1
|
Watanabe M, Salvadori A, Markovic M, Sudo R, Ovsianikov A. Advanced liver-on-chip model mimicking hepatic lobule with continuous microvascular network via high-definition laser patterning. Mater Today Bio 2025; 32:101643. [PMID: 40206147 PMCID: PMC11979415 DOI: 10.1016/j.mtbio.2025.101643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/09/2024] [Accepted: 03/06/2025] [Indexed: 04/11/2025] Open
Abstract
There is a great demand for development of advanced in vitro liver models to predict the efficacy and safety of drug candidates accurately in the preclinical drug development. Despite the great efforts to develop biomimetic models, it remains challenging to precisely mimic a functional unit of the liver (i.e., hepatic lobule) with a continuous microvascular network. Recent progress in laser patterning has allowed us to create arbitrary biomimetic structures with high resolution. Here, we propose an advanced liver-on-chip model mimicking the hepatic lobule with a continuous microvascular network, ranging from the microvessels to the central vein of the liver, utilizing femtosecond laser patterning. Firstly, we optimize the laser power to pattern microchannels mimicking the microvessel and central vein of the hepatic lobule by using a femtosecond laser within a collagen-based hydrogel containing hepatic cells. Secondly, we construct continuous microvessels with luminal structures by comparing different microchannel sizes in diameter. Finally, we assemble a millimeter-scale hepatic lobule-like structure with multiple layers of microvascular networks in the liver-on-chip. Furthermore, our liver-on-chip model exhibits major liver functions and drug-induced hepatotoxicity, as evidenced by albumin and urea productions and by a toxic response to acetaminophen, respectively. Our approach provides valuable strategies for the development of advanced physiological and pathological liver-on-chip models for pharmaceutical and toxicological studies.
Collapse
Affiliation(s)
- Masafumi Watanabe
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, Technische Universität Wien (TU Wien), 1060 Vienna, Austria
- Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Austria
- Japan Society for the Promotion of Science (JSPS) Overseas Research Fellow, Japan
| | - Alice Salvadori
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, Technische Universität Wien (TU Wien), 1060 Vienna, Austria
- Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Austria
| | - Marica Markovic
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, Technische Universität Wien (TU Wien), 1060 Vienna, Austria
- Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Austria
| | - Ryo Sudo
- Department of System Design Engineering, Keio University, 223-8522 Yokohama, Japan
| | - Aleksandr Ovsianikov
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, Technische Universität Wien (TU Wien), 1060 Vienna, Austria
- Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Austria
| |
Collapse
|
2
|
Lang A, Eastburn EA, Younesi M, Nijsure MP, Siciliano C, Pranatharthi Haran A, Panebianco CJ, Seidl E, Tang R, Alsberg E, Willett NJ, Gottardi R, Huh D, Boerckel JD. CYR61 delivery promotes angiogenesis during bone fracture repair. NPJ Regen Med 2025; 10:20. [PMID: 40263309 PMCID: PMC12015299 DOI: 10.1038/s41536-025-00398-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 01/31/2025] [Indexed: 04/24/2025] Open
Abstract
Compromised vascular supply and insufficient neovascularization impede bone repair, increasing risk of non-union. CYR61, Cysteine-rich angiogenic inducer of 61kD (also known as CCN1), is a matricellular growth factor that has been implicated in fracture repair. Here, we map the distribution of endogenous CYR61 during bone repair and evaluate the effects of recombinant CYR61 delivery on vascularized bone regeneration. In vitro, CYR61 treatment did not alter chondrogenesis or osteogenic gene expression, but significantly enhanced angiogenesis. In a mouse femoral fracture model, CYR61 delivery did not alter cartilage or bone formation, but accelerated neovascularization during fracture repair. Early initiation of ambulatory mechanical loading disrupted CYR61-induced neovascularization. Together, these data indicate that CYR61 delivery can enhance angiogenesis during bone repair, particularly for fractures with stable fixation, and may have therapeutic potential for fractures with limited blood vessel supply.
Collapse
Affiliation(s)
- Annemarie Lang
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Emily A Eastburn
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Mousa Younesi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Madhura P Nijsure
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Carly Siciliano
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Annapurna Pranatharthi Haran
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Elizabeth Seidl
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Rui Tang
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Eben Alsberg
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, USA
- Jesse Brown Veterans Affairs Medical Center (JBVAMC), Chicago, IL, USA
| | - Nick J Willett
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, USA
- The Veterans Affairs Portland Health Care System, Portland, OR, USA
| | - Riccardo Gottardi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Joel D Boerckel
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Tiskratok W, Chuinsiri N, Limraksasin P, Kyawsoewin M, Jitprasertwong P. Extracellular Matrix Stiffness: Mechanotransduction and Mechanobiological Response-Driven Strategies for Biomedical Applications Targeting Fibroblast Inflammation. Polymers (Basel) 2025; 17:822. [PMID: 40292716 PMCID: PMC11946729 DOI: 10.3390/polym17060822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
The extracellular matrix (ECM) is a dynamic network providing mechanical and biochemical cues that regulate cellular behavior. ECM stiffness critically influences fibroblasts, the primary ECM producers, particularly in inflammation and fibrosis. This review explores the role of ECM stiffness in fibroblast-driven inflammation and tissue remodeling, focusing on the physicochemical and biological mechanisms involved. Engineered materials, hydrogels, and polydimethylsiloxane (PDMS) are highlighted for replicating tissue-specific stiffness, enabling precise control over cell-matrix interactions. The surface functionalization of substrate materials, including collagen, polydopamine, and fibronectin, enhances bioactivity and fibroblast adhesion. Key mechanotransduction pathways, such as integrin signaling and YAP/TAZ activation, are related to regulating fibroblast behaviors and inflammatory responses. The role of fibroblasts in driving chronic inflammatory diseases emphasizes their therapeutic potentials. Advances in ECM-modifying strategies, including tunable biomaterials and hydrogel-based therapies, are explored for applications in tissue engineering, drug delivery, anti-inflammatory treatments, and diagnostic tools for the accurate diagnosis and prognosis of ECM stiffness-related inflammatory diseases. This review integrates mechanobiology with biomedical innovations, providing a comprehensive prognosis of fibroblast responses to ECM stiffness and outlining future directions for targeted therapies.
Collapse
Affiliation(s)
- Watcharaphol Tiskratok
- Institute of Dentistry, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand; (N.C.); (P.J.)
- Oral Health Centre, Suranaree University of Technology Hospital, Nakhon Ratchasima 30000, Thailand
| | - Nontawat Chuinsiri
- Institute of Dentistry, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand; (N.C.); (P.J.)
- Oral Health Centre, Suranaree University of Technology Hospital, Nakhon Ratchasima 30000, Thailand
| | - Phoonsuk Limraksasin
- Center of Excellence for Dental Stem Cell Biology, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (P.L.); (M.K.)
| | - Maythwe Kyawsoewin
- Center of Excellence for Dental Stem Cell Biology, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (P.L.); (M.K.)
| | - Paiboon Jitprasertwong
- Institute of Dentistry, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand; (N.C.); (P.J.)
- Oral Health Centre, Suranaree University of Technology Hospital, Nakhon Ratchasima 30000, Thailand
| |
Collapse
|
4
|
Agarwal SS, Holter JC, Jones TH, Fuller BT, Tinapple JW, Barlage JM, Song JW. A Modular, Cost-Effective, and Pumpless Perfusion Assembly for the Long-Term Culture of Engineered Microvessels. MICROMACHINES 2025; 16:351. [PMID: 40141962 PMCID: PMC11945127 DOI: 10.3390/mi16030351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025]
Abstract
Continuous perfusion is necessary to sustain microphysiological systems and other microfluidic cell cultures. However, most of the established microfluidic perfusion systems, such as syringe pumps, peristaltic pumps, and rocker plates, have several operational challenges and may be cost-prohibitive, especially for laboratories with no microsystems engineering expertise. Here, we address the need for a cost-efficient, easy-to-implement, and reliable microfluidic perfusion system. Our solution is a modular pumpless perfusion assembly (PPA), which is constructed from commercially available, interchangeable, and aseptically packaged syringes and syringe filters. The total cost for the components of each assembled PPA is USD 1-2. The PPA retains the simplicity of gravity-based pumpless flow systems but incorporates high resistance filters that enable slow and sustained flow for extended periods of time (hours to days). The perfusion characteristics of the PPA were determined by theoretical calculations of the total hydraulic resistance of the assembly and experimental characterization of specific filter resistances. We demonstrated that the PPA enabled reliable long-term culture of engineered endothelialized 3-D microvessels for several weeks. Taken together, our novel PPA solution is simply constructed from extremely low-cost and commercially available laboratory supplies and facilitates robust cell culture and compatibility with current microfluidic setups.
Collapse
Affiliation(s)
- Shashwat S. Agarwal
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH 43210, USA; (S.S.A.); (T.H.J.)
| | - Jacob C. Holter
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; (J.C.H.); (B.T.F.); (J.W.T.)
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Travis H. Jones
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH 43210, USA; (S.S.A.); (T.H.J.)
| | - Brendan T. Fuller
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; (J.C.H.); (B.T.F.); (J.W.T.)
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Joseph W. Tinapple
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; (J.C.H.); (B.T.F.); (J.W.T.)
| | - Joseph M. Barlage
- Department of Biomedical Education and Anatomy, The Ohio State University, Columbus, OH 43210, USA;
| | - Jonathan W. Song
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH 43210, USA; (S.S.A.); (T.H.J.)
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
5
|
Donzanti MJ, Ferrick BJ, Mhatre O, Chernokal B, Renteria DC, Gleghorn JP. Stochastic to Deterministic: A Straightforward Approach to Create Serially Perfusable Multiscale Capillary Beds. ACS Biomater Sci Eng 2025; 11:239-248. [PMID: 39606830 DOI: 10.1021/acsbiomaterials.4c01247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Generation of in vitro tissue models with serially perfused hierarchical vasculature would allow greater control of fluid perfusion throughout the network and enable direct mechanistic investigation of vasculogenesis, angiogenesis, and vascular remodeling. In this work, we have developed a method to produce a closed, serially perfused, multiscale vessel network fully embedded within an acellular hydrogel, where flow through the capillary bed is required prior to fluid exit. We confirmed that the acellular and cellular gel-gel interface was functionally annealed without preventing or biasing cell migration and endothelial self-assembly. Multiscale connectivity of the vessel network was validated via high-resolution microscopy techniques to confirm anastomosis between self-assembled and patterned vessels. Lastly, using a simple acrylic cassette and fluorescently labeled microspheres, the multiscale network was demonstrated to be perfusable. Directed flow from inlet to outlet mandated flow through the capillary bed. This method for producing closed, multiscale vascular networks was developed with the intention of straightforward fabrication and engineering techniques so as to be a low barrier to entry for researchers who wish to investigate mechanistic questions in vascular biology. This ease of use offers a facile extension of these methods for incorporation into organoid culture, organ-on-a-chip (OOC) models, and bioprinted tissues.
Collapse
Affiliation(s)
- Michael J Donzanti
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware United States 19713
| | - Bryan J Ferrick
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware United States 19713
| | - Omkar Mhatre
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware United States 19713
| | - Brea Chernokal
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware United States 19713
| | - Diana C Renteria
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware United States 19713
| | - Jason P Gleghorn
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware United States 19713
| |
Collapse
|
6
|
Sriram G, Makkar H. Microfluidic organ-on-chip systems for periodontal research: advances and future directions. Front Bioeng Biotechnol 2025; 12:1490453. [PMID: 39840127 PMCID: PMC11747509 DOI: 10.3389/fbioe.2024.1490453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025] Open
Abstract
Advances in tissue engineering and microfluidic technologies have enabled the development of sophisticated in vitro models known as organ-on-a-chip (OoC) or microphysiological systems. These systems enable to potential to simulate the dynamic interactions between host tissues and their microenvironment including microbes, biomaterials, mechanical forces, pharmaceutical, and consumer-care products. These fluidic technologies are increasingly being utilized to investigate host-microbe and host-material interactions in oral health and disease. Of interest is their application in understanding periodontal disease, a chronic inflammatory condition marked by the progressive destruction of periodontal tissues, including gingiva, periodontal ligament, and alveolar bone. The pathogenesis of periodontal disease involves a complex interplay between microbial dysbiosis and host immune responses, which can lead to a loss of dental support structures and contribute to systemic conditions such as cardiovascular disease, diabetes, and inflammatory bowel disease. This provides a comprehensive overview of the latest developments in millifluidic and microfluidic systems designed to emulate periodontal host-microbe and host-material interactions. We discuss the critical engineering and biological considerations in designing these platforms, their applications in studying oral biofilms, periodontal tissue responses, and their potential to unravel disease mechanisms and therapeutic targets in periodontal disease.
Collapse
Affiliation(s)
- Gopu Sriram
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Hardik Makkar
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
- Center for Innovation & Precision Dentistry, School of Dental Medicine and School of Engineering & Applied Sciences, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
7
|
Song D, Song CW, Cho SH, Kwon TY, Jung H, Park KH, Kim J, Seo J, Yoo J, Kim M, Lee GR, Hwang J, Lee HM, Shin J, Shin JH, Jung YS, Chang J. Highly Tunable, Nanomaterial-Functionalized Structural Templating of Intracellular Protein Structures Within Biological Species. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406492. [PMID: 39535386 PMCID: PMC11727137 DOI: 10.1002/advs.202406492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/15/2024] [Indexed: 11/16/2024]
Abstract
Inside living organisms, proteins are self-assembled into diverse 3D structures optimized for specific functions. This structure-function relationship can be exploited to synthesize functional materials through biotemplating and depositing functional materials onto protein structures. However, conventional biotemplating faces limitations due to the predominantly intracellular existence of proteins and associated challenges in achieving tunability while preserving functionality. In this study, Conversion to Advanced Materials via labeled Biostructures (CamBio), an integrated biotemplating platform that involves labeling target protein structures with antibodies followed by the growth of functional materials, ensuring outstanding nanostructure tunability is proposed. Protein-derived plasmonic nanostructures created by CamBio can serve as precise quantitative tools for assessing target species is demonstrated. The assessment is achieved through highly tunable and efficient surface-enhanced Raman spectroscopy (SERS). CamBio enables the formation of dense nanogap hot spots among metal nanoparticles, templated by diverse fibrous proteins comprising densely repeated monomers. Furthermore, iterative antibody labeling strategies to adjust the antibody density surrounding targets, amplifying the number of nanogaps and consequently improving SERS performance are employed. Finally, cell-patterned substrates and whole meat sections as SERS substrates, confirming their easily accessible, cost-effective, scalable preparation capabilities and dimensional tunability are incorporated.
Collapse
Affiliation(s)
- Dae‐Hyeon Song
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141South Korea
| | - Chang Woo Song
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141South Korea
| | - Seunghee H. Cho
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141South Korea
| | - Tae Yoon Kwon
- Department of Mechanical EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141South Korea
| | - Hoeyun Jung
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141South Korea
| | - Ki Hyun Park
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141South Korea
| | - Jiyun Kim
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141South Korea
| | - Junyoung Seo
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141South Korea
| | - Jaeyoung Yoo
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141South Korea
| | - Minjoon Kim
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141South Korea
| | - Gyu Rac Lee
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141South Korea
| | - Jisung Hwang
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141South Korea
| | - Hyuck Mo Lee
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141South Korea
| | - Jonghwa Shin
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141South Korea
| | - Jennifer H. Shin
- Department of Mechanical EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141South Korea
| | - Yeon Sik Jung
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141South Korea
| | - Jae‐Byum Chang
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141South Korea
- Department of Biological SciencesKorea Advanced Institute of Science and TechnologyDaejeon34141South Korea
- Bioimaging Data Curation CenterSeoul03760South Korea
| |
Collapse
|
8
|
Belgodere J, Benz MC, Kpeli GW, Elliott JR, Elliott S, North JD, Ponder IJ, Ma P, Dietrich SR, Cheng T, Nguyen K, Tilghman SL, McLachlan JA, Zou B, Anbalagan M, Rowan B, Mondrinos M, Wiese TE, Hoang VT, Collins-Burow BM, Martin EC, Burow ME, Boué SM. Context-Dependent Estrogenic Actions of (+)-Pisatin Produced in Elicited Green or Snow Pea ( Pisum sativum). JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:28255-28269. [PMID: 39665386 PMCID: PMC11674159 DOI: 10.1021/acs.jafc.4c06409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 12/13/2024]
Abstract
Legumes are a predominant source of isoflavones, termed phytoestrogens, that mimic 17β-estradiol (E2). Phytoalexins are inducible isoflavones produced in plants subjected to environmental stressors (e.g., UV, heat, or fungi). This study investigated estrogenic activity of snow and green peas elicited with Aspergillus sojae. Elicited extracts increased estrogenic activity and proliferation of breast cancer cells (MCF-7 or T47D) in a dose-dependent manner but exhibited antiestrogenic activity when combined with synthetic E2. HPLC analysis of elicited pea extracts identified (+)-pisatin as the primary phytoalexin, which was produced significantly (p < 0.0001) more in snow pea compared to green pea. RNA sequencing results suggested potential functional effects on endothelial cells and tissue vascularization. Indeed, (+)-pisatin enhanced metrics of network assembly and maturation in a microphysiological model of bulk tissue vasculogenesis. Thus, context-dependent functional effects of (+)-pisatin and pharmacologically similar phytoestrogens on the entire tissue microenvironment should be considered in preclinical investigation as potential therapeutic agents.
Collapse
Affiliation(s)
- Jorge
A. Belgodere
- Tulane Department
of Medicine, Section of Hematology & Medical Oncology, Tulane University Health Science Center, New Orleans, Louisiana 70112, United States
- Department
of Biological and Agricultural Engineering, Louisiana State University and Agricultural Center, Baton Rouge, Louisiana 70803, United States
- Tulane
Cancer
Center, Tulane University, New Orleans, Louisiana 70112, United States
| | - Megan C. Benz
- Tulane Department
of Medicine, Section of Hematology & Medical Oncology, Tulane University Health Science Center, New Orleans, Louisiana 70112, United States
| | - G. Wills Kpeli
- Department
of Biomedical Engineering, Tulane University, New Orleans, Louisiana 70112, United States
| | - Jack R. Elliott
- Tulane Department
of Medicine, Section of Hematology & Medical Oncology, Tulane University Health Science Center, New Orleans, Louisiana 70112, United States
| | - Steven Elliott
- Tulane Department
of Medicine, Section of Hematology & Medical Oncology, Tulane University Health Science Center, New Orleans, Louisiana 70112, United States
| | - Jack D. North
- Tulane Department
of Medicine, Section of Hematology & Medical Oncology, Tulane University Health Science Center, New Orleans, Louisiana 70112, United States
| | - Isaac J. Ponder
- Department
of Biological and Agricultural Engineering, Louisiana State University and Agricultural Center, Baton Rouge, Louisiana 70803, United States
| | - Peng Ma
- Xavier
University
School of Pharmacy, Xavier University, New Orleans, Louisiana 70125, United States
| | - Sophie R. Dietrich
- Tulane Department
of Medicine, Section of Hematology & Medical Oncology, Tulane University Health Science Center, New Orleans, Louisiana 70112, United States
| | - Thomas Cheng
- Tulane Department
of Medicine, Section of Hematology & Medical Oncology, Tulane University Health Science Center, New Orleans, Louisiana 70112, United States
| | - Khoa Nguyen
- Tulane Department
of Medicine, Section of Hematology & Medical Oncology, Tulane University Health Science Center, New Orleans, Louisiana 70112, United States
| | - Syreeta L. Tilghman
- Pharmaceutical
Sciences Division, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307, United States
| | - John A. McLachlan
- Tulane Department
of Medicine, Section of Hematology & Medical Oncology, Tulane University Health Science Center, New Orleans, Louisiana 70112, United States
| | - Binghao Zou
- Department
of Structural and Cellular Biology, Tulane
University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Muralidharan Anbalagan
- Department
of Structural and Cellular Biology, Tulane
University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Brian Rowan
- Department
of Structural and Cellular Biology, Tulane
University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Mark Mondrinos
- Department
of Biomedical Engineering, Tulane University, New Orleans, Louisiana 70112, United States
| | - Thomas E. Wiese
- Xavier
University
School of Pharmacy, Xavier University, New Orleans, Louisiana 70125, United States
| | - Van T. Hoang
- Tulane Department
of Medicine, Section of Hematology & Medical Oncology, Tulane University Health Science Center, New Orleans, Louisiana 70112, United States
- Tulane
Cancer
Center, Tulane University, New Orleans, Louisiana 70112, United States
| | - Bridgette M. Collins-Burow
- Tulane Department
of Medicine, Section of Hematology & Medical Oncology, Tulane University Health Science Center, New Orleans, Louisiana 70112, United States
- Tulane
Cancer
Center, Tulane University, New Orleans, Louisiana 70112, United States
| | - Elizabeth C. Martin
- Tulane Department
of Medicine, Section of Hematology & Medical Oncology, Tulane University Health Science Center, New Orleans, Louisiana 70112, United States
- Tulane
Cancer
Center, Tulane University, New Orleans, Louisiana 70112, United States
| | - Matthew E. Burow
- Tulane Department
of Medicine, Section of Hematology & Medical Oncology, Tulane University Health Science Center, New Orleans, Louisiana 70112, United States
- Tulane
Cancer
Center, Tulane University, New Orleans, Louisiana 70112, United States
| | - Stephen M. Boué
- U.S.
Department
of Agriculture, Agricultural Research Service, Southern Regional Research
Center, New Orleans, Louisiana 70124, United States
| |
Collapse
|
9
|
Zhukova OA, Ozerskaya IV, Basmanov DV, Stolyarov VY, Bogush VG, Kolesov VV, Zykov KA, Yusubalieva GM, Baklaushev VP. “Lung-on-a-chip” as an instrument for studying the pathophysiology of human respiration. КЛИНИЧЕСКАЯ ПРАКТИКА 2024; 15:70-88. [DOI: 10.17816/clinpract637140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2025] Open
Abstract
“Lung-on-a-chip” (LoC) is a microfluidic device, imitating the gas-fluid interface of the pulmonary alveole in the human lung and intended for pathophysiological, pharmacological and molecular-biological studies of the air-blood barrier in vitro. The LoC device itself contains a system of fluid and gas microchannels, separated with a semipermeable elastic membrane, containing a polymer base and the alveolar cell elements. Depending on the type of LoC (single-, double- and three-channel), the membrane may contain only alveolocytes or alveolocytes combined with other cells — endotheliocytes, fibroblasts, alveolar macrophages or tumor cells. Some LoC models also include proteinic or hydrogel stroma, imitating the pulmonary interstitium. The first double-channel LoC variant, in which one side of the membrane contained an alveolocytic monolayer and the other side — a monolayer of endotheliocytes, was developed in 2010 by a group of scientists from the Harvard University for maximally precise in vitro reproduction of the micro-environment and biomechanics operations of the alveoli. Modern LoC modifications include the same elements and differ only by the construction of the microfluidic system, by the biomaterial of semipermeable membrane, by the composition of cellular and stromal elements and by specific tasks to be solved. Besides the LoC imitating the hematoalveolar barrier, there are modifications for studying the specific pathophysiological processes, for the screening of medicinal products, for modeling specific diseases, for example, lung cancer, chronic obstructive pulmonary disease or asthma. In the present review, we have analyzed the existing types of LoC, the biomaterials used, the methods of detecting molecular processes within the microfluidic devices and the main directions of research to be conducted using the “lung-on-a-chip”.
Collapse
Affiliation(s)
- Oksana A. Zhukova
- Pulmonology Scientific Research Institute
- Federal Center of Brain Research and Neurotechnologies
| | | | - Dmitry V. Basmanov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine
| | | | | | | | - Kirill A. Zykov
- Pulmonology Scientific Research Institute
- Russian University of Medicine
| | - Gaukhar M. Yusubalieva
- Federal Center of Brain Research and Neurotechnologies
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies
- Engelhardt Institute of Molecular Biology
| | - Vladimir P. Baklaushev
- Pulmonology Scientific Research Institute
- Federal Center of Brain Research and Neurotechnologies
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies
- Engelhardt Institute of Molecular Biology
| |
Collapse
|
10
|
Jin G, Huang H, Bao X, Palecek SP. Poly(norepinephrine)-Mediated Universal Surface Modification for Patterning Human Pluripotent Stem Cell Culture and Differentiation. ACS Biomater Sci Eng 2024; 10:7429-7440. [PMID: 39548968 PMCID: PMC11959688 DOI: 10.1021/acsbiomaterials.4c01229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2024]
Abstract
Maintaining undifferentiated states of human pluripotent stem cells (hPSCs) is key to accomplishing successful hPSC research. Specific culture conditions, including hPSC-compatible substrates, are required for the hPSC culture. Over the past two decades, substrates supporting hPSC self-renewal have evolved from undefined and xenogeneic protein components to chemically defined and xenogeneic-free materials. However, these synthetic substrates are often costly and complex to use, leading many laboratories to continue using simpler undefined extracellular matrix (ECM) protein mixtures. In this study, we present a method using poly(norepinephrine) (pNE) for surface modification to enhance the immobilization of ECM proteins on various substrates, including polydimethylsiloxane (PDMS) and ultralow attachment (ULA) hydrogels, thereby supporting hPSC culture and maintenance of pluripotency. The pNE-mediated surface modification enables spatial patterning of ECM proteins on nonadhesive ULA surfaces, facilitating tunable macroscopic cell patterning. This approach improves hPSC attachment and growth and allows for cell patterning to study the effects of anisotropic environments on the hPSC fate. Our findings demonstrate the versatility and simplicity of pNE-mediated surface modification for improving hPSC culture and spatially controlled differentiation into endothelial cells and cardiomyocytes on previously nonamenable substrates, providing a valuable tool for tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Gyuhyung Jin
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Purdue University Institute for Cancer Research, West Lafayette, IN, 47907, USA
- Department of Chemical and Biological Engineering, University of Wisconsin Madison, WI, 53706, USA
| | - Haoning Huang
- Purdue University Institute for Cancer Research, West Lafayette, IN, 47907, USA
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Xiaoping Bao
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Purdue University Institute for Cancer Research, West Lafayette, IN, 47907, USA
| | - Sean P. Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin Madison, WI, 53706, USA
| |
Collapse
|
11
|
Xiao H, Sylla K, Gong X, Wilkowski B, Rossello-Martinez A, Jordan SN, Mintah EY, Zheng A, Sun H, Herzog EL, Mak M. Proteolysis and Contractility Regulate Tissue Opening and Wound Healing by Lung Fibroblasts in 3D Microenvironments. Adv Healthc Mater 2024; 13:e2400941. [PMID: 38967294 PMCID: PMC11617280 DOI: 10.1002/adhm.202400941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/17/2024] [Indexed: 07/06/2024]
Abstract
Damage and repair are recurring processes in tissues, with fibroblasts playing key roles by remodeling extracellular matrices (ECM) through protein synthesis, proteolysis, and cell contractility. Dysregulation of fibroblasts can lead to fibrosis and tissue damage, as seen in idiopathic pulmonary fibrosis (IPF). In advanced IPF, tissue damage manifests as honeycombing, or voids in the lungs. This study explores how transforming growth factor-beta (TGF-β), a crucial factor in IPF, induces lung fibroblast spheroids to create voids in reconstituted collagen through proteolysis and cell contractility, a process we termed as hole formation. These voids reduce when proteases are blocked. Spheroids mimic fibroblast foci observed in IPF. Results indicate that cell contractility mediates tissue opening by stretching fractures in the collagen meshwork. Matrix metalloproteinases (MMPs), including MMP1 and MT1-MMP, are essential for hole formation, with invadopodia playing a significant role. Blocking MMPs reduces hole size and promotes wound healing. This study shows how TGF-β induces excessive tissue destruction and how blocking proteolysis can reverse damage, offering insights into IPF pathology and potential therapeutic interventions.
Collapse
Affiliation(s)
- Hugh Xiao
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | - Kadidia Sylla
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | - Xiangyu Gong
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | - Brendan Wilkowski
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | | | - Seyma Nayir Jordan
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | - Emmanuel Y Mintah
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | - Allen Zheng
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | - Huanxing Sun
- Department of Medicine (Pulmonary, Critical Care and Sleep), Yale School of Medicine, New Haven, CT, 06510, USA
| | - Erica L Herzog
- Department of Medicine (Pulmonary, Critical Care and Sleep), Yale School of Medicine, New Haven, CT, 06510, USA
| | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| |
Collapse
|
12
|
Byrne CE, Martier AT, Kpeli GW, Conrad KM, Bralower W, Olsen E, Fortes G, Culp CC, Wendell M, Boone KA, Burow MR, Mondrinos MJ. Adaptable Manufacturing and Biofabrication of Milliscale Organ Chips With Perfusable Vascular Beds. Biotechnol J 2024; 19:e202400550. [PMID: 39711093 DOI: 10.1002/biot.202400550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/09/2024] [Accepted: 12/04/2024] [Indexed: 12/24/2024]
Abstract
Microphysiological systems (MPS) containing perfusable vascular beds unlock the ability to model tissue-scale elements of vascular physiology and disease in vitro. Access to inexpensive stereolithography (SLA) 3D printers now enables benchtop fabrication of polydimethylsiloxane (PDMS) organ chips, eliminating the need for cleanroom access and microfabrication expertise, and can facilitate broader adoption of MPS approaches in preclinical research. Rapid prototyping of organ chip mold designs accelerates the processes of design, testing, and iteration, but geometric distortion and surface roughness of SLA resin prints can impede the development of standardizable manufacturing workflows. This study reports postprocessing procedures for manufacturing SLA-printed molds that produce fully cured, flat, patently bonded, and optically clear polydimethyl siloxane (PDMS) organ chips. Injection loading tests were conducted to identify milliscale membrane-free organ chip (MFOC) designs that allowed reproducible device loading by target end-users, a key requirement for broad nonexpert adoption in preclinical research. The optimized milliscale MFOC design was used to develop tissue engineering protocols for (i) driving bulk tissue vasculogenesis in MFOC, and (ii) seeding the bulk tissue interfaces with a confluent endothelium to stimulate self-assembly of perfusable anastomoses with the internal vasculature. Comparison of rocker- and pump-based protocols for flow-conditioning of anastomosed vascular beds revealed that continuous pump-driven flow is required for reproducible barrier maturation throughout the 3D tissue bulk. Demonstrated applications include nanoparticle perfusion and engineering perfusable tumor vasculature. These easily adaptable methods for designing and fabricating vascularized microphysiological systems can accelerate their adoption in a diverse range of preclinical laboratory settings.
Collapse
Affiliation(s)
| | - Ashley T Martier
- Department of Biomedical Engineering, Tulane University, New Orleans, USA
| | - Gideon Wills Kpeli
- Department of Biomedical Engineering, Tulane University, New Orleans, USA
| | | | - William Bralower
- Department of Biomedical Engineering, Tulane University, New Orleans, USA
| | - Elisabet Olsen
- Bioinnovation Program, Tulane University, New Orleans, USA
| | - Gabrielle Fortes
- Department of Biomedical Engineering, Tulane University, New Orleans, USA
| | - Caroline C Culp
- Department of Biomedical Engineering, Tulane University, New Orleans, USA
| | - Max Wendell
- Department of Biomedical Engineering, Tulane University, New Orleans, USA
| | - Keefer A Boone
- Department of Biomedical Engineering, Tulane University, New Orleans, USA
| | - Matthew R Burow
- Section of Hematology and Oncology, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, USA
| | - Mark J Mondrinos
- Department of Biomedical Engineering, Tulane University, New Orleans, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, USA
- Department of Physiology, Tulane University School of Medicine, New Orleans, USA
| |
Collapse
|
13
|
Agarwal SS, Cortes-Medina M, Holter JC, Avendano A, Tinapple JW, Barlage JM, Menyhert MM, Onua LM, Song JW. Rapid low-cost assembly of modular microvessel-on-a-chip with benchtop xurography. LAB ON A CHIP 2024; 24:5065-5076. [PMID: 39397763 PMCID: PMC11472271 DOI: 10.1039/d4lc00565a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/06/2024] [Indexed: 10/15/2024]
Abstract
Blood and lymphatic vessels in the body are central to molecular and cellular transport, tissue repair, and pathophysiology. Several approaches have been employed for engineering microfabricated blood and lymphatic vessels in vitro, yet traditionally these approaches require specialized equipment, facilities, and research training beyond the capabilities of many biomedical laboratories. Here we present xurography as an inexpensive, accessible, and versatile rapid prototyping technique for engineering cylindrical and lumenized microvessels. Using a benchtop xurographer, or a cutting plotter, we fabricated modular multi-layer poly(dimethylsiloxane) (PDMS)-based microphysiological systems (MPS) that house endothelial-lined microvessels approximately 260 μm in diameter embedded within a user-defined 3-D extracellular matrix (ECM). We validated the vascularized MPS (or vessel-on-a-chip) by quantifying changes in blood vessel permeability due to the pro-angiogenic chemokine CXCL12. Moreover, we demonstrated the reconfigurable versatility of this approach by engineering a total of four distinct vessel-ECM arrangements, which were obtained by only minor adjustments to a few steps of the fabrication process. Several of these arrangements, such as ones that incorporate close-ended vessel structures and spatially distinct ECM compartments along the same microvessel, have not been widely achieved with other microfabrication strategies. Therefore, we anticipate that our low-cost and easy-to-implement fabrication approach will facilitate broader adoption of MPS with customizable vascular architectures and ECM components while reducing the turnaround time required for iterative designs.
Collapse
Affiliation(s)
- Shashwat S Agarwal
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH 43210, USA.
| | - Marcos Cortes-Medina
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Jacob C Holter
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Alex Avendano
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Joseph W Tinapple
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Joseph M Barlage
- Department of Biomedical Education and Anatomy, The Ohio State University, Columbus, OH 43210, USA
| | - Miles M Menyhert
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Lotanna M Onua
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Jonathan W Song
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH 43210, USA.
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
14
|
Jiao D, Xie L, Xing W. A pumpless liver-on-a-chip for drug hepatotoxicity analysis. Analyst 2024; 149:4675-4686. [PMID: 39086194 DOI: 10.1039/d4an00602j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
This study presents the development and validation of an innovative microfluidic liver-on-a-chip device utilizing gravity-driven perfusion for the evaluation of drug hepatotoxicity. This research involved the construction of a hydrogel-based coculture chip that integrates liver parenchymal and stellate cells within a tri-channel configuration. The assembly and operation of the liver-on-a-chip and its accompanying custom rocker were straightforward. The cells in the chip maintained high viability and continuously synthesized liver albumin over extended culture durations. Acetaminophen (APAP), a hepatic injury-inducing drug, was utilized as a positive control in hepatic toxicity assays on the chip. The liver chip exhibited hepatotoxic responses comparable to those observed in 2D models. Furthermore, in this study we evaluated the effects of two plant-derived natural compounds, aristolochic acid I (AA) and its analog aristolactam AII (AL), in both 2D cell models and the liver-on-a-chip system. AA, known for its hepatorenal toxicity, was observed to cause hepatotoxicity in both the 2D models and on the chip. The flow cytometry and mRNA sequencing results confirmed the propensity of these compounds to induce liver cell apoptosis. Notably, AL, previously considered nontoxic, provoked a significant decrease in the hepatic functionality marker albumin exclusively in the liver chip but not in 2D models, indicating the liver chip's enhanced sensitivity to toxic substances. In summary, this pumpless liver-on-a-chip is a simple yet powerful tool for drug hepatotoxicity studies.
Collapse
Affiliation(s)
- Dian Jiao
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Lan Xie
- Medical Systems Biology Research Center, School of Medicine, Tsinghua University, Beijing, China.
- National Engineering Research Center for Beijing Biochip Technology, Beijing, China
| | - Wanli Xing
- Medical Systems Biology Research Center, School of Medicine, Tsinghua University, Beijing, China.
- National Engineering Research Center for Beijing Biochip Technology, Beijing, China
| |
Collapse
|
15
|
Hernández-Hatibi S, Guerrero PE, García-Aznar JM, García-Gareta E. Polydopamine Interfacial Coating for Stable Tumor-on-a-Chip Models: Application for Pancreatic Ductal Adenocarcinoma. Biomacromolecules 2024; 25:5169-5180. [PMID: 39083627 PMCID: PMC11323005 DOI: 10.1021/acs.biomac.4c00551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024]
Abstract
Addressing current challenges in solid tumor research requires advanced in vitro three-dimensional (3D) cellular models that replicate the inherently 3D architecture and microenvironment of tumor tissue, including the extracellular matrix (ECM). However, tumor cells exert mechanical forces that can disrupt the physical integrity of the matrix in long-term 3D culture. Therefore, it is necessary to find the optimal balance between cellular forces and the preservation of matrix integrity. This work proposes using polydopamine (PDA) coating for 3D microfluidic cultures of pancreatic cancer cells to overcome matrix adhesion challenges to sustain representative tumor 3D cultures. Using PDA's distinctive adhesion and biocompatibility, our model uses type I collagen hydrogels seeded with different pancreatic cancer cell lines, prompting distinct levels of matrix deformation and contraction. Optimizing the PDA coating enhances the adhesion and stability of collagen hydrogels within microfluidic devices, achieving a balance between the disruptive forces of tumor cells on matrix integrity and the maintenance of long-term 3D cultures. The findings reveal how this tension appears to be a critical determinant in spheroid morphology and growth dynamics. Stable and prolonged 3D culture platforms are crucial for understanding solid tumor cell behavior, dynamics, and responses within a controlled microenvironment. This advancement ultimately offers a powerful tool for drug screening, personalized medicine, and wider cancer therapeutics strategies.
Collapse
Affiliation(s)
- Soraya Hernández-Hatibi
- Multiscale
in Mechanical & Biological Engineering Research Group, Aragon
Institute of Engineering Research (I3A), School of Engineering and
Architecture, University of Zaragoza, 50018 Zaragoza, Aragon, Spain
- Department
of Biochemistry and Molecular and Cellular Biology, Faculty of Sciences, University of Zaragoza, 50009 Zaragoza, Aragon, Spain
| | - Pedro Enrique Guerrero
- Multiscale
in Mechanical & Biological Engineering Research Group, Aragon
Institute of Engineering Research (I3A), School of Engineering and
Architecture, University of Zaragoza, 50018 Zaragoza, Aragon, Spain
- Department
of Biochemistry and Molecular and Cellular Biology, Faculty of Sciences, University of Zaragoza, 50009 Zaragoza, Aragon, Spain
| | - José Manuel García-Aznar
- Multiscale
in Mechanical & Biological Engineering Research Group, Aragon
Institute of Engineering Research (I3A), School of Engineering and
Architecture, University of Zaragoza, 50018 Zaragoza, Aragon, Spain
- Aragon
Institute for Health Research (IIS Aragon), Miguel Servet University Hospital, 50009 Zaragoza, Aragon, Spain
| | - Elena García-Gareta
- Multiscale
in Mechanical & Biological Engineering Research Group, Aragon
Institute of Engineering Research (I3A), School of Engineering and
Architecture, University of Zaragoza, 50018 Zaragoza, Aragon, Spain
- Aragon
Institute for Health Research (IIS Aragon), Miguel Servet University Hospital, 50009 Zaragoza, Aragon, Spain
- Division
of Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, University College London, London WC1E 6BT, U.K.
| |
Collapse
|
16
|
Cassel de Camps C, Rostami S, Xu V, Li C, Lépine P, Durcan TM, Moraes C. Microfabricated dynamic brain organoid cocultures to assess the effects of surface geometry on assembloid formation. Biotechnol J 2024; 19:e2400070. [PMID: 39167558 DOI: 10.1002/biot.202400070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024]
Abstract
Organoids have emerged as valuable tools for the study of development and disease. Assembloids are formed by integrating multiple organoid types to create more complex models. However, the process by which organoids integrate to form assembloids remains unclear and may play an important role in the resulting organoid structure. Here, a microfluidic platform is developed that allows separate culture of distinct organoid types and provides the capacity to partially control the geometry of the resulting organoid surfaces. Removal of a microfabricated barrier then allows the shaped and positioned organoids to interact and form an assembloid. When midbrain and unguided brain organoids were allowed to assemble with a defined spacing between them, axonal projections from midbrain organoids and cell migration out of unguided organoids were observed and quantitatively measured as the two types of organoids fused together. Axonal projection directions were statistically biased toward other midbrain organoids, and unguided organoid surface geometry was found to affect cell invasion. This platform provides a tool to observe cellular interactions between organoid surfaces that are spaced apart in a controlled manner, and may ultimately have value in exploring neuronal migration, axon targeting, and assembloid formation mechanisms.
Collapse
Affiliation(s)
| | - Sabra Rostami
- Department of Chemical Engineering, McGill University, Montréal, QC, Canada
| | - Vanessa Xu
- Department of Chemical Engineering, McGill University, Montréal, QC, Canada
| | - Chen Li
- Department of Chemical Engineering, McGill University, Montréal, QC, Canada
| | - Paula Lépine
- Early Drug Discovery Unit (EDDU), Montreal Neurological Institute and Hospital, McGill University, Montréal, QC, Canada
| | - Thomas M Durcan
- Early Drug Discovery Unit (EDDU), Montreal Neurological Institute and Hospital, McGill University, Montréal, QC, Canada
| | - Christopher Moraes
- Department of Biomedical Engineering, McGill University, Montréal, QC, Canada
- Department of Chemical Engineering, McGill University, Montréal, QC, Canada
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
- Division of Experimental Medicine, McGill University, Montréal, QC, Canada
| |
Collapse
|
17
|
Nam U, Kim J, Yi HG, Jeon JS. Investigation of the Dysfunction Caused by High Glucose, Advanced Glycation End Products, and Interleukin-1 Beta and the Effects of Therapeutic Agents on the Microphysiological Artery Model. Adv Healthc Mater 2024; 13:e2302682. [PMID: 38575148 DOI: 10.1002/adhm.202302682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 03/31/2024] [Indexed: 04/06/2024]
Abstract
Diabetes mellitus (DM) has substantial global implications and contributes to vascular inflammation and the onset of atherosclerotic cardiovascular diseases. However, translating the findings from animal models to humans has inherent limitations, necessitating a novel platform. Therefore, herein, an arterial model is established using a microphysiological system. This model successfully replicates the stratified characteristics of human arteries by integrating collagen, endothelial cells (ECs), and vascular smooth muscle cells (VSMCs). Perfusion via a peristaltic pump shows dynamic characteristics distinct from those of static culture models. High glucose, advanced glycation end products (AGEs), and interleukin-1 beta are employed to stimulate diabetic conditions, resulting in notable cellular changes and different levels of cytokines and nitric oxide. Additionally, the interactions between the disease models and oxidized low-density lipoproteins (LDL) are examined. Finally, the potential therapeutic effects of metformin, atorvastatin, and diphenyleneiodonium are investigated. Metformin and diphenyleneiodonium mitigate high-glucose- and AGE-associated pathological changes, whereas atorvastatin affects only the morphology of ECs. Altogether, the arterial model represents a pivotal advancement, offering a robust and insightful platform for investigating cardiovascular diseases and their corresponding drug development.
Collapse
Affiliation(s)
- Ungsig Nam
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
- Center for Scientific Instrumentation, Korea Basic Science Institute (KBSI), Daejeon, 34133, Republic of Korea
| | - Jaesang Kim
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Hee-Gyeong Yi
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jessie S Jeon
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| |
Collapse
|
18
|
Kpeli GW, Conrad KM, Bralower W, Byrne CE, Boue SM, Burow ME, Mondrinos MJ. Xenohormetic Phytochemicals Inhibit Neovascularization in Microphysiological Models of Vasculogenesis and Tumor Angiogenesis. Adv Biol (Weinh) 2024; 8:e2300480. [PMID: 38831745 DOI: 10.1002/adbi.202300480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 04/30/2024] [Indexed: 06/05/2024]
Abstract
Xenohormesis proposes that phytochemicals produced to combat stressors in the host plant exert biochemical effects in animal cells lacking cognate receptors. Xenohormetic phytochemicals such as flavonoids and phytoalexins modulate a range of human cell signaling mechanisms but functional correlations with human pathophysiology are lacking. Here, potent inhibitory effects of grapefruit-derived Naringenin (Nar) and soybean-derived Glyceollins (Gly) in human microphysiological models of bulk tissue vasculogenesis and tumor angiogenesis are reported. Despite this interference of vascular morphogenesis, Nar and Gly are not cytotoxic to endothelial cells and do not prevent cell cycle entry. The anti-vasculogenic effects of Glyceollin are significantly more potent in sex-matched female (XX) models. Nar and Gly do not decrease viability or expression of proangiogenic genes in triple negative breast cancer (TNBC) cell spheroids, suggesting that inhibition of sprouting angiogenesis by Nar and Gly in a MPS model of the (TNBC) microenvironment are mediated via direct effects in endothelial cells. The study supports further research of Naringenin and Glyceollin as health-promoting agents with special attention to mechanisms of action in vascular endothelial cells and the role of biological sex, which can improve the understanding of dietary nutrition and the pharmacology of phytochemical preparations.
Collapse
Affiliation(s)
- G Wills Kpeli
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, 70118, USA
| | - K Michael Conrad
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, 70118, USA
| | - William Bralower
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, 70118, USA
| | - C Ethan Byrne
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, 70118, USA
| | - Stephen M Boue
- Southern Regional Research Center, US Department of Agriculture, New Orleans, LA, 70124, USA
| | - Matthew E Burow
- Section of Hematology & Medical Oncology, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Mark J Mondrinos
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, 70118, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA
- Tulane Center for Excellence in Sex-based Biology and Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| |
Collapse
|
19
|
Asgeirsson DO, Mehta A, Scheeder A, Li F, Wang X, Christiansen MG, Hesse N, Ward R, De Micheli AJ, Ildiz ES, Menghini S, Aceto N, Schuerle S. Magnetically controlled cyclic microscale deformation of in vitro cancer invasion models. Biomater Sci 2023; 11:7541-7555. [PMID: 37855703 DOI: 10.1039/d3bm00583f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Mechanical cues play an important role in the metastatic cascade of cancer. Three-dimensional (3D) tissue matrices with tunable stiffness have been extensively used as model systems of the tumor microenvironment for physiologically relevant studies. Tumor-associated cells actively deform these matrices, providing mechanical cues to other cancer cells residing in the tissue. Mimicking such dynamic deformation in the surrounding tumor matrix may help clarify the effect of local strain on cancer cell invasion. Remotely controlled microscale magnetic actuation of such 3D in vitro systems is a promising approach, offering a non-invasive means for in situ interrogation. Here, we investigate the influence of cyclic deformation on tumor spheroids embedded in matrices, continuously exerted for days by cell-sized anisotropic magnetic probes, referred to as μRods. Particle velocimetry analysis revealed the spatial extent of matrix deformation produced in response to a magnetic field, which was found to be on the order of 200 μm, resembling strain fields reported to originate from contracting cells. Intracellular calcium influx was observed in response to cyclic actuation, as well as an influence on cancer cell invasion from 3D spheroids, as compared to unactuated controls. Furthermore, RNA sequencing revealed subtle upregulation of certain genes associated with migration and stress, such as induced through mechanical deformation, for spheroids exposed to actuation vs. controls. Localized actuation at one side of a tumor spheroid tended to result in anisotropic invasion toward the μRods causing the deformation. In summary, our approach offers a strategy to test and control the influence of non-invasive micromechanical cues on cancer cell invasion and metastasis.
Collapse
Affiliation(s)
- Daphne O Asgeirsson
- Department of Health Sciences and Technology, Responsive Biomedical Systems Laboratory, ETH Zurich, 8093 Zurich, Switzerland.
| | - Avni Mehta
- Department of Health Sciences and Technology, Responsive Biomedical Systems Laboratory, ETH Zurich, 8093 Zurich, Switzerland.
| | - Anna Scheeder
- Department of Health Sciences and Technology, Responsive Biomedical Systems Laboratory, ETH Zurich, 8093 Zurich, Switzerland.
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, U.K
| | - Fan Li
- Department of Health Sciences and Technology, Responsive Biomedical Systems Laboratory, ETH Zurich, 8093 Zurich, Switzerland.
| | - Xiang Wang
- Department of Health Sciences and Technology, Responsive Biomedical Systems Laboratory, ETH Zurich, 8093 Zurich, Switzerland.
| | - Michael G Christiansen
- Department of Health Sciences and Technology, Responsive Biomedical Systems Laboratory, ETH Zurich, 8093 Zurich, Switzerland.
| | - Nicolas Hesse
- Department of Health Sciences and Technology, Responsive Biomedical Systems Laboratory, ETH Zurich, 8093 Zurich, Switzerland.
| | - Rachel Ward
- Department of Health Sciences and Technology, Responsive Biomedical Systems Laboratory, ETH Zurich, 8093 Zurich, Switzerland.
| | - Andrea J De Micheli
- Department of Health Sciences and Technology, Responsive Biomedical Systems Laboratory, ETH Zurich, 8093 Zurich, Switzerland.
- Department of Oncology, Children's Research Center, University Children's Hospital Zurich, Zurich 8032, Switzerland
| | - Ece Su Ildiz
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Stefano Menghini
- Department of Health Sciences and Technology, Responsive Biomedical Systems Laboratory, ETH Zurich, 8093 Zurich, Switzerland.
| | - Nicola Aceto
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Simone Schuerle
- Department of Health Sciences and Technology, Responsive Biomedical Systems Laboratory, ETH Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
20
|
Liu H, Fan P, Jin F, Ren H, Xu F, Li J. Targeting biophysical microenvironment for improved treatment of chronic obstructive pulmonary disease. Trends Mol Med 2023; 29:926-938. [PMID: 37704492 DOI: 10.1016/j.molmed.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/15/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is responsible for high disability rates, high death rates, and significant cost to health systems. Growing evidence in recent decades shows significant biophysical microenvironment changes in COPD, impacting lung tissues, cells, and treatment response. Furthermore, such biophysical changes have shown great potential as novel targets for improved therapeutic strategy of COPD, where both pharmacological and non-pharmacological therapies focusing on repairing the biophysical microenvironment of the lung have emerged. We present the first comprehensive review of four distinct biophysical hallmarks [i.e., extracellular matrix (ECM) microarchitecture, stiffness, fluid shear stress, and mechanical stretch] in COPD, the possible involvement of pathological changes, possible effects, and correlated in vitro models and sum up the emerging COPD treatments targeting these biophysical hallmarks.
Collapse
Affiliation(s)
- Han Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province and Education Ministry of China, Zhengzhou, Henan 450046, China
| | - Pengbei Fan
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province and Education Ministry of China, Zhengzhou, Henan 450046, China
| | - Fanli Jin
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province and Education Ministry of China, Zhengzhou, Henan 450046, China
| | - Hui Ren
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China; Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China; MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Jiansheng Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province and Education Ministry of China, Zhengzhou, Henan 450046, China.
| |
Collapse
|
21
|
Kawara S, Cunningham B, Bezer J, Kc N, Zhu J, Tang MX, Ishihara J, Choi JJ, Au SH. Capillary-Scale Hydrogel Microchannel Networks by Wire Templating. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301163. [PMID: 37267935 DOI: 10.1002/smll.202301163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/08/2023] [Indexed: 06/04/2023]
Abstract
Microvascular networks are essential for the efficient transport of nutrients, waste products, and drugs throughout the body. Wire-templating is an accessible method for generating laboratory models of these blood vessel networks, but it has difficulty fabricating microchannels with diameters of ten microns and narrower, a requirement for modeling human capillaries. This study describes a suite of surface modification techniques to selectively control the interactions amongst wires, hydrogels, and world-to-chip interfaces. This wire templating method enables the fabrication of perfusable hydrogel-based rounded cross-section capillary-scale networks whose diameters controllably narrow at bifurcations down to 6.1 ± 0.3 microns in diameter. Due to its low cost, accessibility, and compatibility with a wide range of common hydrogels of tunable stiffnesses such as collagen, this technique may increase the fidelity of experimental models of capillary networks for the study of human health and disease.
Collapse
Affiliation(s)
- Shusei Kawara
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2AZ, UK
| | - Brian Cunningham
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2AZ, UK
- Cancer Research UK Convergence Science Centre, London, SW7 2AZ, UK
| | - James Bezer
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2AZ, UK
| | - Neelima Kc
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2AZ, UK
| | - Jingwen Zhu
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2AZ, UK
| | - Meng-Xing Tang
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2AZ, UK
| | - Jun Ishihara
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2AZ, UK
| | - James J Choi
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2AZ, UK
| | - Sam H Au
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2AZ, UK
- Cancer Research UK Convergence Science Centre, London, SW7 2AZ, UK
| |
Collapse
|
22
|
Wang D, Brady T, Santhanam L, Gerecht S. The extracellular matrix mechanics in the vasculature. NATURE CARDIOVASCULAR RESEARCH 2023; 2:718-732. [PMID: 39195965 DOI: 10.1038/s44161-023-00311-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 06/20/2023] [Indexed: 08/29/2024]
Abstract
Mechanical stimuli from the extracellular matrix (ECM) modulate vascular differentiation, morphogenesis and dysfunction of the vasculature. With innovation in measurements, we can better characterize vascular microenvironment mechanics in health and disease. Recent advances in material sciences and stem cell biology enable us to accurately recapitulate the complex and dynamic ECM mechanical microenvironment for in vitro studies. These biomimetic approaches help us understand the signaling pathways in disease pathologies, identify therapeutic targets, build tissue replacement and activate tissue regeneration. This Review analyzes how ECM mechanics regulate vascular homeostasis and dysfunction. We highlight approaches to examine ECM mechanics at tissue and cellular levels, focusing on how mechanical interactions between cells and the ECM regulate vascular phenotype, especially under certain pathological conditions. Finally, we explore the development of biomaterials to emulate, measure and alter the physical microenvironment of pathological ECM to understand cell-ECM mechanical interactions toward the development of therapeutics.
Collapse
Affiliation(s)
- Dafu Wang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Travis Brady
- Department of Anesthesiology and Critical Care Medicine and Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Lakshmi Santhanam
- Department of Anesthesiology and Critical Care Medicine and Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Sharon Gerecht
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
23
|
Martier AT, Maurice YV, Conrad KM, Mauvais-Jarvis F, Mondrinos MJ. Sex-specific actions of estradiol and testosterone on human fibroblast and endothelial cell proliferation, bioenergetics, and vasculogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.23.550236. [PMID: 37546849 PMCID: PMC10402012 DOI: 10.1101/2023.07.23.550236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Progress toward the development of sex-specific tissue engineered systems has been hampered by the lack of research efforts to define the effects of sex-specific hormone concentrations on relevant human cell types. Here, we investigated the effects of defined concentrations of estradiol (E2) and dihydrotestosterone (DHT) on primary human dermal and lung fibroblasts (HDF and HLF), and human umbilical vein endothelial cells (HUVEC) from female (XX) and male (XY) donors in both 2D expansion cultures and 3D stromal vascular tissues. Sex-matched E2 and DHT stimulation in 2D expansion cultures significantly increased the proliferation index, mitochondrial membrane potential, and the expression of genes associated with bioenergetics (Na+/K+ ATPase, somatic cytochrome C) and beneficial stress responses (chaperonin) in all cell types tested. Notably, cross sex hormone stimulation, i.e., DHT treatment of XX cells in the absence of E2 and E2 stimulation of XY cells in the absence of DHT, decreased bioenergetic capacity and inhibited cell proliferation. We used a microengineered 3D vasculogenesis assay to assess hormone effects on tissue scale morphogenesis. E2 increased metrics of vascular network complexity compared to vehicle in XX tissues. Conversely, and in line with results from 2D expansion cultures, E2 potently inhibited vasculogenesis compared to vehicle in XY tissues. DHT did not significantly alter vasculogenesis in XX or XY tissues but increased the number of non-participating endothelial cells in both sexes. This study establishes a scientific rationale and adaptable methods for using sex hormone stimulation to develop sex-specific culture systems.
Collapse
Affiliation(s)
- Ashley T. Martier
- Department of Biomedical Engineering, Tulane University School of Science & Engineering, New Orleans, LA, USA
| | - Yasmin V. Maurice
- Department of Biomedical Engineering, Tulane University School of Science & Engineering, New Orleans, LA, USA
| | - K. Michael Conrad
- Department of Biomedical Engineering, Tulane University School of Science & Engineering, New Orleans, LA, USA
| | - Franck Mauvais-Jarvis
- Tulane Center for Excellence in Sex-based Biology and Medicine, New Orleans, LA, USA
- Section of Endocrinology, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
- Southeast Louisiana VA Medical Center, New Orleans, LA, USA
| | - Mark J. Mondrinos
- Department of Biomedical Engineering, Tulane University School of Science & Engineering, New Orleans, LA, USA
- Tulane Center for Excellence in Sex-based Biology and Medicine, New Orleans, LA, USA
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
24
|
Schönherr-Hellec S, Chatzopoulou E, Barnier JP, Atlas Y, Dupichaud S, Guilbert T, Dupraz Y, Meyer J, Chaussain C, Gorin C, Nassif X, Germain S, Muller L, Coureuil M. Implantation of engineered human microvasculature to study human infectious diseases in mouse models. iScience 2023; 26:106286. [PMID: 36942053 PMCID: PMC10024136 DOI: 10.1016/j.isci.2023.106286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 01/10/2023] [Accepted: 02/21/2023] [Indexed: 03/03/2023] Open
Abstract
Animal models for studying human pathogens are crucially lacking. We describe the implantation in mice of engineered human mature microvasculature consisting of endothelial and perivascular cells embedded in collagen hydrogel that allows investigation of pathogen interactions with the endothelium, including in vivo functional studies. Using Neisseria meningitidis as a paradigm of human-restricted infection, we demonstrated the strength and opportunities associated with the use of this approach.
Collapse
Affiliation(s)
- Sophia Schönherr-Hellec
- Université Paris Cité, UFR de Médecine, Paris, France
- Institut Necker Enfants-Malades, Inserm U1151, CNRS UMR 8253, Paris, France
| | - Eirini Chatzopoulou
- Université Paris Cité, UPR2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant, UFR Odontologie, Paris, France
| | - Jean-Philippe Barnier
- Université Paris Cité, UFR de Médecine, Paris, France
- Institut Necker Enfants-Malades, Inserm U1151, CNRS UMR 8253, Paris, France
| | - Yoann Atlas
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France
- Sorbonne Université, Collège doctoral, Paris, France
| | - Sébastien Dupichaud
- Cell Imaging Platform, Structure Fédérative de Recherche Necker INSERM US24/CNRS UMS3633, Paris, France
| | - Thomas Guilbert
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Cité, Paris, France
| | - Yves Dupraz
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Julie Meyer
- Université Paris Cité, UFR de Médecine, Paris, France
- Institut Necker Enfants-Malades, Inserm U1151, CNRS UMR 8253, Paris, France
| | - Catherine Chaussain
- Université Paris Cité, UPR2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant, UFR Odontologie, Paris, France
- AP-HP, Services Médecines bucco-dentaire (GH Paris Sud-Sorbonne Université, Paris Nord-Université Paris Cité), Paris, France
| | - Caroline Gorin
- Université Paris Cité, UPR2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant, UFR Odontologie, Paris, France
- AP-HP, Services Médecines bucco-dentaire (GH Paris Sud-Sorbonne Université, Paris Nord-Université Paris Cité), Paris, France
| | - Xavier Nassif
- Université Paris Cité, UFR de Médecine, Paris, France
- Institut Necker Enfants-Malades, Inserm U1151, CNRS UMR 8253, Paris, France
| | - Stephane Germain
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Laurent Muller
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France
- Corresponding author
| | - Mathieu Coureuil
- Université Paris Cité, UFR de Médecine, Paris, France
- Institut Necker Enfants-Malades, Inserm U1151, CNRS UMR 8253, Paris, France
- Corresponding author
| |
Collapse
|
25
|
Nguyen RY, Cabral AT, Rossello-Martinez A, Zulli A, Gong X, Zhang Q, Yan J, Mak M. Tunable Mesoscopic Collagen Island Architectures Modulate Stem Cell Behavior. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2207882. [PMID: 36895051 PMCID: PMC10166061 DOI: 10.1002/adma.202207882] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/22/2022] [Indexed: 05/10/2023]
Abstract
The extracellular matrix is the biophysical environment that scaffolds mammalian cells in the body. The main constituent is collagen. In physiological tissues, collagen network topology is diverse with complex mesoscopic features. While studies have explored the roles of collagen density and stiffness, the impact of complex architectures remains not well-understood. Developing in vitro systems that recapitulate these diverse collagen architectures is critical for understanding physiologically relevant cell behaviors. Here, methods are developed to induce the formation of heterogeneous mesoscopic architectures, referred to as collagen islands, in collagen hydrogels. These island-containing gels have highly tunable inclusions and mechanical properties. Although these gels are globally soft, there is regional enrichment in the collagen concentration at the cell-scale. Collagen-island architectures are utilized to study mesenchymal stem cell behavior, and it is demonstrated that cell migration and osteogenic differentiation are altered. Finally, induced pluripotent stem cells are cultured in island-containing gels, and it is shown that the architecture is sufficient to induce mesodermal differentiation. Overall, this work highlights complex mesoscopic tissue architectures as bioactive cues in regulating cell behavior and presents a novel collagen-based hydrogel that captures these features for tissue engineering applications.
Collapse
Affiliation(s)
- Ryan Y. Nguyen
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Aidan T. Cabral
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | | | - Alessandro Zulli
- Department of Chemical and Environmental Engineering, Yale University, New Haven, CT, USA
| | - Xiangyu Gong
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Qiuting Zhang
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Jing Yan
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| |
Collapse
|
26
|
Jeong S, Kang HW, Kim SH, Hong GS, Nam MH, Seong J, Yoon ES, Cho IJ, Chung S, Bang S, Kim HN, Choi N. Integration of reconfigurable microchannels into aligned three-dimensional neural networks for spatially controllable neuromodulation. SCIENCE ADVANCES 2023; 9:eadf0925. [PMID: 36897938 PMCID: PMC10005277 DOI: 10.1126/sciadv.adf0925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 02/08/2023] [Indexed: 06/18/2023]
Abstract
Anisotropically organized neural networks are indispensable routes for functional connectivity in the brain, which remains largely unknown. While prevailing animal models require additional preparation and stimulation-applying devices and have exhibited limited capabilities regarding localized stimulation, no in vitro platform exists that permits spatiotemporal control of chemo-stimulation in anisotropic three-dimensional (3D) neural networks. We present the integration of microchannels seamlessly into a fibril-aligned 3D scaffold by adapting a single fabrication principle. We investigated the underlying physics of elastic microchannels' ridges and interfacial sol-gel transition of collagen under compression to determine a critical window of geometry and strain. We demonstrated the spatiotemporally resolved neuromodulation in an aligned 3D neural network by local deliveries of KCl and Ca2+ signal inhibitors, such as tetrodotoxin, nifedipine, and mibefradil, and also visualized Ca2+ signal propagation with a speed of ~3.7 μm/s. We anticipate that our technology will pave the way to elucidate functional connectivity and neurological diseases associated with transsynaptic propagation.
Collapse
Affiliation(s)
- Sohyeon Jeong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Korea
- MEPSGEN Co. Ltd., Seoul 05836, Korea
| | - Hyun Wook Kang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- School of Mechanical Engineering, Korea University, Seoul 02841, Korea
| | - So Hyun Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- SK Biopharmaceuticals Co. Ltd., Seongnam 13494, Korea
| | - Gyu-Sang Hong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Jihye Seong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Korea
- Department of Life Sciences, Korea University, Seoul 02841, Korea
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02453, Korea
| | - Eui-Sung Yoon
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- Division of Nano and Information Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Korea
| | - Il-Joo Cho
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea
- Department of Anatomy, College of Medicine, Korea University, Seoul 02841, Korea
| | - Seok Chung
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- School of Mechanical Engineering, Korea University, Seoul 02841, Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| | - Seokyoung Bang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- Department of Medical Biotechnology, Dongguk University, Goyang 10326, Korea
| | - Hong Nam Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Korea
- School of Mechanical Engineering, Yonsei University, Seoul 03722, Korea
- Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul 03722, Korea
| | - Nakwon Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| |
Collapse
|
27
|
Pan HJ, Lee CW, Wu LY, Hsu HH, Tung YC, Liao WY, Lee CH. A 3D culture system for evaluating the combined effects of cisplatin and anti-fibrotic drugs on the growth and invasion of lung cancer cells co-cultured with fibroblasts. APL Bioeng 2023; 7:016117. [PMID: 37006781 PMCID: PMC10060027 DOI: 10.1063/5.0115464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 02/23/2023] [Indexed: 03/30/2023] Open
Abstract
Fibrosis and fibroblast activation usually occur in the tissues surrounding a malignant tumor; therefore, anti-fibrotic drugs are used in addition to chemotherapy. A reliable technique for evaluating the combined effects of anti-fibrotic drugs and anticancer drugs would be beneficial for the development of an appropriate treatment strategy. In this study, we manufactured a three-dimensional (3D) co-culture system of fibroblasts and lung cancer cell spheroids in Matrigel supplemented with fibrin (fibrin/Matrigel) that simulated the tissue microenvironment around a solid tumor. We compared the efficacy of an anticancer drug (cisplatin) with or without pretreatments of two anti-fibrotic drugs, nintedanib and pirfenidone, on the growth and invasion of cancer cells co-cultured with fibroblasts. The results showed that the addition of nintedanib improved cisplatin's effects on suppressing the growth of cancer cell spheroids and the invasion of cancer cells. In contrast, pirfenidone did not enhance the anticancer activity of cisplatin. Nintedanib also showed higher efficacy than pirfenidone in reducing the expression of four genes in fibroblasts associated with cell adhesion, invasion, and extracellular matrix degradation. This study demonstrated that the 3D co-cultures in fibrin/Matrigel would be useful for assessing the effects of drug combinations on tumor growth and invasion.
Collapse
Affiliation(s)
- Huei-Jyuan Pan
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Chia-Wei Lee
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Li-Yu Wu
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Heng-Hua Hsu
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Yi-Chung Tung
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Wei-Yu Liao
- Authors to whom correspondence should be addressed: and
| | | |
Collapse
|
28
|
Zhou Y, Yang Y, Liu R, Zhou Q, Lu H, Zhang W. Research Progress of Polydopamine Hydrogel in the Prevention and Treatment of Oral Diseases. Int J Nanomedicine 2023; 18:2623-2645. [PMID: 37213351 PMCID: PMC10199686 DOI: 10.2147/ijn.s407044] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 04/12/2023] [Indexed: 05/23/2023] Open
Abstract
Oral diseases represent one of the most prevalent diseases globally and are associated with serious health and economic burdens, greatly altering the quality of life of affected individuals. Various biomaterials play important roles in the treatment of oral diseases. To some extent, the development of biomaterials has promoted progress in clinically available oral medicines. Hydrogels have unique tunable advantages that make them useful in the next generation of regenerative strategies and have been widely applied in both oral soft and hard tissues repair. However, most hydrogels lack self-adhesive properties, which may result in low repair efficacy. Polydopamine (PDA), the primary adhesive component, has attracted increasing attention in recent years. PDA-modified hydrogels exhibit reliable and suitable adherence to tissues and easily integrate into tissues to promote repair efficiency. This paper reviews the latest research progress on PDA hydrogels and elaborates on the mechanism of the reaction between PDA functional groups and hydrogels, and summarizes the biological properties and the applications of PDA hydrogels in the prevention and treatment of the field of oral diseases. It is also proposed that in future research we should simulate the complex microenvironment of the oral cavity as much as possible, coordinate and plan various biological events rationally, and realize the translation from scientific research to clinical practice.
Collapse
Affiliation(s)
- Yuqi Zhou
- Department of Prosthodontics, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yuanmeng Yang
- Department of Preventive Dentistry, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Rongpu Liu
- Department of Prosthodontics, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Qin Zhou
- Department of Oral Surgery, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Haixia Lu
- Department of Preventive Dentistry, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Haixia Lu, Department of Preventive Dentistry, Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, People’s Republic of China, Email
| | - Wenjie Zhang
- Department of Prosthodontics, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Correspondence: Wenjie Zhang, Department of Prosthodontics, Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, People’s Republic of China, Email
| |
Collapse
|
29
|
Deng P, Zhao M, Zhang X, Qin J. A Transwell-Based Vascularized Model to Investigate the Effect of Interstitial Flow on Vasculogenesis. Bioengineering (Basel) 2022; 9:668. [PMID: 36354579 PMCID: PMC9687519 DOI: 10.3390/bioengineering9110668] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/27/2022] [Accepted: 10/30/2022] [Indexed: 09/08/2024] Open
Abstract
Interstitial flow plays a significant role in vascular system development, mainly including angiogenesis and vasculogenesis. However, compared to angiogenesis, the effect of interstitial flow on vasculogenesis is less explored. Current in vitro models for investigating the effect of interstitial flow on vasculogenesis heavily rely on microfluidic chips, which require microfluidic expertise and facilities, and may not be accessible to biological labs. Here, we proposed a facile approach to building perfusable vascular networks through the self-assembly of endothelial cells in a modified transwell format and investigated the effect of interstitial flow on vasculogenesis. We found that the effect of interstitial flow on vasculogenesis was closely related to the existence of VEGF and fibroblasts in the developed model: (1) In the presence of fibroblasts, interstitial flow (within the range of 0.1-0.6 μm/s) facilitated the perfusability of the engineered vasculatures. Additional VEGF in the culture medium further worked synergically with interstitial flow to develop longer, wider, denser, and more perfusable vasculatures than static counterparts; (2) In the absence of fibroblasts, vasculatures underwent severe regression within 7 days under static conditions. However, interstitial flow greatly inhibited vessel regression and enhanced vascular perfusability and morphogenesis without the need for additional VEGF. These results revealed that the effect of interstitial flow might vary depending on the existence of VEGF and fibroblasts, and would provide some guidelines for constructing in vitro self-assembled vasculatures. The established transwell-based vascularized model provides a simple method to build perfusable vasculatures and could also be utilized for creating functional tissues in regenerative medicine.
Collapse
Affiliation(s)
- Pengwei Deng
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengqian Zhao
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xu Zhang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Jianhua Qin
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
30
|
Park J, Kim S, Hong J, Jeon JS. Enabling perfusion through multicellular tumor spheroids promoting lumenization in a vascularized cancer model. LAB ON A CHIP 2022; 22:4335-4348. [PMID: 36226506 DOI: 10.1039/d2lc00597b] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
A tumor is composed of heterogeneous cell population, which is known as tumor stroma. In particular, blood vessels have an indispensable role in the tumor microenvironment acting as a key player in anti-cancer drug delivery. Recently, efforts have been made to accurately recapitulate the microenvironment by employing distinct cell types, however, the proper formation of perfusable tumor tissue is challenging. Here, perfusable tumor tissue is engineered by implanting multicellular tumor spheroids inside the microfluidic devices. Blood perfusion, spheroid growth, and vascular dynamics were monitored according to the spheroid composition and the contribution of internal and external vascular cells to spheroid perfusion was analyzed. Most notably, the increased penetration depth of fluorescence conjugated anti-cancer drug was observed in tri-culture spheroids. The implementation of tumor microenvironment reconstruction developed in this study not only creates a perfusable tumor vascular model but can also be utilized as a novel drug screening platform with patient-derived samples.
Collapse
Affiliation(s)
- Joonha Park
- Department of Mechanical Engineering, KAIST, Daejeon 34141, Korea.
| | - Seunggyu Kim
- Department of Mechanical Engineering, KAIST, Daejeon 34141, Korea.
| | - Jiman Hong
- Department of Mechanical Engineering, KAIST, Daejeon 34141, Korea.
| | - Jessie S Jeon
- Department of Mechanical Engineering, KAIST, Daejeon 34141, Korea.
| |
Collapse
|
31
|
Bulut M, Vila Cuenca M, de Graaf M, van den Hil FE, Mummery CL, Orlova VV. Three-Dimensional Vessels-on-a-Chip Based on hiPSC-derived Vascular Endothelial and Smooth Muscle Cells. Curr Protoc 2022; 2:e564. [PMID: 36250774 PMCID: PMC11648816 DOI: 10.1002/cpz1.564] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Blood vessels are composed of endothelial cells (ECs) that form the inner vessel wall and mural cells that cover the ECs to mediate their stabilization. Crosstalk between ECs and VSMCs while the ECs undergo microfluidic flow is vital for the function and integrity of blood vessels. Here, we describe a protocol to generate three-dimensional (3D) engineered vessels-on-chip (VoCs) composed of vascular cells derived from human induced pluripotent stem cells (hiPSCs). We first describe protocols for robust differentiation of vascular smooth muscle cells (hiPSC-VSMCs) from hiPSCs that are effective across multiple hiPSC lines. Second, we describe the fabrication of a simple microfluidic device consisting of a single collagen lumen that can act as a cell scaffold and support fluid flow using the viscous finger patterning (VFP) technique. After the channel is seeded sequentially with hiPSC-derived ECs (hiPSC-ECs) and hiPSC-VSMCs, a stable EC barrier covered by VSMCs lines the collagen lumen. We demonstrate that this 3D VoC model can recapitulate physiological cell-cell interaction and can be perfused under physiological shear stress using a microfluidic pump. The uniform geometry of the vessel lumens allows precise control of flow dynamics. We have thus developed a robust protocol to generate an entirely isogenic hiPSC-derived 3D VoC model, which could be valuable for studying vessel barrier function and physiology in healthy or disease states. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Differentiation of hiPSC-VSMCs Support Protocol 1: Characterization of hiPSC-NCCs and hiPSC-VSMCs Support Protocol 2: Preparation of cryopreserved hiPSC-VSMCs and hiPSC-ECs for VoC culture Basic Protocol 2: Generation of 3D VoC model composed of hiPSC-ECs and hiPSC-VSMCs Support Protocol 3: Structural characterization of 3D VoC model.
Collapse
Affiliation(s)
- Merve Bulut
- Department of Anatomy and EmbryologyLeiden University Medical CenterLeidenThe Netherlands
| | - Marc Vila Cuenca
- Department of Anatomy and EmbryologyLeiden University Medical CenterLeidenThe Netherlands
| | - Mees de Graaf
- Department of Anatomy and EmbryologyLeiden University Medical CenterLeidenThe Netherlands
| | | | - Christine L. Mummery
- Department of Anatomy and EmbryologyLeiden University Medical CenterLeidenThe Netherlands
- Department of Applied Stem Cell TechnologiesUniversity of TwenteEnschedeThe Netherlands
| | - Valeria V. Orlova
- Department of Anatomy and EmbryologyLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
32
|
Patient-derived microphysiological model identifies the therapeutic potential of metformin for thoracic aortic aneurysm. EBioMedicine 2022; 81:104080. [PMID: 35636318 PMCID: PMC9156889 DOI: 10.1016/j.ebiom.2022.104080] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 05/11/2022] [Accepted: 05/11/2022] [Indexed: 12/20/2022] Open
Abstract
Background Thoracic aortic aneurysm (TAA) is the permanent dilation of the thoracic aortic wall that predisposes patients to lethal events such as aortic dissection or rupture, for which effective medical therapy remains scarce. Human-relevant microphysiological models serve as a promising tool in drug screening and discovery. Methods We developed a dynamic, rhythmically stretching, three-dimensional microphysiological model. Using patient-derived human aortic smooth muscle cells (HAoSMCs), we tested the biological features of the model and compared them with native aortic tissues. Drug testing was performed on the individualized TAA models, and the potentially effective drug was further tested using β-aminopropionitrile-treated mice and retrospective clinical data. Findings The HAoSMCs on the model recapitulated the expressions of many TAA-related genes in tissue. Phenotypic switching and mitochondrial dysfunction, two disease hallmarks of TAA, were highlighted on the microphysiological model: the TAA-derived HAoSMCs exhibited lower alpha-smooth muscle actin expression, lower mitochondrial membrane potential, lower oxygen consumption rate and higher superoxide accumulation than control cells, while these differences were not evidently reflected in two-dimensional culture flasks. Model-based drug testing demonstrated that metformin partially recovered contractile phenotype and mitochondrial function in TAA patients’ cells. Mouse experiment and clinical investigations also demonstrated better preserved aortic microstructure, higher nicotinamide adenine dinucleotide level and lower aortic diameter with metformin treatment. Interpretation These findings support the application of this human-relevant microphysiological model in studying personalized disease characteristics and facilitating drug discovery for TAA. Metformin may regulate contractile phenotypes and metabolic dysfunctions in diseased HAoSMCs and limit aortic dilation. Funding This work was supported by grants from National Key R&D Program of China (2018YFC1005002), National Natural Science Foundation of China (82070482, 81771971, 81772007, 51927805, and 21734003), the Science and Technology Commission of Shanghai Municipality (20ZR1411700, 18ZR1407000, 17JC1400200, and 20YF1406900), Shanghai Municipal Science and Technology Major Project (2017SHZDZX01), and Shanghai Municipal Education Commission (Innovation Program 2017-01-07-00-07-E00027). Y.S.Z. was not supported by any of these funds; instead, the Brigham Research Institute is acknowledged.
Collapse
|
33
|
Kim S, Lee H, Kim JA, Park TH. Prevention of collagen hydrogel contraction using polydopamine-coating and alginate outer shell increases cell contractile force. BIOMATERIALS ADVANCES 2022; 136:212780. [PMID: 35929298 DOI: 10.1016/j.bioadv.2022.212780] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/07/2022] [Accepted: 03/25/2022] [Indexed: 06/15/2023]
Abstract
Collagen is the most abundant protein in the extracellular matrix of mammals and has a great effect on various cell behaviors including adhesion, differentiation, and migration. However, it is difficult to utilize collagen gel as a physical scaffold in vitro because of its severe contraction. Decrease in the overall hydrogel volume induces changes in cell distribution, and mass transfer within the gel. Uncontrolled mechanical and physiological factors in the fibrous matrix result in uncontrolled cell behaviors in the surrounding cells. In this study, two strategies were used to minimize the contraction of collagen gel. A disk-shaped frame made of polydopamine-coated polydimethylsiloxane (PDMS) prevented horizontal contraction at the edge of the hydrogel. The sequentially cross-linked collagen gel with alginate outer shell (CA-shell) structure inhibited the vertical gel contraction. The combined method synergistically prevented the hydrogel from shrinkage in long-term 3D cell culture. We observed the shift in balance of differentiation from adipogenesis to osteogenesis in mesenchymal stem cells under the environment where gel contraction was prevented, and confirmed that this phenomenon is closely associated with the mechanotransduction based on Yes-associated protein (YAP) localization. Development of this contraction inhibition platform made it possible to investigate the influence of regulation of cellular microenvironments. The physical properties of the hydrogel fabricated in this study were similar to that of pure collagen gel but completely changed the cell behavior within the gel by inhibition of gel contraction. The platform can be used to broaden our understanding of the fundamental mechanism underlying cell-matrix interactions and reproduce extracellular matrix in vivo.
Collapse
Affiliation(s)
- Seulha Kim
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| | - Haein Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| | - Jeong Ah Kim
- Center for Scientific Instrumentation, Korea Basic Science Institute, Cheongju, Chungbuk 28119, Republic of Korea.
| | - Tai Hyun Park
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea; Interdisciplinary Program in Bioengineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea; BioMAX/N-Bio Institute, Institute of BioEngineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| |
Collapse
|
34
|
Galateanu B, Hudita A, Biru EI, Iovu H, Zaharia C, Simsensohn E, Costache M, Petca RC, Jinga V. Applications of Polymers for Organ-on-Chip Technology in Urology. Polymers (Basel) 2022; 14:1668. [PMID: 35566836 PMCID: PMC9105302 DOI: 10.3390/polym14091668] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/05/2022] [Accepted: 04/18/2022] [Indexed: 02/07/2023] Open
Abstract
Organ-on-chips (OOCs) are microfluidic devices used for creating physiological organ biomimetic systems. OOC technology brings numerous advantages in the current landscape of preclinical models, capable of recapitulating the multicellular assemblage, tissue-tissue interaction, and replicating numerous human pathologies. Moreover, in cancer research, OOCs emulate the 3D hierarchical complexity of in vivo tumors and mimic the tumor microenvironment, being a practical cost-efficient solution for tumor-growth investigation and anticancer drug screening. OOCs are compact and easy-to-use microphysiological functional units that recapitulate the native function and the mechanical strain that the cells experience in the human bodies, allowing the development of a wide range of applications such as disease modeling or even the development of diagnostic devices. In this context, the current work aims to review the scientific literature in the field of microfluidic devices designed for urology applications in terms of OOC fabrication (principles of manufacture and materials used), development of kidney-on-chip models for drug-toxicity screening and kidney tumors modeling, bladder-on-chip models for urinary tract infections and bladder cancer modeling and prostate-on-chip models for prostate cancer modeling.
Collapse
Affiliation(s)
- Bianca Galateanu
- Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei Street, 050095 Bucharest, Romania; (B.G.); (M.C.)
| | - Ariana Hudita
- Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei Street, 050095 Bucharest, Romania; (B.G.); (M.C.)
| | - Elena Iuliana Biru
- Advanced Polymer Materials Group, Department of Bioresources and Polymer Science, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (H.I.); (C.Z.)
| | - Horia Iovu
- Advanced Polymer Materials Group, Department of Bioresources and Polymer Science, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (H.I.); (C.Z.)
- Academy of Romanian Scientists, Ilfov Street, 50044 Bucharest, Romania
| | - Catalin Zaharia
- Advanced Polymer Materials Group, Department of Bioresources and Polymer Science, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (H.I.); (C.Z.)
| | - Eliza Simsensohn
- “Carol Davila” University of Medicine and Pharmacy Bucharest, 050474 Bucharest, Romania; (E.S.); (R.-C.P.); (V.J.)
| | - Marieta Costache
- Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei Street, 050095 Bucharest, Romania; (B.G.); (M.C.)
| | - Razvan-Cosmin Petca
- “Carol Davila” University of Medicine and Pharmacy Bucharest, 050474 Bucharest, Romania; (E.S.); (R.-C.P.); (V.J.)
| | - Viorel Jinga
- “Carol Davila” University of Medicine and Pharmacy Bucharest, 050474 Bucharest, Romania; (E.S.); (R.-C.P.); (V.J.)
| |
Collapse
|
35
|
Park JY, Mani S, Clair G, Olson HM, Paurus VL, Ansong CK, Blundell C, Young R, Kanter J, Gordon S, Yi AY, Mainigi M, Huh DD. A microphysiological model of human trophoblast invasion during implantation. Nat Commun 2022; 13:1252. [PMID: 35292627 PMCID: PMC8924260 DOI: 10.1038/s41467-022-28663-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 02/01/2022] [Indexed: 12/12/2022] Open
Abstract
Successful establishment of pregnancy requires adhesion of an embryo to the endometrium and subsequent invasion into the maternal tissue. Abnormalities in this critical process of implantation and placentation lead to many pregnancy complications. Here we present a microenigneered system to model a complex sequence of orchestrated multicellular events that plays an essential role in early pregnancy. Our implantation-on-a-chip is capable of reconstructing the three-dimensional structural organization of the maternal-fetal interface to model the invasion of specialized fetal extravillous trophoblasts into the maternal uterus. Using primary human cells isolated from clinical specimens, we demonstrate in vivo-like directional migration of extravillous trophoblasts towards a microengineered maternal vessel and their interactions with the endothelium necessary for vascular remodeling. Through parametric variation of the cellular microenvironment and proteomic analysis of microengineered tissues, we show the important role of decidualized stromal cells as a regulator of extravillous trophoblast migration. Furthermore, our study reveals previously unknown effects of pre-implantation maternal immune cells on extravillous trophoblast invasion. This work represents a significant advance in our ability to model early human pregnancy, and may enable the development of advanced in vitro platforms for basic and clinical research of human reproduction.
Collapse
Affiliation(s)
- Ju Young Park
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Sneha Mani
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Geremy Clair
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Heather M Olson
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Vanessa L Paurus
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Charles K Ansong
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Cassidy Blundell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel Young
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessica Kanter
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Scott Gordon
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alex Y Yi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Monica Mainigi
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Dan Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
36
|
Nguyen RY, Xiao H, Gong X, Arroyo A, Cabral AT, Fischer TT, Flores KM, Zhang X, Robert ME, Ehrlich BE, Mak M. Cytoskeletal dynamics regulates stromal invasion behavior of distinct liver cancer subtypes. Commun Biol 2022; 5:202. [PMID: 35241781 PMCID: PMC8894393 DOI: 10.1038/s42003-022-03121-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 02/08/2022] [Indexed: 02/07/2023] Open
Abstract
Drug treatment against liver cancer has limited efficacy due to heterogeneous response among liver cancer subtypes. In addition, the functional biophysical phenotypes which arise from this heterogeneity and contribute to aggressive invasive behavior remain poorly understood. This study interrogated how heterogeneity in liver cancer subtypes contributes to differences in invasive phenotypes and drug response. Utilizing histological analysis, quantitative 2D invasion metrics, reconstituted 3D hydrogels, and bioinformatics, our study linked cytoskeletal dynamics to differential invasion profiles and drug resistance in liver cancer subtypes. We investigated cytoskeletal regulation in 2D and 3D culture environments using two liver cancer cell lines, SNU-475 and HepG2, chosen for their distinct cytoskeletal features and invasion profiles. For SNU-475 cells, a model for aggressive liver cancer, many cytoskeletal inhibitors abrogated 2D migration but only some suppressed 3D migration. For HepG2 cells, cytoskeletal inhibition did not significantly affect 3D migration but did affect proliferative capabilities and spheroid core growth. This study highlights cytoskeleton driven phenotypic variation, their consequences and coexistence within the same tumor, as well as efficacy of targeting biophysical phenotypes that may be masked in traditional screens against tumor growth.
Collapse
Affiliation(s)
- Ryan Y Nguyen
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Hugh Xiao
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Xiangyu Gong
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Alfredo Arroyo
- Department of Pharmacology, Yale University, New Haven, CT, USA
| | - Aidan T Cabral
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Tom T Fischer
- Department of Pharmacology, Yale University, New Haven, CT, USA
- Institute of Pharmacology, University of Heidelberg, Heidelberg, Germany
| | - Kaitlin M Flores
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Xuchen Zhang
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Marie E Robert
- Department of Pathology, Yale University, New Haven, CT, USA
| | | | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| |
Collapse
|
37
|
Bio-Inspired Proanthocyanidins from Blueberries’ Surface Coating Prevents Red Blood Cell Agglutination on Urinary Silicon-Based Catheters. COATINGS 2022. [DOI: 10.3390/coatings12020172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
Thrombosis can cause the occlusion of implantable medical devices, leading to the rejection of the device and subsequent mortality. Thrombosis is primarily induced by red blood aggregation and coagulation. The administration of anticoagulant drugs is generally used as a treatment to avoid these processes. Adverse effects such as bleeding in the event of an anticoagulant overdose, osteoporosis associated with prolonged use, hypersensitivity, and hives have been reported. New strategies such as biomolecule surface functionalization have recently been studied to overcome these problems. In this study, we report a novel coating composed of polydopamine (PDA) and proanthocyanidins (PACs) from blueberry extract to avoid red blood aggregation in short-term use medical devices such as silicone catheters. We showed that PDA formed stable films on silicone surfaces and PACs could be immobilized on PDA layers using laccase as a catalyst. The PDA–PACs films decreased surface hydrophilicity, increased surface roughness, and decreased plasma protein adsorption. The films were stable in phosphate buffer saline (PBS) and cell culture media. Furthermore, red blood cell adsorption and aggregation decreased. These effects are attributed to changes in the membrane fluidity that influences adhesion, the steric hindrance of the layers, and the low adsorption of plasma proteins on the PAC layer.
Collapse
|
38
|
Yoon D, Son J, Park JK, Nam Y. Development of the micro-patterned 3D neuronal-hydrogel model using soft-lithography for study a 3D neural network on a microelectrode array . ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2021; 2021:1234-1237. [PMID: 34891510 DOI: 10.1109/embc46164.2021.9629822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In vitro patterned neuronal models have been studied as one of the strategies to investigate the relationship between structural connectivity and functional activity of neural network. Despite the importance of three-dimensional (3D) cell models, most of these studies have been performed on two-dimensional models. In this study, we present a technique to construct the micro-pattern to 3D neuronal-hydrogel model using a micromolding in capillaries (MIMIC) technique on microelectrode array (MEA). Our technique was suitable to prevent the deformation of micro-patterned collagen model against the neuronal contracted tension during the network formation. The relationship between the growth directions of glial cells and micro-pattern direction was investigated. Lastly, we confirmed that our 3D model had synchronized activity among neurons in 3D. This model is expected to be used as a tool to study the relationship between structural connectivity and functional activity in the 3D environment.
Collapse
|
39
|
Chen Z, Kheiri S, Gevorkian A, Young EWK, Andre V, Deisenroth T, Kumacheva E. Microfluidic arrays of dermal spheroids: a screening platform for active ingredients of skincare products. LAB ON A CHIP 2021; 21:3952-3962. [PMID: 34636823 DOI: 10.1039/d1lc00619c] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Organotypic micrometre-size 3D aggregates of skin cells (multicellular spheroids) have emerged as a promising in vitro model that can be utilized as an alternative of animal models to test active ingredients (AIs) of skincare products; however, a reliable dermal spheroid-based microfluidic (MF) model with a goal of in vitro AI screening is yet to be developed. Here, we report a MF platform for the growth of massive arrays of dermal fibroblast spheroids (DFSs) in a biomimetic hydrogel under close-to-physiological flow conditions and with the capability of screening AIs for skincare products. The DFSs formed after two days of on-chip culture and, in a case study, were used in a time-efficient manner for screening the effect of vitamin C on the synthesis of collagen type I and fibronectin. The computational simulation showed that the uptake of vitamin C was dominated by the advection flux. The results of screening the benchmark AI, vitamin C, proved that DFSs can serve as a reliable in vitro dermal model. The proposed DFS-based MF platform offers a high screening capacity for AIs of skincare products, as well as drug discovery and development in dermatology.
Collapse
Affiliation(s)
- Zhengkun Chen
- Department of Chemistry, University of Toronto, 80 Saint George Street, Toronto, Ontario, M5S 3H6, Canada.
| | - Sina Kheiri
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, Ontario, M5S 3G8, Canada
| | - Albert Gevorkian
- Department of Chemistry, University of Toronto, 80 Saint George Street, Toronto, Ontario, M5S 3H6, Canada.
| | - Edmond W K Young
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, Ontario, M5S 3G8, Canada
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario, M5S 3G9, Canada
| | - Valerie Andre
- BASF Beauty Care Solutions France S.A.S, 32, rue Saint Jean de Dieu, 69007, Lyon, France
| | - Ted Deisenroth
- BASF Advanced Formulation Research North America, 500 White Plains Road, Tarrytown, New York, 10591, USA
| | - Eugenia Kumacheva
- Department of Chemistry, University of Toronto, 80 Saint George Street, Toronto, Ontario, M5S 3H6, Canada.
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario, M5S 3E5, Canada
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario, M5S 3G9, Canada
| |
Collapse
|
40
|
Pérez-Rodríguez S, Huang SA, Borau C, García-Aznar JM, Polacheck WJ. Microfluidic model of monocyte extravasation reveals the role of hemodynamics and subendothelial matrix mechanics in regulating endothelial integrity. BIOMICROFLUIDICS 2021; 15:054102. [PMID: 34548891 PMCID: PMC8443302 DOI: 10.1063/5.0061997] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/26/2021] [Indexed: 05/08/2023]
Abstract
Extravasation of circulating cells is an essential process that governs tissue inflammation and the body's response to pathogenic infection. To initiate anti-inflammatory and phagocytic functions within tissues, immune cells must cross the vascular endothelial barrier from the vessel lumen to the subluminal extracellular matrix. In this work, we present a microfluidic approach that enables the recreation of a three-dimensional, perfused endothelial vessel formed by human endothelial cells embedded within a collagen-rich matrix. Monocytes are introduced into the vessel perfusate, and we investigate the role of luminal flow and collagen concentration on extravasation. In vessels conditioned with the flow, increased monocyte adhesion to the vascular wall was observed, though fewer monocytes extravasated to the collagen hydrogel. Our results suggest that the lower rates of extravasation are due to the increased vessel integrity and reduced permeability of the endothelial monolayer. We further demonstrate that vascular permeability is a function of collagen hydrogel mass concentration, with increased collagen concentrations leading to elevated vascular permeability and increased extravasation. Collectively, our results demonstrate that extravasation of monocytes is highly regulated by the structural integrity of the endothelial monolayer. The microfluidic approach developed here allows for the dissection of the relative contributions of these cues to further understand the key governing processes that regulate circulating cell extravasation and inflammation.
Collapse
Affiliation(s)
| | - Stephanie A. Huang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27599, USA
| | | | | | | |
Collapse
|
41
|
Engineering a Vascularized Hypoxic Tumor Model for Therapeutic Assessment. Cells 2021; 10:cells10092201. [PMID: 34571851 PMCID: PMC8468635 DOI: 10.3390/cells10092201] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/23/2021] [Accepted: 08/23/2021] [Indexed: 01/23/2023] Open
Abstract
Solid tumors in advanced cancer often feature a structurally and functionally abnormal vasculature through tumor angiogenesis, which contributes to cancer progression, metastasis, and therapeutic resistances. Hypoxia is considered a major driver of angiogenesis in tumor microenvironments. However, there remains a lack of in vitro models that recapitulate both the vasculature and hypoxia in the same model with physiological resemblance to the tumor microenvironment, while allowing for high-content spatiotemporal analyses for mechanistic studies and therapeutic evaluations. We have previously constructed a hypoxia microdevice that utilizes the metabolism of cancer cells to generate an oxygen gradient in the cancer cell layer as seen in solid tumor sections. Here, we have engineered a new composite microdevice-microfluidics platform that recapitulates a vascularized hypoxic tumor. Endothelial cells were seeded in a collagen channel formed by viscous fingering, to generate a rounded vascular lumen surrounding a hypoxic tumor section composed of cancer cells embedded in a 3-D hydrogel extracellular matrix. We demonstrated that the new device can be used with microscopy-based high-content analyses to track the vascular phenotypes, morphology, and sprouting into the hypoxic tumor section over a 7-day culture, as well as the response to different cancer/stromal cells. We further evaluated the integrity/leakiness of the vascular lumen in molecular delivery, and the potential of the platform to study the movement/trafficking of therapeutic immune cells. Therefore, our new platform can be used as a model for understanding tumor angiogenesis and therapeutic delivery/efficacy in vascularized hypoxic tumors.
Collapse
|
42
|
Poventud-Fuentes I, Kwon KW, Seo J, Tomaiuolo M, Stalker TJ, Brass LF, Huh D. A Human Vascular Injury-on-a-Chip Model of Hemostasis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2004889. [PMID: 33150735 PMCID: PMC8049960 DOI: 10.1002/smll.202004889] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/28/2020] [Indexed: 05/02/2023]
Abstract
Hemostasis is an innate protective mechanism that plays a central role in maintaining the homeostasis of the vascular system during vascular injury. Studying this essential physiological process is often challenged by the difficulty of modeling and probing the complex dynamics of hemostatic responses in the native context of human blood vessels. To address this major challenge, this paper describes a microengineering approach for in vitro modeling of hemostasis. This microphysiological model replicates the living endothelium, multilayered microarchitecture, and procoagulant activity of human blood vessels, and is also equipped with a microneedle that is actuated with spatial precision to simulate penetrating vascular injuries. The system recapitulates key features of the hemostatic response to acute vascular injury as observed in vivo, including i) thrombin-driven accumulation of platelets and fibrin, ii) formation of a platelet- and fibrin-rich hemostatic plug that halts blood loss, and iii) matrix deformation driven by platelet contraction for wound closure. Moreover, the potential use of this model for drug testing applications is demonstrated by evaluating the effects of anticoagulants and antiplatelet agents that are in current clinical use. The vascular injury-on-a-chip may serve as an enabling platform for preclinical investigation of hematological disorders and emerging therapeutic approaches against them.
Collapse
Affiliation(s)
| | - Keon Woo Kwon
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jeongyun Seo
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Maurizio Tomaiuolo
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Timothy J Stalker
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Lawrence F Brass
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute of Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
43
|
Mondrinos MJ, Alisafaei F, Yi AY, Ahmadzadeh H, Lee I, Blundell C, Seo J, Osborn M, Jeon TJ, Kim SM, Shenoy VB, Huh D. Surface-directed engineering of tissue anisotropy in microphysiological models of musculoskeletal tissue. SCIENCE ADVANCES 2021; 7:7/11/eabe9446. [PMID: 33712463 PMCID: PMC7954445 DOI: 10.1126/sciadv.abe9446] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/27/2021] [Indexed: 05/11/2023]
Abstract
Here, we present an approach to model and adapt the mechanical regulation of morphogenesis that uses contractile cells as sculptors of engineered tissue anisotropy in vitro. Our method uses heterobifunctional cross-linkers to create mechanical boundary constraints that guide surface-directed sculpting of cell-laden extracellular matrix hydrogel constructs. Using this approach, we engineered linearly aligned tissues with structural and mechanical anisotropy. A multiscale in silico model of the sculpting process was developed to reveal that cell contractility increases as a function of principal stress polarization in anisotropic tissues. We also show that the anisotropic biophysical microenvironment of linearly aligned tissues potentiates soluble factor-mediated tenogenic and myogenic differentiation of mesenchymal stem cells. The application of our method is demonstrated by (i) skeletal muscle arrays to screen therapeutic modulators of acute oxidative injury and (ii) a 3D microphysiological model of lung cancer cachexia to study inflammatory and oxidative muscle injury induced by tumor-derived signals.
Collapse
Affiliation(s)
- Mark J Mondrinos
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Farid Alisafaei
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alex Y Yi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hossein Ahmadzadeh
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Insu Lee
- Department of Mechanical Engineering, Inha University, Incheon, Korea
| | - Cassidy Blundell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jeongyun Seo
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew Osborn
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tae-Joon Jeon
- Department of Biological Engineering, Inha University, Incheon, Korea
| | - Sun Min Kim
- Department of Mechanical Engineering, Inha University, Incheon, Korea
| | - Vivek B Shenoy
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA.
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
44
|
Liang D, Su W, Tan M. Advances of microfluidic intestine-on-a-chip for analyzing anti-inflammation of food. Crit Rev Food Sci Nutr 2021; 62:4418-4434. [PMID: 33480263 DOI: 10.1080/10408398.2021.1875395] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Microfluidic intestine-on-a-chip enables novel means of emulating human intestinal pathophysiology in vitro, which can potentially reduce animal testing and substitute simple 2D culture system. Though a great deal of work has been done in the development of microfluidic platforms for intestinal disease modeling and drug screening, potential investigation of the effect of bioactive food compounds on intestinal inflammation remains largely unexplored. In this review, different biomaterials and chip designs have been explored in the fabrication of intestine-on-a-chip. Other key parameters must be carefully controlled and selected, including shear stress, cell type and cell co-culture spatial configuration, etc. Appropriate techniques to quantify the barrier integrity including trans-epithelial electric resistance, specific tight junction markers and permeability measurements should be standardized and compared with in vivo data. Integration of the gut microbiome and the provision of intestinal-specific environment are the key parameters to realize the in vivo intestinal model simulation and accelerate the screening efficiency of bioactive food compounds.
Collapse
Affiliation(s)
- Duo Liang
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning, China.,National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, Liaoning, China.,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, Liaoning, China
| | - Wentao Su
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning, China.,National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, Liaoning, China.,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, Liaoning, China
| | - Mingqian Tan
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning, China.,National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, Liaoning, China.,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, Liaoning, China
| |
Collapse
|
45
|
Enríquez Á, Libring S, Field TC, Jimenez J, Lee T, Park H, Satoski D, Wendt MK, Calve S, Tepole AB, Solorio L, Lee H. High-Throughput Magnetic Actuation Platform for Evaluating the Effect of Mechanical Force on 3D Tumor Microenvironment. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2005021. [PMID: 34764824 PMCID: PMC8577425 DOI: 10.1002/adfm.202005021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Indexed: 05/03/2023]
Abstract
Accurately replicating and analyzing cellular responses to mechanical cues is vital for exploring metastatic disease progression. However, many of the existing in vitro platforms for applying mechanical stimulation seed cells on synthetic substrates. To better recapitulate physiological conditions, a novel actuating platform is developed with the ability to apply tensile strain on cells at various amplitudes and frequencies in a high-throughput multi-well culture plate using a physiologically-relevant substrate. Suspending fibrillar fibronectin across the body of the magnetic actuator provides a matrix representative of early metastasis for 3D cell culture that is not reliant on a synthetic substrate. This platform enables the culturing and analysis of various cell types in an environment that mimics the dynamic stretching of lung tissue during normal respiration. Metabolic activity, YAP activation, and morphology of breast cancer cells are analyzed within one week of cyclic stretching or static culture. Further, matrix degradation is significantly reduced in breast cancer cell lines with metastatic potential after actuation. These new findings demonstrate a clear suppressive cellular response due to cyclic stretching that has implications for a mechanical role in the dormancy and reactivation of disseminated breast cancer cells to macrometastases.
Collapse
Affiliation(s)
- Ángel Enríquez
- Weldon School of Biomedical Engineering, Birck Nanotechnology Center, Center for Implantable Devices, Purdue University, West Lafayette, IN 47907, USA
| | - Sarah Libring
- Weldon School of Biomedical Engineering, Birck Nanotechnology Center, Purdue University, West Lafayette, IN 47907, USA
| | - Tyler C. Field
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Julian Jimenez
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Taeksang Lee
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Hyunsu Park
- Weldon School of Biomedical Engineering, Birck Nanotechnology Center, Center for Implantable Devices, Purdue University, West Lafayette, IN 47907, USA
| | - Douglas Satoski
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Michael K. Wendt
- Purdue Center for Cancer Research, Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Sarah Calve
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | | | - Luis Solorio
- Purdue Center for Cancer Research, Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Hyowon Lee
- Weldon School of Biomedical Engineering, Birck Nanotechnology Center, Center for Implantable Devices, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
46
|
Akther F, Yakob SB, Nguyen NT, Ta HT. Surface Modification Techniques for Endothelial Cell Seeding in PDMS Microfluidic Devices. BIOSENSORS 2020; 10:E182. [PMID: 33228050 PMCID: PMC7699314 DOI: 10.3390/bios10110182] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/13/2020] [Accepted: 11/15/2020] [Indexed: 12/14/2022]
Abstract
Microfluidic lab-on-a-chip cell culture techniques have been gaining popularity by offering the possibility of reducing the amount of samples and reagents and greater control over cellular microenvironment. Polydimethylsiloxane (PDMS) is the commonly used polymer for microfluidic cell culture devices because of the cheap and easy fabrication techniques, non-toxicity, biocompatibility, high gas permeability, and optical transparency. However, the intrinsic hydrophobic nature of PDMS makes cell seeding challenging when applied on PDMS surface. The hydrophobicity of the PDMS surface also allows the non-specific absorption/adsorption of small molecules and biomolecules that might affect the cellular behaviour and functions. Hydrophilic modification of PDMS surface is indispensable for successful cell seeding. This review collates different techniques with their advantages and disadvantages that have been used to improve PDMS hydrophilicity to facilitate endothelial cells seeding in PDMS devices.
Collapse
Affiliation(s)
- Fahima Akther
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, QLD 4072, Australia;
- Queensland Micro-and Nanotechnology Centre, Griffith University, Brisbane, QLD 4111, Australia;
| | - Shazwani Binte Yakob
- School of Pharmacy, the University of Queensland, Brisbane, QLD 4102, Australia;
| | - Nam-Trung Nguyen
- Queensland Micro-and Nanotechnology Centre, Griffith University, Brisbane, QLD 4111, Australia;
| | - Hang T. Ta
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, QLD 4072, Australia;
- Queensland Micro-and Nanotechnology Centre, Griffith University, Brisbane, QLD 4111, Australia;
- School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia
| |
Collapse
|
47
|
Zhang B, Li J, He L, Huang H, Weng J. Bio-surface coated titanium scaffolds with cancellous bone-like biomimetic structure for enhanced bone tissue regeneration. Acta Biomater 2020; 114:431-448. [PMID: 32682055 DOI: 10.1016/j.actbio.2020.07.024] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023]
Abstract
In view of the fact that titanium (Ti)-based implants still face the problem of loosening and failure of the implants caused by the slow biological response, the low osseointegration rate and the implant bacterial infection in clinical application, we designed a cancellous bone-like biomimetic Ti scaffold using the template accumulated by sugar spheres as a pore-forming agent. And based on a modified surface mineralization process and mussel-like adhesion mechanism, a silicon-doped calcium phosphate composite coating (Van-pBNPs/pep@pSiCaP) with Vancomycin (Van)-loaded polydopamine (pDA)-modified albumin nanoparticles (Van-pBNPs) and cell adhesion peptides (GFOGER) was constructed on the surface of Ti scaffold for mimicking the extracellular matrix (ECM) microenvironment of natural bone matrix to induce greater tissue regeneration. The in vitro study demonstrated that this porous Ti scaffold with functional bio-surface could distinctly facilitate cell early adhesion and spreading, and activate the expression of α2β1 integrin receptor on the cell membrane through promoting the formation of focal adhesions (FAs) in bone marrow stromal cells (BMSCs), thus mediating greater osteogenic cell differentiation. And it could also effectively inhibit the adhesion and growth of Staphylococcus epidermidis, exhibiting good antibacterial properties. Moreover, the Van-pBNPs/pep@pSiCaP-Ti scaffolds showed enhanced in vivo bone-forming ability due to the contributions of bioactive chemical components and the natural cancellous bone-like macrostructure. This work offers a promising structural and functional bio-inspired strategy for designing metal implants with desirable ability of osteoinduction synergistically with antibacterial efficacy for promoting bone regeneration and infection prevention simultaneously. STATEMENT OF SIGNIFICANCE: This manuscript describes a new method for making porous Ti scaffolds with a natural cancellous bone-like structure. Besides, a functional bio-surface was constructed on the bionic structure, mimicking some of the functions of the collagen-rich organic matrix and inorganic CaP nanocrystallites of native ECM of bone in chemical components and biological activities. This interconnected inter-pore opening structure encouraged the migration of cells among open macro-pores within the scaffold. In addition, the functionalized surface not only improved early cell adhesion, spreading, stimulated greater osteogenic differentiation of bone-forming cells, but also endowed the scaffold with excellent antibacterial effect. The biomimetic metal implant with multiple biomedical functions designed in this study has a great clinical application potential. This study represents a feasible method for the preparation of biomimetic structure of metal implants and the improvement of their surface biological activity.
Collapse
Affiliation(s)
- Bingjun Zhang
- Key Laboratory of Advanced Technologies of Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China; State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Jia Li
- Key Laboratory of Advanced Technologies of Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
| | - Lei He
- Key Laboratory of Advanced Technologies of Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
| | - Hao Huang
- Key Laboratory of Advanced Technologies of Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
| | - Jie Weng
- Key Laboratory of Advanced Technologies of Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China.
| |
Collapse
|
48
|
Nam U, Kim S, Park J, Jeon JS. Lipopolysaccharide-Induced Vascular Inflammation Model on Microfluidic Chip. MICROMACHINES 2020; 11:mi11080747. [PMID: 32751936 PMCID: PMC7465530 DOI: 10.3390/mi11080747] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/28/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022]
Abstract
Inflammation is the initiation of defense of our body against harmful stimuli. Lipopolysaccharide (LPS), originating from outer membrane of Gram-negative bacteria, causes inflammation in the animal’s body and can develop several diseases. In order to study the inflammatory response to LPS of blood vessels in vitro, 2D models have been mainly used previously. In this study, a microfluidic device was used to investigate independent inflammatory response of endothelial cells by LPS and interaction of inflamed blood vessel with monocytic THP-1 cells. Firstly, the diffusion of LPS across the collagen gel into blood vessel was simulated using COMSOL. Then, inflammatory response to LPS in engineered blood vessel was confirmed by the expression of Intercellular Adhesion Molecule 1 (ICAM-1) and VE-cadherin of blood vessel, and THP-1 cell adhesion and migration assay. Upregulation of ICAM-1 and downregulation of VE-cadherin in an LPS-treated condition was observed compared to normal condition. In the THP-1 cell adhesion and migration assay, the number of adhered and trans-endothelial migrated THP-1 cells were not different between conditions. However, migration distance of THP-1 was longer in the LPS treatment condition. In conclusion, we recapitulated the inflammatory response of blood vessels and the interaction of THP-1 cells with blood vessels due to the diffusion of LPS.
Collapse
Affiliation(s)
- Ungsig Nam
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141S, Korea; (U.N.); (S.K.); (J.P.)
| | - Seunggyu Kim
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141S, Korea; (U.N.); (S.K.); (J.P.)
| | - Joonha Park
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141S, Korea; (U.N.); (S.K.); (J.P.)
| | - Jessie S. Jeon
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141S, Korea; (U.N.); (S.K.); (J.P.)
- KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
- Correspondence: ; Tel.: +82-42-350-3226
| |
Collapse
|
49
|
Hybrid elastomer-plastic microfluidic device as a convenient model for mimicking the blood-brain barrier in vitro. Biomed Microdevices 2019; 21:90. [PMID: 31686217 DOI: 10.1007/s10544-019-0446-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this study, we fabricated a hybrid elastomer-plastic microdevice using the silicone elastomer poly(dimethylsiloxane) (PDMS) and the plastic polycarbonate (PC), to mimic the human blood-brain barrier (BBB) in vitro. Specifically, the microchannel-imprinted elastomer was first coated with 3-aminopropyltriethoxysilane to produce amine-terminated PDMS. Then, simply by conformal contact at room temperature, the amine-functionalized PDMS was bonded to pristine PC through the formation of urethane linkages. Aside from realizing device bonding, the amine functionalization also assisted in subsequent dopamine coating to form polydopamine and provide a stable surface for culturing human endothelial cells and central nervous system-related cells (e.g., astrocytes) inside the microchannels. Successful mimicking of the BBB-like microenvironment was assessed by 3D co-culturing of human endothelial cells and astrocytes, where the microdevice was verified as an acceptable in vitro BBB model according to the following four criteria: the formation of tight junctions at the cell-cell boundaries of the endothelial cells, evaluated by the expression of the tight junction marker ZO-1; the formation of actin filaments, evaluated using rhodamine phalloidin dye; low permeability, tested using the fluorescent tracer 40-kDa FITC-dextran; and good transendothelial electrical resistance (a measure of the tight junction integrity formed between the endothelial cells). The fabricated PDMS-PC microfluidic device ensured simple yet stable device sealing, and simultaneously enhanced BBB-mimicking cell attachment, thus fulfilling all major criteria for its application as a convenient in vitro BBB model.
Collapse
|