1
|
Vitacolonna M, Bruch R, Schneider R, Jabs J, Hafner M, Reischl M, Rudolf R. A spheroid whole mount drug testing pipeline with machine-learning based image analysis identifies cell-type specific differences in drug efficacy on a single-cell level. BMC Cancer 2024; 24:1542. [PMID: 39696122 DOI: 10.1186/s12885-024-13329-9] [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: 05/02/2024] [Accepted: 12/11/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The growth and drug response of tumors are influenced by their stromal composition, both in vivo and 3D-cell culture models. Cell-type inherent features as well as mutual relationships between the different cell types in a tumor might affect drug susceptibility of the tumor as a whole and/or of its cell populations. However, a lack of single-cell procedures with sufficient detail has hampered the automated observation of cell-type-specific effects in three-dimensional stroma-tumor cell co-cultures. METHODS Here, we developed a high-content pipeline ranging from the setup of novel tumor-fibroblast spheroid co-cultures over optical tissue clearing, whole mount staining, and 3D confocal microscopy to optimized 3D-image segmentation and a 3D-deep-learning model to automate the analysis of a range of cell-type-specific processes, such as cell proliferation, apoptosis, necrosis, drug susceptibility, nuclear morphology, and cell density. RESULTS This demonstrated that co-cultures of KP-4 tumor cells with CCD-1137Sk fibroblasts exhibited a growth advantage compared to tumor cell mono-cultures, resulting in higher cell counts following cytostatic treatments with paclitaxel and doxorubicin. However, cell-type-specific single-cell analysis revealed that this apparent benefit of co-cultures was due to a higher resilience of fibroblasts against the drugs and did not indicate a higher drug resistance of the KP-4 cancer cells during co-culture. Conversely, cancer cells were partially even more susceptible in the presence of fibroblasts than in mono-cultures. CONCLUSION In summary, this underlines that a novel cell-type-specific single-cell analysis method can reveal critical insights regarding the mechanism of action of drug substances in three-dimensional cell culture models.
Collapse
Affiliation(s)
- Mario Vitacolonna
- CeMOS, Mannheim University of Applied Sciences, 68163, Mannheim, Germany.
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163, Mannheim, Germany.
| | - Roman Bruch
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, 76344, Eggen-stein-Leopoldshafen, Germany
| | | | - Julia Jabs
- Merck Healthcare KGaA, 64293, Darmstadt, Germany
| | - Mathias Hafner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163, Mannheim, Germany
- Institute of Medical Technology, Medical Faculty Mannheim of Heidelberg University, Mannheim University of Applied Sciences, 68167, Mannheim, Germany
| | - Markus Reischl
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, 76344, Eggen-stein-Leopoldshafen, Germany
| | - Rüdiger Rudolf
- CeMOS, Mannheim University of Applied Sciences, 68163, Mannheim, Germany
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163, Mannheim, Germany
| |
Collapse
|
2
|
Tavakoli N, Fong EJ, Coleman A, Huang YK, Bigger M, Doche ME, Kim S, Lenz HJ, Graham NA, Macklin P, Finley SD, Mumenthaler SM. Merging Metabolic Modeling and Imaging for Screening Therapeutic Targets in Colorectal Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.595756. [PMID: 38826317 PMCID: PMC11142224 DOI: 10.1101/2024.05.24.595756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Cancer-associated fibroblasts (CAFs) play a key role in metabolic reprogramming and are well-established contributors to drug resistance in colorectal cancer (CRC). To exploit this metabolic crosstalk, we integrated a systems biology approach that identified key metabolic targets in a data-driven method and validated them experimentally. This process involved a novel machine learning-based method to computationally screen, in a high-throughput manner, the effects of enzyme perturbations predicted by a computational model of CRC metabolism. This approach reveals the network-wide effects of metabolic perturbations. Our results highlighted hexokinase (HK) as a crucial target, which subsequently became our focus for experimental validation using patient-derived tumor organoids (PDTOs). Through metabolic imaging and viability assays, we found that PDTOs cultured in CAF-conditioned media exhibited increased sensitivity to HK inhibition, confirming the model predictions. Our approach emphasizes the critical role of integrating computational and experimental techniques in exploring and exploiting CRC-CAF crosstalk.
Collapse
|
3
|
Yoshihara N, Lopes M, Santos I, Kopke B, Almeida C, Araújo J, Fechine PBA, Santos-Oliveira R, Sant'Anna C. Graphitic carbon nitride as a novel anticancer agent: potential mechanisms and efficacy in prostate cancer and glioblastoma treatment. Biomater Sci 2024; 12:5547-5561. [PMID: 39292186 DOI: 10.1039/d4bm01025f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Carbon-derived compounds are gaining traction in the scientific community because of their unique properties, such as conductivity and strength, and promising innovations in technology and medicine. Graphitic nitride carbon (g-C3N4) stands out among these compounds because of its potential in antitumor therapies. This study aimed to assess g-C3N4's antitumor potential and cytotoxic mechanisms. Prostate cancer (DU-145) and glioblastoma (U87) cell lines were used to evaluate antitumor effects, whereas RAW 264.7 and HFF-1 non-tumor cells were used for selectivity evaluation. The synthesized g-C3N4 particles underwent comprehensive characterization, including the assessment of particle size, morphology, and oxygen content, employing various techniques, such as X-ray diffraction, X-ray photoelectron spectroscopy, scanning electron microscopy, energy dispersive X-ray spectroscopy, transmission electron microscopy, and atomic force microscopy. The results indicated that g-C3N4 significantly affected tumor cell proliferation and viability, exhibiting high cytotoxicity within 48 h. In non-tumor cells, minimal effects on proliferation were observed, except for damage to the cell membranes of RAW 264.7 cells. Moreover, g-C3N4 changed the cell morphology and ultrastructure, affecting cell migration in U87 cells and potentially enhancing migration in RAW 264.7 cells. Biochemical assays in Balb/C mice revealed alterations in alanine aminotransferase, aspartate aminotransferase, and amylase levels. In conclusion, g-C3N4 demonstrated promising antitumor effects with minimal toxicity to non-tumor cells, suggesting its potential in neoplasm treatment.
Collapse
Affiliation(s)
- Natalia Yoshihara
- National Institute of Metrology, Quality and Technology, Eukaryotic Cell Biology Laboratory, Duque de Caxias-RJ, 24250020, Brazil.
| | - Michelle Lopes
- National Institute of Metrology, Quality and Technology, Eukaryotic Cell Biology Laboratory, Duque de Caxias-RJ, 24250020, Brazil.
| | - Isabel Santos
- National Institute of Metrology, Quality and Technology, Eukaryotic Cell Biology Laboratory, Duque de Caxias-RJ, 24250020, Brazil.
| | - Beatriz Kopke
- National Institute of Metrology, Quality and Technology, Eukaryotic Cell Biology Laboratory, Duque de Caxias-RJ, 24250020, Brazil.
| | - Clara Almeida
- National Institute of Metrology, Quality and Technology, Laboratory of Microscopy Dimat, Duque de Caxias-RJ, 24250020, Brazil
| | - Joyce Araújo
- National Institute of Metrology, Quality and Technology, Laboratory of Microscopy Dimat, Duque de Caxias-RJ, 24250020, Brazil
| | - Pierre B A Fechine
- Group of Chemistry of Advanced Materials (GQMat)-Department of Analytical Chemistry and Physical-Chemistry, Federal University of Ceará, Fortaleza-CE, 451-970, Brazil
| | - Ralph Santos-Oliveira
- Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Laboratory of Nanoradiopharmacy and Synthesis of New, Brazil
- Radiopharmaceuticals, Rio de Janeiro-RJ, 21941906, Brazil
- Rio de Janeiro State University, Laboratory of Nanoradiopharmaceuticals, Rio de Janeiro, 23070200, Brazil
| | - Celso Sant'Anna
- National Institute of Metrology, Quality and Technology, Eukaryotic Cell Biology Laboratory, Duque de Caxias-RJ, 24250020, Brazil.
| |
Collapse
|
4
|
Ma Y, Shih CH, Cheng J, Chen HC, Wang LJ, Tan Y, Chiu YC, Chen YC. High-Throughput Empirical and Virtual Screening to Discover Novel Inhibitors of Polyploid Giant Cancer Cells in Breast Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614522. [PMID: 39386568 PMCID: PMC11463688 DOI: 10.1101/2024.09.23.614522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Therapy resistance in breast cancer is increasingly attributed to polyploid giant cancer cells (PGCCs), which arise through whole-genome doubling and exhibit heightened resilience to standard treatments. Characterized by enlarged nuclei and increased DNA content, these cells tend to be dormant under therapeutic stress, driving disease relapse. Despite their critical role in resistance, strategies to effectively target PGCCs are limited, largely due to the lack of high-throughput methods for assessing their viability. Traditional assays lack the sensitivity needed to detect PGCC-specific elimination, prompting the development of novel approaches. To address this challenge, we developed a high-throughput single-cell morphological analysis workflow designed to differentiate compounds that selectively inhibit non-PGCCs, PGCCs, or both. Using this method, we screened a library of 2,726 FDA Phase 1-approved drugs, identifying promising anti-PGCC candidates, including proteasome inhibitors, FOXM1, CHK, and macrocyclic lactones. Notably, RNA-Seq analysis of cells treated with the macrocyclic lactone Pyronaridine revealed AXL inhibition as a potential strategy for targeting PGCCs. Although our single-cell morphological analysis pipeline is powerful, empirically testing all existing compounds is impractical and inefficient. To overcome this limitation, we trained a machine learning model to predict anti-PGCC efficacy in silico, integrating chemical fingerprints and compound descriptions from prior publications and databases. The model demonstrated a high correlation with experimental outcomes and predicted efficacious compounds in an expanded library of over 6,000 drugs. Among the top-ranked predictions, we experimentally validated two compounds as potent PGCC inhibitors. These findings underscore the synergistic potential of integrating high-throughput empirical screening with machine learning-based virtual screening to accelerate the discovery of novel therapies, particularly for targeting therapy-resistant PGCCs in breast cancer.
Collapse
Affiliation(s)
- Yushu Ma
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15260, USA
| | - Chien-Hung Shih
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA
| | - Jinxiong Cheng
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O’Hara Street, Pittsburgh, PA 15260, USA
| | - Hsiao-Chun Chen
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15260, USA
| | - Li-Ju Wang
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA
| | - Yanhao Tan
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA
- Division of Malignant Hematology and Medical Oncology, Department of Medicine, University of Pittsburgh, 5150 Centre Avenue, Pittsburgh, PA 15232, USA
| | - Yu-Chiao Chiu
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15260, USA
- Division of Malignant Hematology and Medical Oncology, Department of Medicine, University of Pittsburgh, 5150 Centre Avenue, Pittsburgh, PA 15232, USA
- CMU-Pitt Ph.D. Program in Computational Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15260, USA
| | - Yu-Chih Chen
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15260, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O’Hara Street, Pittsburgh, PA 15260, USA
- CMU-Pitt Ph.D. Program in Computational Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15260, USA
| |
Collapse
|
5
|
Ha DP, Shin WJ, Liu Z, Doche ME, Lau R, Leli NM, Conn CS, Russo M, Lorenzato A, Koumenis C, Yu M, Mumenthaler SM, Lee AS. Targeting stress induction of GRP78 by cardiac glycoside oleandrin dually suppresses cancer and COVID-19. Cell Biosci 2024; 14:115. [PMID: 39238058 PMCID: PMC11378597 DOI: 10.1186/s13578-024-01297-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Despite recent therapeutic advances, combating cancer resistance remains a formidable challenge. The 78-kilodalton glucose-regulated protein (GRP78), a key stress-inducible endoplasmic reticulum (ER) chaperone, plays a crucial role in both cancer cell survival and stress adaptation. GRP78 is also upregulated during SARS-CoV-2 infection and acts as a critical host factor. Recently, we discovered cardiac glycosides (CGs) as novel suppressors of GRP78 stress induction through a high-throughput screen of clinically relevant compound libraries. This study aims to test the possibility that agents capable of blocking stress induction of GRP78 could dually suppress cancer and COVID-19. RESULTS Here we report that oleandrin (OLN), is the most potent among the CGs in inhibiting acute stress induction of total GRP78, which also results in reduced cell surface and nuclear forms of GRP78 in stressed cells. The inhibition of stress induction of GRP78 is at the post-transcriptional level, independent of protein degradation and autophagy and may involve translational control as OLN blocks stress-induced loading of ribosomes onto GRP78 mRNAs. Moreover, the human Na+/K+-ATPase α3 isoform is critical for OLN suppression of GRP78 stress induction. OLN, in nanomolar range, enhances apoptosis, sensitizes colorectal cancer cells to chemotherapeutic agents, and reduces the viability of patient-derived colon cancer organoids. Likewise, OLN, suppresses GRP78 expression and impedes tumor growth in an orthotopic breast cancer xenograft model. Furthermore, OLN blocks infection by SARS-CoV-2 and its variants and enhances existing anti-viral therapies. Notably, GRP78 overexpression mitigates OLN-mediated cancer cell apoptotic onset and suppression of virus release. CONCLUSION Our findings validate GRP78 as a target of OLN anti-cancer and anti-viral activities. These proof-of-principle studies support further investigation of OLN as a readily accessible compound to dually combat cancer and COVID-19.
Collapse
Affiliation(s)
- Dat P Ha
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Woo-Jin Shin
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, 34987, USA
- Department of Cancer Biology, Infection Biology Program, and Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44106, USA
| | - Ze Liu
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Michael E Doche
- Ellison Institute of Technology, Los Angeles, CA, 90064, USA
| | - Roy Lau
- Ellison Institute of Technology, Los Angeles, CA, 90064, USA
| | - Nektaria Maria Leli
- Department of Radiation Oncology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Crystal S Conn
- Department of Radiation Oncology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mariangela Russo
- Dipartimento di Oncologia, Molecular Biotechnology Center, Università di Torino, Turin, Italy
| | - Annalisa Lorenzato
- Dipartimento di Oncologia, Molecular Biotechnology Center, Università di Torino, Turin, Italy
| | - Constantinos Koumenis
- Department of Radiation Oncology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Min Yu
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Shannon M Mumenthaler
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Ellison Institute of Technology, Los Angeles, CA, 90064, USA
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Amy S Lee
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
6
|
Garcia-Fossa F, Moraes-Lacerda T, Rodrigues-da-Silva M, Diaz-Rohrer B, Singh S, Carpenter AE, Cimini BA, de Jesus MB. Live Cell Painting: image-based profiling in live cells using Acridine Orange. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.28.610144. [PMID: 39257795 PMCID: PMC11383656 DOI: 10.1101/2024.08.28.610144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Image-based profiling has been used to analyze cell health, drug mechanism of action, CRISPR-edited cells, and overall cytotoxicity. Cell Painting is a broadly used image-based assay that uses morphological features to capture how cells respond to treatments. However, this method requires cell fixation for staining, which prevents examining live cells. To address this limitation, here we present Live Cell Painting (LCP), a high-content method based on Acridine orange, a metachromatic dye that labels different organelles and cellular structures. We began by showing that LCP can be applied to follow acidic vesicle redistribution of cells exposed to acidic vesicles inhibitors. Next, we show that LCP can identify subtle changes in cells exposed to silver nanoparticles that are not detected by techniques such as MTT assay. In drug treatments, LCP was helpful in assessing the dose-response relationship and creating profiles that allow clustering of drugs that cause liver injury. Here, we present an affordable and easy-to-use image-based assay capable of assessing overall cell health and showing promise for use in various applications such as assessing drugs and nanoparticles. We envisage the use of Live Cell Painting as an initial screening of overall cell health while providing insights into new biological questions.
Collapse
|
7
|
Gourmet L, Walker-Samuel S, Mallick P. Examination of the role of mutualism in immune evasion. Front Oncol 2024; 14:1406744. [PMID: 38779085 PMCID: PMC11109368 DOI: 10.3389/fonc.2024.1406744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Though the earliest stages of oncogenesis, post initiation, are not well understood, it is generally appreciated that a successful transition from a collection of dysregulated cells to an aggressive tumour requires complex ecological interactions between cancer cells and their environment. One key component of tumorigenesis is immune evasion. To investigate the interplay amongst the ecological behaviour of mutualism and immune evasion, we used a computational simulation framework. Sensitivity analyses of the growth of a virtual tumour implemented as a 2D-hexagonal lattice model suggests tumour survival depends on the interplay between growth rates, mutualism and immune evasion. In 60% of simulations, cancer clones with low growth rates, but exhibiting mutualism were able to evade the immune system and continue progressing suggesting that tumours with equivalent growth rates and no mutualism are more likely to be eliminated than tumours with mutualism. Tumours with faster growth rates showed a lower dependence upon mutualism for progression. Geostatistical analysis showed decreased spatial heterogeneity over time for polyclonal tumours with a high division rate. Overall, these results suggest that in slow growing tumours, mutualism is critical for early tumorigenesis.
Collapse
Affiliation(s)
- Lucie Gourmet
- Centre for Computational Medicine, Division of Medicine, University College London, London, United Kingdom
| | - Simon Walker-Samuel
- Centre for Computational Medicine, Division of Medicine, University College London, London, United Kingdom
| | - Parag Mallick
- Canary Center for Cancer Early Detection, Stanford University, Palo Alto, CA, United States
| |
Collapse
|
8
|
Subramani C, Sharma G, Chaira T, Barman TK. High content screening strategies for large-scale compound libraries with a focus on high-containment viruses. Antiviral Res 2024; 221:105764. [PMID: 38008193 DOI: 10.1016/j.antiviral.2023.105764] [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: 09/11/2023] [Revised: 11/14/2023] [Accepted: 11/19/2023] [Indexed: 11/28/2023]
Abstract
A majority of viral diseases do not have FDA-approved drugs. The recent outbreaks caused by SARS-CoV-2, monkeypox, and Sudan ebolavirus have exposed the critical need for rapid screening and identification of antiviral compounds against emerging/re-emerging viral pathogens. A high-content screening (HCS) platform is becoming an essential part of the drug discovery process, thanks to developments in image acquisition and analysis. While HCS has several advantages, its full potential has not been realized in antiviral drug discovery compared to conventional drug screening approaches, such as fluorescence or luminescence-based microplate assays. Therefore, this review aims to summarize HCS workflow, strategies, and developments in image-based drug screening, focusing on high-containment viruses.
Collapse
Affiliation(s)
- Chandru Subramani
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA; Galveston National Laboratory, Galveston, TX, USA
| | - Ghanshyam Sharma
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Tridib Chaira
- Department of Pharmacology, SGT University, Gurugram, Haryana, India
| | - Tarani Kanta Barman
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA; Galveston National Laboratory, Galveston, TX, USA.
| |
Collapse
|
9
|
Zhou M, Ma Y, Chiang CC, Rock EC, Butler SC, Anne R, Yatsenko S, Gong Y, Chen YC. Single-cell morphological and transcriptome analysis unveil inhibitors of polyploid giant breast cancer cells in vitro. Commun Biol 2023; 6:1301. [PMID: 38129519 PMCID: PMC10739852 DOI: 10.1038/s42003-023-05674-5] [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: 06/06/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
Considerable evidence suggests that breast cancer therapeutic resistance and relapse can be driven by polyploid giant cancer cells (PGCCs). The number of PGCCs increases with the stages of disease and therapeutic stress. Given the importance of PGCCs, it remains challenging to eradicate them. To discover effective anti-PGCC compounds, there is an unmet need to rapidly distinguish compounds that kill non-PGCCs, PGCCs, or both. Here, we establish a single-cell morphological analysis pipeline with a high throughput and great precision to characterize dynamics of individual cells. In this manner, we screen a library to identify promising compounds that inhibit all cancer cells or only PGCCs (e.g., regulators of HDAC, proteasome, and ferroptosis). Additionally, we perform scRNA-Seq to reveal altered cell cycle, metabolism, and ferroptosis sensitivity in breast PGCCs. The combination of single-cell morphological and molecular investigation reveals promising anti-PGCC strategies for breast cancer treatment and other malignancies.
Collapse
Affiliation(s)
- Mengli Zhou
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA, 15260, USA
- Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yushu Ma
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA, 15260, USA
| | - Chun-Cheng Chiang
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA, 15260, USA
| | - Edwin C Rock
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA, 15260, USA
| | - Samuel Charles Butler
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
| | - Rajiv Anne
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA, 15260, USA
| | - Svetlana Yatsenko
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
- Magee Womens Research Institute, Pittsburgh, PA, USA
| | - Yinan Gong
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Yu-Chih Chen
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA, 15232, USA.
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA, 15260, USA.
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA, 15260, USA.
- CMU-Pitt Ph.D. Program in Computational Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
10
|
Stossi F, Singh PK, Safari K, Marini M, Labate D, Mancini MA. High throughput microscopy and single cell phenotypic image-based analysis in toxicology and drug discovery. Biochem Pharmacol 2023; 216:115770. [PMID: 37660829 DOI: 10.1016/j.bcp.2023.115770] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/05/2023]
Abstract
Measuring single cell responses to the universe of chemicals (drugs, natural products, environmental toxicants etc.) is of paramount importance to human health as phenotypic variability in sensing stimuli is a hallmark of biology that is considered during high throughput screening. One of the ways to approach this problem is via high throughput, microscopy-based assays coupled with multi-dimensional single cell analysis methods. Here, we will summarize some of the efforts in this vast and growing field, focusing on phenotypic screens (e.g., Cell Painting), single cell analytics and quality control, with particular attention to environmental toxicology and drug screening. We will discuss advantages and limitations of high throughput assays with various end points and levels of complexity.
Collapse
Affiliation(s)
- Fabio Stossi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA.
| | - Pankaj K Singh
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA; Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Kazem Safari
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA; Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Michela Marini
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA; Department of Mathematics, University of Houston, Houston, TX, USA
| | - Demetrio Labate
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA; Department of Mathematics, University of Houston, Houston, TX, USA
| | - Michael A Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA; Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| |
Collapse
|
11
|
Guzzetti S, Morentin Gutierrez P. An integrated modelling approach for targeted degradation: insights on optimization, data requirements and PKPD predictions from semi- or fully-mechanistic models and exact steady state solutions. J Pharmacokinet Pharmacodyn 2023; 50:327-349. [PMID: 37120680 PMCID: PMC10460745 DOI: 10.1007/s10928-023-09857-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/28/2023] [Indexed: 05/01/2023]
Abstract
The value of an integrated mathematical modelling approach for protein degraders which combines the benefits of traditional turnover models and fully mechanistic models is presented. Firstly, we show how exact solutions of the mechanistic models of monovalent and bivalent degraders can provide insight on the role of each system parameter in driving the pharmacological response. We show how on/off binding rates and degradation rates are related to potency and maximal effect of monovalent degraders, and how such relationship can be used to suggest a compound optimization strategy. Even convoluted exact steady state solutions for bivalent degraders provide insight on the type of observations required to ensure the predictive capacity of a mechanistic approach. Specifically for PROTACs, the structure of the exact steady state solution suggests that the total remaining target at steady state, which is easily accessible experimentally, is insufficient to reconstruct the state of the whole system at equilibrium and observations on different species (such as binary/ternary complexes) are necessary. Secondly, global sensitivity analysis of fully mechanistic models for PROTACs suggests that both target and ligase baselines (actually, their ratio) are the major sources of variability in the response of non-cooperative systems, which speaks to the importance of characterizing their distribution in the target patient population. Finally, we propose a pragmatic modelling approach which incorporates the insights generated with fully mechanistic models into simpler turnover models to improve their predictive ability, hence enabling acceleration of drug discovery programs and increased probability of success in the clinic.
Collapse
Affiliation(s)
- Sofia Guzzetti
- DMPK, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | | |
Collapse
|
12
|
Rosenthal MR, Ng CL. High-content imaging as a tool to quantify and characterize malaria parasites. CELL REPORTS METHODS 2023; 3:100516. [PMID: 37533635 PMCID: PMC10391350 DOI: 10.1016/j.crmeth.2023.100516] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 01/18/2023] [Accepted: 06/02/2023] [Indexed: 08/04/2023]
Abstract
In 2021, Plasmodium falciparum was responsible for 619,000 reported malaria-related deaths. Resistance has been detected to every clinically used antimalarial, urging the development of novel antimalarials with uncompromised mechanisms of actions. High-content imaging allows researchers to collect and quantify numerous phenotypic properties at the single-cell level, and machine learning-based approaches enable automated classification and clustering of cell populations. By combining these technologies, we developed a method capable of robustly differentiating and quantifying P. falciparum asexual blood stages. These phenotypic properties also allow for the quantification of changes in parasite morphology. Here, we demonstrate that our analysis can be used to quantify schizont nuclei, a phenotype that previously had to be enumerated manually. By monitoring stage progression and quantifying parasite phenotypes, our method can discern stage specificity of new compounds, thus providing insight into the compound's mode of action.
Collapse
Affiliation(s)
- Melissa R. Rosenthal
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Caroline L. Ng
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Global Center for Health Security, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Biology, University of Omaha, Omaha, NE 68182, USA
| |
Collapse
|
13
|
André O, Kumra Ahnlide J, Norlin N, Swaminathan V, Nordenfelt P. Data-driven microscopy allows for automated context-specific acquisition of high-fidelity image data. CELL REPORTS METHODS 2023; 3:100419. [PMID: 37056378 PMCID: PMC10088093 DOI: 10.1016/j.crmeth.2023.100419] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/20/2022] [Accepted: 02/10/2023] [Indexed: 04/15/2023]
Abstract
Light microscopy is a powerful single-cell technique that allows for quantitative spatial information at subcellular resolution. However, unlike flow cytometry and single-cell sequencing techniques, microscopy has issues achieving high-quality population-wide sample characterization while maintaining high resolution. Here, we present a general framework, data-driven microscopy (DDM) that uses real-time population-wide object characterization to enable data-driven high-fidelity imaging of relevant phenotypes based on the population context. DDM combines data-independent and data-dependent steps to synergistically enhance data acquired using different imaging modalities. As a proof of concept, we develop and apply DDM with plugins for improved high-content screening and live adaptive microscopy for cell migration and infection studies that capture events of interest, rare or common, with high precision and resolution. We propose that DDM can reduce human bias, increase reproducibility, and place single-cell characteristics in the context of the sample population when interpreting microscopy data, leading to an increase in overall data fidelity.
Collapse
Affiliation(s)
- Oscar André
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | | | - Nils Norlin
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Lund University Bioimaging Centre, Lund, Sweden
| | - Vinay Swaminathan
- Department of Clinical Sciences, Wallenberg Centre for Molecular Medicine, Division of Oncology, Lund University, Lund, Sweden
| | - Pontus Nordenfelt
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| |
Collapse
|
14
|
Köhn-Luque A, Myklebust EM, Tadele DS, Giliberto M, Schmiester L, Noory J, Harivel E, Arsenteva P, Mumenthaler SM, Schjesvold F, Taskén K, Enserink JM, Leder K, Frigessi A, Foo J. Phenotypic deconvolution in heterogeneous cancer cell populations using drug-screening data. CELL REPORTS METHODS 2023; 3:100417. [PMID: 37056380 PMCID: PMC10088094 DOI: 10.1016/j.crmeth.2023.100417] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 12/10/2022] [Accepted: 02/08/2023] [Indexed: 03/08/2023]
Abstract
Tumor heterogeneity is an important driver of treatment failure in cancer since therapies often select for drug-tolerant or drug-resistant cellular subpopulations that drive tumor growth and recurrence. Profiling the drug-response heterogeneity of tumor samples using traditional genomic deconvolution methods has yielded limited results, due in part to the imperfect mapping between genomic variation and functional characteristics. Here, we leverage mechanistic population modeling to develop a statistical framework for profiling phenotypic heterogeneity from standard drug-screen data on bulk tumor samples. This method, called PhenoPop, reliably identifies tumor subpopulations exhibiting differential drug responses and estimates their drug sensitivities and frequencies within the bulk population. We apply PhenoPop to synthetically generated cell populations, mixed cell-line experiments, and multiple myeloma patient samples and demonstrate how it can provide individualized predictions of tumor growth under candidate therapies. This methodology can also be applied to deconvolution problems in a variety of biological settings beyond cancer drug response.
Collapse
Affiliation(s)
- Alvaro Köhn-Luque
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
| | - Even Moa Myklebust
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
| | - Dagim Shiferaw Tadele
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318 Oslo, Norway
- Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, OH 44131, USA
| | - Mariaserena Giliberto
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
- KG Jebsen Center for B-Cell Malignancies, Institute for Clinical Medicine, University of Oslo, 0450 Oslo, Norway
| | - Leonard Schmiester
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
| | - Jasmine Noory
- Institute for Mathematics and its Applications, School of Mathematics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Elise Harivel
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- ENSTA, Institut Polytechnique de Paris, Palaiseau, 91120 Paris, France
| | - Polina Arsenteva
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Institut de Matématiques de Bourgogne, Universite de Bourgogne, Dijon Cedex, 21078 Dijon, France
| | - Shannon M. Mumenthaler
- Lawrence J. Ellison Institute for Transformative Medicine, Los Angeles, CA 90064, USA
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Department of Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Fredrik Schjesvold
- KG Jebsen Center for B-Cell Malignancies, Institute for Clinical Medicine, University of Oslo, 0450 Oslo, Norway
- Oslo Myeloma Center, Department of Hematology, Oslo University Hospital, 0450 Oslo, Norway
| | - Kjetil Taskén
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
- KG Jebsen Center for B-Cell Malignancies, Institute for Clinical Medicine, University of Oslo, 0450 Oslo, Norway
| | - Jorrit M. Enserink
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318 Oslo, Norway
- Section for Biochemistry and Molecular Biology, Faculty of Mathematics and Natural Sciences, University of Oslo, 0037 Oslo, Norway
| | - Kevin Leder
- College of Science and Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Arnoldo Frigessi
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, 0372 Oslo, Norway
| | - Jasmine Foo
- Institute for Mathematics and its Applications, School of Mathematics, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
15
|
Guzman G, Creek C, Farley S, Tafesse FG. Genetic Tools for Studying the Roles of Sphingolipids in Viral Infections. Methods Mol Biol 2022; 2610:1-16. [PMID: 36534277 DOI: 10.1007/978-1-0716-2895-9_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Sphingolipids are a critical family of membrane lipids with diverse functions in eukaryotic cells, and a growing body of literature supports that these lipids play essential roles during the lifecycles of viruses. While small molecule inhibitors of sphingolipid synthesis and metabolism are widely used, the advent of CRISPR-based genomic editing techniques allows for nuanced exploration into the manners in which sphingolipids influence various stages of viral infections. Here we describe some of these critical considerations needed in designing studies utilizing genomic editing techniques for manipulating the sphingolipid metabolic pathway, as well as the current body of literature regarding how viruses depend on the products of this pathway. Here, we highlight the ways in which sphingolipids affect viruses as these pathogens interact with and influence their host cell and describe some of the many open questions remaining in the field.
Collapse
Affiliation(s)
- Gaelen Guzman
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Cameron Creek
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Scotland Farley
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Fikadu G Tafesse
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
16
|
Kamiya G, Kitada N, Maki S, Kim SB. Multiplex quadruple bioluminescent assay system. Sci Rep 2022; 12:17485. [PMID: 36261452 PMCID: PMC9581999 DOI: 10.1038/s41598-022-20468-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 09/13/2022] [Indexed: 01/12/2023] Open
Abstract
Bioluminescence (BL) is unique cold body radiation of light, generated by luciferin-luciferase reactions and commonly used in various bioassays and molecular imaging. However, most of the peak emissions of BL populate the blue-yellow region and have broad spectral bandwidths and thus superimpose each other, causing optical cross-leakages in multiplex assays. This study synthesized a new series of coelenterazine (CTZ) analogues, named K-series, that selectively illuminates marine luciferases with unique, blue-shifted spectral properties. The optical property and specificity of the K-series CTZ analogues were characterized by marine luciferases, with K2 and K5 found to specifically luminesce with ALuc- and RLuc-series marine luciferases, respectively. The results confirmed that the luciferase specificity and color variation of the CTZ analogues minimize the cross-leakages of BL signals and enable high-throughput screening of specific ligands in the mixture. The specificity and color variation of the substrates were further tailored to marine luciferases (or single-chain bioluminescent probes) to create a multiplex quadruple assay system with four integrated, single-chain bioluminescent probes, with each probe designed to selectively luminesce only with its specific ligand (first authentication) and a specific CTZ analogue (second authentication). This unique multiplex quadruple bioluminescent assay system is an efficient optical platform for specific and high-throughput imaging of multiple optical markers in bioassays without optical cross-leakages.
Collapse
Affiliation(s)
- Genta Kamiya
- grid.266298.10000 0000 9271 9936Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, Chofu, Tokyo 182-8585 Japan
| | - Nobuo Kitada
- grid.266298.10000 0000 9271 9936Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, Chofu, Tokyo 182-8585 Japan
| | - Shojiro Maki
- grid.266298.10000 0000 9271 9936Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, Chofu, Tokyo 182-8585 Japan
| | - Sung Bae Kim
- grid.208504.b0000 0001 2230 7538Research Institute for Environmental Management Technology, National Institute of Advanced Industrial Science and Technology (AIST), 16-1 Onogawa, Tsukuba, 305-8569 Japan
| |
Collapse
|
17
|
Usher I, Ligammari L, Ahrabi S, Hepburn E, Connolly C, Bond GL, Flanagan AM, Cottone L. Optimizing CRISPR/Cas9 Editing of Repetitive Single Nucleotide Variants. Front Genome Ed 2022; 4:932434. [PMID: 35865001 PMCID: PMC9294353 DOI: 10.3389/fgeed.2022.932434] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
CRISPR/Cas9, base editors and prime editors comprise the contemporary genome editing toolbox. Many studies have optimized the use of CRISPR/Cas9, as the original CRISPR genome editing system, in substituting single nucleotides by homology directed repair (HDR), although this remains challenging. Studies describing modifications that improve editing efficiency fall short of isolating clonal cell lines or have not been validated for challenging loci or cell models. We present data from 95 transfections using a colony forming and an immortalized cell line comparing the effect on editing efficiency of donor template modifications, concentration of components, HDR enhancing agents and cold shock. We found that in silico predictions of guide RNA efficiency correlated poorly withactivity in cells. Using NGS and ddPCR we detected editing efficiencies of 5–12% in the transfected populations which fell to 1% on clonal cell line isolation. Our data demonstrate the variability of CRISPR efficiency by cell model, target locus and other factors. Successful genome editing requires a comparison of systems and modifications to develop the optimal protocol for the cell model and locus. We describe the steps in this process in a flowchart for those embarking on genome editing using any system and incorporate validated HDR-boosting modifications for those using CRISPR/Cas9.
Collapse
Affiliation(s)
- Inga Usher
- Department of Pathology (Research), UCL Cancer Institute, University College London, London, United Kingdom
| | - Lorena Ligammari
- Department of Pathology (Research), UCL Cancer Institute, University College London, London, United Kingdom
| | - Sara Ahrabi
- Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Emily Hepburn
- UCL Medical School, University College London, London, United Kingdom
| | - Calum Connolly
- UCL Medical School, University College London, London, United Kingdom
| | - Gareth L. Bond
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Adrienne M. Flanagan
- Department of Pathology (Research), UCL Cancer Institute, University College London, London, United Kingdom
- Department of Histopathology, Royal National Orthopaedic Hospital, London, United Kingdom
| | - Lucia Cottone
- Department of Pathology (Research), UCL Cancer Institute, University College London, London, United Kingdom
- *Correspondence: Lucia Cottone,
| |
Collapse
|
18
|
Lieschke E, Wang Z, Chang C, Weeden CE, Kelly GL, Strasser A. Flow cytometric single cell-based assay to simultaneously detect cell death, cell cycling, DNA content and cell senescence. Cell Death Differ 2022; 29:1004-1012. [PMID: 35264779 PMCID: PMC9091206 DOI: 10.1038/s41418-022-00964-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 11/09/2022] Open
Abstract
Cell death, cell cycle arrest and cellular senescence are three distinct cellular responses that can be induced by oncogene activation and diverse anti-cancer agents, and this often requires the action of the tumour suppressor TP53. Within a cell population, or even within an individual cell, these processes are not necessarily mutually exclusive. It is therefore important to measure all these processes simultaneously. However, current assays generally visualise only one or at best two responses, often only detecting the dominant one. Here, we present a novel flow cytometric assay that allows simultaneous assessment of cell viability and cell cycling through measurement of DNA content and DNA synthesis, and markers of cell senescence at the single cell level. We demonstrate that this assay can be performed on both human and murine cells, that are either cancerous or non-transformed, and can help to dissect complex cell fate decisions. We believe that this experimental tool will be useful for the study of diverse biological processes.
Collapse
|
19
|
Spiller ER, Ung N, Kim S, Patsch K, Lau R, Strelez C, Doshi C, Choung S, Choi B, Juarez Rosales EF, Lenz HJ, Matasci N, Mumenthaler SM. Imaging-Based Machine Learning Analysis of Patient-Derived Tumor Organoid Drug Response. Front Oncol 2022; 11:771173. [PMID: 34993134 PMCID: PMC8724556 DOI: 10.3389/fonc.2021.771173] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/02/2021] [Indexed: 12/12/2022] Open
Abstract
Three-quarters of compounds that enter clinical trials fail to make it to market due to safety or efficacy concerns. This statistic strongly suggests a need for better screening methods that result in improved translatability of compounds during the preclinical testing period. Patient-derived organoids have been touted as a promising 3D preclinical model system to impact the drug discovery pipeline, particularly in oncology. However, assessing drug efficacy in such models poses its own set of challenges, and traditional cell viability readouts fail to leverage some of the advantages that the organoid systems provide. Consequently, phenotypically evaluating complex 3D cell culture models remains difficult due to intra- and inter-patient organoid size differences, cellular heterogeneities, and temporal response dynamics. Here, we present an image-based high-content assay that provides object level information on 3D patient-derived tumor organoids without the need for vital dyes. Leveraging computer vision, we segment and define organoids as independent regions of interest and obtain morphometric and textural information per organoid. By acquiring brightfield images at different timepoints in a robust, non-destructive manner, we can track the dynamic response of individual organoids to various drugs. Furthermore, to simplify the analysis of the resulting large, complex data files, we developed a web-based data visualization tool, the Organoizer, that is available for public use. Our work demonstrates the feasibility and utility of using imaging, computer vision and machine learning to determine the vital status of individual patient-derived organoids without relying upon vital dyes, thus taking advantage of the characteristics offered by this preclinical model system.
Collapse
Affiliation(s)
- Erin R Spiller
- Lawrence J. Ellison Institute for Transformative Medicine of USC, Los Angeles, CA, United States
| | - Nolan Ung
- Lawrence J. Ellison Institute for Transformative Medicine of USC, Los Angeles, CA, United States
| | - Seungil Kim
- Lawrence J. Ellison Institute for Transformative Medicine of USC, Los Angeles, CA, United States
| | - Katherin Patsch
- Lawrence J. Ellison Institute for Transformative Medicine of USC, Los Angeles, CA, United States
| | - Roy Lau
- Lawrence J. Ellison Institute for Transformative Medicine of USC, Los Angeles, CA, United States
| | - Carly Strelez
- Lawrence J. Ellison Institute for Transformative Medicine of USC, Los Angeles, CA, United States
| | - Chirag Doshi
- Lawrence J. Ellison Institute for Transformative Medicine of USC, Los Angeles, CA, United States
| | - Sarah Choung
- Lawrence J. Ellison Institute for Transformative Medicine of USC, Los Angeles, CA, United States
| | - Brandon Choi
- Lawrence J. Ellison Institute for Transformative Medicine of USC, Los Angeles, CA, United States
| | - Edwin Francisco Juarez Rosales
- Lawrence J. Ellison Institute for Transformative Medicine of USC, Los Angeles, CA, United States.,Department of Medicine, University of California San Diego, La Jolla, CA, United States
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Naim Matasci
- Lawrence J. Ellison Institute for Transformative Medicine of USC, Los Angeles, CA, United States
| | - Shannon M Mumenthaler
- Lawrence J. Ellison Institute for Transformative Medicine of USC, Los Angeles, CA, United States.,Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
20
|
Sarrion-Perdigones A, Gonzalez Y, Chang L, Gallego-Flores T, Young DW, Venken KJT. Multiplex Hextuple Luciferase Assaying. Methods Mol Biol 2022; 2524:433-456. [PMID: 35821491 PMCID: PMC10157609 DOI: 10.1007/978-1-0716-2453-1_33] [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] [Indexed: 11/30/2022]
Abstract
We recently expanded the commonly used dual luciferase assaying method toward multiplex hextuple luciferase assaying, allowing monitoring the activity of five experimental pathways against one control at the same time. In doing so, while our expanded assay utilizes a total of six orthogonal luciferases instead of two, this assay, conveniently, still utilizes the well-established reagents and principles of the widely used dual luciferase assay. Three quenchable D-luciferin-consuming luciferases are measured after addition of D-Luciferin substrate, followed by quenching of their bioluminescence (BL) and the measurement of three coelenterazine (CTZ)-consuming luciferases after addition of CTZ substrate, all in the same vessel. Here, we provide detailed protocols on how to perform such multiplex hextuple luciferase assaying to monitor cellular signal processing upstream of five transcription factors and their corresponding transcription factor-binding motifs, using a constitutive promoter as normalization control. The first protocol is provided on how to perform cell culture in preparation toward genetic or pharmaceutical perturbations, as well as transfecting a multiplex hextuple luciferase reporter vector encoding all luciferase reporter units needed for multiplex hextuple luciferase assaying. The second protocol details on how to execute multiplex hextuple luciferase assaying using a microplate reader appropriately equipped to detect the different BLs emitted by all six luciferases. Finally, the third protocol provides details on analyzing, plotting, and interpreting the data obtained by the microplate reader.
Collapse
Affiliation(s)
- Alejandro Sarrion-Perdigones
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Yezabel Gonzalez
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Lyra Chang
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, USA
| | - Tatiana Gallego-Flores
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | - Damian W Young
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Koen J T Venken
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA.
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, USA.
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, USA.
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- McNair Medical Institute at The Robert and Janice McNair Foundation, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
21
|
From imaging a single cell to implementing precision medicine: an exciting new era. Emerg Top Life Sci 2021; 5:837-847. [PMID: 34889448 PMCID: PMC8786301 DOI: 10.1042/etls20210219] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 11/17/2022]
Abstract
In the age of high-throughput, single-cell biology, single-cell imaging has evolved not only in terms of technological advancements but also in its translational applications. The synchronous advancements of imaging and computational biology have produced opportunities of merging the two, providing the scientific community with tools towards observing, understanding, and predicting cellular and tissue phenotypes and behaviors. Furthermore, multiplexed single-cell imaging and machine learning algorithms now enable patient stratification and predictive diagnostics of clinical specimens. Here, we provide an overall summary of the advances in single-cell imaging, with a focus on high-throughput microscopy phenomics and multiplexed proteomic spatial imaging platforms. We also review various computational tools that have been developed in recent years for image processing and downstream applications used in biomedical sciences. Finally, we discuss how harnessing systems biology approaches and data integration across disciplines can further strengthen the exciting applications and future implementation of single-cell imaging on precision medicine.
Collapse
|
22
|
Müller N, Eugenio M, Romão LF, Marcondes de Souza J, Alves-Leon SV, Campanati L, Sant'Anna C. Assessing the antiproliferative effect of biogenic silver chloride nanoparticles on glioblastoma cell lines by quantitative image-based analysis. IET Nanobiotechnol 2021; 15:558-564. [PMID: 34694742 PMCID: PMC8675776 DOI: 10.1049/nbt2.12038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 11/26/2020] [Accepted: 12/18/2020] [Indexed: 11/30/2022] Open
Abstract
Glioblastoma is the most life‐threatening tumour of the central nervous system. Temozolomide (TMZ) is the first‐choice oral drug for the treatment of glioblastoma, although it shows low efficacy. Silver nanoparticles (AgNPs) have been shown to exhibit biocidal activity in a variety of microorganisms, including some pathogenic microorganisms. Herein, the antiproliferative effect of AgCl‐NPs on glioblastoma cell lines (GBM02 and GBM11) and on astrocytes was evaluated through automated quantitative image‐based analysis (HCA) of the cells. The cells were treated with 0.1‐5.0 μg/ml AgCl‐NPs or with 9.7‐48.5 μg/ml TMZ. Cells that received combined treatment were also analysed. At a maximum tested concentration of AgCl‐NPs, GBM02 and GBM11, the growth decreased by 93% and 40%, respectively, following 72 h of treatment. TMZ treatment decreased the proliferation of GBM02 and GBM11 cells by 58% and 34%, respectively. Combinations of AgCl‐NPs and TMZ showed intermediate antiproliferative effects; the lowest concentrations caused an inhibition similar to that obtained with TMZ, and the highest concentrations caused inhibition similar to that obtained with AgCl‐NPs alone. No significant changes in astrocyte proliferation were observed. The authors’ findings showed that HCA is a fast and reliable approach that can be used to evaluate the antiproliferative effect of the nanoparticles at the single‐cell level and that AgCl‐NPs are promising agents for glioblastoma treatment.
Collapse
Affiliation(s)
- Nathalia Müller
- Laboratory of Microscopy Applied to Life Science - Lamav, National Institute of Metrology, Quality and Technology, Duque de Caxias, RJ, Brazil
| | - Mateus Eugenio
- Laboratory of Microscopy Applied to Life Science - Lamav, National Institute of Metrology, Quality and Technology, Duque de Caxias, RJ, Brazil
| | - Luciana F Romão
- Laboratory of Cellular Morphogenesis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Jorge Marcondes de Souza
- University Hospital Clementino Fraga Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Soniza V Alves-Leon
- University Hospital Clementino Fraga Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Loraine Campanati
- Laboratory of Cellular Morphogenesis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Celso Sant'Anna
- Laboratory of Microscopy Applied to Life Science - Lamav, National Institute of Metrology, Quality and Technology, Duque de Caxias, RJ, Brazil
| |
Collapse
|
23
|
Organoids in image-based phenotypic chemical screens. Exp Mol Med 2021; 53:1495-1502. [PMID: 34663938 PMCID: PMC8569209 DOI: 10.1038/s12276-021-00641-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/08/2021] [Accepted: 05/04/2021] [Indexed: 12/18/2022] Open
Abstract
Image-based phenotypic screening relies on the extraction of multivariate information from cells cultured under a large variety of conditions. Technical advances in high-throughput microscopy enable screening in increasingly complex and biologically relevant model systems. To this end, organoids hold great potential for high-content screening because they recapitulate many aspects of parent tissues and can be derived from patient material. However, screening is substantially more difficult in organoids than in classical cell lines from both technical and analytical standpoints. In this review, we present an overview of studies employing organoids for screening applications. We discuss the promises and challenges of small-molecule treatments in organoids and give practical advice on designing, running, and analyzing high-content organoid-based phenotypic screens.
Collapse
|
24
|
Rens C, Shapira T, Peña-Diaz S, Chao JD, Pfeifer T, Av-Gay Y. Apoptosis assessment in high-content and high-throughput screening assays. Biotechniques 2021; 70:309-318. [PMID: 34114488 DOI: 10.2144/btn-2020-0164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Here the authors describe the development of AUTOptosis, an economical and rapid apoptosis monitoring method suitable for high-content and high-throughput screening assays. AUTOptosis is based on the quantification of nuclei intensity via staining with Hoechst 33342. First, the authors calibrated the method using standard apoptosis inducers in multiple cell lines. Next, the authors validated the applicability of this approach to high-content screening using a small library of compounds and compared it with the terminal deoxynucleotidyl transferase dUTP nick end labeling gold standard. Finally, the authors demonstrated the specificity of the method by using AUTOposis to detect apoptosis triggered by Mycobacterium tuberculosis intracellular infections.
Collapse
Affiliation(s)
- Céline Rens
- Department of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Tirosh Shapira
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Sandra Peña-Diaz
- Department of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Joseph D Chao
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Tom Pfeifer
- Biofactorial High-Throughput Biology Facility, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Yossef Av-Gay
- Department of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, Canada.,Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| |
Collapse
|
25
|
Chiang CT, Lau R, Ghaffarizadeh A, Brovold M, Vyas D, Juárez EF, Atala A, Agus DB, Soker S, Macklin P, Ruderman D, Mumenthaler SM. High-throughput microscopy reveals the impact of multifactorial environmental perturbations on colorectal cancer cell growth. Gigascience 2021; 10:giab026. [PMID: 33871006 PMCID: PMC8054261 DOI: 10.1093/gigascience/giab026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 11/21/2020] [Accepted: 03/15/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Colorectal cancer (CRC) mortality is principally due to metastatic disease, with the most frequent organ of metastasis being the liver. Biochemical and mechanical factors residing in the tumor microenvironment are considered to play a pivotal role in metastatic growth and response to therapy. However, it is difficult to study the tumor microenvironment systematically owing to a lack of fully controlled model systems that can be investigated in rigorous detail. RESULTS We present a quantitative imaging dataset of CRC cell growth dynamics influenced by in vivo-mimicking conditions. They consist of tumor cells grown in various biochemical and biomechanical microenvironmental contexts. These contexts include varying oxygen and drug concentrations, and growth on conventional stiff plastic, softer matrices, and bioengineered acellular liver extracellular matrix. Growth rate analyses under these conditions were performed via the cell phenotype digitizer (CellPD). CONCLUSIONS Our data indicate that the growth of highly aggressive HCT116 cells is affected by oxygen, substrate stiffness, and liver extracellular matrix. In addition, hypoxia has a protective effect against oxaliplatin-induced cytotoxicity on plastic and liver extracellular matrix. This expansive dataset of CRC cell growth measurements under in situ relevant environmental perturbations provides insights into critical tumor microenvironment features contributing to metastatic seeding and tumor growth. Such insights are essential to dynamical modeling and understanding the multicellular tumor-stroma dynamics that contribute to metastatic colonization. It also establishes a benchmark dataset for training and testing data-driven dynamical models of cancer cell lines and therapeutic response in a variety of microenvironmental conditions.
Collapse
Affiliation(s)
- Chun-Te Chiang
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA 90064, USA
| | - Roy Lau
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA 90064, USA
| | - Ahmadreza Ghaffarizadeh
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA 90064, USA
| | - Matthew Brovold
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC 27157, USA
| | - Dipen Vyas
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC 27157, USA
| | - Edwin F Juárez
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA 90064, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC 27157, USA
| | - David B Agus
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA 90064, USA
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC 27157, USA
| | - Paul Macklin
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA 90064, USA
- Intelligent Systems Engineering, Indiana University, Bloomington, IN 47408, USA
| | - Daniel Ruderman
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA 90064, USA
| | - Shannon M Mumenthaler
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA 90064, USA
| |
Collapse
|
26
|
Liu X, Li F, Zhang J, Wang L, Wang J, Wen Z, Wang Z, Shuai L, Wang X, Ge J, Zhao D, Bu Z. The ATPase ATP6V1A facilitates rabies virus replication by promoting virion uncoating and interacting with the viral matrix protein. J Biol Chem 2021; 296:100096. [PMID: 33208464 PMCID: PMC7949080 DOI: 10.1074/jbc.ra120.014190] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 11/12/2020] [Accepted: 11/18/2020] [Indexed: 12/25/2022] Open
Abstract
Rabies virus (RABV) matrix protein (M) plays crucial roles in viral transcription, replication, assembly, and budding; however, its function during the early stage of virus replication remains unknown. Here, we mapped the protein interactome between RABV M and human host factors using a proteomic approach, finding a link to the V-type proton ATPase catalytic subunit A (ATP6V1A), which is located in the endosomes where RABV first enters. By downregulating or upregulating ATP6V1A expression in HEK293T cells, we found that ATP6V1A facilitated RABV replication. We further found that ATP6V1A was involved in the dissociation of incoming viral M proteins during viral uncoating. Coimmunoprecipitation demonstrated that M interacted with the full length or middle domain of ATP6V1A, which was dependent on the lysine residue at position 256 and the glutamic acid residue at position 279. RABV growth and uncoating in ATP6V1A-depleted cells was restored by trans-complementation with the full length or interaction domain of ATP6V1A. Moreover, stably overexpressed ATP6V1A enhanced RABV growth in Vero cells, which are used for the production of rabies vaccine. Our findings identify a new partner for RABV M proteins and establish a new role of ATP6V1A by promoting virion uncoating during RABV replication.
Collapse
Affiliation(s)
- Xing Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Fang Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Jiwen Zhang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Lulu Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Jinliang Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Zhiyuan Wen
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Zilong Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Lei Shuai
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Xijun Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Jinying Ge
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Dongming Zhao
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China; National High Containment Laboratory for Animal Diseases Control and Prevention, Harbin, People's Republic of China.
| | - Zhigao Bu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China; National High Containment Laboratory for Animal Diseases Control and Prevention, Harbin, People's Republic of China.
| |
Collapse
|
27
|
Frieboes HB, Raghavan S, Godin B. Modeling of Nanotherapy Response as a Function of the Tumor Microenvironment: Focus on Liver Metastasis. Front Bioeng Biotechnol 2020; 8:1011. [PMID: 32974325 PMCID: PMC7466654 DOI: 10.3389/fbioe.2020.01011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022] Open
Abstract
The tumor microenvironment (TME) presents a challenging barrier for effective nanotherapy-mediated drug delivery to solid tumors. In particular for tumors less vascularized than the surrounding normal tissue, as in liver metastases, the structure of the organ itself conjures with cancer-specific behavior to impair drug transport and uptake by cancer cells. Cells and elements in the TME of hypovascularized tumors play a key role in the process of delivery and retention of anti-cancer therapeutics by nanocarriers. This brief review describes the drug transport challenges and how they are being addressed with advanced in vitro 3D tissue models as well as with in silico mathematical modeling. This modeling complements network-oriented techniques, which seek to interpret intra-cellular relevant pathways and signal transduction within cells and with their surrounding microenvironment. With a concerted effort integrating experimental observations with computational analyses spanning from the molecular- to the tissue-scale, the goal of effective nanotherapy customized to patient tumor-specific conditions may be finally realized.
Collapse
Affiliation(s)
- Hermann B. Frieboes
- Department of Bioengineering, University of Louisville, Louisville, KY, United States
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
- Center for Predictive Medicine, University of Louisville, Louisville, KY, United States
| | - Shreya Raghavan
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, United States
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States
- Department of Obstetrics and Gynecology, Houston Methodist Hospital, Houston, TX, United States
- Developmental Therapeutics Program, Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
28
|
Sarrion-Perdigones A, Chang L, Gonzalez Y, Gallego-Flores T, Young DW, Venken KJT. Simultaneous Examination of Cellular Pathways using Multiplex Hextuple Luciferase Assaying. ACTA ACUST UNITED AC 2020; 131:e122. [PMID: 32539239 DOI: 10.1002/cpmb.122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Multiplex experimentation that can assay multiple cellular signaling pathways in the same cells requires orthogonal genetically encoded reporters that report over large dynamic ranges. Luciferases are cost-effective, versatile candidates whose output signals can be sensitively detected in a multiplex fashion. Commonly used dual luciferase reporter assays detect one luciferase that is coupled to a single cellular pathway and a second that is coupled to a control pathway for normalization purposes. We have expanded this approach to multiplex hextuple luciferase assays that can report on five cellular signaling pathways and one control, each of which is encoded by a unique luciferase. Light emission by the six luciferases can be distinguished by the use of two distinct substrates, each specific for three luciferases, followed by spectral decomposition of the light emitted by each of the three luciferase enzymes with bandpass filters. Here, we present detailed protocols on how to perform multiplex hextuple luciferase assaying to monitor pathway fluxes through transcriptional response elements for five specific signaling pathways (i.e., c-Myc, NF-κβ, TGF-β, p53, and MAPK/JNK) using the constitutive CMV promoter as normalization control. Protocols are provided for preparing reporter vector plasmids for multiplex reporter assaying, performing cell culture and multiplex luciferase reporter vector plasmid transfection, executing multiplex luciferase assays, and analyzing and interpreting data obtained by a plate reader appropriately equipped to detect the different luminescences. © 2020 Wiley Periodicals LLC. Basic Protocol 1: Preparation of vectors for multiplex hextuple luciferase assaying Basic Protocol 2: Cell culture work for multiplex hextuple luciferase assays Basic Protocol 3: Transfection of luciferase reporter plasmids followed by drug and recombinant protein treatments Basic Protocol 4: Performing the multiplex hextuple luciferase assay.
Collapse
Affiliation(s)
- Alejandro Sarrion-Perdigones
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas
| | - Lyra Chang
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, Texas.,Center for Drug Discovery, Baylor College of Medicine, Houston, Texas
| | - Yezabel Gonzalez
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas
| | - Tatiana Gallego-Flores
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.,Current address: Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | - Damian W Young
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas.,Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, Texas.,Center for Drug Discovery, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Therapeutic Innovation Center, Baylor College of Medicine, Houston, Texas
| | - Koen J T Venken
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas.,Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, Texas.,Center for Drug Discovery, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Therapeutic Innovation Center, Baylor College of Medicine, Houston, Texas.,McNair Medical Institute at The Robert and Janice McNair Foundation, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
29
|
Garvey CM, Lau R, Sanchez A, Sun RX, Fong EJ, Doche ME, Chen O, Jusuf A, Lenz HJ, Larson B, Mumenthaler SM. Anti-EGFR Therapy Induces EGF Secretion by Cancer-Associated Fibroblasts to Confer Colorectal Cancer Chemoresistance. Cancers (Basel) 2020; 12:cancers12061393. [PMID: 32481658 PMCID: PMC7352975 DOI: 10.3390/cancers12061393] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/13/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022] Open
Abstract
Targeted agents have improved the efficacy of chemotherapy for cancer patients, however, there remains a lack of understanding of how these therapies affect the unsuspecting bystanders of the stromal microenvironment. Cetuximab, a monoclonal antibody therapy targeting the epidermal growth factor receptor (EGFR), is given in combination with chemotherapy as the standard of care for a subset of metastatic colorectal cancer patients. The overall response to this treatment is underwhelming and, while genetic mutations that confer resistance have been identified, it is still not known why this drug is ineffective for some patients. We discovered that cancer-associated fibroblasts (CAFs), a major cellular subset of the tumor stroma, can provide a source of cancer cell resistance. Specifically, we observed that upon treatment with cetuximab, CAFs increased their secretion of EGF, which was sufficient to render neighboring cancer cells resistant to cetuximab treatment through sustained mitogen-activated protein kinases (MAPK) signaling. Furthermore, we show the cetuximab-induced EGF secretion to be specific to CAFs and not to cancer cells or normal fibroblasts. Altogether, this work emphasizes the importance of the tumor microenvironment and considering the potential unintended consequences of therapeutically targeting cancer-driving proteins on non-tumorigenic cell types.
Collapse
Affiliation(s)
- Colleen M. Garvey
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA 90033, USA; (C.M.G.); (R.L.); (A.S.); (R.X.S.); (E.J.F.); (M.E.D.); (O.C.); (A.J.)
| | - Roy Lau
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA 90033, USA; (C.M.G.); (R.L.); (A.S.); (R.X.S.); (E.J.F.); (M.E.D.); (O.C.); (A.J.)
| | - Alyssa Sanchez
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA 90033, USA; (C.M.G.); (R.L.); (A.S.); (R.X.S.); (E.J.F.); (M.E.D.); (O.C.); (A.J.)
| | - Ren X. Sun
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA 90033, USA; (C.M.G.); (R.L.); (A.S.); (R.X.S.); (E.J.F.); (M.E.D.); (O.C.); (A.J.)
| | - Emma J. Fong
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA 90033, USA; (C.M.G.); (R.L.); (A.S.); (R.X.S.); (E.J.F.); (M.E.D.); (O.C.); (A.J.)
| | - Michael E. Doche
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA 90033, USA; (C.M.G.); (R.L.); (A.S.); (R.X.S.); (E.J.F.); (M.E.D.); (O.C.); (A.J.)
| | - Oscar Chen
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA 90033, USA; (C.M.G.); (R.L.); (A.S.); (R.X.S.); (E.J.F.); (M.E.D.); (O.C.); (A.J.)
| | - Anthony Jusuf
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA 90033, USA; (C.M.G.); (R.L.); (A.S.); (R.X.S.); (E.J.F.); (M.E.D.); (O.C.); (A.J.)
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
| | - Brent Larson
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Shannon M. Mumenthaler
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA 90033, USA; (C.M.G.); (R.L.); (A.S.); (R.X.S.); (E.J.F.); (M.E.D.); (O.C.); (A.J.)
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
- Correspondence: ; Tel.: +1-323-442-2529
| |
Collapse
|
30
|
Monitoring cell endocytosis of liposomes by real-time electrical impedance spectroscopy. Anal Bioanal Chem 2020; 412:6371-6380. [PMID: 32451643 DOI: 10.1007/s00216-020-02592-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 03/04/2020] [Accepted: 03/09/2020] [Indexed: 10/24/2022]
Abstract
Evaluation and understanding the effect of drug delivery in in vitro systems is fundamental in drug discovery. We present an assay based on real-time electrical impedance spectroscopy (EIS) measurements that can be used to follow the internalisation and cytotoxic effect of a matrix metalloproteinase (MMP)-sensitive liposome formulation loaded with oxaliplatin (OxPt) on colorectal cancer cells. The EIS response identified two different cellular processes: (i) a negative peak in the cell index (CI) within the first 5 h, due to onset of liposome endocytosis, followed by (ii) a subsequent CI increase, due to the reattachment of cells until the onset of cytotoxicity with a decrease in CI. Free OxPt or OxPt-loaded Stealth liposomes did not show this two-stage EIS response; the latter can be due to the fact that Stealth cannot be cleaved by MMPs and thus is not taken up by the cells. Real-time bright-field imaging supported the EIS data, showing variations in cell adherence and cell morphology after exposure to the different liposome formulations. A drastic decrease in cell coverage as well as rounding up of cells during the first 5 h of exposure to OxPt-loaded (MMP)-sensitive liposome formulation is reflected by the first negative EIS response, which indicates the onset of liposome endocytosis. Graphical abstract.
Collapse
|
31
|
Fong EJ, Strelez C, Mumenthaler SM. A Perspective on Expanding Our Understanding of Cancer Treatments by Integrating Approaches from the Biological and Physical Sciences. SLAS DISCOVERY 2020; 25:672-683. [PMID: 32297829 PMCID: PMC7372587 DOI: 10.1177/2472555220915830] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Multicellular systems such as cancer suffer from immense complexity. It is imperative to capture the heterogeneity of these systems across scales to achieve a deeper understanding of the underlying biology and develop effective treatment strategies. In this perspective article, we will discuss how recent technologies and approaches from the biological and physical sciences have transformed traditional ways of measuring, interpreting, and treating cancer. During the SLAS 2019 Annual Meeting, SBI2 hosted a Special Interest Group (SIG) on this topic. Academic and industry leaders engaged in discussions surrounding what biological model systems are appropriate to study cancer complexity, what assays are necessary to interrogate this complexity, and how physical sciences approaches may be useful to detangle this complexity. In particular, we examined the utility of mathematical models in predicting cancer progression and treatment response when tightly integrated with reproducible, quantitative, and dynamic biological measurements achieved using high-content imaging and analysis. The dialogue centered around the impetus for convergent biosciences, bringing new perspectives to cancer research to further understand this complex adaptive system and successfully intervene therapeutically.
Collapse
Affiliation(s)
- Emma J Fong
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Carly Strelez
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shannon M Mumenthaler
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
32
|
HCS Methodology for Helping in Lab Scale Image-Based Assays. Methods Mol Biol 2020. [PMID: 31432486 DOI: 10.1007/978-1-4939-9686-5_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
High-content screening (HCS) automates image acquisition and analysis in microscopy. This technology considers the multiple parameters contained in the images and produces statistically significant results. The recent improvements in image acquisition throughput, image analysis, and machine learning (ML) have popularized this kind of experiments, emphasizing the need for new tools and know-how to help in its design, analysis, and data interpretation. This chapter summarizes HCS recommendations for lab scale assays and provides both macros for HCS-oriented image analysis and user-friendly tools for data mining processes. All the steps described herein are oriented to a wide variety of image cell-based experiments. The workflows are illustrated with practical examples and test images. Their use is expected to help analyze thousands of images, create graphical representations, and apply machine learning models on HCS.
Collapse
|
33
|
Examining multiple cellular pathways at once using multiplex hextuple luciferase assaying. Nat Commun 2019; 10:5710. [PMID: 31836712 PMCID: PMC6911020 DOI: 10.1038/s41467-019-13651-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 11/14/2019] [Indexed: 01/24/2023] Open
Abstract
Sensitive simultaneous assessment of multiple signaling pathways within the same cells requires orthogonal reporters that can assay over large dynamic ranges. Luciferases are such genetically encoded candidates due to their sensitivity, versatility, and cost-effectiveness. We expand luciferase multiplexing in post-lysis endpoint luciferase assays from two to six. Light emissions are distinguished by a combination of distinct substrates and emission spectra deconvolution. All six luciferase reporter units are stitched together into one plasmid facilitating delivery of all reporter units through a process we termed solotransfection, minimizing experimental errors. We engineer a multiplex hextuple luciferase assay to probe pathway fluxes through five transcriptional response elements against a control constitutive promoter. We can monitor effects of siRNA, ligand, and chemical compound treatments on their target pathways along with the four other probed cellular pathways. We demonstrate the effectiveness and adaptiveness of multiplex luciferase assaying, and its broad application across different research fields. Multiplexed detection of luciferase-based sensors in the same sample is challenging and limited by the substrates’ emission spectra. Here the authors establish a system based on three different luciferases and sequential detection to achieve measurements of up to six parameters within the same experiment.
Collapse
|
34
|
Bera T, Xu J, Alusta P, Fong A, Linder SW, Torosian SD. Estimating Bacterial Concentrations in Fibrous Substrates Through a Combination of Scanning Electron Microscopy and ImageJ. Anal Chem 2019; 91:4405-4412. [PMID: 30835114 DOI: 10.1021/acs.analchem.8b04862] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Conventional signal-based microanalytical techniques for estimating bacterial concentrations are often susceptible to false signals. A visual quantification, therefore, may compliment such techniques by providing additional information and support better management decisions in the event of outbreaks. Herein, we explore a method that combines electron microscopy (EM) and image-analysis techniques and allows both visualization and quantification of pathogenic bacteria adherent even to complex nonuniform substrates. Both the estimation and imaging parameters were optimized to reduce the estimation error ( E, %) to close to ±5%. The method was validated against conventional microbiological techniques such as the use of optical density, flow cytometry, and quantitative real-time PCR (qPCR). It could easily be tailored to estimate different species of pathogens, such as Escherichia coli O157, Listeria innocua, Staphylococcus aureus, Enterococcus faecalis, and Bacillus anthracis, on samples similar to those in real-time contamination scenarios. The present method is sensitive enough to detect ∼100 bacterial CFU/mL but has the potential to estimate even lower concentrations with increased imaging and computation times. Overall, this imaging-based method may greatly complement any signal-based pathogen-detection technique, especially in negating false signals, and therefore may significantly contribute to the field of analytical microbiology and biochemistry.
Collapse
Affiliation(s)
- Tanmay Bera
- Arkansas Laboratory-Nanotechnology Core Facility (ARKL-NanoCore), Office of Regulatory Sciences, Office of Regulatory Affairs (ORS, ORA) , U.S. FDA , Jefferson , Arkansas 72079 , United States.,Division of Bioinformatics and Biostatistics , National Center for Toxicological Research (NCTR), U.S. FDA , Jefferson , Arkansas 72079 , United States
| | - Joshua Xu
- Division of Bioinformatics and Biostatistics , National Center for Toxicological Research (NCTR), U.S. FDA , Jefferson , Arkansas 72079 , United States
| | - Pierre Alusta
- Division of Systems Biology , NCTR, U.S. FDA , Jefferson , Arkansas 72079 , United States
| | - Andrew Fong
- Arkansas Laboratory-Nanotechnology Core Facility (ARKL-NanoCore), Office of Regulatory Sciences, Office of Regulatory Affairs (ORS, ORA) , U.S. FDA , Jefferson , Arkansas 72079 , United States
| | - Sean W Linder
- ORS, ORA , U.S. FDA , Jefferson , Arkansas 72079 , United States
| | - Stephen D Torosian
- Winchester Engineering and Analytical Center (WEAC), ORS, ORA , U.S. FDA , Winchester , Massachusetts 01890 , United States
| |
Collapse
|
35
|
St-Georges-Robillard A, Cahuzac M, Péant B, Fleury H, Lateef MA, Ricard A, Sauriol A, Leblond F, Mes-Masson AM, Gervais T. Long-term fluorescence hyperspectral imaging of on-chip treated co-culture tumour spheroids to follow clonal evolution. Integr Biol (Camb) 2019; 11:130-141. [PMID: 31172192 DOI: 10.1093/intbio/zyz012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/09/2019] [Accepted: 05/30/2019] [Indexed: 12/22/2022]
Abstract
Multicellular tumour spheroids are an ideal in vitro tumour model to study clonal heterogeneity and drug resistance in cancer research because different cell types can be mixed at will. However, measuring the individual response of each cell population over time is challenging: current methods are either destructive, such as flow cytometry, or cannot image throughout a spheroid, such as confocal microscopy. Our group previously developed a wide-field fluorescence hyperspectral imaging system to study spheroids formed and cultured in microfluidic chips. In the present study, two subclones of a single parental ovarian cancer cell line transfected to express different fluorophores were produced and co-culture spheroids were formed on-chip using ratios forming highly asymmetric subpopulations. We performed a 3D proliferation assay on each cell population forming the spheroids that matched the 2D growth behaviour. Response assays to PARP inhibitors and platinum-based drugs were also performed to follow the clonal evolution of mixed populations. Our experiments show that hyperspectral imaging can detect spheroid response before observing a decrease in spheroid diameter. Hyperspectral imaging and microfluidic-based spheroid assays provide a versatile solution to study clonal heterogeneity, able to measure response in subpopulations presenting as little as 10% of the initial spheroid.
Collapse
Affiliation(s)
- Amélie St-Georges-Robillard
- Polytechnique Montréal, Department of Engineering Physics and Institute of Biomedical Engineering, Montreal, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Canada
| | - Maxime Cahuzac
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Canada
| | - Benjamin Péant
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Canada
- TransMedTech Institute, Montréal, Canada
| | - Hubert Fleury
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Canada
| | - Muhammad Abdul Lateef
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Canada
| | - Alexis Ricard
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Canada
| | - Alexandre Sauriol
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Canada
| | - Frédéric Leblond
- Polytechnique Montréal, Department of Engineering Physics and Institute of Biomedical Engineering, Montreal, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Canada
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Canada
- Université de Montréal, Department of Medicine, Montreal, Canada
| | - Thomas Gervais
- Polytechnique Montréal, Department of Engineering Physics and Institute of Biomedical Engineering, Montreal, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Canada
| |
Collapse
|
36
|
Identification of pharmacological inhibitors of conventional protein secretion. Sci Rep 2018; 8:14966. [PMID: 30297865 PMCID: PMC6175952 DOI: 10.1038/s41598-018-33378-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/21/2018] [Indexed: 01/29/2023] Open
Abstract
The retention using selective hooks (RUSH) system allows to withhold a fluorescent biosensor such as green fluorescent protein (GFP) fused to a streptavidin-binding peptide (SBP) by an excess of streptavidin molecules that are addressed to different subcellular localizations. Addition of biotin competitively disrupts this interaction, liberating the biosensor from its hook. We constructed a human cell line co-expressing soluble secretory-SBP-GFP (ss-SBP-GFP) and streptavidin within the endoplasmic reticulum (ER) lumen and then used this system to screen a compound library for inhibitors of the biotin-induced release of ss-SBP-GFP via the conventional Golgi-dependent protein secretion pathway into the culture supernatant. We identified and validated a series of molecularly unrelated drugs including antianginal, antidepressant, anthelmintic, antipsychotic, antiprotozoal and immunosuppressive agents that inhibit protein secretion. These compounds vary in their capacity to suppress protein synthesis and to compromise ER morphology and Golgi integrity, as well as in the degree of reversibility of such effects. In sum, we demonstrate the feasibility and utility of a novel RUSH-based phenotypic screening assay.
Collapse
|
37
|
Dorval T, Chanrion B, Cattin ME, Stephan JP. Filling the drug discovery gap: is high-content screening the missing link? Curr Opin Pharmacol 2018; 42:40-45. [DOI: 10.1016/j.coph.2018.07.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 07/01/2018] [Indexed: 11/29/2022]
|
38
|
Chiang CT, Demetriou AN, Ung N, Choudhury N, Ghaffarian K, Ruderman DL, Mumenthaler SM. mTORC2 contributes to the metabolic reprogramming in EGFR tyrosine-kinase inhibitor resistant cells in non-small cell lung cancer. Cancer Lett 2018; 434:152-159. [PMID: 30036610 PMCID: PMC7443389 DOI: 10.1016/j.canlet.2018.07.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/17/2018] [Accepted: 07/17/2018] [Indexed: 12/12/2022]
Abstract
Non-small cell lung cancer (NSCLC) patients with activating EGFR mutations are often successfully treated with EGFR tyrosine kinase inhibitor (TKI) such as erlotinib; however, treatment resistance inevitably occurs. Given tumor metabolism of glucose and therapeutic response are intimately linked, we explored the metabolic differences between isogenic erlotinib-sensitive and -resistant NSCLC cell lines. We discovered that the growth of erlotinib-resistant cells is more sensitive to glucose deprivation. Seahorse metabolic assay revealed erlotinib-resistant cells have lower spare respiratory capacity (SRC), an indicator of metabolic flexibility, compared to erlotinib-sensitive cells. Additionally, we found downstream components of mTORC2 signaling to be phosphorylated in erlotinib-resistant cells. Knockdown of an mTORC2 component, Rictor, enhanced the SRC and rescued the growth rate of erlotinib-resistant cells during glucose deprivation. Among NSCLCs with activating EGFR mutations, gene sets involved in glucose metabolism were enriched in patients with high expression of p-NDGR1, a readout of mTORC2 activity. Furthermore, overall survival was negatively correlated with p-NDRG1. Our work uncovers a link between mTORC2 and metabolic reprogramming in EGFR TKI-resistant cells and highlights the significance of mTORC2 in the progression of EGFR-mutated NSCLC.
Collapse
Affiliation(s)
- Chun-Te Chiang
- Lawrence J. Ellison Institute for Transformative Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Alexandra N Demetriou
- Lawrence J. Ellison Institute for Transformative Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Nolan Ung
- Lawrence J. Ellison Institute for Transformative Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Niharika Choudhury
- Lawrence J. Ellison Institute for Transformative Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Kimya Ghaffarian
- Lawrence J. Ellison Institute for Transformative Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Daniel L Ruderman
- Lawrence J. Ellison Institute for Transformative Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Shannon M Mumenthaler
- Lawrence J. Ellison Institute for Transformative Medicine of USC, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
39
|
Ghaffarizadeh A, Heiland R, Friedman SH, Mumenthaler SM, Macklin P. PhysiCell: An open source physics-based cell simulator for 3-D multicellular systems. PLoS Comput Biol 2018; 14:e1005991. [PMID: 29474446 PMCID: PMC5841829 DOI: 10.1371/journal.pcbi.1005991] [Citation(s) in RCA: 219] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 03/07/2018] [Accepted: 01/19/2018] [Indexed: 02/07/2023] Open
Abstract
Many multicellular systems problems can only be understood by studying how cells move, grow, divide, interact, and die. Tissue-scale dynamics emerge from systems of many interacting cells as they respond to and influence their microenvironment. The ideal "virtual laboratory" for such multicellular systems simulates both the biochemical microenvironment (the "stage") and many mechanically and biochemically interacting cells (the "players" upon the stage). PhysiCell-physics-based multicellular simulator-is an open source agent-based simulator that provides both the stage and the players for studying many interacting cells in dynamic tissue microenvironments. It builds upon a multi-substrate biotransport solver to link cell phenotype to multiple diffusing substrates and signaling factors. It includes biologically-driven sub-models for cell cycling, apoptosis, necrosis, solid and fluid volume changes, mechanics, and motility "out of the box." The C++ code has minimal dependencies, making it simple to maintain and deploy across platforms. PhysiCell has been parallelized with OpenMP, and its performance scales linearly with the number of cells. Simulations up to 105-106 cells are feasible on quad-core desktop workstations; larger simulations are attainable on single HPC compute nodes. We demonstrate PhysiCell by simulating the impact of necrotic core biomechanics, 3-D geometry, and stochasticity on the dynamics of hanging drop tumor spheroids and ductal carcinoma in situ (DCIS) of the breast. We demonstrate stochastic motility, chemical and contact-based interaction of multiple cell types, and the extensibility of PhysiCell with examples in synthetic multicellular systems (a "cellular cargo delivery" system, with application to anti-cancer treatments), cancer heterogeneity, and cancer immunology. PhysiCell is a powerful multicellular systems simulator that will be continually improved with new capabilities and performance improvements. It also represents a significant independent code base for replicating results from other simulation platforms. The PhysiCell source code, examples, documentation, and support are available under the BSD license at http://PhysiCell.MathCancer.org and http://PhysiCell.sf.net.
Collapse
Affiliation(s)
- Ahmadreza Ghaffarizadeh
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Randy Heiland
- Intelligent Systems Engineering, Indiana University, Bloomington, Indiana, United States of America
| | - Samuel H. Friedman
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, California, United States of America
- Opto-Knowledge Systems, Inc., Torrance, California, United States of America
| | - Shannon M. Mumenthaler
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Paul Macklin
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, California, United States of America
- Intelligent Systems Engineering, Indiana University, Bloomington, Indiana, United States of America
| |
Collapse
|
40
|
The emergence of dynamic phenotyping. Cell Biol Toxicol 2017; 33:507-509. [DOI: 10.1007/s10565-017-9413-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 09/18/2017] [Indexed: 02/07/2023]
|
41
|
Garvey CM, Gerhart TA, Mumenthaler SM. Discrimination and Characterization of Heterocellular Populations Using Quantitative Imaging Techniques. J Vis Exp 2017. [PMID: 28715383 PMCID: PMC5608532 DOI: 10.3791/55844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Cellular processes are complex and result from the interplay between multiple cell types and their environment. Existing cell biology techniques often do not allow for accurate interpretation of this interplay. Using a quantitative imaging-based approach, we present a high-content protocol for characterizing the dynamic phenotypic responses (i.e. morphology changes, proliferation, apoptosis) of heterogeneous cell populations to changes in environmental stimuli. We highlight our ability to distinguish between cell types based upon either fluorescence intensity or inherent morphology features depending on the application. This platform allows for a more comprehensive characterization of subpopulation response to perturbation while utilizing shorter time, smaller amounts of reagents, and lower likelihood of error than traditional cell biology assays. However, in some cases, cell populations may be difficult to identify and quantitate based on complex cellular features and will require additional troubleshooting; we highlight some of these circumstances in the protocol. We demonstrate this application using response to drug in a cancer model; however, it can easily be applied more broadly to other physiological processes. This protocol allows one to identify subpopulations within a co-culture system and characterize the particular response of each to external stimuli.
Collapse
Affiliation(s)
- Colleen M Garvey
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California (USC)
| | - Torin A Gerhart
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California (USC)
| | - Shannon M Mumenthaler
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California (USC);
| |
Collapse
|
42
|
Sant GR, Knopf KB, Albala DM. Live-single-cell phenotypic cancer biomarkers-future role in precision oncology? NPJ Precis Oncol 2017; 1:21. [PMID: 29872705 PMCID: PMC5871838 DOI: 10.1038/s41698-017-0025-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 04/21/2017] [Accepted: 05/05/2017] [Indexed: 01/08/2023] Open
Abstract
The promise of precision and personalized medicine is rooted in accurate, highly sensitive, and specific disease biomarkers. This is particularly true for cancer-a disease characterized by marked tumor heterogeneity and diverse molecular signatures. Although thousands of biomarkers have been described, only a very small number have been successfully translated into clinical use. Undoubtedly, there is need for rapid, quantitative, and more cost effective biomarkers for tumor diagnosis and prognosis, to allow for better risk stratification and aid clinicians in making personalized treatment decisions. This is particularly true for cancers where specific biomarkers are either not available (e.g., renal cell carcinoma) or where current biomarkers tend to classify individuals into broad risk categories unable to accurately assess individual tumor aggressiveness and adverse pathology potential (e.g., prostate cancer), thereby leading to problems of over-diagnosis and over-treatment of indolent cancer and under-treatment of aggressive cancer. This perspective highlights an emerging class of cancer biomarkers-live-single-cell phenotypic biomarkers, as compared to genomic biomarkers, and their potential application for cancer diagnosis, risk-stratification, and prognosis.
Collapse
Affiliation(s)
- Grannum R Sant
- Department of Urology, Tufts University School of Medicine, 82 Dennison Street, Gloucester, MA 01930 UK
| | - Kevin B Knopf
- Cancer Commons, 35050 El Camino Real, Los Altos, CA 94022 USA
| | - David M Albala
- 3Department of Urology, Crouse Hospital, Syracuse, NY USA
| |
Collapse
|
43
|
Elitas M, Sadeghi S, Karamahmutoglu H, Gozuacik D, Serdar Turhal N. Microfabricated platforms to quantitatively investigate cellular behavior under the influence of chemical gradients. Biomed Phys Eng Express 2017. [DOI: 10.1088/2057-1976/aa7400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
44
|
Juarez EF, Lau R, Friedman SH, Ghaffarizadeh A, Jonckheere E, Agus DB, Mumenthaler SM, Macklin P. Quantifying differences in cell line population dynamics using CellPD. BMC SYSTEMS BIOLOGY 2016; 10:92. [PMID: 27655224 PMCID: PMC5031291 DOI: 10.1186/s12918-016-0337-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 09/14/2016] [Indexed: 11/10/2022]
Abstract
BACKGROUND The increased availability of high-throughput datasets has revealed a need for reproducible and accessible analyses which can quantitatively relate molecular changes to phenotypic behavior. Existing tools for quantitative analysis generally require expert knowledge. RESULTS CellPD (cell phenotype digitizer) facilitates quantitative phenotype analysis, allowing users to fit mathematical models of cell population dynamics without specialized training. CellPD requires one input (a spreadsheet) and generates multiple outputs including parameter estimation reports, high-quality plots, and minable XML files. We validated CellPD's estimates by comparing it with a previously published tool (cellGrowth) and with Microsoft Excel's built-in functions. CellPD correctly estimates the net growth rate of cell cultures and is more robust to data sparsity than cellGrowth. When we tested CellPD's usability, biologists (without training in computational modeling) ran CellPD correctly on sample data within 30 min. To demonstrate CellPD's ability to aid in the analysis of high throughput data, we created a synthetic high content screening (HCS) data set, where a simulated cell line is exposed to two hypothetical drug compounds at several doses. CellPD correctly estimates the drug-dependent birth, death, and net growth rates. Furthermore, CellPD's estimates quantify and distinguish between the cytostatic and cytotoxic effects of both drugs-analyses that cannot readily be performed with spreadsheet software such as Microsoft Excel or without specialized computational expertise and programming environments. CONCLUSIONS CellPD is an open source tool that can be used by scientists (with or without a background in computational or mathematical modeling) to quantify key aspects of cell phenotypes (such as cell cycle and death parameters). Early applications of CellPD may include drug effect quantification, functional analysis of gene knockout experiments, data quality control, minable big data generation, and integration of biological data with computational models.
Collapse
Affiliation(s)
- Edwin F Juarez
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, California, USA. .,Department of Electrical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA.
| | - Roy Lau
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, California, USA
| | - Samuel H Friedman
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, California, USA
| | - Ahmadreza Ghaffarizadeh
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, California, USA
| | - Edmond Jonckheere
- Department of Electrical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| | - David B Agus
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, California, USA
| | - Shannon M Mumenthaler
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, California, USA
| | - Paul Macklin
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|