1
|
Aye SSS, Fang Z, Wu MCL, Lim KS, Ju LA. Integrating microfluidics, hydrogels, and 3D bioprinting for personalized vessel-on-a-chip platforms. Biomater Sci 2025; 13:1131-1160. [PMID: 39834160 DOI: 10.1039/d4bm01354a] [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: 01/22/2025]
Abstract
Thrombosis, a major cause of morbidity and mortality worldwide, presents a complex challenge in cardiovascular medicine due to the intricacy of clotting mechanisms in living organisms. Traditional research approaches, including clinical studies and animal models, often yield conflicting results due to the inability to control variables in these complex systems, highlighting the need for more precise investigative tools. This review explores the evolution of in vitro thrombosis models, from conventional polydimethylsiloxane (PDMS)-based microfluidic devices to advanced hydrogel-based systems and cutting-edge 3D bioprinted vascular constructs. We discuss how these emerging technologies, particularly vessel-on-a-chip platforms, are enabling researchers to control previously unmanageable factors, thereby offering unprecedented opportunities to pinpoint specific clotting mechanisms. While PDMS-based devices offer optical transparency and fabrication ease, their inherent limitations, including non-physiological rigidity and surface properties, have driven the development of hydrogel-based systems that better mimic the extracellular matrix of blood vessels. The integration of microfluidics with biomimetic materials and tissue engineering approaches has led to the development of sophisticated models capable of simulating patient-specific vascular geometries, flow dynamics, and cellular interactions under highly controlled conditions. The advent of 3D bioprinting further enables the creation of complex, multi-layered vascular structures with precise spatial control over geometry and cellular composition. Despite significant progress, challenges remain in achieving long-term stability, incorporating immune components, and translating these models to clinical applications. By providing a comprehensive overview of current advancements and future prospects, this review aims to stimulate further innovation in thrombosis research and accelerate the development of more effective, personalized approaches to thrombosis prevention and treatment.
Collapse
Affiliation(s)
- San Seint Seint Aye
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia.
| | - Zhongqi Fang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia.
| | - Mike C L Wu
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia.
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia.
| | - Khoon S Lim
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia.
- School of Medical Sciences, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia.
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia.
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW 2006, Australia
- Heart Research Institute, Newtown, NSW 2042, Australia
| |
Collapse
|
2
|
Burčík D, Macko J, Podrojková N, Demeterová J, Stano M, Oriňak A. Role of Cell Adhesion in Cancer Metastasis Formation: A Review. ACS OMEGA 2025; 10:5193-5213. [PMID: 39989825 PMCID: PMC11840620 DOI: 10.1021/acsomega.4c08140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/10/2025] [Accepted: 01/22/2025] [Indexed: 02/25/2025]
Abstract
Intercellular adhesion is accompanied by several physical quantities and actions. In this review, we tried to collect information about the influence of surface energy and its impact on cell-cell adhesion. It still undergoes development for cancer treatment. Data on receptor-ligand interactions that occur on circulating tumor cells (CTCs) are described, and adhesion receptors as therapeutic targets are collected. Additionally, the impact of surface roughness on the interactions between CTC cells and the surface was monitored. The effects of different cell adhesion molecules (CAMs) on cell adhesion, growth, and proliferation were investigated. This review offers general principles of cell adhesion, through the blockade of adhesion with blocking drugs and inhibitors like computational models that describe the process of adhesion. Some theoretical models based on the minimum of the total free energy of interaction between CAMs and selected organic molecules have been presented. The final aim was to find information on how modulation of the surface of CTCs (by medicals or physically) inhibits cancer metastases formation.
Collapse
Affiliation(s)
- Denis Burčík
- University of P. J. Safarik
in Kosice, Faculty of Sciences,
Institute of Chemistry, Department of Physical Chemistry, Moyzesova 11, 041 01 Kosice, Slovakia
| | - Ján Macko
- University of P. J. Safarik
in Kosice, Faculty of Sciences,
Institute of Chemistry, Department of Physical Chemistry, Moyzesova 11, 041 01 Kosice, Slovakia
| | - Natália Podrojková
- University of P. J. Safarik
in Kosice, Faculty of Sciences,
Institute of Chemistry, Department of Physical Chemistry, Moyzesova 11, 041 01 Kosice, Slovakia
| | - Jana Demeterová
- University of P. J. Safarik
in Kosice, Faculty of Sciences,
Institute of Chemistry, Department of Physical Chemistry, Moyzesova 11, 041 01 Kosice, Slovakia
| | - Michal Stano
- University of P. J. Safarik
in Kosice, Faculty of Sciences,
Institute of Chemistry, Department of Physical Chemistry, Moyzesova 11, 041 01 Kosice, Slovakia
| | - Andrej Oriňak
- University of P. J. Safarik
in Kosice, Faculty of Sciences,
Institute of Chemistry, Department of Physical Chemistry, Moyzesova 11, 041 01 Kosice, Slovakia
| |
Collapse
|
3
|
Cai B, Kilian D, Ghorbani S, Roth JG, Seymour AJ, Brunel LG, Mejia DR, Rios RJ, Szabo IM, Iranzo SC, Perez A, Rao RR, Shin S, Heilshorn SC. One-step bioprinting of endothelialized, self-supporting arterial and venous networks. Biofabrication 2025; 17:025012. [PMID: 39819775 DOI: 10.1088/1758-5090/adab26] [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] [Received: 09/24/2024] [Accepted: 01/16/2025] [Indexed: 01/19/2025]
Abstract
Advances in biofabrication have enabled the generation of freeform perfusable networks mimicking vasculature. However, key challenges remain in the effective endothelialization of these complex, vascular-like networks, including cell uniformity, seeding efficiency, and the ability to pattern multiple cell types. To overcome these challenges, we present an integrated fabrication and endothelialization strategy to directly generate branched, endothelial cell-lined networks using a diffusion-based, embedded 3D bioprinting process. In this strategy, a gelatin microparticle sacrificial ink delivering both cells and crosslinkers is extruded into a crosslinkable gel precursor support bath. A self-supporting, perfusable structure is formed by diffusion-induced crosslinking, after which the sacrificial ink is melted to allow cell release and adhesion to the printed lumen. This approach produces a uniform cell lining throughout networks with complex branching geometries, which are challenging to uniformly and efficiently endothelialize using conventional perfusion-based approaches. Furthermore, the biofabrication process enables high cell viability (>90%) and the formation of a confluent endothelial layer providing vascular-mimetic barrier function and shear stress response. Leveraging this strategy, we demonstrate for the first time the patterning of multiple endothelial cell types, including arterial and venous cells, within a single arterial-venous-like network. Altogether, this strategy enables the fabrication of multi-cellular engineered vasculature with enhanced geometric complexity and phenotypic heterogeneity.
Collapse
Affiliation(s)
- Betty Cai
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, United States of America
| | - David Kilian
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, United States of America
| | - Sadegh Ghorbani
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, United States of America
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Julien G Roth
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, United States of America
| | - Alexis J Seymour
- Department of Bioengineering, Stanford University, Stanford, CA 94305, United States of America
| | - Lucia G Brunel
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, United States of America
| | - Daniel Ramos Mejia
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, United States of America
| | - Ricardo J Rios
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, United States of America
| | - Isabella M Szabo
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, United States of America
| | - Sean Chryz Iranzo
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, United States of America
| | - Andy Perez
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, United States of America
| | - Rameshwar R Rao
- Ben Towne Center for Childhood Cancer and Blood Disorders Research, Seattle Children's Research Institute, Seattle, WA 98101, United States of America
- Division of Pediatric Hematology, Oncology, Bone Marrow Transplant, and Cellular Therapies, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98105, United States of America
| | - Sungchul Shin
- Department of Agriculture, Forestry, and Bioresources, Seoul National University, Seoul 08826, Republic of Korea
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, United States of America
| |
Collapse
|
4
|
Capalbo S, Polyakova A, El Imane Z, Khan I, Kawai T, Shindo S, Salinas M. A Comprehensive Review of Contemporary Bioreactors for Vascular Inflammation Studies. Inflammation 2025:10.1007/s10753-024-02231-y. [PMID: 39903422 DOI: 10.1007/s10753-024-02231-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 12/11/2024] [Accepted: 12/26/2024] [Indexed: 02/06/2025]
Abstract
The field of vascular biology has advanced significantly with bioreactor systems, which have become essential tools for investigating the mechanisms of vascular inflammatory diseases such as atherosclerosis, vasculitis, and aneurysms. These bioreactors allow researchers to recreate specific vascular environments, providing a controlled setting for studying the effects of blood flow, mechanical stress, and biochemical factors on vascular tissues. Through these systems, researchers can explore how physical and chemical cues contribute to disease processes and cellular responses, enhancing our understanding of disease progression. Bioreactor studies have demonstrated that hemodynamic forces, particularly shear stress, influence endothelial cell behavior and play a role in vascular pathologies. For instance, in atherosclerosis, disturbed flow patterns are associated with endothelial dysfunction and plaque development. By simulating these conditions, bioreactors provide insight into the effects of mechanical forces on vascular wall biology, highlighting how altered flow can contribute to disease. Bioreactors also support studies on the impacts of pulsatile flow and circumferential stress, allowing a closer approximation of physiological environments. Beyond flow dynamics, these systems facilitate investigation into how vascular cells respond to biochemical signals, inflammatory markers, and therapeutic interventions. This integrated approach allows for a more complete picture of the factors involved in vascular disease. Recent advancements, such as vessel-on-a-chip models and artery-mimicking setups, extend the capabilities of bioreactors by enabling researchers to model a broader range of conditions relevant to human physiology. In vasculitis studies, bioreactors help explore immune interactions with endothelial cells, especially with stem cell-derived cells that replicate patient-specific responses. Bioreactors also play a role in vascular tissue engineering, particularly in assessing materials and scaffold-free designs that may reduce inflammation in vascular grafts. These efforts contribute to the ongoing search for more compatible graft materials, with the potential to improve outcomes in clinical applications. This review provides a comprehensive overview of bioreactor technologies applied in vascular inflammation research, examining their designs, applications, and contributions to disease modeling. Organized into sections on bioreactor configurations, flow dynamics, biochemical interactions, and tissue engineering applications, the review concludes by discussing recent innovations and highlighting directions for future research, underscoring the role of bioreactors in bridging laboratory studies with insights into vascular disease.
Collapse
Affiliation(s)
- Solana Capalbo
- Nova Southeastern University, College of Computing and Engineering, Davie, FL, USA
| | | | - Zayd El Imane
- Nova Southeastern University, College of Computing and Engineering, Davie, FL, USA
| | - Izza Khan
- Nova Southeastern University, College of Computing and Engineering, Davie, FL, USA
| | - Toshihisa Kawai
- Department of Oral Science and Translational Research, Nova Southeastern University College of Dental Medicine, Fort Lauderdale, FL, USA
| | - Satoru Shindo
- Department of Oral Science and Translational Research, Nova Southeastern University College of Dental Medicine, Fort Lauderdale, FL, USA
| | - Manuel Salinas
- Nova Southeastern University, College of Computing and Engineering, Davie, FL, USA.
| |
Collapse
|
5
|
Gu X, Wei S, Lv X. Circulating tumor cells: from new biological insights to clinical practice. Signal Transduct Target Ther 2024; 9:226. [PMID: 39218931 PMCID: PMC11366768 DOI: 10.1038/s41392-024-01938-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 05/31/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
The primary reason for high mortality rates among cancer patients is metastasis, where tumor cells migrate through the bloodstream from the original site to other parts of the body. Recent advancements in technology have significantly enhanced our comprehension of the mechanisms behind the bloodborne spread of circulating tumor cells (CTCs). One critical process, DNA methylation, regulates gene expression and chromosome stability, thus maintaining dynamic equilibrium in the body. Global hypomethylation and locus-specific hypermethylation are examples of changes in DNA methylation patterns that are pivotal to carcinogenesis. This comprehensive review first provides an overview of the various processes that contribute to the formation of CTCs, including epithelial-mesenchymal transition (EMT), immune surveillance, and colonization. We then conduct an in-depth analysis of how modifications in DNA methylation within CTCs impact each of these critical stages during CTC dissemination. Furthermore, we explored potential clinical implications of changes in DNA methylation in CTCs for patients with cancer. By understanding these epigenetic modifications, we can gain insights into the metastatic process and identify new biomarkers for early detection, prognosis, and targeted therapies. This review aims to bridge the gap between basic research and clinical application, highlighting the significance of DNA methylation in the context of cancer metastasis and offering new avenues for improving patient outcomes.
Collapse
Affiliation(s)
- Xuyu Gu
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shiyou Wei
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xin Lv
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
6
|
Fareez UNM, Naqvi SAA, Mahmud M, Temirel M. Computational Fluid Dynamics (CFD) Analysis of Bioprinting. Adv Healthc Mater 2024; 13:e2400643. [PMID: 38648623 DOI: 10.1002/adhm.202400643] [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] [Received: 02/20/2024] [Revised: 04/14/2024] [Indexed: 04/25/2024]
Abstract
Regenerative medicine has evolved with the rise of tissue engineering due to advancements in healthcare and technology. In recent years, bioprinting has been an upcoming approach to traditional tissue engineering practices, through the fabrication of functional tissue by its layer-by-layer deposition process. This overcomes challenges such as irregular cell distribution and limited cell density, and it can potentially address organ shortages, increasing transplant options. Bioprinting fully functional organs is a long stretch but the advancement is rapidly growing due to its precision and compatibility with complex geometries. Computational Fluid Dynamics (CFD), a carestone of computer-aided engineering, has been instrumental in assisting bioprinting research and development by cutting costs and saving time. CFD optimizes bioprinting by testing parameters such as shear stress, diffusivity, and cell viability, reducing repetitive experiments and aiding in material selection and bioprinter nozzle design. This review discusses the current application of CFD in bioprinting and its potential to enhance the technology that can contribute to the evolution of regenerative medicine.
Collapse
Affiliation(s)
- Umar Naseef Mohamed Fareez
- Mechanical Engineering Department, School of Engineering, Abdullah Gul University, Kayseri, 38080, Turkey
| | - Syed Ali Arsal Naqvi
- Mechanical Engineering Department, School of Engineering, Abdullah Gul University, Kayseri, 38080, Turkey
| | - Makame Mahmud
- Mechanical Engineering Department, School of Engineering, Abdullah Gul University, Kayseri, 38080, Turkey
| | - Mikail Temirel
- Mechanical Engineering Department, School of Engineering, Abdullah Gul University, Kayseri, 38080, Turkey
| |
Collapse
|
7
|
He J, Duan P, Liu Y, Feng T, Wang S, Lin X, Xie J, Liu X. Unveiling the Impact of Hemodynamics on Endothelial Inflammation-Mediated Hepatocellular Carcinoma Metastasis Using a Biomimetic Vascular Flow Model. Adv Healthc Mater 2024; 13:e2304439. [PMID: 38486060 DOI: 10.1002/adhm.202304439] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/28/2024] [Indexed: 03/23/2024]
Abstract
Hepatocellular carcinoma (HCC) hematogenous dissemination is a leading cause of HCC-related deaths. The inflammatory facilitates this process by promoting the adhesion and invasion of tumor cells in the circulatory system. But the contribution of hemodynamics to this process remains poorly understood due to the lack of a suitable vascular flow model for investigation. This study develops a vascular flow model to examine the impact of hemodynamics on endothelial inflammation-mediated HCC metastasis. This work finds the increasing shear stress will reduce the recruitment of HCC cells by disturbing adhesion forces between endothelium and HCC cells. However, this reduction will be restored by the inflammation. When applying high FSS (4-6 dyn cm-2) to the inflammatory endothelium, there will be a 4.8-fold increase in HCC cell adhesions compared to normal condition. Nevertheless, the increase fold of cell adhesions is inapparent, around 1.5-fold, with low and medium FSS. This effect can be attributed to the FSS-induced upregulation of ICAM-1 and VCAM-1 of the inflammatory endothelium, which serve to strengthen cell binding forces. These findings indicate that hemodynamics plays a key role in HCC metastasis during endothelial inflammation by regulating the expression of adhesion-related factors.
Collapse
Affiliation(s)
- Jia He
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Peiyan Duan
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Yi Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Tang Feng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shuo Wang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Xinyi Lin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Jing Xie
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
8
|
Pelin G, Sonmez M, Pelin CE. The Use of Additive Manufacturing Techniques in the Development of Polymeric Molds: A Review. Polymers (Basel) 2024; 16:1055. [PMID: 38674976 PMCID: PMC11054453 DOI: 10.3390/polym16081055] [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: 03/09/2024] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
The continuous growth of additive manufacturing in worldwide industrial and research fields is driven by its main feature which allows the customization of items according to the customers' requirements and limitations. There is an expanding competitiveness in the product development sector as well as applicative research that serves special-use domains. Besides the direct use of additive manufacturing in the production of final products, 3D printing is a viable solution that can help manufacturers and researchers produce their support tooling devices (such as molds and dies) more efficiently, in terms of design complexity and flexibility, timeframe, costs, and material consumption reduction as well as functionality and quality enhancements. The compatibility of the features of 3D printing of molds with the requirements of low-volume production and individual-use customized items development makes this class of techniques extremely attractive to a multitude of areas. This review paper presents a synthesis of the use of 3D-printed polymeric molds in the main applications where molds exhibit a major role, from industrially oriented ones (injection, casting, thermoforming, vacuum forming, composite fabrication) to research or single-use oriented ones (tissue engineering, biomedicine, soft lithography), with an emphasis on the benefits of using 3D-printed polymeric molds, compared to traditional tooling.
Collapse
Affiliation(s)
- George Pelin
- INCAS—National Institute for Aerospace Research “Elie Carafoli”, Bd. Iuliu Maniu 220, 061126 Bucharest, Romania;
| | - Maria Sonmez
- INCDTP-ICPI—National Research and Development Institute for Textile and Leather—Division Leather and Footwear Research Institute, Ion Minulescu St. 93, 031215 Bucharest, Romania;
| | - Cristina-Elisabeta Pelin
- INCAS—National Institute for Aerospace Research “Elie Carafoli”, Bd. Iuliu Maniu 220, 061126 Bucharest, Romania;
| |
Collapse
|
9
|
Bersini S, Arrigoni C, Talò G, Candrian C, Moretti M. Complex or not too complex? One size does not fit all in next generation microphysiological systems. iScience 2024; 27:109199. [PMID: 38433912 PMCID: PMC10904982 DOI: 10.1016/j.isci.2024.109199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024] Open
Abstract
In the attempt to overcome the increasingly recognized shortcomings of existing in vitro and in vivo models, researchers have started to implement alternative models, including microphysiological systems. First examples were represented by 2.5D systems, such as microfluidic channels covered by cell monolayers as blood vessel replicates. In recent years, increasingly complex microphysiological systems have been developed, up to multi-organ on chip systems, connecting different 3D tissues in the same device. However, such an increase in model complexity raises several questions about their exploitation and implementation into industrial and clinical applications, ranging from how to improve their reproducibility, robustness, and reliability to how to meaningfully and efficiently analyze the huge amount of heterogeneous datasets emerging from these devices. Considering the multitude of envisaged applications for microphysiological systems, it appears now necessary to tailor their complexity on the intended purpose, being academic or industrial, and possibly combine results deriving from differently complex stages to increase their predictive power.
Collapse
Affiliation(s)
- Simone Bersini
- Regenerative Medicine Technologies Lab, Laboratories for Translational Research, Ente Ospedaliero Cantonale, via Chiesa 5, 6500 Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, via Tesserete 46, 6900 Lugano, Switzerland
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), via Buffi 13, 6900 Lugano, Switzerland
| | - Chiara Arrigoni
- Regenerative Medicine Technologies Lab, Laboratories for Translational Research, Ente Ospedaliero Cantonale, via Chiesa 5, 6500 Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, via Tesserete 46, 6900 Lugano, Switzerland
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), via Buffi 13, 6900 Lugano, Switzerland
| | - Giuseppe Talò
- Cell and Tissue Engineering Laboratory, IRCCS Ospedale Galeazzi – Sant’Ambrogio, via Cristina Belgioioso 173, 20157 Milano, Italy
| | - Christian Candrian
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, via Tesserete 46, 6900 Lugano, Switzerland
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), via Buffi 13, 6900 Lugano, Switzerland
| | - Matteo Moretti
- Regenerative Medicine Technologies Lab, Laboratories for Translational Research, Ente Ospedaliero Cantonale, via Chiesa 5, 6500 Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, via Tesserete 46, 6900 Lugano, Switzerland
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), via Buffi 13, 6900 Lugano, Switzerland
- Cell and Tissue Engineering Laboratory, IRCCS Ospedale Galeazzi – Sant’Ambrogio, via Cristina Belgioioso 173, 20157 Milano, Italy
| |
Collapse
|
10
|
Koskinen LM, Nieminen L, Arjonen A, Guzmán C, Peurla M, Peuhu E. Spatial Engineering of Mammary Epithelial Cell Cultures with 3D Bioprinting Reveals Growth Control by Branch Point Proximity. J Mammary Gland Biol Neoplasia 2024; 29:5. [PMID: 38416267 PMCID: PMC10902034 DOI: 10.1007/s10911-024-09557-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/20/2024] [Indexed: 02/29/2024] Open
Abstract
The three-dimensional (3D) structure of the ductal epithelium and the surrounding extracellular matrix (ECM) are integral aspects of the breast tissue, and they have important roles during mammary gland development, function and malignancy. However, the architecture of the branched mammary epithelial network is poorly recapitulated in the current in vitro models. 3D bioprinting is an emerging approach to improve tissue-mimicry in cell culture. Here, we developed and optimized a protocol for 3D bioprinting of normal and cancerous mammary epithelial cells into a branched Y-shape to study the role of cell positioning in the regulation of cell proliferation and invasion. Non-cancerous cells formed continuous 3D cell networks with several organotypic features, whereas the ductal carcinoma in situ (DCIS) -like cancer cells exhibited aberrant basal polarization and defective formation of the basement membrane (BM). Quantitative analysis over time demonstrated that both normal and cancerous cells proliferate more at the branch tips compared to the trunk region of the 3D-bioprinted cultures, and particularly at the tip further away from the branch point. The location-specific rate of proliferation was independent of TGFβ signaling but invasion of the DCIS-like breast cancer cells was reduced upon the inhibition of TGFβ. Thus, our data demonstrate that the 3D-bioprinted cells can sense their position in the branched network of cells and proliferate at the tips, thus recapitulating this feature of mammary epithelial branching morphogenesis. In all, our results demonstrate the capacity of the developed 3D bioprinting method for quantitative analysis of the relationships between tissue structure and cell behavior in breast morphogenesis and cancer.
Collapse
Affiliation(s)
- Leena M Koskinen
- Institute of Biomedicine, Cancer Laboratory FICAN West, University of Turku, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | | | | | | | - Markus Peurla
- Institute of Biomedicine, Cancer Laboratory FICAN West, University of Turku, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Emilia Peuhu
- Institute of Biomedicine, Cancer Laboratory FICAN West, University of Turku, Turku, Finland.
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
| |
Collapse
|
11
|
Wu Z, Huang D, Wang J, Zhao Y, Sun W, Shen X. Engineering Heterogeneous Tumor Models for Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304160. [PMID: 37946674 PMCID: PMC10767453 DOI: 10.1002/advs.202304160] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/16/2023] [Indexed: 11/12/2023]
Abstract
Tumor tissue engineering holds great promise for replicating the physiological and behavioral characteristics of tumors in vitro. Advances in this field have led to new opportunities for studying the tumor microenvironment and exploring potential anti-cancer therapeutics. However, the main obstacle to the widespread adoption of tumor models is the poor understanding and insufficient reconstruction of tumor heterogeneity. In this review, the current progress of engineering heterogeneous tumor models is discussed. First, the major components of tumor heterogeneity are summarized, which encompasses various signaling pathways, cell proliferations, and spatial configurations. Then, contemporary approaches are elucidated in tumor engineering that are guided by fundamental principles of tumor biology, and the potential of a bottom-up approach in tumor engineering is highlighted. Additionally, the characterization approaches and biomedical applications of tumor models are discussed, emphasizing the significant role of engineered tumor models in scientific research and clinical trials. Lastly, the challenges of heterogeneous tumor models in promoting oncology research and tumor therapy are described and key directions for future research are provided.
Collapse
Affiliation(s)
- Zhuhao Wu
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Danqing Huang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Jinglin Wang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Yuanjin Zhao
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalWenzhou Medical UniversityWenzhou325035China
| | - Weijian Sun
- Department of Gastrointestinal SurgeryThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Xian Shen
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalWenzhou Medical UniversityWenzhou325035China
| |
Collapse
|
12
|
Maung Ye SS, Phng LK. A cell-and-plasma numerical model reveals hemodynamic stress and flow adaptation in zebrafish microvessels after morphological alteration. PLoS Comput Biol 2023; 19:e1011665. [PMID: 38048371 PMCID: PMC10721208 DOI: 10.1371/journal.pcbi.1011665] [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: 05/23/2023] [Revised: 12/14/2023] [Accepted: 11/06/2023] [Indexed: 12/06/2023] Open
Abstract
The development of a functional cardiovascular system ensures a sustainable oxygen, nutrient and hormone delivery system for successful embryonic development and homeostasis in adulthood. While early vessels are formed by biochemical signaling and genetic programming, the onset of blood flow provides mechanical cues that participate in vascular remodeling of the embryonic vascular system. The zebrafish is a prolific animal model for studying the quantitative relationship between blood flow and vascular morphogenesis due to a combination of favorable factors including blood flow visualization in optically transparent larvae. In this study, we have developed a cell-and-plasma blood transport model using computational fluid dynamics (CFD) to understand how red blood cell (RBC) partitioning affect lumen wall shear stress (WSS) and blood pressure in zebrafish trunk blood vascular networks with altered rheology and morphology. By performing live imaging of embryos with reduced hematocrit, we discovered that cardiac output and caudal artery flow rates were maintained. These adaptation trends were recapitulated in our CFD models, which showed reduction in network WSS via viscosity reduction in the caudal artery/vein and via pressure gradient weakening in the intersegmental vessels (ISVs). Embryos with experimentally reduced lumen diameter showed reduced cardiac output and caudal artery flow rate. Factoring in this trend into our CFD models, simulations highlighted that lumen diameter reduction increased vessel WSS but this increase was mitigated by flow reduction due to the adaptive network pressure gradient weakening. Additionally, hypothetical network CFD models with different vessel lumen diameter distribution characteristics indicated the significance of axial variation in lumen diameter and cross-sectional shape for establishing physiological WSS gradients along ISVs. In summary, our work demonstrates how both experiment-driven and hypothetical CFD modeling can be employed for the study of blood flow physiology during vascular remodeling.
Collapse
Affiliation(s)
- Swe Soe Maung Ye
- Laboratory for Vascular Morphogenesis, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe, Japan
| | - Li-Kun Phng
- Laboratory for Vascular Morphogenesis, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe, Japan
| |
Collapse
|
13
|
Park W, Lee JS, Gao G, Kim BS, Cho DW. 3D bioprinted multilayered cerebrovascular conduits to study cancer extravasation mechanism related with vascular geometry. Nat Commun 2023; 14:7696. [PMID: 38001146 PMCID: PMC10673893 DOI: 10.1038/s41467-023-43586-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Cerebral vessels are composed of highly complex structures that facilitate blood perfusion necessary for meeting the high energy demands of the brain. Their geometrical complexities alter the biophysical behavior of circulating tumor cells in the brain, thereby influencing brain metastasis. However, recapitulation of the native cerebrovascular microenvironment that shows continuities between vascular geometry and metastatic cancer development has not been accomplished. Here, we apply an in-bath 3D triaxial bioprinting technique and a brain-specific hybrid bioink containing an ionically crosslinkable hydrogel to generate a mature three-layered cerebrovascular conduit with varying curvatures to investigate the physical and molecular mechanisms of cancer extravasation in vitro. We show that more tumor cells adhere at larger vascular curvature regions, suggesting that prolongation of tumor residence time under low velocity and wall shear stress accelerates the molecular signatures of metastatic potential, including endothelial barrier disruption, epithelial-mesenchymal transition, inflammatory response, and tumorigenesis. These findings provide insights into the underlying mechanisms driving brain metastases and facilitate future advances in pharmaceutical and medical research.
Collapse
Affiliation(s)
- Wonbin Park
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Jae-Seong Lee
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan, Republic of Korea
| | - Ge Gao
- School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan, Republic of Korea.
- Medical Research Institute, Pusan National University, Yangsan, Republic of Korea.
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Republic of Korea.
| |
Collapse
|
14
|
Han X, Saiding Q, Cai X, Xiao Y, Wang P, Cai Z, Gong X, Gong W, Zhang X, Cui W. Intelligent Vascularized 3D/4D/5D/6D-Printed Tissue Scaffolds. NANO-MICRO LETTERS 2023; 15:239. [PMID: 37907770 PMCID: PMC10618155 DOI: 10.1007/s40820-023-01187-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 07/25/2023] [Indexed: 11/02/2023]
Abstract
Blood vessels are essential for nutrient and oxygen delivery and waste removal. Scaffold-repairing materials with functional vascular networks are widely used in bone tissue engineering. Additive manufacturing is a manufacturing technology that creates three-dimensional solids by stacking substances layer by layer, mainly including but not limited to 3D printing, but also 4D printing, 5D printing and 6D printing. It can be effectively combined with vascularization to meet the needs of vascularized tissue scaffolds by precisely tuning the mechanical structure and biological properties of smart vascular scaffolds. Herein, the development of neovascularization to vascularization to bone tissue engineering is systematically discussed in terms of the importance of vascularization to the tissue. Additionally, the research progress and future prospects of vascularized 3D printed scaffold materials are highlighted and presented in four categories: functional vascularized 3D printed scaffolds, cell-based vascularized 3D printed scaffolds, vascularized 3D printed scaffolds loaded with specific carriers and bionic vascularized 3D printed scaffolds. Finally, a brief review of vascularized additive manufacturing-tissue scaffolds in related tissues such as the vascular tissue engineering, cardiovascular system, skeletal muscle, soft tissue and a discussion of the challenges and development efforts leading to significant advances in intelligent vascularized tissue regeneration is presented.
Collapse
Affiliation(s)
- Xiaoyu Han
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, People's Republic of China
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong, People's Republic of China
| | - Qimanguli Saiding
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, People's Republic of China
| | - Xiaolu Cai
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, People's Republic of China
| | - Yi Xiao
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Peng Wang
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong, People's Republic of China
| | - Zhengwei Cai
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, People's Republic of China
| | - Xuan Gong
- University of Texas Southwestern Medical Center, Dallas, TX, 75390-9096, USA
| | - Weiming Gong
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong, People's Republic of China.
| | - Xingcai Zhang
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, People's Republic of China.
| |
Collapse
|
15
|
Schoenborn S, Lorenz T, Kuo K, Fletcher DF, Woodruff MA, Pirola S, Allenby MC. Fluid-structure interactions of peripheral arteries using a coupled in silico and in vitro approach. Comput Biol Med 2023; 165:107474. [PMID: 37703711 DOI: 10.1016/j.compbiomed.2023.107474] [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: 07/12/2023] [Revised: 08/21/2023] [Accepted: 09/04/2023] [Indexed: 09/15/2023]
Abstract
Vascular compliance is considered both a cause and a consequence of cardiovascular disease and a significant factor in the mid- and long-term patency of vascular grafts. However, the biomechanical effects of localised changes in compliance cannot be satisfactorily studied with the available medical imaging technologies or surgical simulation materials. To address this unmet need, we developed a coupled silico-vitro platform which allows for the validation of numerical fluid-structure interaction results as a numerical model and physical prototype. This numerical one-way and two-way fluid-structure interaction study is based on a three-dimensional computer model of an idealised femoral artery which is validated against patient measurements derived from the literature. The numerical results are then compared with experimental values collected from compliant arterial phantoms via direct pressurisation and ring tensile testing. Phantoms within a compliance range of 1.4-68.0%/100 mmHg were fabricated via additive manufacturing and silicone casting, then mechanically characterised via ring tensile testing and optical analysis under direct pressurisation with moderately statistically significant differences in measured compliance ranging between 10 and 20% for the two methods. One-way fluid-structure interaction coupling underestimated arterial wall compliance by up to 14.7% compared with two-way coupled models. Overall, Solaris™ (Smooth-On) matched the compliance range of the numerical and in vivo patient models most closely out of the tested silicone materials. Our approach is promising for vascular applications where mechanical compliance is especially important, such as the study of diseases which commonly affect arterial wall stiffness, such as atherosclerosis, and the model-based design, surgical training, and optimisation of vascular prostheses.
Collapse
Affiliation(s)
- S Schoenborn
- BioMimetic Systems Engineering (BMSE) Lab, School of Chemical Engineering, University of Queensland (UQ), St Lucia, QLD, 4072, Australia; Biofabrication and Tissue Morphology (BTM) Group, Faculty of Engineering, Centre for Biomedical Technologies, Queensland University of Technology (QUT), Kelvin Grove, QLD, 4059, Australia
| | - T Lorenz
- Institute of Textile Technology, RWTH Aachen University, 52074, Aachen, Germany
| | - K Kuo
- Institute of Textile Technology, RWTH Aachen University, 52074, Aachen, Germany
| | - D F Fletcher
- School of Chemical and Biomolecular Engineering, University of Sydney, Darlington, NSW, 2006, Australia
| | - M A Woodruff
- Biofabrication and Tissue Morphology (BTM) Group, Faculty of Engineering, Centre for Biomedical Technologies, Queensland University of Technology (QUT), Kelvin Grove, QLD, 4059, Australia
| | - S Pirola
- BHF Centre of Research Excellence, Faculty of Medicine, Institute of Clinical Sciences, Imperial College London, South Kensington Campus, London, SW7 2AZ, United Kingdom; Department of Biomechanical Engineering, Faculty of Mechanical Engineering (3me), Delft University of Technology (TUD), Delft, the Netherlands
| | - M C Allenby
- BioMimetic Systems Engineering (BMSE) Lab, School of Chemical Engineering, University of Queensland (UQ), St Lucia, QLD, 4072, Australia; Biofabrication and Tissue Morphology (BTM) Group, Faculty of Engineering, Centre for Biomedical Technologies, Queensland University of Technology (QUT), Kelvin Grove, QLD, 4059, Australia.
| |
Collapse
|
16
|
Sigdel I, Ofori-Kwafo A, Heizelman RJ, Nestor-Kalinoski A, Prabhakarpandian B, Tiwari AK, Tang Y. Biomimetic on-chip assay reveals the anti-metastatic potential of a novel thienopyrimidine compound in triple-negative breast cancer cell lines. Front Bioeng Biotechnol 2023; 11:1227119. [PMID: 37840664 PMCID: PMC10569307 DOI: 10.3389/fbioe.2023.1227119] [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: 05/22/2023] [Accepted: 09/19/2023] [Indexed: 10/17/2023] Open
Abstract
Introduction: This study presents a microfluidic tumor microenvironment (TME) model for evaluating the anti-metastatic efficacy of a novel thienopyrimidines analog with anti-cancer properties utilizing an existing commercial platform. The microfluidic device consists of a tissue compartment flanked by vascular channels, allowing for the co-culture of multiple cell types and providing a wide range of culturing conditions in one device. Methods: Human metastatic, drug-resistant triple-negative breast cancer (TNBC) cells (SUM159PTX) and primary human umbilical vein endothelial cells (HUVEC) were used to model the TME. A dynamic perfusion scheme was employed to facilitate EC physiological function and lumen formation. Results: The measured permeability of the EC barrier was comparable to observed microvessels permeability in vivo. The TNBC cells formed a 3D tumor, and co-culture with HUVEC negatively impacted EC barrier integrity. The microfluidic TME was then used to model the intravenous route of drug delivery. Paclitaxel (PTX) and a novel non-apoptotic agent TPH104c were introduced via the vascular channels and successfully reached the TNBC tumor, resulting in both time and concentration-dependent tumor growth inhibition. PTX treatment significantly reduced EC barrier integrity, highlighting the adverse effects of PTX on vascular ECs. TPH104c preserved EC barrier integrity and prevented TNBC intravasation. Discussion: In conclusion, this study demonstrates the potential of microfluidics for studying complex biological processes in a controlled environment and evaluating the efficacy and toxicity of chemotherapeutic agents in more physiologically relevant conditions. This model can be a valuable tool for screening potential anticancer drugs and developing personalized cancer treatment strategies.
Collapse
Affiliation(s)
- Indira Sigdel
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| | - Awurama Ofori-Kwafo
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| | - Robert J. Heizelman
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Andrea Nestor-Kalinoski
- Department of Surgery, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
| | | | - Amit K. Tiwari
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Yuan Tang
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| |
Collapse
|
17
|
Kim SJ, Kim MG, Kim J, Jeon JS, Park J, Yi HG. Bioprinting Methods for Fabricating In Vitro Tubular Blood Vessel Models. CYBORG AND BIONIC SYSTEMS 2023; 4:0043. [PMID: 37533545 PMCID: PMC10393580 DOI: 10.34133/cbsystems.0043] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/26/2023] [Indexed: 08/04/2023] Open
Abstract
Dysfunctional blood vessels are implicated in various diseases, including cardiovascular diseases, neurodegenerative diseases, and cancer. Several studies have attempted to prevent and treat vascular diseases and understand interactions between these diseases and blood vessels across different organs and tissues. Initial studies were conducted using 2-dimensional (2D) in vitro and animal models. However, these models have difficulties in mimicking the 3D microenvironment in human, simulating kinetics related to cell activities, and replicating human pathophysiology; in addition, 3D models involve remarkably high costs. Thus, in vitro bioengineered models (BMs) have recently gained attention. BMs created through biofabrication based on tissue engineering and regenerative medicine are breakthrough models that can overcome limitations of 2D and animal models. They can also simulate the natural microenvironment in a patient- and target-specific manner. In this review, we will introduce 3D bioprinting methods for fabricating bioengineered blood vessel models, which can serve as the basis for treating and preventing various vascular diseases. Additionally, we will describe possible advancements from tubular to vascular models. Last, we will discuss specific applications, limitations, and future perspectives of fabricated BMs.
Collapse
Affiliation(s)
- Seon-Jin Kim
- Department of Rural and Biosystems Engineering, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Min-Gyun Kim
- Department of Convergence Biosystems Engineering, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jangho Kim
- Department of Rural and Biosystems Engineering, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, Republic of Korea
- Department of Convergence Biosystems Engineering, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 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, Daejeon, Republic of Korea
| | - Jinsoo Park
- Department of Mechanical Engineering, Chonnam National University, Republic of Korea
| | - Hee-Gyeong Yi
- Department of Rural and Biosystems Engineering, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, Republic of Korea
- Department of Convergence Biosystems Engineering, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
18
|
Wasson EM, He W, Ahlquist J, Hynes WF, Triplett MG, Hinckley A, Karelehto E, Gray-Sherr DR, Friedman CF, Robertson C, Shusteff M, Warren R, Coleman MA, Moya ML, Wheeler EK. A perfused multi-well bioreactor platform to assess tumor organoid response to a chemotherapeutic gradient. Front Bioeng Biotechnol 2023; 11:1193430. [PMID: 37324446 PMCID: PMC10264793 DOI: 10.3389/fbioe.2023.1193430] [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: 03/24/2023] [Accepted: 05/17/2023] [Indexed: 06/17/2023] Open
Abstract
There is an urgent need to develop new therapies for colorectal cancer that has metastasized to the liver and, more fundamentally, to develop improved preclinical platforms of colorectal cancer liver metastases (CRCLM) to screen therapies for efficacy. To this end, we developed a multi-well perfusable bioreactor capable of monitoring CRCLM patient-derived organoid response to a chemotherapeutic gradient. CRCLM patient-derived organoids were cultured in the multi-well bioreactor for 7 days and the subsequently established gradient in 5-fluorouracil (5-FU) concentration resulted in a lower IC50 in the region near the perfusion channel versus the region far from the channel. We compared behaviour of organoids in this platform to two commonly used PDO culture models: organoids in media and organoids in a static (no perfusion) hydrogel. The bioreactor IC50 values were significantly higher than IC50 values for organoids cultured in media whereas only the IC50 for organoids far from the channel were significantly different than organoids cultured in the static hydrogel condition. Using finite element simulations, we showed that the total dose delivered, calculated using area under the curve (AUC) was similar between platforms, however normalized viability was lower for the organoid in media condition than in the static gel and bioreactor. Our results highlight the utility of our multi-well bioreactor for studying organoid response to chemical gradients and demonstrate that comparing drug response across these different platforms is nontrivial.
Collapse
Affiliation(s)
- Elisa Marie Wasson
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Wei He
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Jesse Ahlquist
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - William Fredrick Hynes
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Michael Gregory Triplett
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Aubree Hinckley
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Eveliina Karelehto
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
- Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, United States
| | | | - Caleb Fisher Friedman
- Department of Computational Media, University of California Santa Cruz, Santa Cruz, CA, United States
| | - Claire Robertson
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
- UC Davis Comprehensive Cancer Center, Davis, CA, United States
| | - Maxim Shusteff
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Robert Warren
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
- Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, United States
| | - Matthew A. Coleman
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Monica Lizet Moya
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Elizabeth K. Wheeler
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| |
Collapse
|
19
|
Wang L, Chen Z, Xu Z, Yang Y, Wang Y, Zhu J, Guo X, Tang D, Gu Z. A new approach of using organ-on-a-chip and fluid-structure interaction modeling to investigate biomechanical characteristics in tissue-engineered blood vessels. Front Physiol 2023; 14:1210826. [PMID: 37275235 PMCID: PMC10237315 DOI: 10.3389/fphys.2023.1210826] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 05/03/2023] [Indexed: 06/07/2023] Open
Abstract
The tissue-engineered blood vessel (TEBV) has been developed and used in cardiovascular disease modeling, preclinical drug screening, and for replacement of native diseased arteries. Increasing attention has been paid to biomechanical cues in TEBV and other tissue-engineered organs to better recapitulate the functional properties of the native organs. Currently, computational fluid dynamics models were employed to reveal the hydrodynamics in TEBV-on-a-chip. However, the biomechanical wall stress/strain conditions in the TEBV wall have never been investigated. In this paper, a straight cylindrical TEBV was placed into a polydimethylsiloxane-made microfluidic device to construct the TEBV-on-a-chip. The chip was then perfused with cell culture media flow driven by a peristaltic pump. A three-dimensional fluid-structure interaction (FSI) model was generated to simulate the biomechanical conditions in TEBV and mimic both the dynamic TEBV movement and pulsatile fluid flow. The material stiffness of the TEBV wall was determined by uniaxial tensile testing, while the viscosity of cell culture media was measured using a rheometer. Comparison analysis between the perfusion experiment and FSI model results showed that the average relative error in diameter expansion of TEBV from both approaches was 10.0% in one period. For fluid flow, the average flow velocity over a period was 2.52 cm/s from the FSI model, 10.5% higher than the average velocity of the observed cell clusters (2.28 mm/s) in the experiment. These results demonstrated the facility to apply the FSI modeling approach in TEBV to obtain more comprehensive biomechanical results for investigating mechanical mechanisms of cardiovascular disease development.
Collapse
Affiliation(s)
- Liang Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, China
| | - Zhuoyue Xu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, China
| | - Yi Yang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, China
| | - Yan Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, China
| | - Jianfeng Zhu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, China
| | - Xiaoya Guo
- School of Science, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Dalin Tang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
- Mathematical Sciences Department, Worcester Polytechnic Institute, Worcester, MA, United States
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, China
| |
Collapse
|
20
|
Kang JH, Jang M, Seo SJ, Choi A, Shin D, Seo S, Lee SH, Kim HN. Mechanobiological Adaptation to Hyperosmolarity Enhances Barrier Function in Human Vascular Microphysiological System. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206384. [PMID: 36808839 PMCID: PMC10161024 DOI: 10.1002/advs.202206384] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/27/2023] [Indexed: 05/06/2023]
Abstract
In infectious disease such as sepsis and COVID-19, blood vessel leakage treatment is critical to prevent fatal progression into multi-organ failure and ultimately death, but the existing effective therapeutic modalities that improve vascular barrier function are limited. Here, this study reports that osmolarity modulation can significantly improve vascular barrier function, even in an inflammatory condition. 3D human vascular microphysiological systems and automated permeability quantification processes for high-throughput analysis of vascular barrier function are utilized. Vascular barrier function is enhanced by >7-folds with 24-48 h hyperosmotic exposure (time window of emergency care; >500 mOsm L-1 ) but is disrupted after hypo-osmotic exposure (<200 mOsm L-1 ). By integrating genetic and protein level analysis, it is shown that hyperosmolarity upregulates vascular endothelial-cadherin, cortical F-actin, and cell-cell junction tension, indicating that hyperosmotic adaptation mechanically stabilizes the vascular barrier. Importantly, improved vascular barrier function following hyperosmotic exposure is maintained even after chronic exposure to proinflammatory cytokines and iso-osmotic recovery via Yes-associated protein signaling pathways. This study suggests that osmolarity modulation may be a unique therapeutic strategy to proactively prevent infectious disease progression into severe stages via vascular barrier function protection.
Collapse
Affiliation(s)
- Joon Ho Kang
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
| | - Minjeong Jang
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
| | - Su Jin Seo
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Department of Chemical EngineeringKwangwoon UniversitySeoul01897Republic of Korea
| | - Andrew Choi
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
| | - Daeeun Shin
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- School of Mechanical EngineeringSungkyunkwan UniversitySuwon16419Republic of Korea
| | - Suyoung Seo
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Program in Nano Science and TechnologyGraduate School of Convergence Science and TechnologySeoul National UniversitySeoul08826Republic of Korea
| | - Soo Hyun Lee
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Division of Bio‐Medical Science & TechnologyKIST SchoolUniversity of Science and Technology (UST)Seoul02792Republic of Korea
| | - Hong Nam Kim
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Division of Bio‐Medical Science & TechnologyKIST SchoolUniversity of Science and Technology (UST)Seoul02792Republic of Korea
- School of Mechanical EngineeringYonsei UniversitySeoul03722Republic of Korea
- Yonsei‐KIST Convergence Research InstituteYonsei UniversitySeoul03722Republic of Korea
| |
Collapse
|
21
|
Mi X, Su Z, Yue X, Ren Y, Yang X, Qiang L, Kong W, Ma Z, Zhang C, Wang J. 3D bioprinting tumor models mimic the tumor microenvironment for drug screening. Biomater Sci 2023; 11:3813-3827. [PMID: 37052182 DOI: 10.1039/d3bm00159h] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Cancer is a severe threat to human life and health and represents the main cause of death globally. Drug therapy is one of the primary means of treating cancer; however, most anticancer medications do not proceed beyond preclinical testing because the conditions of actual human tumors are not effectively mimicked by traditional tumor models. Hence, bionic in vitro tumor models must be developed to screen for anticancer drugs. Three-dimensional (3D) bioprinting technology can produce structures with built-in spatial and chemical complexity and models with accurately controlled structures, a homogeneous size and morphology, less variation across batches, and a more realistic tumor microenvironment (TME). This technology can also rapidly produce such models for high-throughput anticancer medication testing. This review describes 3D bioprinting methods, the use of bioinks in tumor models, and in vitro tumor model design strategies for building complex tumor microenvironment features using biological 3D printing technology. Moreover, the application of 3D bioprinting in vitro tumor models in drug screening is also discussed.
Collapse
Affiliation(s)
- Xuelian Mi
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Zhi Su
- School of Kinesiology, Shanghai University of Sport, 399 Chang Hai Road, Shanghai, 200438, China
| | - Xiaokun Yue
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Ya Ren
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
| | - Xue Yang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
| | - Lei Qiang
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 611756, China
| | - Weiqing Kong
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, Shandong Province, 266000, China
| | - Zhenjiang Ma
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Changru Zhang
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Jinwu Wang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
22
|
de Graaf MNS, Vivas A, Kasi DG, van den Hil FE, van den Berg A, van der Meer AD, Mummery CL, Orlova VV. Multiplexed fluidic circuit board for controlled perfusion of 3D blood vessels-on-a-chip. LAB ON A CHIP 2022; 23:168-181. [PMID: 36484766 PMCID: PMC9764810 DOI: 10.1039/d2lc00686c] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/21/2022] [Indexed: 06/11/2023]
Abstract
Three-dimensional (3D) blood vessels-on-a-chip (VoC) models integrate the biological complexity of vessel walls with dynamic microenvironmental cues, such as wall shear stress (WSS) and circumferential strain (CS). However, these parameters are difficult to control and are often poorly reproducible due to the high intrinsic diameter variation of individual 3D-VoCs. As a result, the throughput of current 3D systems is one-channel-at-a-time. Here, we developed a fluidic circuit board (FCB) for simultaneous perfusion of up to twelve 3D-VoCs using a single set of control parameters. By designing the internal hydraulic resistances in the FCB appropriately, it was possible to provide a pre-set WSS to all connected 3D-VoCs, despite significant variation in lumen diameters. Using this FCB, we found that variation of CS or WSS induce morphological changes to human induced pluripotent stem cell (hiPSC)-derived endothelial cells (ECs) and conclude that control of these parameters using a FCB is necessary to study 3D-VOCs.
Collapse
Affiliation(s)
- Mees N S de Graaf
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Aisen Vivas
- Applied Stem Cell Technologies, University of Twente, 7500AE Enschede, The Netherlands
- BIOS Lab on a Chip Group, MESA+ Institute for Nanotechnology, Technical Medical Centre, Max Planck Institute for Complex Fluid Dynamics, University of Twente, 7500AE Enschede, The Netherlands
| | - Dhanesh G Kasi
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Neurology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Francijna E van den Hil
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Albert van den Berg
- BIOS Lab on a Chip Group, MESA+ Institute for Nanotechnology, Technical Medical Centre, Max Planck Institute for Complex Fluid Dynamics, University of Twente, 7500AE Enschede, The Netherlands
| | | | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
- Applied Stem Cell Technologies, University of Twente, 7500AE Enschede, The Netherlands
| | - Valeria V Orlova
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| |
Collapse
|
23
|
Neufeld L, Yeini E, Pozzi S, Satchi-Fainaro R. 3D bioprinted cancer models: from basic biology to drug development. Nat Rev Cancer 2022; 22:679-692. [PMID: 36280768 DOI: 10.1038/s41568-022-00514-w] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/12/2022] [Indexed: 12/29/2022]
Abstract
Effort invested in the development of new drugs often fails to be translated into meaningful clinical benefits for patients with cancer. The development of more effective anticancer therapeutics and accurate prediction of their clinical merit remain urgent unmet medical needs. As solid cancers have complex and heterogeneous structures composed of different cell types and extracellular matrices, three-dimensional (3D) cancer models hold great potential for advancing our understanding of cancer biology, which has been historically investigated in tumour cell cultures on rigid plastic plates. Advanced 3D bioprinted cancer models have the potential to revolutionize the way we discover therapeutic targets, develop new drugs and personalize anticancer therapies in an accurate, reproducible, clinically translatable and robust manner. These ex vivo cancer models are already replacing existing in vitro systems and could, in the future, diminish or even replace the use of animal models. Therefore, profound understanding of the differences in tumorigenesis between 2D, 3D and animal models of cancer is essential. This Review presents the state of the art of 3D bioprinted cancer modelling, focusing on the biological processes that underlie the molecular mechanisms involved in cancer progression and treatment response as well as on proteomic and genomic signatures.
Collapse
Affiliation(s)
- Lena Neufeld
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eilam Yeini
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
- Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
24
|
Puleri DF, Roychowdhury S, Balogh P, Gounley J, Draeger EW, Ames J, Adebiyi A, Chidyagwai S, Hernández B, Lee S, Moore SV, Vetter JS, Randles A. High Performance Adaptive Physics Refinement to Enable Large-Scale Tracking of Cancer Cell Trajectory. PROCEEDINGS. IEEE INTERNATIONAL CONFERENCE ON CLUSTER COMPUTING 2022; 2022:230-242. [PMID: 38125675 PMCID: PMC10731912 DOI: 10.1109/cluster51413.2022.00036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
The ability to track simulated cancer cells through the circulatory system, important for developing a mechanistic understanding of metastatic spread, pushes the limits of today's supercomputers by requiring the simulation of large fluid volumes at cellular-scale resolution. To overcome this challenge, we introduce a new adaptive physics refinement (APR) method that captures cellular-scale interaction across large domains and leverages a hybrid CPU-GPU approach to maximize performance. Through algorithmic advances that integrate multi-physics and multi-resolution models, we establish a finely resolved window with explicitly modeled cells coupled to a coarsely resolved bulk fluid domain. In this work we present multiple validations of the APR framework by comparing against fully resolved fluid-structure interaction methods and employ techniques, such as latency hiding and maximizing memory bandwidth, to effectively utilize heterogeneous node architectures. Collectively, these computational developments and performance optimizations provide a robust and scalable framework to enable system-level simulations of cancer cell transport.
Collapse
Affiliation(s)
- Daniel F Puleri
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Peter Balogh
- Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - John Gounley
- {Computational Sciences and Engineering, National Center for Computational Sciences, Computer Science and Mathematics}, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Erik W Draeger
- Scientific Computing Group, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Jeff Ames
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Adebayo Adebiyi
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Benjamín Hernández
- {Computational Sciences and Engineering, National Center for Computational Sciences, Computer Science and Mathematics}, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Seyong Lee
- {Computational Sciences and Engineering, National Center for Computational Sciences, Computer Science and Mathematics}, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Shirley V Moore
- Department of Computer Science, University of Texas at El Paso, El Paso, TX, USA
| | - Jeffrey S Vetter
- {Computational Sciences and Engineering, National Center for Computational Sciences, Computer Science and Mathematics}, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Amanda Randles
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| |
Collapse
|
25
|
Bashor CJ, Hilton IB, Bandukwala H, Smith DM, Veiseh O. Engineering the next generation of cell-based therapeutics. Nat Rev Drug Discov 2022; 21:655-675. [PMID: 35637318 PMCID: PMC9149674 DOI: 10.1038/s41573-022-00476-6] [Citation(s) in RCA: 189] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2022] [Indexed: 12/19/2022]
Abstract
Cell-based therapeutics are an emerging modality with the potential to treat many currently intractable diseases through uniquely powerful modes of action. Despite notable recent clinical and commercial successes, cell-based therapies continue to face numerous challenges that limit their widespread translation and commercialization, including identification of the appropriate cell source, generation of a sufficiently viable, potent and safe product that meets patient- and disease-specific needs, and the development of scalable manufacturing processes. These hurdles are being addressed through the use of cutting-edge basic research driven by next-generation engineering approaches, including genome and epigenome editing, synthetic biology and the use of biomaterials.
Collapse
Affiliation(s)
- Caleb J Bashor
- Department of Bioengineering, Rice University, Houston, TX, USA.
- Department of Biosciences, Rice University, Houston, TX, USA.
| | - Isaac B Hilton
- Department of Bioengineering, Rice University, Houston, TX, USA.
- Department of Biosciences, Rice University, Houston, TX, USA.
| | - Hozefa Bandukwala
- Sigilon Therapeutics, Cambridge, MA, USA
- Flagship Pioneering, Cambridge, MA, USA
| | - Devyn M Smith
- Sigilon Therapeutics, Cambridge, MA, USA
- Arbor Biotechnologies, Cambridge, MA, USA
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, USA.
| |
Collapse
|
26
|
Wu Y, Zhou Y, Paul R, Qin X, Islam K, Liu Y. Adaptable Microfluidic Vessel-on-a-Chip Platform for Investigating Tumor Metastatic Transport in Bloodstream. Anal Chem 2022; 94:12159-12166. [PMID: 35998619 DOI: 10.1021/acs.analchem.2c02556] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cancer metastasis counts for 90% of cancer fatalities, and its development process is still a mystery. The dynamic process of tumor metastatic transport in the blood vessel is not well understood, in which some biomechanical factors, such as shear stress and various flow patterns, may have significant impacts. Here, we report a microfluidic vessel-on-a-chip platform for recapitulating several key metastatic steps of tumor cells in blood vessels on the same chip, including intravasation, circulating tumor cell (CTC) vascular adhesion, and extravasation. Due to its excellent adaptability, our system can reproduce various microenvironments to investigate the specific interactions between CTCs and blood vessels. On the basis of this platform, effects of important biomechanical factors on CTC adhesion such as vascular surface properties and vessel geometry-dependent hemodynamics were specifically inspected. We demonstrated that CTC adhesion is more likely to occur under certain mechano-physiological situations, such as vessels with vascular glycocalyx (VGCX) shedding and hemodynamic disturbances. Finally, computational models of both the fluidic dynamics in vessels and CTC adhesion were established based on the confocal scanned 3D images. The modeling results are believed to provide insights into exploring tumor metastasis progression and inspire new ideas for anticancer therapy development.
Collapse
Affiliation(s)
- Yue Wu
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Yuyuan Zhou
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Ratul Paul
- Department of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Xiaochen Qin
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Khayrul Islam
- Department of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Yaling Liu
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States.,Department of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| |
Collapse
|
27
|
Organ-on-a-Chip: Design and Simulation of Various Microfluidic Channel Geometries for the Influence of Fluid Dynamic Parameters. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12083829] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Shear stress, pressure, and flow rate are fluid dynamic parameters that can lead to changes in the morphology, proliferation, function, and survival of many cell types and have a determinant impact on tissue function and viability. Microfluidic devices are promising tools to investigate these parameters and fluid behaviour within different microchannel geometries. This study discusses and analyses different designed microfluidic channel geometries regarding the influence of fluid dynamic parameters on their microenvironment at specified fluidic parameters. The results demonstrate that in the circular microchamber, the velocity and shear stress profiles assume a parabolic shape with a maximum velocity occurring in the centre of the chamber and a minimum velocity at the walls. The longitudinal microchannel shows a uniform velocity and shear stress profile throughout the microchannel. Simulation studies for the two geometries with three parallel microchannels showed that in proximity to the micropillars, the velocity and shear stress profiles decreased. Moreover, the pressure is inversely proportional to the width and directly proportional to the flow rate within the microfluidic channels. The simulations showed that the velocity and wall shear stress indicated different values at different flow rates. It was also found that the width and height of the microfluidic channels could affect both velocity and shear stress profiles, contributing to the control of shear stress. The study has demonstrated strategies to predict and control the effects of these forces and the potential as an alternative to conventional cell culture as well as to recapitulate the cell- and organ-specific microenvironment.
Collapse
|
28
|
Mao M, Chen P, He J, Zhu G, Li X, Li D. Deciphering Fluid Transport Within Leaf-Inspired Capillary Networks Based on a 3D Computational Model. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2108102. [PMID: 35253997 DOI: 10.1002/smll.202108102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/03/2022] [Indexed: 06/14/2023]
Abstract
Leaf venation provides a promising template for engineering capillary-like vasculature in vitro due to its highly efficient fluid transport capability and remarkable similarities to native capillary networks. A key challenge in exploring the potential biological applications of leaf-inspired capillary networks (LICNs) is to accurately and quantitively understand its internal fluid transport characteristics. Here, a centerline-induced partition-assembly modeling strategy is proposed to establish a 3D computational model, which can accurately simulate the flow conditions in LICNs. Based on the 3D flow simulation, the authors demonstrate the excellent defect-resistant fluid transport capability of LICNs. Interestingly, structural defects in the primary channel can effectively accelerate the overall perfusion efficiency. Flow patterns in LICNs with multiple defects can be estimated by simple superposition of the simulation results derived from the corresponding single-defect models. The 3D computational model is further used to determine the optimal perfusion parameter for the in-vitro formation of endothelialized capillary networks by mimicking native microvascular flow conditions. The endothelialized networks can recapitulate the vascular colonization process and reveal a strong correlation between cancer cell adhesion and flow-induced shear stress. This study offers a quantitative tool to scrutinize the fluid and biological transport mechanisms within LICNs for various biomedical applications.
Collapse
Affiliation(s)
- Mao Mao
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Pengyu Chen
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Jiankang He
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Guangyu Zhu
- School of Energy and Power Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Xiao Li
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Dichen Li
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| |
Collapse
|
29
|
Programmed Catalytic Therapy-Mediated ROS Generation and T-Cell Infiltration in Lung Metastasis by a Dual Metal-Organic Framework (MOF) Nanoagent. Pharmaceutics 2022; 14:pharmaceutics14030527. [PMID: 35335903 PMCID: PMC8955711 DOI: 10.3390/pharmaceutics14030527] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/21/2022] [Accepted: 02/25/2022] [Indexed: 12/10/2022] Open
Abstract
Nano-catalytic agents actuating Fenton-like reaction in cancer cells cause intratumoral generation of reactive oxygen species (ROS), allowing the potential for immune therapy of tumor metastasis via the recognition of tumor-associated antigens. However, the self-defense mechanism of cancer cells, known as autophagy, and unsustained ROS generation often restricts efficiency, lowering the immune attack, especially in invading metastatic clusters. Here, a functional core-shell metal-organic framework nanocube (dual MOF) doubling as a catalytic agent and T cell infiltration inducer that programs ROS and inhibits autophagy is reported. The dual MOF integrated a Prussian blue (PB)-coated iron (Fe2+)-containing metal-organic framework (MOF, MIL88) as a programmed peroxide mimic in the cancer cells, facilitating the sustained ROS generation. With the assistance of Chloroquine (CQ), the inhibition of autophagy through lysosomal deacidification breaks off the self-defense mechanism and further improves the cytotoxicity. The purpose of this material design was to inhibit autophagy and ROS efficacy of the tumor, and eventually improve T cell recruitment for immune therapy of lung metastasis. The margination and internalization-mediated cancer cell uptake improve the accumulation of dual MOF of metastatic tumors in vivo. The effective catalytic dual MOF integrated dysfunctional autophagy at the metastasis elicits the ~3-fold recruitment of T lymphocytes. Such synergy of T cell recruitment and ROS generation transported by dual MOF during the metastases successfully suppresses more than 90% of tumor foci in the lung.
Collapse
|
30
|
Live Cell Imaging and Analysis of Cancer-Cell Transmigration Through Endothelial Monolayers. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2441:329-338. [PMID: 35099749 DOI: 10.1007/978-1-0716-2059-5_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
During metastasis, a subset of cancer cells will break away from the primary tumor and invade into the surrounding tissue. Cancer cells which are able to breach the endothelium and enter the blood stream are then transported in the circulation to new target organs where they may seed as a distant metastasis. In order to invade this new organ, the cancer cells must bind to and traverse the vascular wall, a process known as transendothelial migration (TEM) or extravasation. This chapter describes an in vitro approach to automated live cell imaging and analysis of TEM in order to accurately quantify these kinetics and aid the researcher in dissecting the mechanisms of tumor-endothelial interactions during this phase of metastasis.
Collapse
|
31
|
Sunderland K, Jiang J, Zhao F. Disturbed flow's impact on cellular changes indicative of vascular aneurysm initiation, expansion, and rupture: A pathological and methodological review. J Cell Physiol 2022; 237:278-300. [PMID: 34486114 PMCID: PMC8810685 DOI: 10.1002/jcp.30569] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/06/2021] [Accepted: 08/16/2021] [Indexed: 01/03/2023]
Abstract
Aneurysms are malformations within the arterial vasculature brought on by the structural breakdown of the microarchitecture of the vessel wall, with aneurysms posing serious health risks in the event of their rupture. Blood flow within vessels is generally laminar with high, unidirectional wall shear stressors that modulate vascular endothelial cell functionality and regulate vascular smooth muscle cells. However, altered vascular geometry induced by bifurcations, significant curvature, stenosis, or clinical interventions can alter the flow, generating low stressor disturbed flow patterns. Disturbed flow is associated with altered cellular morphology, upregulated expression of proteins modulating inflammation, decreased regulation of vascular permeability, degraded extracellular matrix, and heightened cellular apoptosis. The understanding of the effects disturbed flow has on the cellular cascades which initiate aneurysms and promote their subsequent growth can further elucidate the nature of this complex pathology. This review summarizes the current knowledge about the disturbed flow and its relation to aneurysm pathology, the methods used to investigate these relations, as well as how such knowledge has impacted clinical treatment methodologies. This information can contribute to the understanding of the development, growth, and rupture of aneurysms and help develop novel research and aneurysmal treatment techniques.
Collapse
Affiliation(s)
- Kevin Sunderland
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
| | - Jingfeng Jiang
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931,Corresponding Authors: Feng Zhao, 101 Bizzell Street, College Station, TX 77843-312, Tel : 979-458-1239, , Jingfeng Jiang, 1400 Townsend Dr., Houghton, MI 49931, Tel: 906-487-1943
| | - Feng Zhao
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843,Corresponding Authors: Feng Zhao, 101 Bizzell Street, College Station, TX 77843-312, Tel : 979-458-1239, , Jingfeng Jiang, 1400 Townsend Dr., Houghton, MI 49931, Tel: 906-487-1943
| |
Collapse
|
32
|
Yong KW, Janmaleki M, Pachenari M, Mitha AP, Sanati-Nezhad A, Sen A. Engineering a 3D human intracranial aneurysm model using liquid-assisted injection molding and tuned hydrogels. Acta Biomater 2021; 136:266-278. [PMID: 34547516 DOI: 10.1016/j.actbio.2021.09.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 12/30/2022]
Abstract
Physiologically relevant intracranial aneurysm (IA) models are crucially required to facilitate testing treatment options for IA. Herein, we report the development of a new in vitro tissue-engineered platform, which recapitulates the microenvironment, structure, and cellular complexity of native human IA. A new modified liquid-assisted injection molding technique was developed to fabricate a three-dimensional hollow IA model with clinically relevant IA dimensions within a mechanically tuned Gelatin Methacryloyl (GelMA) hydrogel. An endothelium lining was created inside the IA model by culturing human umbilical vein endothelial cells over pre-cultured human brain vascular smooth muscle cells. These cellularized IA models were subjected to medium perfusion at flow rates between 6.3 and 15.75 mL/min for inducing biomimetic vessel wall shear stress (10-25 dyn/cm2) to the cells for ten days. Both cell types maintained their secretome profiles and showed more than 96% viability, demonstrating the biocompatibility of the hydrogel during perfusion cell culture at such flow rates. Based on the characterized viscoelastic properties of the GelMA hydrogel and with the aid of a fluid-structure interaction model, the capability of the IA model in predicting the response of the IA to different fluid flow profiles was mathematically shown. With physiologically relevant behavior, our developed in vitro human IA model could allow researchers to better understand the pathophysiology and treatment of IA. STATEMENT OF SIGNIFICANCE: A three-dimensional intracranial aneurysm (IA) tissue model recapitulating the microenvironment, structure, and cellular complexity of native human IA was developed. • An endothelium lining was created inside the IA model over pre-cultured human brain vascular smooth muscle cells over at least 10-day successful culture. • The cells maintained their secretome profiles, demonstrating the biocompatibility of hydrogel during a long-term perfusion cell culture. • The IA model showed its capability in predicting the response of IA to different fluid flow profiles. • The cells in the vessel region behaved differently from cells in the aneurysm region due to alteration in hemodynamic shear stress. • The IA model could allow researchers to better understand the pathophysiology and treatment options of IA.
Collapse
|
33
|
Jamee R, Araf Y, Naser IB, Promon SK. The promising rise of bioprinting in revolutionalizing medical science: Advances and possibilities. Regen Ther 2021; 18:133-145. [PMID: 34189195 PMCID: PMC8213915 DOI: 10.1016/j.reth.2021.05.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/19/2021] [Accepted: 05/26/2021] [Indexed: 12/24/2022] Open
Abstract
Bioprinting is a relatively new yet evolving technique predominantly used in regenerative medicine and tissue engineering. 3D bioprinting techniques combine the advantages of creating Extracellular Matrix (ECM)like environments for cells and computer-aided tailoring of predetermined tissue shapes and structures. The essential application of bioprinting is for the regeneration or restoration of damaged and injured tissues by producing implantable tissues and organs. The capability of bioprinting is yet to be fully scrutinized in sectors like the patient-specific spatial distribution of cells, bio-robotics, etc. In this review, currently developed experimental systems and strategies for the bioprinting of different types of tissues as well as for drug delivery and cancer research are explored for potential applications. This review also digs into the most recent opportunities and future possibilities for the efficient implementation of bioprinting to restructure medical and technological practices.
Collapse
Affiliation(s)
- Radia Jamee
- Department of Mathematics and Natural Sciences, School of Data and Sciences, Brac University, Dhaka, Bangladesh
- Mechamind, Dhaka, Bangladesh
| | - Yusha Araf
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Iftekhar Bin Naser
- Department of Mathematics and Natural Sciences, School of Data and Sciences, Brac University, Dhaka, Bangladesh
| | - Salman Khan Promon
- Department of Mathematics and Natural Sciences, School of Data and Sciences, Brac University, Dhaka, Bangladesh
- Mechamind, Dhaka, Bangladesh
| |
Collapse
|
34
|
Haase K, Piatti F, Marcano M, Shin Y, Visone R, Redaelli A, Rasponi M, Kamm RD. Physiologic flow-conditioning limits vascular dysfunction in engineered human capillaries. Biomaterials 2021; 280:121248. [PMID: 34794827 DOI: 10.1016/j.biomaterials.2021.121248] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 02/02/2023]
Abstract
Hemodynamics play a central role in the health and disease of the coronary and peripheral vascular systems. Vessel-lining endothelial cells are known mechanosensors, responding to disturbances in flow - with mechanosensitivity hypothesized to change in response to metabolic demands. The health of our smallest microvessels have been lauded as a prognostic marker for cardiovascular health. Yet, despite numerous animal models, studying these small vessels has proved difficult. Microfluidic technologies have allowed a number of 3D vascular models to be developed and used to investigate human vessels. Here, two such systems are employed for examining 1) interstitial flow effects on neo-vessel formation, and 2) the effects of flow-conditioning on vascular remodeling following sustained static culture. Interstitial flow is shown to enhance early vessel formation via significant remodeling of vessels and interconnected tight junctions of the endothelium. In formed vessels, continuous flow maintains a stable vascular diameter and causes significant remodeling, contrasting the continued anti-angiogenic decline of statically cultured vessels. This study is the first to couple complex 3D computational flow distributions and microvessel remodeling from microvessels grown on-chip (exposed to flow or no-flow conditions). Flow-conditioned vessels (WSS < 1Pa for 30 μm vessels) increase endothelial barrier function, result in significant changes in gene expression and reduce reactive oxygen species and anti-angiogenic cytokines. Taken together, these results demonstrate microvessel mechanosensitivity to flow-conditioning, which limits deleterious vessel regression in vitro, and could have implications for future modeling of reperfusion/no-flow conditions.
Collapse
Affiliation(s)
- Kristina Haase
- Dept. of Mechanical Engineering, MIT, Cambridge, MA, USA
| | - Filippo Piatti
- Dept. of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | | | - Yoojin Shin
- Dept. of Mechanical Engineering, MIT, Cambridge, MA, USA
| | - Roberta Visone
- Dept. of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Alberto Redaelli
- Dept. of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Marco Rasponi
- Dept. of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Roger D Kamm
- Dept. of Mechanical Engineering, MIT, Cambridge, MA, USA; Dept. of Biological Engineering, MIT, Cambridge, MA, USA.
| |
Collapse
|
35
|
Carvalho V, Rodrigues RO, Lima RA, Teixeira S. Computational Simulations in Advanced Microfluidic Devices: A Review. MICROMACHINES 2021; 12:mi12101149. [PMID: 34683199 PMCID: PMC8539624 DOI: 10.3390/mi12101149] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/14/2021] [Accepted: 09/21/2021] [Indexed: 12/11/2022]
Abstract
Numerical simulations have revolutionized research in several engineering areas by contributing to the understanding and improvement of several processes, being biomedical engineering one of them. Due to their potential, computational tools have gained visibility and have been increasingly used by several research groups as a supporting tool for the development of preclinical platforms as they allow studying, in a more detailed and faster way, phenomena that are difficult to study experimentally due to the complexity of biological processes present in these models—namely, heat transfer, shear stresses, diffusion processes, velocity fields, etc. There are several contributions already in the literature, and significant advances have been made in this field of research. This review provides the most recent progress in numerical studies on advanced microfluidic devices, such as organ-on-a-chip (OoC) devices, and how these studies can be helpful in enhancing our insight into the physical processes involved and in developing more effective OoC platforms. In general, it has been noticed that in some cases, the numerical studies performed have limitations that need to be improved, and in the majority of the studies, it is extremely difficult to replicate the data due to the lack of detail around the simulations carried out.
Collapse
Affiliation(s)
- Violeta Carvalho
- MEtRICs, Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal;
- ALGORITMI, Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal;
- Correspondence:
| | - Raquel O. Rodrigues
- Center for MicroElectromechanical Systems (CMEMS-UMinho), Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal;
| | - Rui A. Lima
- MEtRICs, Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal;
- CEFT, R. Dr. Roberto Frias, Faculty of Engineering of the University of Porto (FEUP), 4200-465 Porto, Portugal
| | - Senhorinha Teixeira
- ALGORITMI, Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal;
| |
Collapse
|
36
|
Kim J, Jang J, Cho DW. Recapitulating the Cancer Microenvironment Using Bioprinting Technology for Precision Medicine. MICROMACHINES 2021; 12:1122. [PMID: 34577765 PMCID: PMC8472267 DOI: 10.3390/mi12091122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/09/2021] [Accepted: 09/15/2021] [Indexed: 12/20/2022]
Abstract
The complex and heterogenous nature of cancer contributes to the development of cancer cell drug resistance. The construction of the cancer microenvironment, including the cell-cell interactions and extracellular matrix (ECM), plays a significant role in the development of drug resistance. Traditional animal models used in drug discovery studies have been associated with feasibility issues that limit the recapitulation of human functions; thus, in vitro models have been developed to reconstruct the human cancer system. However, conventional two-dimensional and three-dimensional (3D) in vitro cancer models are limited in their ability to emulate complex cancer microenvironments. Advances in technologies, including bioprinting and cancer microenvironment reconstruction, have demonstrated the potential to overcome some of the limitations of conventional models. This study reviews some representative bioprinted in vitro models used in cancer research, particularly fabrication strategies for modeling and consideration of essential factors needed for the reconstruction of the cancer microenvironment. In addition, we highlight recent studies that applied such models, including application in precision medicine using advanced bioprinting technologies to fabricate biomimetic cancer models. Furthermore, we discuss current challenges in 3D bioprinting and suggest possible strategies to construct in vitro models that better mimic the pathophysiology of the cancer microenvironment for application in clinical settings.
Collapse
Affiliation(s)
- Jisoo Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Jinah Jang
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- Department of Creative IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03722, Korea
| | - Dong-Woo Cho
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
37
|
Wasson EM, Dubbin K, Moya ML. Go with the flow: modeling unique biological flows in engineered in vitro platforms. LAB ON A CHIP 2021; 21:2095-2120. [PMID: 34008661 DOI: 10.1039/d1lc00014d] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Interest in recapitulating in vivo phenomena in vitro using organ-on-a-chip technology has grown rapidly and with it, attention to the types of fluid flow experienced in the body has followed suit. These platforms offer distinct advantages over in vivo models with regards to human relevance, cost, and control of inputs (e.g., controlled manipulation of biomechanical cues from fluid perfusion). Given the critical role biophysical forces play in several tissues and organs, it is therefore imperative that engineered in vitro platforms capture the complex, unique flow profiles experienced in the body that are intimately tied with organ function. In this review, we outline the complex and unique flow regimes experienced by three different organ systems: blood vasculature, lymphatic vasculature, and the intestinal system. We highlight current state-of-the-art platforms that strive to replicate physiological flows within engineered tissues while introducing potential limitations in current approaches.
Collapse
Affiliation(s)
- Elisa M Wasson
- Material Engineering Division, Lawrence Livermore National Laboratory, 7000 East Ave L-222, Livermore, CA 94551, USA.
| | - Karen Dubbin
- Material Engineering Division, Lawrence Livermore National Laboratory, 7000 East Ave L-222, Livermore, CA 94551, USA.
| | - Monica L Moya
- Material Engineering Division, Lawrence Livermore National Laboratory, 7000 East Ave L-222, Livermore, CA 94551, USA.
| |
Collapse
|
38
|
Introduction to bioprinting of in vitro cancer models. Essays Biochem 2021; 65:603-610. [PMID: 34028520 DOI: 10.1042/ebc20200104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/30/2021] [Accepted: 05/06/2021] [Indexed: 12/27/2022]
Abstract
Cancer models are essential in cancer research and for new drug development pipelines. However, conventional cancer tissue models have failed to capture the human cancer physiology, thus hindering drug discovery. The major challenge is the establishment of physiologically relevant cancer models that reflect the complexity of the tumor microenvironment (TME). The TME is a highly complex milieu composed of diverse factors that are associated with cancer progression and metastasis, as well as with the development of cancer resistance to therapeutics. To emulate the TME, 3D bioprinting has emerged as a way to create engineered cancer tissue models. Bioprinted cancer tissue models have the potential to recapitulate cancer pathology and increased drug resistance in an organ-mimicking 3D environment. This review overviews the bioprinting technologies used for the engineering of cancer tissue models and provides a future perspective on bioprinting to further advance cancer research.
Collapse
|
39
|
Priyadarshani J, Roy T, Das S, Chakraborty S. Frugal Approach toward Developing a Biomimetic, Microfluidic Network-on-a-Chip for In Vitro Analysis of Microvascular Physiology. ACS Biomater Sci Eng 2021; 7:1263-1277. [PMID: 33555875 DOI: 10.1021/acsbiomaterials.1c00070] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Several disease conditions, such as cancer metastasis and atherosclerosis, are deeply connected with the complex biophysical phenomena taking place in the complicated architecture of the tiny blood vessels in human circulatory systems. Traditionally, these diseases have been probed by devising various animal models, which are otherwise constrained by ethical considerations as well as limited predictive capabilities. Development of an engineered network-on-a-chip, which replicates not only the functional aspects of the blood-carrying microvessels of human bodies, but also its geometrical complexity and hierarchical microstructure, is therefore central to the evaluation of organ-assist devices and disease models for therapeutic assessment. Overcoming the constraints of reported resource-intensive fabrication techniques, here, we report a facile, simple yet niche combination of surface engineering and microfabrication strategy to devise a highly ordered hierarchical microtubular network embedded within a polydimethylsiloxane (PDMS) slab for dynamic cell culture on a chip, with a vision of addressing the exclusive aspects of the vascular transport processes under medically relevant paradigms. The design consists of hierarchical complexity ranging from capillaries (∼80 μm) to large arteries (∼390 μm) and a simultaneous tuning of the interfacial material chemistry. The fluid flow behavior is characterized numerically within the hierarchical network, and a confluent endothelial layer is realized on the inner wall of microfluidic device. We further explore the efficacy of the device as a vascular deposition assay of circulatory tumor cells (MG-63 osteosarcoma cells) present in whole blood. The proposed paradigm of mimicking an in vitro vascular network in a low-cost paradigm holds further potential for probing cellular dynamics as well as offering critical insights into various vascular transport processes.
Collapse
Affiliation(s)
- Jyotsana Priyadarshani
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Trina Roy
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Soumen Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Suman Chakraborty
- Department of Mechanical Engineering, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| |
Collapse
|
40
|
Wang S, Ye T, Li G, Zhang X, Shi H. Margination and adhesion dynamics of tumor cells in a real microvascular network. PLoS Comput Biol 2021; 17:e1008746. [PMID: 33606686 PMCID: PMC7928530 DOI: 10.1371/journal.pcbi.1008746] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 03/03/2021] [Accepted: 01/27/2021] [Indexed: 01/09/2023] Open
Abstract
In tumor metastasis, the margination and adhesion of tumor cells are two critical and closely related steps, which may determine the destination where the tumor cells extravasate to. We performed a direct three-dimensional simulation on the behaviors of the tumor cells in a real microvascular network, by a hybrid method of the smoothed dissipative particle dynamics and immersed boundary method (SDPD-IBM). The tumor cells are found to adhere at the microvascular bifurcations more frequently, and there is a positive correlation between the adhesion of the tumor cells and the wall-directed force from the surrounding red blood cells (RBCs). The larger the wall-directed force is, the closer the tumor cells are marginated towards the wall, and the higher the probability of adhesion behavior happen is. A relatively low or high hematocrit can help to prevent the adhesion of tumor cells, and similarly, increasing the shear rate of blood flow can serve the same purpose. These results suggest that the tumor cells may be more likely to extravasate at the microvascular bifurcations if the blood flow is slow and the hematocrit is moderate.
Collapse
Affiliation(s)
- Sitong Wang
- Department of Computational Mathematics, School of Mathematics, Jilin University, Changchun, China
| | - Ting Ye
- Department of Computational Mathematics, School of Mathematics, Jilin University, Changchun, China
- * E-mail:
| | - Guansheng Li
- Department of Computational Mathematics, School of Mathematics, Jilin University, Changchun, China
| | - Xuejiao Zhang
- Department of Computational Mathematics, School of Mathematics, Jilin University, Changchun, China
| | - Huixin Shi
- Department of Computational Mathematics, School of Mathematics, Jilin University, Changchun, China
| |
Collapse
|
41
|
Shen WT, Hsu RS, Fang JH, Hu PF, Chiang CS, Hu SH. Marginative Delivery-Mediated Extracellular Leakiness and T Cell Infiltration in Lung Metastasis by a Biomimetic Nanoraspberry. NANO LETTERS 2021; 21:1375-1383. [PMID: 33562964 DOI: 10.1021/acs.nanolett.0c04122] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
T lymphocytes infiltrate the most devastating metastatic tumors for immunotherapy, allowing the potential for tumor metastasis suppression. However, tumor heterogeneity often restricts the infiltration of immune cells and possesses immune privilege that leads to protection from the immune attack, especially for invading metastatic clusters. Here, an exosome-camouflaged nanoraspberry (RB@Exo) doubling as a metastases-targeting agent and T cell-infiltration inducer that delivers an anticancer drug and energy is reported. The RB@Exo integrated an exosome-derived margination effect, and density-mediated nanoparticle-induced extracellular leakiness (nanoEL) exhibited more than a 70% colocalization of the RB@Exo to metastatic tumors in the lung in vivo. The release of cancer cell-cell interactions at the metastasis via nanoEL also elicited the 10-fold infiltration of T lymphocytes. The synergy of the T cell infiltration and photolytic effects transported by the RB@Exo deep into the metastatic tumors effectively inhibited the tumor in 60 days when treated with a single alternating magnetic field (AMF).
Collapse
Affiliation(s)
- Wei-Ting Shen
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Ru-Siou Hsu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Jen-Hung Fang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Pei-Fen Hu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Chi-Shiun Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Shang-Hsiu Hu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
| |
Collapse
|
42
|
Thai VL, Griffin KH, Thorpe SW, Randall RL, Leach JK. Tissue engineered platforms for studying primary and metastatic neoplasm behavior in bone. J Biomech 2021; 115:110189. [PMID: 33385867 PMCID: PMC7855491 DOI: 10.1016/j.jbiomech.2020.110189] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/02/2020] [Accepted: 12/11/2020] [Indexed: 12/19/2022]
Abstract
Cancer is the second leading cause of death in the United States, claiming more than 560,000 lives each year. Osteosarcoma (OS) is the most common primary malignant tumor of bone in children and young adults, while bone is a common site of metastasis for tumors initiating from other tissues. The heterogeneity, continual evolution, and complexity of this disease at different stages of tumor progression drives a critical need for physiologically relevant models that capture the dynamic cancer microenvironment and advance chemotherapy techniques. Monolayer cultures have been favored for cell-based research for decades due to their simplicity and scalability. However, the nature of these models makes it impossible to fully describe the biomechanical and biochemical cues present in 3-dimensional (3D) microenvironments, such as ECM stiffness, degradability, surface topography, and adhesivity. Biomaterials have emerged as valuable tools to model the behavior of various cancers by creating highly tunable 3D systems for studying neoplasm behavior, screening chemotherapeutic drugs, and developing novel treatment delivery techniques. This review highlights the recent application of biomaterials toward the development of tumor models, details methods for their tunability, and discusses the clinical and therapeutic applications of these systems.
Collapse
Affiliation(s)
- Victoria L Thai
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, United States
| | - Katherine H Griffin
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, United States; School of Veterinary Medicine, University of California, Davis, Davis, CA 95616, United States
| | - Steven W Thorpe
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA 95817, United States
| | - R Lor Randall
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA 95817, United States
| | - J Kent Leach
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, United States; Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA 95817, United States.
| |
Collapse
|
43
|
Abstract
Metastasis is the most complex and deadly event. Tumor-stromal interface is a place where invasion of tumor cells in the form of single-cell or collective migration occurs, with the latter being less common but more efficient. Initiation of metastasis relies on the tumor cell cross-talking with stromal cells and taking an epithelial-mesenchymal transition (EMT) in single cells, and a hybrid EMT in collective migratory cells. Stromal cross-talking along with an abnormal leaky vasculature facilitate intravasation of tumor cells, here the cells are called circulating tumor cells (CTCs). Tumor cells isolated from the primary tumor exploit several mechanisms to maintain their survival including rewiring metabolic demands to use sources available within the new environments, avoiding anoikis cell death when cells are detached from extracellular matrix (ECM), adopting flow mechanic by acquiring platelet shielding and immunosuppression by negating the activity of suppressor immune cells, such as natural killer (NK) cells. CTCs will adhere to the interstituim of the secondary organ/s, within which the newly arrived disseminative tumor cells (DTCs) undergo either dormancy or proliferation. Metastatic outgrowth is under the influence of several factors, such as the activity of macrophages, impaired autophagy and secondary site inflammatory events. Metastasis can be targeted by multiple ways, such as repressing the promoters of pre-metastatic niche (PMN) formation, suppressing environmental contributors, such as hypoxia, oxidative and metabolic stressors, and targeting signaling and cell types that take major contribution to the whole process. These strategies can be used in adjuvant with other therapeutics, such as immunotherapy.
Collapse
Affiliation(s)
- Jamal Majidpoor
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Keywan Mortezaee
- Cancer and Immunology Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran.
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
44
|
Clinical Applications of Patient-Specific 3D Printed Models in Cardiovascular Disease: Current Status and Future Directions. Biomolecules 2020; 10:biom10111577. [PMID: 33233652 PMCID: PMC7699768 DOI: 10.3390/biom10111577] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/19/2020] [Accepted: 11/19/2020] [Indexed: 01/09/2023] Open
Abstract
Three-dimensional (3D) printing has been increasingly used in medicine with applications in many different fields ranging from orthopaedics and tumours to cardiovascular disease. Realistic 3D models can be printed with different materials to replicate anatomical structures and pathologies with high accuracy. 3D printed models generated from medical imaging data acquired with computed tomography, magnetic resonance imaging or ultrasound augment the understanding of complex anatomy and pathology, assist preoperative planning and simulate surgical or interventional procedures to achieve precision medicine for improvement of treatment outcomes, train young or junior doctors to gain their confidence in patient management and provide medical education to medical students or healthcare professionals as an effective training tool. This article provides an overview of patient-specific 3D printed models with a focus on the applications in cardiovascular disease including: 3D printed models in congenital heart disease, coronary artery disease, pulmonary embolism, aortic aneurysm and aortic dissection, and aortic valvular disease. Clinical value of the patient-specific 3D printed models in these areas is presented based on the current literature, while limitations and future research in 3D printing including bioprinting of cardiovascular disease are highlighted.
Collapse
|
45
|
Jang LK, Alvarado JA, Pepona M, Wasson EM, Nash LD, Ortega JM, Randles A, Maitland DJ, Moya ML, Hynes WF. Three-dimensional bioprinting of aneurysm-bearing tissue structure for endovascular deployment of embolization coils. Biofabrication 2020; 13. [DOI: 10.1088/1758-5090/abbb9b] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 09/25/2020] [Indexed: 01/30/2023]
|