1
|
Lipreri MV, Totaro MT, Boos JA, Basile MS, Baldini N, Avnet S. A Novel Microfluidic Platform for Personalized Anticancer Drug Screening Through Image Analysis. MICROMACHINES 2024; 15:1521. [PMID: 39770275 PMCID: PMC11677617 DOI: 10.3390/mi15121521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025]
Abstract
The advancement of personalized treatments in oncology has garnered increasing attention, particularly for rare and aggressive cancer with low survival rates like the bone tumors osteosarcoma and chondrosarcoma. This study introduces a novel PDMS-agarose microfluidic device tailored for generating patient-derived tumor spheroids and serving as a reliable tool for personalized drug screening. Using this platform in tandem with a custom imaging index, we evaluated the impact of the anticancer agent doxorubicin on spheroids from both tumor types. The device produces 20 spheroids, each around 300 µm in diameter, within a 24 h timeframe, facilitating assessments of characteristics and reproducibility. Following spheroid generation, we measured patient-derived spheroid diameters in bright-field images, calcein AM-positive areas/volume, and the binary fraction area, a metric analyzing fluorescence intensity. By employing a specially developed equation that combines viability signal extension and intensity, we observed a substantial decrease in spheroid viability of around 75% for both sarcomas at the highest dosage (10 µM). Osteosarcoma spheroids exhibited greater sensitivity to doxorubicin than chondrosarcoma spheroids within 48 h. This approach provides a reliable in vitro model for aggressive sarcomas, representing a personalized approach for drug screening that could lead to more effective cancer treatments tailored to individual patients, despite some implementation challenges.
Collapse
Affiliation(s)
- Maria Veronica Lipreri
- Biomedical Science, Technologies, and Nanobiotecnology Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.V.L.)
| | - Marilina Tamara Totaro
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy;
| | - Julia Alicia Boos
- Bioengineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Klingelbergstrasse 48, 4056 Basel, Switzerland;
| | - Maria Sofia Basile
- Biomedical Science, Technologies, and Nanobiotecnology Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.V.L.)
| | - Nicola Baldini
- Biomedical Science, Technologies, and Nanobiotecnology Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.V.L.)
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy;
| | - Sofia Avnet
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy;
| |
Collapse
|
2
|
Özcan AI, Aquino Lόpez A, Wolff AN, Ma A, Shaw AR, Suzuki M, Brenner MK, McKenna MK. Mesenchymal stromal cells protect combined oncolytic and helper-dependent adenoviruses from humoral immunity. Mol Ther Methods Clin Dev 2024; 32:101279. [PMID: 38993326 PMCID: PMC11238183 DOI: 10.1016/j.omtm.2024.101279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/04/2024] [Indexed: 07/13/2024]
Abstract
Systemic delivery of oncolytic and immunomodulatory adenoviruses may be required for optimal effects on human malignancies. Mesenchymal stromal cells (MSCs) can serve as delivery systems for cancer therapeutics due to their ability to transport and shield these agents while homing to tumors. We now use MSCs to deliver a clinically validated binary oncolytic and helper-dependent adenovirus combination (CAdVEC) to tumor cells. We show successful oncolysis and helper-dependent virus function in tumor cells even in the presence of plasma from adenovirus-seropositive donors. In both two- and three-dimensional cultures, CAdVEC function is eliminated even at high dilutions of seropositive plasma but is well sustained when CAdVEC is delivered by MSCs. These results provide a robust in vitro model to measure oncolytic and helper-dependent virus spread and demonstrate a beneficial role of using MSCs for systemic delivery of CAdVEC even in the presence of a neutralizing humoral response.
Collapse
Affiliation(s)
- Ada Irmak Özcan
- Center for Cell Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Arianexys Aquino Lόpez
- Center for Cell Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA
- Texas Children's Hospital, Department of Pediatrics, Houston, TX 77030, USA
| | - Alexandra N Wolff
- Center for Cell Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA
- Harvard Medical School, Graduate School of Arts and Sciences, Boston, MA 02115, USA
| | - Audrey Ma
- Center for Cell Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Amanda Rosewell Shaw
- Center for Cell Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA
- Benedict College, Columbia, SC 29204, USA
| | - Masataka Suzuki
- Center for Cell Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Malcolm K Brenner
- Center for Cell Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mary K McKenna
- Center for Cell Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
3
|
Franko R, de Almeida Monteiro Melo Ferraz M. Exploring the potential of in vitro extracellular vesicle generation in reproductive biology. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70007. [PMID: 39238549 PMCID: PMC11375532 DOI: 10.1002/jex2.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/11/2024] [Accepted: 08/15/2024] [Indexed: 09/07/2024]
Abstract
The interest in the growing field of extracellular vesicle (EV) research highlights their significance in intercellular signalling and the selective transfer of biological information between donor and recipient cells. EV studies have provided valuable insights into intercellular communication mechanisms, signal identification and their involvement in disease states, offering potential avenues for manipulating pathological conditions, detecting biomarkers and developing drug-delivery systems. While our understanding of EV functions in reproductive tissues has significantly progressed, exploring their potential as biomarkers for infertility, therapeutic interventions and enhancements in assisted reproductive technologies remains to be investigated. This knowledge gap stems partly from the difficulties associated with large-scale EV production relevant to clinical applications. Most existing studies on EV production rely on conventional 2D cell culture systems, characterized by suboptimal EV yields and a failure to replicate in vivo conditions. This results in the generation of EVs that differ from their in vivo counterparts. Hence, this review firstly delves into the importance of EVs in reproduction to then expand on current techniques for in vitro EV production, specifically examining diverse methods of culture and the potential of bioengineering technologies to establish innovative systems for enhanced EV production.
Collapse
Affiliation(s)
- Roksan Franko
- Clinic of Ruminants, Faculty of Veterinary Medicine Ludwig-Maximilians-Universität München Oberschleißheim Germany
- Gene Center Ludwig-Maximilians-Universität München Munich Germany
| | - Marcia de Almeida Monteiro Melo Ferraz
- Clinic of Ruminants, Faculty of Veterinary Medicine Ludwig-Maximilians-Universität München Oberschleißheim Germany
- Gene Center Ludwig-Maximilians-Universität München Munich Germany
| |
Collapse
|
4
|
Zhao Y, Yoon T, Miller J, Montville CP, Bowden AK. A rapid and low-cost method to fabricate well of the well (WOW) dishes with arbitrary 3D microwell shapes for improved embryo culture. Sci Rep 2024; 14:19757. [PMID: 39187532 PMCID: PMC11347645 DOI: 10.1038/s41598-024-70517-0] [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: 05/24/2024] [Accepted: 08/19/2024] [Indexed: 08/28/2024] Open
Abstract
Despite its high cost, the success rate for in vitro fertilization (IVF) remains < 33% in humans, driving the need for new techniques to improve embryo culture outcomes. The well-of-the-well (WOW) culture system is a platform for in-vitro mammalian embryo culture that has been shown to enhance the developmental competence of embryos and clinical pregnancy rates in humans. However, discovery and testing of the best design for optimal embryo culture quality is hindered by the lack of a method to flexibly produce WOW dishes of various designs. Here, we present a low-cost and simple method to fabricate WOW dishes with microwells of arbitrary shapes and dimensions. We use a low-cost 3D printing service to fabricate a poly(dimethylsiloxane) (PDMS)-based WOW insert that can be paired with a standard in vitro fertilization (IVF) dish to create WOW dishes with new microwell shapes, including pyramidal and hemispherical designs. We validate the fabrication quality of the WOW inserts and demonstrate the utility of the assembled WOW dishes for observation and grading of mouse embryo quality. Moreover, our results indicate that WOW dishes with hemispherical microwells result in better culture outcomes than traditional flat-bottomed IVF dishes and those with other microwell shapes, including the semi-elliptical microwells used in commercial WOW dishes. The proposed fabrication strategy thus provides a way to rapidly fabricate and test new WOW dishes that may bolster IVF success rates.
Collapse
Affiliation(s)
- Yunqin Zhao
- Vanderbilt Biophotonics Center and Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37232, USA
| | - Taehyung Yoon
- Vanderbilt Biophotonics Center and Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37232, USA
| | | | | | - Audrey K Bowden
- Vanderbilt Biophotonics Center and Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
5
|
Porter CM, Qian GC, Grindel SH, Hughes AJ. Highly parallel production of designer organoids by mosaic patterning of progenitors. Cell Syst 2024; 15:649-661.e9. [PMID: 38981488 PMCID: PMC11257788 DOI: 10.1016/j.cels.2024.06.004] [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: 11/14/2023] [Revised: 04/09/2024] [Accepted: 06/17/2024] [Indexed: 07/11/2024]
Abstract
Organoids derived from human stem cells are a promising approach for disease modeling, regenerative medicine, and fundamental research. However, organoid variability and limited control over morphological outcomes remain as challenges. One open question is the extent to which engineering control over culture conditions can guide organoids to specific compositions. Here, we extend a DNA "velcro" cell patterning approach, precisely controlling the number and ratio of human induced pluripotent stem cell-derived progenitors contributing to nephron progenitor (NP) organoids and mosaic NP/ureteric bud (UB) tip cell organoids within arrays of microwells. We demonstrate long-term control over organoid size and morphology, decoupled from geometric constraints. We then show emergent trends in organoid tissue proportions that depend on initial progenitor cell composition. These include higher nephron and stromal cell representation in mosaic NP/UB organoids vs. NP-only organoids and a "goldilocks" initial cell ratio in mosaic organoids that optimizes the formation of proximal tubule structures.
Collapse
Affiliation(s)
- Catherine M Porter
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Soft and Living Matter, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Precision Engineering for Health (CPE4H), University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Grace C Qian
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Samuel H Grindel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Soft and Living Matter, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Precision Engineering for Health (CPE4H), University of Pennsylvania, Philadelphia, PA 19104, USA; Materials Research Science and Engineering Center (MRSEC), University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alex J Hughes
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Soft and Living Matter, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Precision Engineering for Health (CPE4H), University of Pennsylvania, Philadelphia, PA 19104, USA; Materials Research Science and Engineering Center (MRSEC), University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
6
|
Layer PG. In a century from agitated cells to human organoids. J Neurosci Methods 2024; 405:110083. [PMID: 38387805 DOI: 10.1016/j.jneumeth.2024.110083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/09/2024] [Accepted: 02/18/2024] [Indexed: 02/24/2024]
Abstract
Reaching back more than a century, suspension cultures have provided major insights into processes of histogenesis; e.g., cell communication, distinction of self/nonself, cell sorting and cell adhesion. Besides studies on lower animals, the vertebrate retina served as excellent reaggregate model to analyze 3D reconstruction of a complex neural laminar tissue. Methodologically, keeping cells under suspension is essential to achieve tissue organisation in vitro; thereby, the environmental conditions direct the emergent histotypic particulars. Recent progress in regenerative medicine is based to a large extent on human induced pluripotent stem cells (hiPSCs), which are cultured under suspension. Following their genetically directed differentiation into various histologic 3D structures, organoids provide excellent multipurpose in vitro assay models, as well as tissues for repair transplantations. Historically, a nearly fully laminated retinal spheroid from avian embryos was achieved already in 1984, foreshadowing the potential of culturing stem cells under suspension for tissue reconstruction purposes.
Collapse
Affiliation(s)
- Paul Gottlob Layer
- Technical University of Darmstadt, Developmental Biology & Neurogenetics, Schnittspahnstrasse 13, Darmstadt 64297, Germany.
| |
Collapse
|
7
|
Nguyen MA, Dinh NT, Do Thi MH, Nguyen Thi D, Pham UT, Tran TQ, Nguyen VM, Le NH, Nguyen DT, Pham DTN. Simple and Rapid Method of Microwell Array Fabrication for Drug Testing on 3D Cancer Spheroids. ACS OMEGA 2024; 9:16949-16958. [PMID: 38645317 PMCID: PMC11024980 DOI: 10.1021/acsomega.3c05873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/30/2023] [Accepted: 12/12/2023] [Indexed: 04/23/2024]
Abstract
Three-dimensional (3D) cell culture systems are becoming increasingly popular due to their ability to mimic the complex process of angiogenesis in cancer, providing more accurate and physiologically relevant data than traditional two-dimensional (2D) cell culture systems. Microwell systems are particularly useful in this context as they provide a microenvironment that more closely resembles the in vivo environment than traditional microwells. Poly(ethylene glycol) (PEG) microwells are particularly advantageous due to their bio-inertness and the ability to tailor their material characteristics depending on the PEG molecular weight. Although there are several methods available for microwell fabrication, most of them are time-consuming and expensive. The current study utilizes a low-cost laser etching technique on poly(methyl methacrylate) materials followed by molding with PDMS to produce microwells. The optimal conditions for making concave microwells are an engraving parameter speed of 600 mm/s, power of 20%, and a design diameter of the microwell of 0.4 mm. The artificial tumor achieved its full size after 7 days of cell growth in a microwell system, and the cells developed drugs through a live/dead assay test. The results of the drug testing revealed that the IC50 value of zerumbone-loaded liposomes in HepG2 was 4.53 pM, which is greater than the IC50 value of zerumbone. The HepG2 cancer sphere's 3D platform for medication testing revealed that zerumbone-loaded liposomes were very effective at high doses. These findings generally imply that zerumbone-loaded liposomes have the capacity to target the liver and maintain medication delivery.
Collapse
Affiliation(s)
- Mai Anh Nguyen
- Institute
for Tropical Technology, Vietnam Academy
of Science and Technology (VAST), 18 Hoang Quoc Viet st., Cau Giay dist., Hanoi 100000, Vietnam
| | - Nhung Thi Dinh
- Hanoi
University of Science and Technology (HUST), 1 Dai Co Viet st., Hai Ba Trung
dist., Hanoi 100000, Vietnam
| | - My Hanh Do Thi
- Institute
for Tropical Technology, Vietnam Academy
of Science and Technology (VAST), 18 Hoang Quoc Viet st., Cau Giay dist., Hanoi 100000, Vietnam
| | - Dung Nguyen Thi
- Institute
for Tropical Technology, Vietnam Academy
of Science and Technology (VAST), 18 Hoang Quoc Viet st., Cau Giay dist., Hanoi 100000, Vietnam
| | - Uyen Thu Pham
- Institute
for Tropical Technology, Vietnam Academy
of Science and Technology (VAST), 18 Hoang Quoc Viet st., Cau Giay dist., Hanoi 100000, Vietnam
- University
of Science and Technology of Hanoi (USTH), Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet st., Cau Giay
dist., Hanoi 100000, Vietnam
| | - Toan Quoc Tran
- Graduate
University of Science and Technology, Vietnam
Academy of Science and Technology (VAST), 18 Hoang Quoc Viet st., Cau Giay dist., Hanoi 100000, Vietnam
- Institute
of Natural Products Chemistry, Vietnam Academy
of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 100000, Vietnam
| | - Vuong Minh Nguyen
- Institute
of Natural Products Chemistry, Vietnam Academy
of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 100000, Vietnam
| | - Nhung Hong Le
- Graduate
University of Science and Technology, Vietnam
Academy of Science and Technology (VAST), 18 Hoang Quoc Viet st., Cau Giay dist., Hanoi 100000, Vietnam
- Institute
of Natural Products Chemistry, Vietnam Academy
of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 100000, Vietnam
| | - Duong Thanh Nguyen
- Institute
for Tropical Technology, Vietnam Academy
of Science and Technology (VAST), 18 Hoang Quoc Viet st., Cau Giay dist., Hanoi 100000, Vietnam
- Graduate
University of Science and Technology, Vietnam
Academy of Science and Technology (VAST), 18 Hoang Quoc Viet st., Cau Giay dist., Hanoi 100000, Vietnam
| | - Dung Thuy Nguyen Pham
- NTT Institute
of Applied Technology and Sustainable Development, Nguyen Tat Thanh University, Ho
Chi Minh City 70000, Vietnam
- Faculty
of Environmental and Food Engineering, Nguyen
Tat Thanh University, Ho Chi Minh
City 70000, Vietnam
| |
Collapse
|
8
|
Tam NW, Schullian O, Cipitria A, Dimova R. Nonspecific membrane-matrix interactions influence diffusivity of lipid vesicles in hydrogels. Biophys J 2024; 123:638-650. [PMID: 38332584 PMCID: PMC10938137 DOI: 10.1016/j.bpj.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/13/2023] [Accepted: 02/05/2024] [Indexed: 02/10/2024] Open
Abstract
The diffusion of extracellular vesicles and liposomes in vivo is affected by different tissue environmental conditions and is of great interest in the development of liposome-based therapeutics and drug-delivery systems. Here, we use a bottom-up biomimetic approach to better isolate and study steric and electrostatic interactions and their influence on the diffusivity of synthetic large unilamellar vesicles in hydrogel environments. Single-particle tracking of these extracellular vesicle-like particles in agarose hydrogels as an extracellular matrix model shows that membrane deformability and surface charge affect the hydrogel pore spaces that vesicles have access to, which determines overall diffusivity. Moreover, we show that passivation of vesicles with PEGylated lipids, as often used in drug-delivery systems, enhances diffusivity, but that this effect cannot be fully explained with electrostatic interactions alone. Finally, we compare our experimental findings with existing computational and theoretical work in the field to help explain the nonspecific interactions between diffusing particles and gel matrix environments.
Collapse
Affiliation(s)
- Nicky W Tam
- Max Planck Institute of Colloids and Interfaces, Science Park Golm, Potsdam, Germany
| | - Otto Schullian
- Max Planck Institute of Colloids and Interfaces, Science Park Golm, Potsdam, Germany; Free University of Berlin, Department of Physics, Berlin, Germany
| | - Amaia Cipitria
- Max Planck Institute of Colloids and Interfaces, Science Park Golm, Potsdam, Germany; Group of Bioengineering in Regeneration and Cancer, Biogipuzkoa Health Research Institute, San Sebastián, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Rumiana Dimova
- Max Planck Institute of Colloids and Interfaces, Science Park Golm, Potsdam, Germany.
| |
Collapse
|
9
|
Zhu L, Tang Q, Mao Z, Chen H, Wu L, Qin Y. Microfluidic-based platforms for cell-to-cell communication studies. Biofabrication 2023; 16:012005. [PMID: 38035370 DOI: 10.1088/1758-5090/ad1116] [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: 07/22/2023] [Accepted: 11/30/2023] [Indexed: 12/02/2023]
Abstract
Intercellular communication is critical to the understanding of human health and disease progression. However, compared to traditional methods with inefficient analysis, microfluidic co-culture technologies developed for cell-cell communication research can reliably analyze crucial biological processes, such as cell signaling, and monitor dynamic intercellular interactions under reproducible physiological cell co-culture conditions. Moreover, microfluidic-based technologies can achieve precise spatial control of two cell types at the single-cell level with high throughput. Herein, this review focuses on recent advances in microfluidic-based 2D and 3D devices developed to confine two or more heterogeneous cells in the study of intercellular communication and decipher the advantages and limitations of these models in specific cellular research scenarios. This review will stimulate the development of more functionalized microfluidic platforms for biomedical research, inspiring broader interests across various disciplines to better comprehend cell-cell communication and other fields, such as tumor heterogeneity and drug screening.
Collapse
Affiliation(s)
- Lvyang Zhu
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, People's Republic of China
| | - Qu Tang
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, People's Republic of China
| | - Zhenzhen Mao
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, People's Republic of China
| | - Huanhuan Chen
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, People's Republic of China
| | - Li Wu
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, People's Republic of China
| | - Yuling Qin
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, People's Republic of China
| |
Collapse
|
10
|
Han X, Zhang Q, He H, Zhao Q, Li G. Reflow-molded deep concave microwell arrays for robust and large-scale production of embryoid bodies. LAB ON A CHIP 2023; 23:4378-4389. [PMID: 37695312 DOI: 10.1039/d3lc00504f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Embryonic stem cell (ESC)-derived aggregates, called embryoid bodies (EBs), are powerful in vitro models used to study human development and disease. However, the cost-effective and large-scale production of homogeneous EBs still remains a challenge. Here, we report a rapid, straightforward method for fabricating closely arrayed deep concave microwells, enabling the mass production of uniform EBs from single cell suspensions. By simply combining micromilling, caramel replica molding, and thermal reflow, we generate convex micromolds with high aspect ratios and excellent surface smoothness. Benefitting from the nature of reflow, this method can produce rounded bottom polydimethylsiloxane (PDMS) microwells, which are not easily achieved with standard soft lithography techniques but critical to producing spherical EBs. To achieve optimal concave microwells, we investigated the effect of thermal reflow temperature and time on the surface smoothness and roundness of the finished microwells. In addition, to further improve the utility of this method, we also investigated the effect of microwell aspect ratio (AR) on the loss of EBs during medium manipulation. The capability of this deep concave microwell system was validated by rapidly generating a large number of human embryonic stem cell (hESC)-derived EBs and then efficiently differentiating them into a cardiac lineage. The proposed fabrication method and deep concave microwell platform are highly practical, and thus will benefit the mass production of EBs for potential tissue regeneration and cell therapy applications.
Collapse
Affiliation(s)
- Xue Han
- Key Laboratory of Optoelectronic Technology and Systems, Ministry of Education, Defense Key Disciplines Lab of Novel Micro-Nano Devices and System Technology, Chongqing University, Chongqing 400044, China.
| | - Qi Zhang
- Key Laboratory of Optoelectronic Technology and Systems, Ministry of Education, Defense Key Disciplines Lab of Novel Micro-Nano Devices and System Technology, Chongqing University, Chongqing 400044, China.
| | - Hui He
- Institute of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiang Zhao
- Key Laboratory of Optoelectronic Technology and Systems, Ministry of Education, Defense Key Disciplines Lab of Novel Micro-Nano Devices and System Technology, Chongqing University, Chongqing 400044, China.
| | - Gang Li
- Key Laboratory of Optoelectronic Technology and Systems, Ministry of Education, Defense Key Disciplines Lab of Novel Micro-Nano Devices and System Technology, Chongqing University, Chongqing 400044, China.
| |
Collapse
|
11
|
Xie Y, Pan R, Wu S, Yang X, Chen F, Sun W, Yu L. Cell repelling agar@paper interface assisted probing of the tumor spheroids infiltrating natural killer cells. BIOMATERIALS ADVANCES 2023; 153:213507. [PMID: 37354744 DOI: 10.1016/j.bioadv.2023.213507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/03/2023] [Accepted: 06/07/2023] [Indexed: 06/26/2023]
Abstract
Scaffold-based culture is one of the effective methods to resemble three-dimensional (3D) cells model in vitro. An agar@lens paper hybrid scaffold was prepared by one-pot dip-coating. The lens paper's cellulose fiber networks are the scaffold's backbone. The agar gel seized the gaps between the fibrous structures that can improve the paper scaffold's optical transparency and prevent cells from spreading on the scaffold. The SEM and light microscope images showed that the agar gel on the bottom of the paper and the cellulose fiber of the paper formed micro-well structures. Without staining, the cells growing on the agar@paper scaffold can be directly observed under a light microscope. Cells aggregated between the cellulose fibers and formed spheroids within 24 h. The cell spheroids can be non-enzymatically retrieved from the agar@paper scaffold because of the cell-repelling property of agar. The agar@paper scaffold was applied for co-culturing tumor cells (MDA-MB-231, DU 145) and natural killer cells (NKs, NK-92). Using the agar@paper scaffolds, the tumor-infiltrating NKs can be separated from floating NKs that did not attack the tumor spheroids. The effect of NKs infiltrating on tumor spheroids size was characterized. The results showed that NKs attacking the spheroids grown on agar@paper scaffold can be readily tracked because of the improved optical transparency. Higher NKs: tumor cells ratio resulted in a high percentage of tumor infiltrating NKs. The separated NKs can be further tested to reveal their biological characteristics. Both agar and lens paper is accessible for most biological labs, highlighting the potential of agar@paper scaffold in 3D culture.
Collapse
Affiliation(s)
- Yuanyuan Xie
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute for Clean Energy and Advanced Materials, School of Materials and Energy, Southwest University, Chongqing 400715, PR China
| | - Rong Pan
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute for Clean Energy and Advanced Materials, School of Materials and Energy, Southwest University, Chongqing 400715, PR China
| | - Shiming Wu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute for Clean Energy and Advanced Materials, School of Materials and Energy, Southwest University, Chongqing 400715, PR China
| | - Xiaoyan Yang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute for Clean Energy and Advanced Materials, School of Materials and Energy, Southwest University, Chongqing 400715, PR China
| | - Feng Chen
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute for Clean Energy and Advanced Materials, School of Materials and Energy, Southwest University, Chongqing 400715, PR China
| | - Wei Sun
- College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, PR China
| | - Ling Yu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute for Clean Energy and Advanced Materials, School of Materials and Energy, Southwest University, Chongqing 400715, PR China.
| |
Collapse
|
12
|
Strand Z, Schrickel F, Dobiasch S, Thomsen AR, Steiger K, Gempt J, Meyer B, Combs SE, Schilling D. Establishment of a 3D Model to Characterize the Radioresponse of Patient-Derived Glioblastoma Cells. Cancers (Basel) 2023; 15:4051. [PMID: 37627079 PMCID: PMC10452456 DOI: 10.3390/cancers15164051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common malignant primary brain tumor in adults. Despite modern, multimodal therapeutic options of surgery, chemotherapy, tumor-treating fields (TTF), and radiotherapy, the 5-year survival is below 10%. In order to develop new therapies, better preclinical models are needed that mimic the complexity of a tumor. In this work, we established a novel three-dimensional (3D) model for patient-derived GBM cell lines. To analyze the volume and growth pattern of primary GBM cells in 3D culture, a CoSeedisTM culture system was used, and radiation sensitivity in comparison to conventional 2D colony formation assay (CFA) was analyzed. Both culture systems revealed a dose-dependent reduction in survival, but the high variance in colony size and shape prevented reliable evaluation of the 2D cultures. In contrast, the size of 3D spheroids could be measured accurately. Immunostaining of spheroids grown in the 3D culture system showed an increase in the DNA double-strand-break marker γH2AX one hour after irradiation. After 24 h, a decrease in DNA damage was observed, indicating active repair mechanisms. In summary, this new translational 3D model may better reflect the tumor complexity and be useful for analyzing the growth, radiosensitivity, and DNA repair of patient-derived GBM cells.
Collapse
Affiliation(s)
- Zoe Strand
- Department of Radiation Oncology, Klinikum Rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Finn Schrickel
- Department of Radiation Oncology, Klinikum Rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Sophie Dobiasch
- Department of Radiation Oncology, Klinikum Rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
- Institute of Radiation Medicine (IRM), Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Andreas R. Thomsen
- Department of Radiation Oncology, University Medical Center, University of Freiburg, 79106 Freiburg, Germany
| | - Katja Steiger
- Institute of Pathology, Technical University of Munich (TUM), 81675 Munich, Germany
- Comparative Experimental Pathology (CEP), Technical University of Munich (TUM), 81675 Munich, Germany
| | - Jens Gempt
- Department of Neurosurgery, Klinikum Rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Bernhard Meyer
- Department of Neurosurgery, Klinikum Rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Stephanie E. Combs
- Department of Radiation Oncology, Klinikum Rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
- Institute of Radiation Medicine (IRM), Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Daniela Schilling
- Department of Radiation Oncology, Klinikum Rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
- Institute of Radiation Medicine (IRM), Helmholtz Zentrum München, 85764 Neuherberg, Germany
| |
Collapse
|
13
|
Pan R, Yang X, Ning K, Xie Y, Chen F, Yu L. Recapitulating the Drifting and Fusion of Two-Generation Spheroids on Concave Agarose Microwells. Int J Mol Sci 2023; 24:11967. [PMID: 37569343 PMCID: PMC10419262 DOI: 10.3390/ijms241511967] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 08/13/2023] Open
Abstract
Cells with various structures and proteins naturally come together to cooperate in vivo. This study used cell spheroids cultured in agarose micro-wells as a 3D model to study the movement of cells or spheroids toward other spheroids. The formation dynamics of tumor spheroids and the interactions of two batches of cells in the agarose micro-wells were studied. The results showed that a concave bottom micro-well (diameter: 2 mm, depth: 2 mm) prepared from 3% agarose could be used to study the interaction of two batches of cells. The initial tumor cell numbers from 5 × 103 cells/well to 6 × 104 cells/well all could form 3D spheroids after 3 days of incubation. Adding the second batch of DU 145 cells to the existing DU 145 spheroid resulted in the formation of satellite cell spheroids around the existing parental tumor spheroid. Complete fusion of two generation cell spheroids was observed when the parental spheroids were formed from 1 × 104 and 2 × 104 cells, and the second batch of cells was 5 × 103 per well. A higher amount of the second batch of cells (1 × 104 cell/well) led to the formation of independent satellite spheroids after 48 h of co-culture, suggesting the behavior of the second batch of cells towards existing parental spheroids depended on various factors, such as the volume of the parental spheroids and the number of the second batch cells. The interactions between the tumor spheroids and Human Umbilical Vein Endothelial Cells (HUVECs) were modeled on concave agarose micro-wells. The HUVECs (3 × 103 cell/well) were observed to gather around the parental tumor spheroids formed from 1 × 104, 2 × 104, and 3 × 104 cells per well rather than aggregate on their own to form HUVEC spheroids. This study highlights the importance of analyzing the biological properties of cells before designing experimental procedures for the sequential fusion of cell spheroids. The study further emphasizes the significant roles that cell density and the volume of the spheroids play in determining the location and movement of cells.
Collapse
Affiliation(s)
| | | | | | | | | | - Ling Yu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute for Clean Energy and Advanced Materials, School of Materials and Energy, Southwest University, Chongqing 400715, China; (R.P.); (X.Y.); (K.N.); (Y.X.); (F.C.)
| |
Collapse
|
14
|
Do TD, Pham UT, Nguyen LP, Nguyen TM, Bui CN, Oliver S, Pham P, Tran TQ, Hoang BT, Pham MTH, Pham DTN, Nguyen DT. Fabrication of a Low-Cost Microfluidic Device for High-Throughput Drug Testing on Static and Dynamic Cancer Spheroid Culture Models. Diagnostics (Basel) 2023; 13:diagnostics13081394. [PMID: 37189495 DOI: 10.3390/diagnostics13081394] [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: 03/02/2023] [Revised: 03/17/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
Drug development is a complex and expensive process from new drug discovery to product approval. Most drug screening and testing rely on in vitro 2D cell culture models; however, they generally lack in vivo tissue microarchitecture and physiological functionality. Therefore, many researchers have used engineering methods, such as microfluidic devices, to culture 3D cells in dynamic conditions. In this study, a simple and low-cost microfluidic device was fabricated using Poly Methyl Methacrylate (PMMA), a widely available material, and the total cost of the completed device was USD 17.75. Dynamic and static cell culture examinations were applied to monitor the growth of 3D cells. α-MG-loaded GA liposomes were used as the drug to test cell viability in 3D cancer spheroids. Two cell culture conditions (i.e., static and dynamic) were also used in drug testing to simulate the effect of flow on drug cytotoxicity. Results from all assays showed that with the velocity of 0.005 mL/min, cell viability was significantly impaired to nearly 30% after 72 h in a dynamic culture. This device is expected to improve in vitro testing models, reduce and eliminate unsuitable compounds, and select more accurate combinations for in vivo testing.
Collapse
Affiliation(s)
- Tung Dinh Do
- Saint Paul General Hospital, No. 12, Chu Van An St., Ba Dinh Dist, Ha Noi 10000, Vietnam
| | - Uyen Thu Pham
- Institute for Tropical Technology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
| | - Linh Phuong Nguyen
- School of Preventive Medicine and Public Health, Hanoi Medical University, 1 Ton That Tung St., Dong Da Dist., Hanoi 10000, Vietnam
| | - Trang Minh Nguyen
- Institute for Tropical Technology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
| | - Cuong Nguyen Bui
- Hung Yen University of Technology and Education (UTEHY), 39A St., Khoai Chau Dist., Hung Yen 17000, Vietnam
| | - Susan Oliver
- Centre for Advanced Macromolecular Design and Australian Centre for NanoMedicine, School of Chemical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Phuong Pham
- Centre for Advanced Macromolecular Design and Australian Centre for NanoMedicine, School of Chemical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Toan Quoc Tran
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
- Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
| | - Bich Thi Hoang
- Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
| | - Minh Thi Hong Pham
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
- Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
| | - Dung Thuy Nguyen Pham
- Institute of Applied Technology and Sustainable Development, Nguyen Tat Thanh University, Ho Chi Minh City 70000, Vietnam
- Faculty of Environmental and Food Engineering, Nguyen Tat Thanh University, Ho Chi Minh City 70000, Vietnam
| | - Duong Thanh Nguyen
- Institute for Tropical Technology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 10000, Vietnam
| |
Collapse
|
15
|
Zhu T, Nie J, Yu T, Zhu D, Huang Y, Chen Z, Gu Z, Tang J, Li D, Fei P. Large-scale high-throughput 3D culture, imaging, and analysis of cell spheroids using microchip-enhanced light-sheet microscopy. BIOMEDICAL OPTICS EXPRESS 2023; 14:1659-1669. [PMID: 37078040 PMCID: PMC10110308 DOI: 10.1364/boe.485217] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/24/2023] [Accepted: 03/06/2023] [Indexed: 05/03/2023]
Abstract
Light sheet microscopy combined with a microchip is an emerging tool in biomedical research that notably improves efficiency. However, microchip-enhanced light-sheet microscopy is limited by noticeable aberrations induced by the complex refractive indices in the chip. Herein, we report a droplet microchip that is specifically engineered to be capable of large-scale culture of 3D spheroids (over 600 samples per chip) and has a polymer index matched to water (difference <1%). When combined with a lab-built open-top light-sheet microscope, this microchip-enhanced microscopy technique allows 3D time-lapse imaging of the cultivated spheroids with ∼2.5-µm single-cell resolution and a high throughput of ∼120 spheroids per minute. This technique was validated by a comparative study on the proliferation and apoptosis rates of hundreds of spheroids with or without treatment with the apoptosis-inducing drug Staurosporine.
Collapse
Affiliation(s)
- Tingting Zhu
- School of Optical and Electronic Information - Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jun Nie
- Institute for Cell Analysis, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Tingting Yu
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Dan Zhu
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Yanyi Huang
- Institute for Cell Analysis, Shenzhen Bay Laboratory, Shenzhen 518132, China
- College of Chemistry, Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing 100871, China
| | - Zaozao Chen
- State Key Laboratory of Bioelectronics School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Jiang Tang
- School of Optical and Electronic Information - Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Dongyu Li
- School of Optical and Electronic Information - Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Peng Fei
- School of Optical and Electronic Information - Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
16
|
Zhang J, Xue J, Luo N, Chen F, Chen B, Zhao Y. Microwell array chip-based single-cell analysis. LAB ON A CHIP 2023; 23:1066-1079. [PMID: 36625143 DOI: 10.1039/d2lc00667g] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Single-cell profiling is key to uncover the cellular heterogeneity and drives deep understanding of cell fate. In recent years, microfluidics has become an ideal tool for single-cell profiling owing to its benefits of high throughput and automation. Among various microfluidic platforms, microwell has the advantages of simple operation and easy integration with in situ analysis ability, making it an ideal technique for single-cell studies. Herein, recent advances of single-cell analysis based on microwell array chips are summarized. We first introduce the design and preparation of different microwell chips. Then microwell-based cell capture and lysis strategies are discussed. We finally focus on advanced microwell-based analysis of single-cell proteins, nucleic acids, and metabolites. The challenges and opportunities for the development of microwell-based single-cell analysis are also presented.
Collapse
Affiliation(s)
- Jin Zhang
- Institute of Analytical Chemistry and Instrument for Life Science, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P. R. China.
| | - Jing Xue
- Institute of Analytical Chemistry and Instrument for Life Science, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P. R. China.
| | - Ningfeng Luo
- Institute of Analytical Chemistry and Instrument for Life Science, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P. R. China.
| | - Feng Chen
- Institute of Analytical Chemistry and Instrument for Life Science, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P. R. China.
| | - Badong Chen
- Institute of Artificial Intelligence and Robotics and the College of Artificial Intelligence, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P. R. China.
| | - Yongxi Zhao
- Institute of Analytical Chemistry and Instrument for Life Science, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P. R. China.
| |
Collapse
|
17
|
McKenna MK, Ozcan A, Brenner D, Watanabe N, Legendre M, Thomas DG, Ashwood C, Cummings RD, Bonifant C, Markovitz DM, Brenner MK. Novel banana lectin CAR-T cells to target pancreatic tumors and tumor-associated stroma. J Immunother Cancer 2023; 11:e005891. [PMID: 36653070 PMCID: PMC9853244 DOI: 10.1136/jitc-2022-005891] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Cell therapies for solid tumors are thwarted by the hostile tumor microenvironment (TME) and by heterogeneous expression of tumor target antigens. We address both limitations with a novel class of chimeric antigen receptors based on plant lectins, which recognize the aberrant sugar residues that are a 'hallmark' of both malignant and associated stromal cells. We have expressed in T cells a modified lectin from banana, H84T BanLec, attached to a chimeric antigen receptor (H84T-CAR) that recognizes high-mannose (asparagine residue with five to nine mannoses). Here, we tested the efficacy of our novel H84T CAR in models of pancreatic ductal adenocarcinoma (PDAC), intractable tumors with aberrant glycosylation and characterized by desmoplastic stroma largely contributed by pancreatic stellate cells (PSCs). METHODS We transduced human T cells with a second-generation retroviral construct expressing the H84T BanLec chimeric receptor, measured T-cell expansion, characterized T-cell phenotype, and tested their efficacy against PDAC tumor cells lines by flow cytometry quantification. In three-dimensional (3D) spheroid models, we measured H84T CAR T-cell disruption of PSC architecture, and T-cell infiltration by live imaging. We tested the activity of H84T CAR T cells against tumor xenografts derived from three PDAC cell lines. Antitumor activity was quantified by caliper measurement and bioluminescence signal and used anti-human vimentin to measure residual PSCs. RESULTS H84T BanLec CAR was successfully transduced and expressed by T cells which had robust expansion and retained central memory phenotype in both CD4 and CD8 compartments. H84T CAR T cells targeted and eliminated PDAC tumor cell lines. They also disrupted PSC architecture in 3D models in vitro and reduced total tumor and stroma cells in mixed co-cultures. H84T CAR T cells exhibited improved T-cell infiltration in multicellular spheroids and had potent antitumor effects in the xenograft models. We observed no adverse effects against normal tissues. CONCLUSIONS T cells expressing H84T CAR target malignant cells and their stroma in PDAC tumor models. The incorporation of glycan-targeting lectins within CARs thus extends their activity to include both malignant cells and their supporting stromal cells, disrupting the TME that otherwise diminishes the activity of cellular therapies against solid tumors.
Collapse
Affiliation(s)
- Mary K McKenna
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Ada Ozcan
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Daniel Brenner
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Norihiro Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Maureen Legendre
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Dafydd G Thomas
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Richard D Cummings
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Challice Bonifant
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David M Markovitz
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Malcolm K Brenner
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
18
|
Guo W, Chen Z, Feng Z, Li H, Zhang M, Zhang H, Cui X. Fabrication of Concave Microwells and Their Applications in Micro-Tissue Engineering: A Review. MICROMACHINES 2022; 13:mi13091555. [PMID: 36144178 PMCID: PMC9505614 DOI: 10.3390/mi13091555] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 05/27/2023]
Abstract
At present, there is an increasing need to mimic the in vivo micro-environment in the culture of cells and tissues in micro-tissue engineering. Concave microwells are becoming increasingly popular since they can provide a micro-environment that is closer to the in vivo environment compared to traditional microwells, which can facilitate the culture of cells and tissues. Here, we will summarize the fabrication methods of concave microwells, as well as their applications in micro-tissue engineering. The fabrication methods of concave microwells include traditional methods, such as lithography and etching, thermal reflow of photoresist, laser ablation, precision-computerized numerical control (CNC) milling, and emerging technologies, such as surface tension methods, the deformation of soft membranes, 3D printing, the molding of microbeads, air bubbles, and frozen droplets. The fabrication of concave microwells is transferring from professional microfabrication labs to common biochemical labs to facilitate their applications and provide convenience for users. Concave microwells have mostly been used in organ-on-a-chip models, including the formation and culture of 3D cell aggregates (spheroids, organoids, and embryoids). Researchers have also used microwells to study the influence of substrate topology on cellular behaviors. We will briefly review their applications in different aspects of micro-tissue engineering and discuss the further applications of concave microwells. We believe that building multiorgan-on-a-chip by 3D cell aggregates of different cell lines will be a popular application of concave microwells, while integrating physiologically relevant molecular analyses with the 3D culture platform will be another popular application in the near future. Furthermore, 3D cell aggregates from these biosystems will find more applications in drug screening and xenogeneic implantation.
Collapse
Affiliation(s)
- Weijin Guo
- Department of Biomedical Engineering, Shantou University, Shantou 515063, China
| | - Zejingqiu Chen
- Department of Biology, Shantou University, Shantou 515063, China
| | - Zitao Feng
- Department of Biomedical Engineering, Shantou University, Shantou 515063, China
| | - Haonan Li
- Department of Electrical Engineering, Shantou University, Shantou 515063, China
| | - Muyang Zhang
- Department of Electrical Engineering, Shantou University, Shantou 515063, China
| | - Huiru Zhang
- Guangdong Foshan Lianchuang Graduate School of Engineering, Foshan 528311, China
| | - Xin Cui
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| |
Collapse
|
19
|
He J, Zhou C, Xu X, Zhou Z, Danoy M, Shinohara M, Xiao W, Zhu D, Zhao X, Feng X, Mao Y, Sun W, Sakai Y, Yang H, Pang Y. Scalable Formation of Highly Viable and Functional Hepatocellular Carcinoma Spheroids in an Oxygen-Permeable Microwell Device for Anti-Tumor Drug Evaluation. Adv Healthc Mater 2022; 11:e2200863. [PMID: 35841538 DOI: 10.1002/adhm.202200863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/30/2022] [Indexed: 01/27/2023]
Abstract
For high-throughput anti-cancer drug screening, microwell arrays may serve as an effective tool to generate uniform and scalable tumor spheroids. However, microwell arrays are commonly anchored in non-oxygen-permeable culture plates, leading to limited oxygen supply for avascular spheroids. Herein, a polydimethylsiloxane (PDMS)-based oxygen-permeable microwell device is introduced for generating highly viable and functional hepatocellular carcinoma (HCC) spheroids. The PDMS sheets at the bottom of the microwell device provide a high flux of oxygen like in vivo neighboring hepatic sinusoids. Owing to the better oxygen supply, the generated HepG2 spheroids are larger in size and exhibit higher viability and proliferation with less cell apoptosis and necrosis. These spheroids also exhibit lower levels of anaerobic cellular respiration and express higher levels of liver-related functions. In anti-cancer drug testing, spheroids cultured in PDMS plates show a significantly stronger resistance against doxorubicin because of the stronger stem-cell and multidrug resistance phenotype. Moreover, higher expression of vascular endothelial growth factor-A produces a stronger angiogenesis capability of the spheroids. Overall, compared to the spheroids cultured in conventional non-oxygen-permeable plates, these spheroids can be used as a more favorable model for early-stage HCCs and be applied in high-throughput anti-cancer drug screening.
Collapse
Affiliation(s)
- Jianyu He
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, 100084, P. R. China.,Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Tsinghua University, Beijing, 100084, P. R. China.,Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, 100084, P. R. China
| | - Chang Zhou
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, 100084, P. R. China.,Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Tsinghua University, Beijing, 100084, P. R. China.,Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, 100084, P. R. China
| | - Xiaolei Xu
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, 100084, P. R. China.,Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Tsinghua University, Beijing, 100084, P. R. China.,Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, 100084, P. R. China.,Department of Hepatobiliary Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Changping District, Beijing, 102218, P. R. China
| | - Zhenzhen Zhou
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, 100084, P. R. China.,Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Tsinghua University, Beijing, 100084, P. R. China.,Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, 100084, P. R. China
| | - Mathieu Danoy
- Department of Chemical System Engineering, Graduate School of Engineering, University of Tokyo, Tokyo, 113-033, Japan
| | - Marie Shinohara
- Institute of Industrial Science, University of Tokyo, Tokyo, 153-8505, Japan
| | - Wenjin Xiao
- Centre de Recherche des Cordeliers, INSERM UMR-S1138, CNRS SNC5014, University of Paris, Paris, 75006, France
| | - Dong Zhu
- Clinical Laboratory, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Changping District, Beijing, 102218, P. R. China
| | - Xiuying Zhao
- Clinical Laboratory, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Changping District, Beijing, 102218, P. R. China
| | - Xiaobin Feng
- Department of Hepatobiliary Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Changping District, Beijing, 102218, P. R. China
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Dongcheng District, Beijing, 100005, P. R. China
| | - Wei Sun
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, 100084, P. R. China.,Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Tsinghua University, Beijing, 100084, P. R. China.,Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, 100084, P. R. China.,Department of Mechanical Engineering and Mechanics, College of Engineering, Drexel University, Philadelphia, PA, 19104, USA
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, Graduate School of Engineering, University of Tokyo, Tokyo, 113-033, Japan
| | - Huayu Yang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Dongcheng District, Beijing, 100005, P. R. China
| | - Yuan Pang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, 100084, P. R. China.,Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Tsinghua University, Beijing, 100084, P. R. China.,Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, 100084, P. R. China
| |
Collapse
|
20
|
Human Mesenchymal Stromal Cells Do Not Cause Radioprotection of Head-and-Neck Squamous Cell Carcinoma. Int J Mol Sci 2022; 23:ijms23147689. [PMID: 35887032 PMCID: PMC9323822 DOI: 10.3390/ijms23147689] [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: 04/29/2022] [Revised: 07/03/2022] [Accepted: 07/09/2022] [Indexed: 02/01/2023] Open
Abstract
Radiotherapy of head-and-neck squamous cell carcinoma (HNSCC) can cause considerable normal tissue injuries, and mesenchymal stromal cells (MSCs) have been shown to aid regeneration of irradiation-damaged normal tissues. However, utilization of MSC-based treatments for HNSCC patients undergoing radiotherapy is hampered by concerns regarding potential radioprotective effects. We therefore investigated the influence of MSCs on the radiosensitivity of HNSCCs. Several human papillomavirus (HPV)-negative and HPV-positive HNSCCs were co-cultured with human bone marrow-derived MSCs using two-dimensional and three-dimensional assays. Clonogenic survival, proliferation, and viability of HNSCCs after radiotherapy were assessed depending on MSC co-culture. Flow cytometry analyses were conducted to examine the influence of MSCs on irradiation-induced cell cycle distribution and apoptosis induction in HNSCCs. Immunofluorescence stainings of γH2AX were conducted to determine the levels of residual irradiation-induced DNA double-strand breaks. Levels of connective tissue growth factor (CTGF), a multifunctional pro-tumorigenic cytokine, were analyzed using enzyme-linked immunosorbent assays. Neither direct MSC co-culture nor MSC-conditioned medium exerted radioprotective effects on HNSCCs as determined by clonogenic survival, proliferation, and viability assays. Consistently, three-dimensional microwell arrays revealed no radioprotective effects of MSCs. Irradiation resulted in a G2/M arrest of HNSCCs at 96 h independently of MSC co-culture. HNSCCs’ apoptosis rates were increased by irradiation irrespective of MSCs. Numbers of residual γH2AX foci after irradiation with 2 or 8 Gy were comparable between mono- and co-cultures. MSC mono-cultures and HNSCC-MSC co-cultures exhibited comparable CTGF levels. We did not detect radioprotective effects of human MSCs on HNSCCs. Our results suggest that the usage of MSC-based therapies for radiotherapy-related toxicities in HNSCC patients may be safe in the context of absent radioprotection.
Collapse
|
21
|
Coplanar embedding of multiple 3D cell models in hydrogel towards high-throughput micro-histology. Sci Rep 2022; 12:9991. [PMID: 35705590 PMCID: PMC9200833 DOI: 10.1038/s41598-022-13987-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 05/31/2022] [Indexed: 11/25/2022] Open
Abstract
Standardised and high-throughput methods have been developed for the production and experimental handling of some 3D in vitro models. However, adapted analytical tools are still missing for scientists and researchers to fully exploit the potential of complex cellular models in pre-clinical drug testing and precision medicine. Histology is the established, cost-effective and gold standard method for structural and functional tissue analysis. However, standard histological processes are challenging and costly to apply to 3D cell models, as their small size often leads to poor alignment of samples, which lowers analysis throughput. This body of work proposes a new approach: HistoBrick facilitates histological processing of spheroids and organoids by enabling gel embedding of 3D cell models with precise coplanar alignment, parallel to the sectioning plane, thus minimising the loss of sample material. HistoBrick’s features are compatible with automation standards, potentially allowing automated sample transfer from a multi-well plate to the gel device. Moreover, HistoBrick’s technology was validated by demonstrating the alignment of HepG2 cultured spheroids measuring 150–200 µm in diameter with a height precision of ± 80 µm. HistoBrick allows up to 96 samples to be studied across minimal sections, paving the way towards high-throughput micro-histology.
Collapse
|
22
|
Zimmermann CE, Mackens-Kiani L, Acil Y, Terheyden H. Characterization of porcine mesenchymal stromal cells and their proliferative and osteogenic potential in long-term culture. J Stem Cells Regen Med 2022; 17:49-55. [PMID: 35250201 DOI: 10.46582/jsrm.1702008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/07/2021] [Indexed: 12/29/2022]
Abstract
Background: Porcine mesenchymal stromal cells (pMSCs) are considered a valuable research model for bone tissue engineering, which requires adequate amounts of viable cells with sufficient potential for osteogenic differentiation. For isolation and expansion of these cells through long-term culture, appropriate culture conditions are needed. Objective: To study the effect of extended in vitro cultivation on pMSC proliferation and differentiation potential using different osteogenic and adipogenic induction media. Methods: pMSCs were isolated from the bone marrow of adult Göttingen minipigs, cultured, expanded to passage 20 (~160 days) and characterized by their expression of cell surface markers (wCD44, CD45, CD90, SWC9, fibronectin), alkaline phosphatase (ALP), and osteocalcin and their potential for osteogenic and adipogenic differentiation using different induction media. Results: pMSCs retained their capacity for proliferation and osteogenic differentiation, and the number of CD90-positive cells increased significantly over more than 60 population doublings. CD90 expression in uninduced cells correlated strongly with ALP expression following osteogenic induction. Medium enriched with calcium yielded a stronger osteogenic response. Conclusion: The selection of CD90-positive MSCs and adequate levels of calcium seem to enhance the osteogenic phenotype for bone tissue engineering.
Collapse
Affiliation(s)
- Corinna E Zimmermann
- Department of Craniomaxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Strasse 3, 24105 Kiel, Germany.,University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | | | - Yahya Acil
- Department of Craniomaxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Strasse 3, 24105 Kiel, Germany
| | - Hendrik Terheyden
- Department of Craniomaxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Strasse 3, 24105 Kiel, Germany
| |
Collapse
|
23
|
Thomsen A, Aldrian C, Luka B, Hornhardt S, Gomolka M, Moertl S, Hess J, Zitzelsberger H, Heider T, Schlueter N, Rau S, Monroy Ordonez B, Schäfer H, Rücker G, Henke M. Biopsy-Derived Oral Keratinocytes – a Model to Potentially Test for Oral Mucosa Radiation Sensitivity. Clin Transl Radiat Oncol 2022; 34:51-56. [PMID: 35345866 PMCID: PMC8956846 DOI: 10.1016/j.ctro.2022.03.007] [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: 01/19/2022] [Revised: 03/13/2022] [Accepted: 03/14/2022] [Indexed: 11/29/2022] Open
Abstract
Human oral keratinocytes – the key players in radiation mucositis in head and neck cancer treatment – are established ex vivo from patient-derived micro-biopsies. Individual radiosensitivity of primary oral keratinocytes is measured by a novel assay for cellular proliferation and spreading. The keratinocyte model also supports classical functional assays such as clonogenic survival and DNA double strand repair.
Purpose To establish stable in vitro growth of keratinocytes from very small biopsy specimens and successfully apply new test systems to determine their radiosensitivity. Materials and Methods Oral mucosa biopsies (diameter: 1.7 mm) from 15 subjects were immobilized with custom-made cups onto culture plates. Outgrowing cells were tested for cytokeratin 5/14 and Ki67, expanded, radiated at different doses, and seeded onto circumscribed areas before being allowed to spread centrifugally. In this newly developed spreading assay, cell-covered areas were measured by image analysis. For statistical analysis, a linear mixed regression model was used; additionally, results were correlated to the radiation dose applied. Colony forming efficiency (CFE) was used to validate the results. DNA damage repair was analysed by gammaH2AX and 53BP1 foci quantification using immunofluorescence microscopy 24 h and 96 h after irradiation. Results Stable keratinocyte growth continued for up to 7 weeks in 14 biopsies. Cells spread reliably from an initial 16.6 mm2 up to a median of 119.2 mm2 (range: 54.4–290). Radiated cells spread to only 100.7 mm2 (2 Gy; range: 55.3–266.7); 73.2 mm2 (4 Gy; 15–240.4); 47 mm2 (6 Gy; 2–111.9), and 22.7 mm2 (8 Gy; 0–80). Similarly, CFE decreased from 0.223 (0 Gy) to 0.0028 (8 Gy). Using an individual donor as a random factor, cell spread correlated with CFE, where radiation dose was the main driver (decrease by 0.50, adjusted for area). Upon irradiation with 6 Gy, radiation-induced DNA damage was increased after 24 h in all samples, and even after 96 h in 5 out of 7 samples, as detected by a higher number of gammaH2AX/53BP1 foci in irradiated cells (mean 3.7 for 24 h; mean 0.6 for 96 h). Conclusion In vitro propagation of keratinocytes derived from a small biopsy is feasible. Radiation impairs cellular migration and proliferation, and the newly described spreading assay allows ranking for cellular radioresistance. The keratinocyte model also supports classical functional assays such as clonogenic survival and DNA double strand repair. The clinical relevance awaits upcoming investigations.
Collapse
Affiliation(s)
- A.R. Thomsen
- Department of Radiation Oncology, University Medical Center, University of Freiburg, Freiburg/Breisgau, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (dkfz), Heidelberg, Germany
- Corresponding author at: Department of Radiation Oncology, University Medical Center, University of Freiburg, Freiburg/Breisgau, Germany.
| | - C. Aldrian
- Department of Radiation Oncology, University Medical Center, University of Freiburg, Freiburg/Breisgau, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (dkfz), Heidelberg, Germany
| | - B. Luka
- Division for Cariology, Department of Operative Dentistry and Periodontology, Center for Dental Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - S. Hornhardt
- Federal Office for Radiation Protection, Ingolstädter Landstr. 1, 85764 Oberschleißheim, Germany
| | - M. Gomolka
- Federal Office for Radiation Protection, Ingolstädter Landstr. 1, 85764 Oberschleißheim, Germany
| | - S. Moertl
- Federal Office for Radiation Protection, Ingolstädter Landstr. 1, 85764 Oberschleißheim, Germany
| | - J. Hess
- Research Unit Radiation Cytogenetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
- Clinical Cooperation Group “Personalized Radiotherapy in Head and Neck Cancer”, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - H. Zitzelsberger
- Research Unit Radiation Cytogenetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - T. Heider
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - N. Schlueter
- Division for Cariology, Department of Operative Dentistry and Periodontology, Center for Dental Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - S. Rau
- Division for Cariology, Department of Operative Dentistry and Periodontology, Center for Dental Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - B. Monroy Ordonez
- Department of Radiation Oncology, University Medical Center, University of Freiburg, Freiburg/Breisgau, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (dkfz), Heidelberg, Germany
| | - H. Schäfer
- Department of Radiation Oncology, University Medical Center, University of Freiburg, Freiburg/Breisgau, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (dkfz), Heidelberg, Germany
| | - G. Rücker
- Institute for Medical Biometry and Statistics, Medical Center – University of Freiburg, Germany
| | - M. Henke
- Department of Radiation Oncology, University Medical Center, University of Freiburg, Freiburg/Breisgau, Germany
| |
Collapse
|
24
|
Barhouse PS, Andrade MJ, Smith Q. Home Away From Home: Bioengineering Advancements to Mimic the Developmental and Adult Stem Cell Niche. FRONTIERS IN CHEMICAL ENGINEERING 2022. [DOI: 10.3389/fceng.2022.832754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The inherent self-organizing capacity of pluripotent and adult stem cell populations has advanced our fundamental understanding of processes that drive human development, homeostasis, regeneration, and disease progression. Translating these principles into in vitro model systems has been achieved with the advent of organoid technology, driving innovation to harness patient-specific, cell-laden regenerative constructs that can be engineered to augment or replace diseased tissue. While developmental organization and regenerative adult stem cell niches are tightly regulated in vivo, in vitro analogs lack defined architecture and presentation of physicochemical cues, leading to the unhindered arrangement of mini-tissues that lack complete physiological mimicry. This review aims to highlight the recent integrative engineering approaches that elicit spatio-temporal control of the extracellular niche to direct the structural and functional maturation of pluripotent and adult stem cell derivatives. While the advances presented here leverage multi-pronged strategies ranging from synthetic biology to microfabrication technologies, the methods converge on recreating the biochemical and biophysical milieu of the native tissue to be modeled or regenerated.
Collapse
|
25
|
Waldschmidt JM, Fruttiger SJ, Wider D, Jung J, Thomsen AR, Hartmann TN, Duyster J, Hug MJ, Azab KA, Jung M, Wäsch R, Engelhardt M. Ex vivo propagation in a novel 3D high-throughput co-culture system for multiple myeloma. J Cancer Res Clin Oncol 2022; 148:1045-1055. [PMID: 35072775 PMCID: PMC9016043 DOI: 10.1007/s00432-021-03854-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 11/04/2021] [Indexed: 12/28/2022]
Abstract
Purpose Multiple myeloma (MM) remains an incurable hematologic malignancy which ultimately develops drug resistance and evades treatment. Despite substantial therapeutic advances over the past years, the clinical failure rate of preclinically promising anti-MM drugs remains substantial. More realistic in vitro models are thus required to better predict clinical efficacy of a preclinically active compound. Methods Here, we report on the establishment of a conical agarose 3D co-culture platform for the preclinical propagation of primary MM cells ex vivo. Cell growth was compared to yet established 2D and liquid overlay systems. MM cell lines (MMCL: RPMI-8226, U266, OPM-2) and primary patient specimens were tested. Drug sensitivity was examined by exploring the cytotoxic effect of bortezomib and the deubiquitinase inhibitor auranofin under various conditions. Results In contrast to 2D and liquid overlay, cell proliferation in the 3D array followed a sigmoidal curve characterized by an initial growth delay but more durable proliferation of MMCL over 12 days of culture. Primary MM specimens did not expand in ex vivo monoculture, but required co-culture support by a human stromal cell line (HS-5, MSP-1). HS-5 induced a > fivefold increase in cluster volume and maintained long-term viability of primary MM cells for up to 21 days. Bortezomib and auranofin induced less cytotoxicity under 3D vs. 2D condition and in co- vs. monoculture, respectively. Conclusions This study introduces a novel model that is capable of long-term propagation and drug testing of primary MM specimens ex vivo overcoming some of the pitfalls of currently available in vitro models. Supplementary Information The online version contains supplementary material available at 10.1007/s00432-021-03854-6.
Collapse
Affiliation(s)
- Johannes M Waldschmidt
- Department of Internal Medicine I, Faculty of Medicine and Medical Center, University of Freiburg, Hugstetterstr. 53, 79106, Freiburg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Freiburg University Medical Center, Freiburg, Germany
| | - Stefan J Fruttiger
- Department of Internal Medicine I, Faculty of Medicine and Medical Center, University of Freiburg, Hugstetterstr. 53, 79106, Freiburg, Germany
- Pharmacy, Freiburg University Medical Center, Freiburg, Germany
| | - Dagmar Wider
- Department of Internal Medicine I, Faculty of Medicine and Medical Center, University of Freiburg, Hugstetterstr. 53, 79106, Freiburg, Germany
| | - Johannes Jung
- Department of Internal Medicine I, Faculty of Medicine and Medical Center, University of Freiburg, Hugstetterstr. 53, 79106, Freiburg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Freiburg University Medical Center, Freiburg, Germany
| | - Andreas R Thomsen
- Department of Radiation Oncology, Freiburg University Medical Center, Freiburg, Germany
| | - Tanja N Hartmann
- Department of Internal Medicine I, Faculty of Medicine and Medical Center, University of Freiburg, Hugstetterstr. 53, 79106, Freiburg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Freiburg University Medical Center, Freiburg, Germany
| | - Justus Duyster
- Department of Internal Medicine I, Faculty of Medicine and Medical Center, University of Freiburg, Hugstetterstr. 53, 79106, Freiburg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Freiburg University Medical Center, Freiburg, Germany
| | - Martin J Hug
- Pharmacy, Freiburg University Medical Center, Freiburg, Germany
| | - Kareem A Azab
- Department of Radiation Oncology, Washington University, St. Louis, MO, USA
| | - Manfred Jung
- Institute of Pharmaceutical Sciences, University of Freiburg, Freiburg, Germany
| | - Ralph Wäsch
- Department of Internal Medicine I, Faculty of Medicine and Medical Center, University of Freiburg, Hugstetterstr. 53, 79106, Freiburg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Freiburg University Medical Center, Freiburg, Germany
| | - Monika Engelhardt
- Department of Internal Medicine I, Faculty of Medicine and Medical Center, University of Freiburg, Hugstetterstr. 53, 79106, Freiburg, Germany.
- Comprehensive Cancer Center Freiburg (CCCF), Freiburg University Medical Center, Freiburg, Germany.
| |
Collapse
|
26
|
Decarli MC, de Castro MV, Nogueira JA, Nagahara MHT, Westin CB, de Oliveira ALR, Silva JVL, Moroni L, Mota C, Moraes ÂM. Development of a device useful to reproducibly produce large quantities of viable and uniform stem cell spheroids with controlled diameters. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 135:112685. [DOI: 10.1016/j.msec.2022.112685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/20/2021] [Accepted: 01/22/2022] [Indexed: 01/08/2023]
|
27
|
Development of a Simple Spheroid Production Method Using Fluoropolymers with Reduced Chemical and Physical Damage. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app112110495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Establishing an in vitro–based cell culture system that can realistically simulate in vivo cell dynamics is desirable. It is thus necessary to develop a method for producing a large amount of cell aggregates (i.e., spheroids) that are uniform in size and quality. Various methods have been proposed for the preparation of spheroids; however, none of them satisfy all requirements, such as cost, size uniformity, and throughput. Herein, we successfully developed a new cell culture method by combining fluoropolymers and dot patterned extracellular matrix substrates to achieve size-controlled spheroids. First, the spheroids were spontaneously formed by culturing them two-dimensionally, after which the cells were detached with a weak liquid flow and cultured in suspension without enzyme treatment. Stable quality spheroids were easily produced, and it is expected that the introduction and running costs of the technique will be low; therefore, this method shows potential for application in the field of regenerative medicine.
Collapse
|
28
|
Zhuang P, Chiang YH, Fernanda MS, He M. Using Spheroids as Building Blocks Towards 3D Bioprinting of Tumor Microenvironment. Int J Bioprint 2021; 7:444. [PMID: 34805601 PMCID: PMC8600307 DOI: 10.18063/ijb.v7i4.444] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/02/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer still ranks as a leading cause of mortality worldwide. Although considerable efforts have been dedicated to anticancer therapeutics, progress is still slow, partially due to the absence of robust prediction models. Multicellular tumor spheroids, as a major three-dimensional (3D) culture model exhibiting features of avascular tumors, gained great popularity in pathophysiological studies and high throughput drug screening. However, limited control over cellular and structural organization is still the key challenge in achieving in vivo like tissue microenvironment. 3D bioprinting has made great strides toward tissue/organ mimicry, due to its outstanding spatial control through combining both cells and materials, scalability, and reproducibility. Prospectively, harnessing the power from both 3D bioprinting and multicellular spheroids would likely generate more faithful tumor models and advance our understanding on the mechanism of tumor progression. In this review, the emerging concept on using spheroids as a building block in 3D bioprinting for tumor modeling is illustrated. We begin by describing the context of the tumor microenvironment, followed by an introduction of various methodologies for tumor spheroid formation, with their specific merits and drawbacks. Thereafter, we present an overview of existing 3D printed tumor models using spheroids as a focus. We provide a compilation of the contemporary literature sources and summarize the overall advancements in technology and possibilities of using spheroids as building blocks in 3D printed tissue modeling, with a particular emphasis on tumor models. Future outlooks about the wonderous advancements of integrated 3D spheroidal printing conclude this review.
Collapse
Affiliation(s)
- Pei Zhuang
- Department of Pharmaceutics, University of Florida, Gainesville, Florida, 32610, USA
| | - Yi-Hua Chiang
- Department of Pharmaceutics, University of Florida, Gainesville, Florida, 32610, USA
| | | | - Mei He
- Department of Pharmaceutics, University of Florida, Gainesville, Florida, 32610, USA
| |
Collapse
|
29
|
Goudar VS, Koduri MP, Ta YNN, Chen Y, Chu LA, Lu LS, Tseng FG. Impact of a Desmoplastic Tumor Microenvironment for Colon Cancer Drug Sensitivity: A Study with 3D Chimeric Tumor Spheroids. ACS APPLIED MATERIALS & INTERFACES 2021; 13:48478-48491. [PMID: 34633791 DOI: 10.1021/acsami.1c18249] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Three-dimensional (3D) spheroid culture provides opportunities to model tumor growth closer to its natural context. The collagen network in the extracellular matrix supports autonomic tumor cell proliferation, but its presence and role in tumor spheroids remain unclear. In this research, we developed an in vitro 3D co-culture model in a microwell 3D (μ-well 3D) cell-culture array platform to mimic the tumor microenvironment (TME). The modular setup is used to characterize the paracrine signaling molecules and the role of the intraspheroidal collagen network in cancer drug resistance. The μ-well 3D platform is made up of poly(dimethylsiloxane) that contains 630 round wells for individual spheroid growth. Inside each well, the growth surface measured 500 μm in diameter and was functionalized with the amphiphilic copolymer. HCT-8 colon cancer cells and/or NIH3T3 fibroblasts were seeded in each well and incubated for up to 9 days for TME studies. It was observed that NIH3T3 cells promoted the kinetics of tumor organoid formation. The two types of cells self-organized into core-shell chimeric tumor spheroids (CTSs) with fibroblasts confined to the shell and cancer cells localized to the core. Confocal microscopy analysis indicated that a type-I collagen network developed inside the CTS along with increased TGF-β1 and α-SMA proteins. The results were correlated with a significantly increased stiffness in 3D co-cultured CTS up to 52 kPa as compared to two-dimensional (2D) co-culture. CTS was more resistant to 5-FU (IC50 = 14.0 ± 3.9 μM) and Regorafenib (IC50 = 49.8 ± 9.9 μM) compared to cells grown under the 2D condition 5-FU (IC50 = 12.2 ± 3.7 μM) and Regorafenib (IC50 = 5.9 ± 1.9 μM). Targeted collagen homeostasis with Sclerotiorin led to damaged collagen structure and disrupted the type-I collagen network within CTS. Such a treatment significantly sensitized collagen-supported CTS to 5-FU (IC50 = 4.4 ± 1.3 μM) and to Regorafenib (IC50 = 0.5 ± 0.2 μM). In summary, the efficient formation of colon cancer CTSs in a μ-well 3D culture platform allows exploration of the desmoplastic TME. The novel role of intratumor collagen quality as a drug sensitization target warrants further investigation.
Collapse
Affiliation(s)
- Venkanagouda S Goudar
- Department of Engineering and System Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan, ROC
| | - Manohar Prasad Koduri
- International Intercollegiate Ph.D. Program, National Tsing Hua University, Hsinchu 30013, Taiwan, ROC
- Department of Mechanical, Materials, and Aerospace, School of Engineering, University of Liverpool, Harrison Hughes Building, Liverpool L693GH, U.K
| | - Yen-Nhi Ngoc Ta
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan, ROC
| | - Yunching Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan, ROC
| | - Li-An Chu
- Department of Biomedical and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan, ROC
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan, ROC
| | - Long-Sheng Lu
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan, ROC
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan, ROC
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan, ROC
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 11031, Taiwan, ROC
| | - Fan-Gang Tseng
- Department of Engineering and System Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan, ROC
- International Intercollegiate Ph.D. Program, National Tsing Hua University, Hsinchu 30013, Taiwan, ROC
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan, ROC
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan, ROC
| |
Collapse
|
30
|
Sen D, Voulgaropoulos A, Keung AJ. Effects of early geometric confinement on the transcriptomic profile of human cerebral organoids. BMC Biotechnol 2021; 21:59. [PMID: 34641840 PMCID: PMC8507123 DOI: 10.1186/s12896-021-00718-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 09/28/2021] [Indexed: 12/21/2022] Open
Abstract
Background Human cerebral organoids (hCO) are attractive systems due to their ability to model important brain regions and transcriptomics of early in vivo brain development. To date, they have been used to understand the effects of genetics and soluble factors on neurodevelopment. Interestingly, one of the main advantages of hCOs are that they provide three dimensionality that better mimics the in vivo environment; yet, despite this central feature it remains unclear how spatial and mechanical properties regulate hCO and neurodevelopment. While biophysical factors such as shape and mechanical forces are known to play crucial roles in stem cell differentiation, embryogenesis and neurodevelopment, much of this work investigated two dimensional systems or relied on correlative observations of native developing tissues in three dimensions. Using hCOs to establish links between spatial factors and neurodevelopment will require the use of new approaches and could reveal fundamental principles of brain organogenesis as well as improve hCOs as an experimental model. Results Here, we investigated the effects of early geometric confinements on transcriptomic changes during hCO differentiation. Using a custom and tunable agarose microwell platform we generated embryoid bodies (EB) of diverse shapes mimicking several structures from embryogenesis and neurodevelopment and then further differentiated those EBs to whole brain hCOs. Our results showed that the microwells did not have negative gross impacts on the ability of the hCOs to differentiate towards neural fates, and there were clear shape dependent effects on neural lineage specification. In particular we observed that non-spherical shapes showed signs of altered neurodevelopmental kinetics and favored the development of medial ganglionic eminence-associated brain regions and cell types over cortical regions. Transcriptomic analysis suggests these mechanotransducive effects may be mediated by integrin and Wnt signaling. Conclusions The findings presented here suggest a role for spatial factors in brain region specification during hCO development. Understanding these spatial patterning factors will not only improve understanding of in vivo development and differentiation, but also provide important handles with which to advance and improve control over human model systems for in vitro applications. Supplementary Information The online version contains supplementary material available at 10.1186/s12896-021-00718-2.
Collapse
Affiliation(s)
- Dilara Sen
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, Raleigh, NC, 27695-7905, USA
| | - Alexis Voulgaropoulos
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, Raleigh, NC, 27695-7905, USA
| | - Albert J Keung
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, Raleigh, NC, 27695-7905, USA.
| |
Collapse
|
31
|
Jia Y, Shen P, Yan T, Zhou W, Sun J, Han X. Microfluidic Tandem Mechanical Sorting System for Enhanced Cancer Stem Cell Isolation and Ingredient Screening. Adv Healthc Mater 2021; 10:e2100985. [PMID: 34486235 DOI: 10.1002/adhm.202100985] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/13/2021] [Indexed: 12/13/2022]
Abstract
Robust isolation of cancer stem cells (CSCs) in a high-throughput, label-free manner is critical for understanding tumor heterogeneity and developing therapeutic strategies targeting CSCs. Cell-mechanics-based microfluidic sorting systems provide efficient and specific platforms for investigation of stem cell-like characteristics on the basis of cell deformability and cell-substrate adhesion properties. In the present study, a microfluidic tandem mechanical sorting system is developed to enrich CSCs with high flexibility and low adhesive capacity. In the integrated microfluidic system, cancer cells are driven by hydrodynamic forces to flow continuously through two featured devices, which are functionalized with sequentially variable microbarriers and surface-coated fluid mixing microchannels, respectively. Collected deformable and low-adhesive cancer cells exhibit enhanced stem cell-like properties with higher stemness and metastasis capacity both in vitro and in vivo, compared with each single device separation. Using these devices, bioactive natural compound screening targeting CSCs is performed and a potent therapeutic compound isoliquiritigenin from licorice is identified to inhibit the lung cancer stem cell phenotype. Taken together, this microfluidic tandem mechanical sorting system can facilitate drug screening targeting CSCs and the analysis of signals regulating CSC function in drug resistance.
Collapse
Affiliation(s)
- Yuanyuan Jia
- Department of Biochemistry and Molecular Biology School of Medicine & Holistic Integrative Medicine Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Peiliang Shen
- Department of Biochemistry and Molecular Biology School of Medicine & Holistic Integrative Medicine Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Tao Yan
- Department of Biochemistry and Molecular Biology School of Medicine & Holistic Integrative Medicine Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Weijia Zhou
- Department of Biochemistry and Molecular Biology School of Medicine & Holistic Integrative Medicine Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Jia Sun
- Department of Biochemistry and Molecular Biology School of Medicine & Holistic Integrative Medicine Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Xin Han
- Department of Biochemistry and Molecular Biology School of Medicine & Holistic Integrative Medicine Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization Nanjing University of Chinese Medicine Nanjing 210023 China
| |
Collapse
|
32
|
Kim D, Lee SJ, Youn J, Hong H, Eom S, Kim DS. A deep and permeable nanofibrous oval-shaped microwell array for the stable formation of viable and functional spheroids. Biofabrication 2021; 13. [PMID: 34030141 DOI: 10.1088/1758-5090/ac044c] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/24/2021] [Indexed: 12/26/2022]
Abstract
Despite the potential of a nanofibrous (NF) microwell array as a permeable microwell array to improve the viability and functions of spheroids, thanks to the superior permeability to both gases and solutes, there have still been difficulties regarding the stable formation of spheroids in the NF microwell array due to the low aspect ratio (AR) and the large interspacing between microwells. This study proposes a nanofibrous oval-shaped microwell array, named the NOVA microwell array, with both a high AR and a high well density, enabling us to not only collect cells in the microwell with a high cell seeding efficiency, but also to generate multiple viable and functional spheroids in a uniform and stable manner. To realize a deep NOVA microwell array with a high aspect ratio (AR = 0.9) and a high well density (494 wells cm-2), we developed a matched-mold thermoforming process for the fabrication of both size- and AR-controllable NOVA microwell arrays with various interspacing between microwells while maintaining the porous nature of the NF membrane. The human hepatocellular carcinoma (HepG2) cell spheroids cultured on the deep NOVA microwell array not only had uniform size and shape, with a spheroid circularity of 0.80 ± 0.03 at a cell seeding efficiency of 94.29 ± 9.55%, but also exhibited enhanced viability with a small fraction of dead cells and promoted functionality with increased albumin secretion, compared with the conventional impermeable microwell array. The superior characteristics of the deep NOVA microwell array, i.e. a high AR, a high well density, and a high permeability, pave the way to the production of various viable and functional spheroids and even organoids in a scalable manner.
Collapse
Affiliation(s)
- Dohui Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Seong Jin Lee
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Jaeseung Youn
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Hyeonjun Hong
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Seongsu Eom
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Dong Sung Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea.,Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea.,Institute for Convergence Research and Education in Advanced Technology, Yonsei University, 50, Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|
33
|
Visualizing Extracellular Vesicles and Their Function in 3D Tumor Microenvironment Models. Int J Mol Sci 2021; 22:ijms22094784. [PMID: 33946403 PMCID: PMC8125158 DOI: 10.3390/ijms22094784] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are cell-derived nanostructures that mediate intercellular communication by delivering complex signals in normal tissues and cancer. The cellular coordination required for tumor development and maintenance is mediated, in part, through EV transport of molecular cargo to resident and distant cells. Most studies on EV-mediated signaling have been performed in two-dimensional (2D) monolayer cell cultures, largely because of their simplicity and high-throughput screening capacity. Three-dimensional (3D) cell cultures can be used to study cell-to-cell and cell-to-matrix interactions, enabling the study of EV-mediated cellular communication. 3D cultures may best model the role of EVs in formation of the tumor microenvironment (TME) and cancer cell-stromal interactions that sustain tumor growth. In this review, we discuss EV biology in 3D culture correlates of the TME. This includes EV communication between cell types of the TME, differences in EV biogenesis and signaling associated with differing scaffold choices and in scaffold-free 3D cultures and cultivation of the premetastatic niche. An understanding of EV biogenesis and signaling within a 3D TME will improve culture correlates of oncogenesis, enable molecular control of the TME and aid development of drug delivery tools based on EV-mediated signaling.
Collapse
|
34
|
Kreutzer FP, Meinecke A, Schmidt K, Fiedler J, Thum T. Alternative strategies in cardiac preclinical research and new clinical trial formats. Cardiovasc Res 2021; 118:746-762. [PMID: 33693475 PMCID: PMC7989574 DOI: 10.1093/cvr/cvab075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
An efficient and safe drug development process is crucial for the establishment of new drugs on the market aiming to increase quality of life and life-span of our patients. Despite technological advances in the past decade, successful launches of drug candidates per year remain low. We here give an overview about some of these advances and suggest improvements for implementation to boost preclinical and clinical drug development with a focus on the cardiovascular field. We highlight advantages and disadvantages of animal experimentation and thoroughly review alternatives in the field of three-dimensional cell culture as well as preclinical use of spheroids and organoids. Microfluidic devices and their potential as organ-on-a-chip systems, as well as the use of living animal and human cardiac tissues are additionally introduced. In the second part, we examine recent gold standard randomized clinical trials and present possible modifications to increase lead candidate throughput: adaptive designs, master protocols, and drug repurposing. In silico and N-of-1 trials have the potential to redefine clinical drug candidate evaluation. Finally, we briefly discuss clinical trial designs during pandemic times.
Collapse
Affiliation(s)
- Fabian Philipp Kreutzer
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Anna Meinecke
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Kevin Schmidt
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Jan Fiedler
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.,Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.,Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| |
Collapse
|
35
|
McKenna MK, Englisch A, Brenner B, Smith T, Hoyos V, Suzuki M, Brenner MK. Mesenchymal stromal cell delivery of oncolytic immunotherapy improves CAR-T cell antitumor activity. Mol Ther 2021; 29:1808-1820. [PMID: 33571680 DOI: 10.1016/j.ymthe.2021.02.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 01/05/2021] [Accepted: 02/01/2021] [Indexed: 12/20/2022] Open
Abstract
The immunosuppressive tumor microenvironment (TME) is a formidable barrier to the success of adoptive cell therapies for solid tumors. Oncolytic immunotherapy with engineered adenoviruses (OAd) may disrupt the TME by infecting tumor cells, as well as surrounding stroma, to improve the functionality of tumor-directed chimeric antigen receptor (CAR)-T cells, yet efficient delivery of OAds to solid tumors has been challenging. Here we describe how mesenchymal stromal cells (MSCs) can be used to systemically deliver a binary vector containing an OAd together with a helper-dependent Ad (HDAd; combinatorial Ad vector [CAd]) that expresses interleukin-12 (IL-12) and checkpoint PD-L1 (programmed death-ligand 1) blocker. CAd-infected MSCs deliver and produce functional virus to infect and lyse lung tumor cells while stimulating CAR-T cell anti-tumor activity by release of IL-12 and PD-L1 blocker. The combination of this approach with administration of HER.2-specific CAR-T cells eliminates 3D tumor spheroids in vitro and suppresses tumor growth in two orthotopic lung cancer models in vivo. Treatment with CAd MSCs increases the overall numbers of human T cells in vivo compared to CAR-T cell only treatment and enhances their polyfunctional cytokine secretion. These studies combine the predictable targeting of CAR-T cells with the advantages of cancer cell lysis and TME disruption by systemic MSC delivery of oncolytic virotherapy: incorporation of immunostimulation by cytokine and checkpoint inhibitor production through the HDAd further enhances anti-tumor activity.
Collapse
Affiliation(s)
- Mary K McKenna
- Baylor College of Medicine, Center for Cell Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Alexander Englisch
- Baylor College of Medicine, Center for Cell Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA; Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany
| | - Benjamin Brenner
- Baylor College of Medicine, Center for Cell Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA; Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Rd., Evanston, IL 60208, USA
| | - Tyler Smith
- Baylor College of Medicine, Center for Cell Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Valentina Hoyos
- Baylor College of Medicine, Center for Cell Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Masataka Suzuki
- Baylor College of Medicine, Center for Cell Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Malcolm K Brenner
- Baylor College of Medicine, Center for Cell Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA.
| |
Collapse
|
36
|
Wu K, Kuo C, Tu T. A Highly Reproducible Micro U-Well Array Plate Facilitating High-Throughput Tumor Spheroid Culture and Drug Assessment. GLOBAL CHALLENGES (HOBOKEN, NJ) 2021; 5:2000056. [PMID: 33552551 PMCID: PMC7857131 DOI: 10.1002/gch2.202000056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/30/2020] [Indexed: 05/05/2023]
Abstract
3D multicellular tumor spheroids (MCTSs) have recently emerged as a landmark for cancer research due to their inherent traits that are physiologically relevant to primary tumor microenvironments. A facile approach-laser-ablated micro U-wells-has been widely adopted in the past decade. However, the differentiation of microwell uniformities and the construction of arrays have all remained elusive. Herein, an improved laser-ablated microwell array technique is proposed that can not only achieve arrayed MCTSs with identical sizes but can also perform high-throughput drug assessments in situ. Three critical laser ablation parameters, including frequency, duty cycle, and pulse number, are investigated to generate microwells flexibly with a range from 170 to 400 μm. The choice of microwells is optimally arranged into an array via precise control of horizontal spacing (d x) and vertical spacing (d y) amenable of cell-loss-free culture during cell seeding. Harvested T24, A549 and Huh-7 MCTSs from the microwell array correspond to approximately 75 to 140 μm in diameter. Anticancer drug screening of cisplatin validated IC50 values in 2D and MCTS conditions are 3.5 versus 9.1 μM (T24), 11.8 versus 277.7 μM (A549) and 33.5 versus 52.8 μM (Huh-7), and the permeability is measured to range from 0.042 to 0.58 μm min-1.
Collapse
Affiliation(s)
- Kuang‐Wei Wu
- Department of Biomedical EngineeringNational Cheng Kung UniversityTainan70101Taiwan
| | - Ching‐Te Kuo
- Department of Mechanical and Electro‐Mechanical EngineeringNational Sun Yat‐sen UniversityKaohsiung80400Taiwan
| | - Ting‐Yuan Tu
- Department of Biomedical EngineeringNational Cheng Kung UniversityTainan70101Taiwan
- Medical Device Innovation CenterNational Cheng Kung UniversityTainan70101Taiwan
- International Center for Wound Repair and RegenerationNational Cheng Kung UniversityTainan70101Taiwan
| |
Collapse
|
37
|
Kang SM, Kim D, Lee JH, Takayama S, Park JY. Engineered Microsystems for Spheroid and Organoid Studies. Adv Healthc Mater 2021; 10:e2001284. [PMID: 33185040 PMCID: PMC7855453 DOI: 10.1002/adhm.202001284] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/01/2020] [Indexed: 01/09/2023]
Abstract
3D in vitro model systems such as spheroids and organoids provide an opportunity to extend the physiological understanding using recapitulated tissues that mimic physiological characteristics of in vivo microenvironments. Unlike 2D systems, 3D in vitro systems can bridge the gap between inadequate 2D cultures and the in vivo environments, providing novel insights on complex physiological mechanisms at various scales of organization, ranging from the cellular, tissue-, to organ-levels. To satisfy the ever-increasing need for highly complex and sophisticated systems, many 3D in vitro models with advanced microengineering techniques have been developed to answer diverse physiological questions. This review summarizes recent advances in engineered microsystems for the development of 3D in vitro model systems. The relationship between the underlying physics behind the microengineering techniques, and their ability to recapitulate distinct 3D cellular structures and functions of diverse types of tissues and organs are highlighted and discussed in detail. A number of 3D in vitro models and their engineering principles are also introduced. Finally, current limitations are summarized, and perspectives for future directions in guiding the development of 3D in vitro model systems using microengineering techniques are provided.
Collapse
Affiliation(s)
- Sung-Min Kang
- Department of Green Chemical Engineering, Sangmyung University, Cheonan, Chungnam, 31066, Republic of Korea
| | - Daehan Kim
- Department of Mechanical Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Ji-Hoon Lee
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Shuichi Takayama
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Joong Yull Park
- Department of Mechanical Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| |
Collapse
|
38
|
Behroodi E, Latifi H, Bagheri Z, Ermis E, Roshani S, Salehi Moghaddam M. A combined 3D printing/CNC micro-milling method to fabricate a large-scale microfluidic device with the small size 3D architectures: an application for tumor spheroid production. Sci Rep 2020; 10:22171. [PMID: 33335148 PMCID: PMC7747638 DOI: 10.1038/s41598-020-79015-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 12/02/2020] [Indexed: 02/06/2023] Open
Abstract
The fabrication of a large-scale microfluidic mold with 3D microstructures for manufacturing of the conical microwell chip using a combined projection micro-stereolithography (PµSL) 3D printing/CNC micro-milling method for tumor spheroid formation is presented. The PµSL technique is known as the most promising method of manufacturing microfluidic chips due to the possibility of creating complex three-dimensional microstructures with high resolution in the range of several micrometers. The purpose of applying the proposed method is to investigate the influence of microwell depths on the formation of tumor spheroids. In the conventional methods, the construction of three-dimensional microstructures and multi-height chips is difficult, time-consuming, and is performed using a multi-step lithography process. Microwell depth is an essential parameter for microwell design since it directly affects the shear stress of the fluid flow and the diffusion of nutrients, respiratory gases, and growth factors. In this study, a chip was made with microwells of different depth varying from 100 to 500 µm. The mold of the microwell section is printed by the lab-made PµSL printer with 6 and 1 µm lateral and vertical resolutions. Other parts of the mold, such as the main chamber and micro-channels, were manufactured using the CNC micro-milling method. Finally, different parts of the master mold were assembled and used for PDMS casting. The proposed technique drastically simplifies the fabrication and rapid prototyping of large-scale microfluidic devices with high-resolution microstructures by combining 3D printing with the CNC micro-milling method.
Collapse
Affiliation(s)
- Ebrahim Behroodi
- Laser and Plasma Research Institute, Shahid Beheshti University, 1983963113, Tehran, Iran
| | - Hamid Latifi
- Laser and Plasma Research Institute, Shahid Beheshti University, 1983963113, Tehran, Iran. .,Department of Physics, Shahid Beheshti University, 1983963113, Tehran, Iran.
| | - Zeinab Bagheri
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, 1983963113, Tehran, Iran
| | - Esra Ermis
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, 1983963113, Tehran, Iran
| | - Shabnam Roshani
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, 1983963113, Tehran, Iran
| | | |
Collapse
|
39
|
Li Y, Wang Y, Shen C, Meng Q. Non-swellable F127-DA hydrogel with concave microwells for formation of uniform-sized vascular spheroids. RSC Adv 2020; 10:44494-44502. [PMID: 35517174 PMCID: PMC9058638 DOI: 10.1039/d0ra06188c] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/03/2020] [Indexed: 01/02/2023] Open
Abstract
Hydrogels with concave microwells are one of the simplest means to obtain uniform-sized cellular spheroids. However, the inherent swelling of hydrogels leads to reduced mechanical strength and thus deforms the structure of the microwells. In this study, we developed a hydrogel with microwells for formation of vascular spheroids via non-swellable di-acrylated Pluronic F127 (F127-DA), which showed higher mechanical strength than a conventional di-acrylated polyethylene glycol (PEG-DA) hydrogel. The uniform-sized vascular spheroids were spontaneously generated by human umbilical vein endothelial cells (HUVECs) and fibroblasts in the microwells. The endothelial functions of vascular spheroids were about 1-fold higher than those in two-dimensional (2D) culture, as indicated by secretion of nitric oxide (NO), prostacyclin (PGI2) and tissue factor pathway inhibitor (TFPI). Interestingly, the vascular spheroids with large diameter showed higher sensitivity to ethanol toxicity than those with small diameter, possibly due to the higher endothelial functions of large spheroids. Hence, F127-DA hydrogel with concave microwells provides a convenient way of forming uniform-sized spheroids that are useful for high throughput screening of drug/food toxicity. Hydrogels with concave microwells are one of the simplest means to obtain uniform-sized cellular spheroids.![]()
Collapse
Affiliation(s)
- Yingjun Li
- College of Chemical and Biological Engineering, Zhejiang University Hangzhou 310027 China
| | - Ying Wang
- College of Chemical and Biological Engineering, Zhejiang University Hangzhou 310027 China
| | - Chong Shen
- College of Chemical and Biological Engineering, Zhejiang University Hangzhou 310027 China
| | - Qin Meng
- College of Chemical and Biological Engineering, Zhejiang University Hangzhou 310027 China
| |
Collapse
|
40
|
Manzoor AA, Romita L, Hwang DK. A review on microwell and microfluidic geometric array fabrication techniques and its potential applications in cellular studies. CAN J CHEM ENG 2020. [DOI: 10.1002/cjce.23875] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Ahmad Ali Manzoor
- Department of Chemical Engineering Ryerson University Toronto Ontario Canada
- Keenan Research Centre for Biomedical Science St. Michael's Hospital Toronto Ontario Canada
- Institute for Biomedical Engineering Science and Technology (iBEST) A partnership between Ryerson University and St. Michael's Hospital Toronto Ontario Canada
| | - Lauren Romita
- Department of Chemical Engineering Ryerson University Toronto Ontario Canada
- Keenan Research Centre for Biomedical Science St. Michael's Hospital Toronto Ontario Canada
- Institute for Biomedical Engineering Science and Technology (iBEST) A partnership between Ryerson University and St. Michael's Hospital Toronto Ontario Canada
| | - Dae Kun Hwang
- Department of Chemical Engineering Ryerson University Toronto Ontario Canada
- Keenan Research Centre for Biomedical Science St. Michael's Hospital Toronto Ontario Canada
- Institute for Biomedical Engineering Science and Technology (iBEST) A partnership between Ryerson University and St. Michael's Hospital Toronto Ontario Canada
| |
Collapse
|
41
|
Three-dimensional cell models for extracellular vesicles production, isolation, and characterization. Methods Enzymol 2020; 645:209-230. [PMID: 33565973 DOI: 10.1016/bs.mie.2020.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Extracellular vesicles (EVs) play a pivotal role in cancer progression. However, the majority of functional studies performed so far relies on data acquired in traditional 2D cultures. Because the spatial architecture of tissue is decisive for the cell fate, new cell models to study EV functions in the 3D environment must approximate in vitro models to the physiological conditions. Several models were developed during the last years, which may be suitable to serve as 3D models to study EVs; among them are hydrogels, solid scaffolds, bioreactors, and 3D CoSeedis™ inserts. We present in this chapter a protocol for a 3D cell model based on the 3D CoSeedis™ agarose inserts, allowing for a long-term culture of cells of different origins under serum-free conditions and easy EV recovery. Additionally, information on individual culture conditions in 3D CoSeedis™ for different cell lines, protocols for model evaluation, and quality controls are included. We hope that our suggestions and experience will be useful to carry out EV study under more physiological conditions and contribute to the EV research field's progress.
Collapse
|
42
|
FAK inhibition radiosensitizes pancreatic ductal adenocarcinoma cells in vitro. Strahlenther Onkol 2020; 197:27-38. [PMID: 32705304 PMCID: PMC7801360 DOI: 10.1007/s00066-020-01666-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 06/29/2020] [Indexed: 12/16/2022]
Abstract
Introduction Focal adhesion kinase (FAK) is a nonreceptor tyrosine kinase protein frequently overexpressed in cancer and has been linked to an increase in the stem cell population of tumors, resistance to therapy, and metastatic spread. Pharmacological FAK inhibition in pancreatic cancer has received increased attention over the last few years, either alone or in combination with other therapeutics including chemotherapy and immunotherapy. However, its prognostic value and its role in radioresistance of pancreatic ducal adenocarcinoma (PDAC) is unknown. Methods and materials Using the TCGA and GTEx databases, we investigated the genetic alterations and mRNA expression levels of PTK2 (the encoding-gene for FAK) in normal pancreatic tissue and pancreatic cancer and its impact on patient survival. Furthermore, we evaluated the expression of FAK and its tyrosine domain Ty-397 in three pancreatic cancer cell lines. We went further and evaluated the role of a commercial FAK tyrosine kinase inhibitor VS-4718 on the viability and radiosensitization of the pancreatic cell lines as well as its effect on the extracellular matrix (ECM) production from the pancreatic stellate cells. Furthermore, we tested the effect of combining radiation with VS-4718 in a three-dimensional (3D) multicellular pancreatic tumor spheroid model. Results A database analysis revealed a relevant increase in genetic alterations and mRNA expression of the PTK2 in PDAC, which were associated with lower progression-free survival. In vitro, there was only variation in the basal phosphorylation level of FAK in cell lines. VS-4718 radiosensitized pancreatic cell lines only in the presence of ECM-producing pancreatic stellate cells and markedly reduced the ECM production in the stromal cells. Finally, using a 3D multicellular tumor model, the combination of VS-4718 and radiotherapy significantly reduced the growth of tumor aggregates. Conclusion Pharmacological inhibition of FAK in pancreatic cancer could be a novel therapeutic strategy as our results show a radiosensitization effect of VS-4718 in vitro in a multicellular 2D- and in a 3D-model of pancreatic cancer. Electronic supplementary material The online version of this article (10.1007/s00066-020-01666-0) contains supplementary material, which is available to authorized users.
Collapse
|
43
|
Rahman SM, Campbell JM, Coates RN, Render KM, Byrne CE, Martin EC, Melvin AT. Evaluation of intercellular communication between breast cancer cells and adipose-derived stem cells via passive diffusion in a two-layer microfluidic device. LAB ON A CHIP 2020; 20:2009-2019. [PMID: 32379852 PMCID: PMC7331673 DOI: 10.1039/d0lc00142b] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Breast cancer tumorigenesis and response to therapy is regulated by cancer cell interactions with the tumor microenvironment (TME). Breast cancer signaling to the surrounding TME results in a heterogeneous and diverse tumor microenvironment, which includes the production of cancer-associated fibroblasts, macrophages, adipocytes, and stem cells. The secretory profile of these cancer-associated cell types results in elevated chemokines and growth factors that promote cell survival and proliferation within the tumor. Current co-culture approaches mostly rely on transwell chambers to study intercellular signaling between adipose-derived stem cells (ASCs) and cancer cells; however, these methods are limited to endpoint measurements and lack dynamic control. In this study, a 4-channel, "flow-free" microfluidic device was developed to co-culture triple-negative MDA-MB-231 breast cancer cells and ASCs to study intercellular communication between two distinct cell types found in the TME. The device consists of two layers: a top PDMS layer with four imprinted channels coupled with a bottom agarose slab enclosed in a Plexiglas chamber. For dynamic co-culture, the device geometry contained two centered, flow-free channels, which were supplied with media from two outer flow channels via orthogonal diffusion through the agarose. Continuous fresh media was provided to the cell culture channel via passive diffusion without creating any shearing effect on the cells. The device geometry also allowed for the passive diffusion of cytokines and growth factors between the two cell types cultured in parallel channels to initiate cell-to-cell crosstalk. The device was used to show that MDA-MB-231 cells co-cultured with ASCs exhibited enhanced growth, a more aggressive morphology, and polarization toward the ASCs. The MDA-MB-231 cells were found to exhibit a greater degree of resistance to the drug paclitaxel when co-cultured with ASCs when compared to single culture studies. This microfluidic device is an ideal platform to study intercellular communication for many types of cells during co-culture experiments and allows for new investigations into stromal cell-mediated drug resistance in the tumor microenvironment.
Collapse
Affiliation(s)
- Sharif M Rahman
- Cain Department of Chemical Engineering, Louisiana State University, Baton Rouge, LA, 70803 USA.
| | | | | | | | | | | | | |
Collapse
|
44
|
Chao C, Ngo Le P, Engelward BP. SpheroidChip: Patterned Agarose Microwell Compartments Harboring HepG2 Spheroids are Compatible with Genotoxicity Testing. ACS Biomater Sci Eng 2020; 6:2427-2439. [PMID: 33145399 DOI: 10.1021/acsbiomaterials.9b01951] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Three-dimensional tissue culture models are emerging as effective alternatives to animal testing. They are especially beneficial for liver toxicity studies, enabling hepatocytes to display improved levels of liver-specific functions. One common model is hepatocyte spheroids, which are spontaneously formed cell aggregates. Techniques for spheroid formation include the use of ultralow attachment plates and the hanging drop method, both of which are technically challenging and relatively low throughput. Here, we describe a simple-to-use platform that improves spheroid production and is compatible with genotoxicity testing by the comet assay. To achieve this, we created a chip containing a microwell array where dozens of spheroids are contained within a single well of a 96-well plate. The microwells are made from agarose, a nontoxic material suitable for cell growth and spheroid formation. HepG2 cells loaded into customizable microwells formed spheroids through agarose-assisted aggregation within one to two days. In addition, the agarose matrix allows the same platform to be used in DNA damage analysis. Specifically, the comet assay enables quantification of DNA breaks based on the increased migration of damaged DNA through agarose during electrophoresis. Here, we developed a modified comet assay and show that intact HepG2 spheroids cultured in microwells can be electrophoresed to reveal the extent of DNA damage following exposure to inflammatory chemicals (H2O2 and SIN-1). With this SpheroidChip analysis method, we detected a dose-dependent increase in DNA damage and observed rapid repair of H2O2-induced DNA damage. In summary, we utilized an agarose microarray to condense what had required an entire 96-well plate into a single well, enabling analysis techniques that were cumbersome or impossible under conditions of a single spheroid per well of a 96-well plate.
Collapse
Affiliation(s)
- Christy Chao
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - P Ngo Le
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Bevin P Engelward
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| |
Collapse
|
45
|
Brüningk SC, Rivens I, Box C, Oelfke U, Ter Haar G. 3D tumour spheroids for the prediction of the effects of radiation and hyperthermia treatments. Sci Rep 2020; 10:1653. [PMID: 32015396 PMCID: PMC6997397 DOI: 10.1038/s41598-020-58569-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 01/17/2020] [Indexed: 01/08/2023] Open
Abstract
For multimodality therapies such as the combination of hyperthermia and radiation, quantification of biological effects is key for dose prescription and response prediction. Tumour spheroids have a microenvironment that more closely resembles that of tumours in vivo and may thus be a superior in vitro cancer model than monolayer cultures. Here, the response of tumour spheroids formed from two established human cancer cell lines (HCT116 and CAL27) to single and combination treatments of radiation (0-20 Gy), and hyperthermia at 47 °C (0-780 CEM43) has been evaluated. Response was analysed in terms of spheroid growth, cell viability and the distribution of live/dead cells. Time-lapse imaging was used to evaluate mechanisms of cell death and cell detachment. It was found that sensitivity to heat in spheroids was significantly less than that seen in monolayer cultures. Spheroids showed different patterns of shrinkage and regrowth when exposed to heat or radiation: heated spheroids shed dead cells within four days of heating and displayed faster growth post-exposure than samples that received radiation or no treatment. Irradiated spheroids maintained a dense structure and exhibited a longer growth delay than spheroids receiving hyperthermia or combination treatment at (thermal) doses that yielded equivalent levels of clonogenic cell survival. We suggest that, unlike radiation, which kills dividing cells, hyperthermia-induced cell death affects cells independent of their proliferation status. This induces microenvironmental changes that promote spheroid growth. In conclusion, 3D tumour spheroid growth studies reveal differences in response to heat and/or radiation that were not apparent in 2D clonogenic assays but that may significantly influence treatment efficacy.
Collapse
Affiliation(s)
- Sarah C Brüningk
- Joint Department of Physics at The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, SM25NG, UK.
| | - Ian Rivens
- Joint Department of Physics at The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, SM25NG, UK
| | - Carol Box
- Joint Department of Physics at The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, SM25NG, UK
| | - Uwe Oelfke
- Joint Department of Physics at The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, SM25NG, UK
| | - Gail Ter Haar
- Joint Department of Physics at The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, SM25NG, UK
| |
Collapse
|
46
|
Nguyen TPT, Le NXT, Lee NY. Microfluidic Approach to Generate a Tadpole-Egg-Shaped Alginate Fiber and Its Application in Tissue Engineering. ACS Biomater Sci Eng 2019; 6:1663-1670. [PMID: 33455358 DOI: 10.1021/acsbiomaterials.9b01753] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Herein, we introduce a facile microfluidic technique to produce a hybrid alginate fiber with a tadpole-egg shape. A triple-flow polydimethylsiloxane microfluidic device was constructed to allow the formation of oil droplets inside the alginate stream and was instantaneously gelated with the coaxially adjacent CaCl2. The fiber entrapping the uniform oil droplets was dehydrated, leading to the formation of a distinct tadpole-egg-shaped structure. A series of diverse fiber architectures was fabricated in a controlled manner based on the flow rates of the relevant flows. The tadpole-egg-shaped alginate fibers were employed as building blocks to create a three-dimensional microwell template for cell cultures. First, the tadpole-egg-shaped alginate fibers containing the oil droplets were half-dipped into a melted agarose solution. After the solidification of the agarose gel, the alginate fibers were degraded by an ethylenediaminetetraacetic acid (EDTA) solution to generate the hemispherical microwells. Mesenchymal stem cells (MSCs) were cultured in the microwells to generate spheroids, which were induced into chondrocytes using transforming growth factor-β3. The formed MSC spheroids exhibited a relatively high ratio of cell viability with more than 95% live cells after 14 days of culture. The success of the chondrogenic differentiation was proven based on staining (Safranin O) and the glycosaminoglycan levels. The latter was significantly higher in spheroids that were induced to form chondrocytes compared to those that were not induced after 21 days of differentiation. Second, we investigated the potential of the tadpole-egg-shaped alginate fibers as microcarriers for applications in drug delivery and implantable technologies. It was revealed that the degradation of the Ca-alginate wall of the hybrid fibers to release the oil droplets required an EDTA solution with a concentration of 500 mM for a 15 min period. This result can be used to further develop the tadpole-egg-shaped alginate fibers as uniform microcarriers with multiple compartments.
Collapse
Affiliation(s)
- Thi Phuong Thuy Nguyen
- Department of BioNano Technology, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si, Gyeonggi-do 13120, Republic of Korea
| | - Nguyen Xuan Thanh Le
- Department of BioNano Technology, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si, Gyeonggi-do 13120, Republic of Korea
| | - Nae Yoon Lee
- Department of BioNano Technology, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si, Gyeonggi-do 13120, Republic of Korea
| |
Collapse
|
47
|
Ravi M, Sneka MK, Joshipura A. The culture conditions and outputs from breast cancer cell line in vitro experiments. Exp Cell Res 2019; 383:111548. [PMID: 31398351 DOI: 10.1016/j.yexcr.2019.111548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 08/02/2019] [Accepted: 08/03/2019] [Indexed: 10/26/2022]
Abstract
One of the major cancer types that have gained significant importance globally is the breast cancer due to its socio-economic impact. Breast cancer research is an area of considerable importance and several types of material are available for research applications. These include cancer cell lines which can be utilized in several ways. Cell lines are convenient to use and recently about 84 human breast cancer cell lines were classified by molecular sub-typing. These cells lines come under five major molecular subtypes namely the luminal A and B, HER-2+, triple- A and B subtypes. These cell lines have been well characterized and were utilized for understanding various aspects of breast cancers. Also, apart from providing an understanding of the molecular mechanisms associated with breast cancers, these cell lines have contributed significantly to areas such as drug testing. We present in this review the features of these cell lines, the studies conducted using them and the outcome of such studies. Also, the details about the culture conditions and study outcomes of the cell lines grown in 3-dimensional (3D) systems are presented.
Collapse
Affiliation(s)
- Maddaly Ravi
- Department of Human Genetics, Faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, 600116, India.
| | - M Kaviya Sneka
- Department of Human Genetics, Faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, 600116, India
| | - Aastha Joshipura
- Department of Human Genetics, Faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, 600116, India
| |
Collapse
|
48
|
Lee SW, Jeong SY, Shin TH, Min J, Lee D, Jeong GS. A cell-loss-free concave microwell array based size-controlled multi-cellular tumoroid generation for anti-cancer drug screening. PLoS One 2019; 14:e0219834. [PMID: 31344058 PMCID: PMC6658056 DOI: 10.1371/journal.pone.0219834] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 07/03/2019] [Indexed: 12/20/2022] Open
Abstract
The 3D multi-cellular tumoroid (MCT) model is an in vivo-like, avascular tumor model that has received much attention as a refined screening platform for drug therapies. Several types of research have been efforted to improve the physiological characteristics of the tumor microenvironment (TME) of the in vivo-like MCTs. Size-controlled MCTs have received much attention for obtaining highly reproducible results in drug screening assays and achieving a homogeneous and meaningful level of biological activities. Here, we describe an effective method for fabricating the size-controlled in vivo-like MCTs using a cell-loss-free (CLF) microwell arrays. The CLF microwell arrays was fabricated by using the simple operation of laser carving of a poly (methyl methacrylate) (PMMA) master mold. We also demonstrated the biophysicochemical effect of tumor microenvironment (TME) resident fibroblasts through the expression of TGFβ, αSMA, Type I-, IV collagen, angiogenesis related markers on tumorigenesis, and confirmed the drug response of MCTs with anti-cancer agents. This technology for the fabrication of CLF microwell arrays could be used as an effective method to produce an in vitro tumor model for cancer research and drug discovery.
Collapse
Affiliation(s)
- Sang Woo Lee
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Soo Yeon Jeong
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Tae Hoon Shin
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Junhong Min
- School of Integrative Engineering, Department of Biomedical Engineering, Chung-Ang University, Seoul, Korea
| | - Donghyun Lee
- School of Integrative Engineering, Department of Biomedical Engineering, Chung-Ang University, Seoul, Korea
| | - Gi Seok Jeong
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| |
Collapse
|
49
|
Voglstaetter M, Thomsen AR, Nouvel J, Koch A, Jank P, Navarro EG, Gainey-Schleicher T, Khanduri R, Groß A, Rossner F, Blaue C, Franz CM, Veil M, Puetz G, Hippe A, Dindorf J, Kashef J, Thiele W, Homey B, Greco C, Boucheix C, Baur A, Erbes T, Waller CF, Follo M, Hossein G, Sers C, Sleeman J, Nazarenko I. Tspan8 is expressed in breast cancer and regulates E-cadherin/catenin signalling and metastasis accompanied by increased circulating extracellular vesicles. J Pathol 2019; 248:421-437. [PMID: 30982971 PMCID: PMC6771825 DOI: 10.1002/path.5281] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 03/23/2019] [Accepted: 03/27/2019] [Indexed: 01/02/2023]
Abstract
Tspan8 exhibits a functional role in many cancer types including pancreatic, colorectal, oesophagus carcinoma, and melanoma. We present a first study on the expression and function of Tspan8 in breast cancer. Tspan8 protein was present in the majority of human primary breast cancer lesions and metastases in the brain, bone, lung, and liver. In a syngeneic rat breast cancer model, Tspan8+ tumours formed multiple liver and spleen metastases, while Tspan8− tumours exhibited a significantly diminished ability to metastasise, indicating a role of Tspan8 in metastases. Addressing the underlying molecular mechanisms, we discovered that Tspan8 can mediate up‐regulation of E‐cadherin and down‐regulation of Twist, p120‐catenin, and β‐catenin target genes accompanied by the change of cell phenotype, resembling the mesenchymal–epithelial transition. Furthermore, Tspan8+ cells exhibited enhanced cell–cell adhesion, diminished motility, and decreased sensitivity to irradiation. As a regulator of the content and function of extracellular vesicles (EVs), Tspan8 mediated a several‐fold increase in EV number in cell culture and the circulation of tumour‐bearing animals. We observed increased protein levels of E‐cadherin and p120‐catenin in these EVs; furthermore, Tspan8 and p120‐catenin were co‐immunoprecipitated, indicating that they may interact with each other. Altogether, our findings show the presence of Tspan8 in breast cancer primary lesion and metastases and indicate its role as a regulator of cell behaviour and EV release in breast cancer. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Maren Voglstaetter
- Institute for Infection Prevention and Hospital Epidemiology; Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andreas R Thomsen
- Department of Radiation Oncology, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jerome Nouvel
- Institute for Infection Prevention and Hospital Epidemiology; Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Arend Koch
- Institute of Neuropathology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Paul Jank
- Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Elena Grueso Navarro
- Institute for Infection Prevention and Hospital Epidemiology; Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tanja Gainey-Schleicher
- Institute for Infection Prevention and Hospital Epidemiology; Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Richa Khanduri
- Institute for Infection Prevention and Hospital Epidemiology; Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andrea Groß
- Institute for Infection Prevention and Hospital Epidemiology; Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Florian Rossner
- Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Carina Blaue
- DFG-Center for Functional Nanostructures, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Clemens M Franz
- DFG-Center for Functional Nanostructures, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Marina Veil
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Gerhard Puetz
- Institute of Clinical Chemistry and Laboratory Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Andreas Hippe
- Department of Dermatology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany
| | - Jochen Dindorf
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Department of Dermatology, University Hospital Erlangen, Erlangen, Germany.,Translational Research Center, Friedrich-Alexander-University of Erlangen-Nuernberg, Erlangen, Germany
| | - Jubin Kashef
- Institute for Photon Science and Synchrotron Radiation, Karlsruhe Institute of Technology (KIT), Eggenstein-Leopoldshafen, Germany
| | - Wilko Thiele
- Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Bernhard Homey
- Department of Dermatology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany
| | - Celine Greco
- UMR-S935, Inserm, Université Paris Sud, Université Paris Saclay, Villejuif, France.,Department of Pain Management and Palliative Care, Necker Hospital, Paris, France
| | - Claude Boucheix
- UMR-S935, Inserm, Université Paris Sud, Université Paris Saclay, Villejuif, France.,Department of Pain Management and Palliative Care, Necker Hospital, Paris, France
| | - Andreas Baur
- Department of Dermatology, University Hospital Erlangen, Erlangen, Germany.,Translational Research Center, Friedrich-Alexander-University of Erlangen-Nuernberg, Erlangen, Germany
| | - Thalia Erbes
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Cornelius F Waller
- Institute for Infection Prevention and Hospital Epidemiology; Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marie Follo
- Department of Medicine I, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ghamartaj Hossein
- Institute for Infection Prevention and Hospital Epidemiology; Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Animal Physiology, Laboratory of Developmental Biology, University of Tehran, Tehran, Iran
| | - Christine Sers
- Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Jonathan Sleeman
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Irina Nazarenko
- Institute for Infection Prevention and Hospital Epidemiology; Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
50
|
Rocha S, Carvalho J, Oliveira P, Voglstaetter M, Schvartz D, Thomsen AR, Walter N, Khanduri R, Sanchez J, Keller A, Oliveira C, Nazarenko I. 3D Cellular Architecture Affects MicroRNA and Protein Cargo of Extracellular Vesicles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1800948. [PMID: 30828519 PMCID: PMC6382357 DOI: 10.1002/advs.201800948] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 11/30/2018] [Indexed: 05/05/2023]
Abstract
The success of malignant tumors is conditioned by the intercellular communication between tumor cells and their microenvironment, with extracellular vesicles (EVs) acting as main mediators. While the value of 3D conditions to study tumor cells is well established, the impact of cellular architecture on EV content and function is not investigated yet. Here, a recently developed 3D cell culture microwell array is adapted for EV production and a comprehensive comparative analysis of biochemical features, RNA and proteomic profiles of EVs secreted by 2D vs 3D cultures of gastric cancer cells, is performed. 3D cultures are significantly more efficient in producing EVs than 2D cultures. Global upregulation of microRNAs and downregulation of proteins in 3D are observed, indicating their dynamic coregulation in response to cellular architecture, with the ADP-ribosylation factor 6 signaling pathway significantly downregulated in 3D EVs. The data strengthen the biological relevance of cellular architecture for production and cargo of EVs.
Collapse
Affiliation(s)
- Sara Rocha
- i3S—Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen 2084200‐135PortoPortugal
- Ipatimup—Institute of Molecular Pathology and ImmunologyUniversity of PortoRua Júlio Amaral de Carvalho 454200‐135PortoPortugal
- ICBAS—Instituto de Ciências Biomédicas Abel SalazarUniversidade do PortoR. Jorge de Viterbo Ferreira 2284050‐313PortoPortugal
| | - Joana Carvalho
- i3S—Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen 2084200‐135PortoPortugal
- Ipatimup—Institute of Molecular Pathology and ImmunologyUniversity of PortoRua Júlio Amaral de Carvalho 454200‐135PortoPortugal
| | - Patrícia Oliveira
- i3S—Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen 2084200‐135PortoPortugal
- Ipatimup—Institute of Molecular Pathology and ImmunologyUniversity of PortoRua Júlio Amaral de Carvalho 454200‐135PortoPortugal
| | - Maren Voglstaetter
- Institute for Infection Prevention and Hospital EpidemiologyMedical Center—University of FreiburgFaculty of MedicineUniversity of FreiburgBreisacherstr. 115b79106FreiburgGermany
| | - Domitille Schvartz
- Department of Human Protein SciencesCentre Médical UniversitaireRue Michel‐Servet 1CH1211GenevaSwitzerland
| | - Andreas R. Thomsen
- Department of Radiation OncologyMedical Center—University of FreiburgHugstaetterstr 55Freiburg79106Germany
- German Cancer Consortium (DKTK)Partner Site Freiburg and German Cancer Research Center (DKFZ)Im Neuenheimer Feld 28069120HeidelbergGermany
| | - Nadia Walter
- Department of Human Protein SciencesCentre Médical UniversitaireRue Michel‐Servet 1CH1211GenevaSwitzerland
| | - Richa Khanduri
- Institute for Infection Prevention and Hospital EpidemiologyMedical Center—University of FreiburgFaculty of MedicineUniversity of FreiburgBreisacherstr. 115b79106FreiburgGermany
| | - Jean‐Charles Sanchez
- Department of Human Protein SciencesCentre Médical UniversitaireRue Michel‐Servet 1CH1211GenevaSwitzerland
| | - Andreas Keller
- Clinical BioinformaticsUniversity HospitalSaarland UniversityKirrberger Straße, Building E2.166123SaarbrückenGermany
| | - Carla Oliveira
- i3S—Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen 2084200‐135PortoPortugal
- Ipatimup—Institute of Molecular Pathology and ImmunologyUniversity of PortoRua Júlio Amaral de Carvalho 454200‐135PortoPortugal
- Department Pathology and OncologyFaculty of MedicineUniversity of PortoAlameda Prof. Hernâni Monteiro4200‐319PortoPortugal
| | - Irina Nazarenko
- Institute for Infection Prevention and Hospital EpidemiologyMedical Center—University of FreiburgFaculty of MedicineUniversity of FreiburgBreisacherstr. 115b79106FreiburgGermany
- German Cancer Consortium (DKTK)Partner Site Freiburg and German Cancer Research Center (DKFZ)Im Neuenheimer Feld 28069120HeidelbergGermany
| |
Collapse
|