1
|
Flörkemeier I, Antons LK, Weimer JP, Hedemann N, Rogmans C, Krüger S, Scherließ R, Dempfle A, Arnold N, Maass N, Bauerschlag DO. Multicellular ovarian cancer spheroids: novel 3D model to mimic tumour complexity. Sci Rep 2024; 14:23526. [PMID: 39384844 PMCID: PMC11464915 DOI: 10.1038/s41598-024-73680-6] [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: 06/13/2024] [Accepted: 09/19/2024] [Indexed: 10/11/2024] Open
Abstract
In vitro, spheroid models have become well established in cancer research because they can better mimic certain characteristics of in vivo tumours. However, interaction with the tumour microenvironment, such as cancer-associated fibroblasts, plays a key role in tumour progression. We initially focused on the interaction of tumour cells with fibroblasts. To model this interaction, we developed a spheroid model of ovarian cancer and fibroblasts. To this end, ovarian cancer cell lines and ex vivo primary cells were simultaneously and sequentially seeded with fibroblasts in a scaffold-free system at different ratios and subsequently characterized with respect to changes in morphology, proliferation, and viability. We demonstrated that co-cultures are able to form by far more compact spheroids, especially in cells that form aggregates in mono-culture. In addition, the co-cultures were able to increase proliferation and sensitivity to cisplatin. Simultaneous seeding led fibroblasts invade the core in both cell lines and primary cells. These results show differences in formation, firmness, and size between co-culture and mono-culture. Our model is designed to better represent and characterize the mutual influencing factors of fibroblasts and tumour cells. Fibroblast-supplemented multicellular spheroids are a valuable tool for tumour microenvironment interaction and new drug discovery.
Collapse
Affiliation(s)
- Inken Flörkemeier
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany.
- KiNSIS Priority Research Area, Kiel University, Kiel, Germany.
| | - Lisa K Antons
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Jörg P Weimer
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Nina Hedemann
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Christoph Rogmans
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Sandra Krüger
- Department of Pathology, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Regina Scherließ
- Department of Pharmaceutics and Biopharmaceutics, Kiel University, Kiel, Germany
- KiNSIS Priority Research Area, Kiel University, Kiel, Germany
| | - Astrid Dempfle
- Institute of Medical Informatics and Statistics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Norbert Arnold
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Nicolai Maass
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Dirk O Bauerschlag
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
- Department of Gynaecology, Jena University Hospital, Jena, Germany
| |
Collapse
|
2
|
Sahu P, Camarillo IG, Dettin M, Zamuner A, Teresa Conconi M, Barozzi M, Giri P, Sundararajan R, Sieni E. Electroporation enhances cell death in 3D scaffold-based MDA-MB-231 cells treated with metformin. Bioelectrochemistry 2024; 159:108734. [PMID: 38762949 DOI: 10.1016/j.bioelechem.2024.108734] [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: 03/22/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 05/21/2024]
Abstract
Triple-negative breast cancer (TNBC), the most aggressive subtype of breast cancer lacks estrogen, progesterone, and HER2 receptors and hence, is therapeutically challenging. Towards this, we studied an alternate therapy by repurposing metformin (FDA-approved type-2 diabetic drug with anticancer properties) in a 3D-scaffold culture, with electrical pulses. 3D cell culture was used to simulate the tumor microenvironment more closely and MDA-MB-231, human TNBC cells, treated with both 5 mM metformin (Met) and 8 electrical pulses at 2500 V/cm, 10 µs (EP1) and 800 V/cm, 100 µs (EP2) at 1 Hz were studied in 3D and 2D. They were characterized using cell viability, reactive oxygen species (ROS), glucose uptake, and lactate production assays at 24 h. Cell viability, as low as 20 % was obtained with EP1 + 5 mM Met. They exhibited 1.65-fold lower cell viability than 2D with EP1 + 5 mM Met. ROS levels indicated a 2-fold increase in oxidative stress for EP1 + 5 mM Met, while the glucose uptake was limited to only 9 %. No significant change in the lactate production indicated glycolytic arrest and a non-conducive environment for MDA-MB-231 growth. Our results indicate that 3D cell culture, with a more realistic tumor environment that enhances cell death using metformin and electrical pulses could be a promising approach for TNBC therapeutic intervention studies.
Collapse
Affiliation(s)
- Praveen Sahu
- School of Engineering Technology, Purdue University, West Lafayette, IN 47907, USA
| | - Ignacio G Camarillo
- Deptartment of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; Purdue University Center for Cancer Research, West Lafayette, IN 47907, USA
| | - Monica Dettin
- Department of Industrial Engineering, University of Padova, Padova 35122, Italy
| | - Annj Zamuner
- Department of Industrial Engineering, University of Padova, Padova 35122, Italy; Department of Civil, Environmental, and Architectural Engineering, University of Padova, Italy
| | - Maria Teresa Conconi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova 35131, Italy
| | - Marco Barozzi
- Department of Theoretical and Applied Sciences, University of Insubria, Varese 21100, Italy
| | - Pragatheiswar Giri
- School of Engineering Technology, Purdue University, West Lafayette, IN 47907, USA
| | - Raji Sundararajan
- School of Engineering Technology, Purdue University, West Lafayette, IN 47907, USA
| | - Elisabetta Sieni
- Department of Theoretical and Applied Sciences, University of Insubria, Varese 21100, Italy.
| |
Collapse
|
3
|
Feng Y, Che B, Fu J, Sun Y, Ma W, Tian J, Dai L, Jing G, Zhao W, Sun D, Zhang C. From Chips-in-Lab to Point-of-Care Live Cell Device: Development of a Microfluidic Device for On-Site Cell Culture and High-Throughput Drug Screening. ACS Biomater Sci Eng 2024; 10:5399-5408. [PMID: 39031055 DOI: 10.1021/acsbiomaterials.4c00766] [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] [Indexed: 07/22/2024]
Abstract
Live cell assays provide real-time data of cellular responses. In combination with microfluidics, applications such as automated and high-throughput drug screening on live cells can be accomplished in small devices. However, their application in point-of-care testing (POCT) is limited by the requirement for bulky equipment to maintain optimal cell culture conditions. In this study, we propose a POCT device that allows on-site cell culture and high-throughput drug screening on live cells. We first observe that cell viabilities are substantially affected by liquid evaporation within the microfluidic device, which is intrinsic to the polydimethylsiloxane (PDMS) material due to its hydrophobic nature and nanopatterned surface. The unwanted PDMS-liquid-air interface in the cell culture environment can be eliminated by maintaining a persistent humidity of 95-100% or submerging the whole microfluidic device under water. Our results demonstrate that in the POCT device equipped with a water tank, both primary cells and cell lines can be maintained for up to 1 week without the need for external cell culture equipment. Moreover, this device is powered by a standard alkali battery and can automatically screen over 5000 combinatorial drug conditions for regulating neural stem cell differentiation. By monitoring dynamic variations in fluorescent markers, we determine the optimal doses of platelet-derived growth factor and epidermal growth factor to suppress proinflammatory S100A9-induced neuronal toxicities. Overall, this study presents an opportunity to transform lab-on-a-chip technology from a laboratory-based approach to actual point-of-care devices capable of performing complex experimental procedures on-site and offers significant advancements in the fields of personalized medicine and rapid clinical diagnostics.
Collapse
Affiliation(s)
- Yibo Feng
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, No. 1, Xuefu Avenue, Xi'an 710127, Shaanxi, China
| | - Bingchen Che
- School of Physics, Northwest University, No. 1 Xuefu Avenue, Xi'an 710127, Shaanxi, China
| | - Jiahao Fu
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, No. 1, Xuefu Avenue, Xi'an 710127, Shaanxi, China
| | - Yu Sun
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an 710127, China
| | - Wenju Ma
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, No. 1, Xuefu Avenue, Xi'an 710127, Shaanxi, China
| | - Jing Tian
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an 710127, China
- Center for Automated and Innovative Drug Discovery, Northwest University, No. 1, Xuefu Avenue, Xi'an 710127, Shaanxi, China
| | - Liang Dai
- Department of Physics, City University of Hong Kong, Hong Kong 999077, China
| | - Guangyin Jing
- School of Physics, Northwest University, No. 1 Xuefu Avenue, Xi'an 710127, Shaanxi, China
| | - Wei Zhao
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, No. 1, Xuefu Avenue, Xi'an 710127, Shaanxi, China
| | - Dan Sun
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, No. 1, Xuefu Avenue, Xi'an 710127, Shaanxi, China
- Center for Automated and Innovative Drug Discovery, Northwest University, No. 1, Xuefu Avenue, Xi'an 710127, Shaanxi, China
| | - Ce Zhang
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, No. 1, Xuefu Avenue, Xi'an 710127, Shaanxi, China
| |
Collapse
|
4
|
Leippe D, Choy R, Vidugiris G, Merritt H, Mellem KT, Beattie DT, Ullman JC, Vidugiriene J. Bioluminescent Assay for the Quantification of Cellular Glycogen Levels. ACS OMEGA 2024; 9:33072-33080. [PMID: 39100309 PMCID: PMC11292620 DOI: 10.1021/acsomega.4c04190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/19/2024] [Accepted: 07/08/2024] [Indexed: 08/06/2024]
Abstract
Glycogen is a large polymer of glucose that functions as an important means of storing energy and maintaining glucose homeostasis. Glycogen synthesis and degradation pathways are highly regulated and their dysregulation can contribute to disease. Glycogen storage diseases are a set of disorders that arise from improper glycogen metabolism. Glycogen storage disease II, known as Pompe disease, is caused by a genetic mutation that leads to increased glycogen storage in cells and tissues, resulting in progressive muscle atrophy and respiratory decline for patients. One approach for treating Pompe disease is to reduce glycogen levels by interfering with the glycogen synthesis pathway through glycogen synthase inhibitors. To facilitate the study of glycogen synthase inhibitors in biological samples, such as cultured cells, a high-throughput approach for measuring cellular glycogen was developed. A bioluminescent glycogen detection assay was automated and used to measure the glycogen content in cells grown in 384-well plates. The assay successfully quantified reduced glycogen stores in cells treated with a series of glycogen synthase 1 inhibitors, validating the utility of the assay for drug screening efforts and demonstrating its value for therapy development and glycogen metabolism research.
Collapse
Affiliation(s)
- Donna Leippe
- Research
and Development, Promega Corporation, Madison, Wisconsin 53711, United States
| | - Rebeca Choy
- Maze
Therapeutics, South
San Francisco, California 94080, United States
| | - Gediminas Vidugiris
- Research
and Development, Promega Corporation, Madison, Wisconsin 53711, United States
| | - Hanne Merritt
- Maze
Therapeutics, South
San Francisco, California 94080, United States
| | - Kevin T. Mellem
- Maze
Therapeutics, South
San Francisco, California 94080, United States
| | - David T. Beattie
- Maze
Therapeutics, South
San Francisco, California 94080, United States
| | - Julie C. Ullman
- Maze
Therapeutics, South
San Francisco, California 94080, United States
| | - Jolanta Vidugiriene
- Research
and Development, Promega Corporation, Madison, Wisconsin 53711, United States
| |
Collapse
|
5
|
Hamar J, Cnaani A, Kültz D. Effects of CRISPR/Cas9 targeting of the myo-inositol biosynthesis pathway on hyper-osmotic tolerance of tilapia cells. Genomics 2024; 116:110833. [PMID: 38518899 DOI: 10.1016/j.ygeno.2024.110833] [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: 12/13/2023] [Revised: 03/05/2024] [Accepted: 03/19/2024] [Indexed: 03/24/2024]
Abstract
Myo-inositol is an important compatible osmolyte in vertebrates. This osmolyte is produced by the myo-inositol biosynthesis (MIB) pathway composed of myo-inositol phosphate synthase and inositol monophosphatase. These enzymes are among the highest upregulated proteins in tissues and cell cultures from teleost fish exposed to hyperosmotic conditions indicating high importance of this pathway for tolerating this type of stress. CRISPR/Cas9 gene editing of tilapia cells produced knockout lines of MIB enzymes and control genes. Metabolic activity decreased significantly for MIB KO lines in hyperosmotic media. Trends of faster growth of the MIB knockout lines in isosmotic media and faster decline of MIB knockout lines in hyperosmotic media were also observed. These results indicate a decline in metabolic fitness but only moderate effects on cell survival when tilapia cells with disrupted MIB genes are exposed to hyperosmolality. Therefore MIB genes are required for full osmotolerance of tilapia cells.
Collapse
Affiliation(s)
- Jens Hamar
- Department of Animal Sciences & Genome Center, University of California Davis, Meyer Hall, One Shields Avenue, Davis, CA 95616, USA
| | - Avner Cnaani
- Department of Poultry and Aquaculture, Institute of Animal Sciences, Agricultural Research Organization, Volcani Center, P.O. Box 15159, Rishon LeZion 7528809, Israel
| | - Dietmar Kültz
- Department of Animal Sciences & Genome Center, University of California Davis, Meyer Hall, One Shields Avenue, Davis, CA 95616, USA.
| |
Collapse
|
6
|
Dougherty BV, Moore CJ, Rawls KD, Jenior ML, Chun B, Nagdas S, Saucerman JJ, Kolling GL, Wallqvist A, Papin JA. Identifying metabolic adaptations characteristic of cardiotoxicity using paired transcriptomics and metabolomics data integrated with a computational model of heart metabolism. PLoS Comput Biol 2024; 20:e1011919. [PMID: 38422168 PMCID: PMC10931521 DOI: 10.1371/journal.pcbi.1011919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/12/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024] Open
Abstract
Improvements in the diagnosis and treatment of cancer have revealed long-term side effects of chemotherapeutics, particularly cardiotoxicity. Here, we present paired transcriptomics and metabolomics data characterizing in vitro cardiotoxicity to three compounds: 5-fluorouracil, acetaminophen, and doxorubicin. Standard gene enrichment and metabolomics approaches identify some commonly affected pathways and metabolites but are not able to readily identify metabolic adaptations in response to cardiotoxicity. The paired data was integrated with a genome-scale metabolic network reconstruction of the heart to identify shifted metabolic functions, unique metabolic reactions, and changes in flux in metabolic reactions in response to these compounds. Using this approach, we confirm previously seen changes in the p53 pathway by doxorubicin and RNA synthesis by 5-fluorouracil, we find evidence for an increase in phospholipid metabolism in response to acetaminophen, and we see a shift in central carbon metabolism suggesting an increase in metabolic demand after treatment with doxorubicin and 5-fluorouracil.
Collapse
Affiliation(s)
- Bonnie V. Dougherty
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Connor J. Moore
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Kristopher D. Rawls
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Matthew L. Jenior
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Bryan Chun
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Sarbajeet Nagdas
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, Virginia, United States of America
| | - Jeffrey J. Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Glynis L. Kolling
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, United States of America
| | - Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Maryland, United States of America
| | - Jason A. Papin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
7
|
Orfanoudaki M, Smith EA, Hill NT, Garman KA, Brownell I, Copp BR, Grkovic T, Henrich CJ. An Investigation of Structure-Activity Relationships and Cell Death Mechanisms of the Marine Alkaloids Discorhabdins in Merkel Cell Carcinoma Cells. Mar Drugs 2023; 21:474. [PMID: 37755087 PMCID: PMC10532587 DOI: 10.3390/md21090474] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/28/2023] Open
Abstract
A library of naturally occurring and semi-synthetic discorhabdins was assessed for their effects on Merkel cell carcinoma (MCC) cell viability. The set included five new natural products and semi-synthetic compounds whose structures were elucidated with NMR, HRMS, and ECD techniques. Several discorhabdins averaged sub-micromolar potency against the MCC cell lines tested and most of the active compounds showed selectivity towards virus-positive MCC cell lines. An investigation of structure-activity relationships resulted in an expanded understanding of the crucial structural features of the discorhabdin scaffold. Mechanistic cell death assays suggested that discorhabdins, unlike many other MCC-active small molecules, do not induce apoptosis, as shown by the lack of caspase activation, annexin V staining, and response to caspase inhibition. Similarly, discorhabdin treatment failed to increase MCC intracellular calcium and ROS levels. In contrast, the rapid loss of cellular reducing potential and mitochondrial membrane potential suggested that discorhabdins induce mitochondrial dysfunction leading to non-apoptotic cell death.
Collapse
Affiliation(s)
- Maria Orfanoudaki
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; (M.O.); (E.A.S.)
| | - Emily A. Smith
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; (M.O.); (E.A.S.)
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Natasha T. Hill
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD 20891, USA; (N.T.H.); (K.A.G.); (I.B.)
| | - Khalid A. Garman
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD 20891, USA; (N.T.H.); (K.A.G.); (I.B.)
| | - Isaac Brownell
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD 20891, USA; (N.T.H.); (K.A.G.); (I.B.)
| | - Brent R. Copp
- School of Chemical Sciences, University of Auckland, Auckland 1142, New Zealand;
| | - Tanja Grkovic
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; (M.O.); (E.A.S.)
- Natural Products Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD 21702, USA
| | - Curtis J. Henrich
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; (M.O.); (E.A.S.)
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| |
Collapse
|
8
|
A dual-functional module cellular electrochemical sensing platform for simultaneous detection guanine and xanthine. Biosens Bioelectron 2023; 226:115104. [PMID: 36764126 DOI: 10.1016/j.bios.2023.115104] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/30/2022] [Accepted: 01/24/2023] [Indexed: 01/27/2023]
Abstract
The separation of the superimposed electrochemical signals of intracellular guanine (G) and xanthine (X) is difficult, which is great obstacle to the application of cell electrochemistry. In this paper, independent functional modules, G-functional module (G-FM) and X-functional module (X-FM), were constructed by molecular imprinting technology for sensitive detection of G and X without mutual interference, then integrated in dual-functional module cellular electrochemical sensing platform (DMCEP) as signal sensing units. DMCEP transmitted signals of G and X in cells synchronously to two windows by two signal sensing channels, and achieved the separation of superimposed signals of G and X in cells. DMCEP exhibited satisfactory reproducibility with relative standard deviation (RSD) of 3.10 and 2.22 %, repeatability with RSD of 3.72 and 3.05 % for G and X detection, and detection limit 0.05 μΜ for G and 0.06 μΜ for X. Good linear relationships between cell concentrations and the signals of G and X on DMCEP were shown in range of 0.75-85 × 106 and 3-85 × 106 cells/mL, respectively. The growth of MCF-7 cells was tracked by DMCEP, and showed consistent trend with the cell counting method, while the change of cell viability from lag to logarithmic phase captured by DMCEP was earlier than that of cell counting method. This strategy provided the foundation for the establishment of the cell viability electrochemical detection method, and new insights into the simultaneous recording of other analyses with superimposed peak positions and the simultaneous tracking of multiple biomarkers.
Collapse
|
9
|
Serioli L, Gruzinskyte L, Zappalà G, Hwu ET, Laksafoss TZ, Jensen PL, Demarchi D, Müllertz A, Boisen A, Zór K. Moving perfusion culture and live-cell imaging from lab to disc: proof of concept toxicity assay with AI-based image analysis. LAB ON A CHIP 2023; 23:1603-1612. [PMID: 36790123 DOI: 10.1039/d2lc00984f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
In vitro, cell-based assays are essential in diagnostics and drug development. There are ongoing efforts to establish new technologies that enable real-time detection of cell-drug interaction during culture under flow conditions. Our compact (10 × 10 × 8.5 cm) cell culture and microscope on disc (CMoD) platform aims to decrease the application barriers of existing lab-on-a-chip (LoC) approaches. For the first time in a centrifugal device, (i) cells were cultured for up to six days while a spindle motor facilitated culture medium perfusion, and (ii) an onboard microscope enabled live bright-field imaging of cells while the data wirelessly transmitted to a computer. The quantification of cells from the acquired images was done using artificial intelligence (AI) software. After optimization, the obtained cell viability data from the AI-based image analysis proved to correlate well with data collected from commonly used image analysis software. The CMoD was also suitable for conducting a proof-of-concept toxicity assay with HeLa cells under continuous flow. The half-maximal inhibitory time (IT50) for various concentrations of doxorubicin (DOX) in the case of HeLa cells in flow, was shown to be lower than the IT50 obtained from a static cytotoxicity assay, indicating a faster onset of cell death in flow. The CMoD proved to be easy to handle, enabled cell culture and monitoring without assistance, and is a promising tool for examining the dynamic processes of cells in real-time assays.
Collapse
Affiliation(s)
- Laura Serioli
- The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Denmark.
- BioInnovation Institute Foundation, Copenhagen N 2800, Denmark
| | - Lina Gruzinskyte
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Denmark.
| | - Giulia Zappalà
- The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Denmark.
- Department of Electronics and Telecommunications, Politecnico di Torino, Italy
| | - En Te Hwu
- The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Denmark.
| | - Trygvi Zachariassen Laksafoss
- The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Denmark.
| | | | - Danilo Demarchi
- Department of Electronics and Telecommunications, Politecnico di Torino, Italy
| | - Anette Müllertz
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Denmark.
| | - Anja Boisen
- The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Denmark.
- BioInnovation Institute Foundation, Copenhagen N 2800, Denmark
| | - Kinga Zór
- The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Denmark.
- BioInnovation Institute Foundation, Copenhagen N 2800, Denmark
| |
Collapse
|
10
|
Baljinnyam B, Ronzetti M, Simeonov A. Advances in luminescence-based technologies for drug discovery. Expert Opin Drug Discov 2023; 18:25-35. [PMID: 36562206 PMCID: PMC9892298 DOI: 10.1080/17460441.2023.2160441] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Luminescence-based technologies, specifically bioluminescence and chemiluminescence, are powerful tools with extensive use in drug discovery. Production of light during chemiluminescence and bioluminescence, unlike fluorescence, doesn't require an excitation light source, resulting in high signal-to-noise ratio, less background interference, and no issues from phototoxicity and photobleaching. These characteristics of luminescence technologies offer unique advantages for experimental designs, allowing for greater flexibility to target a wide range of proteins and biological processes for drug discovery at different stages. AREAS COVERED This review provides a basic overview of luciferase-based technologies and details recent advances and use cases of luciferase and luciferin variations and their applicability in the drug discovery toolset. The authors expand upon specific applications of luciferase technologies, including chemiluminescent and bioluminescent-based microscopy. Finally, the authors lay out forward-looking statements on the field of luminescence and how it may shape the translational scientists' work moving forward. EXPERT OPINION The demand for improved luciferase and luciferin pairs correlates strongly with efforts to improve the sensitivity and robustness of high-throughput assays. As luminescent reporter systems improve, so will the expansion of use cases for luminescence-based technologies in early-stage drug discovery. With the synthesis of novel, non-enzymatic chemiluminescence-based probes, which previously were restrained to only basic research applications, they may now be readily implemented in drug discovery campaigns.
Collapse
Affiliation(s)
- Bolormaa Baljinnyam
- Staff Scientist, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Michael Ronzetti
- Predoctoral IRTA Fellow, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Anton Simeonov
- Group Leader, Scientific Director, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
11
|
Wandishin CM, Robbins CJ, Tyson DR, Harris LA, Quaranta V. Real-time luminescence enables continuous drug-response analysis in adherent and suspension cell lines. Cancer Biol Ther 2022; 23:358-368. [PMID: 35443861 PMCID: PMC9037430 DOI: 10.1080/15384047.2022.2065182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/30/2022] [Accepted: 04/04/2022] [Indexed: 11/11/2022] Open
Abstract
The drug-induced proliferation (DIP) rate is a metric of in vitro drug response that avoids inherent biases in commonly used metrics such as 72 h viability. However, DIP rate measurements rely on direct cell counting over time, a laborious task that is subject to numerous challenges, including the need to fluorescently label cells and automatically segment nuclei. Moreover, it is incredibly difficult to directly count cells and accurately measure DIP rates for cell populations in suspension. As an alternative, we use real-time luminescence measurements derived from the cellular activity of NAD(P)H oxidoreductase to efficiently estimate drug response in both adherent and suspension cell populations to a panel of known anticancer agents. For the adherent cell lines, we collect both luminescence reads and direct cell counts over time simultaneously to assess their congruency. Our results demonstrate that the proposed approach significantly speeds up data collection, avoids the need for cellular labels and image segmentation, and opens the door to significant advances in high-throughput screening of anticancer drugs.
Collapse
Affiliation(s)
| | - Charles John Robbins
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TNUSA
| | - Darren R. Tyson
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TNUSA
| | - Leonard A. Harris
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, ARUSA
| | - Vito Quaranta
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TNUSA
| |
Collapse
|
12
|
Current methodologies for the assessment of deep eutectic systems toxicology: Challenges and perspectives. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.119675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
13
|
Reiterer M, Eakin A, Johnson RS, Branco CM. Hyperoxia Reprogrammes Microvascular Endothelial Cell Response to Hypoxia in an Organ-Specific Manner. Cells 2022; 11:cells11162469. [PMID: 36010546 PMCID: PMC9406746 DOI: 10.3390/cells11162469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 11/16/2022] Open
Abstract
Organ function relies on microvascular networks to maintain homeostatic equilibrium, which varies widely in different organs and during different physiological challenges. The endothelium role in this critical process can only be evaluated in physiologically relevant contexts. Comparing the responses to oxygen flux in primary murine microvascular EC (MVEC) obtained from brain and lung tissue reveals that supra-physiological oxygen tensions can compromise MVEC viability. Brain MVEC lose mitochondrial activity and undergo significant alterations in electron transport chain (ETC) composition when cultured under standard, non-physiological atmospheric oxygen levels. While glycolytic capacity of both lung and brain MVEC are unchanged by environmental oxygen, the ability to trigger a metabolic shift when oxygen levels drop is greatly compromised following exposure to hyperoxia. This is particularly striking in MVEC from the brain. This work demonstrates that the unique metabolism and function of organ-specific MVEC (1) can be reprogrammed by external oxygen, (2) that this reprogramming can compromise MVEC survival and, importantly, (3) that ex vivo modelling of endothelial function is significantly affected by culture conditions. It further demonstrates that physiological, metabolic and functional studies performed in non-physiological environments do not represent cell function in situ, and this has serious implications in the interpretation of cell-based pre-clinical models.
Collapse
Affiliation(s)
- Moritz Reiterer
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast BT9 7AE, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Amanda Eakin
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast BT9 7AE, UK
| | - Randall S. Johnson
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Cristina M. Branco
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast BT9 7AE, UK
- Correspondence:
| |
Collapse
|
14
|
Al-madani H, Du H, Yao J, Peng H, Yao C, Jiang B, Wu A, Yang F. Living Sample Viability Measurement Methods from Traditional Assays to Nanomotion. BIOSENSORS 2022; 12:453. [PMID: 35884256 PMCID: PMC9313330 DOI: 10.3390/bios12070453] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 12/18/2022]
Abstract
Living sample viability measurement is an extremely common process in medical, pharmaceutical, and biological fields, especially drug pharmacology and toxicology detection. Nowadays, there are a number of chemical, optical, and mechanical methods that have been developed in response to the growing demand for simple, rapid, accurate, and reliable real-time living sample viability assessment. In parallel, the development trend of viability measurement methods (VMMs) has increasingly shifted from traditional assays towards the innovative atomic force microscope (AFM) oscillating sensor method (referred to as nanomotion), which takes advantage of the adhesion of living samples to an oscillating surface. Herein, we provide a comprehensive review of the common VMMs, laying emphasis on their benefits and drawbacks, as well as evaluating the potential utility of VMMs. In addition, we discuss the nanomotion technique, focusing on its applications, sample attachment protocols, and result display methods. Furthermore, the challenges and future perspectives on nanomotion are commented on, mainly emphasizing scientific restrictions and development orientations.
Collapse
Affiliation(s)
- Hamzah Al-madani
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS), Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China; (H.A.-m.); (H.D.); (J.Y.); (H.P.); (C.Y.); (B.J.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hui Du
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS), Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China; (H.A.-m.); (H.D.); (J.Y.); (H.P.); (C.Y.); (B.J.)
- College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junlie Yao
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS), Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China; (H.A.-m.); (H.D.); (J.Y.); (H.P.); (C.Y.); (B.J.)
- College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hao Peng
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS), Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China; (H.A.-m.); (H.D.); (J.Y.); (H.P.); (C.Y.); (B.J.)
- College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chenyang Yao
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS), Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China; (H.A.-m.); (H.D.); (J.Y.); (H.P.); (C.Y.); (B.J.)
- College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bo Jiang
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS), Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China; (H.A.-m.); (H.D.); (J.Y.); (H.P.); (C.Y.); (B.J.)
| | - Aiguo Wu
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS), Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China; (H.A.-m.); (H.D.); (J.Y.); (H.P.); (C.Y.); (B.J.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, China
| | - Fang Yang
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS), Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China; (H.A.-m.); (H.D.); (J.Y.); (H.P.); (C.Y.); (B.J.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, China
| |
Collapse
|
15
|
Cromwell EF, Sirenko O, Nikolov E, Hammer M, Brock CK, Matossian MD, Alzoubi MS, Collins-Burow BM, Burow ME. Multifunctional profiling of triple-negative breast cancer patient-derived tumoroids for disease modeling. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2022; 27:191-200. [PMID: 35124274 DOI: 10.1016/j.slasd.2022.01.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
3D cell models derived from patient tumors are highly translational tools that can recapitulate the complex genetic and molecular compositions of solid cancers and accelerate identification of drug targets and drug testing. However, the complexity of performing assays with such models remains a hurdle for their wider adoption. In the present study, we describe methods for processing and multi-functional profiling of tumoroid samples to test compound effects using a novel flowchip system in combination with high content imaging and metabolite analysis. Tumoroids were formed from primary cells isolated from a patient-derived tumor explant, TU-BcX-4IC, that represents metaplastic breast cancer with a triple-negative breast cancer subtype. Assays were performed in a microfluidics-based device (Pu⋅MA System) that allows automated exchange of media and treatments of tumoroids in a tissue culture incubator environment. Multi-functional assay profiling was performed on tumoroids treated with anti-cancer drugs. High-content imaging was used to evaluate drug effects on cell viability and expression of E-cadherin and CD44. Lactate secretion was used to measure tumoroid metabolism as a function of time and drug concentration. Observed responses included loss of cell viability, decrease in E-cadherin expression, and increase of lactate production. Importantly, the tumoroids were sensitive to romidepsin and trametinib, while showed significantly reduced sensitivity to paclitaxel and cytarabine, consistent with the primary tumor response. These methods for multi-parametric profiling of drug effects in patient-derived tumoroids provide an in depth understanding of drug sensitivity of individual tumor types, with important implications for the future development of personalized medicine.
Collapse
|
16
|
Sung HW, Choi SE, Chu CH, Ouyang M, Kalyan S, Scott N, Hur SC. Sensitizing drug-resistant cancer cells from blood using microfluidic electroporator. PLoS One 2022; 17:e0264907. [PMID: 35259174 PMCID: PMC8903260 DOI: 10.1371/journal.pone.0264907] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 02/21/2022] [Indexed: 02/06/2023] Open
Abstract
Direct assessment of patient samples holds unprecedented potential in the treatment of cancer. Circulating tumor cells (CTCs) in liquid biopsies are a rapidly evolving source of primary cells in the clinic and are ideal candidates for functional assays to uncover real-time tumor information in real-time. However, a lack of routines allowing direct and active interrogation of CTCs directly from liquid biopsy samples represents a bottleneck for the translational use of liquid biopsies in clinical settings. To address this, we present a workflow for using a microfluidic vortex-assisted electroporation system designed for the functional assessment of CTCs purified from blood. Validation of this approach was assessed through drug response assays on wild-type (HCC827 wt) and gefitinib-resistant (HCC827 GR6) non-small cell lung cancer (NSCLC) cells. HCC827 cells trapped within microscale vortices were electroporated to sequentially deliver drug agents into the cytosol. Electroporation conditions facilitating multi-agent delivery were characterized for both cell lines using an automatic single-cell image fluorescence intensity algorithm. HCC827 GR6 cells spiked into the blood to emulate drug-resistant CTCs were able to be collected with high purity, demonstrating the ability of the device to minimize background cell impact for downstream sensitive cell assays. Using our proposed workflow, drug agent combinations to restore gefitinib sensitivity reflected the anticipated cytotoxic response. Taken together, these results represent a microfluidics multi-drug screening panel workflow that can enable functional interrogation of patient CTCs in situ, thereby accelerating the clinical standardization of liquid biopsies.
Collapse
Affiliation(s)
- Hyun Woo Sung
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Sung-Eun Choi
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Chris H. Chu
- Department of Internal Medicine, Virginia Mason Medical Center, Seattle, Washington, United States of America
| | - Mengxing Ouyang
- Department of Bioengineering, University of California, Los Angeles, California, United States of America
| | - Srivathsan Kalyan
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Nathan Scott
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Soojung Claire Hur
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland, United States of America
- Institute of NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
17
|
Damodarasamy M, Khaing ZZ, Hyde J, Keene CD, Bentov I, Banks WA, Reed MJ. Viable human brain microvessels for the study of aging and neurodegenerative diseases. Microvasc Res 2022; 140:104282. [PMID: 34813858 PMCID: PMC8846932 DOI: 10.1016/j.mvr.2021.104282] [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: 06/09/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 01/28/2023]
Abstract
The brain microvasculature is altered in normal aging and in the presence of disease processes, such as neurodegeneration or ischemia, but there are few methods for studying living tissues. We now report that viable microvessels (MV) are readily isolated from brain tissue of subjects enrolled in studies of neurodegenerative diseases who undergo rapid autopsy (performed with <12 h postmortem interval - PMI). We find that these MV retain their morphology and cellular components and are fairly uniform in size. Sufficient MV (~3-5000) are obtained from 3 to 4 g of tissue to allow for studies of cellular composition as well as extracellular matrix (ECM). Using live/dead assays, these MV are viable for up to 5 days in tissue culture media (2D) designed to support endothelial cells and up to 11 days post-isolation in a 3-dimensional (3D) matrix (Low Growth Factor Matrigel™). Assays that measure the reducing potential of live cells \demonstrated that the majority of the MV maintain high levels of metabolic activity for a similar number of days as the live/dead assays. Functional cellular components (such as tight junctions and transporter proteins) and ECM of MV in tissue culture media, and to a lesser extent in 3D matrices, were readily visualized using immunofluorescence techniques. MV in tissue culture media are lysed and protein content analyzed, but MV in 3D matrix first require removal of the supporting matrix, which can confound the analysis of MV ECM. Finally, MV can be preserved in cryoprotective media, whereby over 50% retain their baseline viability upon thawing. In summary, we find that MV isolated from human brains undergoing rapid autopsy are viable in standard tissue culture for up to 5 days and the timeframe for experiments can be extended up to 11 days by use of a supportive 3D matrix. Viable human MV allow for temporal and spatial analysis of relevant cellular and ECM components that have implications for microvascular function in neurodegenerative diseases, vascular brain injury, and neurotrauma.
Collapse
Affiliation(s)
- Mamatha Damodarasamy
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA, USA,VA Puget Sound Health Care System, Geriatric Research Education and Clinical Center, Seattle, WA, USA
| | - Zin Z Khaing
- Department of Neurosurgery, University of Washington, Seattle, WA, USA
| | - Jeffrey Hyde
- Department of Neurosurgery, University of Washington, Seattle, WA, USA
| | - C Dirk Keene
- Department of Laboratory Medicine and Pathology, Division of Neuropathology, University of Washington, Seattle, WA, USA
| | - Itay Bentov
- Department of Pain and Anesthesia, University of Washington, Seattle, WA, USA
| | - William A Banks
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA, USA,VA Puget Sound Health Care System, Geriatric Research Education and Clinical Center, Seattle, WA, USA
| | - May J Reed
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA, USA; VA Puget Sound Health Care System, Geriatric Research Education and Clinical Center, Seattle, WA, USA.
| |
Collapse
|
18
|
Oudebrouckx G, Goossens J, Bormans S, Vandenryt T, Wagner P, Thoelen R. Integrating Thermal Sensors in a Microplate Format: Simultaneous Real-Time Quantification of Cell Number and Metabolic Activity. ACS APPLIED MATERIALS & INTERFACES 2022; 14:2440-2451. [PMID: 34990545 DOI: 10.1021/acsami.1c14668] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Microplates have become a standard tool in the pharmaceutical industry and academia for a broad range of screening assays. One of the most commonly performed assays is the cell proliferation assay, which is often used for the purpose of drug discovery. Microplate readers play a crucial role in this field, as they enable high-throughput testing of large sample numbers. Common drawbacks of the most popular plate reader technologies are that they are end-point-based and most often require the use of detection reagents. As a solution, with this work, we aim to expand the possibilities of real-time and label-free monitoring of cell proliferation inside a microplate format by introducing a novel thermal-based sensing approach. For this purpose, we have developed thin-film sensors that can easily be integrated into the bottom of standard 96-well plates. First, the accuracy and precision of the sensors for measuring temperature and thermal effusivity are assessed via characterization experiments. These experiments highlight the fast response of the sensors to changes in temperature and thermal effusivity, as well as the excellent reproducibility between different sensors. Later, proof-of-principle measurements were performed on the proliferation of Saccharomyces cerevisiae. The proliferation measurements show that the thermal sensors were able to simultaneously detect relative changes in cell number as well as changes in metabolic activity. This dual functionality makes the presented sensor technology a promising candidate for monitoring microplate assays.
Collapse
Affiliation(s)
- Gilles Oudebrouckx
- Institute for Materials Research (IMO), Hasselt University, 3500 Hasselt, Belgium
- Division IMOMEC, IMEC vzw, 3590 Diepenbeek, Belgium
| | - Juul Goossens
- Institute for Materials Research (IMO), Hasselt University, 3500 Hasselt, Belgium
- Division IMOMEC, IMEC vzw, 3590 Diepenbeek, Belgium
| | - Seppe Bormans
- Institute for Materials Research (IMO), Hasselt University, 3500 Hasselt, Belgium
- Division IMOMEC, IMEC vzw, 3590 Diepenbeek, Belgium
| | - Thijs Vandenryt
- Institute for Materials Research (IMO), Hasselt University, 3500 Hasselt, Belgium
- Division IMOMEC, IMEC vzw, 3590 Diepenbeek, Belgium
| | - Patrick Wagner
- Laboratory for Soft Matter and Biophysics, KU Leuven, 3001 Leuven, Belgium
| | - Ronald Thoelen
- Institute for Materials Research (IMO), Hasselt University, 3500 Hasselt, Belgium
- Division IMOMEC, IMEC vzw, 3590 Diepenbeek, Belgium
| |
Collapse
|
19
|
Péter B, Boldizsár I, Kovács GM, Erdei A, Bajtay Z, Vörös A, Ramsden JJ, Szabó I, Bősze S, Horvath R. Natural Compounds as Target Biomolecules in Cellular Adhesion and Migration: From Biomolecular Stimulation to Label-Free Discovery and Bioactivity-Based Isolation. Biomedicines 2021; 9:1781. [PMID: 34944597 PMCID: PMC8698624 DOI: 10.3390/biomedicines9121781] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 01/07/2023] Open
Abstract
Plants and fungi can be used for medical applications because of their accumulation of special bioactive metabolites. These substances might be beneficial to human health, exerting also anti-inflammatory and anticancer (antiproliferative) effects. We propose that they are mediated by influencing cellular adhesion and migration via various signaling pathways and by directly inactivating key cell adhesion surface receptor sites. The evidence for this proposition is reviewed (by summarizing the natural metabolites and their effects influencing cellular adhesion and migration), along with the classical measuring techniques used to gain such evidence. We systematize existing knowledge concerning the mechanisms of how natural metabolites affect adhesion and movement, and their role in gene expression as well. We conclude by highlighting the possibilities to screen natural compounds faster and more easily by applying new label-free methods, which also enable a far greater degree of quantification than the conventional methods used hitherto. We have systematically classified recent studies regarding the effects of natural compounds on cellular adhesion and movement, characterizing the active substances according to their organismal origin (plants, animals or fungi). Finally, we also summarize the results of recent studies and experiments on SARS-CoV-2 treatments by natural extracts affecting mainly the adhesion and entry of the virus.
Collapse
Affiliation(s)
- Beatrix Péter
- Nanobiosensorics Group, Research Centre for Energy Research, Institute for Technical Physics and Materials Science, Konkoly-Thege u 29-33, 1120 Budapest, Hungary; (A.V.); (R.H.)
| | - Imre Boldizsár
- Department of Plant Anatomy, Institute of Biology, Eötvös Loránd University, 1117 Budapest, Hungary; (I.B.); (G.M.K.)
- Department of Pharmacognosy, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary
| | - Gábor M. Kovács
- Department of Plant Anatomy, Institute of Biology, Eötvös Loránd University, 1117 Budapest, Hungary; (I.B.); (G.M.K.)
- Plant Protection Institute, Centre for Agricultural Research, Hungarian Academy of Sciences, 1022 Budapest, Hungary
| | - Anna Erdei
- Department of Immunology, Eötvös Loránd University, 1117 Budapest, Hungary; (A.E.); (Z.B.)
- MTA-ELTE Immunology Research Group, Eötvös Loránd Research Network (ELKH), Eötvös Loránd University, 1117 Budapest, Hungary
| | - Zsuzsa Bajtay
- Department of Immunology, Eötvös Loránd University, 1117 Budapest, Hungary; (A.E.); (Z.B.)
- MTA-ELTE Immunology Research Group, Eötvös Loránd Research Network (ELKH), Eötvös Loránd University, 1117 Budapest, Hungary
| | - Alexandra Vörös
- Nanobiosensorics Group, Research Centre for Energy Research, Institute for Technical Physics and Materials Science, Konkoly-Thege u 29-33, 1120 Budapest, Hungary; (A.V.); (R.H.)
| | - Jeremy J. Ramsden
- Clore Laboratory, University of Buckingham, Buckingham MK18 1EG, UK;
| | - Ildikó Szabó
- MTA-ELTE Research Group of Peptide Chemistry, Eötvös Loránd Research Network (ELKH), Institute of Chemistry, Eötvös Loránd University, 1117 Budapest, Hungary; (I.S.); (S.B.)
- National Public Health Center, Albert Flórián út 2-6, 1097 Budapest, Hungary
| | - Szilvia Bősze
- MTA-ELTE Research Group of Peptide Chemistry, Eötvös Loránd Research Network (ELKH), Institute of Chemistry, Eötvös Loránd University, 1117 Budapest, Hungary; (I.S.); (S.B.)
- National Public Health Center, Albert Flórián út 2-6, 1097 Budapest, Hungary
| | - Robert Horvath
- Nanobiosensorics Group, Research Centre for Energy Research, Institute for Technical Physics and Materials Science, Konkoly-Thege u 29-33, 1120 Budapest, Hungary; (A.V.); (R.H.)
| |
Collapse
|
20
|
Hyaluronic Acid-Functionalized Nanomicelles Enhance SAHA Efficacy in 3D Endometrial Cancer Models. Cancers (Basel) 2021; 13:cancers13164032. [PMID: 34439185 PMCID: PMC8394402 DOI: 10.3390/cancers13164032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/28/2021] [Accepted: 08/09/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary One of the major limitations to cancer therapies are the side effects caused by the drug interacting with any tissue in the body. There is often a balance between patient health and effectively treating the disease. To by-pass this balancing act nanoparticles are being used to deliver therapeutics straight to the tumors, acting as “Trojan Horses”. Endometrial cancers are known to have more of the cell surface protein CD44 than healthy tissues. Here, to efficiently target endometrial cancer, hyaluronic acid, which naturally binds to the CD44 protein was attached to the surface of nanoparticles and tested on microtissues or spheroids to better model a tumor and understand drug delivery performance. We show that our hyaluronic acid-nanoparticle formulations improve drug effects and interact with the cancer cells more than without this targeting agent. Abstract Histone Deacetylase (HDAC) enzymes are upregulated in cancer leading to the development of HDAC inhibiting compounds, several of which are currently in clinical trials. Side effects associated with toxicity and non-specific targeting indicate the need for efficient drug delivery approaches and tumor specific targeting to enhance HDAC efficacy in solid tumor cancers. SAHA encapsulation within F127 micelles functionalized with a surface hyaluronic acid moiety, was developed to target endometrial cancer cells expressing elevated levels of CD44. In vitro viability and morphology analyses was conducted in both 2D and 3D models to assess the translational potential of this approach. Encapsulation enhanced SAHA delivery and activity, demonstrating increased cytotoxic efficacy in 2D and 3D endometrial cancer models. High-content imaging showed improved nanoparticle internalization in 2D and CD44 enhanced penetration in 3D models. In addition, the nano-delivery system enhanced spheroid penetration resulting in cell growth suppression, p21 associated cell cycle arrest, as well as overcoming the formation of an EMT associated phenotype observed in free drug treated type II endometrial cancer cells. This study demonstrates that targeted nanoparticle delivery of SAHA could provide the basis for improving its efficacy in endometrial cancer. Using 3D models for endometrial cancer allows the elucidation of nanoparticle performance and CD44 targeting, likely through penetration and retention within the tumor model.
Collapse
|
21
|
Mohtar N, Parumasivam T, Gazzali AM, Tan CS, Tan ML, Othman R, Fazalul Rahiman SS, Wahab HA. Advanced Nanoparticle-Based Drug Delivery Systems and Their Cellular Evaluation for Non-Small Cell Lung Cancer Treatment. Cancers (Basel) 2021; 13:3539. [PMID: 34298753 PMCID: PMC8303683 DOI: 10.3390/cancers13143539] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/18/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
Lung cancers, the number one cancer killer, can be broadly divided into small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC), with NSCLC being the most commonly diagnosed type. Anticancer agents for NSCLC suffer from various limitations that can be partly overcome by the application of nanomedicines. Nanoparticles is a branch within nanomedicine that can improve the delivery of anticancer drugs, whilst ensuring the stability and sufficient bioavailability following administration. There are many publications available in the literature exploring different types of nanoparticles from different materials. The effectiveness of a treatment option needs to be validated in suitable in vitro and/or in vivo models. This includes the developed nanoparticles, to prove their safety and efficacy. Many researchers have turned towards in vitro models that use normal cells or specific cells from diseased tissues. However, in cellular works, the physiological dynamics that is available in the body could not be mimicked entirely, and hence, there is still possible development of false positive or false negative results from the in vitro models. This article provides an overview of NSCLC, the different nanoparticles available to date, and in vitro evaluation of the nanoparticles. Different types of cells suitable for in vitro study and the important precautions to limit the development of false results are also extensively discussed.
Collapse
Affiliation(s)
- Noratiqah Mohtar
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor 11800, Penang, Malaysia; (N.M.); (T.P.); (A.M.G.); (C.S.T.); (M.L.T.); (H.A.W.)
| | - Thaigarajan Parumasivam
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor 11800, Penang, Malaysia; (N.M.); (T.P.); (A.M.G.); (C.S.T.); (M.L.T.); (H.A.W.)
| | - Amirah Mohd Gazzali
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor 11800, Penang, Malaysia; (N.M.); (T.P.); (A.M.G.); (C.S.T.); (M.L.T.); (H.A.W.)
| | - Chu Shan Tan
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor 11800, Penang, Malaysia; (N.M.); (T.P.); (A.M.G.); (C.S.T.); (M.L.T.); (H.A.W.)
| | - Mei Lan Tan
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor 11800, Penang, Malaysia; (N.M.); (T.P.); (A.M.G.); (C.S.T.); (M.L.T.); (H.A.W.)
| | - Rozana Othman
- Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur 50603, Malaysia
- Center for Natural Products Research and Drug Discovery (CENAR), Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Siti Sarah Fazalul Rahiman
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor 11800, Penang, Malaysia; (N.M.); (T.P.); (A.M.G.); (C.S.T.); (M.L.T.); (H.A.W.)
| | - Habibah A. Wahab
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor 11800, Penang, Malaysia; (N.M.); (T.P.); (A.M.G.); (C.S.T.); (M.L.T.); (H.A.W.)
| |
Collapse
|
22
|
Beloglazova Y, Nikitiuk A, Voronina A, Gagarskikh O, Bayandin Y, Naimark O, Grishko V. Label-Free Single Cell Viability Assay Using Laser Interference Microscopy. BIOLOGY 2021; 10:590. [PMID: 34206974 PMCID: PMC8301067 DOI: 10.3390/biology10070590] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/21/2021] [Accepted: 06/24/2021] [Indexed: 12/20/2022]
Abstract
Laser interference microscopy (LIM) is a promising label-free method for single-cell research applicable to cell viability assessment in the studies of mammalian cells. This paper describes the development of a sensitive and reproducible method for assessing cell viability using LIM. The method, based on associated signal processing techniques, has been developed as a result of real-time investigation in phase thickness fluctuations of viable and non-viable MCF-7 cells, reflecting the presence and absence of their metabolic activity. As evinced by the values of the variable vc, this variable determines the viability of a cell only in the attached state (vc exceeds 20 nm2 for viable attached cells). The critical value of the power spectrum slope βc of the phase thickness fluctuations equals 1.00 for attached MCF-7 cells and 0.71 for suspended cells. The slope of the phase fluctuations' power spectrum for MCF-7 cells was determined to exceed the threshold value of βc for a living cell, otherwise the cell is dead. The results evince the power spectrum slope as the most appropriate indicator of cell viability, while the integrated evaluation criterion (vc and βc values) can be used to assay the viability of attached cells.
Collapse
Affiliation(s)
- Yulia Beloglazova
- Perm Federal Scientific Centre, Institute of Technical Chemistry UB RAS, Academician Korolev St. 3, 614013 Perm, Russia; (Y.B.); (A.V.); (O.G.)
| | - Aleksandr Nikitiuk
- Perm Federal Scientific Centre, Institute of Continuous Media Mechanics UB RAS, Academician Korolev St. 1, 614013 Perm, Russia; (A.N.); (Y.B.); (O.N.)
| | - Anna Voronina
- Perm Federal Scientific Centre, Institute of Technical Chemistry UB RAS, Academician Korolev St. 3, 614013 Perm, Russia; (Y.B.); (A.V.); (O.G.)
| | - Olga Gagarskikh
- Perm Federal Scientific Centre, Institute of Technical Chemistry UB RAS, Academician Korolev St. 3, 614013 Perm, Russia; (Y.B.); (A.V.); (O.G.)
| | - Yuriy Bayandin
- Perm Federal Scientific Centre, Institute of Continuous Media Mechanics UB RAS, Academician Korolev St. 1, 614013 Perm, Russia; (A.N.); (Y.B.); (O.N.)
| | - Oleg Naimark
- Perm Federal Scientific Centre, Institute of Continuous Media Mechanics UB RAS, Academician Korolev St. 1, 614013 Perm, Russia; (A.N.); (Y.B.); (O.N.)
| | - Victoria Grishko
- Perm Federal Scientific Centre, Institute of Technical Chemistry UB RAS, Academician Korolev St. 3, 614013 Perm, Russia; (Y.B.); (A.V.); (O.G.)
| |
Collapse
|
23
|
Hofsteen P, Karassina N, Cali JJ, Vidugiriene J. A Luminescence Assay to Quantify Cell Viability in Real Time. Methods Mol Biol 2021; 2255:187-196. [PMID: 34033104 DOI: 10.1007/978-1-0716-1162-3_16] [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] [Indexed: 06/12/2023]
Abstract
Comprehensive understanding of cellular responses to changes in the cellular environment or by drug treatment requires time-dependent analysis ranging from hours to several days. Here, we describe a sensitive, nonlytic live-cell assay that allows continuous or 'real-time' monitoring of cell viability, growth, and cytotoxicity over an extended period of time. We illustrate the use of the assay for small drug molecule and antibody-dependent cytotoxicity studies using cancer cells in 384-well plates. We show that the ability to measure changes in live cells over time provides instantaneous information on the biological status of the cells, information about the mode of action of the drug, and offers an added advantage of preserving the cells for multiplexing with downstream applications.
Collapse
Affiliation(s)
- Peter Hofsteen
- Research and Development, Promega Corporation, Madison, WI, USA
| | | | - James J Cali
- Research and Development, Promega Corporation, Madison, WI, USA
| | | |
Collapse
|
24
|
Van den Bossche S, Vandeplassche E, Ostyn L, Coenye T, Crabbé A. Bacterial Interference With Lactate Dehydrogenase Assay Leads to an Underestimation of Cytotoxicity. Front Cell Infect Microbiol 2020; 10:494. [PMID: 33042868 PMCID: PMC7523407 DOI: 10.3389/fcimb.2020.00494] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 08/10/2020] [Indexed: 12/21/2022] Open
Abstract
Models to study host-pathogen interactions in vitro are an important tool for investigating the infectious disease process and evaluating the efficacy of antimicrobial compounds. In these models, the viability of mammalian cells is often determined using the lactate dehydrogenase (LDH) cytotoxicity assay. In the present study we evaluated whether bacteria could interfere with the LDH assay. As a model for host-pathogen interactions, we co-cultured lung epithelial cells with eight bacteria encountered in the lower respiratory tract. We show that LDH activity is affected by Pseudomonas aeruginosa, Klebsiella pneumoniae, Stenotrophomonas maltophilia, and Streptococcus pneumoniae, and that this depends on the density of the start inoculum and the duration of infection. Two different mechanisms were discovered through which bacteria interfered with LDH activity, i.e., acidification of the cell culture medium (by K. pneumoniae and S. pneumoniae) and protease production (by P. aeruginosa and S. maltophilia). In addition, we developed and validated a modified protocol to evaluate cytotoxicity using the LDH assay, where bacterial interference with LDH quantification is avoided.
Collapse
Affiliation(s)
| | - Eva Vandeplassche
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Lisa Ostyn
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Aurélie Crabbé
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
25
|
Imidazo[1,2- b]pyrazole-7-Carboxamide Derivative Induces Differentiation-Coupled Apoptosis of Immature Myeloid Cells Such as Acute Myeloid Leukemia and Myeloid-Derived Suppressor Cells. Int J Mol Sci 2020; 21:ijms21145135. [PMID: 32698503 PMCID: PMC7404197 DOI: 10.3390/ijms21145135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/15/2020] [Accepted: 07/17/2020] [Indexed: 12/29/2022] Open
Abstract
Chemotherapy-induced differentiation of immature myeloid progenitors, such as acute myeloid leukemia (AML) cells or myeloid-derived suppressor cells (MDSCs), has remained a challenge for the clinicians. Testing our imidazo[1,2-b]pyrazole-7-carboxamide derivative on HL-60 cells, we obtained ERK phosphorylation as an early survival response to treatment followed by the increase of the percentage of the Bcl-xlbright and pAktbright cells. Following the induction of Vav1 and the AP-1 complex, a driver of cellular differentiation, FOS, JUN, JUNB, and JUND were elevated on a concentration and time-dependent manner. As a proof of granulocytic differentiation, the cells remained non-adherent, the expression of CD33 decreased; the granularity, CD11b expression, and MPO activity of HL-60 cells increased upon treatment. Finally, viability of HL-60 cells was hampered shown by the depolarization of mitochondria, activation of caspase-3, cleavage of Z-DEVD-aLUC, appearance of the sub-G1 population, and the leakage of the lactate-dehydrogenase into the supernatant. We confirmed the differentiating effect of our drug candidate on human patient-derived AML cells shown by the increase of CD11b and decrease of CD33+, CD7+, CD206+, and CD38bright cells followed apoptosis (IC50: 80 nM) after treatment ex vivo. Our compound reduced both CD11b+/Ly6C+ and CD11b+/Ly6G+ splenic MDSCs from the murine 4T1 breast cancer model ex vivo.
Collapse
|
26
|
Liu H, Dilger JP, Lin J. Effects of local anesthetics on cancer cells. Pharmacol Ther 2020; 212:107558. [PMID: 32343985 DOI: 10.1016/j.pharmthera.2020.107558] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/15/2020] [Indexed: 02/06/2023]
Abstract
Local anesthetics are widely used during clinical cancer surgeries. Studies have suggested that the use and the type of anesthesia affect cancer outcomes. In vivo studies and clinical data show that the use of local anesthetics is potentially beneficial for cancer treatment. However, the effect of the use of local anesthetics on the survival rate of cancer patients following surgery is controversial and, so far, little is known about the direct effects of local anesthetics on cancer cells. This work reviews and summarizes the published literature regarding the preclinical research methods and findings on the influence of local anesthetics on cancer cells. We hope that a thorough understanding of this subject will help to define optimal anesthetic regimens that lead to better outcomes for clinical cancer patients.
Collapse
Affiliation(s)
- Hengrui Liu
- Department of Anesthesiology, Health Science Center, Stony Brook University, Stony Brook, NY 11794, USA
| | - James P Dilger
- Department of Anesthesiology, Health Science Center, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jun Lin
- Department of Anesthesiology, Health Science Center, Stony Brook University, Stony Brook, NY 11794, USA.
| |
Collapse
|
27
|
Abstract
Apoptosis is an important and necessary cell death program which promotes homeostasis and organismal survival. When dysregulated, however, it can lead to a myriad of pathologies from neurodegenerative diseases to cancer. Apoptosis is therefore the subject of intense study aimed at dissecting its pathways and molecular mechanisms. Although many assay methods exist for confirming whether an apoptotic response has occurred in vitro, most methods are destructive and involve laborious operator effort or specialized instrumentation. Here we describe a real-time, no-wash, microplate method which utilizes recombinant annexin V fusion proteins containing evolved binary subunits of NanoBiT™ luciferase. The fusion proteins, a time-released enzymatic substrate, a necrosis detection dye and exogenous calcium ions are delivered via an optimized and physiologically inert reagent directly to cells in culture at the time of treatment or dosing. Luminescent signals proportional to phosphatidylserine (PS) exposure and fluorescent signals generated as a result of loss of membrane integrity are then collected using a standard multimode plate reader at scheduled intervals over the exposure. The resulting luminescent and fluorescent data are then used to define the kinetics and magnitude of an apoptotic response. This study details our efforts to develop, characterize, and demonstrate the features of the assay by providing relevant examples from diverse cell models for programmed cell death.
Collapse
|
28
|
Teow SY, Liew K, Che Mat MF, Marzuki M, Abdul Aziz N, Chu TL, Ahmad M, Khoo ASB. Development of a luciferase/luciferin cell proliferation (XenoLuc) assay for real-time measurements of Gfp-Luc2-modified cells in a co-culture system. BMC Biotechnol 2019; 19:34. [PMID: 31200673 PMCID: PMC6570829 DOI: 10.1186/s12896-019-0528-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 05/20/2019] [Indexed: 11/30/2022] Open
Abstract
Background In vitro modelling of cancer cells is becoming more complex due to prevailing evidence of intimate interactions between cancer cells and their surrounding stroma. A co-culture system which consists of more than one cell type is physiologically more relevant and thus, could serve as a useful model for various biological studies. An assay that specifically detects the phenotypic changes of cancer cells in a multi-cellular system is lacking for nasopharyngeal carcinoma (NPC). Results Here, we describe a luciferase/luciferin (XenoLuc) assay that could specifically measure changes in the proliferation of cancer cells in the co-culture system using two modified NPC patient-derived tumour xenograft (PDTXs) cells: Xeno284-gfp-luc2 and XenoB110-gfp-luc2. Through this assay, we are able to show that the growth of NPC xenograft cells in both two-dimensional (2D) and three-dimensional (3D) models was enhanced when co-cultured with normal human dermal fibroblasts (NHDFs). In addition, potential applications of this assay in in vitro drug or inhibitor screening experiments are also illustrated. Conclusions XenoLuc assay is specific, sensitive, rapid and cost-effective for measuring the growth of luciferase-expressing cells in a co- or multiple-culture system. This assay may also be adapted for tumour microenvironment studies as well as drug screening experiments in more complex 3D co-culture systems. Electronic supplementary material The online version of this article (10.1186/s12896-019-0528-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sin-Yeang Teow
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, National Institutes of Health (NIH Complex), Ministry of Health Malaysia, Level 4, Block C7, No: 1, Jalan Setia Murni U13/52, Section U13, Setia Alam, 40170 Shah Alam, 50588, Kuala Lumpur, Selangor, Malaysia.,Present Address: Department of Medical Sciences, School of Healthcare and Medical Sciences, Sunway University, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia
| | - Kitson Liew
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, National Institutes of Health (NIH Complex), Ministry of Health Malaysia, Level 4, Block C7, No: 1, Jalan Setia Murni U13/52, Section U13, Setia Alam, 40170 Shah Alam, 50588, Kuala Lumpur, Selangor, Malaysia
| | - Mohd Firdaus Che Mat
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, National Institutes of Health (NIH Complex), Ministry of Health Malaysia, Level 4, Block C7, No: 1, Jalan Setia Murni U13/52, Section U13, Setia Alam, 40170 Shah Alam, 50588, Kuala Lumpur, Selangor, Malaysia
| | - Marini Marzuki
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, National Institutes of Health (NIH Complex), Ministry of Health Malaysia, Level 4, Block C7, No: 1, Jalan Setia Murni U13/52, Section U13, Setia Alam, 40170 Shah Alam, 50588, Kuala Lumpur, Selangor, Malaysia
| | - Norazlin Abdul Aziz
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, National Institutes of Health (NIH Complex), Ministry of Health Malaysia, Level 4, Block C7, No: 1, Jalan Setia Murni U13/52, Section U13, Setia Alam, 40170 Shah Alam, 50588, Kuala Lumpur, Selangor, Malaysia
| | - Tai-Lin Chu
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, National Institutes of Health (NIH Complex), Ministry of Health Malaysia, Level 4, Block C7, No: 1, Jalan Setia Murni U13/52, Section U13, Setia Alam, 40170 Shah Alam, 50588, Kuala Lumpur, Selangor, Malaysia
| | - Munirah Ahmad
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, National Institutes of Health (NIH Complex), Ministry of Health Malaysia, Level 4, Block C7, No: 1, Jalan Setia Murni U13/52, Section U13, Setia Alam, 40170 Shah Alam, 50588, Kuala Lumpur, Selangor, Malaysia
| | - Alan Soo-Beng Khoo
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, National Institutes of Health (NIH Complex), Ministry of Health Malaysia, Level 4, Block C7, No: 1, Jalan Setia Murni U13/52, Section U13, Setia Alam, 40170 Shah Alam, 50588, Kuala Lumpur, Selangor, Malaysia.
| |
Collapse
|
29
|
Stereochemistry as a determining factor for the effect of a cell-penetrating peptide on cellular viability and epithelial integrity. Biochem J 2018; 475:1773-1788. [PMID: 29686042 DOI: 10.1042/bcj20180155] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/19/2018] [Accepted: 04/20/2018] [Indexed: 01/07/2023]
Abstract
Cell-penetrating peptides (CPPs) comprise efficient peptide-based delivery vectors. Owing to the inherent poor enzymatic stability of peptides, CPPs displaying partial or full replacement of l-amino acids with the corresponding d-amino acids might possess advantages as delivery vectors. Thus, the present study aims to elucidate the membrane- and metabolism-associated effects of l-Penetratin (l-PEN) and its corresponding all-d analog (d-PEN). These effects were investigated when exerted on hepatocellular (HepG2) or intestinal (Caco-2 and IEC-6) cell culture models. The head-to-head comparison of these enantiomeric CPPs included evaluation of their effects on cell viability and morphology, epithelial membrane integrity, and cellular ultrastructure. In all investigated cell models, a rapid decrease in cell viability, pronounced membrane perturbation and an altered ultrastructure were detected upon exposure to d-PEN. At equimolar concentrations, these observations were less pronounced or even absent for cells exposed to l-PEN. Both CPPs remained stable for at least 2 h during exposure to proliferating cells (cultured for 24 h), although d-PEN exhibited a longer half-life when compared with that of l-PEN when exposed to well-differentiated cell monolayers (cultured for 18-20 days). Thus, the stereochemistry of the CPP penetratin significantly influences its effects on cell viability and epithelial integrity when profiled against a panel of mammalian cells.
Collapse
|
30
|
Orbach SM, Ehrich MF, Rajagopalan P. High-throughput toxicity testing of chemicals and mixtures in organotypic multi-cellular cultures of primary human hepatic cells. Toxicol In Vitro 2018; 51:83-94. [PMID: 29751030 DOI: 10.1016/j.tiv.2018.05.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 05/06/2018] [Accepted: 05/07/2018] [Indexed: 02/07/2023]
Abstract
High-throughput screening (HTS) of liver toxicants can bridge the gap in understanding adverse effects of chemicals on humans. Toxicity testing of mixtures is time consuming and expensive, since the number of possible combinations increases exponentially with the number of chemicals. The combination of organotypic culture models (OCMs) and HTS assays can lead to the rapidly evaluation of chemical toxicity in a cost and time-effective manner while prioritizing chemicals that warrant additional investigation. We describe the design, assembly and toxicant response of multi-cellular hepatic organotypic culture models comprised of primary human or rat cells assembled in 96-well plates (denoted as μOCMs). These models were assembled using automated procedures that did not affect hepatocyte function or viability, rendering them ideal for large-scale toxicity evaluations. Rat μOCMs were assembled to obtain insights into deviations from human toxicity. Four test chemicals (acetaminophen, ethanol, isoniazid, and perfluorooctanoic acid) were added to the μOCMs individually or in mixtures. HTS assays were utilized to measure cell death, apoptosis, glutathione depletion, mitochondrial membrane damage, and cytochrome P450 2E1 activity. The μOCMs exhibited increased toxicant sensitivity compared to hepatocyte sandwich cultures. Synergistic and non-synergistic interactions were observed when the toxicants were added as mixtures. Specifically, chemical interactions in the μOCMs were manifested by changes in apoptosis and decreased glutathione. The μOCMs accurately predicted hepatotoxicity for individual and mixtures of toxicants, demonstrating their potential for large-scale toxicity evaluations in the future.
Collapse
Affiliation(s)
- Sophia M Orbach
- Department of Chemical Engineering, Virginia Tech, Suite 245 Goodwin Hall, 635 Prices Fork Road, Blacksburg, VA 24061, USA.
| | - Marion F Ehrich
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, 205 Duck Pond Drive, Blacksburg, VA 24061, USA.
| | - Padmavathy Rajagopalan
- Department of Chemical Engineering, Virginia Tech, Suite 245 Goodwin Hall, 635 Prices Fork Road, Blacksburg, VA 24061, USA; ICTAS Center for Systems Biology of Engineered Tissue, Virginia Tech, 333 Kelly Hall, 325 Stanger Street, Blacksburg, VA 24061, USA; School of Biomedical Engineering and Sciences, Virginia Tech, 333 Kelly Hall, 325 Stanger Street, Blacksburg, VA 24061, USA.
| |
Collapse
|
31
|
Lee HS, Park EJ, Han S, Oh GY, Kang HS, Suh JH, Shin MK, Oh HS, Hwang MS, Moon G, Koh YH, Park Y, Hong JH, Koo YE. Assessment of androgen receptor agonistic/antagonistic effects on 25 chemicals in household applicants by OECD in vitro stably transfected transcriptional activation assays. CHEMOSPHERE 2018; 191:589-596. [PMID: 29073568 DOI: 10.1016/j.chemosphere.2017.10.084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 10/10/2017] [Accepted: 10/14/2017] [Indexed: 06/07/2023]
Abstract
The aim of this study is to assess the androgen receptor (AR) agonistic/antagonistic effects on various chemicals, which are used in household products including cleaning agents and wetted tissues by in vitro OECD test guideline No. 458 (using AR-EcoScreen™ cell line) and the me-too test method (using 22Rv1cell line), which was adopted as OECD project No. 4.99. All chemicals were not determined as AR agonists. However α-dodecyl-ω-hydroxypoly (oxyethylene) and 3-iodo-2-propynyl butylcarbamate have shown a weak AR antagonistic effects with IC50 values of 2.18 ± 0.12 and 4.26 ± 0.17 μg/ml via binding affinity to AR in only 22Rv1/mouse mammary tumor virus using AR transcriptional activation assay, because of their different cytotoxicity on each applied cell line. This report firstly provides information about agonistic/antagonistic effects against human AR of various chemicals including surfactants and biocides by OECD in vitro stably transfected transcriptional activation assays. However, further in vivo and human model studies are needed to confirm their adverse effects.
Collapse
Affiliation(s)
- Hee-Seok Lee
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea.
| | - Eun-Jung Park
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Songyi Han
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Gyeong-Yong Oh
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Hui-Seung Kang
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Jin-Hyang Suh
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Min-Ki Shin
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Hyun-Suk Oh
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Myung-Sil Hwang
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Guiim Moon
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Young-Ho Koh
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Yooheon Park
- Dongguk University Research Institute of Biotechnology & Medical Converged Science, Dongguk University, Gyeonggi 10325, South Korea
| | - Jin-Hwan Hong
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea
| | - Yong Eui Koo
- Food Safety Risk Assessment Division, National Institute of Food and Drug Safety Evaluation, Korean Ministry of Food and Drug Safety, Chungcheongbuk-do 28159, South Korea.
| |
Collapse
|
32
|
Hassan F, El-Hiti GA, Abd-Allateef M, Yousif E. Cytotoxicity anticancer activities of anastrozole against breast, liver hepatocellular, and prostate cancer cells. Saudi Med J 2017; 38:359-365. [PMID: 28397941 PMCID: PMC5447187 DOI: 10.15537/smj.2017.4.17061] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVES To investigate the cytotoxic effect of anastrozole on breast (MCF7), liver hepatocellular (HepG2), and prostate (PC3) cancer cells. Methods: This is a prospective study. Anastrozole's mechanism of apoptosis in living cells was also determined by high content screening (HCS) assay. Methylthiazol tetrazolium (MTT) assay was carried out at the Centre of Biotechnology Research's, Al-Nahrain University, Baghdad, Iraq between July 2015 and October 2015. The HCS assay was performed at the Centre for Natural Product Research and Drug Discovery, Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia between November 2015 and February 2016. Results: The most significant cytotoxic effect of anastrozole towards 3 cancer cell lines was obtained when its concentration was 400 µg/mL. The MCF7 cells were more sensitive to anastrozole compared with the HepG2 and PC-3 cells. There was a significant increase in membrane permeability, cytochrome c and nuclear intensity when anastrozole (200 µg/mL) was used compared with doxorubicin (20 µg/mL) as a standard. Also, there was a significant decrease in cell viability and mitochondrial membrane permeability when anastrozole (200 µg/mL) was used compared with positive control. Conclusion: Anastrozole showed cytotoxic effects against the MCF7, HepG2, and PC3 cell lines as determined in-vitro by the MTT assay. The HCS technique also showed toxic effect towards MCF7. It is evident that anastrozole inhibits the aromatase enzyme preventing the aromatization mechanism; however, it has a toxic effect.
Collapse
Affiliation(s)
- Firas Hassan
- Department of Chemistry, College of Science, Al-Nahrain University, Baghdad, Iraq. E-mail.
| | | | | | | |
Collapse
|
33
|
Methods to Evaluate Cell Growth, Viability, and Response to Treatment in a Tissue Engineered Breast Cancer Model. Sci Rep 2017; 7:14167. [PMID: 29074857 PMCID: PMC5658356 DOI: 10.1038/s41598-017-14326-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/09/2017] [Indexed: 01/01/2023] Open
Abstract
The use of in vitro, engineered surrogates in the field of cancer research is of interest for studies involving mechanisms of growth and metastasis, and response to therapeutic intervention. While biomimetic surrogates better model human disease, their complex composition and dimensionality make them challenging to evaluate in a real-time manner. This feature has hindered the broad implementation of these models, particularly in drug discovery. Herein, several methods and approaches for the real-time, non-invasive analysis of cell growth and response to treatment in tissue-engineered, three-dimensional models of breast cancer are presented. The tissue-engineered surrogates used to demonstrate these methods consist of breast cancer epithelial cells and fibroblasts within a three dimensional volume of extracellular matrix and are continuously perfused with nutrients via a bioreactor system. Growth of the surrogates over time was measured using optical in vivo (IVIS) imaging. Morphologic changes in specific cell populations were evaluated by multi-photon confocal microscopy. Response of the surrogates to treatment with paclitaxel was measured by optical imaging and by analysis of lactate dehydrogenase and caspase-cleaved cytokeratin 18 in the perfused medium. Each method described can be repeatedly performed during culture, allowing for real-time, longitudinal analysis of cell populations within engineered tumor models.
Collapse
|
34
|
Veyrand J, Marin-Kuan M, Bezencon C, Frank N, Guérin V, Koster S, Latado H, Mollergues J, Patin A, Piguet D, Serrant P, Varela J, Schilter B. Integrating bioassays and analytical chemistry as an improved approach to support safety assessment of food contact materials. Food Addit Contam Part A Chem Anal Control Expo Risk Assess 2017; 34:1807-1816. [PMID: 28730944 DOI: 10.1080/19440049.2017.1358466] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Food contact materials (FCM) contain chemicals which can migrate into food and result in human exposure. Although it is mandatory to ensure that migration does not endanger human health, there is still no consensus on how to pragmatically assess the safety of FCM since traditional approaches would require extensive toxicological and analytical testing which are expensive and time consuming. Recently, the combination of bioassays, analytical chemistry and risk assessment has been promoted as a new paradigm to identify toxicologically relevant molecules and address safety issues. However, there has been debate on the actual value of bioassays in that framework. In the present work, a FCM anticipated to release the endocrine active chemical 4-nonyphenol (4NP) was used as a model. In a migration study, the leaching of 4NP was confirmed by LC-MS/MS and GC-MS. This was correlated with an increase in both estrogenic and anti-androgenic activities as measured with bioassays. A standard risk assessment indicated that according to the food intake scenario applied, the level of 4NP measured was lower, close or slightly above the acceptable daily intake. Altogether these results show that bioassays could reveal the presence of an endocrine active chemical in a real-case FCM migration study. The levels reported were relevant for safety assessment. In addition, this work also highlighted that bioactivity measured in migrate does not necessarily represent a safety issue. In conclusion, together with analytics, bioassays contribute to identify toxicologically relevant molecules leaching from FCM and enable improved safety assessment.
Collapse
Affiliation(s)
- Julien Veyrand
- a Institute of Food Safety and Analytical Science , Nestlé Research Center , Lausanne , Switzerland
| | - Maricel Marin-Kuan
- a Institute of Food Safety and Analytical Science , Nestlé Research Center , Lausanne , Switzerland
| | - Claudine Bezencon
- a Institute of Food Safety and Analytical Science , Nestlé Research Center , Lausanne , Switzerland
| | - Nancy Frank
- a Institute of Food Safety and Analytical Science , Nestlé Research Center , Lausanne , Switzerland
| | - Violaine Guérin
- a Institute of Food Safety and Analytical Science , Nestlé Research Center , Lausanne , Switzerland
| | - Sander Koster
- a Institute of Food Safety and Analytical Science , Nestlé Research Center , Lausanne , Switzerland
| | - Hélia Latado
- a Institute of Food Safety and Analytical Science , Nestlé Research Center , Lausanne , Switzerland
| | - Julie Mollergues
- a Institute of Food Safety and Analytical Science , Nestlé Research Center , Lausanne , Switzerland
| | - Amaury Patin
- a Institute of Food Safety and Analytical Science , Nestlé Research Center , Lausanne , Switzerland
| | - Dominique Piguet
- a Institute of Food Safety and Analytical Science , Nestlé Research Center , Lausanne , Switzerland
| | - Patrick Serrant
- a Institute of Food Safety and Analytical Science , Nestlé Research Center , Lausanne , Switzerland
| | - Jesus Varela
- a Institute of Food Safety and Analytical Science , Nestlé Research Center , Lausanne , Switzerland
| | - Benoît Schilter
- a Institute of Food Safety and Analytical Science , Nestlé Research Center , Lausanne , Switzerland
| |
Collapse
|
35
|
Keidar M, Yan D, Beilis II, Trink B, Sherman JH. Plasmas for Treating Cancer: Opportunities for Adaptive and Self-Adaptive Approaches. Trends Biotechnol 2017; 36:586-593. [PMID: 28755977 DOI: 10.1016/j.tibtech.2017.06.013] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/14/2017] [Accepted: 06/15/2017] [Indexed: 01/31/2023]
Abstract
Plasma is an ionized gas that is typically formed under high-temperature laboratory conditions. Recent progress in atmospheric plasmas has led to cold atmospheric plasma (CAP) devices with ion temperatures close to room temperature. The unique chemical and physical properties of CAP have led to its use in various biomedical applications including cancer therapy. CAP exhibits a spontaneous transition from a spatially homogeneous state to a modifiable pattern that is subject to self-organization. In this Opinion article, we discuss some new applications for plasma in cancer therapy based on plasma self-organization, which enables adaptive features in plasma-based therapeutic systems.
Collapse
Affiliation(s)
- Michael Keidar
- The George Washington University, Washington, DC 20052, USA.
| | - Dayun Yan
- The George Washington University, Washington, DC 20052, USA
| | | | | | | |
Collapse
|
36
|
Hsieh JH, Huang R, Lin JA, Sedykh A, Zhao J, Tice RR, Paules RS, Xia M, Auerbach SS. Real-time cell toxicity profiling of Tox21 10K compounds reveals cytotoxicity dependent toxicity pathway linkage. PLoS One 2017; 12:e0177902. [PMID: 28531190 PMCID: PMC5439695 DOI: 10.1371/journal.pone.0177902] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 05/04/2017] [Indexed: 01/01/2023] Open
Abstract
Cytotoxicity is a commonly used in vitro endpoint for evaluating chemical toxicity. In support of the U.S. Tox21 screening program, the cytotoxicity of ~10K chemicals was interrogated at 0, 8, 16, 24, 32, & 40 hours of exposure in a concentration dependent fashion in two cell lines (HEK293, HepG2) using two multiplexed, real-time assay technologies. One technology measures the metabolic activity of cells (i.e., cell viability, glo) while the other evaluates cell membrane integrity (i.e., cell death, flor). Using glo technology, more actives and greater temporal variations were seen in HEK293 cells, while results for the flor technology were more similar across the two cell types. Chemicals were grouped into classes based on their cytotoxicity kinetics profiles and these classes were evaluated for their associations with activity in the Tox21 nuclear receptor and stress response pathway assays. Some pathways, such as the activation of H2AX, were associated with the fast-responding cytotoxicity classes, while others, such as activation of TP53, were associated with the slow-responding cytotoxicity classes. By clustering pathways based on their degree of association to the different cytotoxicity kinetics labels, we identified clusters of pathways where active chemicals presented similar kinetics of cytotoxicity. Such linkages could be due to shared underlying biological processes between pathways, for example, activation of H2AX and heat shock factor. Others involving nuclear receptor activity are likely due to shared chemical structures rather than pathway level interactions. Based on the linkage between androgen receptor antagonism and Nrf2 activity, we surmise that a subclass of androgen receptor antagonists cause cytotoxicity via oxidative stress that is associated with Nrf2 activation. In summary, the real-time cytotoxicity screen provides informative chemical cytotoxicity kinetics data related to their cytotoxicity mechanisms, and with our analysis, it is possible to formulate mechanism-based hypotheses on the cytotoxic properties of the tested chemicals.
Collapse
Affiliation(s)
- Jui-Hua Hsieh
- Kelly Government Solutions, Durham, North Carolina, United States of America
| | - Ruili Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, United States of America
| | - Ja-An Lin
- US Food and Drug Administration, Silver Spring, Maryland, United States of America
| | | | - Jinghua Zhao
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, United States of America
| | - Raymond R. Tice
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina, United States of America
| | - Richard S. Paules
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina, United States of America
| | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, United States of America
| | - Scott S. Auerbach
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina, United States of America
| |
Collapse
|
37
|
A novel method for quantitative measurements of gene expression in single living cells. Methods 2017; 120:65-75. [PMID: 28456689 DOI: 10.1016/j.ymeth.2017.04.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/12/2017] [Accepted: 04/14/2017] [Indexed: 12/13/2022] Open
Abstract
Gene expression is at the heart of virtually any biological process, and its deregulation is at the source of numerous pathological conditions. While impressive progress has been made in genome-wide measurements of mRNA and protein expression levels, it is still challenging to obtain highly quantitative measurements in single living cells. Here we describe a novel approach based on internal tagging of endogenous proteins with a reporter allowing luminescence and fluorescence time-lapse microscopy. Using luminescence microscopy, fluctuations of protein expression levels can be monitored in single living cells with high sensitivity and temporal resolution over extended time periods. The integrated protein decay reporter allows measuring protein degradation rates in the absence of protein synthesis inhibitors, and in combination with absolute protein levels allows determining absolute amounts of proteins synthesized over the cell cycle. Finally, the internal tag can be excised by inducible expression of Cre recombinase, which enables to estimate endogenous mRNA half-lives. Our method thus opens new avenues in quantitative analysis of gene expression in single living cells.
Collapse
|
38
|
Walker JR, Hall MP, Zimprich CA, Robers MB, Duellman SJ, Machleidt T, Rodriguez J, Zhou W. Highly Potent Cell-Permeable and Impermeable NanoLuc Luciferase Inhibitors. ACS Chem Biol 2017; 12:1028-1037. [PMID: 28195704 DOI: 10.1021/acschembio.6b01129] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Novel engineered NanoLuc (Nluc) luciferase being smaller, brighter, and superior to traditional firefly (Fluc) or Renilla (Rluc) provides a great opportunity for the development of numerous biological, biomedical, clinical, and food and environmental safety applications. This new platform created an urgent need for Nluc inhibitors that could allow selective bioluminescent suppression and multiplexing compatibility with existing luminescence or fluorescence assays. Starting from thienopyrrole carboxylate 1, a hit from a 42 000 PubChem compound library with a low micromolar IC50 against Nluc, we derivatized four different structural fragments to discover a family of potent, single digit nanomolar, cell permeable inhibitors. Further elaboration revealed a channel that allowed access to the external Nluc surface, resulting in a series of highly potent cell impermeable Nluc inhibitors with negatively charged groups likely extending to the protein surface. The permeability was evaluated by comparing EC50 shifts calculated from both live and lysed cells expressing Nluc cytosolically. Luminescence imaging further confirmed that cell permeable compounds inhibit both intracellular and extracellular Nluc, whereas less permeable compounds differentially inhibit extracellular Nluc and Nluc on the cell surface. The compounds displayed little to no toxicity to cells and high luciferase specificity, showing no activity against firefly luciferase or even the closely related NanoBit system. Looking forward, the structural motifs used to gain access to the Nluc surface can also be appended with other functional groups, and therefore interesting opportunities for developing assays based on relief-of-inhibition can be envisioned.
Collapse
Affiliation(s)
- Joel R. Walker
- Promega Biosciences LLC, 277
Granada Drive, San Luis Obispo, California 93401, United States
- Promega Corporation, 2800 Woods
Hollow Road, Madison, Wisconsin 53711-5399, United States
| | - Mary P. Hall
- Promega Biosciences LLC, 277
Granada Drive, San Luis Obispo, California 93401, United States
- Promega Corporation, 2800 Woods
Hollow Road, Madison, Wisconsin 53711-5399, United States
| | - Chad A. Zimprich
- Promega Biosciences LLC, 277
Granada Drive, San Luis Obispo, California 93401, United States
- Promega Corporation, 2800 Woods
Hollow Road, Madison, Wisconsin 53711-5399, United States
| | - Matthew B. Robers
- Promega Biosciences LLC, 277
Granada Drive, San Luis Obispo, California 93401, United States
- Promega Corporation, 2800 Woods
Hollow Road, Madison, Wisconsin 53711-5399, United States
| | - Sarah J. Duellman
- Promega Biosciences LLC, 277
Granada Drive, San Luis Obispo, California 93401, United States
- Promega Corporation, 2800 Woods
Hollow Road, Madison, Wisconsin 53711-5399, United States
| | - Thomas Machleidt
- Promega Biosciences LLC, 277
Granada Drive, San Luis Obispo, California 93401, United States
- Promega Corporation, 2800 Woods
Hollow Road, Madison, Wisconsin 53711-5399, United States
| | - Jacquelynn Rodriguez
- Promega Biosciences LLC, 277
Granada Drive, San Luis Obispo, California 93401, United States
- Promega Corporation, 2800 Woods
Hollow Road, Madison, Wisconsin 53711-5399, United States
| | - Wenhui Zhou
- Promega Biosciences LLC, 277
Granada Drive, San Luis Obispo, California 93401, United States
- Promega Corporation, 2800 Woods
Hollow Road, Madison, Wisconsin 53711-5399, United States
| |
Collapse
|
39
|
Marin-Kuan M, Fussell KC, Riederer N, Latado H, Serrant P, Mollergues J, Coulet M, Schilter B. Differentiating true androgen receptor inhibition from cytotoxicity-mediated reduction of reporter-gene transactivation in-vitro. Toxicol In Vitro 2017; 45:359-365. [PMID: 28377212 DOI: 10.1016/j.tiv.2017.03.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 03/17/2017] [Accepted: 03/30/2017] [Indexed: 12/22/2022]
Abstract
In vitro effect-based reporter assays are applied as biodetection tools designed to address nuclear receptor mediated-modulation. While such assays detect receptor modulating potential, cell viability needs to be addressed, preferably in the same well. Some assays circumvent this by co-transfecting a second constitutively-expressed marker gene or by multiplexing a cytotoxicity assay. Some assays, such as the CALUX®, lack this feature. The cytotoxic effects of unknown substances can confound in vitro assays, making the interpretation of results difficult and uncertain, particularly when assessing antagonistic activity. It's necessary to determine whether the cause of the reporter signal decrease is an antagonistic effect or a non-specific cytotoxic effect. To remedy this, we assessed the suitability of multiplexing a cell viability assay within the CALUX® transcriptional activation test for anti-androgenicity. Tests of both well-characterized anti-androgens and cytotoxic compounds demonstrated the suitability of this approach for discerning between the molecular mechanisms of action without altering the nuclear receptor assay; though some compounds were both cytotoxic and anti-androgenic. The optimized multiplexed assay was then applied to an uncharacterized set of polycyclic aromatic compounds. These results better characterized the mode of action and the classification of effects. Overall, the multiplexed protocol added value to CALUX test performance.
Collapse
Affiliation(s)
- Maricel Marin-Kuan
- Chemical Food Safety, Nestlé Research Centre, P.O. Box 44, CH-1000 Lausanne 26, Switzerland.
| | - Karma C Fussell
- Chemical Food Safety, Nestlé Research Centre, P.O. Box 44, CH-1000 Lausanne 26, Switzerland
| | - Nicolas Riederer
- Chemical Food Safety, Nestlé Research Centre, P.O. Box 44, CH-1000 Lausanne 26, Switzerland
| | - Helia Latado
- Chemical Food Safety, Nestlé Research Centre, P.O. Box 44, CH-1000 Lausanne 26, Switzerland
| | - Patrick Serrant
- Chemical Food Safety, Nestlé Research Centre, P.O. Box 44, CH-1000 Lausanne 26, Switzerland
| | - Julie Mollergues
- Chemical Food Safety, Nestlé Research Centre, P.O. Box 44, CH-1000 Lausanne 26, Switzerland
| | - Myriam Coulet
- Chemical Food Safety, Nestlé Research Centre, P.O. Box 44, CH-1000 Lausanne 26, Switzerland
| | - Benoit Schilter
- Chemical Food Safety, Nestlé Research Centre, P.O. Box 44, CH-1000 Lausanne 26, Switzerland
| |
Collapse
|
40
|
Leippe D, Sobol M, Vidugiris G, Cali JJ, Vidugiriene J. Bioluminescent Assays for Glucose and Glutamine Metabolism: High-Throughput Screening for Changes in Extracellular and Intracellular Metabolites. SLAS DISCOVERY 2016; 22:366-377. [DOI: 10.1177/1087057116675612] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cancer cell metabolism is a complex, dynamic network of regulated pathways. Interrogation of this network would benefit from rapid, sensitive techniques that are adaptable to high-throughput formats, facilitating novel compound screening. This requires assays that have minimal sample preparation and are adaptable to lower-volume 384-well formats and automation. Here we describe bioluminescent glucose, lactate, glutamine, and glutamate detection assays that are well suited for high-throughput analysis of two major metabolic pathways in cancer cells: glycolysis and glutaminolysis. The sensitivity (1–5 pmol/sample), broad linear range (0.1–100 µM), and wide dynamic range (>100-fold) are advantageous for measuring both extracellular and intracellular metabolites. Importantly, the assays incorporate rapid inactivation of endogenous enzymes, eliminating deproteinization steps required by other methods. Using ovarian cancer cell lines as a model system, the assays were used to monitor changes in glucose and glutamine consumption and lactate and glutamate secretion over time. Homogeneous formats of the lactate and glutamate assays were robust (Z′ = 0.6–0.9) and could be multiplexed with a real-time viability assay to generate internally controlled data. Screening a small-compound library with these assays resulted in the identification of both inhibitors and activators of lactate and glutamate production.
Collapse
Affiliation(s)
- Donna Leippe
- Research and Development, Promega Corporation, Madison, WI, USA
| | - Mary Sobol
- Research and Development, Promega Corporation, Madison, WI, USA
| | | | - James J. Cali
- Research and Development, Promega Corporation, Madison, WI, USA
| | | |
Collapse
|
41
|
A bioluminescent assay for measuring glucose uptake. Anal Biochem 2016; 505:43-50. [DOI: 10.1016/j.ab.2016.04.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 04/12/2016] [Accepted: 04/15/2016] [Indexed: 11/18/2022]
|
42
|
Kulesskiy E, Saarela J, Turunen L, Wennerberg K. Precision Cancer Medicine in the Acoustic Dispensing Era: Ex Vivo Primary Cell Drug Sensitivity Testing. ACTA ACUST UNITED AC 2015; 21:27-36. [PMID: 26721820 DOI: 10.1177/2211068215618869] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Indexed: 12/12/2022]
Abstract
Cancer therapy is increasingly becoming individualized, but there are also big gaps between the molecular knowledge of individual cancers we can generate today and what can be applied in the clinic. In an attempt to bridge this knowledge gap between cancer genetic and molecular profiling and clinically useful information, an individualized systems medicine program has been established at the Institute for Molecular Medicine Finland (FIMM), University of Helsinki, and the Helsinki University Hospital. Central to this program is drug sensitivity and resistance testing (DSRT), in which responses of primary cancer cells to a comprehensive clinical oncology and signal transduction drug collection are monitored. The drug sensitivity information is used with molecular profiling to establish hypotheses on individual cancer-selective targeting drug combinations and their predictive biomarkers, which can be explored in the clinic. Here, we describe how acoustic droplet ejection is enabling DSRT in our cancer individualized systems medicine program to (1) generate consistent but configurable assay-ready plates and determine how this affects data quality, (2) flexibly prepare drug combination testing plates, (3) dispense reagents and cells to the assay plates, and (4) perform ultra-miniaturized follow-up assays on the cells from DSRT plates.
Collapse
Affiliation(s)
- Evgeny Kulesskiy
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Finland
| | - Jani Saarela
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Finland
| | - Laura Turunen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Finland
| | - Krister Wennerberg
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Finland
| |
Collapse
|