301
|
Ouyang L, Yao R, Chen X, Na J, Sun W. 3D printing of HEK 293FT cell-laden hydrogel into macroporous constructs with high cell viability and normal biological functions. Biofabrication 2015; 7:015010. [PMID: 25691496 DOI: 10.1088/1758-5090/7/1/015010] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
3D printing has evolved into a versatile technology for fabricating tissue-engineered constructs with spatially controlled cells and biomaterial distribution to allow biomimicking of in vivo tissues. In this paper, we reported a novel study of 3D printing of cell lines derived from human embryonic kidney tissue into a macroporous tissue-like construct. Nozzle temperature, chamber temperature and the composition of the matrix material were studied to achieve high cell viability (>90%) after 3D printing and construct formation. Long-term construct stability with a clear grid structure up to 30 days was observed. Cells continued to grow as cellular spheroids with strong cell-cell interactions. Two transfected cell lines of HEK 293FT were also 3D printed and showed normal biological functions, i.e. protein synthesis and gene activation in responding to small molecule stimulus. With further refinement, this 3D cell printing technology may lead to a practical fabrication of functional embryonic tissues in vitro.
Collapse
Affiliation(s)
- Liliang Ouyang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China. Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, People's Republic of China
| | | | | | | | | |
Collapse
|
302
|
Adjei IM, Blanka S. Modulation of the tumor microenvironment for cancer treatment: a biomaterials approach. J Funct Biomater 2015; 6:81-103. [PMID: 25695337 PMCID: PMC4384103 DOI: 10.3390/jfb6010081] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 10/07/2014] [Accepted: 02/12/2015] [Indexed: 12/26/2022] Open
Abstract
Tumors are complex tissues that consist of stromal cells, such as fibroblasts, immune cells and mesenchymal stem cells, as well as non-cellular components, in addition to neoplastic cells. Increasingly, there is evidence to suggest that these non-neoplastic cell components support cancer initiation, progression and metastasis and that their ablation or reprogramming can inhibit tumor growth. Our understanding of the activities of different parts of the tumor stroma in advancing cancer has been improved by the use of scaffold and matrix-based 3D systems originally developed for regenerative medicine. Additionally, drug delivery systems made from synthetic and natural biomaterials deliver drugs to kill stromal cells or reprogram the microenvironment for tumor inhibition. In this article, we review the impact of 3D tumor models in increasing our understanding of tumorigenesis. We also discuss how different drug delivery systems aid in the reprogramming of tumor stroma for cancer treatment.
Collapse
Affiliation(s)
- Isaac M Adjei
- Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA.
| | - Sharma Blanka
- Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
303
|
Lei H, Hofferberth SC, Liu R, Colby A, Tevis KM, Catalano P, Grinstaff MW, Colson YL. Paclitaxel-loaded expansile nanoparticles enhance chemotherapeutic drug delivery in mesothelioma 3-dimensional multicellular spheroids. J Thorac Cardiovasc Surg 2015; 149:1417-24; discussion 1424-25.e1. [PMID: 25841659 DOI: 10.1016/j.jtcvs.2015.02.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 02/02/2015] [Accepted: 02/07/2015] [Indexed: 12/28/2022]
Abstract
OBJECTIVES Intraperitoneal administration of paclitaxel-loaded expansile nanoparticles (Pax-eNPs) significantly improves survival in an in vivo model of malignant mesothelioma compared with conventional drug delivery with the clinically utilized Cremophor EL/ethanol (C/E) excipient. However, in vitro monolayer cell culture experiments do not replicate this superior efficacy, suggesting Pax-eNPs utilize a unique mechanism of drug delivery. Using a mesothelioma spheroid model, we characterized the mechanisms of enhanced tumor cytotoxicity leveraged by Pax-eNPs. METHODS Human malignant mesothelioma (MSTO-211H) spheroids were co-incubated for 24 hours with Oregon Green-conjugated paclitaxel dissolved in C/E or loaded into eNPs. Oregon Green-paclitaxel uptake was measured as Oregon Green intensity via confocal microscopy and kinetics of tumor cytotoxicity were assessed via propidium iodide staining. Pharmacologic endocytotic inhibitors were used to elucidate mechanisms of eNP uptake into spheroids. RESULTS Increased drug penetration and a 38-fold higher intraspheroidal drug concentration were observed 24 hours after MSTO-211H spheroids were treated with Oregon Green-conjugated paclitaxel loaded into eNPs compared with Oregon Green-conjugated paclitaxel dissolved in C/E (P < .01). Macropinocytosis was the dominant endocytotic pathway of eNP uptake. Spheroids were more susceptible to paclitaxel when delivered via eNP, exhibiting more than twice the propidium iodine intensity compared with an equivalent paclitaxel-C/E dose. CONCLUSIONS Compared with monolayer cell culture, the in vitro 3-D tumor spheroid model better reflects the superior in vivo efficacy of Pax-eNPs. Persistent tumor penetration and prolonged intratumoral release are unique mechanisms of Pax-eNP cytotoxicity. 3-D spheroid models are valuable tools for investigating cytotoxic mechanisms and nanoparticle-tumor interactions, particularly given the costs and limitations of in vivo animal studies.
Collapse
Affiliation(s)
- Hongyi Lei
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, Mass; Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Sophie C Hofferberth
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, Mass
| | - Rong Liu
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, Mass
| | - Aaron Colby
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, Mass
| | - Kristie M Tevis
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, Mass
| | - Paul Catalano
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, Mass
| | - Mark W Grinstaff
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, Mass
| | - Yolonda L Colson
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, Mass.
| |
Collapse
|
304
|
Mabry KM, Lawrence RL, Anseth KS. Dynamic stiffening of poly(ethylene glycol)-based hydrogels to direct valvular interstitial cell phenotype in a three-dimensional environment. Biomaterials 2015; 49:47-56. [PMID: 25725554 PMCID: PMC4346780 DOI: 10.1016/j.biomaterials.2015.01.047] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 12/19/2014] [Accepted: 01/20/2015] [Indexed: 01/13/2023]
Abstract
Valvular interstitial cells (VICs) are active regulators of valve homeostasis and disease, responsible for secreting and remodeling the valve tissue matrix. As a result of VIC activity, the valve modulus can substantially change during development, injury and repair, and disease progression. While two-dimensional biomaterial substrates have been used to study mechanosensing and its influence on VIC phenotype, less is known about how these cells respond to matrix modulus in a three-dimensional environment. Here, we synthesized MMP-degradable poly(ethylene glycol) (PEG) hydrogels with elastic moduli ranging from 0.24 kPa to 12 kPa and observed that cell morphology was constrained in stiffer gels. To vary gel stiffness without substantially changing cell morphology, cell-laden hydrogels were cultured in the 0.24 kPa gels for 3 days to allow VIC spreading, and then stiffened in situ via a second, photoinitiated thiol-ene polymerization such that the gel modulus increased from 0.24 kPa to 1.2 kPa or 13 kPa. VICs encapsulated within soft gels exhibited αSMA stress fibers (∼ 40%), a hallmark of the myofibroblast phenotype. Interestingly, in stiffened gels, VICs became deactivated to a quiescent fibroblast phenotype, suggesting that matrix stiffness directs VIC phenotype independent of morphology, but in a manner that depends on the dimensionality of the culture platform. Collectively, these studies present a versatile method for dynamic stiffening of hydrogels and demonstrate the significant effects of matrix modulus on VIC myofibroblast properties in three-dimensional environments.
Collapse
Affiliation(s)
- Kelly M Mabry
- Department of Chemical and Biological Engineering, University of Colorado at Boulder, Boulder, CO 80309, USA
| | | | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado at Boulder, Boulder, CO 80309, USA; Howard Hughes Medical Institute and the BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO 80309, USA.
| |
Collapse
|
305
|
Subia B, Dey T, Sharma S, Kundu SC. Target specific delivery of anticancer drug in silk fibroin based 3D distribution model of bone-breast cancer cells. ACS APPLIED MATERIALS & INTERFACES 2015; 7:2269-2279. [PMID: 25557227 DOI: 10.1021/am506094c] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
To avoid the indiscriminating action of anticancer drugs, the cancer cell specific targeting of drug molecule becomes a preferred choice for the treatment. The successful screening of the drug molecules in 2D culture system requires further validation. The failure of target specific drug in animal model raises the issue of creating a platform in between the in vitro (2D) and in vivo animal testing. The metastatic breast cancer cells migrate and settle at different sites such as bone tissue. This work evaluates the in vitro 3D model of the breast cancer and bone cells to understand the cellular interactions in the presence of a targeted anticancer drug delivery system. The silk fibroin based cytocompatible 3D scaffold is used as in vitro 3D distribution model. Human breast adenocarcinoma and osteoblast like cells are cocultured to evaluate the efficiency of doxorubicin loaded folic acid conjugated silk fibroin nanoparticle as drug delivery system. Decreasing population of the cancer cells, which lower the levels of vascular endothelial growth factors, glucose consumption, and lactate production are observed in the drug treated coculture constructs. The drug treated constructs do not show any major impact on bone mineralization. The diminished expression of osteogenic markers such as osteocalcein and alkaline phosphatase are recorded. The result indicates that this type of silk based 3D in vitro coculture model may be utilized as a bridge between the traditional 2D and animal model system to evaluate the new drug molecule (s) or to reassay the known drug molecules or to develop target specific drug in cancer research.
Collapse
Affiliation(s)
- Bano Subia
- Department of Biotechnology, Indian Institute of Technology Kharagpur , Kharagpur, West Bengal 721302, India
| | | | | | | |
Collapse
|
306
|
Edmondson R, Broglie JJ, Adcock AF, Yang L. Three-dimensional cell culture systems and their applications in drug discovery and cell-based biosensors. Assay Drug Dev Technol 2015; 12:207-18. [PMID: 24831787 DOI: 10.1089/adt.2014.573] [Citation(s) in RCA: 1603] [Impact Index Per Article: 160.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Three-dimensional (3D) cell culture systems have gained increasing interest in drug discovery and tissue engineering due to their evident advantages in providing more physiologically relevant information and more predictive data for in vivo tests. In this review, we discuss the characteristics of 3D cell culture systems in comparison to the two-dimensional (2D) monolayer culture, focusing on cell growth conditions, cell proliferation, population, and gene and protein expression profiles. The innovations and development in 3D culture systems for drug discovery over the past 5 years are also reviewed in the article, emphasizing the cellular response to different classes of anticancer drugs, focusing particularly on similarities and differences between 3D and 2D models across the field. The progression and advancement in the application of 3D cell cultures in cell-based biosensors is another focal point of this review.
Collapse
Affiliation(s)
- Rasheena Edmondson
- Biomanufacturing Research Institute and Technology Enterprises (BRITE), and Department of Pharmaceutical Sciences, North Carolina Central University , Durham, North Carolina
| | | | | | | |
Collapse
|
307
|
Wang G, Zhao T, Song X, Zhong W, Yu L, Hua W, Xing MMQ, Qiu X. A 3-D multicellular tumor spheroid on ultrathin matrix coated single cancer cells provides a tumor microenvironment model to study epithelial-to-mesenchymal transitions. Polym Chem 2015; 6:283-293. [DOI: 10.1039/c4py01161a] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2024]
Abstract
We report a 3D tumor spheroid model to study epithelial-to-mesenchymal transitions (EMT) using ultra-thin matrix coated single cancer cells.
Collapse
Affiliation(s)
- Guobao Wang
- Department of Anatomy
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering
- Southern Medical University
- Guangzhou 510515
- P.R. China
| | - Tingting Zhao
- Department of Anatomy
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering
- Southern Medical University
- Guangzhou 510515
- P.R. China
| | - Xiaoping Song
- Department of Anatomy
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering
- Southern Medical University
- Guangzhou 510515
- P.R. China
| | - Wen Zhong
- Department of Textile Sciences
- University of Manitoba
- Winnipeg
- Canada
| | - Lei Yu
- Department of Anatomy
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering
- Southern Medical University
- Guangzhou 510515
- P.R. China
| | - Wenxi Hua
- Department of Anatomy
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering
- Southern Medical University
- Guangzhou 510515
- P.R. China
| | - Malcolm M. Q. Xing
- Department of Mechanical and Manitoba Institute of Child Health
- University of Manitoba
- Winnipeg, MB R3T 2N2
- Canada
| | - Xiaozhong Qiu
- Department of Anatomy
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering
- Southern Medical University
- Guangzhou 510515
- P.R. China
| |
Collapse
|
308
|
Li CC, Kharaziha M, Min C, Maas R, Nikkhah M. Microfabrication of Cell-Laden Hydrogels for Engineering Mineralized and Load Bearing Tissues. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 881:15-31. [DOI: 10.1007/978-3-319-22345-2_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
309
|
Abstract
The epidermis and associated appendages of the skin represent a multi-lineage tissue that is maintained by perpetual rounds of renewal. During homeostasis, turnover of epidermal lineages is achieved by input from regionalized keratinocytes stem or progenitor populations with little overlap from neighboring niches. Over the last decade, molecular markers selectively expressed by a number of these stem or progenitor pools have been identified, allowing for the isolation and functional assessment of stem cells and genetic lineage tracing analysis within intact skin. These advancements have led to many fundamental observations about epidermal stem cell function such as the identification of their progeny, their role in maintenance of skin homeostasis, or their contribution to wound healing. In this chapter, we provide a methodology to identify and isolate epidermal stem cells and to assess their functional role in their respective niche. Furthermore, recent evidence has shown that the microenvironment also plays a crucial role in stem cell function. Indeed, epidermal cells are under the influence of surrounding fibroblasts, adipocytes, and sensory neurons that provide extrinsic signals and mechanical cues to the niche and contribute to skin morphogenesis and homeostasis. A better understanding of these microenvironmental cues will help engineer in vitro experimental models with more relevance to in vivo skin biology. New approaches to address and study these environmental cues in vitro will also be addressed.
Collapse
Affiliation(s)
- Yanne S Doucet
- Department of Dermatology, College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | | |
Collapse
|
310
|
Bakmand T, Kwasny D, Dimaki M, Svendsen WE. Fabrication and Characterisation of Membrane-Based Gold Electrodes. ELECTROANAL 2014. [DOI: 10.1002/elan.201400407] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
311
|
Bartlett R, Everett W, Lim S, G N, Loizidou M, Jell G, Tan A, Seifalian AM. Personalized in vitro cancer modeling - fantasy or reality? Transl Oncol 2014; 7:657-64. [PMID: 25500073 PMCID: PMC4311045 DOI: 10.1016/j.tranon.2014.10.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 10/06/2014] [Accepted: 10/13/2014] [Indexed: 01/06/2023] Open
Abstract
With greater technological advancements and understanding of pathophysiology, “personalized medicine” has become a more realistic goal. In the field of cancer, personalized medicine is the ultimate objective, as each cancer is unique and each tumor is heterogeneous. For many decades, researchers have relied upon studying the histopathology of tumors in the hope that it would provide clues to understanding the pathophysiology of cancer. Current preclinical research relies heavily upon two-dimensional culture models. However, these models have had limited success in recreating the complex interactions between cancer cells and the stroma environment in vivo. Thus, there is increasing impetus to shift to three-dimensional models, which more accurately reflect this phenomenon. With a more accurate in vitro tumor model, drug sensitivity can be tested to determine the best treatment option based on the tumor characteristics. Many methods have been developed to create tumor models or “tumoroids,” each with its advantages and limitations. One significant problem faced is the replication of angiogenesis that is characteristic of tumors in vivo. Nonetheless, if three-dimensional models could be standardized and implemented as a preclinical research tool for therapeutic testing, we would be taking a step towards making personalized cancer medicine a reality.
Collapse
Affiliation(s)
- Richard Bartlett
- Centre for Nanotechnology & Regenerative Medicine, Research Department of Nanotechnology, UCL Division of Surgery & Interventional Science, University College London (UCL), London, UK; UCL Medical School, University College London (UCL), London, UK
| | - William Everett
- Centre for Nanotechnology & Regenerative Medicine, Research Department of Nanotechnology, UCL Division of Surgery & Interventional Science, University College London (UCL), London, UK; UCL Medical School, University College London (UCL), London, UK
| | - Santi Lim
- Centre for Nanotechnology & Regenerative Medicine, Research Department of Nanotechnology, UCL Division of Surgery & Interventional Science, University College London (UCL), London, UK; UCL Medical School, University College London (UCL), London, UK
| | - Natasha G
- Centre for Nanotechnology & Regenerative Medicine, Research Department of Nanotechnology, UCL Division of Surgery & Interventional Science, University College London (UCL), London, UK; UCL Medical School, University College London (UCL), London, UK
| | - Marilena Loizidou
- Centre for Nanotechnology & Regenerative Medicine, Research Department of Nanotechnology, UCL Division of Surgery & Interventional Science, University College London (UCL), London, UK
| | - Gavin Jell
- Centre for Nanotechnology & Regenerative Medicine, Research Department of Nanotechnology, UCL Division of Surgery & Interventional Science, University College London (UCL), London, UK
| | - Aaron Tan
- Centre for Nanotechnology & Regenerative Medicine, Research Department of Nanotechnology, UCL Division of Surgery & Interventional Science, University College London (UCL), London, UK; UCL Medical School, University College London (UCL), London, UK; Biomaterials & Advanced Drug Delivery Laboratory (BioADD), Stanford School of Medicine, Stanford University, Stanford, CA, USA
| | - Alexander M Seifalian
- Centre for Nanotechnology & Regenerative Medicine, Research Department of Nanotechnology, UCL Division of Surgery & Interventional Science, University College London (UCL), London, UK; Royal Free London NHS Foundation Trust Hospital, London, UK.
| |
Collapse
|
312
|
Alemany-Ribes M, Semino CE. Bioengineering 3D environments for cancer models. Adv Drug Deliv Rev 2014; 79-80:40-9. [PMID: 24996134 DOI: 10.1016/j.addr.2014.06.004] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 06/16/2014] [Accepted: 06/24/2014] [Indexed: 11/26/2022]
Abstract
Tumor development is a dynamic process where cancer cells differentiate, proliferate and migrate interacting among each other and with the surrounding matrix in a three-dimensional (3D) context. Interestingly, the process follows patterns similar to those involved in early tissue formation by accessing specific genetic programs to grow and disseminate. Thus, the complex biological mechanisms driving tumor progression cannot easily be recreated in the laboratory. Yet, essential tumor stages, including epithelial-mesenchymal transition (EMT), tumor-induced angiogenesis and metastasis, urgently need more realistic models in order to unravel the underlying molecular and cellular mechanisms that govern them. The latest implementation of successful 3D models is having a positive impact on the fight against cancer by obtaining more predictive systems for pre-clinical research, therapeutic drug screening, and early cancer diagnosis. In this review we explore the latest advances and challenges in tumor tissue engineering, by accessing knowledge and tools from cancer biology, material science and bioengineering.
Collapse
|
313
|
Montani C, Steimberg N, Boniotti J, Biasiotto G, Zanella I, Diafera G, Biunno I, Caimi L, Mazzoleni G, Di Lorenzo D. Fibroblasts maintained in 3 dimensions show a better differentiation state and higher sensitivity to estrogens. Toxicol Appl Pharmacol 2014; 280:421-33. [DOI: 10.1016/j.taap.2014.08.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 07/24/2014] [Accepted: 08/12/2014] [Indexed: 01/07/2023]
|
314
|
Boutin ME, Hoffman-Kim D. Application and assessment of optical clearing methods for imaging of tissue-engineered neural stem cell spheres. Tissue Eng Part C Methods 2014; 21:292-302. [PMID: 25128373 DOI: 10.1089/ten.tec.2014.0296] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Three-dimensional (3D) cell culture is an important tool that facilitates biological discoveries by bridging the divide between standard two-dimensional cell culture and the complex, high-cell-density in vivo environment. Typically, the internal structures of 3D tissue-engineered samples are visualized through an involved process of physical sectioning, immunostaining, imaging, and computational reconstruction. However, recent progress in tissue-clearing methods has improved optical-imaging-depth capabilities in whole embryos and brains by reducing tissue opacity and light scattering, thus decreasing the need for physical sectioning. In this study, we assessed the application of the recently published clearing techniques Clear(T2), Scale, and SeeDB to tissue-engineered neural spheres. We found that scaffold-free self-assembled adult hippocampal neural stem cell spheres of 100-μm diameter could be optically cleared and imaged using either Clear(T2) or Scale, while SeeDB only marginally improved imaging depth. The Clear(T2) protocol maintained sphere size, while Scale led to sample expansion, and SeeDB led to sample shrinkage. Additionally, using Clear(T2) we cleared and successfully imaged spheres of C6 glioma cells and spheres of primary cortical neurons. We conclude that Clear(T2) is the most effective protocol of those tested at clearing neural spheres of various cell types and could be applied to better understand neural cell interactions in 3D tissue-engineered samples.
Collapse
Affiliation(s)
- Molly E Boutin
- 1 Center for Biomedical Engineering, Brown University , Providence, Rhode Island
| | | |
Collapse
|
315
|
Guided and magnetic self-assembly of tunable magnetoceptive gels. Nat Commun 2014; 5:4702. [PMID: 25175148 PMCID: PMC4153407 DOI: 10.1038/ncomms5702] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 07/15/2014] [Indexed: 12/19/2022] Open
Abstract
Self-assembly of components into complex functional patterns at microscale is common in nature, and used increasingly in numerous disciplines such as optoelectronics, microfabrication, sensors, tissue engineering and computation. Here, we describe the use of stable radicals to guide the self-assembly of magnetically tunable gels, which we call ‘magnetoceptive’ materials at the scale of hundreds of microns to a millimeter, each can be programmed by shape and composition, into heterogeneous complex structures. Using paramagnetism of free radicals as a driving mechanism, complex heterogeneous structures are built in the magnetic field generated by permanent magnets. The overall magnetic signature of final structure is erased via an antioxidant vitamin E, subsequent to guided self-assembly. We demonstrate unique capabilities of radicals and antioxidants in fabrication of soft systems with heterogeneity in material properties, such as porosity, elastic modulus and mass density; then in bottom-up tissue engineering and finally, levitational and selective assembly of microcomponents. Self-assembly of micrometre-sized building blocks into complex functional patterns provides a useful tool for tissue engineering applications. Here, Tasoglu et al. present a practical method to guide the assembly of magnetically tunable gels in a magnetic field without physical contact.
Collapse
|
316
|
Rauh J, Jacobi A, Stiehler M. Identification of stable reference genes for gene expression analysis of three-dimensional cultivated human bone marrow-derived mesenchymal stromal cells for bone tissue engineering. Tissue Eng Part C Methods 2014; 21:192-206. [PMID: 25000821 DOI: 10.1089/ten.tec.2014.0230] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The principles of tissue engineering (TE) are widely used for bone regeneration concepts. Three-dimensional (3D) cultivation of autologous human mesenchymal stromal cells (MSCs) on porous scaffolds is the basic prerequisite to generate newly formed bone tissue. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) is a specific and sensitive analytical tool for the measurement of mRNA-levels in cells or tissues. For an accurate quantification of gene expression levels, stably expressed reference genes (RGs) are essential to obtain reliable results. Since the 3D environment can affect a cell's morphology, proliferation, and gene expression profile compared with two-dimensional (2D) cultivation, there is a need to identify robust RGs for the quantification of gene expression. So far, this issue has not been adequately investigated. The aim of this study was to identify the most stably expressed RGs for gene expression analysis of 3D-cultivated human bone marrow-derived MSCs (BM-MSCs). For this, we analyzed the gene expression levels of n=31 RGs in 3D-cultivated human BM-MSCs from six different donors compared with conventional 2D cultivation using qRT-PCR. MSCs isolated from bone marrow aspirates were cultivated on human cancellous bone cube scaffolds for 14 days. Osteogenic differentiation was assessed by cell-specific alkaline phosphatase (ALP) activity and expression of osteogenic marker genes. Expression levels of potential reference and target genes were quantified using commercially available TaqMan(®) assays. mRNA expression stability of RGs was determined by calculating the coefficient of variation (CV) and using the algorithms of geNorm and NormFinder. Using both algorithms, we identified TATA box binding protein (TBP), transferrin receptor (p90, CD71) (TFRC), and hypoxanthine phosphoribosyltransferase 1 (HPRT1) as the most stably expressed RGs in 3D-cultivated BM-MSCs. Notably, genes that are routinely used as RGs, for example, beta actin (ACTB) and ribosomal protein L37a (RPL37A), were among the least stable genes. We recommend the combined use of TBP, TFRC, and HPRT1 for the accurate and robust normalization of qRT-PCR data of 3D-cultivated human BM-MSCs.
Collapse
Affiliation(s)
- Juliane Rauh
- University Center for Orthopedics and Trauma Surgery, Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus at Technische Universität Dresden , Dresden, Germany
| | | | | |
Collapse
|
317
|
Higgins CA, Christiano AM. Regenerative medicine and hair loss: how hair follicle culture has advanced our understanding of treatment options for androgenetic alopecia. Regen Med 2014; 9:101-11. [PMID: 24351010 DOI: 10.2217/rme.13.87] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Many of the current drug therapies for androgenetic alopecia were discovered serendipitously, with hair growth observed as an off-target effect when drugs were used to treat a different disorder. Subsequently, several studies using cultured cells have enabled identification of hair growth modulators with similar properties to the currently available drugs, which may also provide clinical benefit. In situations where the current therapeutics do not work, follicular unit transplantation is an alternative surgical option. More recently, the concept of follicular cell implantation, or hair follicle neogenesis, has been attempted, exploiting the inherent properties of cultured hair follicle cells to induce de novo hair growth in balding scalp. In this review, we discuss both the advances in cell culture techniques that have led to a wider range of potential therapeutics to promote hair growth, in addition to detailing current knowledge on follicular cell implantation, and the challenges in making this approach a reality.
Collapse
Affiliation(s)
- Claire A Higgins
- Department of Dermatology, Columbia University, New York, NY, USA
| | | |
Collapse
|
318
|
Brito IR, Lima IMT, Xu M, Shea LD, Woodruff TK, Figueiredo JR. Three-dimensional systems for in vitro follicular culture: overview of alginate-based matrices. Reprod Fertil Dev 2014; 26:915-30. [PMID: 23866836 PMCID: PMC11287383 DOI: 10.1071/rd12401] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 06/12/2013] [Indexed: 08/01/2024] Open
Abstract
The in vitro culture of ovarian follicles has provided critical insight into the biology of the follicle and its enclosed oocyte and the physical interaction and communication between the theca and granulosa cells and the oocyte that is necessary to produce meiotically competent oocytes. Various two-dimensional (2D) and three-dimensional (3D) culture systems have been developed to evaluate the effect of growth factors, hormones, extracellular matrix components and culture conditions on follicle development and oocyte growth and maturation. Among these culture systems, 3D systems make it possible to maintain follicle structure and support communication between the various cell compartments within the follicle. In this review article, we will discuss the three main approaches to ovarian follicle culture: 2D attachment systems, 3D floating systems and 3D encapsulated systems. We will specifically emphasise the development of and advances in alginate-based encapsulated systems for in vitro follicle culture.
Collapse
Affiliation(s)
- Ivina R. Brito
- Faculty of Veterinary, Laboratory of Manipulation of Oocyte and Preantral Follicles (LAMOFOPA), PPGCV, State University of Ceará, Fortaleza, CE 60740-930, Brazil
| | | | - Min Xu
- Division of Reproductive Biology and Clinical Research, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Lonnie D. Shea
- Division of Reproductive Biology and Clinical Research, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Teresa K. Woodruff
- Division of Reproductive Biology and Clinical Research, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - José R. Figueiredo
- Faculty of Veterinary, Laboratory of Manipulation of Oocyte and Preantral Follicles (LAMOFOPA), PPGCV, State University of Ceará, Fortaleza, CE 60740-930, Brazil
| |
Collapse
|
319
|
Yamamoto S, Hotta MM, Okochi M, Honda H. Effect of vascular formed endothelial cell network on the invasive capacity of melanoma using the in vitro 3D co-culture patterning model. PLoS One 2014; 9:e103502. [PMID: 25058006 PMCID: PMC4110033 DOI: 10.1371/journal.pone.0103502] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 07/02/2014] [Indexed: 12/12/2022] Open
Abstract
In vitro three dimensional (3D) cancer models were developed to observe the invasive capacity of melanoma cell spheroids co-cultured with the vascular-formed endothelial cell network. An array-like multicellular pattern of mouse melanoma cell line B16F1 was developed by magnetic cell labeling using a pin-holder device for allocation of magnetic force. When the B16F1 patterned together with a vascular network of human umbilical vein epithelial cells (HUVEC), spreading and progression were observed along the HUVEC network. The B16F1 cells over 80 µm distance from HUVEC remain in a compact spheroid shape, while B16F1 in the proximity of HUVEC aggressively changed their morphology and migrated. The mRNA expression levels of IL-6, MDR-1 and MMP-9 in B16F1 increased along with the distance the HUVEC network, and these expressions were increased by 5, 3 and 2-fold in the B16F1 close to HUVEC (within 80 µm distance) as compared to that far from HUVEC (over 80 µm distance). Our results clearly show that malignancy of tumor cells is enhanced in proximity to vascular endothelial cells and leads to intravasation.
Collapse
Affiliation(s)
- Shuhei Yamamoto
- Department of Biotechnology, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Michael Masakuni Hotta
- Department of Biotechnology, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Mina Okochi
- Department of Biotechnology, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Hiroyuki Honda
- Department of Biotechnology, Graduate School of Engineering, Nagoya University, Nagoya, Japan
- * E-mail:
| |
Collapse
|
320
|
Tourovskaia A, Fauver M, Kramer G, Simonson S, Neumann T. Tissue-engineered microenvironment systems for modeling human vasculature. Exp Biol Med (Maywood) 2014; 239:1264-71. [PMID: 25030480 DOI: 10.1177/1535370214539228] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The high attrition rate of drug candidates late in the development process has led to an increasing demand for test assays that predict clinical outcome better than conventional 2D cell culture systems and animal models. Government agencies, the military, and the pharmaceutical industry have started initiatives for the development of novel in-vitro systems that recapitulate functional units of human tissues and organs. There is growing evidence that 3D cell arrangement, co-culture of different cell types, and physico-chemical cues lead to improved predictive power. A key element of all tissue microenvironments is the vasculature. Beyond transporting blood the microvasculature assumes important organ-specific functions. It is also involved in pathologic conditions, such as inflammation, tumor growth, metastasis, and degenerative diseases. To provide a tool for modeling this important feature of human tissue microenvironments, we developed a microfluidic chip for creating tissue-engineered microenvironment systems (TEMS) composed of tubular cell structures. Our chip design encompasses a small chamber that is filled with an extracellular matrix (ECM) surrounding one or more tubular channels. Endothelial cells (ECs) seeded into the channels adhere to the ECM walls and grow into perfusable tubular tissue structures that are fluidically connected to upstream and downstream fluid channels in the chip. Using these chips we created models of angiogenesis, the blood-brain barrier (BBB), and tumor-cell extravasation. Our angiogenesis model recapitulates true angiogenesis, in which sprouting occurs from a "parent" vessel in response to a gradient of growth factors. Our BBB model is composed of a microvessel generated from brain-specific ECs within an ECM populated with astrocytes and pericytes. Our tumor-cell extravasation model can be utilized to visualize and measure tumor-cell migration through vessel walls into the surrounding matrix. The described technology can be used to create TEMS that recapitulate structural, functional, and physico-chemical elements of vascularized human tissue microenvironments in vitro.
Collapse
|
321
|
Alépée N, Bahinski A, Daneshian M, De Wever B, Fritsche E, Goldberg A, Hansmann J, Hartung T, Haycock J, Hogberg H, Hoelting L, Kelm JM, Kadereit S, McVey E, Landsiedel R, Leist M, Lübberstedt M, Noor F, Pellevoisin C, Petersohn D, Pfannenbecker U, Reisinger K, Ramirez T, Rothen-Rutishauser B, Schäfer-Korting M, Zeilinger K, Zurich MG. State-of-the-art of 3D cultures (organs-on-a-chip) in safety testing and pathophysiology. ALTEX-ALTERNATIVES TO ANIMAL EXPERIMENTATION 2014. [PMID: 25027500 DOI: 10.14573/altex1406111] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Integrated approaches using different in vitro methods in combination with bioinformatics can (i) increase the success rate and speed of drug development; (ii) improve the accuracy of toxicological risk assessment; and (iii) increase our understanding of disease. Three-dimensional (3D) cell culture models are important building blocks of this strategy which has emerged during the last years. The majority of these models are organotypic, i.e., they aim to reproduce major functions of an organ or organ system. This implies in many cases that more than one cell type forms the 3D structure, and often matrix elements play an important role. This review summarizes the state of the art concerning commonalities of the different models. For instance, the theory of mass transport/metabolite exchange in 3D systems and the special analytical requirements for test endpoints in organotypic cultures are discussed in detail. In the next part, 3D model systems for selected organs--liver, lung, skin, brain--are presented and characterized in dedicated chapters. Also, 3D approaches to the modeling of tumors are presented and discussed. All chapters give a historical background, illustrate the large variety of approaches, and highlight up- and downsides as well as specific requirements. Moreover, they refer to the application in disease modeling, drug discovery and safety assessment. Finally, consensus recommendations indicate a roadmap for the successful implementation of 3D models in routine screening. It is expected that the use of such models will accelerate progress by reducing error rates and wrong predictions from compound testing.
Collapse
|
322
|
Faghihi F, Papadimitropoulos A, Martin I, Eslaminejad MB. Effect of Purmorphamine on Osteogenic Differentiation of Human Mesenchymal Stem Cells in a Three-Dimensional Dynamic Culture System. Cell Mol Bioeng 2014. [DOI: 10.1007/s12195-014-0343-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
323
|
Forbes B, O'Lone R, Allen PP, Cahn A, Clarke C, Collinge M, Dailey LA, Donnelly LE, Dybowski J, Hassall D, Hildebrand D, Jones R, Kilgour J, Klapwijk J, Maier CC, McGovern T, Nikula K, Parry JD, Reed MD, Robinson I, Tomlinson L, Wolfreys A. Challenges for inhaled drug discovery and development: Induced alveolar macrophage responses. Adv Drug Deliv Rev 2014; 71:15-33. [PMID: 24530633 DOI: 10.1016/j.addr.2014.02.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 02/01/2014] [Accepted: 02/03/2014] [Indexed: 12/27/2022]
Abstract
Alveolar macrophage (AM) responses are commonly induced in inhalation toxicology studies, typically being observed as an increase in number or a vacuolated 'foamy' morphology. Discriminating between adaptive AM responses and adverse events during nonclinical and clinical development is a major scientific challenge. When measuring and interpreting induced AM responses, an understanding of macrophage biology is essential; this includes 'sub-types' of AMs with different roles in health and disease and mechanisms of induction/resolution of AM responses to inhalation of pharmaceutical aerosols. In this context, emerging assay techniques, the utility of toxicokinetics and the requirement for new biomarkers are considered. Risk assessment for nonclinical toxicology findings and their translation to effects in humans is discussed from a scientific and regulatory perspective. At present, when apparently adaptive macrophage-only responses to inhaled investigational products are observed in nonclinical studies, this poses a challenge for risk assessment and an improved understanding of induced AM responses to inhaled pharmaceuticals is required.
Collapse
|
324
|
Dunne LW, Huang Z, Meng W, Fan X, Zhang N, Zhang Q, An Z. Human decellularized adipose tissue scaffold as a model for breast cancer cell growth and drug treatments. Biomaterials 2014; 35:4940-9. [PMID: 24661550 DOI: 10.1016/j.biomaterials.2014.03.003] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 03/03/2014] [Indexed: 12/23/2022]
Abstract
Human adipose tissue extracellular matrix, derived through decellularization processing, has been shown to provide a biomimetic microenvironment for adipose tissue regeneration. This study reports the use of human adipose tissue-derived extracellular matrix (hDAM) scaffolds as a three-dimensional cell culturing system for the investigation of breast cancer growth and drug treatments. The hDAM scaffolds have similar extracellular matrix composition to the microenvironment of breast tissues. Breast cancer cells were cultured in hDAM scaffolds, and cell proliferation, migration, morphology, and drug responses were investigated. The growth profiles of multiple breast cancer cell lines cultured in hDAM scaffolds differed from the growth of those cultured on two-dimensional surfaces and more closely resembled the growth of xenografts. hDAM-cultured breast cancer cells also differed from those cultured on two-dimensional surfaces in terms of cell morphology, migration, expression of adhesion molecules, and sensitivity to drug treatment. Our results demonstrated that the hDAM system provides breast cancer cells with a biomimetic microenvironment in vitro that more closely mimics the in vivo microenvironment than existing two-dimensional and Matrigel three-dimensional cultures do, and thus can provide vital information for the characterization of cancer cells and screening of cancer therapeutics.
Collapse
Affiliation(s)
- Lina W Dunne
- Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Zhao Huang
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, TX 77030, USA
| | - Weixu Meng
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, TX 77030, USA
| | - Xuejun Fan
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, TX 77030, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, TX 77030, USA
| | - Qixu Zhang
- Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA.
| | - Zhiqiang An
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, TX 77030, USA.
| |
Collapse
|
325
|
Gurkan UA, El Assal R, Yildiz SE, Sung Y, Trachtenberg AJ, Kuo WP, Demirci U. Engineering anisotropic biomimetic fibrocartilage microenvironment by bioprinting mesenchymal stem cells in nanoliter gel droplets. Mol Pharm 2014; 11:2151-9. [PMID: 24495169 PMCID: PMC4096228 DOI: 10.1021/mp400573g] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Over the past decade, bioprinting has emerged as a promising patterning strategy to organize cells and extracellular components both in two and three dimensions (2D and 3D) to engineer functional tissue mimicking constructs. So far, tissue printing has neither been used for 3D patterning of mesenchymal stem cells (MSCs) in multiphase growth factor embedded 3D hydrogels nor been investigated phenotypically in terms of simultaneous differentiation into different cell types within the same micropatterned 3D tissue constructs. Accordingly, we demonstrated a biochemical gradient by bioprinting nanoliter droplets encapsulating human MSCs, bone morphogenetic protein 2 (BMP-2), and transforming growth factor β1 (TGF- β1), engineering an anisotropic biomimetic fibrocartilage microenvironment. Assessment of the model tissue construct displayed multiphasic anisotropy of the incorporated biochemical factors after patterning. Quantitative real time polymerase chain reaction (qRT-PCR) results suggested genomic expression patterns leading to simultaneous differentiation of MSC populations into osteogenic and chondrogenic phenotype within the multiphasic construct, evidenced by upregulation of osteogenesis and condrogenesis related genes during in vitro culture. Comprehensive phenotypic network and pathway analysis results, which were based on genomic expression data, indicated activation of differentiation related mechanisms, via signaling pathways, including TGF, BMP, and vascular endothelial growth factor.
Collapse
Affiliation(s)
- Umut A Gurkan
- Case Biomanufacturing and Microfabrication Laboratory, Mechanical and Aerospace Engineering Department, Department of Orthopaedics, Case Western Reserve University , Cleveland, Ohio 44106, United States
| | | | | | | | | | | | | |
Collapse
|
326
|
Bloom AB, Zaman MH. Influence of the microenvironment on cell fate determination and migration. Physiol Genomics 2014; 46:309-14. [PMID: 24619520 DOI: 10.1152/physiolgenomics.00170.2013] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Several critical cell functions are influenced not only by internal cellular machinery but also by external mechanical and biochemical cues from the surrounding microenvironment. Slight changes to the microenvironment can result in dramatic changes to the cell's phenotype; for example, a change in the nutrients or pH of a tumor microenvironment can result in increased tumor metastasis. While cellular fate and the regulators of cell fate have been studied in detail for several decades now, our understanding of the extracellular regulators remains qualitative and far from comprehensive. In this review, we discuss the microenvironment influence on cell fate in terms of adhesion, migration, and differentiation and focus on both developments in experimental and computation tools to analyze cellular fate.
Collapse
Affiliation(s)
- Alexander B Bloom
- Department of Molecular Biology, Cell Biology, and Biochemistry, Boston University, Boston, Massachusetts; and
| | | |
Collapse
|
327
|
Stanton MM, Rankenberg JM, Park BW, McGimpsey WG, Malcuit C, Lambert CR. Cell Behavior on Surface Modified Polydimethylsiloxane (PDMS). Macromol Biosci 2014; 14:953-64. [DOI: 10.1002/mabi.201300504] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 01/17/2014] [Indexed: 12/23/2022]
Affiliation(s)
- Morgan M. Stanton
- Worcester Polytechnic Institute; Department of Chemistry and Biochemistry; 100 Institute Road Worcester MA 01609 USA
| | - Johanna M. Rankenberg
- Kent State University; Department of Biological Sciences; 256 Cunningham Hall Kent OH 44242 USA
| | - Byung-Wook Park
- Kent State University; Department of Chemistry and Biochemistry; 214 Williams Hall Kent OH 44242 USA
| | - W. Grant McGimpsey
- Kent State University; Department of Chemistry and Biochemistry; 214 Williams Hall Kent OH 44242 USA
| | - Christopher Malcuit
- Kent State University; Department of Biological Sciences; 256 Cunningham Hall Kent OH 44242 USA
| | - Christopher R. Lambert
- Worcester Polytechnic Institute; Department of Chemistry and Biochemistry; 100 Institute Road Worcester MA 01609 USA
| |
Collapse
|
328
|
Pan D, Xue G, Zhu J, Hu B. Ionizing radiation induced biological effects in three-dimensional cell cultures. RENDICONTI LINCEI 2014. [DOI: 10.1007/s12210-013-0260-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
329
|
Jeremias TDS, Machado RG, Visoni SBC, Pereima MJ, Leonardi DF, Trentin AG. Dermal substitutes support the growth of human skin-derived mesenchymal stromal cells: potential tool for skin regeneration. PLoS One 2014; 9:e89542. [PMID: 24586857 PMCID: PMC3935879 DOI: 10.1371/journal.pone.0089542] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 01/22/2014] [Indexed: 12/25/2022] Open
Abstract
New strategies for skin regeneration are needed in order to provide effective treatment for cutaneous wounds and disease. Mesenchymal stem cells (MSCs) are an attractive source of cells for tissue engineering because of their prolonged self-renewal capacity, multipotentiality, and ability to release active molecules important for tissue repair. In this paper, we show that human skin-derived mesenchymal stromal cells (SD-MSCs) display similar characteristics to the multipotent MSCs. We also evaluate their growth in a three-dimensional (3D) culture system with dermal substitutes (Integra and Pelnac). When cultured in monolayers, SD-MSCs expressed mesenchymal markers, such as CD105, Fibronectin, and α-SMA; and neural markers, such as Nestin and βIII-Tubulin; at transcriptional and/or protein level. Integra and Pelnac equally supported the adhesion, spread and growth of human SD-MSCs in 3D culture, maintaining the MSC characteristics and the expression of multilineage markers. Therefore, dermal substitutes support the growth of mesenchymal stromal cells from human skin, promising an effective tool for tissue engineering and regenerative technology.
Collapse
Affiliation(s)
- Talita da Silva Jeremias
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brasil
| | - Rafaela Grecco Machado
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brasil
| | - Silvia Beatriz Coutinho Visoni
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brasil
| | - Maurício José Pereima
- Departamento de Pediatria, Centro de Ciências da Saúde, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brasil
- Hospital Infantil Joana de Gusmão, Florianópolis, Santa Catarina, Brasil
| | - Dilmar Francisco Leonardi
- Hospital Governador Celso Ramos, Florianópolis, Santa Catarina, Brasil
- Departamento de Cirurgia, Universidade do Sul de Santa Catarina, Florianópolis, Santa Catarina, Brasil
| | - Andrea Gonçalves Trentin
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brasil
- * E-mail:
| |
Collapse
|
330
|
Alépée N, Bahinski A, Daneshian M, De Wever B, Fritsche E, Goldberg A, Hansmann J, Hartung T, Haycock J, Hogberg HT, Hoelting L, Kelm JM, Kadereit S, McVey E, Landsiedel R, Leist M, Lübberstedt M, Noor F, Pellevoisin C, Petersohn D, Pfannenbecker U, Reisinger K, Ramirez T, Rothen-Rutishauser B, Schäfer-Korting M, Zeilinger K, Zurich MG. State-of-the-art of 3D cultures (organs-on-a-chip) in safety testing and pathophysiology. ALTEX 2014; 31:441-77. [PMID: 25027500 PMCID: PMC4783151 DOI: 10.14573/altex.1406111] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 06/30/2014] [Indexed: 02/02/2023]
Abstract
Integrated approaches using different in vitro methods in combination with bioinformatics can (i) increase the success rate and speed of drug development; (ii) improve the accuracy of toxicological risk assessment; and (iii) increase our understanding of disease. Three-dimensional (3D) cell culture models are important building blocks of this strategy which has emerged during the last years. The majority of these models are organotypic, i.e., they aim to reproduce major functions of an organ or organ system. This implies in many cases that more than one cell type forms the 3D structure, and often matrix elements play an important role. This review summarizes the state of the art concerning commonalities of the different models. For instance, the theory of mass transport/metabolite exchange in 3D systems and the special analytical requirements for test endpoints in organotypic cultures are discussed in detail. In the next part, 3D model systems for selected organs--liver, lung, skin, brain--are presented and characterized in dedicated chapters. Also, 3D approaches to the modeling of tumors are presented and discussed. All chapters give a historical background, illustrate the large variety of approaches, and highlight up- and downsides as well as specific requirements. Moreover, they refer to the application in disease modeling, drug discovery and safety assessment. Finally, consensus recommendations indicate a roadmap for the successful implementation of 3D models in routine screening. It is expected that the use of such models will accelerate progress by reducing error rates and wrong predictions from compound testing.
Collapse
Affiliation(s)
| | - Anthony Bahinski
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, USA
| | - Mardas Daneshian
- Center for Alternatives to Animal Testing – Europe (CAAT-Europe), University of Konstanz, Konstanz, Germany
| | | | - Ellen Fritsche
- Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Alan Goldberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, USA
| | - Jan Hansmann
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Thomas Hartung
- Center for Alternatives to Animal Testing – Europe (CAAT-Europe), University of Konstanz, Konstanz, Germany,Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, USA
| | - John Haycock
- Department of Materials Science of Engineering, University of Sheffield, Sheffield, UK
| | - Helena T. Hogberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, USA
| | - Lisa Hoelting
- Doerenkamp-Zbinden Chair of in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | | | - Suzanne Kadereit
- Doerenkamp-Zbinden Chair of in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Emily McVey
- Board for the Authorization of Plant Protection Products and Biocides, Wageningen, The Netherlands
| | | | - Marcel Leist
- Center for Alternatives to Animal Testing – Europe (CAAT-Europe), University of Konstanz, Konstanz, Germany,Doerenkamp-Zbinden Chair of in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Marc Lübberstedt
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Campus Virchow-Klinikum, Berlin, Germany
| | - Fozia Noor
- Biochemical Engineering, Saarland University, Saarbruecken, Germany
| | | | | | | | | | - Tzutzuy Ramirez
- BASF SE, Experimental Toxicology and Ecology, Ludwigshafen, Germany
| | | | - Monika Schäfer-Korting
- Institute for Pharmacy (Pharmacology and Toxicology), Freie Universität Berlin, Berlin, Germany
| | - Katrin Zeilinger
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Campus Virchow-Klinikum, Berlin, Germany
| | - Marie-Gabriele Zurich
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland,Swiss Center for Applied Human Toxicology (SCAHT), University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
331
|
Guo Z, Higgins CA, Gillette BM, Itoh M, Umegaki N, Gledhill K, Sia SK, Christiano AM. Building a microphysiological skin model from induced pluripotent stem cells. Stem Cell Res Ther 2013; 4 Suppl 1:S2. [PMID: 24564920 PMCID: PMC4029476 DOI: 10.1186/scrt363] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The discovery of induced pluripotent stem cells (iPSCs) in 2006 was a major breakthrough for regenerative medicine. The establishment of patient-specific iPSCs has created the opportunity to model diseases in culture systems, with the potential to rapidly advance the drug discovery field. Current methods of drug discovery are inefficient, with a high proportion of drug candidates failing during clinical trials due to low efficacy and/or high toxicity. Many drugs fail toxicity testing during clinical trials, since the cells on which they have been tested do not adequately model three-dimensional tissues or their interaction with other organs in the body. There is a need to develop microphysiological systems that reliably represent both an intact tissue and also the interaction of a particular tissue with other systems throughout the body. As the port of entry for many drugs is via topical delivery, the skin is the first line of exposure, and also one of the first organs to demonstrate a reaction after systemic drug delivery. In this review, we discuss our strategy to develop a microphysiological system using iPSCs that recapitulates human skin for analyzing the interactions of drugs with the skin.
Collapse
|
332
|
Charoen KM, Fallica B, Colson YL, Zaman MH, Grinstaff MW. Embedded multicellular spheroids as a biomimetic 3D cancer model for evaluating drug and drug-device combinations. Biomaterials 2013; 35:2264-71. [PMID: 24360576 DOI: 10.1016/j.biomaterials.2013.11.038] [Citation(s) in RCA: 146] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 11/14/2013] [Indexed: 01/17/2023]
Abstract
Multicellular aggregates of cells, termed spheroids, are of interest for studying tumor behavior and for evaluating the response of pharmacologically active agents. Spheroids more faithfully reproduce the tumor macrostructure found in vivo compared to classical 2D monolayers. We present a method for embedding spheroids within collagen gels followed by quantitative and qualitative whole spheroid and single cell analyses enabling characterization over the length scales from molecular to macroscopic. Spheroid producing and embedding capabilities are demonstrated for U2OS and MDA-MB-231 cell lines, of osteosarcoma and breast adenocarcinoma origin, respectively. Finally, using the MDA-MB-231 tumor model, the chemotherapeutic response between paclitaxel delivery as a bolus dose, as practiced in the clinic, is compared to delivery within an expansile nanoparticle. The expansile nanoparticle delivery route provides a superior outcome and the results mirror those observed in a murine xenograft model. These findings highlight the synergistic beneficial results that may arise from the use of a drug delivery system, and the need to evaluate both drug candidates and delivery systems in the research and preclinical screening phases of a new cancer therapy development program.
Collapse
Affiliation(s)
- Kristie M Charoen
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Brian Fallica
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Yolonda L Colson
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA 02215, USA
| | - Muhammad H Zaman
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| | - Mark W Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Department of Chemistry, Boston University, Boston, MA 02215, USA.
| |
Collapse
|
333
|
Alassaf A, Aleid A, Frenkel V. In vitro methods for evaluating therapeutic ultrasound exposures: present-day models and future innovations. J Ther Ultrasound 2013; 1:21. [PMID: 25093079 PMCID: PMC4109267 DOI: 10.1186/2050-5736-1-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 09/09/2013] [Indexed: 11/30/2022] Open
Abstract
Although preclinical experiments are ultimately required to evaluate new therapeutic ultrasound exposures and devices prior to clinical trials, in vitro experiments can play an important role in the developmental process. A variety of in vitro methods have been developed, where each of these has demonstrated their utility for various test purposes. These include inert tissue-mimicking phantoms, which can incorporate thermocouples or cells and ex vivo tissue. Cell-based methods have also been used, both in monolayer and suspension. More biologically relevant platforms have also shown utility, such as blood clots and collagen gels. Each of these methods possesses characteristics that are well suited for various well-defined investigative goals. None, however, incorporate all the properties of real tissues, which include a 3D environment and live cells that may be maintained long-term post-treatment. This review is intended to provide an overview of the existing application-specific in vitro methods available to therapeutic ultrasound investigators, highlighting their advantages and limitations. Additional reporting is presented on the exciting and emerging field of 3D biological scaffolds, employing methods and materials adapted from tissue engineering. This type of platform holds much promise for achieving more representative conditions of those found in vivo, especially important for the newest sphere of therapeutic applications, based on molecular changes that may be generated in response to non-destructive exposures.
Collapse
Affiliation(s)
- Ahmad Alassaf
- Department of Biomedical Engineering, Catholic University of America, 620 Michigan Ave NE, Washington, DC 20064, USA
| | - Adham Aleid
- Department of Biomedical Engineering, Catholic University of America, 620 Michigan Ave NE, Washington, DC 20064, USA
| | - Victor Frenkel
- Department of Biomedical Engineering, Catholic University of America, 620 Michigan Ave NE, Washington, DC 20064, USA
| |
Collapse
|
334
|
Moad M, Pal D, Hepburn AC, Williamson SC, Wilson L, Lako M, Armstrong L, Hayward SW, Franco OE, Cates JM, Fordham SE, Przyborski S, Carr-Wilkinson J, Robson CN, Heer R. A novel model of urinary tract differentiation, tissue regeneration, and disease: reprogramming human prostate and bladder cells into induced pluripotent stem cells. Eur Urol 2013; 64:753-61. [PMID: 23582880 PMCID: PMC3819995 DOI: 10.1016/j.eururo.2013.03.054] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 03/25/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND Primary culture and animal and cell-line models of prostate and bladder development have limitations in describing human biology, and novel strategies that describe the full spectrum of differentiation from foetal through to ageing tissue are required. Recent advances in biology demonstrate that direct reprogramming of somatic cells into pluripotent embryonic stem cell (ESC)-like cells is possible. These cells, termed induced pluripotent stem cells (iPSCs), could theoretically generate adult prostate and bladder tissue, providing an alternative strategy to study differentiation. OBJECTIVE To generate human iPSCs derived from normal, ageing, human prostate (Pro-iPSC), and urinary tract (UT-iPSC) tissue and to assess their capacity for lineage-directed differentiation. DESIGN, SETTING, AND PARTICIPANTS Prostate and urinary tract stroma were transduced with POU class 5 homeobox 1 (POU5F1; formerly OCT4), SRY (sex determining region Y)-box 2 (SOX2), Kruppel-like factor 4 (gut) (KLF4), and v-myc myelocytomatosis viral oncogene homolog (avian) (MYC, formerly C-MYC) genes to generate iPSCs. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The potential for differentiation into prostate and bladder lineages was compared with classical skin-derived iPSCs. The student t test was used. RESULTS AND LIMITATIONS Successful reprogramming of prostate tissue into Pro-iPSCs and bladder and ureter into UT-iPSCs was demonstrated by characteristic ESC morphology, marker expression, and functional pluripotency in generating all three germ-layer lineages. In contrast to conventional skin-derived iPSCs, Pro-iPSCs showed a vastly increased ability to generate prostate epithelial-specific differentiation, as characterised by androgen receptor and prostate-specific antigen induction. Similarly, UT-iPSCs were shown to be more efficient than skin-derived iPSCs in undergoing bladder differentiation as demonstrated by expression of urothelial-specific markers: uroplakins, claudins, and cytokeratin; and stromal smooth muscle markers: α-smooth-muscle actin, calponin, and desmin. These disparities are likely to represent epigenetic differences between individual iPSC lines and highlight the importance of organ-specific iPSCs for tissue-specific studies. CONCLUSIONS IPSCs provide an exciting new model to characterise mechanisms regulating prostate and bladder differentiation and to develop novel approaches to disease modelling. Regeneration of bladder cells also provides an exceptional opportunity for translational tissue engineering.
Collapse
Affiliation(s)
- Mohammad Moad
- Northern Institute for Cancer Research, Newcastle University, UK
| | - Deepali Pal
- Northern Institute for Cancer Research, Newcastle University, UK
| | | | | | - Laura Wilson
- Northern Institute for Cancer Research, Newcastle University, UK
| | - Majlinda Lako
- Institute of Genetic Medicine, Newcastle University, UK
| | | | - Simon W. Hayward
- Department of Urological Surgery, Vanderbilt University Medical Centre, TN, USA
| | - Omar E. Franco
- Department of Urological Surgery, Vanderbilt University Medical Centre, TN, USA
| | - Justin M. Cates
- Department of Urological Surgery, Vanderbilt University Medical Centre, TN, USA
| | - Sarah E. Fordham
- Northern Institute for Cancer Research, Newcastle University, UK
| | | | | | - Craig N. Robson
- Northern Institute for Cancer Research, Newcastle University, UK
| | - Rakesh Heer
- Northern Institute for Cancer Research, Newcastle University, UK
| |
Collapse
|
335
|
Li Z, Cui Z. Three-dimensional perfused cell culture. Biotechnol Adv 2013; 32:243-54. [PMID: 24184152 DOI: 10.1016/j.biotechadv.2013.10.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 02/14/2013] [Accepted: 10/07/2013] [Indexed: 12/17/2022]
Abstract
Compelling evidence suggests the limitation and shortcomings of the current and well established cell culture method using multi-well plates, flasks and Petri dishes. These are particularly important when cell functions are sensitive to the local microenvironment, cell-cell and cell-extracellular matrix interactions. There is a clear need for advanced cell culture systems which mimic in vivo and more physiological conditions. This review summarises and analyses recent progress in three dimensional (3D) cell culture with perfusion as the next generation cell culture tools, while excluding engineered tissue culture where three dimensional scaffold has to be used for structural support and perfusion for overcoming mass transfer control. Apart from research activities in academic community, product development in industry is also included in this review.
Collapse
Affiliation(s)
- Zhaohui Li
- Institute of Biomedical Engineering, Department of Engineering Science, Oxford University, Oxford, UK
| | - Zhanfeng Cui
- Institute of Biomedical Engineering, Department of Engineering Science, Oxford University, Oxford, UK.
| |
Collapse
|
336
|
Microenvironmental reprogramming by three-dimensional culture enables dermal papilla cells to induce de novo human hair-follicle growth. Proc Natl Acad Sci U S A 2013; 110:19679-88. [PMID: 24145441 DOI: 10.1073/pnas.1309970110] [Citation(s) in RCA: 272] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
De novo organ regeneration has been observed in several lower organisms, as well as rodents; however, demonstrating these regenerative properties in human cells and tissues has been challenging. In the hair follicle, rodent hair follicle-derived dermal cells can interact with local epithelia and induce de novo hair follicles in a variety of hairless recipient skin sites. However, multiple attempts to recapitulate this process in humans using human dermal papilla cells in human skin have failed, suggesting that human dermal papilla cells lose key inductive properties upon culture. Here, we performed global gene expression analysis of human dermal papilla cells in culture and discovered very rapid and profound molecular signature changes linking their transition from a 3D to a 2D environment with early loss of their hair-inducing capacity. We demonstrate that the intact dermal papilla transcriptional signature can be partially restored by growth of papilla cells in 3D spheroid cultures. This signature change translates to a partial restoration of inductive capability, and we show that human dermal papilla cells, when grown as spheroids, are capable of inducing de novo hair follicles in human skin.
Collapse
|
337
|
Prasad A, Kumar SS, Dessimoz C, Bleuler S, Laule O, Hruz T, Gruissem W, Zimmermann P. Global regulatory architecture of human, mouse and rat tissue transcriptomes. BMC Genomics 2013; 14:716. [PMID: 24138449 PMCID: PMC4008137 DOI: 10.1186/1471-2164-14-716] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 09/25/2013] [Indexed: 01/30/2023] Open
Abstract
Background Predicting molecular responses in human by extrapolating results from model organisms requires a precise understanding of the architecture and regulation of biological mechanisms across species. Results Here, we present a large-scale comparative analysis of organ and tissue transcriptomes involving the three mammalian species human, mouse and rat. To this end, we created a unique, highly standardized compendium of tissue expression. Representative tissue specific datasets were aggregated from more than 33,900 Affymetrix expression microarrays. For each organism, we created two expression datasets covering over 55 distinct tissue types with curated data from two independent microarray platforms. Principal component analysis (PCA) revealed that the tissue-specific architecture of transcriptomes is highly conserved between human, mouse and rat. Moreover, tissues with related biological function clustered tightly together, even if the underlying data originated from different labs and experimental settings. Overall, the expression variance caused by tissue type was approximately 10 times higher than the variance caused by perturbations or diseases, except for a subset of cancers and chemicals. Pairs of gene orthologs exhibited higher expression correlation between mouse and rat than with human. Finally, we show evidence that tissue expression profiles, if combined with sequence similarity, can improve the correct assignment of functionally related homologs across species. Conclusion The results demonstrate that tissue-specific regulation is the main determinant of transcriptome composition and is highly conserved across mammalian species.
Collapse
|
338
|
Whitney J, DeWitt M, Whited BM, Carswell W, Simon A, Rylander CG, Rylander MN. 3D viability imaging of tumor phantoms treated with single-walled carbon nanohorns and photothermal therapy. NANOTECHNOLOGY 2013; 24:275102. [PMID: 23780336 PMCID: PMC3786715 DOI: 10.1088/0957-4484/24/27/275102] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
A new image analysis method called the spatial phantom evaluation of cellular thermal response in layers (SPECTRL) is presented for assessing spatial viability response to nanoparticle enhanced photothermal therapy in tissue representative phantoms. Sodium alginate phantoms seeded with MDA-MB-231 breast cancer cells and single-walled nanohorns were laser irradiated with an ytterbium fiber laser at a wavelength of 1064 nm and irradiance of 3.8 W cm(-2) for 10-80 s. SPECTRL quantitatively assessed and correlated 3D viability with spatiotemporal temperature. Based on this analysis, kill and transition zones increased from 3.7 mm(3) and 13 mm(3) respectively to 44.5 mm(3) and 44.3 mm(3) as duration was increased from 10 to 80 s. SPECTRL provides a quantitative tool for measuring precise spatial treatment regions, providing information necessary to tailor therapy protocols.
Collapse
Affiliation(s)
- Jon Whitney
- Department of Mechanical Engineering, Virgina Tech., Blacksburg, VA 24061, USA
| | | | | | | | | | | | | |
Collapse
|
339
|
Burgstaller G, Oehrle B, Koch I, Lindner M, Eickelberg O. Multiplex profiling of cellular invasion in 3D cell culture models. PLoS One 2013; 8:e63121. [PMID: 23671660 PMCID: PMC3650046 DOI: 10.1371/journal.pone.0063121] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 03/28/2013] [Indexed: 12/20/2022] Open
Abstract
To-date, most invasion or migration assays use a modified Boyden chamber-like design to assess migration as single-cell or scratch assays on coated or uncoated planar plastic surfaces. Here, we describe a 96-well microplate-based, high-content, three-dimensional cell culture assay capable of assessing invasion dynamics and molecular signatures thereof. On applying our invasion assay, we were able to demonstrate significant effects on the invasion capacity of fibroblast cell lines, as well as primary lung fibroblasts. Administration of epidermal growth factor resulted in a substantial increase of cellular invasion, thus making this technique suitable for high-throughput pharmacological screening of novel compounds regulating invasive and migratory pathways of primary cells. Our assay also correlates cellular invasiveness to molecular events. Thus, we argue of having developed a powerful and versatile toolbox for an extensive profiling of invasive cells in a 96-well format. This will have a major impact on research in disease areas like fibrosis, metastatic cancers, or chronic inflammatory states.
Collapse
Affiliation(s)
- Gerald Burgstaller
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany
| | - Bettina Oehrle
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany
| | - Ina Koch
- Center for Thoracic Surgery, Asklepios Biobank for Lung Diseases, Comprehensive Pneumology Center, Asklepios Clinic Munich-Gauting, Munich, Germany
| | - Michael Lindner
- Center for Thoracic Surgery, Asklepios Biobank for Lung Diseases, Comprehensive Pneumology Center, Asklepios Clinic Munich-Gauting, Munich, Germany
| | - Oliver Eickelberg
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany
| |
Collapse
|
340
|
Craniosynostosis-associated Fgfr2(C342Y) mutant bone marrow stromal cells exhibit cell autonomous abnormalities in osteoblast differentiation and bone formation. BIOMED RESEARCH INTERNATIONAL 2013; 2013:292506. [PMID: 23762837 PMCID: PMC3665166 DOI: 10.1155/2013/292506] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 03/18/2013] [Accepted: 03/29/2013] [Indexed: 11/22/2022]
Abstract
We recently reported that cranial bones of Fgfr2C342Y/+ craniosynostotic mice are diminished in density when compared to those of wild type mice, and that cranial bone cells isolated from the mutant mice exhibit inhibited late stage osteoblast differentiation. To provide further support for the idea that craniosynostosis-associated Fgfr mutations lead to cell autonomous defects in osteoblast differentiation and mineralized tissue formation, here we tested bone marrow stromal cells isolated from Fgfr2C342Y/+ mice for their ability to differentiate into osteoblasts. Additionally, to determine if the low bone mass phenotype of Crouzon syndrome includes the appendicular skeleton, long bones were assessed by micro CT. Fgfr2C342Y/+ cells showed increased osteoblastic gene expression during early osteoblastic differentiation but decreased expression of alkaline phosphatase mRNA and enzyme activity, and decreased mineralization during later stages of differentiation, when cultured under 2D in vitro conditions. Cells isolated from Fgfr2C342Y/+ mice also formed less bone when allowed to differentiate in a 3D matrix in vivo. Cortical bone parameters were diminished in long bones of Fgfr2C342Y/+ mice. These results demonstrate that marrow stromal cells of Fgfr2C342Y/+ mice have an autonomous defect in osteoblast differentiation and bone mineralization, and that the Fgfr2C342Y mutation influences both the axial and appendicular skeletons.
Collapse
|
341
|
Qiu P, Qu X, Brackett DJ, Lerner MR, Li D, Mao C. Silica-based branched hollow microfibers as a biomimetic extracellular matrix for promoting tumor cell growth in vitro and in vivo. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2013; 25:2492-6. [PMID: 23450784 PMCID: PMC3731149 DOI: 10.1002/adma.201204472] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 11/25/2012] [Indexed: 05/27/2023]
Abstract
A novel scaffold composed of loosely branched hollow silica microfibers that has been proven to be highly biocompatible is proposed for the 3D culture of cancer cells. The MCF-7 cancer cells can grow and proliferate freely inside the scaffold in the form of multicellular spheroids. MCF-7 cancer cells cultured on the current 3D silica scaffold retained significantly more oncological characters than those cultured on the conventional 2D substrate and can serve as in vitro tumor model for studying cancer treatment.
Collapse
Affiliation(s)
- Penghe Qiu
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, OK, 73019, USA
| | - Xuewei Qu
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, OK, 73019, USA
| | - Daniel J. Brackett
- Health Science Center, University of Oklahoma and Veterans Research and Education Foundation, Oklahoma City, OK 73104, USA
| | - Megan R. Lerner
- Health Science Center, University of Oklahoma and Veterans Research and Education Foundation, Oklahoma City, OK 73104, USA
| | - Dong Li
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, OK, 73019, USA
| | - Chuanbin Mao
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, OK, 73019, USA
| |
Collapse
|
342
|
Qutachi O, Shakesheff KM, Buttery LD. Delivery of definable number of drug or growth factor loaded poly(dl-lactic acid-co-glycolic acid) microparticles within human embryonic stem cell derived aggregates. J Control Release 2013; 168:18-27. [DOI: 10.1016/j.jconrel.2013.02.029] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 02/14/2013] [Accepted: 02/24/2013] [Indexed: 10/27/2022]
|
343
|
Bartel C, Tichy A, Schoenkypl S, Aurich C, Walter I. Effects of steroid hormones on differentiated glandular epithelial and stromal cells in a three dimensional cell culture model of the canine endometrium. BMC Vet Res 2013; 9:86. [PMID: 23618385 PMCID: PMC3660264 DOI: 10.1186/1746-6148-9-86] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 04/22/2013] [Indexed: 12/21/2022] Open
Abstract
Background Oestrogens and progesterone have a significant impact on the endometrium during the canine oestrous cycle. Their receptors mediate plasma steroid hormone levels and are expressed in several endometrial cell types. Altered steroid receptor expression patterns are involved in serious uterine diseases; however the mechanisms of hormone action during pathogenesis in these tissues remain unclear. The development of 3D culture systems of canine endometrial cells provides an opportunity for the effects of steroid hormones to be quantitatively assessed in a more in vivo-like setting. The present study aimed to determine the effects of the steroid hormones 17β-estradiol (E) and progesterone (P) on the expression of the oestrogen and progesterone receptors (ER and PR), and on proliferative activity, in a 3D co-culture system of canine uterine origin, comprising differentiated endometrial glands, and stromal cells (SCs). Results Morphology, differentiation, and apical-basolateral polarity of cultured glandular epithelial cells (GECs) were comparable to those in native uterine tissue as assessed by immunohistochemistry using differentiation markers (β-catenin, laminin), lectin histochemistry, and transmission electron microscopy. Supplementation of our 3D-culture system with E (at 15, 30 and 100 pg/mL) resulted in constant levels of ER expression in GECs, but reduced expression levels in SCs. PR expression was reduced in both GECs and SCs following treatment with E. 3 ng/mL P resulted in increased ER expression in GECs, but a decrease in SCs. PR expression in GECs increased in all P-treated groups, whereas PRs in SCs decreased with the lowest and highest doses, but increased with the middle dose of treatment. Proliferative activity, assessed by Ki67 staining, remained below 1% in all assays and cell types. Conclusions The present study demonstrates the applicability of our 3D organotypic canine endometrium-derived culture system for cellular-level studies. 3D cultures represent near-physiological systems allowing reproducible quantitative experimentation, thus reducing the need to experiment on living animals. The results of the present investigation emphasize the importance of co-culture of the uterine glands with SCs, as it was shown that the responsiveness of the different cell types to steroid hormones were divergent in the 3D cell culture model.
Collapse
Affiliation(s)
- Cordula Bartel
- Department of Pathobiology, Institute of Anatomy, Histology and Embryology, University of Veterinary Medicine, Veterinaerplatz 1, Vienna A - 1210, Austria.
| | | | | | | | | |
Collapse
|
344
|
The effect of forced growth of cells into 3D spheres using low attachment surfaces on the acquisition of stemness properties. Biomaterials 2013; 34:3215-22. [DOI: 10.1016/j.biomaterials.2013.01.044] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 01/07/2013] [Indexed: 01/16/2023]
|
345
|
Abstract
BACKGROUND Microfluidic technology emerges as a convenient route to applying automated and reliable assays in a high-throughput manner with low cost. OBJECTIVE This review aims to answer questions related to the capabilities and potential applications of microfluidic assays that can benefit the drug development process and extends an outlook on its future trends. METHODS This article reviews recent publications in the field of microfluidics, with an emphasis on novel applications for drug development. RESULTS/CONCLUSION Microfluidics affords unique capabilities in sample preparation and separation, combinatorial synthesis and array formation, and incorporating nanotechnology for more functionalities. The pharmaceutical industry, facing challenges from limited productivity and accelerated competition, can thus greatly benefit from applying new microfluidic assays in various drug development stages, from target screening and lead optimization to absorption distribution metabolism elimination and toxicity studies in preclinical evaluations, diagnostics in clinical trials and drug formulation and manufacturing process optimization.
Collapse
Affiliation(s)
- Yuan Wen
- The Ohio State University, Department of Chemical and Biomolecular Engineering, 140 West 19th Avenue, Columbus, Ohio 43210, USA +1 614 2926611 ; +1 614 2923769 ;
| | | |
Collapse
|
346
|
Gurkan UA, Fan Y, Xu F, Erkmen B, Urkac ES, Parlakgul G, Bernstein J, Xing W, Boyden ES, Demirci U. Simple precision creation of digitally specified, spatially heterogeneous, engineered tissue architectures. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2013; 25:1192-8. [PMID: 23192949 PMCID: PMC3842103 DOI: 10.1002/adma.201203261] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 10/04/2012] [Indexed: 05/04/2023]
Affiliation(s)
- Umut Atakan Gurkan
- Harvard Medical School, Division of Biomedical Engineering at Brigham and Women's Hospital, Bio-Acoustic-MEMS in Medicine (BAMM) Laboratory, Harvard-MIT Health Sciences & Technology, 65 Landsdowne St. PRB 252, Cambridge, MA 02139, USA
| | - Yantao Fan
- Harvard Medical School, Division of Biomedical Engineering at Brigham and Women's Hospital, Bio-Acoustic-MEMS in Medicine (BAMM) Laboratory, Harvard-MIT Health Sciences & Technology, 65 Landsdowne St. PRB 252, Cambridge, MA 02139, USA
| | - Feng Xu
- Harvard Medical School, Division of Biomedical Engineering at Brigham and Women's Hospital, Bio-Acoustic-MEMS in Medicine (BAMM) Laboratory, Harvard-MIT Health Sciences & Technology, 65 Landsdowne St. PRB 252, Cambridge, MA 02139, USA
| | - Burcu Erkmen
- Harvard Medical School, Division of Biomedical Engineering at Brigham and Women's Hospital, Bio-Acoustic-MEMS in Medicine (BAMM) Laboratory, Harvard-MIT Health Sciences & Technology, 65 Landsdowne St. PRB 252, Cambridge, MA 02139, USA
| | - Emel Sokullu Urkac
- Harvard Medical School, Division of Biomedical Engineering at Brigham and Women's Hospital, Bio-Acoustic-MEMS in Medicine (BAMM) Laboratory, Harvard-MIT Health Sciences & Technology, 65 Landsdowne St. PRB 252, Cambridge, MA 02139, USA
| | - Gunes Parlakgul
- Harvard Medical School, Division of Biomedical Engineering at Brigham and Women's Hospital, Bio-Acoustic-MEMS in Medicine (BAMM) Laboratory, Harvard-MIT Health Sciences & Technology, 65 Landsdowne St. PRB 252, Cambridge, MA 02139, USA
| | - Jacob Bernstein
- Media Lab and McGovern Institute, Departments of Brain and Cognitive Sciences and Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Wangli Xing
- Medical Systems Biology Research Center, School of Medicine, Tsinghua University, Beijing 100084, PR China, National Engineering Research Center for Beijing Biochip Technology, 18 Life Science Parkway, Beijing, 102206, P. R. China
| | - Edward S. Boyden
- Media Lab and McGovern Institute, Departments of Brain and Cognitive Sciences and Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Utkan Demirci
- Harvard Medical School, Brigham and Women's Hospital, Harvard-MIT Health Sciences & Technology, 65 Landsdowne St. PRB 252, Cambridge, MA 02139, USA
| |
Collapse
|
347
|
Cai S, Xu H, Jiang Q, Yang Y. Novel 3D electrospun scaffolds with fibers oriented randomly and evenly in three dimensions to closely mimic the unique architectures of extracellular matrices in soft tissues: fabrication and mechanism study. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2013; 29:2311-2318. [PMID: 23390966 DOI: 10.1021/la304414j] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
In this work, novel electrospun scaffolds with fibers oriented randomly and evenly in three dimensions (3D) including in the thickness direction were developed based on the principle of electrostatic repulsion. This unique structure is different from most electrospun scaffolds with fibers oriented mainly in one direction. The structure of novel 3D scaffolds could more closely mimic the 3D randomly oriented fibrous architectures in many native extracellular matrices (ECMs). The cell culture results of this study indicated that, instead of becoming flattened cells when cultured in conventional electrospun scaffolds, the cells cultured on novel 3D scaffolds could develop into stereoscopic topographies, which highly simulated in vivo 3D cellular morphologies and are believed to be of vital importance for cells to function and differentiate appropriately. Also, due to the randomly oriented fibrous structure, improvement of nearly 5 times in cell proliferation could be observed when comparing our 3D scaffolds with 2D counterparts after 7 days of cell culture, while most currently reported 3D scaffolds only showed 1.5- to 2.5-fold improvement for the similar comparison. One mechanism of this fabrication process has also been proposed and showed that the rapid delivery of electrons on the fibers was the crucial factor for formation of 3D architectures.
Collapse
Affiliation(s)
- Shaobo Cai
- Department of Textiles, Merchandising and Fashion Design, University of Nebraska-Lincoln, Lincoln, Nebraska 68583-0802, United States
| | | | | | | |
Collapse
|
348
|
Long-term three-dimensional cell culture and anticancer drug activity evaluation in a microfluidic chip. Biosens Bioelectron 2013; 40:68-74. [DOI: 10.1016/j.bios.2012.06.017] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Revised: 06/11/2012] [Accepted: 06/12/2012] [Indexed: 12/21/2022]
|
349
|
Zimmermann M, Box C, Eccles SA. Two-dimensional vs. three-dimensional in vitro tumor migration and invasion assays. Methods Mol Biol 2013; 986:227-52. [PMID: 23436416 DOI: 10.1007/978-1-62703-311-4_15] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Motility and invasion are key hallmarks that distinguish benign from malignant tumors, enabling cells to cross tissue boundaries, disseminate in blood and lymph and establish metastases at distant sites. Similar properties are also utilized by activated endothelial cells during tumor-induced angiogenesis. It is now appreciated that these processes might provide a rich source of novel molecular targets with the potential for inhibitors to restrain both metastasis and neoangiogenesis. Such therapeutic strategies require assays that can rapidly and quantitatively measure cell movement and the ability to traverse physiological barriers. The need for high-throughput, however, must be balanced by assay designs that accommodate, as far as possible, the complexity of the in vivo tumor microenvironment. This chapter aims to give an overview of some commonly used migration and invasion assays to aid in the selection of a balanced portfolio of techniques for the rapid and accurate evaluation of novel therapeutic agents.
Collapse
Affiliation(s)
- Miriam Zimmermann
- Tumour Biology and Metastasis, Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, McElwain Laboratories, The Institute of Cancer Research, Surrey, UK
| | | | | |
Collapse
|
350
|
Zang R, Zhang X, Li M, Yang ST. Microwell bioreactor system for cell-based high throughput proliferation and cytotoxicity assays. Process Biochem 2013. [DOI: 10.1016/j.procbio.2012.11.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|