1
|
Lee J, Jung S, Hong HK, Jo H, Rhee S, Jeong YL, Ko J, Cho YB, Jeon NL. Vascularized tissue on mesh-assisted platform (VT-MAP): a novel approach for diverse organoid size culture and tailored cancer drug response analysis. LAB ON A CHIP 2024; 24:2208-2223. [PMID: 38533822 DOI: 10.1039/d3lc01055d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
This study presents the vascularized tissue on mesh-assisted platform (VT-MAP), a novel microfluidic in vitro model that uses an open microfluidic principle for cultivating vascularized organoids. Addressing the gap in 3D high-throughput platforms for drug response analysis, the VT-MAP can host tumor clusters of various sizes, allowing for precise, size-dependent drug interaction assessments. Key features include capability for forming versatile co-culture conditions (EC, fibroblasts and colon cancer organoids) that enhance tumor organoid viability and a perfusable vessel network that ensures efficient drug delivery and maintenance of organoid health. The VT-MAP enables the culture and analysis of organoids across a diverse size spectrum, from tens of microns to several millimeters. The VT-MAP addresses the inconsistencies in traditional organoid testing related to organoid size, which significantly impacts drug response and viability. Its ability to handle various organoid sizes leads to results that more accurately reflect patient-derived xenograft (PDX) models and differ markedly from traditional in vitro well plate-based methods. We introduce a novel image analysis algorithm that allows for quantitative analysis of organoid size-dependent drug responses, marking a significant step forward in replicating PDX models. The PDX sample from a positive responder exhibited a significant reduction in cell viability across all organoid sizes when exposed to chemotherapeutic agents (5-FU, oxaliplatin, and irinotecan), as expected for cytotoxic drugs. In sharp contrast, PDX samples of a negative responder showed little to no change in viability in smaller clusters and only a slight reduction in larger clusters. This differential response, accurately replicated in the VT-MAP, underscores its ability to generate data that align with PDX models and in vivo findings. Its capacity to handle various organoid sizes leads to results that more accurately reflect PDX models and differ markedly from traditional in vitro methods. The platform's distinct advantage lies in demonstrating how organoid size can critically influence drug response, revealing insights into cancer biology previously unattainable with conventional techniques.
Collapse
Affiliation(s)
- Jungseub Lee
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea.
| | - Sangmin Jung
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea.
| | - Hye Kyoung Hong
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Hyeonsu Jo
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea.
| | - Stephen Rhee
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea.
| | - Ye-Lin Jeong
- Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Jihoon Ko
- Department of Bionano Technology, Gachon University, Seoul, Republic of Korea
| | - Yong Beom Cho
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea.
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Seoul, Republic of Korea
| | - Noo Li Jeon
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea.
- Institute of Advanced Machines and Design, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
2
|
Park W, Kim EM, Jeon Y, Lee J, Yi J, Jeong J, Kim B, Jeong BG, Kim DR, Kong H, Lee CH. Transparent Intracellular Sensing Platform with Si Needles for Simultaneous Live Imaging. ACS NANO 2023; 17:25014-25026. [PMID: 38059775 DOI: 10.1021/acsnano.3c07527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
Vertically ordered Si needles are of particular interest for long-term intracellular recording owing to their capacity to infiltrate living cells with negligible damage and minimal toxicity. Such intracellular recordings could greatly benefit from simultaneous live cell imaging without disrupting their culture, contributing to an in-depth understanding of cellular function and activity. However, the use of standard live imaging techniques, such as inverted and confocal microscopy, is currently impeded by the opacity of Si wafers, typically employed for fabricating vertical Si needles. Here, we introduce a transparent intracellular sensing platform that combines vertical Si needles with a percolated network of Au-Ag nanowires on a transparent elastomeric substrate. This sensing platform meets all prerequisites for simultaneous intracellular recording and imaging, including electrochemical impedance, optical transparency, mechanical compliance, and cell viability. Proof-of-concept demonstrations of this sensing platform include monitoring electrical potentials in cardiomyocyte cells and in three-dimensionally engineered cardiovascular tissue, all while conducting live imaging with inverted and confocal microscopes. This sensing platform holds wide-ranging potential applications for intracellular research across various disciplines such as neuroscience, cardiology, muscle physiology, and drug screening.
Collapse
Affiliation(s)
- Woohyun Park
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Eun Mi Kim
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Yale Jeon
- School of Mechanical Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Junsang Lee
- School of Mechanical Engineering, Hanyang University, Seoul 04763, Republic of Korea
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Jonghun Yi
- School of Mechanical Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Jinheon Jeong
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Bongjoong Kim
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
- Department of Mechanical and System Design Engineering, Hongik University, Seoul 04066, Republic of Korea
| | - Byeong Guk Jeong
- School of Mechanical Engineering, Hanyang University, Seoul 04763, Republic of Korea
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Dong Rip Kim
- School of Mechanical Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Hyunjoon Kong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Chi Hwan Lee
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
- Department of Materials Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
3
|
Chai XX, Liu J, Yu TY, Zhang G, Sun WJ, Zhou Y, Ren L, Cao HL, Yin DC, Zhang CY. Recent progress of mechanosensitive mechanism on breast cancer. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 185:1-16. [PMID: 37793504 DOI: 10.1016/j.pbiomolbio.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/10/2023] [Accepted: 09/25/2023] [Indexed: 10/06/2023]
Abstract
The mechanical environment is important for tumorigenesis and progression. Tumor cells can sense mechanical signals by mechanosensitive receptors, and these mechanical signals can be converted to biochemical signals to regulate cell behaviors, such as cell differentiation, proliferation, migration, apoptosis, and drug resistance. Here, we summarized the effects of the mechanical microenvironment on breast cancer cell activity, and mechanotransduction mechanism from cellular microenvironment to cell membrane, and finally to the nucleus, and also relative mechanosensitive proteins, ion channels, and signaling pathways were elaborated, therefore the mechanical signal could be transduced to biochemical or molecular signal. Meanwhile, the mechanical models commonly used for biomechanics study in vitro and some quantitative descriptions were listed. It provided an essential theoretical basis for the occurrence and development of mechanosensitive breast cancer, and also some potential drug targets were proposed to treat such disease.
Collapse
Affiliation(s)
- Xiao-Xia Chai
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Jie Liu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Tong-Yao Yu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Ge Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Wen-Jun Sun
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Yan Zhou
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Li Ren
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China; Key Laboratory of Flexible Electronics of Zhejiang Province, Ningbo Institute of Northwestern Polytechnical University, Ningbo, 315103, Zhejiang, PR China
| | - Hui-Ling Cao
- Xi'an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, School of Pharmacy, Xi'an Medical University, Xi'an, 710021, Shaanxi, PR China.
| | - Da-Chuan Yin
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China.
| | - Chen-Yan Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China.
| |
Collapse
|
4
|
Slay EE, Meldrum FC, Pensabene V, Amer MH. Embracing Mechanobiology in Next Generation Organ-On-A-Chip Models of Bone Metastasis. FRONTIERS IN MEDICAL TECHNOLOGY 2021; 3:722501. [PMID: 35047952 PMCID: PMC8757701 DOI: 10.3389/fmedt.2021.722501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/09/2021] [Indexed: 12/31/2022] Open
Abstract
Bone metastasis in breast cancer is associated with high mortality. Biomechanical cues presented by the extracellular matrix play a vital role in driving cancer metastasis. The lack of in vitro models that recapitulate the mechanical aspects of the in vivo microenvironment hinders the development of novel targeted therapies. Organ-on-a-chip (OOAC) platforms have recently emerged as a new generation of in vitro models that can mimic cell-cell interactions, enable control over fluid flow and allow the introduction of mechanical cues. Biomaterials used within OOAC platforms can determine the physical microenvironment that cells reside in and affect their behavior, adhesion, and localization. Refining the design of OOAC platforms to recreate microenvironmental regulation of metastasis and probe cell-matrix interactions will advance our understanding of breast cancer metastasis and support the development of next-generation metastasis-on-a-chip platforms. In this mini-review, we discuss the role of mechanobiology on the behavior of breast cancer and bone-residing cells, summarize the current capabilities of OOAC platforms for modeling breast cancer metastasis to bone, and highlight design opportunities offered by the incorporation of mechanobiological cues in these platforms.
Collapse
Affiliation(s)
- Ellen E. Slay
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | | | - Virginia Pensabene
- School of School of Electronic and Electrical Engineering, University of Leeds, Leeds, United Kingdom
- School of Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | - Mahetab H. Amer
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
5
|
Li W, Li P, Li N, Du Y, Lü S, Elad D, Long M. Matrix stiffness and shear stresses modulate hepatocyte functions in a fibrotic liver sinusoidal model. Am J Physiol Gastrointest Liver Physiol 2021; 320:G272-G282. [PMID: 33296275 PMCID: PMC8609567 DOI: 10.1152/ajpgi.00379.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Extracellular matrix (ECM) rigidity has important effects on cell behaviors and increases sharply in liver fibrosis and cirrhosis. Hepatic blood flow is essential in maintaining hepatocytes' (HCs) functions. However, it is still unclear how matrix stiffness and shear stresses orchestrate HC phenotype in concert. A fibrotic three-dimensional (3-D) liver sinusoidal model is constructed using a porous membrane sandwiched between two polydimethylsiloxane (PDMS) layers with respective flow channels. The HCs are cultured in collagen gels of various stiffnesses in the lower channel, whereas the upper channel is pre-seeded with liver sinusoidal endothelial cells (LSECs) and accessible to shear flow. The results reveal that HCs cultured within stiffer matrices exhibit reduced albumin production and cytochrome P450 (CYP450) reductase expression. Low shear stresses enhance synthetic and metabolic functions of HC, whereas high shear stresses lead to the loss of HC phenotype. Furthermore, both mechanical factors regulate HC functions by complementing each other. These observations are likely attributed to mechanically induced mass transport or key signaling molecule of hepatocyte nuclear factor 4α (HNF4α). The present study results provide an insight into understanding the mechanisms of HC dysfunction in liver fibrosis and cirrhosis, especially from the viewpoint of matrix stiffness and blood flow.NEW & NOTEWORTHY A fibrotic three-dimensional (3-D) liver sinusoidal model was constructed to mimic different stages of liver fibrosis in vivo and to explore the cooperative effects of matrix stiffness and shear stresses on hepatocyte (HC) functions. Mechanically induced alterations of mass transport mainly contributed to HC functions via typical mechanosensitive signaling.
Collapse
Affiliation(s)
- Wang Li
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Peiwen Li
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Ning Li
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Yu Du
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Shouqin Lü
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - David Elad
- 5Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Mian Long
- 1Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,2Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,3Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, People’s Republic of China,4School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| |
Collapse
|
6
|
Thai VL, Griffin KH, Thorpe SW, Randall RL, Leach JK. Tissue engineered platforms for studying primary and metastatic neoplasm behavior in bone. J Biomech 2021; 115:110189. [PMID: 33385867 PMCID: PMC7855491 DOI: 10.1016/j.jbiomech.2020.110189] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/02/2020] [Accepted: 12/11/2020] [Indexed: 12/19/2022]
Abstract
Cancer is the second leading cause of death in the United States, claiming more than 560,000 lives each year. Osteosarcoma (OS) is the most common primary malignant tumor of bone in children and young adults, while bone is a common site of metastasis for tumors initiating from other tissues. The heterogeneity, continual evolution, and complexity of this disease at different stages of tumor progression drives a critical need for physiologically relevant models that capture the dynamic cancer microenvironment and advance chemotherapy techniques. Monolayer cultures have been favored for cell-based research for decades due to their simplicity and scalability. However, the nature of these models makes it impossible to fully describe the biomechanical and biochemical cues present in 3-dimensional (3D) microenvironments, such as ECM stiffness, degradability, surface topography, and adhesivity. Biomaterials have emerged as valuable tools to model the behavior of various cancers by creating highly tunable 3D systems for studying neoplasm behavior, screening chemotherapeutic drugs, and developing novel treatment delivery techniques. This review highlights the recent application of biomaterials toward the development of tumor models, details methods for their tunability, and discusses the clinical and therapeutic applications of these systems.
Collapse
Affiliation(s)
- Victoria L Thai
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, United States
| | - Katherine H Griffin
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, United States; School of Veterinary Medicine, University of California, Davis, Davis, CA 95616, United States
| | - Steven W Thorpe
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA 95817, United States
| | - R Lor Randall
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA 95817, United States
| | - J Kent Leach
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, United States; Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA 95817, United States.
| |
Collapse
|
7
|
Sullivan KM, Park CG, Ito JD, Kandel M, Popescu G, Kim YJ, Kong H. Matrix Softness-Mediated 3D Zebrafish Hepatocyte Modulates Response to Endocrine Disrupting Chemicals. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:13797-13806. [PMID: 32975940 PMCID: PMC8202163 DOI: 10.1021/acs.est.0c01988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Endocrine disrupting chemicals (EDC) include synthetic compounds that mimic the structure or function of natural hormones. While most studies utilize live embryos or primary cells from adult fish, these cells rapidly lose functionality when cultured on plastic or glass substrates coated with extracellular matrix proteins. This study hypothesizes that the softness of a matrix with adhered fish cells can regulate the intercellular organization and physiological function of engineered hepatoids during EDC exposure. We scrutinized this hypothesis by culturing zebrafish hepatocytes (ZF-L) on collagen-based hydrogels with controlled elastic moduli by examining morphology, urea production, and intracellular oxidative stress of hepatoids exposed to 17β-estradiol. Interestingly, the softer gel drove cells to form a cell sheet with a canaliculi-like structure compared to its stiffer gel counterpart. The hepatoids cultured on the softer gel exhibited more active urea production upon exposure to 17β-estradiol and displayed faster recovery of intracellular reactive oxygen species level confirmed by gradient light interference microscopy (GLIM), a live-cell imaging technique. These results are broadly useful to improve screening and understanding of potential EDC impacts on aquatic organisms and human health.
Collapse
Affiliation(s)
- Kathryn M Sullivan
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Institute of Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Chang Gyun Park
- Environmental Safety Group, Korea Institute of Science and Technology (KIST) Europe, Campus E 7.1, 66123 Saarbrücken, Germany
| | - John D Ito
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Mikhail Kandel
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Gabriel Popescu
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Young Jun Kim
- Environmental Safety Group, Korea Institute of Science and Technology (KIST) Europe, Campus E 7.1, 66123 Saarbrücken, Germany
| | - Hyunjoon Kong
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Institute of Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
8
|
Liu H, Wang Y, Cui K, Guo Y, Zhang X, Qin J. Advances in Hydrogels in Organoids and Organs-on-a-Chip. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1902042. [PMID: 31282047 DOI: 10.1002/adma.201902042] [Citation(s) in RCA: 202] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/25/2019] [Indexed: 05/10/2023]
Abstract
Significant advances in materials, microscale technology, and stem cell biology have enabled the construction of 3D tissues and organs, which will ultimately lead to more effective diagnostics and therapy. Organoids and organs-on-a-chip (OOC), evolved from developmental biology and bioengineering principles, have emerged as major technological breakthrough and distinct model systems to revolutionize biomedical research and drug discovery by recapitulating the key structural and functional complexity of human organs in vitro. There is growing interest in the development of functional biomaterials, especially hydrogels, for utilization in these promising systems to build more physiologically relevant 3D tissues with defined properties. The remarkable properties of defined hydrogels as proper extracellular matrix that can instruct cellular behaviors are presented. The recent trend where functional hydrogels are integrated into organoids and OOC systems for the construction of 3D tissue models is highlighted. Future opportunities and perspectives in the development of advanced hydrogels toward accelerating organoids and OOC research in biomedical applications are also discussed.
Collapse
Affiliation(s)
- Haitao Liu
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yaqing Wang
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kangli Cui
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yaqiong Guo
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xu Zhang
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Jianhua Qin
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| |
Collapse
|
9
|
Feng X, Dixon H, Glen‐Ravenhill H, Karaosmanoglu S, Li Q, Yan L, Chen X. Smart Nanotechnologies to Target Tumor with Deep Penetration Depth for Efficient Cancer Treatment and Imaging. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900093] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Xue Feng
- School of EngineeringInstitute for BioengineeringThe University of Edinburgh King's Buildings, Mayfield Road Edinburgh EH9 3JL UK
| | - Hannah Dixon
- School of EngineeringInstitute for BioengineeringThe University of Edinburgh King's Buildings, Mayfield Road Edinburgh EH9 3JL UK
| | - Harriet Glen‐Ravenhill
- School of EngineeringInstitute for BioengineeringThe University of Edinburgh King's Buildings, Mayfield Road Edinburgh EH9 3JL UK
| | - Sena Karaosmanoglu
- School of EngineeringInstitute for BioengineeringThe University of Edinburgh King's Buildings, Mayfield Road Edinburgh EH9 3JL UK
| | - Quan Li
- School of EngineeringInstitute for Energy SystemsThe University of Edinburgh King's Buildings, Mayfield Road Edinburgh EH9 3JL UK
| | - Li Yan
- Monash Institute of Pharmaceutical SciencesMonash University Parkville Victoria 3052 Australia
| | - Xianfeng Chen
- School of EngineeringInstitute for BioengineeringThe University of Edinburgh King's Buildings, Mayfield Road Edinburgh EH9 3JL UK
- Translational Medicine CenterThe Second Affiliated HospitalGuangzhou Medical University Guangzhou 510182 P. R. China
| |
Collapse
|
10
|
Ozcelikkale A, Moon HR, Linnes M, Han B. In vitro microfluidic models of tumor microenvironment to screen transport of drugs and nanoparticles. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 9:10.1002/wnan.1460. [PMID: 28198106 PMCID: PMC5555839 DOI: 10.1002/wnan.1460] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 11/14/2016] [Accepted: 12/17/2016] [Indexed: 12/16/2022]
Abstract
Advances in nanotechnology have enabled numerous types of nanoparticles (NPs) to improve drug delivery to tumors. While many NP systems have been proposed, their clinical translation has been less than anticipated primarily due to failure of current preclinical evaluation techniques to adequately model the complex interactions between the NP and physiological barriers of tumor microenvironment. This review focuses on microfluidic tumor models for characterization of delivery efficacy and toxicity of cancer nanomedicine. Microfluidics offer significant advantages over traditional macroscale cell cultures by enabling recapitulation of tumor microenvironment through precise control of physiological cues such as hydrostatic pressure, shear stress, oxygen, and nutrient gradients. Microfluidic systems have recently started to be adapted for screening of drugs and NPs under physiologically relevant settings. So far the two primary application areas of microfluidics in this area have been high-throughput screening using traditional culture settings such as single cells or multicellular tumor spheroids, and mimicry of tumor microenvironment for study of cancer-related cell-cell and cell-matrix interactions. These microfluidic technologies are also useful in modeling specific steps in NP delivery to tumor and characterize NP transport properties and outcomes by systematic variation of physiological conditions. Ultimately, it will be possible to design drug-screening platforms uniquely tailored for individual patient physiology using microfluidics. These in vitro models can contribute to development of precision medicine by enabling rapid and patient-specific evaluation of cancer nanomedicine. WIREs Nanomed Nanobiotechnol 2017, 9:e1460. doi: 10.1002/wnan.1460 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Altug Ozcelikkale
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Hye-ran Moon
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Michael Linnes
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Bumsoo Han
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA,
| |
Collapse
|
11
|
Wan Q, TruongVo T, Steele HE, Ozcelikkale A, Han B, Wang Y, Oh J, Yokota H, Na S. Subcellular domain-dependent molecular hierarchy of SFK and FAK in mechanotransduction and cytokine signaling. Sci Rep 2017; 7:9033. [PMID: 28831165 PMCID: PMC5567257 DOI: 10.1038/s41598-017-09495-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/26/2017] [Indexed: 01/23/2023] Open
Abstract
Focal adhesion kinase (FAK) and Src family kinases (SFK) are known to play critical roles in mechanotransduction and other crucial cell functions. Recent reports indicate that they reside in different microdomains of the plasma membrane. However, little is known about their subcellular domain-dependent roles and responses to extracellular stimuli. Here, we employed fluorescence resonance energy transfer (FRET)-based biosensors in conjunction with collagen-coupled agarose gels to detect subcellular activities of SFK and FAK in three-dimensional (3D) settings. We observed that SFK and FAK in the lipid rafts and nonrafts are differently regulated by fluid flow and pro-inflammatory cytokines. Inhibition of FAK in the lipid rafts blocked SFK response to fluid flow, while inhibition of SFK in the non-rafts blocked FAK activation by the cytokines. Ex-vivo FRET imaging of mouse cartilage explants showed that intermediate level of interstitial fluid flow selectively decreased cytokine-induced SFK/FAK activation. These findings suggest that SFK and FAK exert distinctive molecular hierarchy depending on their subcellular location and extracellular stimuli.
Collapse
Affiliation(s)
- Qiaoqiao Wan
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, 46202, USA
- School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, 47907, USA
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, 02115, USA
| | - ThucNhi TruongVo
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, 46202, USA
| | - Hannah E Steele
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, 46202, USA
| | - Altug Ozcelikkale
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana, 47907, USA
| | - Bumsoo Han
- School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, 47907, USA
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana, 47907, USA
| | - Yingxiao Wang
- Department of Bioengineering, University of California San Diego, La Jolla, California, 92093, USA
| | - Junghwan Oh
- Department of Biomedical Engineering, Pukyong National University, Busan, 48513, Republic of Korea
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, 46202, USA
- School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, 47907, USA
| | - Sungsoo Na
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, 46202, USA.
- School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, 47907, USA.
| |
Collapse
|
12
|
Rich MH, Lee MK, Ballance WC, Boppart M, Kong H. Poly(ethylene glycol)-Mediated Collagen Gel Mechanics Regulates Cellular Phenotypes in a Microchanneled Matrix. Biomacromolecules 2017. [DOI: 10.1021/acs.biomac.7b00476] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Max H. Rich
- Department of Chemical and Biomolecular Engineering, ‡Institute for Genomic Biology, §Department of Kinesiology, and ∥Beckman Institute, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Min Kyung Lee
- Department of Chemical and Biomolecular Engineering, ‡Institute for Genomic Biology, §Department of Kinesiology, and ∥Beckman Institute, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - William C. Ballance
- Department of Chemical and Biomolecular Engineering, ‡Institute for Genomic Biology, §Department of Kinesiology, and ∥Beckman Institute, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Marni Boppart
- Department of Chemical and Biomolecular Engineering, ‡Institute for Genomic Biology, §Department of Kinesiology, and ∥Beckman Institute, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Hyunjoon Kong
- Department of Chemical and Biomolecular Engineering, ‡Institute for Genomic Biology, §Department of Kinesiology, and ∥Beckman Institute, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|