medicine industry that aims to produce stem cells and their progeny on a large scale will benefit from moving away from existing destructive biochemical assays towards data-driven automation and control at the industrial scale. Impedance-based cellular assays (IBCA) have emerged as an alternative approach to study stem-cell properties and cumulative studies, reviewed here, have shown their potential to monitor stem-cell renewal, differentiation and maturation. They offer a novel method to non-destructively assess and quality-control stem-cell cultures. In addition, when combined with in vitro disease models they provide complementary insights as label-free phenotypic assays. IBCA provide quantitative and very sensitive results that can easily be automated and up-scaled in multi-well format. When facing the emerging challenge of real-time monitoring of three-dimensional cell culture dielectric spectroscopy and electrical impedance tomography represent viable alternatives to two-dimensional impedance sensing.This article is part of the theme issue 'Designer human tissue: coming to a lab near you'.
Collapse
Affiliation(s)
- W Gamal
- School of Electronic Engineering, Bangor University, Bangor LL57 1UT, UK
| | - H Wu
- School of Engineering, University of Edinburgh, Edinburgh EH9 3FB, UK
| | - I Underwood
- School of Engineering, University of Edinburgh, Edinburgh EH9 3FB, UK
| | - J Jia
- School of Engineering, University of Edinburgh, Edinburgh EH9 3FB, UK
| | - S Smith
- School of Engineering, University of Edinburgh, Edinburgh EH9 3FB, UK
| | - P O Bagnaninchi
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| |
Collapse
|
13
|
Franke R, Hinkelmann B, Fetz V, Stradal T, Sasse F, Klawonn F, Brönstrup M. xCELLanalyzer: A Framework for the Analysis of Cellular Impedance Measurements for Mode of Action Discovery. SLAS DISCOVERY 2019; 24:213-223. [PMID: 30681906 DOI: 10.1177/2472555218819459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Mode of action (MoA) identification of bioactive compounds is very often a challenging and time-consuming task. We used a label-free kinetic profiling method based on an impedance readout to monitor the time-dependent cellular response profiles for the interaction of bioactive natural products and other small molecules with mammalian cells. Such approaches have been rarely used so far due to the lack of data mining tools to properly capture the characteristics of the impedance curves. We developed a data analysis pipeline for the xCELLigence Real-Time Cell Analysis detection platform to process the data, assess and score their reproducibility, and provide rank-based MoA predictions for a reference set of 60 bioactive compounds. The method can reveal additional, previously unknown targets, as exemplified by the identification of tubulin-destabilizing activities of the RNA synthesis inhibitor actinomycin D and the effects on DNA replication of vioprolide A. The data analysis pipeline is based on the statistical programming language R and is available to the scientific community through a GitHub repository.
Collapse
Affiliation(s)
- Raimo Franke
- 1 Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Bettina Hinkelmann
- 1 Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Verena Fetz
- 1 Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Theresia Stradal
- 2 Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Florenz Sasse
- 1 Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Frank Klawonn
- 3 Biostatistics Group, Helmholtz Centre for Infection Research, Braunschweig, Germany.,4 Department of Computer Science, Ostfalia University, Wolfenbuettel, Germany
| | - Mark Brönstrup
- 1 Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,5 Center of Biomolecular Drug Research (BMWZ), Institute of Organic Chemistry, Leibniz Universität, Hannover, Germany
| |
Collapse
|
14
|
Batista L, Bastogne T, Delaunois A, Valentin JP, Atienzar F. A novel statistical signal processing method to estimate effects of compounds on contractility of cardiomyocytes using impedance assays. Biomed Signal Process Control 2018. [DOI: 10.1016/j.bspc.2018.05.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
15
|
In vitro screening of cell bioenergetics to assess mitochondrial dysfunction in drug development. Toxicol In Vitro 2018; 52:374-383. [PMID: 30030051 DOI: 10.1016/j.tiv.2018.07.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 05/28/2018] [Accepted: 07/15/2018] [Indexed: 12/17/2022]
Abstract
Drug-induced mitochondrial toxicity is considered as a common cellular mechanism that can induce a variety of organ toxicities. In the present manuscript, 17 in vitro mitochondrial toxic drugs, reported to induce Drug-Induced Liver Injury (DILI) and 6 non-mitochondrial toxic drugs (3 with DILI and 3 without DILI concern), were tested in HepG2 cells using a bioenergetics system. The 17 mitochondrial toxic drugs represent a wide variety of mitochondrial dysfunctions as well as DILI and include 4 pairs of drugs which are structurally related but associated with different DILI concerns in human. Cell bioenergetics were measured using the XF96e analyzer which simultaneous monitor oxygen consumption rate (OCR) and extracellular acidification rate (ECAR), indirect measurements of oxidative phosphorylation and glycolysis, respectively. OCR associated with ATP production, maximal respiration, proton leak and spare respiratory capacity, were also assessed. Duplicate experiments resulted in a sensitivity of 82% (14/17) and specificity of 83% (5/6). The addition of stressors improved specificity considerably. Cut-offs, statistics and rules are clearly discussed to facilitate the use of this assay for screening purposes. Overall, the authors consider that this assay should be part of the battery of safety screening assays at early stages of drug development.
Collapse
|
16
|
Abstract
The use of impedance-based label free cell analysis is increasingly popular and has many different applications. Here, we report that a real-time cell analyzer (RTCA) can be used to study the stimulation of Natural Killer (NK) cells. Engagement of NK cells via plate-bound antibodies directed against different activating surface receptors could be measured in real time using the label-free detection of impedance. The change in impedance was dependent on early signal transduction events in the NK cells as it was blocked by inhibitors of Src-family kinases and by inhibiting actin polymerization. While CD16 was the only receptor that could induce a strong change in impedance in primary NK cells, several activating receptors induced changes in impedance in expanded NK cells. Using PBMCs we could detect T cell receptor-mediated T cell activation and CD16-mediated NK cell activation in the same sample. Performing a dose-response analysis for the Src-family kinases inhibitor PP1 we show that T cells are more sensitive to inhibition compared to NK cells. Our data demonstrate that the RTCA can be used to detect physiological activation events in NK cells in a label-free and real-time fashion.
Collapse
|
17
|
Loiodice S, Nogueira da Costa A, Atienzar F. Current trends in in silico, in vitro toxicology, and safety biomarkers in early drug development. Drug Chem Toxicol 2017; 42:113-121. [DOI: 10.1080/01480545.2017.1400044] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Simon Loiodice
- Department of Non-Clinical Development, UCB Biopharma SPRL, Braine-l’Alleud, Belgium
| | | | - Franck Atienzar
- Department of Non-Clinical Development, UCB Biopharma SPRL, Braine-l’Alleud, Belgium
| |
Collapse
|
18
|
Pisani C, Rascol E, Dorandeu C, Charnay C, Guari Y, Chopineau J, Devoisselle JM, Prat O. Biocompatibility assessment of functionalized magnetic mesoporous silica nanoparticles in human HepaRG cells. Nanotoxicology 2017; 11:871-890. [PMID: 28937306 DOI: 10.1080/17435390.2017.1378749] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Magnetic mesoporous silica nanoparticles (M-MSNs) are a promising class of nanoparticles for drug delivery. However, a deep understanding of the toxicological mechanisms of action of these nanocarriers is essential, especially in the liver. The potential toxicity on HepaRG cells of pristine, pegylated (PEG), and lipid (DMPC) M-MSNs were compared. Based on MTT assay and real-time cell impedance, none of these NPs presented an extensive toxicity on hepatic cells. However, we observed by transmission electron microscopy (TEM) that the DMPC and pristine M-MSNs were greatly internalized. In comparison, PEG M-MSNs showed a slower cellular uptake. Whole gene expression profiling revealed the M-MSNs molecular modes of action in a time- and dose-dependent manner. The lowest dose tested (1.6 µg/cm2) induced no molecular effect and was defined as 'No Observed Transcriptional Effect level.' The dose 16 µg/cm2 revealed nascent but transient effects. At the highest dose (80 µg/cm2), adverse effects have clearly arisen and increased over time. The limit of biocompatibility for HepaRG cells could be set at 16 µg/cm2 for these NPs. Thanks to a comparative pathway-driven analysis, we highlighted the sequence of events that leads to the disruption of hepatobiliary system, elicited by the three types of M-MSNs, at the highest dose. The Adverse Outcome Pathway of hepatic cholestasis was implicated. Toxicogenomics applied to cell cultures is an effective tool to characterize and compare the modes of action of many substances. We propose this strategy as an asset for upstream selection of the safest nanocarriers in the framework of regulation for nanobiosafety.
Collapse
Affiliation(s)
- Cédric Pisani
- a MACS, UMR 5253 CNRS-ENSCM-UM , Institut Charles Gerhardt de Montpellier , Montpellier , France.,b Direction de la Recherche Fondamentale-BIAM , CEA , Bagnols-sur-Cèze , France
| | - Estelle Rascol
- a MACS, UMR 5253 CNRS-ENSCM-UM , Institut Charles Gerhardt de Montpellier , Montpellier , France
| | - Christophe Dorandeu
- a MACS, UMR 5253 CNRS-ENSCM-UM , Institut Charles Gerhardt de Montpellier , Montpellier , France
| | - Clarence Charnay
- c IMNO, UMR 5253 CNRS-ENSCM-UM , Institut Charles Gerhardt de Montpellier , Montpellier , France
| | - Yannick Guari
- c IMNO, UMR 5253 CNRS-ENSCM-UM , Institut Charles Gerhardt de Montpellier , Montpellier , France
| | - Joël Chopineau
- a MACS, UMR 5253 CNRS-ENSCM-UM , Institut Charles Gerhardt de Montpellier , Montpellier , France
| | - Jean-Marie Devoisselle
- a MACS, UMR 5253 CNRS-ENSCM-UM , Institut Charles Gerhardt de Montpellier , Montpellier , France
| | - Odette Prat
- b Direction de la Recherche Fondamentale-BIAM , CEA , Bagnols-sur-Cèze , France
| |
Collapse
|
19
|
Pisani C, Rascol E, Dorandeu C, Gaillard JC, Charnay C, Guari Y, Chopineau J, Armengaud J, Devoisselle JM, Prat O. The species origin of the serum in the culture medium influences the in vitro toxicity of silica nanoparticles to HepG2 cells. PLoS One 2017; 12:e0182906. [PMID: 28796831 PMCID: PMC5552166 DOI: 10.1371/journal.pone.0182906] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 07/26/2017] [Indexed: 01/08/2023] Open
Abstract
The formation of a protein corona around nanoparticles can influence their toxicity, triggering cellular responses that may be totally different from those elicited by pristine nanoparticles. The main objective of this study was to investigate whether the species origin of the serum proteins forming the corona influences the in vitro toxicity assessment of silica nanoparticles. Coronas were preformed around nanoparticles before cell exposures by incubation in fetal bovine (FBS) or human (HS) serum. The compositions of these protein coronas were assessed by nano-LC MS/MS. The effects of these protein-coated nanoparticles on HepG2 cells were monitored using real-time cell impedance technology. The nanoparticle coronas formed in human or fetal bovine serum comprised many homologous proteins. Using human compared with fetal bovine serum, nanoparticle toxicity in HepG2 cells decreased by 4-fold and 1.5-fold, when used at 50 and 10μg/mL, respectively. It is likely that "markers of self" are present in the serum and are recognized by human cell receptors. Preforming a corona with human serum seems to be more appropriate for in vitro toxicity testing of potential nanocarriers using human cells. In vitro cytotoxicity assays must reflect in vivo conditions as closely as possible to provide solid and useful results.
Collapse
Affiliation(s)
- Cédric Pisani
- Institut Charles Gerhardt de Montpellier, MACS, UMR 5253 CNRS-ENSCM-UM, Place Eugène Bataillon, Montpellier, France
- CEA, Direction de la Recherche Fondamentale—BIAM, Site de Marcoule, Bagnols-sur-Cèze, France
| | - Estelle Rascol
- Institut Charles Gerhardt de Montpellier, MACS, UMR 5253 CNRS-ENSCM-UM, Place Eugène Bataillon, Montpellier, France
| | - Christophe Dorandeu
- Institut Charles Gerhardt de Montpellier, MACS, UMR 5253 CNRS-ENSCM-UM, Place Eugène Bataillon, Montpellier, France
| | - Jean-Charles Gaillard
- CEA, Direction de la Recherche Fondamentale—IBITECS, Site de Marcoule, Bagnols-sur-Cèze, France
| | - Clarence Charnay
- Institut Charles Gerhardt de Montpellier, IMNO, UMR 5253 CNRS-ENSCM-UM, 1701, Place Eugène Bataillon, Montpellier, France
| | - Yannick Guari
- Institut Charles Gerhardt de Montpellier, IMNO, UMR 5253 CNRS-ENSCM-UM, 1701, Place Eugène Bataillon, Montpellier, France
| | - Joël Chopineau
- Institut Charles Gerhardt de Montpellier, MACS, UMR 5253 CNRS-ENSCM-UM, Place Eugène Bataillon, Montpellier, France
- Université de Nîmes Rue Georges Salan, Nîmes, France
| | - Jean Armengaud
- CEA, Direction de la Recherche Fondamentale—IBITECS, Site de Marcoule, Bagnols-sur-Cèze, France
| | - Jean-Marie Devoisselle
- Institut Charles Gerhardt de Montpellier, MACS, UMR 5253 CNRS-ENSCM-UM, Place Eugène Bataillon, Montpellier, France
| | - Odette Prat
- CEA, Direction de la Recherche Fondamentale—BIAM, Site de Marcoule, Bagnols-sur-Cèze, France
| |
Collapse
|
20
|
Weaver RJ, Betts C, Blomme EAG, Gerets HHJ, Gjervig Jensen K, Hewitt PG, Juhila S, Labbe G, Liguori MJ, Mesens N, Ogese MO, Persson M, Snoeys J, Stevens JL, Walker T, Park BK. Test systems in drug discovery for hazard identification and risk assessment of human drug-induced liver injury. Expert Opin Drug Metab Toxicol 2017; 13:767-782. [PMID: 28604124 DOI: 10.1080/17425255.2017.1341489] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The liver is an important target for drug-induced toxicities. Early detection of hepatotoxic drugs requires use of well-characterized test systems, yet current knowledge, gaps and limitations of tests employed remains an important issue for drug development. Areas Covered: The current state of the science, understanding and application of test systems in use for the detection of drug-induced cytotoxicity, mitochondrial toxicity, cholestasis and inflammation is summarized. The test systems highlighted herein cover mostly in vitro and some in vivo models and endpoint measurements used in the assessment of small molecule toxic liabilities. Opportunities for research efforts in areas necessitating the development of specific tests and improved mechanistic understanding are highlighted. Expert Opinion: Use of in vitro test systems for safety optimization will remain a core activity in drug discovery. Substantial inroads have been made with a number of assays established for human Drug-induced Liver Injury. There nevertheless remain significant gaps with a need for improved in vitro tools and novel tests to address specific mechanisms of human Drug-Induced Liver Injury. Progress in these areas will necessitate not only models fit for application, but also mechanistic understanding of how chemical insult on the liver occurs in order to identify translational and quantifiable readouts for decision-making.
Collapse
Affiliation(s)
- Richard J Weaver
- a Research & Biopharmacy, Institut de Recherches Internationales Servier , Suresnes , France
| | - Catherine Betts
- b Pathology Sciences, Drug Safety and Metabolism , AstraZeneca R&D , Cambridge , UK
| | | | - Helga H J Gerets
- d Non Clinical Development, Chemin du Foriest , UCB BioPharma SPRL , Braine L'Alleud , Belgium
| | | | - Philip G Hewitt
- f Non-Clinical Development, Merck KGaA , Darmstadt , Germany
| | - Satu Juhila
- g In Vitro Biology , Orion Pharma , Espoo , Finland
| | - Gilles Labbe
- h Investigative Toxicology, Preclinical Safety , Sanofi R&D , Paris , France
| | | | - Natalie Mesens
- i Preclinical Development & Safety, Janssen (Pharmaceutical Companies of Johnson & Johnson) Turnhoutseweg 30 , Beerse , Belgium
| | - Monday O Ogese
- j Pathology Sciences, Drug Safety and Metabolism , AstraZeneca R&D , Cambridge , UK
| | - Mikael Persson
- k Innovative Medicines and Early Clinical Development, Drug Safety and Metabolism, Discovery Safety , AstraZeneca R&D , Mölndal , Sweden
| | - Jan Snoeys
- l Pharmacokinetics Dynamics & Metabolism, Janssen (Pharmaceutical Companies of Johnson & Johnson) Turnhoutseweg 30 , Beerse , Belgium
| | - James L Stevens
- m Dept of Toxicology , Lilly Research Laboratories, Eli Lilly and Company , Indianapolis , Indiana , USA
| | - Tracy Walker
- n Investigative Safety & Drug Metabolism , GlaxoSmithKline, David Jack Centre for Research and Development , Ware , Herts , Hertfordshire, UK
| | - B Kevin Park
- o Institute of Translational Medicine , University of Liverpool , Liverpool , UK
| |
Collapse
|
21
|
Gamal W, Treskes P, Samuel K, Sullivan GJ, Siller R, Srsen V, Morgan K, Bryans A, Kozlowska A, Koulovasilopoulos A, Underwood I, Smith S, Del-Pozo J, Moss S, Thompson AI, Henderson NC, Hayes PC, Plevris JN, Bagnaninchi PO, Nelson LJ. Low-dose acetaminophen induces early disruption of cell-cell tight junctions in human hepatic cells and mouse liver. Sci Rep 2017; 7:37541. [PMID: 28134251 PMCID: PMC5278402 DOI: 10.1038/srep37541] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 10/31/2016] [Indexed: 02/07/2023] Open
Abstract
Dysfunction of cell-cell tight junction (TJ) adhesions is a major feature in the pathogenesis of various diseases. Liver TJs preserve cellular polarity by delimiting functional bile-canalicular structures, forming the blood-biliary barrier. In acetaminophen-hepatotoxicity, the mechanism by which tissue cohesion and polarity are affected remains unclear. Here, we demonstrate that acetaminophen, even at low-dose, disrupts the integrity of TJ and cell-matrix adhesions, with indicators of cellular stress with liver injury in the human hepatic HepaRG cell line, and primary hepatocytes. In mouse liver, at human-equivalence (therapeutic) doses, dose-dependent loss of intercellular hepatic TJ-associated ZO-1 protein expression was evident with progressive clinical signs of liver injury. Temporal, dose-dependent and specific disruption of the TJ-associated ZO-1 and cytoskeletal-F-actin proteins, correlated with modulation of hepatic ultrastructure. Real-time impedance biosensing verified in vitro early, dose-dependent quantitative decreases in TJ and cell-substrate adhesions. Whereas treatment with NAPQI, the reactive metabolite of acetaminophen, or the PKCα-activator and TJ-disruptor phorbol-12-myristate-13-acetate, similarly reduced TJ integrity, which may implicate oxidative stress and the PKC pathway in TJ destabilization. These findings are relevant to the clinical presentation of acetaminophen-hepatotoxicity and may inform future mechanistic studies to identify specific molecular targets and pathways that may be altered in acetaminophen-induced hepatic depolarization.
Collapse
Affiliation(s)
- Wesam Gamal
- MRC Centre for Regenerative Medicine, SCRM Building, The University of Edinburgh, Edinburgh BioQuarter, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Philipp Treskes
- Hepatology Laboratory, University of Edinburgh, Royal Infirmary of Edinburgh, 49 Little France Crescent EH16 4SB, UK
| | - Kay Samuel
- Scottish National Blood Transfusion Service, Research, Development and Innovation Directorate, Cell Therapy Group, Ellens Glen Road, Edinburgh, EH17 7QT, UK
| | - Gareth J Sullivan
- Department of Biochemistry, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, PO Box 1112 Blindern, 0317 Oslo, Norway, UK.,Norwegian Center for Stem Cell Research, PO Box 1112 Blindern, 0317 Oslo, Norway.,Institute of Immunology, Oslo University Hospital-Rikshospitalet, PO Box 4950 Nydalen, Oslo 0424, Norway
| | - Richard Siller
- Department of Biochemistry, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, PO Box 1112 Blindern, 0317 Oslo, Norway, UK
| | - Vlastimil Srsen
- Institute for Bioengineering, University of Edinburgh, King's Buildings, Colin MacLaurin Road, EH9 3DW, UK
| | - Katie Morgan
- Hepatology Laboratory, University of Edinburgh, Royal Infirmary of Edinburgh, 49 Little France Crescent EH16 4SB, UK
| | - Anna Bryans
- Hepatology Laboratory, University of Edinburgh, Royal Infirmary of Edinburgh, 49 Little France Crescent EH16 4SB, UK
| | - Ada Kozlowska
- Hepatology Laboratory, University of Edinburgh, Royal Infirmary of Edinburgh, 49 Little France Crescent EH16 4SB, UK
| | - Andreas Koulovasilopoulos
- Hepatology Laboratory, University of Edinburgh, Royal Infirmary of Edinburgh, 49 Little France Crescent EH16 4SB, UK
| | - Ian Underwood
- Institute for Integrated Micro and Nano systems, University of Edinburgh, Scottish Micro Electronic Centre, Alexander Crum Brown Road, EH9 3FF, UK
| | - Stewart Smith
- Institute for Bioengineering, University of Edinburgh, King's Buildings, Colin MacLaurin Road, EH9 3DW, UK
| | - Jorge Del-Pozo
- Easter Bush Pathology, The Royal (Dick) School of Veterinary Studies and The Roslin Institute, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | - Sharon Moss
- Easter Bush Pathology, The Royal (Dick) School of Veterinary Studies and The Roslin Institute, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | - Alexandra Inés Thompson
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Neil C Henderson
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Peter C Hayes
- Hepatology Laboratory, University of Edinburgh, Royal Infirmary of Edinburgh, 49 Little France Crescent EH16 4SB, UK
| | - John N Plevris
- Hepatology Laboratory, University of Edinburgh, Royal Infirmary of Edinburgh, 49 Little France Crescent EH16 4SB, UK
| | - Pierre-Olivier Bagnaninchi
- MRC Centre for Regenerative Medicine, SCRM Building, The University of Edinburgh, Edinburgh BioQuarter, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Leonard J Nelson
- Hepatology Laboratory, University of Edinburgh, Royal Infirmary of Edinburgh, 49 Little France Crescent EH16 4SB, UK
| |
Collapse
|
22
|
Selli C, Erac Y, Tosun M. Effects of cell seeding density on real-time monitoring of anti-proliferative effects of transient gene silencing. ACTA ACUST UNITED AC 2016; 23:20. [PMID: 27981039 PMCID: PMC5133759 DOI: 10.1186/s40709-016-0057-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 11/25/2016] [Indexed: 01/15/2023]
Abstract
Background Real-time cellular analysis systems enable impedance-based label-free and dynamic monitoring of various cellular events such as proliferation. In this study, we describe the effects of initial cell seeding density on the anti-proliferative effects of transient gene silencing monitored via real-time cellular analysis. We monitored the real-time changes in proliferation of Huh7 hepatocellular carcinoma and A7r5 vascular smooth muscle cells with different initial seeding densities following transient receptor potential canonical 1 (TRPC1) silencing using xCELLigence system. Huh7 and A7r5 cells were seeded on E-plate 96 at 10,000, 5000, 1250 and 5000, 2500 cells well−1, respectively, following silencing vector transfection. The inhibitory effects of transient silencing on cell proliferation monitored every 30 min for 72 h. Results TRPC1 silencing did not inhibit the proliferation rates of Huh7 cells at 10,000 cells well−1 seeding density. However, a significant anti-proliferative effect was observed at 1250 cells well−1 density at each time point throughout 72 h. Furthermore, significant inhibitory effects on A7r5 proliferation were observed at both 5000 and 2500 cells well−1 for 72 h. Conclusions Data suggest that the effects of transient silencing on cell proliferation differ depending on the initial cell seeding density. While high seeding densities mask the significant changes in proliferation, the inhibitory effects of silencing become apparent at lower seeding densities as the entry into log phase is delayed. Using the optimal initial seeding density is crucial when studying the effects of transient gene silencing. In addition, the results suggest that TRPC1 may contribute to proliferation and phenotypic switching of vascular smooth muscle cells.
Collapse
Affiliation(s)
- Cigdem Selli
- Applied Bioinformatics of Cancer, Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, The University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR UK ; Department of Pharmacology, Faculty of Pharmacy, Ege University, 35040 Izmir, Turkey
| | - Yasemin Erac
- Department of Pharmacology, Faculty of Pharmacy, Ege University, 35040 Izmir, Turkey
| | - Metiner Tosun
- Department of Pharmacology, Faculty of Pharmacy, Ege University, 35040 Izmir, Turkey ; Faculty of Medicine, Izmir University of Economics, 35330 Izmir, Turkey
| |
Collapse
|
23
|
Richard S, Saric A, Boucher M, Slomianny C, Geffroy F, Mériaux S, Lalatonne Y, Petit PX, Motte L. Antioxidative Theranostic Iron Oxide Nanoparticles toward Brain Tumors Imaging and ROS Production. ACS Chem Biol 2016; 11:2812-2819. [PMID: 27513597 DOI: 10.1021/acschembio.6b00558] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Gliomas are the most common primary brain tumor in humans. To date, the only treatment of care consists of surgical removal of the tumor bulk, irradiation, and chemotherapy, finally resulting in a very poor prognosis due to the lack of efficiency in diagnostics. In this context, nanomedicine combining both diagnostic and magnetic resonance imaging (MRI) and therapeutic applications is a relevant strategy referred to theranostic. Magnetic nanoparticles (NP) are excellent MRI contrast agents because of their large magnetic moment, which induces high transverse relaxivity (r2) characteristic and increased susceptibility effect (T2*). NP can be also used for drug delivery by coating their surface with therapeutic molecules. Preliminary in vitro studies show the high potential of caffeic acid (CA), a natural polyphenol, as a promising anticancer drug due to its antioxidant, anti-inflammatory, and antimetastatic properties. In this study, the antioxidative properties of iron oxide NP functionalized with caffeic acid (γFe2O3@CA NP) are investigated in vitro on U87-MG brain cancer cell lines. After intravenous injection of these NP in mice bearing a U87 glioblastoma, a negative contrast enhancement was specifically observed on 11.7 T MRI images in cancerous tissue, demonstrating a passive targeting of the tumor with these nanoplatforms.
Collapse
Affiliation(s)
- Sophie Richard
- Laboratoire CSPBAT, CNRS UMR 7244 UFR SMBH, Université Paris 13 Sorbonne Paris Cité, F-93017 Bobigny, France
| | - Ana Saric
- Laboratoire de Toxicologie, Pharmacologie et Signalisation Cellulaire,
INSERM U1124, Université Paris-Descartes, Centre Universitaire des Saints-Pères, F-75270 Paris Cedex 06, France
- Division of Molecular Medicine, Rudger Boskivic Institute, Zagreb, Croatia
| | - Marianne Boucher
- Unité d’Imagerie par Résonance
Magnétique et de Spectroscopie, CEA/DRF/I2BM/NeuroSpin, F-91191 Gif-sur-Yvette, France
| | - Christian Slomianny
- Inserm, U100, Laboratoire de Physiologie Cellulaire, Université Lille 1, F-59655 Villeneuve d’Ascq, France
| | - Françoise Geffroy
- Unité d’Imagerie par Résonance
Magnétique et de Spectroscopie, CEA/DRF/I2BM/NeuroSpin, F-91191 Gif-sur-Yvette, France
| | - Sébastien Mériaux
- Unité d’Imagerie par Résonance
Magnétique et de Spectroscopie, CEA/DRF/I2BM/NeuroSpin, F-91191 Gif-sur-Yvette, France
| | - Yoann Lalatonne
- Inserm, U1148, Laboratory for Vascular Translational
Science, UFR SMBH, Université Paris 13, Sorbonne Paris Cité, F-93017 Bobigny, France
- Service de Médecine Nucléaire, Hôpital Avicenne
Assistance Publique-Hôpitaux de Paris, F-93009 Bobigny, France
| | - Patrice X. Petit
- Laboratoire de Toxicologie, Pharmacologie et Signalisation Cellulaire,
INSERM U1124, Université Paris-Descartes, Centre Universitaire des Saints-Pères, F-75270 Paris Cedex 06, France
| | - Laurence Motte
- Inserm, U1148, Laboratory for Vascular Translational
Science, UFR SMBH, Université Paris 13, Sorbonne Paris Cité, F-93017 Bobigny, France
| |
Collapse
|
24
|
Karthivashan G, Masarudin MJ, Kura AU, Abas F, Fakurazi S. Optimization, formulation, and characterization of multiflavonoids-loaded flavanosome by bulk or sequential technique. Int J Nanomedicine 2016; 11:3417-34. [PMID: 27555765 PMCID: PMC4968855 DOI: 10.2147/ijn.s112045] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
This study involves adaptation of bulk or sequential technique to load multiple flavonoids in a single phytosome, which can be termed as "flavonosome". Three widely established and therapeutically valuable flavonoids, such as quercetin (Q), kaempferol (K), and apigenin (A), were quantified in the ethyl acetate fraction of Moringa oleifera leaves extract and were commercially obtained and incorporated in a single flavonosome (QKA-phosphatidylcholine) through four different methods of synthesis - bulk (M1) and serialized (M2) co-sonication and bulk (M3) and sequential (M4) co-loading. The study also established an optimal formulation method based on screening the synthesized flavonosomes with respect to their size, charge, polydispersity index, morphology, drug-carrier interaction, antioxidant potential through in vitro 1,1-diphenyl-2-picrylhydrazyl kinetics, and cytotoxicity evaluation against human hepatoma cell line (HepaRG). Furthermore, entrapment and loading efficiency of flavonoids in the optimal flavonosome have been identified. Among the four synthesis methods, sequential loading technique has been optimized as the best method for the synthesis of QKA-phosphatidylcholine flavonosome, which revealed an average diameter of 375.93±33.61 nm, with a zeta potential of -39.07±3.55 mV, and the entrapment efficiency was >98% for all the flavonoids, whereas the drug-loading capacity of Q, K, and A was 31.63%±0.17%, 34.51%±2.07%, and 31.79%±0.01%, respectively. The in vitro 1,1-diphenyl-2-picrylhydrazyl kinetics of the flavonoids indirectly depicts the release kinetic behavior of the flavonoids from the carrier. The QKA-loaded flavonosome had no indication of toxicity toward human hepatoma cell line as shown by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide result, wherein even at the higher concentration of 200 µg/mL, the flavonosomes exert >85% of cell viability. These results suggest that sequential loading technique may be a promising nanodrug delivery system for loading multiflavonoids in a single entity with sustained activity as an antioxidant, hepatoprotective, and hepatosupplement candidate.
Collapse
Affiliation(s)
| | - Mas Jaffri Masarudin
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences
| | - Aminu Umar Kura
- Laboratory of Vaccines and Immunotherapeutics, Institute of Bioscience
| | - Faridah Abas
- Department of Food Science, Faculty of Food Science and Technology
- Laboratory of Natural Products, Institute of Bioscience
| | - Sharida Fakurazi
- Laboratory of Vaccines and Immunotherapeutics, Institute of Bioscience
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
25
|
Zhou W, Feng X, Han Han, Guo S, Wang G. Synergistic effects of combined treatment with histone deacetylase inhibitor suberoylanilide hydroxamic acid and TRAIL on human breast cancer cells. Sci Rep 2016; 6:28004. [PMID: 27292433 PMCID: PMC4904277 DOI: 10.1038/srep28004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 05/20/2016] [Indexed: 12/20/2022] Open
Abstract
Previous studies showed that either histone deacetylase (HDAC) inhibitors or tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can induce apoptosis in tumor cells including breast cancer. However, the underling mechanisms of combining HDAC inhibitors with TRAIL in the treatment of breast cancer are poorly understood. In this study, we determined the ability of SAHA and TRAIL as single agents or in combination to inhibit the growth and survival of MCF-7 and MDA-MB-231 breast cancer cells. Our results demonstrate that the distinct effects of SAHA or TRAIL individually and in combination on the proliferation, cell viability, apoptosis, cell cycle distribution, and morphological changes of MDA-MB-231 and MCF-7 cells. We further determined the different effects of SAHA or TRAIL alone and combining SAHA with TRAIL on the expression of a number of apoptosis-related molecules, cell cycle, growth factors and their receptors in cancer cells. Our results demonstrated that the combinatorial treatment of SAHA and TRAIL may target multiple pathways and serve as an effective therapeutic strategy against breast cancer. An improved understanding of the molecular mechanisms may facilitate either SAHA or TRAIL targeted use and the selection of suitable combinations.
Collapse
Affiliation(s)
- Weiqiang Zhou
- Key Laboratory of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146 North Huanghe St, Huanggu Dis, Shenyang City, Liaoning Pro 110034, P. R. China
| | - Xiuyan Feng
- Key Laboratory of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146 North Huanghe St, Huanggu Dis, Shenyang City, Liaoning Pro 110034, P. R. China
- The Second Affiliated Hospital of Shenyang Medical College, No. 20 North 9th St, Heping Dis, Shenyang City, Liaoning Pro 110002, P. R. China
| | - Han Han
- Key Laboratory of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146 North Huanghe St, Huanggu Dis, Shenyang City, Liaoning Pro 110034, P. R. China
| | - Shanchun Guo
- RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA 70125, USA
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA
| | - Guangdi Wang
- RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA 70125, USA
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA
| |
Collapse
|
26
|
Novel Method Based on Real-Time Cell Analysis for Drug Susceptibility Testing of Herpes Simplex Virus and Human Cytomegalovirus. J Clin Microbiol 2016; 54:2120-7. [PMID: 27252463 DOI: 10.1128/jcm.03274-15] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 05/26/2016] [Indexed: 01/23/2023] Open
Abstract
The plaque reduction assay (PRA) is the gold standard phenotypic method to determine herpes simplex virus (HSV) and human cytomegalovirus (HCMV) susceptibilities to antiviral drugs. However, this assay is subjective and labor intensive. Here, we describe a novel antiviral phenotypic method based on real-time cell analysis (RTCA) that measures electronic impedance over time. The effective drug concentrations that reduced by 50% (EC50s) the cytopathic effects induced by HSV-1 and HCMV were evaluated by both methods. The EC50s of acyclovir and foscarnet against a reference wild-type (WT) HSV-1 strain in Vero cells were, respectively, 0.5 μM and 32.6 μM by PRA and 0.8 μM and 93.6 μM by RTCA. The EC50 ratios for acyclovir against several HSV-1 thymidine kinase (TK) mutants were 101.8×, 73.4×, 28.8×, and 35.4× (PRA) and 18.0×, 52.0×, 5.5×, and 87.8× (RTCA) compared to those for the WT. The EC50 ratios for acyclovir and foscarnet against the HSV-1 TK/DNA polymerase mutant were 182.8× and 9.7× (PRA) and >125.0× and 10.8× (RTCA) compared to the WT. The EC50s of ganciclovir and foscarnet against WT HCMV strain AD169 in fibroblasts were, respectively, 1.6 μM and 27.8 μM by PRA and 5.0 μM and 111.4 μM by RTCA. The EC50 ratios of ganciclovir against the HCMV UL97 mutant were 3.8× (PRA) and 8.2× (RTCA) compared to those for the WT. The EC50 ratios of ganciclovir and foscarnet against the HCMV UL97/DNA polymerase mutant were 17.1× and 12.1× (PRA) and 14.7× and 4.6× (RTCA) compared to those for the WT. RTCA allows objective drug susceptibility testing of HSV and HCMV and could permit high-throughput screening of new antivirals.
Collapse
|
27
|
A real-time assay for neutrophil chemotaxis. Biotechniques 2016; 60:245-51. [PMID: 27177817 DOI: 10.2144/000114416] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 02/05/2016] [Indexed: 11/23/2022] Open
Abstract
Neutrophils are the predominant cells during acute phases of inflammation, and it is now recognized that these leukocytes play an important role in modulation of the immune response. Directed migration of these cells to the sites of injury, known as chemotaxis, is driven by chemoattractants present at the endothelial cell surface or in the extracellular matrix (ECM). Since uncontrolled or excessive neutrophil chemotaxis is involved in pathological conditions such as atherosclerosis and severe asthma, studying the chemical cues triggering neutrophil migration is essential for understanding the biology of these cells and developing new anti-inflammatory therapies. Although several methods have been developed to evaluate neutrophil chemotaxis, these techniques are generally labor-intensive or alter the native form of these cells and their physiological response. Here we report a rapid, non-invasive, impedance-based, and label-free assay for real-time assessment of neutrophil chemotaxis.
Collapse
|
28
|
Hoang NTM, Phuong TT, Nguyen TTN, Tran YTH, Nguyen ATN, Nguyen TL, Bui KTV. In Vitro Characterization of Derrone as an Aurora Kinase Inhibitor. Biol Pharm Bull 2016; 39:935-45. [PMID: 26983907 DOI: 10.1248/bpb.b15-00835] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Among mitotic kinases, Aurora kinases are the most widely studied, since their expression is restricted to mitosis. They play a key role in chromosome segregation and cell polyploidy. Aurora kinases are important therapeutic targets, and several research groups have directed their efforts toward the identification of kinase inhibitors. The aim of this study is to screen and characterize Aurora kinase inhibitors from natural substances extracted from plants that are used in the Vietnamese pharmacopoeia. We have characterized in vitro Derrone, extracted from Erythrina orientalis L. MURR, as a novel Aurora kinase inhibitor. This compound exhibited an ability to inhibit the phosphorylation of histone H3 at ser10 both in kinase assay and at the cellular level. The compound was more effective against Aurora kinase B, with a lower IC50 value as compared to Aurora A. Moreover, it impaired the mitotic spindle checkpoint and led to endoreduplication in cancer cells, a phenomenon caused by an Aurora B inhibitor. Interestingly, using the xCelligence system and real-time cell analysis (RTCA) software, we set up a comparison of cell proliferation profiles between cancer cells treated with Derrone and VX680-a well-known Aurora kinase inhibitor-and we found that these profiles exhibited considerable similarity in cell morphology, growth, and death. Additionally, Derrone significantly inhibited the formation and growth of MCF7 tumor spheroids.
Collapse
|
29
|
Morales P, Whyte LS, Chicharro R, Gómez-Cañas M, Pazos MR, Goya P, Irving AJ, Fernández-Ruiz J, Ross RA, Jagerovic N. Identification of Novel GPR55 Modulators Using Cell-Impedance-Based Label-Free Technology. J Med Chem 2016; 59:1840-53. [DOI: 10.1021/acs.jmedchem.5b01331] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Paula Morales
- Instituto de Química Médica, CSIC, Calle Juan de la Cierva, 3, 28006 Madrid, Spain
| | - Lauren S. Whyte
- Department
of Pharmacology and Toxicology, Medical Sciences Building, University of Toronto, 1 King’s College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Roberto Chicharro
- Instituto de Química Médica, CSIC, Calle Juan de la Cierva, 3, 28006 Madrid, Spain
| | - María Gómez-Cañas
- Departamento
de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28040 Madrid, Spain
- Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), 28034 Madrid, Spain
| | - M. Ruth Pazos
- Departamento
de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28040 Madrid, Spain
- Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), 28034 Madrid, Spain
| | - Pilar Goya
- Instituto de Química Médica, CSIC, Calle Juan de la Cierva, 3, 28006 Madrid, Spain
| | - Andrew J. Irving
- School of
Biomolecular and Biomedical Science, University College Dublin, Dublin D4, Ireland
| | - Javier Fernández-Ruiz
- Departamento
de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28040 Madrid, Spain
- Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), 28034 Madrid, Spain
| | - Ruth A. Ross
- Department
of Pharmacology and Toxicology, Medical Sciences Building, University of Toronto, 1 King’s College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Nadine Jagerovic
- Instituto de Química Médica, CSIC, Calle Juan de la Cierva, 3, 28006 Madrid, Spain
| |
Collapse
|
30
|
Abstract
The aim of this study was to investigate the toxic effect of the metal salt cadmium chloride dihydrate on the rabbit kidney cell line using the xCELLigence system or real-time cell analyser (RTCA), and to compare this relatively new method with standard biological cytotoxicity assays. This system provides real-time monitoring of cell behaviour and proliferative activity during the whole time of experiment. Moreover, after 24 h exposure of cells to cadmium, colorimetric 3-[4,5-dimethylthiazol-2-yl]-2,5-difenyl tetrazolium bromide (MTT) test was used to measure the metabolic activity and cytotoxicity was determined by measurement of lactate dehydrogenase (LDH) leaked from damaged cells. We found that renal cells exposed to lower concentrations (5–10 mg·l-1) of cadmium tend to grow similarly to control cells, however, cell index was significantly different (P < 0.05) after 24 h. With increasing concentration of cadmium (15–50 mg·l-1) significantly lower proliferative (P < 0.05) and metabolic activity (P < 0.05) of cells was observed and cytotoxicity increased simultaneously (P < 0.001). In addition, we found that the real-time monitoring of the cell response was significantly correlated with commonly used biological methods for toxicity measurement, for MTT assay R2 was 0.9448 (P < 0.01) and for LDH assay R2 was 0.9466 (P < 0.01), respectively. The present study is the first report when combination of RTCA, MTT assay and LDH test was used for cadmium nephrotoxicity assessment. In all these methods, the toxic effect of cadmium on rabbit kidney cells increased in a concentration-dependent manner.
Collapse
|
31
|
Atienzar FA, Novik EI, Gerets HH, Parekh A, Delatour C, Cardenas A, MacDonald J, Yarmush ML, Dhalluin S. Predictivity of dog co-culture model, primary human hepatocytes and HepG2 cells for the detection of hepatotoxic drugs in humans. Toxicol Appl Pharmacol 2013; 275:44-61. [PMID: 24333257 DOI: 10.1016/j.taap.2013.11.022] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 11/20/2013] [Accepted: 11/27/2013] [Indexed: 12/19/2022]
Abstract
Drug induced liver injury (DILI) is a major cause of attrition during early and late stage drug development. Consequently, there is a need to develop better in vitro primary hepatocyte models from different species for predicting hepatotoxicity in both animals and humans early in drug development. Dog is often chosen as the non-rodent species for toxicology studies. Unfortunately, dog in vitro models allowing long term cultures are not available. The objective of the present manuscript is to describe the development of a co-culture dog model for predicting hepatotoxic drugs in humans and to compare the predictivity of the canine model along with primary human hepatocytes and HepG2 cells. After rigorous optimization, the dog co-culture model displayed metabolic capacities that were maintained up to 2 weeks which indicates that such model could be also used for long term metabolism studies. Most of the human hepatotoxic drugs were detected with a sensitivity of approximately 80% (n=40) for the three cellular models. Nevertheless, the specificity was low approximately 40% for the HepG2 cells and hepatocytes compared to 72.7% for the canine model (n=11). Furthermore, the dog co-culture model showed a higher superiority for the classification of 5 pairs of close structural analogs with different DILI concerns in comparison to both human cellular models. Finally, the reproducibility of the canine system was also satisfactory with a coefficient of correlation of 75.2% (n=14). Overall, the present manuscript indicates that the dog co-culture model may represent a relevant tool to perform chronic hepatotoxicity and metabolism studies.
Collapse
Affiliation(s)
- Franck A Atienzar
- UCB Pharma SA, Non-Clinical Development, Chemin du Foriest, 1420 Braine-l'Alleud, Belgium.
| | - Eric I Novik
- Hμrel Corporation, 675 U.S. Highway 1, North Brunswick, NJ 08902, USA
| | - Helga H Gerets
- UCB Pharma SA, Non-Clinical Development, Chemin du Foriest, 1420 Braine-l'Alleud, Belgium
| | - Amit Parekh
- Hμrel Corporation, 675 U.S. Highway 1, North Brunswick, NJ 08902, USA
| | - Claude Delatour
- UCB Pharma SA, Non-Clinical Development, Chemin du Foriest, 1420 Braine-l'Alleud, Belgium
| | - Alvaro Cardenas
- UCB Pharma SA, Non-Clinical Development, Chemin du Foriest, 1420 Braine-l'Alleud, Belgium
| | - James MacDonald
- Chrysalis Pharma Consulting, LLC, 385 Route 24, Suite 1G, Chester, NJ 07930, USA
| | - Martin L Yarmush
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA
| | - Stéphane Dhalluin
- UCB Pharma SA, Non-Clinical Development, Chemin du Foriest, 1420 Braine-l'Alleud, Belgium
| |
Collapse
|