201
|
Ishihara K, Narita Y, Teramura Y, Fukazawa K. Preparation of Magnetic Hydrogel Microparticles with Cationic Surfaces and Their Cell-Assembling Performance. ACS Biomater Sci Eng 2021; 7:5107-5117. [PMID: 34677934 DOI: 10.1021/acsbiomaterials.1c01150] [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: 11/29/2022]
Abstract
Cationic magnetic hydrogel microparticles with high retention on cell surfaces were prepared using a two-step procedure. Using these magnetic hydrogel microparticles, cells were clustered with each other, and cell aggregates were prepared effectively. Cross-linked poly(vinyl alcohol) (PVA) hydrogel microparticles containing iron oxide nanoparticles were prepared. The diameter of the microparticles was in the range of 200-500 nm. Water-soluble cationic polymers containing both trimethyl ammonium (TMA) groups and phenylboronic acid (PBA) groups were synthesized for the surface modification of the microparticles. To regulate the composition, electrically neutral phosphorylcholine groups were introduced into the polymer. Covalent bonds were formed between the hydroxy groups of PVA microparticles and PBA groups in the polymer. The surface zeta potential of the microparticles reflected the composition of the TMA groups. The particles responded to an external magnetic field and clustered rapidly. Microparticles were adsorbed on the floating cell surface and induced cell aggregation quickly when a magnetic field was applied. Under the most effective conditions, the diameter of the cell aggregates increased to approximately 1 mm after 30 min. Denser cell aggregates were formed by the synergistic effects of the magnetic field and the properties of the microparticles. The formed cell aggregates continued to grow for more than 4 days under an applied magnetic field, indicating that the ability of the cells in the aggregate to proliferate was well maintained.
Collapse
Affiliation(s)
- Kazuhiko Ishihara
- Department of Materials Engineering, School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.,Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Yusuke Narita
- Department of Materials Engineering, School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Yuji Teramura
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Kyoko Fukazawa
- Department of Materials Engineering, School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
202
|
Sun B, Zhao Y, Wu W, Zhao Q, Li G. A superhydrophobic chip integrated with an array of medium reservoirs for long-term hanging drop spheroid culture. Acta Biomater 2021; 135:234-242. [PMID: 34389482 DOI: 10.1016/j.actbio.2021.08.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 01/24/2023]
Abstract
Hanging drop (HD) is one of the most popular methods used for forming three-dimensional (3D) cell spheroids. However, conventional hanging drop systems are only applicable for short-term spheroid culture due to their inconvenience in exchanging cell culture media. Here we present a medium-reservoir-integrated superhydrophobic (MRI-SH) chip for long-term HD spheroid cultures. The device consists of two main components: i) a patterned superhydrophobic (SH) surface containing an array of wettable spots which anchor arrays of droplets of cell suspension, and ii) an array of chambers that serve as medium reservoirs, both interconnected via an array of thru-holes. This configuration provides two distinct advantages over conventional HD configurations: i) the high wettability contrast of the SH pattern on the chip leads to the formation and adhesion of nearly spherical hanging droplets on its surface, which minimizes interactions between the liquid and the substrate; ii) the integrated chambers provide large volumes of medium to maintain longer culture durations. Using this device, spheroids of MHCC97H cells were successfully formed, and the cultured spheroids could maintain high viability for up to 30 days and exhibited enhanced spheroid morphology compared to those cultured in the conventional HD systems. STATEMENT OF SIGNIFICANCE: This paper presents a medium-reservoir-integrated superhydrophobic hanging drop (HD) platform for the long-term culture of spheroids with enhanced morphology. By monolithically integrating medium reservoirs and a patterned SH surface into a single device, this HD platform can not only produce high-quality spheroids, but also permit them to sustain high viability for up to 30 days without the need for tedious medium replenishment. We believe that such a platform will be valuable in a wide range of biological or biomedical applications, including tissue engineering, regenerative medicine, and drug discovery.
Collapse
Affiliation(s)
- Bangyong Sun
- Key Laboratory of Optoelectronic Technology and Systems, Ministry of Education, Defence Key Disciplines Lab of Novel Micro-Nano Devices and System Technology, Chongqing University, Chongqing 400044, China
| | - Yi Zhao
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Weimin Wu
- School of Mechanical and Power Engineering, Chongqing University of Science and Technology, Chongqing 401331, China
| | - Qiang Zhao
- Key Laboratory of Optoelectronic Technology and Systems, Ministry of Education, Defence 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, Defence Key Disciplines Lab of Novel Micro-Nano Devices and System Technology, Chongqing University, Chongqing 400044, China.
| |
Collapse
|
203
|
3D Cell Culture Technology – A New Insight Into in Vitro Research – A Review. ANNALS OF ANIMAL SCIENCE 2021. [DOI: 10.2478/aoas-2021-0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Abstract
Most in vitro cell-based research is based on two-dimensional (2D) systems where growth and development take place on a flat surface, which does not reflect the natural environment of the cells. The imperfection and limitations of culture in 2D systems eventually led to the creation of three-dimensional (3D) culture models that more closely reproduce the actual conditions of physiological cell growth. Since the inception of 3D culture technology, many culture models have been developed, such as technologies of multicellular spheroids, organoids, and organs on chips in the technology of scaffolding, hydrogels, bio-printing and liquid media. In this review we will focus on the advantages and disadvantages of the 2D vs. 3D cell cultures technologies. We will also try to sum up available 3D culture systems and materials for building 3D scaffolds.
Collapse
|
204
|
Recent Advances in Three-Dimensional Stem Cell Culture Systems and Applications. Stem Cells Int 2021; 2021:9477332. [PMID: 34671401 PMCID: PMC8523294 DOI: 10.1155/2021/9477332] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/26/2021] [Accepted: 09/20/2021] [Indexed: 12/17/2022] Open
Abstract
Cell culture is one of the most core and fundamental techniques employed in the fields of biology and medicine. At present, although the two-dimensional cell culture method is commonly used in vitro, it is quite different from the cell growth microenvironment in vivo. In recent years, the limitations of two-dimensional culture and the advantages of three-dimensional culture have increasingly attracted more and more attentions. Compared to two-dimensional culture, three-dimensional culture system is better to realistically simulate the local microenvironment of cells, promote the exchange of information among cells and the extracellular matrix (ECM), and retain the original biological characteristics of stem cells. In this review, we first present three-dimensional cell culture methods from two aspects: a scaffold-free culture system and a scaffold-based culture system. The culture method and cell characterizations will be summarized. Then the application of three-dimensional cell culture system is further explored, such as in the fields of drug screening, organoids and assembloids. Finally, the directions for future research of three-dimensional cell culture are stated briefly.
Collapse
|
205
|
Modulation of Inherent Niches in 3D Multicellular MSC Spheroids Reconfigures Metabolism and Enhances Therapeutic Potential. Cells 2021; 10:cells10102747. [PMID: 34685727 PMCID: PMC8534378 DOI: 10.3390/cells10102747] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/05/2021] [Accepted: 10/12/2021] [Indexed: 01/02/2023] Open
Abstract
Multicellular spheroids show three-dimensional (3D) organization with extensive cell–cell and cell–extracellular matrix interactions. Owing to their native tissue-mimicking characteristics, mesenchymal stem cell (MSC) spheroids are considered promising as implantable therapeutics for stem cell therapy. Herein, we aim to further enhance their therapeutic potential by tuning the cultivation parameters and thus the inherent niche of 3D MSC spheroids. Significantly increased expression of multiple pro-regenerative paracrine signaling molecules and immunomodulatory factors by MSCs was observed after optimizing the conditions for spheroid culture. Moreover, these alterations in cellular behaviors may be associated with not only the hypoxic niche developed in the spheroid core but also with the metabolic reconfiguration of MSCs. The present study provides efficient methods for manipulating the therapeutic capacity of 3D MSC spheroids, thus laying solid foundations for future development and clinical application of spheroid-based MSC therapy for regenerative medicine.
Collapse
|
206
|
Guenther R, Dreschers S, Maassen J, Reibert D, Skazik-Voogt C, Gutermuth A. The Treasury of Wharton's Jelly. Stem Cell Rev Rep 2021; 18:1627-1638. [PMID: 34647276 PMCID: PMC9209346 DOI: 10.1007/s12015-021-10217-8] [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] [Accepted: 06/25/2021] [Indexed: 11/11/2022]
Abstract
Background Postnatal umbilical cord tissue contains valuable mesenchymal progenitor cells of various differentiation stages. While mesenchymal stem cells are plastic-adherent and tend to differentiate into myofibroblastic phenotypes, some round cells detach, float above the adherent cells, and build up cell aggregates, or form spheroids spontaneously. Very small luminescent cells are always involved as single cells or within collective forms and resemble the common well-known very small embryonic-like cells (VSELs). In this study, we investigated these VSELs-like cells in terms of their pluripotency phenotype and tri-lineage differentiation potential. Methods VSELs-like cells were isolated from cell-culture supernatants by a process that combines filtering, up concentration, and centrifugation. To determine their pluripotency character, we measured the expression of Nanog, Sox-2, Oct-4, SSEA-1, CXCR4, SSEA-4 on gene and protein level. In addition, the cultured cells derived from UC tissue were examined regarding their potential to differentiate into three germ layers. Result The VSELs-like cells express all of the pluripotency-associated markers we investigated and are able to differentiate into meso- endo- and ectodermal precursor cells. Conclusions Umbilical cord tissue hosts highly potent VSELs-like stem cells. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1007/s12015-021-10217-8.
Collapse
Affiliation(s)
- Rebecca Guenther
- Department for Applied Cell Biology, Fraunhofer Institute for Production Technology, Steinbachstr. 17, 52074, Aachen, Germany
| | - Stephan Dreschers
- Clinic for Gynaecology, University Hospital Aachen, Pauwelsstr 30, 52074, Aachen, Germany
| | - Jessika Maassen
- Department for Applied Cell Biology, Fraunhofer Institute for Production Technology, Steinbachstr. 17, 52074, Aachen, Germany
| | - Daniel Reibert
- Department for Applied Cell Biology, Fraunhofer Institute for Production Technology, Steinbachstr. 17, 52074, Aachen, Germany
| | - Claudia Skazik-Voogt
- Department for Applied Cell Biology, Fraunhofer Institute for Production Technology, Steinbachstr. 17, 52074, Aachen, Germany
| | - Angela Gutermuth
- Department for Applied Cell Biology, Fraunhofer Institute for Production Technology, Steinbachstr. 17, 52074, Aachen, Germany.
| |
Collapse
|
207
|
Zahn I, Braun T, Gögele C, Schulze-Tanzil G. Minispheroids as a Tool for Ligament Tissue Engineering: Do the Self-Assembly Techniques and Spheroid Dimensions Influence the Cruciate Ligamentocyte Phenotype? Int J Mol Sci 2021; 22:11011. [PMID: 34681672 PMCID: PMC8537246 DOI: 10.3390/ijms222011011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 12/14/2022] Open
Abstract
Spheroid culture might stabilize the ligamentocyte phenotype. Therefore, the phenotype of lapine cruciate ligamentocyte (L-CLs) minispheroids prepared either by hanging drop (HD) method or by using a novel spheroid plate (SP) and the option of methyl cellulose (MC) for tuning spheroid formation was tested. A total of 250 and 1000 L-CLs per spheroid were seeded as HDs or on an SP before performing cell viability assay, morphometry, gene expression (qRT-PCR) and protein immunolocalization after 7 (HD/SP) and 14 (SP) days. Stable and viable spheroids of both sizes could be produced with both methods, but more rapidly with SP. MC accelerated the formation of round spheroids (HD). Their circular areas decreased significantly during culturing. After 7 days, the diameters of HD-derived spheroids were significantly larger compared to those harvested from the SP, with a tendency of lower circularity suggesting an ellipsoid shape. Gene expression of decorin increased significantly after 7 days (HD, similar trend in SP), tenascin C tended to increase after 7 (HD/SP) and 14 days (SP), whereas collagen type 1 decreased (HD/SP) compared to the monolayer control. The cruciate ligament extracellular matrix components could be localized in all mini-spheroids, confirming their conserved expression profile and their suitability for ligament tissue engineering.
Collapse
Affiliation(s)
- Ingrid Zahn
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, Prof. Ernst Nathan Str.1, 90419 Nuremberg, Germany; (I.Z.); (T.B.); (C.G.)
- Institute of Functional and Clinical Anatomy, Friedrich Alexander University, Erlangen-Nuremberg, Universitätsstraße 19, 91054 Erlangen, Germany
- Department of Applied Chemistry, Nuremberg Institute of Technology Georg Simon Ohm, Keßlerplatz 12, 90489 Nuremberg, Germany
| | - Tobias Braun
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, Prof. Ernst Nathan Str.1, 90419 Nuremberg, Germany; (I.Z.); (T.B.); (C.G.)
- Department of Applied Chemistry, Nuremberg Institute of Technology Georg Simon Ohm, Keßlerplatz 12, 90489 Nuremberg, Germany
- Department of Cardiac Surgery (Cardiovascular Center), Klinikum Nürnberg Süd, Breslauer Str. 201, 90471 Nuremberg, Germany
| | - Clemens Gögele
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, Prof. Ernst Nathan Str.1, 90419 Nuremberg, Germany; (I.Z.); (T.B.); (C.G.)
- Department of Biosciences, Paris Lodron University of Salzburg, Hellbrunnerstr 34, 5020 Salzburg, Austria
| | - Gundula Schulze-Tanzil
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, Prof. Ernst Nathan Str.1, 90419 Nuremberg, Germany; (I.Z.); (T.B.); (C.G.)
| |
Collapse
|
208
|
Manzan-Martins C, Paulesu L. Impact of bisphenol A (BPA) on cells and tissues at the human materno-fetal interface. Tissue Cell 2021; 73:101662. [PMID: 34628212 DOI: 10.1016/j.tice.2021.101662] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 02/07/2023]
Abstract
Bisphenol A (BPA) is an endocrine disruptor extensively used in the production of polycarbonate plastics and epoxy resins and a component of liquid and food containers. It is a hazard in the prenatal period because of its presence in the placenta, fetal membranes, amniotic fluid, maternal and fetal blood and its ability to cross the placenta and reach the fetus. Estimation of the risk of BPA exposure during in utero life is extremely important in order to prevent complications of pregnancy and fetal growth. This review describes in vitro models of the human materno-fetal interface. It also outlines the effects of BPA at doses indicated as "physiological", namely at the concentrations found in the general population, and at "supraphysiological" and "subphysiological" doses, i.e. above and below the physiological range. This work will help clarify the discrepancies observed in studies on the effects of BPA on human reproduction and pregnancy, and it will be useful for the choice of appropriate in vitro models for future studies aimed at identifying the potential impact of BPA on specific functional processes.
Collapse
Affiliation(s)
| | - L Paulesu
- Department of Life Sciences, University of Siena, Siena, Italy.
| |
Collapse
|
209
|
Zamorano M, Castillo RL, Beltran JF, Herrera L, Farias JA, Antileo C, Aguilar-Gallardo C, Pessoa A, Calle Y, Farias JG. Tackling Ischemic Reperfusion Injury With the Aid of Stem Cells and Tissue Engineering. Front Physiol 2021; 12:705256. [PMID: 34603075 PMCID: PMC8484708 DOI: 10.3389/fphys.2021.705256] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/11/2021] [Indexed: 01/14/2023] Open
Abstract
Ischemia is a severe condition in which blood supply, including oxygen (O), to organs and tissues is interrupted and reduced. This is usually due to a clog or blockage in the arteries that feed the affected organ. Reinstatement of blood flow is essential to salvage ischemic tissues, restoring O, and nutrient supply. However, reperfusion itself may lead to major adverse consequences. Ischemia-reperfusion injury is often prompted by the local and systemic inflammatory reaction, as well as oxidative stress, and contributes to organ and tissue damage. In addition, the duration and consecutive ischemia-reperfusion cycles are related to the severity of the damage and could lead to chronic wounds. Clinical pathophysiological conditions associated with reperfusion events, including stroke, myocardial infarction, wounds, lung, renal, liver, and intestinal damage or failure, are concomitant in due process with a disability, morbidity, and mortality. Consequently, preventive or palliative therapies for this injury are in demand. Tissue engineering offers a promising toolset to tackle ischemia-reperfusion injuries. It devises tissue-mimetics by using the following: (1) the unique therapeutic features of stem cells, i.e., self-renewal, differentiability, anti-inflammatory, and immunosuppressants effects; (2) growth factors to drive cell growth, and development; (3) functional biomaterials, to provide defined microarchitecture for cell-cell interactions; (4) bioprocess design tools to emulate the macroscopic environment that interacts with tissues. This strategy allows the production of cell therapeutics capable of addressing ischemia-reperfusion injury (IRI). In addition, it allows the development of physiological-tissue-mimetics to study this condition or to assess the effect of drugs. Thus, it provides a sound platform for a better understanding of the reperfusion condition. This review article presents a synopsis and discusses tissue engineering applications available to treat various types of ischemia-reperfusions, ultimately aiming to highlight possible therapies and to bring closer the gap between preclinical and clinical settings.
Collapse
Affiliation(s)
- Mauricio Zamorano
- Department of Chemical Engineering, Universidad de La Frontera, Temuco, Chile
| | | | - Jorge F Beltran
- Department of Chemical Engineering, Universidad de La Frontera, Temuco, Chile
| | - Lisandra Herrera
- Department of Chemical Engineering, Universidad de La Frontera, Temuco, Chile
| | - Joaquín A Farias
- Facultad de Ingeniería y Ciencias, Universidad Adolfo Ibíñtez, Santiago, Chile
| | - Christian Antileo
- Department of Chemical Engineering, Universidad de La Frontera, Temuco, Chile
| | - Cristobal Aguilar-Gallardo
- Hematological Transplant and Cell Therapy Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Adalberto Pessoa
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Yolanda Calle
- Department of Life Sciences, Whitelands College, University of Roehampton, London, United Kingdom
| | - Jorge G Farias
- Department of Chemical Engineering, Universidad de La Frontera, Temuco, Chile
| |
Collapse
|
210
|
Gravity-Based Flow Efficient Perfusion Culture System for Spheroids Mimicking Liver Inflammation. Biomedicines 2021; 9:biomedicines9101369. [PMID: 34680487 PMCID: PMC8533112 DOI: 10.3390/biomedicines9101369] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 02/07/2023] Open
Abstract
The spheroid culture system provides an efficient method to emulate organ-specific pathophysiology, overcoming the traditional two-dimensional (2D) cell culture limitations. The intervention of microfluidics in the spheroid culture platform has the potential to enhance the capacity of in vitro microphysiological tissues for disease modeling. Conventionally, spheroid culture is carried out in static conditions, making the media nutrient-deficient around the spheroid periphery. The current approach tries to enhance the capacity of the spheroid culture platform by integrating the perfusion channel for dynamic culture conditions. A pro-inflammatory hepatic model was emulated using a coculture of HepG2 cell line, fibroblasts, and endothelial cells for validating the spheroid culture plate with a perfusable channel across the spheroid well. Enhanced proliferation and metabolic capacity of the microphysiological model were observed and further validated by metabolic assays. A comparative analysis of static and dynamic conditions validated the advantage of spheroid culture with dynamic media flow. Hepatic spheroids were found to have improved proliferation in dynamic flow conditions as compared to the static culture platform. The perfusable culture system for spheroids is more physiologically relevant as compared to the static spheroid culture system for disease and drug analysis.
Collapse
|
211
|
Rasouli R, Tabrizian M. Rapid Formation of Multicellular Spheroids in Boundary-Driven Acoustic Microstreams. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101931. [PMID: 34418307 DOI: 10.1002/smll.202101931] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/25/2021] [Indexed: 06/13/2023]
Abstract
3D cell spheroid culture has emerged as a more faithful recreation of cell growth environment compared to conventional 2D culture, as it can maintain tissue structures, physicochemical characteristics, and cell phenotypes. The majority of current spheroid formation methods are limited to a physical agglomeration of the desired cell type, and then relying on cell capacity to secrete extracellular matrix to form coherent spheroids. Hence, apart from being time-consuming, their success in leading to functional spheroid formation is also cell-type dependent. In this study, a boundary-driven acoustic microstreaming tool is presented that can simultaneously congregate cells and generate sturdy cell clusters through incorporating a bioadhesive such as collagen for rapid production of spheroids. The optimized mixture of type I collagen (0.42 mg mL-1 ) and methylcellulose (0.4% w/v ) accelerates the coagulation of cell-matrix as fast as 10 s while avoiding their adhesion to the device, and thereby offering easy spheroid retrieval. The versatility of the platform is shown for the production of MDA-MB-231 and MCF-7 spheroids, multicellular spheroids, and composite spheroids made of cells and microparticles. The ability to produce densely packed spheroids embedded within a biomimetic extracellular matrix component, along with rapid formation and easy collection of spheroids render the proposed device a step in technology development required to realize potentials of 3D constructs such as building blocks for the emerging field of bottom-up tissue engineering.
Collapse
Affiliation(s)
- Reza Rasouli
- Biomedical Engineering Department, Faculty of Medicine, McGill University, Montreal, Quebec, H3A 2B4, Canada
| | - Maryam Tabrizian
- Biomedical Engineering Department, Faculty of Medicine, McGill University, Montreal, Quebec, H3A 2B4, Canada
- Faculty of Dentistry, McGill University, Montreal, Quebec, H3A 1G1, Canada
| |
Collapse
|
212
|
Yadav S, Majumder A. Biomimicked hierarchical 2D and 3D structures from natural templates: applications in cell biology. Biomed Mater 2021; 16. [PMID: 34438385 DOI: 10.1088/1748-605x/ac21a7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 08/26/2021] [Indexed: 11/11/2022]
Abstract
Intricate structures of natural surfaces and materials have amazed people over the ages. The unique properties of various surfaces also created interest and curiosity in researchers. In the recent past, with the advent of superior microscopy techniques, we have started to understand how these complex structures provide superior properties. With that knowledge, scientists have developed various biomimicked and bio-inspired surfaces for different non-biological applications. In the last two decades, we have also started to learn how structures of the tissue microenvironment influence cell function and behaviour, both in physiological and pathological conditions. Hence, it became essential to decipher the role and importance of structural hierarchy in the cellular context. With advances in microfabricated techniques, such complex structures were made by superimposing features of different dimensions. However, the fabricated topographies are far from matching the complexities presentin vivo. Hence, the need of biomimicking the natural surfaces for cellular applications was felt. In this review, we discuss a few examples of hierarchical surfaces found in plants, insects, and vertebrates. Such structures have been widely biomimicked for various applications but rarely studied for cell-substrate interaction and cellular response. Here, we discuss the research work wherein 2D hierarchical substrates were prepared using biomimicking to understand cellular functions such as adhesion, orientation, differentiation, and formation of spheroids. Further, we also present the status of ongoing research in mimicking 3D tissue architecture using de-cellularized plant-based and tissue/organ-based scaffolds. We will also discuss 3D printing for fabricating 2D and 3D hierarchical structures. The review will end by highlighting the various advantages and research challenges in this approach. The biomimickedin-vivolike substrate can be used to better understand cellular physiology, and for tissue engineering.
Collapse
Affiliation(s)
- Shital Yadav
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Abhijit Majumder
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| |
Collapse
|
213
|
Bello AB, Kim Y, Park S, Muttigi MS, Kim J, Park H, Lee S. Matrilin3/TGFβ3 gelatin microparticles promote chondrogenesis, prevent hypertrophy, and induce paracrine release in MSC spheroid for disc regeneration. NPJ Regen Med 2021; 6:50. [PMID: 34480032 PMCID: PMC8417285 DOI: 10.1038/s41536-021-00160-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 08/04/2021] [Indexed: 12/21/2022] Open
Abstract
Degenerative disc disease (DDD) is the leading cause of excruciating lower back pain and disability in adults worldwide. Among the current treatments for DDD, cell-based therapies such as the injection of both disc- and non-disc-derived chondrocytes have shown significant improvements in the patients’ condition. However, further advancement of these therapies is required to not only ensure a supply of healthy chondrocytes but also to promote regeneration of the defective cells in the injury site. Here, we report that the incorporation of gelatin microparticles coloaded with transforming growth factor beta 3 and matrilin 3 promoted chondrogenic differentiation of adipose-derived mesenchymal stem cell spheroids while preventing hypertrophy and terminal differentiation of cells. Moreover, these composite spheroids induced the release of chondrogenic cytokines that, in turn, promoted regeneration of degenerative chondrocytes in vitro. Finally, injections of these composite spheroids in a rat model of intervertebral disc disease promoted restoration of the chondrogenic properties of the cells, thereby allowing regeneration of the chondrogenic tissue in vivo.
Collapse
Affiliation(s)
- Alvin Bacero Bello
- School of Integrative Engineering, Chung-Ang University, Seoul, 06911, Korea.,Department of Medical Biotechnology, Dongguk University, Seoul, 04620, Korea
| | - Yunkyung Kim
- School of Integrative Engineering, Chung-Ang University, Seoul, 06911, Korea
| | - Sunghyun Park
- Department of Life Science, CHA University, Seongnam, 13488, Korea
| | | | - Jiseong Kim
- Department of Medical Biotechnology, Dongguk University, Seoul, 04620, Korea
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul, 06911, Korea.
| | - Soohong Lee
- Department of Medical Biotechnology, Dongguk University, Seoul, 04620, Korea.
| |
Collapse
|
214
|
Nalbach L, Müller D, Wrublewsky S, Metzger W, Menger MD, Laschke MW, Ampofo E. Microvascular fragment spheroids: Three-dimensional vascularization units for tissue engineering and regeneration. J Tissue Eng 2021; 12:20417314211035593. [PMID: 34471514 PMCID: PMC8404660 DOI: 10.1177/20417314211035593] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 07/12/2021] [Indexed: 11/29/2022] Open
Abstract
Adipose tissue-derived microvascular fragments (MVF) serve as vascularization units in tissue engineering and regenerative medicine. Because a three-dimensional cellular arrangement has been shown to improve cell function, we herein generated for the first time MVF spheroids to investigate whether this further increases their vascularization potential. These spheroids exhibited a morphology, size, and viability comparable to that of previously introduced stromal vascular fraction (SVF) spheroids. However, MVF spheroids contained a significantly higher number of CD31-positive endothelial cells and α-smooth muscle actin (SMA)-positive perivascular cells, resulting in an enhanced angiogenic sprouting activity. Accordingly, they also exhibited an improved in vivo vascularization and engraftment after transplantation into mouse dorsal skinfold chambers. These findings indicate that MVF spheroids are superior to SVF spheroids and, thus, may be highly suitable to improve the vascularization of tissue defects and implanted tissue constructs.
Collapse
Affiliation(s)
- Lisa Nalbach
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| | - Danièle Müller
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| | - Selina Wrublewsky
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| | - Wolfgang Metzger
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| | - Emmanuel Ampofo
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| |
Collapse
|
215
|
Park Y, Ji ST, Yong U, Das S, Jang WB, Ahn G, Kwon SM, Jang J. 3D bioprinted tissue-specific spheroidal multicellular microarchitectures for advanced cell therapy. Biofabrication 2021; 13. [PMID: 34433153 DOI: 10.1088/1758-5090/ac212e] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/25/2021] [Indexed: 01/05/2023]
Abstract
Intercellular interaction is the most crucial factor in promoting cell viability and functionality in an engineered tissue system. Of the various shapes available for cell-laden constructs, spheroidal multicellular microarchitectures (SMMs) have been introduced as building blocks and injectable cell carriers with substantial cell-cell and cell-extracellular matrix (ECM) interactions. Here, we developed a precise and expeditious SMM printing method that can create a tissue-specific microenvironment and thus be potentially useful for cell therapy. This printing strategy is designed to manufacture SMMs fabricated with optimal bioink blended with decellularized ECM and alginate to enhance the functional performance of the encapsulated cells. Experimental results showed that the proposed method allowed for size controllability and mass production of SMMs with high cell viability. Moreover, SMMs co-cultured with endothelial cells promoted lineage-specific maturation and increased functionality compared to monocultured SMMs. Overall, it was concluded that SMMs have the potential for use in cell therapy due to their high cell retention and proliferation rate compared to single-cell injection, particularly for efficient tissue regeneration after myocardial infarction. This study suggests that utilizing microextrusion-based 3D bioprinting technology to encapsulate cells in cell-niche-standardized SMMs can expand the range of possible applications.
Collapse
Affiliation(s)
- Yejin Park
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk 37673, Republic of Korea
| | - Seung Taek Ji
- Stem Cell Research Center, Medical Research Institute, Pusan National University School of Medicine, Yangsan, Kyungnam 50612, Republic of Korea
| | - Uijung Yong
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk 37673, Republic of Korea
| | - Sanskrita Das
- Department of Biomedical Engineering, Emory University, Atlanta, GA 30322, United States of America
| | - Woong Bi Jang
- Stem Cell Research Center, Medical Research Institute, Pusan National University School of Medicine, Yangsan, Kyungnam 50612, Republic of Korea
| | - Geunseon Ahn
- Research Institute, Sphebio Co., Ltd, Pohang, Kyungbuk 37666, Republic of Korea
| | - Sang-Mo Kwon
- Stem Cell Research Center, Medical Research Institute, Pusan National University School of Medicine, Yangsan, Kyungnam 50612, Republic of Korea
| | - Jinah Jang
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk 37673, Republic of Korea.,School of Interdisciplinary Bioscience and Bioengineering, POSTECH, Pohang, Kyungbuk 37673, Republic of Korea.,Department of Mechanical Engineering, POSTECH, Pohang, Kyungbuk 37673, Republic of Korea
| |
Collapse
|
216
|
Scalzone A, Wang XN, Dalgarno K, Ferreira AM, Gentile P. A Chondrosphere-Based Scaffold Free Approach to Manufacture an In Vitro Articular Cartilage Model. Tissue Eng Part A 2021; 28:84-93. [PMID: 34114497 DOI: 10.1089/ten.tea.2021.0061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
In vitro engineering of human articular cartilage (AC) is a regenerative medicine challenge. The main objective of this study was the development of a repeatable scaffold-free in vitro model of chondrocyte spheroid-based treatments of cartilage defects, to allow for systematic study and further optimization of this type of treatment. Human articular chondrocytes (HC) and immortalized mesenchymal cells differentiated in chondrocytes (Y201-Cs) were cultured in round-bottom 96-well plates to produce multicellular spheroids and their growth kinetics, and viability was evaluated over 7 days of culture. Then, the spheroids were assembled and cultured for 21 days on a gelatin-coated poly(lactic-co-glycolic acid) electrospun membrane (10 spheroids/cm2), following a protocol in line with the clinically approved Chondrosphere® (CO.DON AG) technique. Both HC and Y201-C cells formed compact and viable spheroids after 7 days of culture with a reduction of diameter over the 7 days from 1300 ± 150 μm to 600 ± 90 μm and from 1250 ± 60 μm to 800 ± 20 μm for HC and Y201-C, respectively. When the spheroids were transferred onto the support membrane, these adhered on the membrane itself and fused themselves, producing collagen type II (COL2A1) and aggrecan (ACAN), according to gene expression and glycosaminoglycans quantification analyses. We detected higher expression of COL2A1 in HC cells, while the Y201-C constructs were characterized by an increased ACAN expression. The approach we presented allows a standardizable production of spheroids with predictable geometry and the creation of a reproducible scaffold-free in vitro AC-like construct showing high expression of chondrogenic markers, using both HC and Y201-C. In addition, the bankable Y201-C cells provide an effective base model for experimentation with the spheroid approach to further enhance the process. Impact statement This is first work on the development of a repeatable scaffold-free in vitro model based on an optimized protocol in line with a recent clinically approved Chondrosphere® (CO.DON AG) technique. In addition, we demonstrated that a bankable cell type (Y201-C) could produce an engineered cartilage-like construct, giving a repeatable model as a key tool for experimentation of therapeutic treatment ahead of studies with heterogeneous cell populations.
Collapse
Affiliation(s)
- Annachiara Scalzone
- School of Engineering and Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Xiao N Wang
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Kenny Dalgarno
- School of Engineering and Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Ana M Ferreira
- School of Engineering and Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Piergiorgio Gentile
- School of Engineering and Newcastle University, Newcastle Upon Tyne, United Kingdom
| |
Collapse
|
217
|
Di Mattia M, Mauro A, Citeroni MR, Dufrusine B, Peserico A, Russo V, Berardinelli P, Dainese E, Cimini A, Barboni B. Insight into Hypoxia Stemness Control. Cells 2021; 10:cells10082161. [PMID: 34440930 PMCID: PMC8394199 DOI: 10.3390/cells10082161] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 01/10/2023] Open
Abstract
Recently, the research on stemness and multilineage differentiation mechanisms has greatly increased its value due to the potential therapeutic impact of stem cell-based approaches. Stem cells modulate their self-renewing and differentiation capacities in response to endogenous and/or extrinsic factors that can control stem cell fate. One key factor controlling stem cell phenotype is oxygen (O2). Several pieces of evidence demonstrated that the complexity of reproducing O2 physiological tensions and gradients in culture is responsible for defective stem cell behavior in vitro and after transplantation. This evidence is still worsened by considering that stem cells are conventionally incubated under non-physiological air O2 tension (21%). Therefore, the study of mechanisms and signaling activated at lower O2 tension, such as those existing under native microenvironments (referred to as hypoxia), represent an effective strategy to define if O2 is essential in preserving naïve stemness potential as well as in modulating their differentiation. Starting from this premise, the goal of the present review is to report the status of the art about the link existing between hypoxia and stemness providing insight into the factors/molecules involved, to design targeted strategies that, recapitulating naïve O2 signals, enable towards the therapeutic use of stem cell for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Miriam Di Mattia
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (M.D.M.); (M.R.C.); (A.P.); (V.R.); (P.B.); (E.D.); (B.B.)
| | - Annunziata Mauro
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (M.D.M.); (M.R.C.); (A.P.); (V.R.); (P.B.); (E.D.); (B.B.)
- Correspondence: ; Tel.: +39-086-1426-6888; Fax: +39-08-6126-6860
| | - Maria Rita Citeroni
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (M.D.M.); (M.R.C.); (A.P.); (V.R.); (P.B.); (E.D.); (B.B.)
| | - Beatrice Dufrusine
- Department of Innovative Technologies in Medicine & Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy;
- Center of Advanced Studies and Technology (CAST), 66100 Chieti, Italy
| | - Alessia Peserico
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (M.D.M.); (M.R.C.); (A.P.); (V.R.); (P.B.); (E.D.); (B.B.)
| | - Valentina Russo
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (M.D.M.); (M.R.C.); (A.P.); (V.R.); (P.B.); (E.D.); (B.B.)
| | - Paolo Berardinelli
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (M.D.M.); (M.R.C.); (A.P.); (V.R.); (P.B.); (E.D.); (B.B.)
| | - Enrico Dainese
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (M.D.M.); (M.R.C.); (A.P.); (V.R.); (P.B.); (E.D.); (B.B.)
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, PA 19122, USA
| | - Barbara Barboni
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (M.D.M.); (M.R.C.); (A.P.); (V.R.); (P.B.); (E.D.); (B.B.)
| |
Collapse
|
218
|
Shrestha S, Lekkala VKR, Acharya P, Siddhpura D, Lee MY. Recent advances in microarray 3D bioprinting for high-throughput spheroid and tissue culture and analysis. Essays Biochem 2021; 65:481-489. [PMID: 34296737 PMCID: PMC9270997 DOI: 10.1042/ebc20200150] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/26/2022]
Abstract
Three-dimensional (3D) cell culture in vitro has proven to be more physiologically relevant than two-dimensional (2D) culture of cell monolayers, thus more predictive in assessing efficacy and toxicity of compounds. There have been several 3D cell culture techniques developed, which include spheroid and multicellular tissue cultures. Cell spheroids have been generated from single or multiple cell types cultured in ultralow attachment (ULA) well plates and hanging droplet plates. In general, cell spheroids are formed in a relatively short period of culture, in the absence of extracellular matrices (ECMs), via gravity-driven self-aggregation, thus having limited ability to self-organization in layered structure. On the other hand, multicellular tissue cultures including miniature tissues derived from pluripotent stem cells and adult stem cells (a.k.a. 'organoids') and 3D bioprinted tissue constructs require biomimetic hydrogels or ECMs and show highly ordered structure due to spontaneous self-organization of cells during differentiation and maturation processes. In this short review article, we summarize traditional methods of spheroid and multicellular tissue cultures as well as their technical challenges, and introduce how droplet-based, miniature 3D bioprinting ('microarray 3D bioprinting') can be used to improve assay throughput and reproducibility for high-throughput, predictive screening of compounds. Several platforms including a micropillar chip and a 384-pillar plate developed to facilitate miniature spheroid and tissue cultures via microarray 3D bioprinting are introduced. We excluded microphysiological systems (MPSs) in this article although they are important tissue models to simulate multiorgan interactions.
Collapse
Affiliation(s)
- Sunil Shrestha
- Department of Biomedical Engineering, University of North Texas, 1155 Union Circle, Denton, Texas 76203, United States
| | - Vinod Kumar Reddy Lekkala
- Department of Biomedical Engineering, University of North Texas, 1155 Union Circle, Denton, Texas 76203, United States
| | - Prabha Acharya
- Department of Biomedical Engineering, University of North Texas, 1155 Union Circle, Denton, Texas 76203, United States
| | - Darshita Siddhpura
- Department of Chemical and Biomedical Engineering, Cleveland State University, 2121 Euclid Ave, Cleveland, Ohio 44115, United States
| | - Moo-Yeal Lee
- Department of Biomedical Engineering, University of North Texas, 1155 Union Circle, Denton, Texas 76203, United States
| |
Collapse
|
219
|
Co-Stimulation of Oxytocin and Arginine-Vasopressin Receptors Affect Hypothalamic Neurospheroid Size. Int J Mol Sci 2021; 22:ijms22168464. [PMID: 34445168 PMCID: PMC8395152 DOI: 10.3390/ijms22168464] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 12/15/2022] Open
Abstract
Oxytocin (OXT) is a neuropeptide involved in a plethora of behavioral and physiological processes. However, there is a prominent lack of 3D cell culture models that investigate the effects of OXT on a cellular/molecular level. In this study, we established a hypothalamic neuronal spheroid model to investigate the cellular response in a more realistic 3D setting. Our data indicate that the formation of spheroids itself does not alter the basic characteristics of the cell line and that markers of cellular morphology and connectivity are stably expressed. We found that both OXT and arginine vasopressin (AVP) treatment increase spheroid size (surface area and volume), as well as individual nucleus size, which serves as an indicator for cellular proliferation. The cellular response to both OXT and AVP seems mainly to be mediated by the AVP receptor 1a (V1aR); however, the OXT receptor (OXTR) contributes significantly to the observed proliferative effect. When we blocked the OXTR pharmacologically or knocked down the OXTR by siRNA, the OXT- or AVP-induced cellular proliferation decreased. In summary, we established a 3D cell culture model of the neuronal response to OXT and AVP and found that spheroids react to the treatment via their respective receptors but also via cross-talk between the two receptor types.
Collapse
|
220
|
Abstract
Cell-based therapy is a promising approach in the field of regenerative medicine. As cells are formed into spheroids, their survival, functions, and engraftment in the transplanted site are significantly improved compared to single cell transplantation. To improve the therapeutic effect of cell spheroids even further, various biomaterials (e.g., nano- or microparticles, fibers, and hydrogels) have been developed for spheroid engineering. These biomaterials not only can control the overall spheroid formation (e.g., size, shape, aggregation speed, and degree of compaction), but also can regulate cell-to-cell and cell-to-matrix interactions in spheroids. Therefore, cell spheroids in synergy with biomaterials have recently emerged for cell-based regenerative therapy. Biomaterials-assisted spheroid engineering has been extensively studied for regeneration of bone or/and cartilage defects, critical limb ischemia, and myocardial infarction. Furthermore, it has been expanded to pancreas islets and hair follicle transplantation. This paper comprehensively reviews biomaterials-assisted spheroid engineering for regenerative therapy.
Collapse
Affiliation(s)
- Na-Hyun Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Oyunchimeg Bayaraa
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Zhou Zechu
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | | |
Collapse
|
221
|
Nagano H, Suematsu Y, Takuma M, Aoki S, Satoh A, Takayama E, Kinoshita M, Morimoto Y, Takeoka S, Fujie T, Kiyosawa T. Enhanced cellular engraftment of adipose-derived mesenchymal stem cell spheroids by using nanosheets as scaffolds. Sci Rep 2021; 11:14500. [PMID: 34262089 PMCID: PMC8280158 DOI: 10.1038/s41598-021-93642-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 06/28/2021] [Indexed: 02/07/2023] Open
Abstract
The short survival time of transplanted adipose-derived mesenchymal stem cells (ASCs) is a problem for skin wound healing. Transplantation after the formation of cellular spheroids has been investigated as a promising method for prolonging cellular survival. However, there have been technical restrictions for transplantation of spheroids in clinical practice. Here, we show an effective method for transplantation of ASC spheroids onto skin wounds in order to efficiently cure refractory ulcers. To assist anchoring of spheroids onto skin wounds, we used a 120-nm-thick free-standing film (nanosheet) that has a highly adhesive property. Bioluminescence imaging showed that ASC spheroids carried by the nanosheet survived for 14 days, which is about two-times longer than that previously reported. Wounds treated with a nanosheet carrying ASC spheroids were 4-times smaller than untreated wounds on day 14. This method for transplantation of spheroids could be applied to cell therapy for various refractory skin wounds.
Collapse
Affiliation(s)
- Hisato Nagano
- Department of Plastic and Reconstructive Surgery, National Defense Medical College, Tokorozawa, Saitama, 359-8513, Japan
| | - Yoshitaka Suematsu
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 162-8480, Japan
| | - Megumi Takuma
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, 226-8501, Japan
| | - Shimpo Aoki
- Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Ayano Satoh
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-0082, Japan
| | - Eiji Takayama
- Department of Oral Biochemistry, Asahi University School of Dentistry, Gifu, 501-0296, Japan
| | - Manabu Kinoshita
- Department of Immunology and Microbiology, National Defense Medical College, Tokorozawa, Saitama, 359-8513, Japan
| | - Yuji Morimoto
- Department of Physiology, National Defense Medical College, Tokorozawa, Saitama, 359-8513, Japan
| | - Shinji Takeoka
- Institute for Advanced Research of Biosystem Dynamics, Research Institute for Science and Engineering, Waseda University, 3-4-1 Ohkubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Toshinori Fujie
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, 226-8501, Japan
| | - Tomoharu Kiyosawa
- Department of Plastic and Reconstructive Surgery, National Defense Medical College, Tokorozawa, Saitama, 359-8513, Japan.
| |
Collapse
|
222
|
Barbet V, Broutier L. Future Match Making: When Pediatric Oncology Meets Organoid Technology. Front Cell Dev Biol 2021; 9:674219. [PMID: 34327198 PMCID: PMC8315550 DOI: 10.3389/fcell.2021.674219] [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: 02/28/2021] [Accepted: 05/10/2021] [Indexed: 11/13/2022] Open
Abstract
Unlike adult cancers that frequently result from the accumulation in time of mutational “hits” often linked to lifestyle, childhood cancers are emerging as diseases of dysregulated development through massive epigenetic alterations. The ability to reconstruct these differences in cancer models is therefore crucial for better understanding the uniqueness of pediatric cancer biology. Cancer organoids (i.e., tumoroids) represent a promising approach for creating patient-derived in vitro cancer models that closely recapitulate the overall pathophysiological features of natural tumorigenesis, including intra-tumoral heterogeneity and plasticity. Though largely applied to adult cancers, this technology is scarcely used for childhood cancers, with a notable delay in technological transfer. However, tumoroids could provide an unprecedented tool to unravel the biology of pediatric cancers and improve their therapeutic management. We herein present the current state-of-the-art of a long awaited and much needed matchmaking.
Collapse
Affiliation(s)
- Virginie Barbet
- Childhood Cancer & Cell Death (C3), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Lyon, France
| | - Laura Broutier
- Childhood Cancer & Cell Death (C3), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Lyon, France
| |
Collapse
|
223
|
Intramyocardial delivery of human cardiac stem cell spheroids with enhanced cell engraftment ability and cardiomyogenic potential for myocardial infarct repair. J Control Release 2021; 336:499-509. [PMID: 34224774 DOI: 10.1016/j.jconrel.2021.06.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 06/18/2021] [Accepted: 06/25/2021] [Indexed: 12/13/2022]
Abstract
Strategies for stem cell-based cardiac regeneration and repair are key issues for the ischemic heart disease (IHD) patients with chronic complications related to ischemic necrosis. Cardiac stem cells (CSCs) have demonstrated high therapeutic efficacy for IHD treatment owing to their specific cardiac-lineage commitment. The therapeutic potential of CSCs could be further enhanced by designing a cellular spheroid formulation. The spheroid culture condition of CSCs was optimized to ensure regulated size and minimal core necrosis in the spheroids. The CSC spheroids revealed mRNA profiles of the factors related to cardiac regeneration, angiogenesis, anti-inflammatory, and cardiomyocyte differentiation with a higher expression level than the CSCs. Intramyocardially delivered CSC spheroids in the rat IHD model resulted in a significant increase in retention rate by 1.82-fold (day 3) and 1.98-fold (day 14) compared to CSCs. Endothelial cell differentiation and neovascularization of the engrafted CSC spheroids were noted in the infarcted myocardium. CSC spheroids significantly promoted cardiac regeneration: i.e., decreased infarction and fibrotic area (11.22% and 4.18%) and increased left ventricle thickness (0.62 mm) compared to the untreated group. Cardiac performance was also improved by 2.04-fold and 1.44-fold increase in the ejection fraction and fractional shortening, respectively. Intramyocardial administration of CSC spheroids might serve as an advanced therapeutic modality with enhanced cell engraftment and regenerative abilities for cardiac repair after myocardial infarction.
Collapse
|
224
|
Li M, Fu T, Yang S, Pan L, Tang J, Chen M, Liang P, Gao Z, Guo L. Agarose-based spheroid culture enhanced stemness and promoted odontogenic differentiation potential of human dental follicle cells in vitro. In Vitro Cell Dev Biol Anim 2021; 57:620-630. [PMID: 34212339 PMCID: PMC8247612 DOI: 10.1007/s11626-021-00591-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/09/2021] [Indexed: 01/01/2023]
Abstract
Human dental follicle cells (HDFCs) are an ideal cell source of stem cells for dental tissue repair and regeneration and they have great potential for regenerative medicine applications. However, the conventional monolayer culture usually reduces cell proliferation and differentiation potential due to the continuous passage during in vitro expansion. In this study, primary HDFC spheroids were generated on 1% agarose, and the HDFCs spontaneously formed cell spheroids in the agarose-coated dishes. Compared with monolayer culture, the spheroid-derived HDFCs exhibited increased proliferative ability for later passage HDFCs as analysed by Cell Counting Kit-8 (CCK-8). The transcription-quantitative polymerase chain reaction (qRT-PCR), western blot and immunofluorescence assay showed that the expression of stemness marker genes Sox2, Oct4 and Nanog was increased significantly in the HDFC spheroids. Furthermore, we found that the odontogenic differentiation capability of HDFCs was significantly improved by spheroid culture in the agarose-coated dishes. On the other hand, the osteogenic differentiation capability was weakened compared with monolayer culture. Our results suggest that spheroid formation of HDFCs in agarose-coated dishes partially restores the proliferative ability of HDFCs at later passages, enhances their stemness and improves odontogenic differentiation capability in vitro. Therefore, spheroid formation of HDFCs has great therapeutic potential for stem cell clinical therapy.
Collapse
Affiliation(s)
- Min Li
- Department of Stomatology, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, People's Republic of China
| | - Tiwei Fu
- Chongqing Medical University Stomatology College, Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, People's Republic of China
| | - Sen Yang
- Stomatology Centre, Suining Central Hospital, Suining, 629000, People's Republic of China
| | - Lanlan Pan
- Department of Periodontics, Stomatology Hospital of Chongqing Medical University, Chongqing, 401147, People's Republic of China
| | - Jing Tang
- Department of Stomatology, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, People's Republic of China
| | - Meng Chen
- Department of Endodontics, Stomatology Hospital of Chongqing Medical University, Chongqing, 401147, People's Republic of China
| | - Panpan Liang
- Chongqing Medical University Stomatology College, Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, People's Republic of China
| | - Zhi Gao
- Department of Stomatology, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, People's Republic of China.
| | - Lijuan Guo
- Department of Stomatology, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, People's Republic of China.
- Department of Medical Cosmetology, Suining Central Hospital, Suining, 629000, People's Republic of China.
| |
Collapse
|
225
|
Heinrich MA, Mostafa AMRH, Morton JP, Hawinkels LJAC, Prakash J. Translating complexity and heterogeneity of pancreatic tumor: 3D in vitro to in vivo models. Adv Drug Deliv Rev 2021; 174:265-293. [PMID: 33895214 DOI: 10.1016/j.addr.2021.04.018] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 02/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive type of cancer with an overall survival rate of less than 7-8%, emphasizing the need for novel effective therapeutics against PDAC. However only a fraction of therapeutics which seemed promising in the laboratory environment will eventually reach the clinic. One of the main reasons behind this low success rate is the complex tumor microenvironment (TME) of PDAC, a highly fibrotic and dense stroma surrounding tumor cells, which supports tumor progression as well as increases the resistance against the treatment. In particular, the growing understanding of the PDAC TME points out a different challenge in the development of efficient therapeutics - a lack of biologically relevant in vitro and in vivo models that resemble the complexity and heterogeneity of PDAC observed in patients. The purpose and scope of this review is to provide an overview of the recent developments in different in vitro and in vivo models, which aim to recapitulate the complexity of PDAC in a laboratory environment, as well to describe how 3D in vitro models can be integrated into drug development pipelines that are already including sophisticated in vivo models. Hereby a special focus will be given on the complexity of in vivo models and the challenges in vitro models face to reach the same levels of complexity in a controllable manner. First, a brief introduction of novel developments in two dimensional (2D) models and ex vivo models is provided. Next, recent developments in three dimensional (3D) in vitro models are described ranging from spheroids, organoids, scaffold models, bioprinted models to organ-on-chip models including a discussion on advantages and limitations for each model. Furthermore, we will provide a detailed overview on the current PDAC in vivo models including chemically-induced models, syngeneic and xenogeneic models, highlighting hetero- and orthotopic, patient-derived tissues (PDX) models, and genetically engineered mouse models. Finally, we will provide a discussion on overall limitations of both, in vitro and in vivo models, and discuss necessary steps to overcome these limitations to reach an efficient drug development pipeline, as well as discuss possibilities to include novel in silico models in the process.
Collapse
Affiliation(s)
- Marcel A Heinrich
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands
| | - Ahmed M R H Mostafa
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands
| | - Jennifer P Morton
- Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Rd, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Rd, Glasgow G61 1QH, UK
| | - Lukas J A C Hawinkels
- Department of Gastroenterology-Hepatology, Leiden University Medical Centre, PO-box 9600, 2300 RC Leiden, the Netherlands
| | - Jai Prakash
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands.
| |
Collapse
|
226
|
Lee NH, Bayaraa O, Zechu Z, Kim HS. Biomaterials-assisted spheroid engineering for regenerative therapy. BMB Rep 2021; 54:356-367. [PMID: 34154700 PMCID: PMC8328824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/23/2021] [Accepted: 06/15/2021] [Indexed: 04/04/2024] Open
Abstract
Cell-based therapy is a promising approach in the field of regenerative medicine. As cells are formed into spheroids, their survival, functions, and engraftment in the transplanted site are significantly improved compared to single cell transplantation. To improve the therapeutic effect of cell spheroids even further, various biomaterials (e.g., nano- or microparticles, fibers, and hydrogels) have been developed for spheroid engineering. These biomaterials not only can control the overall spheroid formation (e.g., size, shape, aggregation speed, and degree of compaction), but also can regulate cell-to-cell and cell-to-matrix interactions in spheroids. Therefore, cell spheroids in synergy with biomaterials have recently emerged for cell-based regenerative therapy. Biomaterials-assisted spheroid engineering has been extensively studied for regeneration of bone or/and cartilage defects, critical limb ischemia, and myocardial infarction. Furthermore, it has been expanded to pancreas islets and hair follicle transplantation. This paper comprehensively reviews biomaterials-assisted spheroid engineering for regenerative therapy. [BMB Reports 2021; 54(7): 356-367].
Collapse
Affiliation(s)
- Na-Hyun Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Oyunchimeg Bayaraa
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Zhou Zechu
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Hye Sung Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan 31116, Korea
| |
Collapse
|
227
|
Fracture Healing Research-Shift towards In Vitro Modeling? Biomedicines 2021; 9:biomedicines9070748. [PMID: 34203470 PMCID: PMC8301383 DOI: 10.3390/biomedicines9070748] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/18/2021] [Accepted: 06/23/2021] [Indexed: 01/07/2023] Open
Abstract
Fractures are one of the most frequently occurring traumatic events worldwide. Approximately 10% of fractures lead to bone healing disorders, resulting in strain for affected patients and enormous costs for society. In order to shed light into underlying mechanisms of bone regeneration (habitual or disturbed), and to develop new therapeutic strategies, various in vivo, ex vivo and in vitro models can be applied. Undeniably, in vivo models include the systemic and biological situation. However, transferability towards the human patient along with ethical concerns regarding in vivo models have to be considered. Fostered by enormous technical improvements, such as bioreactors, on-a-chip-technologies and bone tissue engineering, sophisticated in vitro models are of rising interest. These models offer the possibility to use human cells from individual donors, complex cell systems and 3D models, therefore bridging the transferability gap, providing a platform for the introduction of personalized precision medicine and finally sparing animals. Facing diverse processes during fracture healing and thus various scientific opportunities, the reliability of results oftentimes depends on the choice of an appropriate model. Hence, we here focus on categorizing available models with respect to the requirements of the scientific approach.
Collapse
|
228
|
Fontana F, Marzagalli M, Sommariva M, Gagliano N, Limonta P. In Vitro 3D Cultures to Model the Tumor Microenvironment. Cancers (Basel) 2021; 13:cancers13122970. [PMID: 34199324 PMCID: PMC8231786 DOI: 10.3390/cancers13122970] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/06/2021] [Accepted: 06/09/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Tumor stroma is known to significantly influence cancer initiation and progression. In the last decade, 3D cell cultures have shown potential in modeling the tumor microenvironment. This review summarizes the main features of current 3D models, shedding light on their importance in the study of cancer biology and treatment. Abstract It is now well established that the tumor microenvironment plays a key role in determining cancer growth, metastasis and drug resistance. Thus, it is fundamental to understand how cancer cells interact and communicate with their stroma and how this crosstalk regulates disease initiation and progression. In this setting, 3D cell cultures have gained a lot of interest in the last two decades, due to their ability to better recapitulate the complexity of tumor microenvironment and therefore to bridge the gap between 2D monolayers and animal models. Herein, we present an overview of the 3D systems commonly used for studying tumor–stroma interactions, with a focus on recent advances in cancer modeling and drug discovery and testing.
Collapse
Affiliation(s)
- Fabrizio Fontana
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy; (M.M.); (P.L.)
- Correspondence: ; Tel.: +39-02-503-18427
| | - Monica Marzagalli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy; (M.M.); (P.L.)
| | - Michele Sommariva
- Department of Biomedical Sciences for Health, University of Milan, Via Mangiagalli 31, 20133 Milan, Italy; (M.S.); (N.G.)
| | - Nicoletta Gagliano
- Department of Biomedical Sciences for Health, University of Milan, Via Mangiagalli 31, 20133 Milan, Italy; (M.S.); (N.G.)
| | - Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy; (M.M.); (P.L.)
| |
Collapse
|
229
|
Liu X, Lin H, Song J, Zhang T, Wang X, Huang X, Zheng C. A Novel SimpleDrop Chip for 3D Spheroid Formation and Anti-Cancer Drug Assay. MICROMACHINES 2021; 12:681. [PMID: 34200752 PMCID: PMC8230402 DOI: 10.3390/mi12060681] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/03/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023]
Abstract
Cell culture is important for the rapid screening of anti-cancer drug candidates, attracting intense interest. Traditional 2D cell culture has been widely utilized in cancer biological research. However, 3D cellular spheroids are able to recapitulate the in vivo microenvironment of tissues or tumors. Thus far, several 3D cell culture methods have been developed, for instance, the hanging drop method, spinner flasks and micropatterned plates. Nevertheless, these methods have been reported to have some disadvantages, for example, medium replacement is inconvenient or causes cellular damage. Here, we report on an easy-to-operate and useful micro-hole culture chip (SimpleDrop) for 3D cellular spheroid formation and culture and drug analysis, which has advantages over the traditional method in terms of its ease of operation, lack of shear force and environmentally friendliness. On this chip, we observed the formation of a 3D spheroid clearly. Three drugs (paclitaxel, cisplatin and methotrexate) were tested by both cell viability assay and drug-induced apoptotic assay. The results show that the three drugs present a similar conclusion: cell viability decreased over time and concentration. Moreover, the apoptotic experiment showed a similar trend to the live/dead cell assay, in that the fraction of the apoptotic and necrotic cells correlated with the concentration and time. All these results prove that our SimpleDrop method is a useful and easy method for the formation of 3D cellular spheroids, which shows its potential for both cell-cell interaction research, tissue engineering and anticancer drug screening.
Collapse
Affiliation(s)
- Xiaoli Liu
- Department of Hematology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, China;
| | - Huichao Lin
- State Key Laboratory of Biobased Material and Green Papermaking, Department of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250300, China; (H.L.); (J.S.); (T.Z.)
| | - Jiaao Song
- State Key Laboratory of Biobased Material and Green Papermaking, Department of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250300, China; (H.L.); (J.S.); (T.Z.)
| | - Taiyi Zhang
- State Key Laboratory of Biobased Material and Green Papermaking, Department of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250300, China; (H.L.); (J.S.); (T.Z.)
| | - Xiaoying Wang
- Department of Pathology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, China;
| | - Xiaowen Huang
- State Key Laboratory of Biobased Material and Green Papermaking, Department of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250300, China; (H.L.); (J.S.); (T.Z.)
| | - Chengyun Zheng
- Department of Hematology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, China;
| |
Collapse
|
230
|
Lee JH, Song YM, Min SK, Lee HJ, Lee HL, Kim MJ, Park YH, Park JU, Park JB. NELL-1 Increased the Osteogenic Differentiation and mRNA Expression of Spheroids Composed of Stem Cells. ACTA ACUST UNITED AC 2021; 57:medicina57060586. [PMID: 34201046 PMCID: PMC8229008 DOI: 10.3390/medicina57060586] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/21/2021] [Accepted: 06/04/2021] [Indexed: 11/23/2022]
Abstract
Background and objectives: NELL-1 is a competent growth factor and it reported to target cells committed to the osteochondral lineage. The secreted, osteoinductive glycoproteins are reported to rheostatically control skeletal ossification. This study was performed to determine the effects of NELL-1 on spheroid morphology and cell viability and the promotion of osteogenic differentiation of stem cell spheroids. Materials and Methods: Cultures of stem cell spheroids of gingiva-derived stem cells were grown in the presence of NELL-1 at concentrations of 1, 10, 100, and 500 ng/mL. Evaluations of cell morphology were performed using a microscope, and cell viability was assessed using a two-color assay and Cell Counting Kit-8. Evaluation of the activity of alkaline phosphatase and calcium deposition assays involved anthraquinone dye assay to determine the level of osteogenic differentiation of cell spheroids treated with NELL-1. Real-time quantitative polymerase chain reaction (qPCR) was used to evaluate the expressions of RUNX2, BSP, OCN, COL1A1, and β-actin mRNAs. Results: The applied stem cells produced well-formed spheroids, and the addition of NELL-1 at tested concentrations did not show any apparent changes in spheroid shape. There were no significant changes in diameter with addition of NELL-1 at 0, 1, 10, 100, and 500 ng/mL concentrations. The quantitative cell viability results derived on Days 1, 3, and 7 did not show significant disparities among groups (p > 0.05). There was statistically higher alkaline phosphatase activity in the 10 ng/mL group compared with the unloaded control on Day 7 (p < 0.05). A significant increase in anthraquinone dye staining was observed with the addition of NELL-1, and the highest value was noted at 10 ng/mL (p < 0.05). qPCR results demonstrated that the mRNA expression levels of RUNX2 and BSP were significantly increased when NELL-1 was added to the culture. Conclusions: Based on these findings, we conclude that NELL-1 can be applied for increased osteogenic differentiation of stem cell spheroids.
Collapse
Affiliation(s)
- Jong-Ho Lee
- Department of Oral and Maxillofacial Surgery, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Young-Min Song
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (Y.-M.S.); (S.-K.M.); (H.-J.L.)
| | - Sae-Kyung Min
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (Y.-M.S.); (S.-K.M.); (H.-J.L.)
| | - Hyun-Jin Lee
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (Y.-M.S.); (S.-K.M.); (H.-J.L.)
| | - Hye-Lim Lee
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697, USA;
| | - Min-Ji Kim
- College of Dentistry, Chosun University, Gwangju 61452, Korea;
| | - Yoon-Hee Park
- Ebiogen, #405, Sungsu A1 Center 48 Ttukseom-ro 17-ga-gil, Seongdong-gu, Seoul 04785, Korea;
| | - Je-Uk Park
- Department of Oral and Maxillofacial Surgery, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
- Correspondence: (J.-U.P.); (J.-B.P.); Tel.: +82-2-2258-6291 (J.-U.P.); +82-2-2258-6290 (J.-B.P.)
| | - Jun-Beom Park
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (Y.-M.S.); (S.-K.M.); (H.-J.L.)
- Correspondence: (J.-U.P.); (J.-B.P.); Tel.: +82-2-2258-6291 (J.-U.P.); +82-2-2258-6290 (J.-B.P.)
| |
Collapse
|
231
|
Gibler P, Gimble J, Hamel K, Rogers E, Henderson M, Wu X, Olesky S, Frazier T. Human Adipose-Derived Stromal/Stem Cell Culture and Analysis Methods for Adipose Tissue Modeling In Vitro: A Systematic Review. Cells 2021; 10:1378. [PMID: 34204869 PMCID: PMC8227575 DOI: 10.3390/cells10061378] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/11/2022] Open
Abstract
Human adipose-derived stromal/stem cells (hASC) are widely used for in vitro modeling of physiologically relevant human adipose tissue. These models are useful for the development of tissue constructs for soft tissue regeneration and 3-dimensional (3D) microphysiological systems (MPS) for drug discovery. In this systematic review, we report on the current state of hASC culture and assessment methods for adipose tissue engineering using 3D MPS. Our search efforts resulted in the identification of 184 independent records, of which 27 were determined to be most relevant to the goals of the present review. Our results demonstrate a lack of consensus on methods for hASC culture and assessment for the production of physiologically relevant in vitro models of human adipose tissue. Few studies have assessed the impact of different 3D culture conditions on hASC adipogenesis. Additionally, there has been a limited use of assays for characterizing the functionality of adipose tissue in vitro. Results from this study suggest the need for more standardized culture methods and further analysis on in vitro tissue functionality. These will be necessary to validate the utility of 3D MPS as an in vitro model to reduce, refine, and replace in vivo experiments in the drug discovery regulatory process.
Collapse
Affiliation(s)
- Peyton Gibler
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Jeffrey Gimble
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
- Department of Structural and Cell Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Katie Hamel
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Emma Rogers
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Michael Henderson
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Xiying Wu
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Spencer Olesky
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Trivia Frazier
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
- Department of Structural and Cell Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
232
|
Lohberger B, Glaenzer D, Eck N, Steinecker-Frohnwieser B, Leithner A, Rinner B, Kerschbaum-Gruber S, Georg D. Effects of a combined therapy of bortezomib and ionizing radiation on chondrosarcoma three-dimensional spheroid cultures. Oncol Lett 2021; 21:428. [PMID: 33868466 PMCID: PMC8045153 DOI: 10.3892/ol.2021.12689] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 03/03/2021] [Indexed: 12/27/2022] Open
Abstract
Chondrosarcomas represent a heterogeneous group of primary bone cancers that are characterized by hyaline cartilaginous neoplastic tissue and are predominantly resistant to radiation and chemotherapy. However, adjuvant radiotherapy is often recommended in inoperable cases or after incomplete resections. To improve the efficiency of treatment, the present study tested a combination therapy with ionizing radiation (IR) and the proteasome inhibitor bortezomib. Using a three-dimensional (3D) spheroid model, 0-20 Gy of IR was applied to chondrosarcoma cells and healthy human chondrocytes. Following combined treatment with IR and bortezomib, the cell cycle distribution, apoptotic induction, the survivin pathway, autophagy and DNA damage were evaluated. Both cell types exhibited a slight decrease in viability following increasing doses of IR; the chondrosarcoma cells demonstrated a significant dose-dependent increase in the expression levels of the DNA damage marker histone H2AX phosphorylation at serine 139 (γH2AX). The combination treatment with bortezomib significantly decreased the cell viability after 48 h compared with that in irradiated cells. High-dose IR induced a G2/M phase arrest, which was accompanied by a decrease in the number of cells at the G1 and S phase. Co-treatment with bortezomib changed the distribution of the cell cycle phases. The mRNA expression levels of the proapoptotic genes Bcl-2-associated X protein (Bax) and Bak were significantly increased by bortezomib treatment and combination therapy with IR. In addition, the combination therapy resulted in a synergistic decrease of the expression levels of survivin and its corresponding downstream pathway molecules, including heat shock protein 90, X-linked inhibitor of apoptosis protein, smad 2 and smad 3. Comparative analyses of γH2AX at 1 and 24 h post-IR revealed efficient DNA repair in human chondrosarcoma cells. Therefore, additional bortezomib treatment may only temporarily improve the radiation sensitivity of chondrosarcoma cells. However, the inhibition of the survivin pathway by the combined treatment with IR and bortezomib, observed in the present study, revealed a novel aspect in the tumor biology of chondrosarcoma 3D spheroid cultures and may represent a potential target for therapy.
Collapse
Affiliation(s)
- Birgit Lohberger
- Department of Orthopedics and Trauma, Medical University of Graz, A-8036 Graz, Austria
| | - Dietmar Glaenzer
- Department of Orthopedics and Trauma, Medical University of Graz, A-8036 Graz, Austria
- Department for Rehabilitation, Ludwig Boltzmann Institute for Arthritis and Rehabilitation, A-5760 Saalfelden, Austria
| | - Nicole Eck
- Department of Orthopedics and Trauma, Medical University of Graz, A-8036 Graz, Austria
- Department for Rehabilitation, Ludwig Boltzmann Institute for Arthritis and Rehabilitation, A-5760 Saalfelden, Austria
| | | | - Andreas Leithner
- Department of Orthopedics and Trauma, Medical University of Graz, A-8036 Graz, Austria
| | - Beate Rinner
- Division of Biomedical Research, Medical University of Graz, A-8036 Graz, Austria
| | | | - Dietmar Georg
- Department of Radiation Oncology, Medical University of Vienna, A-1090 Vienna, Austria
- MedAustron Ion Therapy Center, A-2700 Wiener Neustadt, Austria
| |
Collapse
|
233
|
Jiang W, Yang Y, Mercer-Smith AR, Valdivia A, Bago JR, Woodell AS, Buckley AA, Marand MH, Qian L, Anders CK, Hingtgen SD. Development of next-generation tumor-homing induced neural stem cells to enhance treatment of metastatic cancers. SCIENCE ADVANCES 2021; 7:eabf1526. [PMID: 34108203 PMCID: PMC8189583 DOI: 10.1126/sciadv.abf1526] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 04/23/2021] [Indexed: 05/08/2023]
Abstract
Engineered tumor-homing neural stem cells (NSCs) have shown promise in treating cancer. Recently, we transdifferentiated skin fibroblasts into human-induced NSCs (hiNSC) as personalized NSC drug carriers. Here, using a SOX2 and spheroidal culture-based reprogramming strategy, we generated a new hiNSC variant, hiNeuroS, that was genetically distinct from fibroblasts and first-generation hiNSCs and had significantly enhanced tumor-homing and antitumor properties. In vitro, hiNeuroSs demonstrated superior migration to human triple-negative breast cancer (TNBC) cells and in vivo rapidly homed to TNBC tumor foci following intracerebroventricular (ICV) infusion. In TNBC parenchymal metastasis models, ICV infusion of hiNeuroSs secreting the proapoptotic agent TRAIL (hiNeuroS-TRAIL) significantly reduced tumor burden and extended median survival. In models of TNBC leptomeningeal carcinomatosis, ICV dosing of hiNeuroS-TRAIL therapy significantly delayed the onset of tumor formation and extended survival when administered as a prophylactic treatment, as well as reduced tumor volume while prolonging survival when delivered as established tumor therapy.
Collapse
Affiliation(s)
- Wulin Jiang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
| | - Yuchen Yang
- Department of Pathology and Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
| | - Alison R Mercer-Smith
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
| | - Alain Valdivia
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
| | - Juli R Bago
- Department of Hemato-Oncology, University Hospital of Ostrava, 708 52 Ostrava, Czech Republic
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic
| | - Alex S Woodell
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
| | - Andrew A Buckley
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
| | - Michael H Marand
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
| | - Carey K Anders
- Department of Medicine, Duke University, North Carolina, 27710, USA
| | - Shawn D Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA.
- Department of Neurosurgery, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27588, USA
| |
Collapse
|
234
|
Abstract
Cell spheroids have been studied as a biomimic medicine for tissue healing using cell sources. Rapid cell spheroid production increases cell survival and activity as well as the efficiency of mass production by reducing processing time. In this study, two-dimensional MXene (Ti3C2) particles were used to form mesenchymal stem cell spheroids, and the optimal MXene concentration, spheroid-production times, and bioactivity levels of spheroid cells during this process were assessed. A MXene concentration range of 1 to 10 μg/mL induced spheroid formation within 6 h. The MXene-induced spheroids exhibited osteogenic-differentiation behavior, with the highest activity levels at a concentration of 5 μg/mL. We report a novel and effective method for the rapid formation of stem cell spheroids using MXene.
Collapse
|
235
|
Kawaguchi H, Sakamoto T, Koya T, Togi M, Date I, Watanabe A, Yoshida K, Kato T, Nakamura Y, Ishigaki Y, Shimodaira S. Quality Verification with a Cluster-Controlled Manufacturing System to Generate Monocyte-Derived Dendritic Cells. Vaccines (Basel) 2021; 9:vaccines9050533. [PMID: 34065520 PMCID: PMC8160655 DOI: 10.3390/vaccines9050533] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 12/22/2022] Open
Abstract
Dendritic cell (DC) vaccines for cancer immunotherapy have been actively developed to improve clinical efficacy. In our previous report, monocyte−derived DCs induced by interleukin (IL)−4 with a low−adherence dish (low−adherent IL-4−DCs: la−IL-4−DCs) improved the yield and viability, as well as relatively prolonged survival in vitro, compared to IL-4−DCs developed using an adherent culture protocol. However, la−IL-4−DCs exhibit remarkable cluster formation and display heterogeneous immature phenotypes. Therefore, cluster formation in la−IL-4−DCs needs to be optimized for the clinical development of DC vaccines. In this study, we examined the effects of cluster control in the generation of mature IL-4−DCs, using cell culture vessels and measuring spheroid formation, survival, cytokine secretion, and gene expression of IL-4−DCs. Mature IL-4−DCs in cell culture vessels (cluster−controlled IL-4−DCs: cc−IL-4−DCs) displayed increased levels of CD80, CD86, and CD40 compared with that of la−IL-4−DCs. cc−IL-4−DCs induced antigen−specific cytotoxic T lymphocytes (CTLs) with a human leukocyte antigen (HLA)−restricted melanoma antigen recognized by T cells 1 (MART−1) peptide. Additionally, cc−IL-4−DCs produced higher levels of IFN−γ, possessing the CTL induction. Furthermore, DNA microarrays revealed the upregulation of BCL2A1, a pro−survival gene. According to these findings, the cc−IL-4−DCs are useful for generating homogeneous and functional IL-4−DCs that would be expected to promote long−lasting effects in DC vaccines.
Collapse
Affiliation(s)
- Haruhiko Kawaguchi
- Department of Regenerative Medicine, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa 920-0293, Japan; (H.K.); (T.S.); (T.K.); (M.T.); (I.D.); (A.W.)
| | - Takuya Sakamoto
- Department of Regenerative Medicine, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa 920-0293, Japan; (H.K.); (T.S.); (T.K.); (M.T.); (I.D.); (A.W.)
- Center for Regenerative Medicine, Kanazawa Medical University Hospital, Uchinada, Kahoku, Ishikawa 920-0293, Japan;
| | - Terutsugu Koya
- Department of Regenerative Medicine, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa 920-0293, Japan; (H.K.); (T.S.); (T.K.); (M.T.); (I.D.); (A.W.)
- Center for Regenerative Medicine, Kanazawa Medical University Hospital, Uchinada, Kahoku, Ishikawa 920-0293, Japan;
| | - Misa Togi
- Department of Regenerative Medicine, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa 920-0293, Japan; (H.K.); (T.S.); (T.K.); (M.T.); (I.D.); (A.W.)
- Center for Regenerative Medicine, Kanazawa Medical University Hospital, Uchinada, Kahoku, Ishikawa 920-0293, Japan;
| | - Ippei Date
- Department of Regenerative Medicine, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa 920-0293, Japan; (H.K.); (T.S.); (T.K.); (M.T.); (I.D.); (A.W.)
| | - Asuka Watanabe
- Department of Regenerative Medicine, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa 920-0293, Japan; (H.K.); (T.S.); (T.K.); (M.T.); (I.D.); (A.W.)
| | - Kenichi Yoshida
- Center for Regenerative Medicine, Kanazawa Medical University Hospital, Uchinada, Kahoku, Ishikawa 920-0293, Japan;
| | - Tomohisa Kato
- Medical Research Institute, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa 920-0293, Japan; (T.K.J.); (Y.N.); (Y.I.)
| | - Yuka Nakamura
- Medical Research Institute, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa 920-0293, Japan; (T.K.J.); (Y.N.); (Y.I.)
| | - Yasuhito Ishigaki
- Medical Research Institute, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa 920-0293, Japan; (T.K.J.); (Y.N.); (Y.I.)
| | - Shigetaka Shimodaira
- Department of Regenerative Medicine, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa 920-0293, Japan; (H.K.); (T.S.); (T.K.); (M.T.); (I.D.); (A.W.)
- Center for Regenerative Medicine, Kanazawa Medical University Hospital, Uchinada, Kahoku, Ishikawa 920-0293, Japan;
- Correspondence: ; Tel.: +81-76-218-8304
| |
Collapse
|
236
|
Meeremans M, Van de Walle GR, Van Vlierberghe S, De Schauwer C. The Lack of a Representative Tendinopathy Model Hampers Fundamental Mesenchymal Stem Cell Research. Front Cell Dev Biol 2021; 9:651164. [PMID: 34012963 PMCID: PMC8126669 DOI: 10.3389/fcell.2021.651164] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Overuse tendon injuries are a major cause of musculoskeletal morbidity in both human and equine athletes, due to the cumulative degenerative damage. These injuries present significant challenges as the healing process often results in the formation of inferior scar tissue. The poor success with conventional therapy supports the need to search for novel treatments to restore functionality and regenerate tissue as close to native tendon as possible. Mesenchymal stem cell (MSC)-based strategies represent promising therapeutic tools for tendon repair in both human and veterinary medicine. The translation of tissue engineering strategies from basic research findings, however, into clinical use has been hampered by the limited understanding of the multifaceted MSC mechanisms of action. In vitro models serve as important biological tools to study cell behavior, bypassing the confounding factors associated with in vivo experiments. Controllable and reproducible in vitro conditions should be provided to study the MSC healing mechanisms in tendon injuries. Unfortunately, no physiologically representative tendinopathy models exist to date. A major shortcoming of most currently available in vitro tendon models is the lack of extracellular tendon matrix and vascular supply. These models often make use of synthetic biomaterials, which do not reflect the natural tendon composition. Alternatively, decellularized tendon has been applied, but it is challenging to obtain reproducible results due to its variable composition, less efficient cell seeding approaches and lack of cell encapsulation and vascularization. The current review will overview pros and cons associated with the use of different biomaterials and technologies enabling scaffold production. In addition, the characteristics of the ideal, state-of-the-art tendinopathy model will be discussed. Briefly, a representative in vitro tendinopathy model should be vascularized and mimic the hierarchical structure of the tendon matrix with elongated cells being organized in a parallel fashion and subjected to uniaxial stretching. Incorporation of mechanical stimulation, preferably uniaxial stretching may be a key element in order to obtain appropriate matrix alignment and create a pathophysiological model. Together, a thorough discussion on the current status and future directions for tendon models will enhance fundamental MSC research, accelerating translation of MSC therapies for tendon injuries from bench to bedside.
Collapse
Affiliation(s)
- Marguerite Meeremans
- Comparative Physiology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Sandra Van Vlierberghe
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Catharina De Schauwer
- Comparative Physiology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
237
|
Badea MA, Balas M, Dinischiotu A. Biological properties and development of hypoxia in a breast cancer 3D model generated by hanging drop technique. Cell Biochem Biophys 2021; 80:63-73. [PMID: 33904026 DOI: 10.1007/s12013-021-00982-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 03/31/2021] [Indexed: 01/29/2023]
Abstract
Hanging drop represents a simple approach designed for the generation of 3D models that have potential to be used for the study of solid tumors characteristics. The aim of the study was to develop and characterize the breast cancer 3D cellular models obtained through hanging drop technique using MDA-MB-231 cells. The biological characteristics such as: morphology, cellular viability, proliferation capacity and hypoxia, were monitored for a six-day time period. The morphological evaluation indicated that the 3D models presented the aspect of compact (seeding density of 2500 and 5000 cells/drop) and loose (seeding density of 8000 cells/drop) aggregates, with a decrease in diameter and an increase of their circularity. The cellular viability and proliferation capacity decreased in time and the level of lactate dehydrogenase (LDH) increased in a time-dependent manner, suggesting the presence of necrotic cells that were dispersed in the cellular aggregates. The occurrence of hypoxia process was suggested by the up-regulation of Hsp70 protein expression and increased level of nitric oxide (NO). Moreover, the up-regulation of HIF-1α and poli-ubiquitinated Nrf2 protein expressions and decreased level of reduced glutathione (GSH) indicated the presence of an acute hypoxic environment in MDA-MB-231 3D aggregates. In conclusion, the MDA-MB-231 3D models generated through hanging drop are compact and loose aggregates characterized by an acute hypoxic condition.
Collapse
Affiliation(s)
- Madalina Andreea Badea
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, Bucharest, R-050095, Romania
| | - Mihaela Balas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, Bucharest, R-050095, Romania.
| | - Anca Dinischiotu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, Bucharest, R-050095, Romania
| |
Collapse
|
238
|
Chan YH, Lee YC, Hung CY, Yang PJ, Lai PC, Feng SW. Three-dimensional Spheroid Culture Enhances Multipotent Differentiation and Stemness Capacities of Human Dental Pulp-derived Mesenchymal Stem Cells by Modulating MAPK and NF-kB Signaling Pathways. Stem Cell Rev Rep 2021; 17:1810-1826. [PMID: 33893620 DOI: 10.1007/s12015-021-10172-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND Three-dimensional (3D) culture of mesenchymal stem cells has become an important research and development topic. However, comprehensive analysis of human dental pulp-derived mesenchymal stem cells (DPSCs) in 3D-spheroid culture remains unexplored. Thus, we evaluated the cellular characteristics, multipotent differentiation, gene expression, and related-signal transduction pathways of DPSCs in 3D-spheroid culture via magnetic levitation (3DM), compared with 2D-monolayer (2D) and 3D-aggregate (3D) cultures. METHODS The gross morphology and cellular ultrastructure were observed in the 2D, 3D, and 3DM experimental groups using scanning electron microscopy (SEM) and transmission electron microscopy (TEM). Surface markers and trilineage differentiation were evaluated using flow cytometry and staining analysis. Quantitative reverse transcription-polymerase chain reaction and immunofluorescence staining (IF) were performed to investigate the expression of differentiation and stemness markers. Signaling transduction pathways were evaluated using western blot analysis. RESULTS The morphology of cell aggregates and spheroids was largely influenced by the types of cell culture plates and initial cell seeding density. SEM and TEM experiments confirmed that the solid and firm structure of spheroids was quickly formed in the 3DM-medium without damaging cells. In addition, these three groups all expressed multilineage differentiation capabilities and surface marker expression. The trilineage differentiation capacities of the 3DM-group were significantly superior to the 2D and 3D-groups. The osteogenesis, angiogenesis, adipogenesis, and stemness-related genes were significantly enhanced in the 3D and 3DM-groups. The IF analysis showed that the extracellular matrix expression, osteogenesis, and angiogenesis proteins of the 3DM-group were significantly higher than those in the 2D and 3D-groups. Finally, 3DM-culture significantly activated the MAPK and NF-kB signaling transduction pathways and ameliorated the apoptosis effects of 3D-culture. CONCLUSIONS This study confirmed that 3DM-spheroids efficiently enhanced the therapeutic efficiency of DPSCs.
Collapse
Affiliation(s)
- Ya-Hui Chan
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chieh Lee
- Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chia-Yi Hung
- School of Dentistry, College of Oral Medicine, Taipei Medical University, No. 250, Wuxing St, Taipei, 11031, Taiwan
| | - Pi-Ju Yang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Pin-Chuang Lai
- Department of Diagnosis and Oral Health, School of Dentistry, University of Louisville, Louisville, KY, USA
| | - Sheng-Wei Feng
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan. .,School of Dentistry, College of Oral Medicine, Taipei Medical University, No. 250, Wuxing St, Taipei, 11031, Taiwan. .,Division of Prosthodontics, Department of Dentistry, Taipei Medical University Hospital, Taipei, Taiwan.
| |
Collapse
|
239
|
Kim D, Lee AE, Xu Q, Zhang Q, Le AD. Gingiva-Derived Mesenchymal Stem Cells: Potential Application in Tissue Engineering and Regenerative Medicine - A Comprehensive Review. Front Immunol 2021; 12:667221. [PMID: 33936109 PMCID: PMC8085523 DOI: 10.3389/fimmu.2021.667221] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/30/2021] [Indexed: 12/15/2022] Open
Abstract
A unique subpopulation of mesenchymal stem cells (MSCs) has been isolated and characterized from human gingival tissues (GMSCs). Similar to MSCs derived from other sources of tissues, e.g. bone marrow, adipose or umbilical cord, GMSCs also possess multipotent differentiation capacities and potent immunomodulatory effects on both innate and adaptive immune cells through the secretion of various types of bioactive factors with immunosuppressive and anti-inflammatory functions. Uniquely, GMSCs are highly proliferative and have the propensity to differentiate into neural cell lineages due to the neural crest-origin. These properties have endowed GMSCs with potent regenerative and therapeutic potentials in various preclinical models of human disorders, particularly, some inflammatory and autoimmune diseases, skin diseases, oral and maxillofacial disorders, and peripheral nerve injuries. All types of cells release extracellular vesicles (EVs), including exosomes, that play critical roles in cell-cell communication through their cargos containing a variety of bioactive molecules, such as proteins, nucleic acids, and lipids. Like EVs released by other sources of MSCs, GMSC-derived EVs have been shown to possess similar biological functions and therapeutic effects on several preclinical diseases models as GMSCs, thus representing a promising cell-free platform for regenerative therapy. Taken together, due to the easily accessibility and less morbidity of harvesting gingival tissues as well as the potent immunomodulatory and anti-inflammatory functions, GMSCs represent a unique source of MSCs of a neural crest-origin for potential application in tissue engineering and regenerative therapy.
Collapse
Affiliation(s)
- Dane Kim
- Department of Oral & Maxillofacial Surgery & Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Alisa E Lee
- Department of Oral & Maxillofacial Surgery & Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Qilin Xu
- Department of Oral & Maxillofacial Surgery & Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Qunzhou Zhang
- Department of Oral & Maxillofacial Surgery & Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Anh D Le
- Department of Oral & Maxillofacial Surgery & Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Center of Innovation & Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, United States.,Department of Oral & Maxillofacial Surgery, Penn Medicine Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
240
|
Ganguli A, Mostafa A, Saavedra C, Kim Y, Le P, Faramarzi V, Feathers RW, Berger J, Ramos-Cruz KP, Adeniba O, Diaz GJP, Drnevich J, Wright CL, Hernandez AG, Lin W, Smith AM, Kosari F, Vasmatzis G, Anastasiadis PZ, Bashir R. Three-dimensional microscale hanging drop arrays with geometric control for drug screening and live tissue imaging. SCIENCE ADVANCES 2021; 7:7/17/eabc1323. [PMID: 33893093 PMCID: PMC8064630 DOI: 10.1126/sciadv.abc1323] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 03/05/2021] [Indexed: 05/09/2023]
Abstract
Existing three-dimensional (3D) culture techniques are limited by trade-offs between throughput, capacity for high-resolution imaging in living state, and geometric control. Here, we introduce a modular microscale hanging drop culture where simple design elements allow high replicates for drug screening, direct on-chip real-time or high-resolution confocal microscopy, and geometric control in 3D. Thousands of spheroids can be formed on our microchip in a single step and without any selective pressure from specific matrices. Microchip cultures from human LN229 glioblastoma and patient-derived mouse xenograft cells retained genomic alterations of originating tumors based on mate pair sequencing. We measured response to drugs over time with real-time microscopy on-chip. Last, by engineering droplets to form predetermined geometric shapes, we were able to manipulate the geometry of cultured cell masses. These outcomes can enable broad applications in advancing personalized medicine for cancer and drug discovery, tissue engineering, and stem cell research.
Collapse
Affiliation(s)
- A Ganguli
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - A Mostafa
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - C Saavedra
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Y Kim
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - P Le
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - V Faramarzi
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - R W Feathers
- Mayo-Illinois Alliance for Technology-Based Healthcare, Urbana, IL, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - J Berger
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - K P Ramos-Cruz
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - O Adeniba
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - G J Pagan Diaz
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - J Drnevich
- High-Performance Biological Computing, Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - C L Wright
- DNA Services Lab, Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - A G Hernandez
- DNA Services Lab, Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - W Lin
- Mayo-Illinois Alliance for Technology-Based Healthcare, Urbana, IL, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - A M Smith
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, Urbana, IL 61820, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - F Kosari
- Mayo-Illinois Alliance for Technology-Based Healthcare, Urbana, IL, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - G Vasmatzis
- Mayo-Illinois Alliance for Technology-Based Healthcare, Urbana, IL, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - P Z Anastasiadis
- Mayo-Illinois Alliance for Technology-Based Healthcare, Urbana, IL, USA.
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - R Bashir
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Mayo-Illinois Alliance for Technology-Based Healthcare, Urbana, IL, USA
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, Urbana, IL 61820, USA
| |
Collapse
|
241
|
Eibl R, Senn Y, Gubser G, Jossen V, van den Bos C, Eibl D. Cellular Agriculture: Opportunities and Challenges. Annu Rev Food Sci Technol 2021; 12:51-73. [PMID: 33770467 DOI: 10.1146/annurev-food-063020-123940] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cellular agriculture is the controlled and sustainable manufacture of agricultural products with cells and tissues without plant or animal involvement. Today, microorganisms cultivated in bioreactors already produce egg and milk proteins, sweeteners, and flavors for human nutrition as well as leather and fibers for shoes, bags, and textiles. Furthermore, plant cell and tissue cultures provide ingredients that stimulate the immune system and improve skin texture, with another precommercial cellular agriculture product, in vitro meat, currently receiving a great deal of attention. All these approaches could assist traditional agriculture in continuing to provide for the dietary requirements of a growing world population while freeing up important resources such as arable land. Despite early successes, challenges remain and are discussed in this review, with a focus on production processes involving plant and animal cell and tissue cultures.
Collapse
Affiliation(s)
- Regine Eibl
- Institute of Chemistry and Biotechnology, Department of Life Sciences and Facility Management, Zurich University of Applied Sciences, Wädenswil 8820, Switzerland;
| | - Yannick Senn
- Institute of Chemistry and Biotechnology, Department of Life Sciences and Facility Management, Zurich University of Applied Sciences, Wädenswil 8820, Switzerland;
| | - Géraldine Gubser
- Institute of Chemistry and Biotechnology, Department of Life Sciences and Facility Management, Zurich University of Applied Sciences, Wädenswil 8820, Switzerland;
| | - Valentin Jossen
- Institute of Chemistry and Biotechnology, Department of Life Sciences and Facility Management, Zurich University of Applied Sciences, Wädenswil 8820, Switzerland;
| | | | - Dieter Eibl
- Institute of Chemistry and Biotechnology, Department of Life Sciences and Facility Management, Zurich University of Applied Sciences, Wädenswil 8820, Switzerland;
| |
Collapse
|
242
|
Sun M, Liu A, Yang X, Gong J, Yu M, Yao X, Wang H, He Y. 3D Cell Culture—Can It Be As Popular as 2D Cell Culture? ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202000066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Miao Sun
- The Affiliated Hospital of Stomatology School of Stomatology Zhejiang University School of Medicine and Key Laboratory of Oral Biomedical Research of Zhejiang Province Hangzhou Zhejiang 310000 China
| | - An Liu
- Department of Orthopaedic Surgery Second Affiliated Hospital School of Medicine Zhejiang University Hangzhou 310000 China
| | - Xiaofu Yang
- The Affiliated Hospital of Stomatology School of Stomatology Zhejiang University School of Medicine and Key Laboratory of Oral Biomedical Research of Zhejiang Province Hangzhou Zhejiang 310000 China
| | - Jiaxing Gong
- The Affiliated Hospital of Stomatology School of Stomatology Zhejiang University School of Medicine and Key Laboratory of Oral Biomedical Research of Zhejiang Province Hangzhou Zhejiang 310000 China
| | - Mengfei Yu
- The Affiliated Hospital of Stomatology School of Stomatology Zhejiang University School of Medicine and Key Laboratory of Oral Biomedical Research of Zhejiang Province Hangzhou Zhejiang 310000 China
| | - Xinhua Yao
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province School of Mechanical Engineering Zhejiang University Hangzhou 310000 China
| | - Huiming Wang
- The Affiliated Hospital of Stomatology School of Stomatology Zhejiang University School of Medicine and Key Laboratory of Oral Biomedical Research of Zhejiang Province Hangzhou Zhejiang 310000 China
| | - Yong He
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province School of Mechanical Engineering Zhejiang University Hangzhou 310000 China
- State Key Laboratory of Fluid Power and Mechatronic Systems School of Mechanical Engineering Zhejiang University Hangzhou 310000 China
| |
Collapse
|
243
|
Harman RM, Marx C, Van de Walle GR. Translational Animal Models Provide Insight Into Mesenchymal Stromal Cell (MSC) Secretome Therapy. Front Cell Dev Biol 2021; 9:654885. [PMID: 33869217 PMCID: PMC8044970 DOI: 10.3389/fcell.2021.654885] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
The therapeutic potential of the mesenchymal stromal cell (MSC) secretome, consisting of all molecules secreted by MSCs, is intensively studied. MSCs can be readily isolated, expanded, and manipulated in culture, and few people argue with the ethics of their collection. Despite promising pre-clinical studies, most MSC secretome-based therapies have not been implemented in human medicine, in part because the complexity of bioactive factors secreted by MSCs is not completely understood. In addition, the MSC secretome is variable, influenced by individual donor, tissue source of origin, culture conditions, and passage. An increased understanding of the factors that make up the secretome and the ability to manipulate MSCs to consistently secrete factors of biologic importance will improve MSC therapy. To aid in this goal, we can draw from the wealth of information available on secreted factors from MSC isolated from veterinary species. These translational animal models will inspire efforts to move human MSC secretome therapy from bench to bedside.
Collapse
Affiliation(s)
| | | | - Gerlinde R. Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| |
Collapse
|
244
|
Macrophage Plasticity and Function in the Lung Tumour Microenvironment Revealed in 3D Heterotypic Spheroid and Explant Models. Biomedicines 2021; 9:biomedicines9030302. [PMID: 33804204 PMCID: PMC7999110 DOI: 10.3390/biomedicines9030302] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 11/16/2022] Open
Abstract
In non-small cell lung cancer (NSCLC), stroma-resident and tumour-infiltrating macrophages may facilitate an immunosuppressive tumour microenvironment (TME) and hamper immunotherapeutic responses. Analysis of tumour-associated macrophage (TAM) plasticity in NSCLC is largely lacking. We established a novel, multi-marker, dual analysis approach for assessing monocyte-derived macrophage (Mφ) polarisation and M1/M2 phenotypic plasticity. We developed a flow cytometry-based, two-marker analysis (CD64 and CD206) of CD14+ cells. The phenotype and immune function of in vitro-induced TAMs was studied in a heterotypic spheroid and tumour-derived explant model of NSCLC. Heterotypic spheroids and NSCLC explants skewed Mφs from an M1- (CD206loCD64hi) to M2-like (CD206hiCD64lo) phenotype. Lipopolysaccharide (LPS) and IFNγ treatment reversed M2-like Mφ polarisation, indicating the plasticity of Mφs. Importantly, antigen-specific CD8+ T cell responses were reduced in the presence of tumour explant-conditioned Mφs, but not spheroid-conditioned Mφs, suggesting explants are likely a more relevant model of the immune TME than cell line-derived spheroids. Our data indicates the importance of multi-marker, functional analyses within Mφ subsets and the advantages of the ex vivo NSCLC explant model in immunomodulation studies. We highlight the plasticity of the M1/M2 phenotype using the explant model and provide a tool for studying therapeutic interventions designed to reprogram M2-like Mφ-induced immunosuppression.
Collapse
|
245
|
Tieu T, Irani S, Bremert KL, Ryan NK, Wojnilowicz M, Helm M, Thissen H, Voelcker NH, Butler LM, Cifuentes‐Rius A. Patient-Derived Prostate Cancer Explants: A Clinically Relevant Model to Assess siRNA-Based Nanomedicines. Adv Healthc Mater 2021; 10:e2001594. [PMID: 33274851 DOI: 10.1002/adhm.202001594] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/05/2020] [Indexed: 01/27/2023]
Abstract
Over the last thirty years, research in nanomedicine has widely been focused on applications in cancer therapeutics. However, despite the plethora of reported nanoscale drug delivery systems that can successfully eradicate solid tumor xenografts in vivo, many of these formulations have not yet achieved clinical translation. This issue particularly pertains to the delivery of small interfering RNA (siRNA), a highly attractive tool for selective gene targeting. One of the likely reasons behind the lack of translation is that current in vivo models fail to recapitulate critical elements of clinical solid tumors that may influence drug response, such as cellular heterogeneity in the tumor microenvironment. This study incorporates a more clinically relevant model for assessing siRNA delivery systems; ex vivo culture of prostate cancer harvested from patients who have undergone radical prostatectomy, denoted patient-derived explants (PDE). The model retains native human tissue architecture, microenvironment, and cell signaling pathways. Porous silicon nanoparticles (pSiNPs) behavior in this model is investigated and compared with commonly used 3D cancer cell spheroids for their efficacy in the delivery of siRNA directed against the androgen receptor (AR), a key driver of prostate cancer.
Collapse
Affiliation(s)
- Terence Tieu
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
- CSIRO Manufacturing Bayview Avenue Clayton Victoria 3168 Australia
| | - Swati Irani
- South Australian Health & Medical Research Institute Adelaide SA 5001 Australia
- Adelaide Medical School & Freemasons Foundation Centre for Men's Health University of Adelaide Adelaide SA 5005 Australia
| | - Kayla L. Bremert
- South Australian Health & Medical Research Institute Adelaide SA 5001 Australia
- Adelaide Medical School & Freemasons Foundation Centre for Men's Health University of Adelaide Adelaide SA 5005 Australia
| | - Natalie K. Ryan
- South Australian Health & Medical Research Institute Adelaide SA 5001 Australia
- Adelaide Medical School & Freemasons Foundation Centre for Men's Health University of Adelaide Adelaide SA 5005 Australia
| | | | - Madison Helm
- South Australian Health & Medical Research Institute Adelaide SA 5001 Australia
- Adelaide Medical School & Freemasons Foundation Centre for Men's Health University of Adelaide Adelaide SA 5005 Australia
| | - Helmut Thissen
- CSIRO Manufacturing Bayview Avenue Clayton Victoria 3168 Australia
| | - Nicolas H. Voelcker
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
- CSIRO Manufacturing Bayview Avenue Clayton Victoria 3168 Australia
- Melbourne Centre for Nanofabrication Victorian Node of the Australian National Fabrication Facility 151 Wellington Road Clayton Victoria 3168 Australia
| | - Lisa M. Butler
- South Australian Health & Medical Research Institute Adelaide SA 5001 Australia
- Adelaide Medical School & Freemasons Foundation Centre for Men's Health University of Adelaide Adelaide SA 5005 Australia
| | - Anna Cifuentes‐Rius
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
| |
Collapse
|
246
|
Luo L, Zhang W, Chen W, Fu X, Wang X, Xu R, Hu D. Based on a Self-Feeder Layer, a Novel 3D Culture Model of Human ADSCs Facilitates Trans-Differentiation of the Spheroid Cells into Neural Progenitor-Like Cells Using siEID3 with a Laminin/Poly-d-lysine Matrix. Cells 2021; 10:493. [PMID: 33668931 PMCID: PMC7996540 DOI: 10.3390/cells10030493] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/18/2021] [Accepted: 02/21/2021] [Indexed: 12/21/2022] Open
Abstract
Human adipose-derived stromal cells (ADSCs) are receiving unprecedented attention as a potential cellular source for regenerative medicine-based therapies against various diseases and conditions. However, there still have significant issues concerning the translational development of ADSC-based therapies, such as its heterogeneity and being prone to aging. We developed a new simple and economical 3D semi-suspended expansion method in which 3D spheroids reside on an ADSC-derived self-feeder cell layer, producing cells with increased population homogeneity and strong stemness and ensuring that the proliferation and differentiation potency of the cells does not become notably reduced after at least ten passages in culture. To check the potential application of the 3D ADSC spheroids, we discovered that the combination of siEID3, which is a small interfering RNA of EP300 inhibitor of differentiation 3 (EID3), and laminin/poly-d-lysine matrix can rapidly result in trans-differentiation of the 3D spheroid cells to neural progenitor-like cells (NPLCs) in approximately 9 days in vitro. This approach provides a multidisciplinary tool for stem cell research and production in mesenchymal stem cell-related fields.
Collapse
Affiliation(s)
- Liang Luo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi’an 710003, China; (W.Z.); (X.W.)
- Stem Cell Research Center, Neurosurgery Institute of PLA Army, Beijing 100700, China; (W.C.); (X.F.)
| | - Wei Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi’an 710003, China; (W.Z.); (X.W.)
- Department of Plastics and Aesthetic Surgery, the First Affiliated Hospital of Xi’an Medical University, Xi’an 710077, China
| | - Wenjin Chen
- Stem Cell Research Center, Neurosurgery Institute of PLA Army, Beijing 100700, China; (W.C.); (X.F.)
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510000, China
| | - Xiaojun Fu
- Stem Cell Research Center, Neurosurgery Institute of PLA Army, Beijing 100700, China; (W.C.); (X.F.)
- Bayi Brain Hospital, General Hospital of PLA Army, Beijing 100700, China
| | - Xujie Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi’an 710003, China; (W.Z.); (X.W.)
| | - Ruxiang Xu
- Stem Cell Research Center, Neurosurgery Institute of PLA Army, Beijing 100700, China; (W.C.); (X.F.)
- Department of Plastics and Aesthetic Surgery, the First Affiliated Hospital of Xi’an Medical University, Xi’an 710077, China
- The Department of Neurosurgery, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu 610072, China
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi’an 710003, China; (W.Z.); (X.W.)
| |
Collapse
|
247
|
Organoid and Spheroid Tumor Models: Techniques and Applications. Cancers (Basel) 2021; 13:cancers13040874. [PMID: 33669619 PMCID: PMC7922036 DOI: 10.3390/cancers13040874] [Citation(s) in RCA: 201] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/05/2021] [Accepted: 02/11/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Cell cultures can be carried out in three dimensions (3D). Organoids and spheroids are different 3D cell culture models that can be cultured with different techniques. These 3D cell culture units established from a patient tumor have several similarities to the original tumor tissue and possess several advantages in conducting basic and clinical cancer research. Organoids prepared from a patient tissue can be preserved in a living biobank. Testing chemo-, radio- and immuno-therapies on these organoids has the potential to predict the patient responses and these models have incredible promise for personalized medicine. This review presents different organoid models, the techniques to prepare them and recent advances in their applications. Abstract Techniques to develop three-dimensional cell culture models are rapidly expanding to bridge the gap between conventional cell culture and animal models. Organoid and spheroid cultures have distinct and overlapping purposes and differ in cellular sources and protocol for establishment. Spheroids are of lower complexity structurally but are simple and popular models for drug screening. Organoids histologically and genetically resemble the original tumor from which they were derived. Ease of generation, ability for long-term culture and cryopreservation make organoids suitable for a wide range of applications. Organoids-on-chip models combine organoid methods with powerful designing and fabrication of micro-chip technology. Organoid-chip models can emulate the dynamic microenvironment of tumor pathophysiology as well as tissue–tissue interactions. In this review, we outline different tumor spheroid and organoid models and techniques to establish them. We also discuss the recent advances and applications of tumor organoids with an emphasis on tumor modeling, drug screening, personalized medicine and immunotherapy.
Collapse
|
248
|
Maury P, Porcel E, Mau A, Lux F, Tillement O, Mahou P, Schanne-Klein MC, Lacombe S. Rapid Evaluation of Novel Therapeutic Strategies Using a 3D Collagen-Based Tissue-Like Model. Front Bioeng Biotechnol 2021; 9:574035. [PMID: 33681152 PMCID: PMC7929985 DOI: 10.3389/fbioe.2021.574035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 01/18/2021] [Indexed: 11/25/2022] Open
Abstract
2D cell cultures are commonly used to rapidly evaluate the therapeutic potential of various treatments on living cells. However, the effects of the extracellular matrix (ECM) including the 3D arrangement of cells and the complex physiology of native environment are missing, which makes these models far from in vivo conditions. 3D cell models have emerged in preclinical studies to simulate the impact of the ECM and partially bridge the gap between monolayer cultures and in vivo tissues. To date, the difficulty to handle the existing 3D models, the cost of their production and their poor reproducibility have hindered their use. Here, we present a reproducible and commercially available "3D cell collagen-based model" (3D-CCM) that allows to study the influence of the matrix on nanoagent uptake and radiation effects. The cell density in these samples is homogeneous. The oxygen concentration in the 3D-CCM is tunable, which opens the opportunity to investigate hypoxic effects. In addition, thanks to the intrinsic properties of the collagen, the second harmonic imaging microscopy may be used to probe the whole volume and visualize living cells in real-time. Thus, the architecture and composition of 3D-CCMs as well as the impact of various therapeutic strategies on cells embedded in the ECM is observed directly. Moreover, the disaggregation of the collagen matrix allows recovering of cells without damaging them. It is a major advantage that makes possible single cell analysis and quantification of treatment effects using clonogenic assay. In this work, 3D-CCMs were used to evaluate the correlative efficacies of nanodrug exposure and medical radiation on cells contained in a tumor like sample. A comparison with monolayer cell cultures was performed showing the advantageous outcome and the higher potential of 3D-CCMs. This cheap and easy to handle approach is more ethical than in vivo experiments, thus, giving a fast evaluation of cellular responses to various treatments.
Collapse
Affiliation(s)
- Pauline Maury
- Université Paris-Saclay, CNRS, Institut des Sciences Moléculaires d’Orsay, Orsay, France
| | - Erika Porcel
- Université Paris-Saclay, CNRS, Institut des Sciences Moléculaires d’Orsay, Orsay, France
| | - Adrien Mau
- Université Paris-Saclay, CNRS, Institut des Sciences Moléculaires d’Orsay, Orsay, France
| | - François Lux
- Institut Universitaire de France, Paris, France
- Institut Lumière Matière, Université Claude Bernard Lyon 1, UMR 5306 CNRS-UCBL, Villeurbanne, France
| | - Olivier Tillement
- Institut Lumière Matière, Université Claude Bernard Lyon 1, UMR 5306 CNRS-UCBL, Villeurbanne, France
| | - Pierre Mahou
- Laboratoire d’Optique et Biosciences, Ecole Polytechnique, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut Polytechnique de Paris, Palaiseau, France
| | - Marie-Claire Schanne-Klein
- Laboratoire d’Optique et Biosciences, Ecole Polytechnique, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut Polytechnique de Paris, Palaiseau, France
| | - Sandrine Lacombe
- Université Paris-Saclay, CNRS, Institut des Sciences Moléculaires d’Orsay, Orsay, France
| |
Collapse
|
249
|
Kulkarni NS, Vaidya B, Gupta V. Nano-synergistic combination of Erlotinib and Quinacrine for non-small cell lung cancer (NSCLC) therapeutics - Evaluation in biologically relevant in-vitro models. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 121:111891. [PMID: 33579503 DOI: 10.1016/j.msec.2021.111891] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/07/2021] [Accepted: 01/10/2021] [Indexed: 01/10/2023]
Abstract
Non-small cell lung cancer (NSCLC), pre-dominant subtype of lung cancer, is a global disorder affecting millions worldwide. One of the early treatments for NSCLC was use of a first-generation tyrosine kinase inhibitor, Erlotinib (Erlo). However, chronic exposure to Erlo led to development of acquired drug resistance (ADR) in NSCLC, limiting the clinical use of Erlo. A potential approach to overcome development of ADR is a multi-drug therapy. It has been previously reported that Erlo and Quinacrine (QA), an anti-malarial drug, can work synergistically to inhibit tumor progression in NSCLC. However, the combination failed at clinical stages, citing lack of efficacy. In this study, an effort has been made to improve the efficacy of Erlo-QA combination via development of nanoformulations, known to enhance therapeutic efficacy of potent chemotherapies. Synergy between Erlo and QA was measured via estimating the combination indices (CI). It was seen that established combination of nanoformulations (CI: 0.25) had better synergy than plain drug solutions (CI: 0.85) in combination. Following extensive in-vitro testing, data were simulated in biologically relevant 3D tumor models. Two tumor models were developed for extensive in-vitro testing, 3D-Spheroids grown in ultra-low attachment culture plates for efficacy evaluation and a 5D-spheroid model in 5D-sphericalplate with capability of growing 750 spheroids/well for protein expression analysis. Extensive studies on these models revealed that combination of Erlo and QA nanoformulations overall had a better effect in terms of synergy enhancement as compared to plain drug combination. Further, effect of combinatorial therapy on molecular markers was evaluated on 5D-Sphericalplate leading to similar effects on synergy enhancement. Results from present study suggests that combination of nanoformulations can improve the synergy between Erlo and QA while reducing the overall therapeutic dose.
Collapse
Affiliation(s)
- Nishant S Kulkarni
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | | | - Vivek Gupta
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA; School of Pharmacy, Keck Graduate Institute, Claremont, CA, USA.
| |
Collapse
|
250
|
Schmitz C, Potekhina E, Belousov VV, Lavrentieva A. Hypoxia Onset in Mesenchymal Stem Cell Spheroids: Monitoring With Hypoxia Reporter Cells. Front Bioeng Biotechnol 2021; 9:611837. [PMID: 33614611 PMCID: PMC7892969 DOI: 10.3389/fbioe.2021.611837] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/08/2021] [Indexed: 12/20/2022] Open
Abstract
The therapeutic and differentiation potential of human mesenchymal stems cells (hMSCs) makes these cells a promising candidate for cellular therapies and tissue engineering. On the path of a successful medical application of hMSC, the cultivation of cells in a three-dimensional (3D) environment was a landmark for the transition from simple two-dimensional (2D) testing platforms to complex systems that mimic physiological in vivo conditions and can improve hMSC curative potential as well as survival after implantation. A 3D arrangement of cells can be mediated by scaffold materials where cells get entrapped in pores, or by the fabrication of spheroids, scaffold-free self-organized cell aggregates that express their own extracellular matrix. Independently from the cultivation method, cells expanded in 3D experience an inhomogeneous microenvironment. Many gradients in nutrient supply, oxygen supply, and waste disposal from one hand mimic in vivo microenvironment, but also put every cell in the 3D construct in a different context. Since oxygen concentration in spheroids is compromised in a size-dependent manner, it is crucial to have a closer insight on the thresholds of hypoxic response in such systems. In this work, we want to improve our understanding of oxygen availability and consequensing hypoxia onset in hMSC spheroids. Therefore, we utilized human adipose tissue-derived MSCs (hAD-MSCs) modified with a genetical sensor construct to reveal (I) the influence of spheroid production methods and (II) hMSCs cell number per spheroid to detect the onset of hypoxia in aggregates. We could demonstrate that not only higher cell numbers of MSCs, but also spheroid formation method plays a critical role in onset of hypoxia.
Collapse
Affiliation(s)
- Carola Schmitz
- Institute of Technical Chemistry, Gottfried Wilhelm Leibniz University Hannover, Hanover, Germany
| | - Ekaterina Potekhina
- Department of Metabolism and Redox Biology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Vsevolod V Belousov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia.,Federal Center of Brain Research and Neurotechnologies, Federal Biomedical Agency, Moscow, Russia
| | - Antonina Lavrentieva
- Institute of Technical Chemistry, Gottfried Wilhelm Leibniz University Hannover, Hanover, Germany
| |
Collapse
|