1
|
Takagi M, Yamada M, Utoh R, Seki M. A multiscale, vertical-flow perfusion system with integrated porous microchambers for upgrading multicellular spheroid culture. LAB ON A CHIP 2023; 23:2257-2267. [PMID: 37038847 DOI: 10.1039/d3lc00168g] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Spheroid formation assisted by microengineered chambers is a versatile approach for morphology-controlled three-dimensional (3D) cell cultivation with physiological relevance to human tissues. However, the limitation in diffusion-based oxygen/nutrient transport has been a critical issue for the densely packed cells in spheroids, preventing maximization of cellular functions and thus limiting their biomedical applications. Here, we have developed a multiscale microfluidic system for the perfusion culture of spheroids, in which porous microchambers, connected with microfluidic channels, were engineered. A newly developed process of centrifugation-assisted replica molding and salt-leaching enabled the formation of single micrometer-sized pores on the chamber surface and in the substrate. The porous configuration generates a vertical flow to directly supply the medium to the spheroids, while avoiding the formation of stagnant flow regions. We created seamlessly integrated, all PDMS/silicone-based microfluidic devices with an array of microchambers. Spheroids of human liver cells (HepG2 cells) were formed and cultured under vertical-flow perfusion, and the proliferation ability and liver cell-specific functions were compared with those of cells cultured in non-porous chambers with a horizontal flow. The presented system realizes both size-controlled formation of spheroids and direct medium supply, making it suitable as a precision cell culture platform for drug development, disease modelling, and regenerative medicine.
Collapse
Affiliation(s)
- Mai Takagi
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan.
| | - Masumi Yamada
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan.
| | - Rie Utoh
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan.
| | - Minoru Seki
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan.
| |
Collapse
|
2
|
Guo W, Chen Z, Feng Z, Li H, Zhang M, Zhang H, Cui X. Fabrication of Concave Microwells and Their Applications in Micro-Tissue Engineering: A Review. MICROMACHINES 2022; 13:mi13091555. [PMID: 36144178 PMCID: PMC9505614 DOI: 10.3390/mi13091555] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 05/27/2023]
Abstract
At present, there is an increasing need to mimic the in vivo micro-environment in the culture of cells and tissues in micro-tissue engineering. Concave microwells are becoming increasingly popular since they can provide a micro-environment that is closer to the in vivo environment compared to traditional microwells, which can facilitate the culture of cells and tissues. Here, we will summarize the fabrication methods of concave microwells, as well as their applications in micro-tissue engineering. The fabrication methods of concave microwells include traditional methods, such as lithography and etching, thermal reflow of photoresist, laser ablation, precision-computerized numerical control (CNC) milling, and emerging technologies, such as surface tension methods, the deformation of soft membranes, 3D printing, the molding of microbeads, air bubbles, and frozen droplets. The fabrication of concave microwells is transferring from professional microfabrication labs to common biochemical labs to facilitate their applications and provide convenience for users. Concave microwells have mostly been used in organ-on-a-chip models, including the formation and culture of 3D cell aggregates (spheroids, organoids, and embryoids). Researchers have also used microwells to study the influence of substrate topology on cellular behaviors. We will briefly review their applications in different aspects of micro-tissue engineering and discuss the further applications of concave microwells. We believe that building multiorgan-on-a-chip by 3D cell aggregates of different cell lines will be a popular application of concave microwells, while integrating physiologically relevant molecular analyses with the 3D culture platform will be another popular application in the near future. Furthermore, 3D cell aggregates from these biosystems will find more applications in drug screening and xenogeneic implantation.
Collapse
Affiliation(s)
- Weijin Guo
- Department of Biomedical Engineering, Shantou University, Shantou 515063, China
| | - Zejingqiu Chen
- Department of Biology, Shantou University, Shantou 515063, China
| | - Zitao Feng
- Department of Biomedical Engineering, Shantou University, Shantou 515063, China
| | - Haonan Li
- Department of Electrical Engineering, Shantou University, Shantou 515063, China
| | - Muyang Zhang
- Department of Electrical Engineering, Shantou University, Shantou 515063, China
| | - Huiru Zhang
- Guangdong Foshan Lianchuang Graduate School of Engineering, Foshan 528311, China
| | - Xin Cui
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| |
Collapse
|
3
|
He J, Zhou C, Xu X, Zhou Z, Danoy M, Shinohara M, Xiao W, Zhu D, Zhao X, Feng X, Mao Y, Sun W, Sakai Y, Yang H, Pang Y. Scalable Formation of Highly Viable and Functional Hepatocellular Carcinoma Spheroids in an Oxygen-Permeable Microwell Device for Anti-Tumor Drug Evaluation. Adv Healthc Mater 2022; 11:e2200863. [PMID: 35841538 DOI: 10.1002/adhm.202200863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/30/2022] [Indexed: 01/27/2023]
Abstract
For high-throughput anti-cancer drug screening, microwell arrays may serve as an effective tool to generate uniform and scalable tumor spheroids. However, microwell arrays are commonly anchored in non-oxygen-permeable culture plates, leading to limited oxygen supply for avascular spheroids. Herein, a polydimethylsiloxane (PDMS)-based oxygen-permeable microwell device is introduced for generating highly viable and functional hepatocellular carcinoma (HCC) spheroids. The PDMS sheets at the bottom of the microwell device provide a high flux of oxygen like in vivo neighboring hepatic sinusoids. Owing to the better oxygen supply, the generated HepG2 spheroids are larger in size and exhibit higher viability and proliferation with less cell apoptosis and necrosis. These spheroids also exhibit lower levels of anaerobic cellular respiration and express higher levels of liver-related functions. In anti-cancer drug testing, spheroids cultured in PDMS plates show a significantly stronger resistance against doxorubicin because of the stronger stem-cell and multidrug resistance phenotype. Moreover, higher expression of vascular endothelial growth factor-A produces a stronger angiogenesis capability of the spheroids. Overall, compared to the spheroids cultured in conventional non-oxygen-permeable plates, these spheroids can be used as a more favorable model for early-stage HCCs and be applied in high-throughput anti-cancer drug screening.
Collapse
Affiliation(s)
- Jianyu He
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, 100084, P. R. China.,Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Tsinghua University, Beijing, 100084, P. R. China.,Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, 100084, P. R. China
| | - Chang Zhou
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, 100084, P. R. China.,Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Tsinghua University, Beijing, 100084, P. R. China.,Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, 100084, P. R. China
| | - Xiaolei Xu
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, 100084, P. R. China.,Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Tsinghua University, Beijing, 100084, P. R. China.,Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, 100084, P. R. China.,Department of Hepatobiliary Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Changping District, Beijing, 102218, P. R. China
| | - Zhenzhen Zhou
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, 100084, P. R. China.,Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Tsinghua University, Beijing, 100084, P. R. China.,Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, 100084, P. R. China
| | - Mathieu Danoy
- Department of Chemical System Engineering, Graduate School of Engineering, University of Tokyo, Tokyo, 113-033, Japan
| | - Marie Shinohara
- Institute of Industrial Science, University of Tokyo, Tokyo, 153-8505, Japan
| | - Wenjin Xiao
- Centre de Recherche des Cordeliers, INSERM UMR-S1138, CNRS SNC5014, University of Paris, Paris, 75006, France
| | - Dong Zhu
- Clinical Laboratory, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Changping District, Beijing, 102218, P. R. China
| | - Xiuying Zhao
- Clinical Laboratory, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Changping District, Beijing, 102218, P. R. China
| | - Xiaobin Feng
- Department of Hepatobiliary Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Changping District, Beijing, 102218, P. R. China
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Dongcheng District, Beijing, 100005, P. R. China
| | - Wei Sun
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, 100084, P. R. China.,Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Tsinghua University, Beijing, 100084, P. R. China.,Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, 100084, P. R. China.,Department of Mechanical Engineering and Mechanics, College of Engineering, Drexel University, Philadelphia, PA, 19104, USA
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, Graduate School of Engineering, University of Tokyo, Tokyo, 113-033, Japan
| | - Huayu Yang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Dongcheng District, Beijing, 100005, P. R. China
| | - Yuan Pang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, 100084, P. R. China.,Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Tsinghua University, Beijing, 100084, P. R. China.,Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, 100084, P. R. China
| |
Collapse
|
4
|
Wang AJ, Allen A, Sofman M, Sphabmixay P, Yildiz E, Griffith LG. Engineering Modular 3D Liver Culture Microenvironments In Vitro to Parse the Interplay between Biophysical and Biochemical Microenvironment Cues on Hepatic Phenotypes. ADVANCED NANOBIOMED RESEARCH 2022; 2:2100049. [PMID: 35872804 PMCID: PMC9307216 DOI: 10.1002/anbr.202100049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
In vitro models of human liver functions are used across a diverse range of applications in preclinical drug development and disease modeling, with particular increasing interest in models that capture facets of liver inflammatory status. This study investigates how the interplay between biophysical and biochemical microenvironment cues influence phenotypic responses, including inflammation signatures, of primary human hepatocytes (PHH) cultured in a commercially available perfused bioreactor. A 3D printing-based alginate microwell system was designed to form thousands of hepatic spheroids in a scalable manner as a comparator 3D culture modality to the bioreactor. Soft, synthetic extracellular matrix (ECM) hydrogel scaffolds with biophysical properties mimicking features of liver were engineered to replace polystyrene scaffolds, and the biochemical microenvironment was modulated with a defined set of growth factors and signaling modulators. The supplemented media significantly increased tissue density, albumin secretion, and CYP3A4 activity but also upregulated inflammatory markers. Basal inflammatory markers were lower for cells maintained in ECM hydrogel scaffolds or spheroid formats than polystyrene scaffolds, while hydrogel scaffolds exhibited the most sensitive response to inflammation as assessed by multiplexed cytokine and RNA-seq analyses. Together, these engineered 3D liver microenvironments provide insights for probing human liver functions and inflammatory response in vitro.
Collapse
Affiliation(s)
- Alex J Wang
- Biological Engineering Department, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Allysa Allen
- Biological Engineering Department, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Marianna Sofman
- Biological Engineering Department, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Pierre Sphabmixay
- Mechanical Engineering Department, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA; Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA, 02142, USA
| | - Ece Yildiz
- Biological Engineering Department, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA; Institute of Bioengineering, School of Life Science, École Polytechnique Fédérale de Lausanne, Route Cantonale, 1015 Lausanne, Switzerland
| | - Linda G. Griffith
- Biological Engineering Department, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA; Center for Gynepathology Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| |
Collapse
|
5
|
de Hoyos-Vega JM, Hong HJ, Stybayeva G, Revzin A. Hepatocyte cultures: From collagen gel sandwiches to microfluidic devices with integrated biosensors. APL Bioeng 2021; 5:041504. [PMID: 34703968 PMCID: PMC8519630 DOI: 10.1063/5.0058798] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocytes are parenchymal cells of the liver responsible for drug detoxification, urea and bile production, serum protein synthesis, and glucose homeostasis. Hepatocytes are widely used for drug toxicity studies in bioartificial liver devices and for cell-based liver therapies. Because hepatocytes are highly differentiated cells residing in a complex microenvironment in vivo, they tend to lose hepatic phenotype and function in vitro. This paper first reviews traditional culture approaches used to rescue hepatic function in vitro and then discusses the benefits of emerging microfluidic-based culture approaches. We conclude by reviewing integration of hepatocyte cultures with bioanalytical or sensing approaches.
Collapse
Affiliation(s)
- Jose M. de Hoyos-Vega
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Hye Jin Hong
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| |
Collapse
|
6
|
Dellaquila A, Le Bao C, Letourneur D, Simon‐Yarza T. In Vitro Strategies to Vascularize 3D Physiologically Relevant Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100798. [PMID: 34351702 PMCID: PMC8498873 DOI: 10.1002/advs.202100798] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/23/2021] [Indexed: 05/04/2023]
Abstract
Vascularization of 3D models represents a major challenge of tissue engineering and a key prerequisite for their clinical and industrial application. The use of prevascularized models built from dedicated materials could solve some of the actual limitations, such as suboptimal integration of the bioconstructs within the host tissue, and would provide more in vivo-like perfusable tissue and organ-specific platforms. In the last decade, the fabrication of vascularized physiologically relevant 3D constructs has been attempted by numerous tissue engineering strategies, which are classified here in microfluidic technology, 3D coculture models, namely, spheroids and organoids, and biofabrication. In this review, the recent advancements in prevascularization techniques and the increasing use of natural and synthetic materials to build physiological organ-specific models are discussed. Current drawbacks of each technology, future perspectives, and translation of vascularized tissue constructs toward clinics, pharmaceutical field, and industry are also presented. By combining complementary strategies, these models are envisioned to be successfully used for regenerative medicine and drug development in a near future.
Collapse
Affiliation(s)
- Alessandra Dellaquila
- Université de ParisINSERM U1148X Bichat HospitalParisF‐75018France
- Elvesys Microfluidics Innovation CenterParis75011France
- Biomolecular PhotonicsDepartment of PhysicsUniversity of BielefeldBielefeld33615Germany
| | - Chau Le Bao
- Université de ParisINSERM U1148X Bichat HospitalParisF‐75018France
- Université Sorbonne Paris NordGalilée InstituteVilletaneuseF‐93430France
| | | | | |
Collapse
|
7
|
Velasco V, Shariati SA, Esfandyarpour R. Microtechnology-based methods for organoid models. MICROSYSTEMS & NANOENGINEERING 2020; 6:76. [PMID: 34567686 PMCID: PMC8433138 DOI: 10.1038/s41378-020-00185-3] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/10/2020] [Accepted: 06/03/2020] [Indexed: 05/03/2023]
Abstract
Innovations in biomaterials and stem cell technology have allowed for the emergence of novel three-dimensional (3D) tissue-like structures known as organoids and spheroids. As a result, compared to conventional 2D cell culture and animal models, these complex 3D structures have improved the accuracy and facilitated in vitro investigations of human diseases, human development, and personalized medical treatment. Due to the rapid progress of this field, numerous spheroid and organoid production methodologies have been published. However, many of the current spheroid and organoid production techniques are limited by complexity, throughput, and reproducibility. Microfabricated and microscale platforms (e.g., microfluidics and microprinting) have shown promise to address some of the current limitations in both organoid and spheroid generation. Microfabricated and microfluidic devices have been shown to improve nutrient delivery and exchange and have allowed for the arrayed production of size-controlled culture areas that yield more uniform organoids and spheroids for a higher throughput at a lower cost. In this review, we discuss the most recent production methods, challenges currently faced in organoid and spheroid production, and microfabricated and microfluidic applications for improving spheroid and organoid generation. Specifically, we focus on how microfabrication methods and devices such as lithography, microcontact printing, and microfluidic delivery systems can advance organoid and spheroid applications in medicine.
Collapse
Affiliation(s)
- Vanessa Velasco
- Biochemistry Department, Stanford University, Palo Alto, CA USA
| | - S. Ali Shariati
- Department of Biomolecular Engineering, Institute for the Biology of Stem Cells, University of California, Santa Cruz, CA USA
| | - Rahim Esfandyarpour
- Department of Electrical Engineering, University of California, Irvine, CA USA
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA USA
- Henry Samueli School of Engineering, University of California, Irvine, CA USA
| |
Collapse
|
8
|
Li Z, Guo X, Sun L, Xu J, Liu W, Li T, Wang J. A simple microsphere‐based mold to rapidly fabricate microwell arrays for multisize 3D tumor culture. Biotechnol Bioeng 2020; 117:1092-1100. [DOI: 10.1002/bit.27257] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/21/2019] [Accepted: 12/15/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Zixiu Li
- College of Chemistry & Pharmacy Northwest A&F University Yangling Shaanxi P. R. China
| | - Xiaofang Guo
- College of Chemistry & Pharmacy Northwest A&F University Yangling Shaanxi P. R. China
| | - Lili Sun
- College of Chemistry & Pharmacy Northwest A&F University Yangling Shaanxi P. R. China
| | - Juan Xu
- College of Chemistry & Pharmacy Northwest A&F University Yangling Shaanxi P. R. China
| | - Wenming Liu
- School of Basic Medical Science Central South University Changsha Hunan P. R. China
| | - Tianbao Li
- College of Chemistry & Pharmacy Northwest A&F University Yangling Shaanxi P. R. China
| | - Jinyi Wang
- College of Chemistry & Pharmacy Northwest A&F University Yangling Shaanxi P. R. China
| |
Collapse
|
9
|
Sun J, Warden AR, Ding X. Recent advances in microfluidics for drug screening. BIOMICROFLUIDICS 2019; 13:061503. [PMID: 31768197 PMCID: PMC6870548 DOI: 10.1063/1.5121200] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 11/07/2019] [Indexed: 05/03/2023]
Abstract
With ever increasing drug resistance and emergence of new diseases, demand for new drug development is at an unprecedented urgency. This fact has led to extensive recent efforts to develop new drugs and novel techniques for efficient drug screening. However, new drug development is commonly hindered by cost and time span. Thus, developing more accessible, cost-effective methods for drug screening is necessary. Compared with conventional drug screening methods, a microfluidic-based system has superior advantages in sample consumption, reaction time, and cost of the operation. In this paper, the advantages of microfluidic technology in drug screening as well as the critical factors for device design are described. The strategies and applications of microfluidics for drug screening are reviewed. Moreover, current limitations and future prospects for a drug screening microdevice are also discussed.
Collapse
Affiliation(s)
- Jiahui Sun
- State Key Laboratory of Oncogenes and Related Genes, Institute for
Personalized Medicine and School of Biomedical Engineering, Shanghai Jiao Tong
University, Shanghai 200030, China
| | - Antony R. Warden
- State Key Laboratory of Oncogenes and Related Genes, Institute for
Personalized Medicine and School of Biomedical Engineering, Shanghai Jiao Tong
University, Shanghai 200030, China
| | - Xianting Ding
- State Key Laboratory of Oncogenes and Related Genes, Institute for
Personalized Medicine and School of Biomedical Engineering, Shanghai Jiao Tong
University, Shanghai 200030, China
| |
Collapse
|
10
|
Underhill GH, Khetani SR. Advances in Engineered Human Liver Platforms for Drug Metabolism Studies. Drug Metab Dispos 2018; 46:1626-1637. [PMID: 30135245 DOI: 10.1124/dmd.118.083295] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/17/2018] [Indexed: 12/27/2022] Open
Abstract
Metabolism in the liver often determines the overall clearance rates of many pharmaceuticals. Furthermore, induction or inhibition of the liver drug metabolism enzymes by perpetrator drugs can influence the metabolism of victim drugs (drug-drug interactions). Therefore, determining liver-drug interactions is critical during preclinical drug development. Unfortunately, studies in animals are often of limited value because of significant differences in the metabolic pathways of the liver across different species. To mitigate such limitations, the pharmaceutical industry uses a continuum of human liver models, ranging from microsomes to transfected cell lines and cultures of primary human hepatocytes (PHHs). Of these models, PHHs provide a balance of high-throughput testing capabilities together with a physiologically relevant cell type that exhibits all the characteristic enzymes, cofactors, and transporters. However, PHH monocultures display a rapid decline in metabolic capacity. Consequently, bioengineers have developed several tools, such as cellular microarrays, micropatterned cocultures, self-assembled and bioprinted spheroids, and perfusion devices, to enhance and stabilize PHH functions for ≥2 weeks. Many of these platforms have been validated for drug studies, whereas some have been adapted to include liver nonparenchymal cells that can influence hepatic drug metabolism in health and disease. Here, we focus on the design features of such platforms and their representative drug metabolism validation datasets, while discussing emerging trends. Overall, the use of engineered human liver platforms in the pharmaceutical industry has been steadily rising over the last 10 years, and we anticipate that these platforms will become an integral part of drug development with continued commercialization and validation for routine screening use.
Collapse
Affiliation(s)
- Gregory H Underhill
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois; and Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| | - Salman R Khetani
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois; and Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
11
|
|
12
|
Zavrel EA, Shuler ML, Shen X. A SIMPLE ASPECT RATIO DEPENDENT METHOD OF PATTERNING MICROWELLS FOR SELECTIVE CELL ATTACHMENT. PROCEEDINGS OF THE ... DESIGN OF MEDICAL DEVICES CONFERENCE. DESIGN OF MEDICAL DEVICES CONFERENCE 2018; 2018:V001T05A001. [PMID: 30854173 PMCID: PMC6408146 DOI: 10.1115/dmd2018-6811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
3-D culture has been shown to provide cells with a more physiologically authentic environment than traditional 2-D (planar) culture [1, 2]. 3-D cues allow cells to exhibit more realistic functions and behaviors, e.g., adhesion, spreading, migration, metabolic activity, and differentiation. Knowledge of changes in cell morphology, mechanics, and mobility in response to geometrical cues and topological stimuli is important for understanding normal and pathological cell development [3]. Microfabrication provides unique in vitro approaches to recapitulating in vivo conditions due to the ability to precisely control the cellular microenvironment [4, 5]. Microwell arrays have emerged as robust alternatives to traditional 2D cell culture substrates as they are relatively simple and compatible with existing laboratory techniques and instrumentation [6, 7]. In particular, microwells have been adopted as a biomimetic approach to modeling the unique micro-architecture of the epithelial lining of the gastrointestinal (GI) tract [8–10]. The inner (lumen-facing) surface of the intestine has a convoluted topography consisting of finger-like projections (villi) with deep well-like invaginations (crypts) between them. The dimensions of villi and crypts are on the order of hundreds of microns (100–700 μm in height and 50–250 μm in diameter) [11]. While microwells have proven important in the development of physiologically realistic in vitro models of human intestine, existing methods of ensuring their surface is suitable for cell culture are lacking. Sometimes it is desirable to selectively seed cells within microwells and confine or restrict them to the microwells in which they are seeded. Existing methods of patterning microwells for cell attachment either lack selectivity, meaning cells can adhere and migrate anywhere on the microwell array, i.e., inside microwells or outside of them, or necessitate sophisticated techniques such as micro-contact printing, which requires precise alignment and control to selectively pattern the bottoms of microwells for cell attachment [12, 13].
Collapse
Affiliation(s)
- Erik A Zavrel
- Department of Biomedical Engineering, Cornell University Ithaca, NY, United States
| | - Michael L Shuler
- Department of Biomedical Engineering, Cornell University Ithaca, NY, United States
| | - Xiling Shen
- Department of Biomedical Engineering, Duke University Durham, NC, United States
| |
Collapse
|
13
|
Underhill GH, Khetani SR. Bioengineered Liver Models for Drug Testing and Cell Differentiation Studies. Cell Mol Gastroenterol Hepatol 2018; 5:426-439.e1. [PMID: 29675458 PMCID: PMC5904032 DOI: 10.1016/j.jcmgh.2017.11.012] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 11/21/2017] [Indexed: 12/19/2022]
Abstract
In vitro models of the human liver are important for the following: (1) mitigating the risk of drug-induced liver injury to human beings, (2) modeling human liver diseases, (3) elucidating the role of single and combinatorial microenvironmental cues on liver cell function, and (4) enabling cell-based therapies in the clinic. Methods to isolate and culture primary human hepatocytes (PHHs), the gold standard for building human liver models, were developed several decades ago; however, PHHs show a precipitous decline in phenotypic functions in 2-dimensional extracellular matrix-coated conventional culture formats, which does not allow chronic treatment with drugs and other stimuli. The development of several engineering tools, such as cellular microarrays, protein micropatterning, microfluidics, biomaterial scaffolds, and bioprinting, now allow precise control over the cellular microenvironment for enhancing the function of both PHHs and induced pluripotent stem cell-derived human hepatocyte-like cells; long-term (4+ weeks) stabilization of hepatocellular function typically requires co-cultivation with liver-derived or non-liver-derived nonparenchymal cell types. In addition, the recent development of liver organoid culture systems can provide a strategy for the enhanced expansion of therapeutically relevant cell types. Here, we discuss advances in engineering approaches for constructing in vitro human liver models that have utility in drug screening and for determining microenvironmental determinants of liver cell differentiation/function. Design features and validation data of representative models are presented to highlight major trends followed by the discussion of pending issues that need to be addressed. Overall, bioengineered liver models have significantly advanced our understanding of liver function and injury, which will prove useful for drug development and ultimately cell-based therapies.
Collapse
Key Words
- 3D, 3-dimensional
- BAL, bioartificial liver
- Bioprinting
- CRP, C-reactive protein
- CYP450, cytochrome P450
- Cellular Microarrays
- DILI, drug-induced liver injury
- ECM, extracellular matrix
- HSC, hepatic stellate cell
- Hepatocytes
- IL, interleukin
- KC, Kupffer cell
- LSEC, liver sinusoidal endothelial cell
- MPCC, micropatterned co-culture
- Microfluidics
- Micropatterned Co-Cultures
- NPC, nonparenchymal cell
- PEG, polyethylene glycol
- PHH, primary human hepatocyte
- Spheroids
- iHep, induced pluripotent stem cell-derived human hepatocyte-like cell
- iPS, induced pluripotent stem
Collapse
Affiliation(s)
- Gregory H. Underhill
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Salman R. Khetani
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
14
|
|
15
|
Shinohara M, Komori K, Fujii T, Sakai Y. Enhanced self-organization of size-controlled hepatocyte aggregates on oxygen permeable honeycomb microwell sheets. Biomed Phys Eng Express 2017. [DOI: 10.1088/2057-1976/aa7c3d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
16
|
All-in-one 3D printed microscopy chamber for multidimensional imaging, the UniverSlide. Sci Rep 2017; 7:42378. [PMID: 28186188 PMCID: PMC5301227 DOI: 10.1038/srep42378] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 12/29/2016] [Indexed: 12/11/2022] Open
Abstract
While live 3D high resolution microscopy techniques are developing rapidly, their use for biological applications is partially hampered by practical difficulties such as the lack of a versatile sample chamber. Here, we propose the design of a multi-usage observation chamber adapted for live 3D bio-imaging. We show the usefulness and practicality of this chamber, which we named the UniverSlide, for live imaging of two case examples, namely multicellular systems encapsulated in sub-millimeter hydrogel shells and zebrafish larvae. We also demonstrate its versatility and compatibility with all microscopy devices by using upright or inverted microscope configurations after loading the UniverSlide with fixed or living samples. Further, the device is applicable for medium/high throughput screening and automatized multi-position image acquisition, providing a constraint-free but stable and parallelized immobilization of the samples. The frame of the UniverSlide is fabricated using a stereolithography 3D printer, has the size of a microscopy slide, is autoclavable and sealed with a removable lid, which makes it suitable for use in a controlled culture environment. We describe in details how to build this chamber and we provide all the files necessary to print the different pieces in the lab.
Collapse
|
17
|
Priwitaningrum DL, Blondé JBG, Sridhar A, van Baarlen J, Hennink WE, Storm G, Le Gac S, Prakash J. Tumor stroma-containing 3D spheroid arrays: A tool to study nanoparticle penetration. J Control Release 2016; 244:257-268. [PMID: 27616660 DOI: 10.1016/j.jconrel.2016.09.004] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/05/2016] [Accepted: 09/07/2016] [Indexed: 12/14/2022]
Abstract
Nanoparticle penetration through tumor tissue after extravasation is considered as a key issue for tumor distribution and therapeutic effects. Most tumors possess abundant stroma, a fibrotic tissue composed of cancer-associated fibroblasts (CAFs) and extracellular matrix (ECM), which acts as a barrier for nanoparticle penetration. There is however a lack of suitable in vitro systems to study the tumor stroma penetration of nanoparticles. In the present study, we developed and thoroughly characterized a 3D co-culture spheroidal array to mimic tumor stroma and investigated the penetration of silica and PLGA nanoparticles in these spheroids. First, we examined human breast tumor patient biopsies to characterize the content and organization of stroma and found a high expression of alpha-smooth muscle actin (α-SMA; 40% positive area) and collagen-1 (50% positive area). Next, we prepared homospheroids of 4T1 mouse breast cancer cells or 3T3 mouse fibroblasts alone as well as heterospheroids combining 3T3 and 4T1 cells in different ratios (1:1 and 5:1) using a microwell array platform. Confocal live imaging revealed that fibroblasts distributed and reorganized within 48h in heterospheroids. Furthermore, immunohistochemical staining and gene expression analysis showed a proportional increase of α-SMA and collagen in heterospheroids with higher fibroblast ratios attaining 35% and 45% positive area at 5:1 (3T3:4T1) ratio, in a good match with the clinical breast tumor stroma. Subsequently, we studied the penetration of high and low negatively charged fluorescent silica nanoparticles (30nm; red and 100 or 70nm; green; zeta potential: -40mV and -20mV) and as well as Cy5-conjugated pegylated PLGA nanoparticles (200nm, -7mV) in both homo- and heterospheroid models. Fluorescent microscopy on spheroid cryosections after incubation with silica nanoparticles showed that 4T1 homospheroids allowed a high penetration of about 75-80% within 24h, with higher penetration in case of the 30nm nanoparticles. In contrast, spheroids with increasing fibroblast amounts significantly inhibited NP penetration. Silica nanoparticles with a less negative zeta potential exhibited lesser penetration compared to highly negative charged nanoparticles. Subsequently, similar experiments were conducted using Cy5-conjugated pegylated PLGA nanoparticles and confocal laser scanning microscopy; an increased nanoparticle penetration was found in 4T1 homospheroids until 48h, but significantly lower penetration in heterospheroids. Furthermore, we also developed human homospheroids (MDA-MB-231 or Panc-1 tumor cells) and heterospheroids (MDA-MB-231/BJ-hTert and Panc-1/pancreatic stellate cells) and performed silica nanoparticle (30 and 100nm) penetration studies. As a result, heterospheroids had significantly a lesser penetration of the nanoparticles compared to homospheroids. In conclusion, our data demonstrate that tumor stroma acts as a strong barrier for nanoparticle penetration. The 30-nm nanoparticles with low zeta potential favor deeper penetration. Furthermore, the herein proposed 3D co-culture platform that mimics the tumor stroma, is ideally suited to systematically investigate the factors influencing the penetration characteristics of newly developed nanomedicines to allow the design of nanoparticles with optimal penetration characteristics.
Collapse
Affiliation(s)
- Dwi L Priwitaningrum
- Targeted Therapeutics, Department of Biomaterials Science and Technology, MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Jean-Baptiste G Blondé
- Applied Microfluidics for BioEngineering Research, MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Adithya Sridhar
- Applied Microfluidics for BioEngineering Research, MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Joop van Baarlen
- Laboratorium Pathologie Oost-Nederland (LabPON), Hengelo, The Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht University, Utrecht, The Netherlands
| | - Gert Storm
- Targeted Therapeutics, Department of Biomaterials Science and Technology, MIRA Institute, University of Twente, Enschede, The Netherlands; Department of Pharmaceutics, Utrecht University, Utrecht, The Netherlands
| | - Séverine Le Gac
- Applied Microfluidics for BioEngineering Research, MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Jai Prakash
- Targeted Therapeutics, Department of Biomaterials Science and Technology, MIRA Institute, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
18
|
Fagoonee S, Famulari ES, Silengo L, Camussi G, Altruda F. Prospects for Adult Stem Cells in the Treatment of Liver Diseases. Stem Cells Dev 2016; 25:1471-1482. [PMID: 27503633 DOI: 10.1089/scd.2016.0144] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hepatocytes constitute the main bulk of the liver and perform several essential functions. After injury, the hepatocytes have a remarkable capacity to regenerate and restore functionality. However, in some cases, the endogenous hepatocytes cannot replicate or restore the function, and liver transplantation, which is not exempt of complications, is required. Stem cells offer in theory the possibility of generating unlimited supply of hepatocytes in vitro due to their capacity to self-renew and differentiate when given the right cues. Stem cells isolated from an array of tissues have been investigated for their capacity to differentiate into hepatocyte-like cells in vitro and are employed in rescue experiments in vivo. Adult stem cells have gained in attractiveness over embryonic stem cells for liver cell therapy due to their origin, multipotentiality, and the possibility of autologous transplantation. This review deals with the promise and limitations of adult stem cells in clinically restoring liver functionality.
Collapse
Affiliation(s)
- Sharmila Fagoonee
- 1 Institute of Biostructure and Bioimaging , CNR, Turin, Italy .,2 Molecular Biotechnology Center, University of Turin , Turin, Italy .,3 Department of Molecular Biotechnology and Health Sciences, University of Turin , Turin, Italy
| | - Elvira Smeralda Famulari
- 2 Molecular Biotechnology Center, University of Turin , Turin, Italy .,3 Department of Molecular Biotechnology and Health Sciences, University of Turin , Turin, Italy
| | - Lorenzo Silengo
- 2 Molecular Biotechnology Center, University of Turin , Turin, Italy .,3 Department of Molecular Biotechnology and Health Sciences, University of Turin , Turin, Italy
| | - Giovanni Camussi
- 2 Molecular Biotechnology Center, University of Turin , Turin, Italy .,4 Department of Medical Sciences, University of Torino , Torino, Italy
| | - Fiorella Altruda
- 2 Molecular Biotechnology Center, University of Turin , Turin, Italy .,3 Department of Molecular Biotechnology and Health Sciences, University of Turin , Turin, Italy
| |
Collapse
|
19
|
Ware BR, Khetani SR. Engineered Liver Platforms for Different Phases of Drug Development. Trends Biotechnol 2016; 35:172-183. [PMID: 27592803 DOI: 10.1016/j.tibtech.2016.08.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/26/2016] [Accepted: 08/02/2016] [Indexed: 12/12/2022]
Abstract
Drug-induced liver injury (DILI) remains a leading cause of drug withdrawal from human clinical trials or the marketplace. Owing to species-specific differences in liver pathways, predicting human-relevant DILI using in vitro human liver models is crucial. Microfabrication tools allow precise control over the cellular microenvironment towards stabilizing liver functions for weeks. These tools are used to engineer human liver models with different complexities and throughput using cell lines, primary cells, and stem cell-derived hepatocytes. Including multiple human liver cell types can mimic cell-cell interactions in specific types of DILI. Finally, organ-on-a-chip models demonstrate how drug metabolism in the liver affects multi-organ toxicities. In this review we survey engineered human liver platforms within the needs of different phases of drug development.
Collapse
Affiliation(s)
- Brenton R Ware
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA; Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Salman R Khetani
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA.
| |
Collapse
|
20
|
Munaz A, Vadivelu RK, John JAS, Nguyen NT. A lab-on-a-chip device for investigating the fusion process of olfactory ensheathing cell spheroids. LAB ON A CHIP 2016; 16:2946-54. [PMID: 27387270 DOI: 10.1039/c6lc00815a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Understanding the process of fusion of olfactory ensheathing cell spheroids will lead to improvement of cell transplantation therapies to repair spinal cord injuries. The successful fusion of transplanted spheroids will enable alternative transplantation strategies to be developed for in vivo applications. This paper describes the use of a microfluidic device to trap and fuse olfactory ensheathing cell spheroids. The velocity, the pressure distribution in the device were simulated numerically to predict the trapping location. The simulation predicted the optimum flow rates for trapping the spheroids in the later experiments. Simulated particle trajectories were verified experimentally with tracing of fluorescent micro particles. The fusion process of the spheroids was investigated over a period of 48 hours. The microfluidic platform presented here can be used for testing potential drugs that can promote the fusion process and improve the transplantation therapy.
Collapse
Affiliation(s)
- Ahmed Munaz
- Queensland Micro- and Nanotechnology Centre, Griffith University, Brisbane, QLD 4111, Australia.
| | | | | | | |
Collapse
|
21
|
Hannoun Z, Steichen C, Dianat N, Weber A, Dubart-Kupperschmitt A. The potential of induced pluripotent stem cell derived hepatocytes. J Hepatol 2016; 65:182-199. [PMID: 26916529 DOI: 10.1016/j.jhep.2016.02.025] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 01/12/2016] [Accepted: 02/09/2016] [Indexed: 12/21/2022]
Abstract
Orthotopic liver transplantation remains the only curative treatment for liver disease. However, the number of patients who die while on the waiting list (15%) has increased in recent years as a result of severe organ shortages; furthermore the incidence of liver disease is increasing worldwide. Clinical trials involving hepatocyte transplantation have provided encouraging results. However, transplanted cell function appears to often decline after several months, necessitating liver transplantation. The precise aetiology of the loss of cell function is not clear, but poor engraftment and immune-mediated loss appear to be important factors. Also, primary human hepatocytes (PHH) are not readily available, de-differentiate, and die rapidly in culture. Hepatocytes are available from other sources, such as tumour-derived human hepatocyte cell lines and immortalised human hepatocyte cell lines or porcine hepatocytes. However, all these cells suffer from various limitations such as reduced or differences in functions or risk of zoonotic infections. Due to their significant potential, one possible inexhaustible source of hepatocytes is through the directed differentiation of human induced pluripotent stem cells (hiPSCs). This review will discuss the potential applications and existing limitations of hiPSC-derived hepatocytes in regenerative medicine, drug screening, in vitro disease modelling and bioartificial livers.
Collapse
Affiliation(s)
- Zara Hannoun
- INSERM U1193, Hôpital Paul Brousse, Villejuif F-94807, France; UMR_S1193, Université Paris-Sud, Hôpital Paul Brousse, Villejuif F-94800, France; Département hospitalo-universitaire Hepatinov, Hôpital Paul Brousse, Villejuif F-94807, France
| | - Clara Steichen
- INSERM U1193, Hôpital Paul Brousse, Villejuif F-94807, France; UMR_S1193, Université Paris-Sud, Hôpital Paul Brousse, Villejuif F-94800, France; Département hospitalo-universitaire Hepatinov, Hôpital Paul Brousse, Villejuif F-94807, France
| | - Noushin Dianat
- INSERM U1193, Hôpital Paul Brousse, Villejuif F-94807, France; UMR_S1193, Université Paris-Sud, Hôpital Paul Brousse, Villejuif F-94800, France; Département hospitalo-universitaire Hepatinov, Hôpital Paul Brousse, Villejuif F-94807, France
| | - Anne Weber
- INSERM U1193, Hôpital Paul Brousse, Villejuif F-94807, France; UMR_S1193, Université Paris-Sud, Hôpital Paul Brousse, Villejuif F-94800, France; Département hospitalo-universitaire Hepatinov, Hôpital Paul Brousse, Villejuif F-94807, France
| | - Anne Dubart-Kupperschmitt
- INSERM U1193, Hôpital Paul Brousse, Villejuif F-94807, France; UMR_S1193, Université Paris-Sud, Hôpital Paul Brousse, Villejuif F-94800, France; Département hospitalo-universitaire Hepatinov, Hôpital Paul Brousse, Villejuif F-94807, France.
| |
Collapse
|
22
|
Alhasan L, Qi A, Al-Abboodi A, Rezk A, Chan PP, Iliescu C, Yeo LY. Rapid Enhancement of Cellular Spheroid Assembly by Acoustically Driven Microcentrifugation. ACS Biomater Sci Eng 2016; 2:1013-1022. [DOI: 10.1021/acsbiomaterials.6b00144] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Layla Alhasan
- Biotechnology & Biological Sciences, School of Applied Science, RMIT University, Melbourne, Victoria 3000, Australia
| | - Aisha Qi
- Micro/Nanophysics
Research Laboratory, RMIT University, Melbourne, Victoria 3000, Australia
| | - Aswan Al-Abboodi
- Department
of Chemical Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Amgad Rezk
- Micro/Nanophysics
Research Laboratory, RMIT University, Melbourne, Victoria 3000, Australia
| | - Peggy P.Y. Chan
- Micro/Nanophysics
Research Laboratory, RMIT University, Melbourne, Victoria 3000, Australia
- Department
of Biomedical Engineering, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Ciprian Iliescu
- Institute
of Bioengineering and Nanotechnology, A*STAR, Singapore 138669, Singapore
| | - Leslie Y. Yeo
- Micro/Nanophysics
Research Laboratory, RMIT University, Melbourne, Victoria 3000, Australia
| |
Collapse
|
23
|
Tripathi A, Melo JS. Preparation of a sponge-like biocomposite agarose–chitosan scaffold with primary hepatocytes for establishing an in vitro 3D liver tissue model. RSC Adv 2015. [DOI: 10.1039/c5ra04153h] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Sponge-like agarose–chitosan scaffold synthesized by cryo-polymerization andin vitroevaluation of interfacial cell–material interaction and liver-like functions of impregnate primary hepatocytes.
Collapse
Affiliation(s)
- Anuj Tripathi
- Nuclear Agriculture and Biotechnology Division
- Bhabha Atomic Research Centre
- Mumbai – 400 085
- India
| | - Jose Savio Melo
- Nuclear Agriculture and Biotechnology Division
- Bhabha Atomic Research Centre
- Mumbai – 400 085
- India
| |
Collapse
|
24
|
Poly(ethylene glycol) (PEG) microwells in microfluidics: Fabrication methods and applications. BIOCHIP JOURNAL 2014. [DOI: 10.1007/s13206-014-8401-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
25
|
Patra B, Peng YS, Peng CC, Liao WH, Chen YA, Lin KH, Tung YC, Lee CH. Migration and vascular lumen formation of endothelial cells in cancer cell spheroids of various sizes. BIOMICROFLUIDICS 2014; 8:052109. [PMID: 25332736 PMCID: PMC4189544 DOI: 10.1063/1.4895568] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 09/01/2014] [Indexed: 05/16/2023]
Abstract
We developed a microfluidic device to culture cellular spheroids of controlled sizes and suitable for live cell imaging by selective plane illumination microscopy (SPIM). We cocultured human umbilical vein endothelial cells (HUVECs) within the spheroids formed by hepatocellular carcinoma cells, and studied the distributions of the HUVECs over time. We observed that the migration of HUVECs depended on the size of spheroids. In the spheroids of ∼200 μm diameters, HUVECs migrated outwards to the edges within 48 h; while in the spheroids of ∼250 μm diameters, there was no outward migration of the HUVECs up to 72 h. In addition, we studied the effects of pro-angiogenic factors, namely, vascular endothelial growth factor (VEGF) and fibroblast growth factor (β-FGF), on the migration of HUVECs in the carcinoma cell spheroid. The outward migration of HUVECs in 200 μm spheroids was hindered by the treatment with VEGF and β-FGF. Moreover, some of the HUVECs formed hollow lumen within 72 h under VEGF and β-FGF treatment. The combination of SPIM and microfluidic devices gives high resolution in both spatial and temporal domains. The observation of HUVECs in spheroids provides us insight on tumor vascularization, an ideal disease model for drug screening and fundamental studies.
Collapse
Affiliation(s)
| | | | - Chien-Chung Peng
- Research Center for Applied Sciences , Academia Sinica, Taipei 11529, Taiwan
| | - Wei-Hao Liao
- Research Center for Applied Sciences , Academia Sinica, Taipei 11529, Taiwan
| | | | - Keng-Hui Lin
- Institute of Physics , Academia Sinica, Taipei 11529, Taiwan
| | - Yi-Chung Tung
- Research Center for Applied Sciences , Academia Sinica, Taipei 11529, Taiwan
| | | |
Collapse
|
26
|
Ruppen J, Cortes-Dericks L, Marconi E, Karoubi G, Schmid RA, Peng R, Marti TM, Guenat OT. A microfluidic platform for chemoresistive testing of multicellular pleural cancer spheroids. LAB ON A CHIP 2014; 14:1198-1205. [PMID: 24496222 DOI: 10.1039/c3lc51093j] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
This study reports on a microfluidic platform on which single multicellular spheroids from malignant pleural mesothelioma (MPM), an aggressive tumor with poor prognosis, can be loaded, trapped and tested for chemotherapeutic drug response. A new method to detect the spheroid viability cultured on the microfluidic chip as a function of the drug concentration is presented. This approach is based on the evaluation of the caspase activity in the supernatant sampled from the chip and tested using a microplate reader. This simple and time-saving method does only require a minimum amount of manipulations and was established for very low numbers of cells. This feature is particularly important in view of personalised medicine applications for which the number of cells obtained from the patients is low. MPM spheroids were continuously perfused for 48 hours with cisplatin, one of the standard chemotherapeutic drugs used to treat MPM. The 50% growth inhibitory concentration of cisplatin in perfused MPM spheroids was found to be twice as high as in spheroids cultured under static conditions. This chemoresistance increase might be due to the continuous support of nutrients and oxygen to the perfused spheroids.
Collapse
Affiliation(s)
- Janine Ruppen
- ARTORG Lung Regeneration Technologies Lab, University of Berne, Murtenstrasse 50, CH-3010 Berne, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Liu Y, Li H, Yan S, Wei J, Li X. Hepatocyte cocultures with endothelial cells and fibroblasts on micropatterned fibrous mats to promote liver-specific functions and capillary formation capabilities. Biomacromolecules 2014; 15:1044-54. [PMID: 24547870 DOI: 10.1021/bm401926k] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The maintenance of hepatocyte phenotype and functions remains as a great challenge in the generation of functional liver tissue and in vitro model for drug metabolism studies. The use of hepatocyte coculture systems plays essential roles in the establishment of cell-cell and cell-extracellular matrix communications similar to native liver tissues. In the current study, micropatterned electrospun fibrous mats were created to load hepatocytes, fibroblasts, and endothelial cells (ECs), which were precisely assembled to establish their spatially controlled coculture for mimicking the in vivo structure of hepatic lobules. Hepatocytes formed compact polyhedral spheroids with an average diameter of 80-100 μm, reorganized actin filaments in the cell-cell contact regions, and well-developed bile canaliculi. Compared with hepatocytes cultured alone, the coculture of hepatocytes with either fibroblasts or ECs led to significantly higher albumin secretion, urea synthesis and cytochrome P-450 expression, which were dramatically improved by the coculture of hepatocytes with both fibroblasts and ECs. The cocultured ECs well spread on patterned regions with little organized filamentous actin, and significantly higher densities and deeper penetration into patterned scaffolds were determined for ECs after coculture with fibroblasts and hepatocytes compared with those after cultured alone or coculture with either fibroblasts or hepatocytes. A Matrigel overlay assay showed that the capabilities of ECs to form capillary tubes were significantly enhanced by micropatterned coculture with fibroblasts and hepatocytes. Thus, the coculture of hepatocytes, fibroblasts, and ECs on micropatterned fibrous mats helps both hepatocytes in the maintenance of hepatic functions and ECs in the formation of capillary-like structures. It is suggested that the micropatterned coculture model described here not only provides functional hepatic tissues for predictions of drug metabolism profiles, but also will enable investigations on more complex and physiological cell-cell communications.
Collapse
Affiliation(s)
- Yaowen Liu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University , Chengdu 610031, People's Republic of China
| | | | | | | | | |
Collapse
|
28
|
Abstract
The liver is a target of in vitro tissue engineering despite its capability to regenerate in vivo. The construction of liver tissues in vitro remains challenging. In this review, conventional 3D cultures of hepatocytes are first discussed. Recent advances in the 3D culturing of liver cells are then summarized in the context of in vitro liver tissue reconstruction at the micro- and macroscales. The application of microfluidics technology to liver tissue engineering has been introduced as a bottom-up approach performed at the microscale, whereas whole-organ bioengineering technology was introduced as a top-down approach performed at the macroscale. Mesoscale approaches are also discussed in considering the integration of micro- and macroscale approaches. Multiple parallel multiscale liver tissue engineering studies are ongoing; however, no tissue-engineered liver that is appropriate for clinical use has yet been realized. The integration of multiscale tissue engineering studies is essential for further understanding of liver reconstruction strategies.
Collapse
Affiliation(s)
- Ryo Sudo
- Department of System Design Engineering; Keio University; Yokohama, Japan
| |
Collapse
|
29
|
Liu Y, Zhang L, Wei J, Yan S, Yu J, Li X. Promoting hepatocyte spheroid formation and functions by coculture with fibroblasts on micropatterned electrospun fibrous scaffolds. J Mater Chem B 2014; 2:3029-3040. [DOI: 10.1039/c3tb21779e] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
30
|
Godoy P, Hewitt NJ, Albrecht U, Andersen ME, Ansari N, Bhattacharya S, Bode JG, Bolleyn J, Borner C, Böttger J, Braeuning A, Budinsky RA, Burkhardt B, Cameron NR, Camussi G, Cho CS, Choi YJ, Craig Rowlands J, Dahmen U, Damm G, Dirsch O, Donato MT, Dong J, Dooley S, Drasdo D, Eakins R, Ferreira KS, Fonsato V, Fraczek J, Gebhardt R, Gibson A, Glanemann M, Goldring CEP, Gómez-Lechón MJ, Groothuis GMM, Gustavsson L, Guyot C, Hallifax D, Hammad S, Hayward A, Häussinger D, Hellerbrand C, Hewitt P, Hoehme S, Holzhütter HG, Houston JB, Hrach J, Ito K, Jaeschke H, Keitel V, Kelm JM, Kevin Park B, Kordes C, Kullak-Ublick GA, LeCluyse EL, Lu P, Luebke-Wheeler J, Lutz A, Maltman DJ, Matz-Soja M, McMullen P, Merfort I, Messner S, Meyer C, Mwinyi J, Naisbitt DJ, Nussler AK, Olinga P, Pampaloni F, Pi J, Pluta L, Przyborski SA, Ramachandran A, Rogiers V, Rowe C, Schelcher C, Schmich K, Schwarz M, Singh B, Stelzer EHK, Stieger B, Stöber R, Sugiyama Y, Tetta C, Thasler WE, Vanhaecke T, Vinken M, Weiss TS, Widera A, Woods CG, Xu JJ, Yarborough KM, Hengstler JG. Recent advances in 2D and 3D in vitro systems using primary hepatocytes, alternative hepatocyte sources and non-parenchymal liver cells and their use in investigating mechanisms of hepatotoxicity, cell signaling and ADME. Arch Toxicol 2013; 87:1315-530. [PMID: 23974980 PMCID: PMC3753504 DOI: 10.1007/s00204-013-1078-5] [Citation(s) in RCA: 1062] [Impact Index Per Article: 96.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 05/06/2013] [Indexed: 12/15/2022]
Abstract
This review encompasses the most important advances in liver functions and hepatotoxicity and analyzes which mechanisms can be studied in vitro. In a complex architecture of nested, zonated lobules, the liver consists of approximately 80 % hepatocytes and 20 % non-parenchymal cells, the latter being involved in a secondary phase that may dramatically aggravate the initial damage. Hepatotoxicity, as well as hepatic metabolism, is controlled by a set of nuclear receptors (including PXR, CAR, HNF-4α, FXR, LXR, SHP, VDR and PPAR) and signaling pathways. When isolating liver cells, some pathways are activated, e.g., the RAS/MEK/ERK pathway, whereas others are silenced (e.g. HNF-4α), resulting in up- and downregulation of hundreds of genes. An understanding of these changes is crucial for a correct interpretation of in vitro data. The possibilities and limitations of the most useful liver in vitro systems are summarized, including three-dimensional culture techniques, co-cultures with non-parenchymal cells, hepatospheres, precision cut liver slices and the isolated perfused liver. Also discussed is how closely hepatoma, stem cell and iPS cell-derived hepatocyte-like-cells resemble real hepatocytes. Finally, a summary is given of the state of the art of liver in vitro and mathematical modeling systems that are currently used in the pharmaceutical industry with an emphasis on drug metabolism, prediction of clearance, drug interaction, transporter studies and hepatotoxicity. One key message is that despite our enthusiasm for in vitro systems, we must never lose sight of the in vivo situation. Although hepatocytes have been isolated for decades, the hunt for relevant alternative systems has only just begun.
Collapse
Affiliation(s)
- Patricio Godoy
- Leibniz Research Centre for Working Environment and Human Factors (IFADO), 44139 Dortmund, Germany
| | | | - Ute Albrecht
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Melvin E. Andersen
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Nariman Ansari
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Sudin Bhattacharya
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Johannes Georg Bode
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Jennifer Bolleyn
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Christoph Borner
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany
| | - Jan Böttger
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Albert Braeuning
- Department of Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Wilhelmstr. 56, 72074 Tübingen, Germany
| | - Robert A. Budinsky
- Toxicology and Environmental Research and Consulting, The Dow Chemical Company, Midland, MI USA
| | - Britta Burkhardt
- BG Trauma Center, Siegfried Weller Institut, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Neil R. Cameron
- Department of Chemistry, Durham University, Durham, DH1 3LE UK
| | - Giovanni Camussi
- Department of Medical Sciences, University of Torino, 10126 Turin, Italy
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921 Korea
| | - Yun-Jaie Choi
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921 Korea
| | - J. Craig Rowlands
- Toxicology and Environmental Research and Consulting, The Dow Chemical Company, Midland, MI USA
| | - Uta Dahmen
- Experimental Transplantation Surgery, Department of General Visceral, and Vascular Surgery, Friedrich-Schiller-University Jena, 07745 Jena, Germany
| | - Georg Damm
- Department of General-, Visceral- and Transplantation Surgery, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Olaf Dirsch
- Institute of Pathology, Friedrich-Schiller-University Jena, 07745 Jena, Germany
| | - María Teresa Donato
- Unidad de Hepatología Experimental, IIS Hospital La Fe Avda Campanar 21, 46009 Valencia, Spain
- CIBERehd, Fondo de Investigaciones Sanitarias, Barcelona, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Jian Dong
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Steven Dooley
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Dirk Drasdo
- Interdisciplinary Center for Bioinformatics (IZBI), University of Leipzig, 04107 Leipzig, Germany
- INRIA (French National Institute for Research in Computer Science and Control), Domaine de Voluceau-Rocquencourt, B.P. 105, 78153 Le Chesnay Cedex, France
- UPMC University of Paris 06, CNRS UMR 7598, Laboratoire Jacques-Louis Lions, 4, pl. Jussieu, 75252 Paris cedex 05, France
| | - Rowena Eakins
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Karine Sá Ferreira
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany
- GRK 1104 From Cells to Organs, Molecular Mechanisms of Organogenesis, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Valentina Fonsato
- Department of Medical Sciences, University of Torino, 10126 Turin, Italy
| | - Joanna Fraczek
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Rolf Gebhardt
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Andrew Gibson
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Matthias Glanemann
- Department of General-, Visceral- and Transplantation Surgery, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Chris E. P. Goldring
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - María José Gómez-Lechón
- Unidad de Hepatología Experimental, IIS Hospital La Fe Avda Campanar 21, 46009 Valencia, Spain
- CIBERehd, Fondo de Investigaciones Sanitarias, Barcelona, Spain
| | - Geny M. M. Groothuis
- Department of Pharmacy, Pharmacokinetics Toxicology and Targeting, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Lena Gustavsson
- Department of Laboratory Medicine (Malmö), Center for Molecular Pathology, Lund University, Jan Waldenströms gata 59, 205 02 Malmö, Sweden
| | - Christelle Guyot
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | - David Hallifax
- Centre for Applied Pharmacokinetic Research (CAPKR), School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT UK
| | - Seddik Hammad
- Department of Forensic Medicine and Veterinary Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Adam Hayward
- Biological and Biomedical Sciences, Durham University, Durham, DH13LE UK
| | - Dieter Häussinger
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Claus Hellerbrand
- Department of Medicine I, University Hospital Regensburg, 93053 Regensburg, Germany
| | | | - Stefan Hoehme
- Interdisciplinary Center for Bioinformatics (IZBI), University of Leipzig, 04107 Leipzig, Germany
| | - Hermann-Georg Holzhütter
- Institut für Biochemie Abteilung Mathematische Systembiochemie, Universitätsmedizin Berlin (Charité), Charitéplatz 1, 10117 Berlin, Germany
| | - J. Brian Houston
- Centre for Applied Pharmacokinetic Research (CAPKR), School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT UK
| | | | - Kiyomi Ito
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo, 202-8585 Japan
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160 USA
| | - Verena Keitel
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | | | - B. Kevin Park
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Claus Kordes
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Gerd A. Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | - Edward L. LeCluyse
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Peng Lu
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | | | - Anna Lutz
- Department of Pharmaceutical Biology and Biotechnology, University of Freiburg, Freiburg, Germany
| | - Daniel J. Maltman
- Reinnervate Limited, NETPark Incubator, Thomas Wright Way, Sedgefield, TS21 3FD UK
| | - Madlen Matz-Soja
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Patrick McMullen
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Irmgard Merfort
- Department of Pharmaceutical Biology and Biotechnology, University of Freiburg, Freiburg, Germany
| | | | - Christoph Meyer
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jessica Mwinyi
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | - Dean J. Naisbitt
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Andreas K. Nussler
- BG Trauma Center, Siegfried Weller Institut, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Peter Olinga
- Division of Pharmaceutical Technology and Biopharmacy, Department of Pharmacy, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Francesco Pampaloni
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Jingbo Pi
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Linda Pluta
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Stefan A. Przyborski
- Reinnervate Limited, NETPark Incubator, Thomas Wright Way, Sedgefield, TS21 3FD UK
- Biological and Biomedical Sciences, Durham University, Durham, DH13LE UK
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160 USA
| | - Vera Rogiers
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Cliff Rowe
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Celine Schelcher
- Department of Surgery, Liver Regeneration, Core Facility, Human in Vitro Models of the Liver, Ludwig Maximilians University of Munich, Munich, Germany
| | - Kathrin Schmich
- Department of Pharmaceutical Biology and Biotechnology, University of Freiburg, Freiburg, Germany
| | - Michael Schwarz
- Department of Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Wilhelmstr. 56, 72074 Tübingen, Germany
| | - Bijay Singh
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921 Korea
| | - Ernst H. K. Stelzer
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Bruno Stieger
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | - Regina Stöber
- Leibniz Research Centre for Working Environment and Human Factors (IFADO), 44139 Dortmund, Germany
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Yokohama Biopharmaceutical R&D Center, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045 Japan
| | - Ciro Tetta
- Fresenius Medical Care, Bad Homburg, Germany
| | - Wolfgang E. Thasler
- Department of Surgery, Ludwig-Maximilians-University of Munich Hospital Grosshadern, Munich, Germany
| | - Tamara Vanhaecke
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Mathieu Vinken
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Thomas S. Weiss
- Department of Pediatrics and Juvenile Medicine, University of Regensburg Hospital, Regensburg, Germany
| | - Agata Widera
- Leibniz Research Centre for Working Environment and Human Factors (IFADO), 44139 Dortmund, Germany
| | - Courtney G. Woods
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | | | | | - Jan G. Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IFADO), 44139 Dortmund, Germany
| |
Collapse
|
31
|
Patra B, Chen YH, Peng CC, Lin SC, Lee CH, Tung YC. A microfluidic device for uniform-sized cell spheroids formation, culture, harvesting and flow cytometry analysis. BIOMICROFLUIDICS 2013; 7:54114. [PMID: 24396525 PMCID: PMC3808411 DOI: 10.1063/1.4824480] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 09/24/2013] [Indexed: 05/10/2023]
Abstract
Culture of cells as three-dimensional (3D) aggregates, named spheroids, possesses great potential to improve in vitro cell models for basic biomedical research. However, such cell spheroid models are often complicated, cumbersome, and expensive compared to conventional Petri-dish cell cultures. In this work, we developed a simple microfluidic device for cell spheroid formation, culture, and harvesting. Using this device, cells could form uniformly sized spheroids due to strong cell-cell interactions and the spatial confinement of microfluidic culture chambers. We demonstrated cell spheroid formation and culture in the designed devices using embryonic stem cells, carcinoma cells, and fibroblasts. We further scaled up the device capable of simultaneously forming and culturing 5000 spheroids in a single chip. Finally, we demonstrated harvesting of the cultured spheroids from the device with a simple setup. The harvested spheroids possess great integrity, and the cells can be exploited for further flow cytometry assays due to the ample cell numbers.
Collapse
Affiliation(s)
- Bishnubrata Patra
- Institute of Biophotonics, National Yang-Ming University, Taipei 11221, Taiwan ; Biophotonics & Molecular Imaging Research Center (BMIRC), National Yang-Ming University, Taipei 11221, Taiwan
| | - Ying-Hua Chen
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Chien-Chung Peng
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Shiang-Chi Lin
- Graduate Institute of Electronics Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Chau-Hwang Lee
- Institute of Biophotonics, National Yang-Ming University, Taipei 11221, Taiwan ; Biophotonics & Molecular Imaging Research Center (BMIRC), National Yang-Ming University, Taipei 11221, Taiwan ; Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Yi-Chung Tung
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|
32
|
Luongo K, Holton A, Kaushik A, Spence P, Ng B, Deschenes R, Sundaram S, Bhansali S. Microfluidic device for trapping and monitoring three dimensional multicell spheroids using electrical impedance spectroscopy. BIOMICROFLUIDICS 2013; 7:34108. [PMID: 24404028 PMCID: PMC3689825 DOI: 10.1063/1.4809590] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 05/22/2013] [Indexed: 05/11/2023]
Abstract
In this paper, we report the design, fabrication, and testing of a lab-on-a-chip based microfluidic device for application of trapping and measuring the dielectric properties of microtumors over time using electrical impedance spectroscopy (EIS). Microelectromechanical system (MEMS) techniques were used to embed opposing electrodes onto the top and bottom surfaces of a microfluidic channel fabricated using Pyrex substrate, chrome gold, SU-8, and polydimethylsiloxane. Differing concentrations of cell culture medium, differing sized polystyrene beads, and MCF-7 microtumor spheroids were used to validate the designs ability to detect background conductivity changes and dielectric particle diameter changes between electrodes. The observed changes in cell medium concentrations demonstrated a linear relation to extracted solution resistance (Rs), while polystyrene beads and multicell spheroids induced changes in magnitude consistent with diameter increase. This design permits optical correlation between electrical measurements and EIS spectra.
Collapse
Affiliation(s)
- Kevin Luongo
- BioMEMs and Microfabrication system Laboratory, Department of Electrical and Computer Engineering, Florida International University, Miami, Florida 33172, USA ; Bioengineering Center, Draper Laboratory, Tampa, Florida 33612, USA ; Electrical Engineering, University of South Florida, Tampa, Florida 33620, USA
| | - Angela Holton
- Bioengineering Center, Draper Laboratory, Tampa, Florida 33612, USA
| | - Ajeet Kaushik
- BioMEMs and Microfabrication system Laboratory, Department of Electrical and Computer Engineering, Florida International University, Miami, Florida 33172, USA
| | - Paige Spence
- Bioengineering Center, Draper Laboratory, Tampa, Florida 33612, USA
| | - Beng Ng
- Department of Molecular Medicine, University of South Florida, Tampa, Florida 33620, USA
| | - Robert Deschenes
- Department of Molecular Medicine, University of South Florida, Tampa, Florida 33620, USA
| | - Shankar Sundaram
- Bioengineering Center, Draper Laboratory, Tampa, Florida 33612, USA
| | - Shekhar Bhansali
- BioMEMs and Microfabrication system Laboratory, Department of Electrical and Computer Engineering, Florida International University, Miami, Florida 33172, USA
| |
Collapse
|
33
|
Kim T, Doh I, Cho YH. On-chip three-dimensional tumor spheroid formation and pump-less perfusion culture using gravity-driven cell aggregation and balanced droplet dispensing. BIOMICROFLUIDICS 2012; 6:34107. [PMID: 23882300 PMCID: PMC3416850 DOI: 10.1063/1.4739460] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 07/12/2012] [Indexed: 05/19/2023]
Abstract
This paper presents a spheroid chip in which three-dimensional (3D) tumor spheroids are not only formed by gravity-driven cell aggregation but also cultured at the perfusion rates controlled by balanced droplet dispensing without fluidic pumps. The previous spheroid chips require additional off-chip processes of spheroid formation and extraction as well as bulky components of fluidic pumps. However, the present spheroid chip, where autonomous medium droplet dispensers are integrated on a well array, achieves the on-chip 3D tumor spheroid formation and perfusion culture using simple structure without bulky fluidic pumps. In the experimental study, we demonstrated that the spheroid chip successfully forms 3D tumor spheroids in the wide diameter range of 220 μm-3.2 mm (uniformity > 90%) using H358, H23, and A549 non-small cell lung cancer cells. At the pump-less perfusion culture (Q = 0.1-0.3 μl/min) of spheroids, the number of H358 cells in the spheroid increased up to 50% from the static culture (Q = 0 μl/min) and the viability of the cultured cells also increased about 10%. Therefore, we experimentally verified that the perfusion environment created by the spheroid chip offers a favourable condition to the spheroids with high increase rate and viability. The present chip achieves on-chip 3D tumor spheroid formation and pump-less perfusion culture with simple structure, thereby exhibiting potential for use in integrated in-vivo-like cell culture systems.
Collapse
Affiliation(s)
- Taeyoon Kim
- Cell Bench Research Center, KAIST, Daejeon, South Korea
| | | | | |
Collapse
|
34
|
Controlled formation of heterotypic hepatic micro-organoids in anisotropic hydrogel microfibers for long-term preservation of liver-specific functions. Biomaterials 2012; 33:8304-15. [PMID: 22906609 DOI: 10.1016/j.biomaterials.2012.07.068] [Citation(s) in RCA: 165] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 07/31/2012] [Indexed: 12/25/2022]
Abstract
We have developed a hydrogel-based cell cultivation platform for forming 3D restiform hepatic micro-organoids consisting of primary rat hepatocytes and feeder cells (Swiss 3T3 cells). Sodium alginate solutions containing hepatocytes/3T3 cells were continuously introduced into a microfluidic channel to produce cell-incorporating anisotropic Ba-alginate hydrogel microfibers, where hepatocytes at the center were closely sandwiched by 3T3 cells. Hydrogel fiber-based cultivation under high oxygen tension enabled the formation of heterotypic micro-organoids with a length of up to 1 mm and a diameter of ∼50 μm, mimicking the hepatic cord structures found in the liver, while maintaining a high hepatocyte viability (∼80%) over 30 days. Long-term observation of up to 90 days revealed a significant enhancement of hepatic functions because of heterotypic and homotypic cell-cell interactions, including albumin secretion and urea synthesis as well as expression of hepatocyte-specific genes, compared with conventional monolayer culture and single cultivation in the hydrogel fibers. The encapsulated hepatic constructs were recovered as scaffold-free micro-organoids by enzymatically digesting the hydrogel matrices using alginate lyase. This technique for creating heterotypic micro-organoids with precisely ordered multiple cell types will be useful for the development of a new liver tissue engineering approach and may be applicable to the fabrication of extracorporeal bioartificial liver (BAL) devices and assessment tools for drug development and testing.
Collapse
|
35
|
Skardal A, Smith L, Bharadwaj S, Atala A, Soker S, Zhang Y. Tissue specific synthetic ECM hydrogels for 3-D in vitro maintenance of hepatocyte function. Biomaterials 2012; 33:4565-75. [PMID: 22475531 DOI: 10.1016/j.biomaterials.2012.03.034] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 03/10/2012] [Indexed: 01/19/2023]
Abstract
Despite recent advances in biomaterial science, there is yet no culture system that supports long-term culture expansion of human adult hepatocytes, while preserving continued function. Previous studies suggested that acellular liver extracellular matrix (ECM), employed as a substrate, improved proliferation and function of liver cells. Here we investigated whether extracts prepared from acellular liver ECM (liver ECM extract, LEE), or from whole (fresh) liver tissue (liver tissue extract, LTE), could be combined with collagen Type I, hyaluronic acid (HA), or heparin-conjugated HA (HP) hydrogels to enhance survival and functional output of primary human hepatocytes. The liver-specific semi-synthetic ECMs (sECMs) were prepared by incorporating LEE or LTE into the gel matrices. Subsequently, primary human hepatocytes were maintained in sandwich-style hydrogel cultures for 4 weeks. Progressive increase in hepatocyte metabolism was observed in all HA and HP groups. Hepatocytes cultured in HA and HP hydrogels containing LEE or LTE synthesized and secreted steady levels of albumin and urea and sustained cytochrome p450-dependent drug metabolism of ethoxycoumarin. Collectively, these results indicate that customized HA hydrogels with liver-specific ECM components may be an efficient method for expansion human hepatocytes in vitro for cell therapy and drug and toxicology screening purposes.
Collapse
Affiliation(s)
- Aleksander Skardal
- Wake Forest Institute for Regenerative Medicine, 391 Technology Way, Winston-Salem, NC 27101, USA
| | | | | | | | | | | |
Collapse
|
36
|
Dokmeci MR, Khademhosseini A. Preface to Special Topic: Microfluidics in cell biology and tissue engineering. BIOMICROFLUIDICS 2011; 5:22101. [PMID: 21799707 PMCID: PMC3145226 DOI: 10.1063/1.3594781] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Indexed: 05/31/2023]
Abstract
In this special issue of Biomicrofluidics, a wide variety of applications of microfluidics to tissue engineering and cell biology are presented. The articles illustrate the benefits of using microfluidics for controlling the cellular environment in a precise yet high rate manner using minimum reagents. The topic is very timely and takes a stab at portraying a glimpse of what is to come in this exciting and emerging field of research.
Collapse
|