1
|
Aw WY, Sawhney A, Rathod M, Whitworth CP, Doherty EL, Madden E, Lu J, Westphal K, Stack R, Polacheck WJ. Dysfunctional mechanotransduction regulates the progression of PIK3CA-driven vascular malformations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.22.609165. [PMID: 39229154 PMCID: PMC11370454 DOI: 10.1101/2024.08.22.609165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Somatic activating mutations in PIK3CA are common drivers of vascular and lymphatic malformations. Despite common biophysical signatures of tissues susceptible to lesion formation, including compliant extracellular matrix and low rates of perfusion, lesions vary in clinical presentation from localized cystic dilatation to diffuse and infiltrative vascular dysplasia. The mechanisms driving the differences in disease severity and variability in clinical presentation and the role of the biophysical microenvironment in potentiating progression are poorly understood. Here, we investigate the role of hemodynamic forces and the biophysical microenvironment in the pathophysiology of vascular malformations, and we identify hemodynamic shear stress and defective endothelial cell mechanotransduction as key regulators of lesion progression. We found that constitutive PI3K activation impaired flow-mediated endothelial cell alignment and barrier function. We show that defective shear stress sensing in PIK3CA E542K endothelial cells is associated with reduced myosin light chain phosphorylation, junctional instability, and defective recruitment of vinculin to cell-cell junctions. Using 3D microfluidic models of the vasculature, we demonstrate that PIK3CA E542K microvessels apply reduced traction forces and are unaffected by flow interruption. We further found that draining transmural flow resulted in increased sprouting and invasion responses in PIK3CA E542K microvessels. Mechanistically, constitutive PI3K activation decreased cellular and nuclear elasticity resulting in defective cellular tensional homeostasis in endothelial cells which may underlie vascular dilation, tissue hyperplasia, and hypersprouting in PIK3CA-driven venous and lymphatic malformations. Together, these results suggest that defective nuclear mechanics, impaired cellular mechanotransduction, and maladaptive hemodynamic responses contribute to the development and progression of PIK3CA-driven vascular malformations.
Collapse
Affiliation(s)
- Wen Yih Aw
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC and Raleigh, NC, USA
| | - Aanya Sawhney
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC and Raleigh, NC, USA
| | - Mitesh Rathod
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC and Raleigh, NC, USA
| | - Chloe P. Whitworth
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC and Raleigh, NC, USA
- Department of Genetics and Molecular Biology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Elizabeth L. Doherty
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC and Raleigh, NC, USA
| | - Ethan Madden
- Department of Genetics and Molecular Biology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Jingming Lu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC and Raleigh, NC, USA
| | - Kaden Westphal
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC and Raleigh, NC, USA
| | - Ryan Stack
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC and Raleigh, NC, USA
| | - William J. Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC and Raleigh, NC, USA
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- McAllister Heart Institute, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
2
|
Hsu YH, Yang WC, Chen YT, Lin CY, Yang CF, Liu WW, Shivani S, Li PC. Spatially controlled diffusion range of tumor-associated angiogenic factors to develop a tumor model using a microfluidic resistive circuit. LAB ON A CHIP 2024; 24:2644-2657. [PMID: 38576341 DOI: 10.1039/d3lc00891f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Developing a tumor model with vessels has been a challenge in microfluidics. This difficulty is because cancer cells can overgrow in a co-culture system. The up-regulation of anti-angiogenic factors during the initial tumor development can hinder neovascularization. The standard method is to develop a quiescent vessel network before loading a tumor construct in an adjacent chamber, which simulates the interaction between a tumor and its surrounding vessels. Here, we present a new method that allows a vessel network and a tumor to develop simultaneously in two linked chambers. The physiological environment of these two chambers is controlled by a microfluidic resistive circuit using two symmetric long microchannels. Applying the resistive circuit, a diffusion-dominated environment with a small 2-D pressure gradient is created across the two chambers with velocity <10.9 nm s-1 and Péclet number <6.3 × 10-5. This 2-D pressure gradient creates a V-shaped velocity clamp to confine the tumor-associated angiogenic factors at pores between the two chambers, and it has two functions. At the early stage, vasculogenesis is stimulated to grow a vessel network in the vessel chamber with minimal influence from the tumor that is still developed in the adjacent chamber. At the post-tumor-development stage, the induced steep concentration gradient at pores mimics vessel-tumor interactions to stimulate angiogenesis to grow vessels toward the tumor. Applying this method, we demonstrate that vasculogenic vessels can grow first, followed by stimulating angiogenesis. Angiogenic vessels can grow into stroma tissue up to 1.3 mm long, and vessels can also grow into or wrap around a 625 μm tumor spheroid or a tumor tissue developed from a cell suspension. In summary, our study suggests that the interactions between a developing vasculature and a growing tumor must be controlled differently throughout the tissue development process, including at the early stage when vessels are still forming and at the later stage when the tumor needs to interact with the vessels.
Collapse
Affiliation(s)
- Yu-Hsiang Hsu
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C.
- Graduate School of Advanced Technology, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C
| | - Wen-Chih Yang
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C.
| | - Yi-Ting Chen
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C.
| | - Che-Yu Lin
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C.
| | - Chiou-Fong Yang
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C.
| | - Wei-Wen Liu
- Graduate Institute of Oral Biology, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C
| | - Subhashree Shivani
- Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei, 10617, Taiwan, R.O.C
| | - Pai-Chi Li
- Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei, 10617, Taiwan, R.O.C
| |
Collapse
|
3
|
Konopka J, Żuchowska A, Jastrzębska E. Vascularized tumor-on-chip microplatforms for the studies of neovasculature as hope for more effective cancer treatments. Biosens Bioelectron 2024; 249:115986. [PMID: 38194813 DOI: 10.1016/j.bios.2023.115986] [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: 10/31/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/11/2024]
Abstract
Angiogenesis is the development of new blood vessels from pre-existing vasculature. Multiple factors control its course. Disorders of the distribution of angiogenic agents are responsible for development of solid tumors and its metastases. Understanding of the molecular interactions regulating pathological angiogenesis will allow for development of more effective, even personalized treatment. A simulation of angiogenesis under microflow conditions is a promising alternative to previous studies conducted on animals and on 2D cell cultures. In this review, we summarize what has been discovered so far in the field of vascularized tumor-on-a-chip platforms. For this purpose, we describe different vascularization techniques used in microfluidics, present various attempts to induce angiogenesis-on-a-chip and report some approaches to recapitulate vascularized tumor microenvironment under microflow conditions.
Collapse
Affiliation(s)
- Joanna Konopka
- Warsaw University of Technology, Faculty of Chemistry, Medical Biotechnology, 00-664, Warsaw, Poland
| | - Agnieszka Żuchowska
- Warsaw University of Technology, Faculty of Chemistry, Medical Biotechnology, 00-664, Warsaw, Poland
| | - Elżbieta Jastrzębska
- Warsaw University of Technology, Faculty of Chemistry, Medical Biotechnology, 00-664, Warsaw, Poland.
| |
Collapse
|
4
|
Cherubini M, Erickson S, Padmanaban P, Haberkant P, Stein F, Beltran-Sastre V, Haase K. Flow in fetoplacental-like microvessels in vitro enhances perfusion, barrier function, and matrix stability. SCIENCE ADVANCES 2023; 9:eadj8540. [PMID: 38134282 PMCID: PMC10745711 DOI: 10.1126/sciadv.adj8540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023]
Abstract
Proper placental vascularization is vital for pregnancy outcomes, but assessing it with animal models and human explants has limitations. We introduce a 3D in vitro model of human placenta terminal villi including fetal mesenchyme and vascular endothelium. By coculturing HUVEC, placental fibroblasts, and pericytes in a macrofluidic chip with a flow reservoir, we generate fully perfusable fetal microvessels. Pressure-driven flow facilitates microvessel growth and remodeling, resulting in early formation of interconnected and lasting placental-like vascular networks. Computational fluid dynamics simulations predict shear forces, which increase microtissue stiffness, decrease diffusivity, and enhance barrier function as shear stress rises. Mass spectrometry analysis reveals enhanced protein expression with flow, including matrix stability regulators, proteins associated with actin dynamics, and cytoskeleton organization. Our model provides a powerful tool for deducing complex in vivo parameters, such as shear stress on developing vascularized placental tissue, and holds promise for unraveling gestational disorders related to the vasculature.
Collapse
Affiliation(s)
- Marta Cherubini
- European Molecular Biology Laboratory (EMBL), Barcelona, Spain
| | - Scott Erickson
- European Molecular Biology Laboratory (EMBL), Barcelona, Spain
| | | | - Per Haberkant
- Proteomics Core Facility, EMBL Heidelberg, Heidelberg, Germany
| | - Frank Stein
- Proteomics Core Facility, EMBL Heidelberg, Heidelberg, Germany
| | | | - Kristina Haase
- European Molecular Biology Laboratory (EMBL), Barcelona, Spain
| |
Collapse
|
5
|
Santamaría R, Cruz-Caballero J, Gkontra P, Jiménez-Montiel A, Clemente C, López JA, Villalba-Orero M, Vázquez J, Hutloff A, Lara-Pezzi E, Arroyo AG. Capillary pruning couples tissue perfusion and oxygenation with cardiomyocyte maturation in the postnatal mouse heart. Front Cell Dev Biol 2023; 11:1256127. [PMID: 38020883 PMCID: PMC10661946 DOI: 10.3389/fcell.2023.1256127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction: Removal of poorly perfused capillaries by pruning contributes to remodeling the microvasculature to optimize oxygen and nutrient delivery. Blood flow drives this process by promoting the intravascular migration of endothelial cells in developing networks, such as in the yolk sac, zebrafish brain or postnatal mouse retina. Methods: In this study, we have implemented innovative tools to recognize capillary pruning in the complex 3D coronary microvasculature of the postnatal mouse heart. We have also experimentally tested the impact of decreasing pruning on the structure and function of this network by altering blood flow with two different vasodilators: losartan and prazosin. Results: Although both drugs reduced capillary pruning, a combination of experiments based on ex vivo imaging, proteomics, electron microscopy and in vivo functional approaches showed that losartan treatment resulted in an inefficient coronary network, reduced myocardial oxygenation and metabolic changes that delayed the arrest of cardiomyocyte proliferation, in contrast to the effects of prazosin, probably due to its concomitant promotion of capillary expansion. Discussion: Our work demonstrates that capillary pruning contributes to proper maturation and function of the heart and that manipulation of blood flow may be a novel strategy to refine the microvasculature and improve tissue perfusion after damage.
Collapse
Affiliation(s)
- Ricardo Santamaría
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Polyxeni Gkontra
- Artificial Intelligence in Medicine Lab (BCN-AIM), Departament de Matemàtiques i Informàtica, Universitat de Barcelona, Barcelona, Spain
| | | | - Cristina Clemente
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Juan A. López
- Cardiovascular Proteomics Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - María Villalba-Orero
- Myocardial Pathology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Jesús Vázquez
- Cardiovascular Proteomics Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Andreas Hutloff
- Institute of Immunology, University Hospital Schleswig-Holstein, Kiel, Germany
- German Rheumatism Research Centre, A Leibniz Institute, Berlin, Germany
| | - Enrique Lara-Pezzi
- Myocardial Pathology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Alicia G. Arroyo
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| |
Collapse
|
6
|
Russell T, Dirar Q, Li Y, Chiang C, Laskowitz DT, Yun Y. Cortical spheroid on perfusable microvascular network in a microfluidic device. PLoS One 2023; 18:e0288025. [PMID: 37856438 PMCID: PMC10586606 DOI: 10.1371/journal.pone.0288025] [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: 01/06/2023] [Accepted: 06/18/2023] [Indexed: 10/21/2023] Open
Abstract
Human induced pluripotent stem cell (hiPSC)-derived brain spheroids can recapitulate the complex cytoarchitecture of the brain, as well as the genetic/epigenetic footprint of human brain development. However, hiPSC-derived 3D models such as spheroid and organoids does not have a perfusable microvascular network, which plays a vital role in maintaining homeostasis in vivo. With the critical balance of positive and negative angiogenic modulators, 3D microvascular network can be achieved by angiogenesis. This paper reports on a microfluidic-based three-dimensional, cortical spheroid grafted on the vascular-network. Vascular network was formed by inducing angiogenic sprouting using concentration gradient-driven angiogenic factors in the microfluidic device. We investigate critical factors for angiogenic vascular network formation with spheroid placement, including 1) a PKCα activator, phorbol-12-myristate-13-acetate (PMA); 2) orientation of endothelial cells under perfusion and permeability of vascular network; 3) effect of extracellular matrix (ECM) types and their densities on angiogenesis; and 4) integration with cortical spheroid on vascular network. This paper demonstrates proof of concept for the potential utility of a membrane-free in vitro cortical spheroid tissue construct with perfusable microvascular network that can be scaled up to a high throughput platform. It can provide a cost-effective alternative platform to animal testing by modeling brain diseases and disorders, and screening drugs.
Collapse
Affiliation(s)
- Teal Russell
- Fostering Innovation Through Biosystems for Enhanced Scientific Technologies (FIT BEST) Laboratory, Department of Chemical, Biological, and Bio Engineering, College of Engineering, North Carolina A&T State University, Greensboro, NC, United States of America
| | - Qassim Dirar
- Fostering Innovation Through Biosystems for Enhanced Scientific Technologies (FIT BEST) Laboratory, Department of Chemical, Biological, and Bio Engineering, College of Engineering, North Carolina A&T State University, Greensboro, NC, United States of America
| | - Yan Li
- Chemical & Biomedical Engineering, College of Engineering, Florida A&M University-Florida State University, Tallahassee, FL, United States of America
| | | | - Daniel T. Laskowitz
- Department of Neurology, Duke University Medical Center, Durham, NC, United States of America
| | - Yeoheung Yun
- Fostering Innovation Through Biosystems for Enhanced Scientific Technologies (FIT BEST) Laboratory, Department of Chemical, Biological, and Bio Engineering, College of Engineering, North Carolina A&T State University, Greensboro, NC, United States of America
| |
Collapse
|
7
|
Wang H, Lu J, Rathod M, Aw WY, Huang SA, Polacheck WJ. A facile fluid pressure system reveals differential cellular response to interstitial pressure gradients and flow. BIOMICROFLUIDICS 2023; 17:054103. [PMID: 37781136 PMCID: PMC10539030 DOI: 10.1063/5.0165119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/09/2023] [Indexed: 10/03/2023]
Abstract
Interstitial fluid pressure gradients and interstitial flow have been shown to drive morphogenic processes that shape tissues and influence progression of diseases including cancer. The advent of porous media microfluidic approaches has enabled investigation of the cellular response to interstitial flow, but questions remain as to the critical biophysical and biochemical signals imparted by interstitial fluid pressure gradients and resulting flow on resident cells and extracellular matrix (ECM). Here, we introduce a low-cost method to maintain physiological interstitial fluid pressures that is built from commonly accessible laboratory equipment, including a laser pointer, camera, Arduino board, and a commercially available linear actuator. We demonstrate that when the system is connected to a microfluidic device containing a 3D porous hydrogel, physiologic pressure is maintained with sub-Pascal resolution and when basic feedback control is directed using an Arduino, constant pressure and pressure gradient can be maintained even as cells remodel and degrade the ECM hydrogel over time. Using this model, we characterized breast cancer cell growth and ECM changes to ECM fibril structure and porosity in response to constant interstitial fluid pressure or constant interstitial flow. We observe increased collagen fibril bundling and the formation of porous structures in the vicinity of cancer cells in response to constant interstitial fluid pressure as compared to constant interstitial flow. Collectively, these results further define interstitial fluid pressure as a driver of key pathogenic responses in cells, and the systems and methods developed here will allow for future mechanistic work investigating mechanotransduction of interstitial fluid pressures and flows.
Collapse
Affiliation(s)
- Hao Wang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | - Jingming Lu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | - Mitesh Rathod
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | - Wen Yih Aw
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | - Stephanie A. Huang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | | |
Collapse
|
8
|
Juste-Lanas Y, Hervas-Raluy S, García-Aznar JM, González-Loyola A. Fluid flow to mimic organ function in 3D in vitro models. APL Bioeng 2023; 7:031501. [PMID: 37547671 PMCID: PMC10404142 DOI: 10.1063/5.0146000] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/20/2023] [Indexed: 08/08/2023] Open
Abstract
Many different strategies can be found in the literature to model organ physiology, tissue functionality, and disease in vitro; however, most of these models lack the physiological fluid dynamics present in vivo. Here, we highlight the importance of fluid flow for tissue homeostasis, specifically in vessels, other lumen structures, and interstitium, to point out the need of perfusion in current 3D in vitro models. Importantly, the advantages and limitations of the different current experimental fluid-flow setups are discussed. Finally, we shed light on current challenges and future focus of fluid flow models applied to the newest bioengineering state-of-the-art platforms, such as organoids and organ-on-a-chip, as the most sophisticated and physiological preclinical platforms.
Collapse
Affiliation(s)
| | - Silvia Hervas-Raluy
- Department of Mechanical Engineering, Engineering Research Institute of Aragón (I3A), University of Zaragoza, Zaragoza, Spain
| | | | | |
Collapse
|
9
|
Lee S, Ahn J, Kim SM, Kim D, Yeom J, Kim J, Park JY, Ryu BY. Fluid dynamic design for mitigating undesired cell effects and its application to testis cell response testing to endocrine disruptors. J Biol Eng 2023; 17:51. [PMID: 37550751 PMCID: PMC10408176 DOI: 10.1186/s13036-023-00369-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/16/2023] [Indexed: 08/09/2023] Open
Abstract
Microfluidic devices have emerged as powerful tools for cell-based experiments, offering a controlled microenvironment that mimic the conditions within the body. Numerous cell experiment studies have successfully utilized microfluidic channels to achieve various new scientific discoveries. However, it has been often overlooked that undesired and unnoticed propagation of cellular molecules in such bio-microfluidic channel systems can have a negative impact on the experimental results. Thus, more careful designing is required to minimize such unwanted issues through deeper understanding and careful control of chemically and physically predominant factors at the microscopic scale. In this paper, we introduce a new approach to improve microfluidic channel design, specifically targeting the mitigation of the aforementioned challenges. To minimize the occurrence of undesired cell positioning upstream from the main test section where a concentration gradient field locates, an additional narrow port structure was devised between the microfluidic upstream channel and each inlet reservoir. This port also functioned as a passive lock that hold the flow at rest via fluid-air surface tension, which facilitated manual movement of the device even when cell attachment was not achieved completely. To demonstrate the practicability of the system, we conducted experiments and diffusion simulations on the effect of endocrine disruptors on germ cells. To this end, a bisphenol-A (BPA) concentration gradient was generated in the main channel of the system at BPA concentrations ranging from 120.8 μM to 79.3 μM, and the proliferation of GC-1 cells in the BPA gradient environment was quantitatively evaluated. The features and concepts of the introduced design is to minimize unexpected and ignored error sources, which will be one of the issues to be considered in the development of microfluidic systems to explore extremely delicate cellular phenomena.
Collapse
Affiliation(s)
- Seungjin Lee
- School of Mechanical Engineering, College of Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jinseop Ahn
- Present address: Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Animal Science and Technology, BET Research Institute, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Seok-Man Kim
- Department of Animal Science and Technology, BET Research Institute, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Daehan Kim
- School of Mechanical Engineering, College of Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jiun Yeom
- School of Mechanical Engineering, College of Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jeongmok Kim
- School of Mechanical Engineering, College of Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Joong Yull Park
- School of Mechanical Engineering, College of Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea.
- Department of Intelligent Energy and Industry, Graduate School, Chung-Ang University, Seoul, 06974, Republic of Korea.
| | - Buom-Yong Ryu
- Department of Animal Science and Technology, BET Research Institute, Chung-Ang University, Anseong, 17546, Republic of Korea.
| |
Collapse
|
10
|
Arslan U, Brescia M, Meraviglia V, Nahon DM, van Helden RWJ, Stein JM, van den Hil FE, van Meer BJ, Vila Cuenca M, Mummery CL, Orlova VV. Vascularized hiPSC-derived 3D cardiac microtissue on chip. Stem Cell Reports 2023; 18:1394-1404. [PMID: 37390826 PMCID: PMC10362508 DOI: 10.1016/j.stemcr.2023.06.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 06/02/2023] [Accepted: 06/02/2023] [Indexed: 07/02/2023] Open
Abstract
Functional vasculature is essential for delivering nutrients, oxygen, and cells to the heart and removing waste products. Here, we developed an in vitro vascularized human cardiac microtissue (MT) model based on human induced pluripotent stem cells (hiPSCs) in a microfluidic organ-on-chip by coculturing hiPSC-derived, pre-vascularized, cardiac MTs with vascular cells within a fibrin hydrogel. We showed that vascular networks spontaneously formed in and around these MTs and were lumenized and interconnected through anastomosis. Anastomosis was fluid flow dependent: continuous perfusion increased vessel density and thus enhanced the formation of the hybrid vessels. Vascularization further improved endothelial cell (EC)-cardiomyocyte communication via EC-derived paracrine factors, such as nitric oxide, and resulted in an enhanced inflammatory response. The platform sets the stage for studies on how organ-specific EC barriers respond to drugs or inflammatory stimuli.
Collapse
Affiliation(s)
- Ulgu Arslan
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2333ZC Leiden, the Netherlands
| | - Marcella Brescia
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2333ZC Leiden, the Netherlands
| | - Viviana Meraviglia
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2333ZC Leiden, the Netherlands
| | - Dennis M Nahon
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2333ZC Leiden, the Netherlands
| | - Ruben W J van Helden
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2333ZC Leiden, the Netherlands
| | - Jeroen M Stein
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2333ZC Leiden, the Netherlands
| | - Francijna E van den Hil
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2333ZC Leiden, the Netherlands
| | - Berend J van Meer
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2333ZC Leiden, the Netherlands
| | - Marc Vila Cuenca
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2333ZC Leiden, the Netherlands; Department of Clinical Genetics, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2333ZC Leiden, the Netherlands
| | - Valeria V Orlova
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2333ZC Leiden, the Netherlands.
| |
Collapse
|
11
|
Winkelman MA, Dai G. Bioengineered perfused human brain microvascular networks enhance neural progenitor cell survival, neurogenesis, and maturation. SCIENCE ADVANCES 2023; 9:eaaz9499. [PMID: 37163593 PMCID: PMC10171804 DOI: 10.1126/sciadv.aaz9499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 04/10/2023] [Indexed: 05/12/2023]
Abstract
Neural progenitor cells (NPCs) have the capability to self-renew and differentiate into neurons and glial cells. In the adult brain, NPCs are found near brain microvascular networks (BMVNs) in specialized microenvironments called the neurovascular niche (NVN). Although several in vitro NVN models have been previously reported, most do not properly recapitulate the intimate cellular interactions between NPCs and perfused brain microvessels. Here, we developed perfused BMVNs composed of primary human brain endothelial cells, pericytes, and astrocytes within microfluidic devices. When induced pluripotent stem cell-derived NPCs were introduced into BMVNs, we found that NPC survival, neurogenesis, and maturation were enhanced. The application of flow during BMVN coculture was also beneficial for neuron differentiation. Collectively, our work highlighted the important role of BMVNs and flow in NPC self-renewal and neurogenesis, as well as demonstrated our model's potential to study the biological and physical interactions of human NVN in vitro.
Collapse
Affiliation(s)
- Max A. Winkelman
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | | |
Collapse
|
12
|
Wan HY, Chen JCH, Xiao Q, Wong CW, Yang B, Cao B, Tuan RS, Nilsson SK, Ho YP, Raghunath M, Kamm RD, Blocki A. Stabilization and improved functionality of three-dimensional perfusable microvascular networks in microfluidic devices under macromolecular crowding. Biomater Res 2023; 27:32. [PMID: 37076899 PMCID: PMC10116810 DOI: 10.1186/s40824-023-00375-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 04/04/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND There is great interest to engineer in vitro models that allow the study of complex biological processes of the microvasculature with high spatiotemporal resolution. Microfluidic systems are currently used to engineer microvasculature in vitro, which consists of perfusable microvascular networks (MVNs). These are formed through spontaneous vasculogenesis and exhibit the closest resemblance to physiological microvasculature. Unfortunately, under standard culture conditions and in the absence of co-culture with auxiliary cells as well as protease inhibitors, pure MVNs suffer from a short-lived stability. METHODS Herein, we introduce a strategy for stabilization of MVNs through macromolecular crowding (MMC) based on a previously established mixture of Ficoll macromolecules. The biophysical principle of MMC is based on macromolecules occupying space, thus increasing the effective concentration of other components and thereby accelerating various biological processes, such as extracellular matrix deposition. We thus hypothesized that MMC will promote the accumulation of vascular ECM (basement membrane) components and lead to a stabilization of MVN with improved functionality. RESULTS MMC promoted the enrichment of cellular junctions and basement membrane components, while reducing cellular contractility. The resulting advantageous balance of adhesive forces over cellular tension resulted in a significant stabilization of MVNs over time, as well as improved vascular barrier function, closely resembling that of in vivo microvasculature. CONCLUSION Application of MMC to MVNs in microfluidic devices provides a reliable, flexible and versatile approach to stabilize engineered microvessels under simulated physiological conditions.
Collapse
Affiliation(s)
- Ho-Ying Wan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jack Chun Hin Chen
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qinru Xiao
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Christy Wingtung Wong
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Boguang Yang
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Benjamin Cao
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organisation (CSIRO), Melbourne, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia
| | - Rocky S Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine (CNRM), Hong Kong Science Park, Shatin, New Territories, Hong Kong SAR, China
| | - Susan K Nilsson
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organisation (CSIRO), Melbourne, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia
| | - Yi-Ping Ho
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Michael Raghunath
- Institute for Chemistry and Biotechnology, Zurich University of Applied Sciences, Wädenswil, Switzerland
| | - Roger D Kamm
- Department of Biology and Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anna Blocki
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
- Center for Neuromusculoskeletal Restorative Medicine (CNRM), Hong Kong Science Park, Shatin, New Territories, Hong Kong SAR, China.
| |
Collapse
|
13
|
Chang CW, Shih HC, Cortes-Medina MG, Beshay PE, Avendano A, Seibel AJ, Liao WH, Tung YC, Song JW. Extracellular Matrix-Derived Biophysical Cues Mediate Interstitial Flow-Induced Sprouting Angiogenesis. ACS APPLIED MATERIALS & INTERFACES 2023; 15:15047-15058. [PMID: 36916875 PMCID: PMC11078157 DOI: 10.1021/acsami.2c15180] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Sprouting angiogenesis is orchestrated by an intricate balance of biochemical and mechanical cues in the local tissue microenvironment. Interstitial flow has been established as a potent regulator of angiogenesis. Similarly, extracellular matrix (ECM) physical properties, such as stiffness and microarchitecture, have also emerged as important mediators of angiogenesis. However, the interplay between interstitial flow and ECM physical properties in the initiation and control of angiogenesis is poorly understood. Using a three-dimensional (3D) microfluidic tissue analogue of angiogenic sprouting with defined interstitial flow superimposed over ECM with well-characterized physical properties, we found that the addition of hyaluronan (HA) to collagen-based matrices significantly enhances sprouting induced by interstitial flow compared to responses in collagen-only hydrogels. We confirmed that both the stiffness and matrix pore size of collagen-only hydrogels were increased by the addition of HA. Interestingly, interstitial flow-potentiated sprouting responses in collagen/HA matrices were not affected when functionally blocking the HA receptor CD44. In contrast, enzymatic depletion of HA in collagen/HA matrices with hyaluronidase (HAdase) resulted in decreased stiffness, pore size, and interstitial flow-mediated sprouting to the levels observed in collagen-only matrices. Taken together, these results suggest that HA enhances interstitial flow-mediated angiogenic sprouting through its alterations to collagen ECM stiffness and pore size.
Collapse
Affiliation(s)
- Chia-Wen Chang
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Hsiu-Chen Shih
- Research Center for Applied Science, Academia Sinica, Taipei 115-29, Taiwan
| | - Marcos G Cortes-Medina
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Peter E Beshay
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Alex Avendano
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Alex J Seibel
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Wei-Hao Liao
- Research Center for Applied Science, Academia Sinica, Taipei 115-29, Taiwan
| | - Yi-Chung Tung
- Research Center for Applied Science, Academia Sinica, Taipei 115-29, Taiwan
| | - Jonathan W Song
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, Ohio 43210, United States
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
14
|
Cabral KA, Srivastava V, Graham AJ, Coyle MC, Stashko C, Weaver V, Gartner ZJ. Programming the Self-Organization of Endothelial Cells into Perfusable Microvasculature. Tissue Eng Part A 2023; 29:80-92. [PMID: 36181350 PMCID: PMC10266707 DOI: 10.1089/ten.tea.2022.0072] [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: 04/01/2022] [Accepted: 09/15/2022] [Indexed: 11/12/2022] Open
Abstract
The construction of three-dimensional (3D) microvascular networks with defined structures remains challenging. Emerging bioprinting strategies provide a means of patterning endothelial cells (ECs) into the geometry of 3D microvascular networks, but the microenvironmental cues necessary to promote their self-organization into cohesive and perfusable microvessels are not well known. To this end, we reconstituted microvessel formation in vitro by patterning thin lines of closely packed ECs fully embedded within a 3D extracellular matrix (ECM) and observed how different microenvironmental parameters influenced EC behaviors and their self-organization into microvessels. We found that the inclusion of fibrillar matrices, such as collagen I, into the ECM positively influenced cell condensation into extended geometries such as cords. We also identified the presence of a high-molecular-weight protein(s) in fetal bovine serum that negatively influenced EC condensation. This component destabilized cord structure by promoting cell protrusions and destabilizing cell-cell adhesions. Endothelial cords cultured in the presence of fibrillar collagen and in the absence of this protein activity were able to polarize, lumenize, incorporate mural cells, and support fluid flow. These optimized conditions allowed for the construction of branched and perfusable microvascular networks directly from patterned cells in as little as 3 days. These findings reveal important design principles for future microvascular engineering efforts based on bioprinting and micropatterning techniques. Impact statement Bioprinting is a potential strategy to achieve microvascularization in engineered tissues. However, the controlled self-organization of patterned endothelial cells into perfusable microvasculature remains challenging. We used DNA Programmed Assembly of Cells to create cell-dense, capillary-sized cords of endothelial cells with complete control over their structure. We optimized the matrix and media conditions to promote self-organization and maturation of these endothelial cords into stable and perfusable microvascular networks.
Collapse
Affiliation(s)
- Katelyn A. Cabral
- Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, Berkeley, California, USA
| | - Vasudha Srivastava
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
| | - Austin J. Graham
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
- Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, California, USA
| | - Maxwell C. Coyle
- Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley, California, USA
| | - Connor Stashko
- Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, Berkeley, California, USA
| | - Valerie Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Zev J. Gartner
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
- Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, California, USA
- Center for Cellular Construction, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
15
|
Vascularized Tissue Organoids. Bioengineering (Basel) 2023; 10:bioengineering10020124. [PMID: 36829618 PMCID: PMC9951914 DOI: 10.3390/bioengineering10020124] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/19/2023] Open
Abstract
Tissue organoids hold enormous potential as tools for a variety of applications, including disease modeling and drug screening. To effectively mimic the native tissue environment, it is critical to integrate a microvasculature with the parenchyma and stroma. In addition to providing a means to physiologically perfuse the organoids, the microvasculature also contributes to the cellular dynamics of the tissue model via the cells of the perivascular niche, thereby further modulating tissue function. In this review, we discuss current and developing strategies for vascularizing organoids, consider tissue-specific vascularization approaches, discuss the importance of perfusion, and provide perspectives on the state of the field.
Collapse
|
16
|
Mathur T, Tronolone JJ, Jain A. AngioMT: An in silico platform for digital sensing of oxygen transport through heterogenous microvascular networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.09.523275. [PMID: 36711826 PMCID: PMC9881947 DOI: 10.1101/2023.01.09.523275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Measuring the capacity of microvascular networks in delivering soluble oxygen and nutrients to its organs is essential in health, disease, and surgical interventions. Here, a finite element method-based oxygen transport program, AngioMT, is designed and validated to predict spatial oxygen distribution and other physiologically relevant transport metrics within both the vascular network and the surrounding tissue. The software processes acquired images of microvascular networks and produces a digital mesh which is used to predict vessel and tissue oxygenation. The image-to-physics translation by AngioMT correlated with results from commercial software, however only AngioMT could provide predictions within the solid tissue in addition to vessel oxygenation. AngioMT predictions were sensitive and positively correlated to spatial heterogeneity and extent of vascularization of 500 different vascular networks formed with variable vasculogenic conditions. The predictions of AngioMT cross-correlate with experimentally-measured oxygen distributions in vivo. The computational power of the software is increased by including calculations of higher order reaction mechanisms, and the program includes defining additional organ and tissue structures for a more physiologically relevant analysis of tissue oxygenation in complex co-cultured systems, or in vivo. AngioMT may serve as a digital performance measuring tool of vascular networks in microcirculation, experimental models of vascularized tissues and organs, and in clinical applications, such as organ transplants.
Collapse
Affiliation(s)
- Tanmay Mathur
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St, College Station, TX 77843, USA
| | - James J. Tronolone
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St, College Station, TX 77843, USA
| | - Abhishek Jain
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St, College Station, TX 77843, USA
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, TX, USA
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
| |
Collapse
|
17
|
Konopka J, Kołodziejek D, Flont M, Żuchowska A, Jastrzębska E, Brzózka Z. Exploring Endothelial Expansion on a Chip. SENSORS (BASEL, SWITZERLAND) 2022; 22:9414. [PMID: 36502120 PMCID: PMC9741423 DOI: 10.3390/s22239414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/22/2022] [Accepted: 11/29/2022] [Indexed: 06/17/2023]
Abstract
Angiogenesis is the development of new blood vessels from the existing vasculature. Its malfunction leads to the development of cancers and cardiovascular diseases qualified by the WHO as a leading cause of death worldwide. A better understanding of mechanisms regulating physiological and pathological angiogenesis will potentially contribute to developing more effective treatments for those urgent issues. Therefore, the main goal of the following study was to design and manufacture an angiogenesis-on-a-chip microplatform, including cylindrical microvessels created by Viscous Finger Patterning (VFP) technique and seeded with HUVECs. While optimizing the VFP procedure, we have observed that lumen's diameter decreases with a diminution of the droplet's volume. The influence of Vascular Endothelial Growth Factor (VEGF) with a concentration of 5, 25, 50, and 100 ng/mL on the migration of HUVECs was assessed. VEGF's solution with concentrations varying from 5 to 50 ng/mL reveals high angiogenic potential. The spatial arrangement of cells and their morphology were visualized by fluorescence and confocal microscopy. Migration of HUVECs toward loaded angiogenic stimuli has been initiated after overnight incubation. This research is the basis for developing more complex vascularized multi-organ-on-a-chip microsystems that could potentially be used for drug screening.
Collapse
Affiliation(s)
- Joanna Konopka
- Faculty of Chemistry, Warsaw University of Technology, 00-661 Warszawa, Poland
| | - Dominik Kołodziejek
- Faculty of Chemistry, Warsaw University of Technology, 00-661 Warszawa, Poland
| | - Magdalena Flont
- Centre for Advanced Materials and Technologies CEZAMAT, Warsaw University of Technology, 02-822 Warszawa, Poland
| | - Agnieszka Żuchowska
- Faculty of Chemistry, Warsaw University of Technology, 00-661 Warszawa, Poland
| | - Elżbieta Jastrzębska
- Faculty of Chemistry, Warsaw University of Technology, 00-661 Warszawa, Poland
- Centre for Advanced Materials and Technologies CEZAMAT, Warsaw University of Technology, 02-822 Warszawa, Poland
| | - Zbigniew Brzózka
- Faculty of Chemistry, Warsaw University of Technology, 00-661 Warszawa, Poland
| |
Collapse
|
18
|
Barrasa-Ramos S, Dessalles CA, Hautefeuille M, Barakat AI. Mechanical regulation of the early stages of angiogenesis. J R Soc Interface 2022; 19:20220360. [PMID: 36475392 PMCID: PMC9727679 DOI: 10.1098/rsif.2022.0360] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Favouring or thwarting the development of a vascular network is essential in fields as diverse as oncology, cardiovascular disease or tissue engineering. As a result, understanding and controlling angiogenesis has become a major scientific challenge. Mechanical factors play a fundamental role in angiogenesis and can potentially be exploited for optimizing the architecture of the resulting vascular network. Largely focusing on in vitro systems but also supported by some in vivo evidence, the aim of this Highlight Review is dual. First, we describe the current knowledge with particular focus on the effects of fluid and solid mechanical stimuli on the early stages of the angiogenic process, most notably the destabilization of existing vessels and the initiation and elongation of new vessels. Second, we explore inherent difficulties in the field and propose future perspectives on the use of in vitro and physics-based modelling to overcome these difficulties.
Collapse
Affiliation(s)
- Sara Barrasa-Ramos
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Claire A. Dessalles
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Mathieu Hautefeuille
- Laboratoire de Biologie du Développement (UMR7622), Institut de Biologie Paris Seine, Sorbonne Université, Paris, France,Facultad de Ciencias, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Abdul I. Barakat
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| |
Collapse
|
19
|
Deng P, Zhao M, Zhang X, Qin J. A Transwell-Based Vascularized Model to Investigate the Effect of Interstitial Flow on Vasculogenesis. Bioengineering (Basel) 2022; 9:668. [PMID: 36354579 PMCID: PMC9687519 DOI: 10.3390/bioengineering9110668] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/27/2022] [Accepted: 10/30/2022] [Indexed: 09/08/2024] Open
Abstract
Interstitial flow plays a significant role in vascular system development, mainly including angiogenesis and vasculogenesis. However, compared to angiogenesis, the effect of interstitial flow on vasculogenesis is less explored. Current in vitro models for investigating the effect of interstitial flow on vasculogenesis heavily rely on microfluidic chips, which require microfluidic expertise and facilities, and may not be accessible to biological labs. Here, we proposed a facile approach to building perfusable vascular networks through the self-assembly of endothelial cells in a modified transwell format and investigated the effect of interstitial flow on vasculogenesis. We found that the effect of interstitial flow on vasculogenesis was closely related to the existence of VEGF and fibroblasts in the developed model: (1) In the presence of fibroblasts, interstitial flow (within the range of 0.1-0.6 μm/s) facilitated the perfusability of the engineered vasculatures. Additional VEGF in the culture medium further worked synergically with interstitial flow to develop longer, wider, denser, and more perfusable vasculatures than static counterparts; (2) In the absence of fibroblasts, vasculatures underwent severe regression within 7 days under static conditions. However, interstitial flow greatly inhibited vessel regression and enhanced vascular perfusability and morphogenesis without the need for additional VEGF. These results revealed that the effect of interstitial flow might vary depending on the existence of VEGF and fibroblasts, and would provide some guidelines for constructing in vitro self-assembled vasculatures. The established transwell-based vascularized model provides a simple method to build perfusable vasculatures and could also be utilized for creating functional tissues in regenerative medicine.
Collapse
Affiliation(s)
- Pengwei Deng
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengqian Zhao
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xu Zhang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Jianhua Qin
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
20
|
LaMontagne E, Muotri AR, Engler AJ. Recent advancements and future requirements in vascularization of cortical organoids. Front Bioeng Biotechnol 2022; 10:1048731. [PMID: 36406234 PMCID: PMC9669755 DOI: 10.3389/fbioe.2022.1048731] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/18/2022] [Indexed: 07/23/2023] Open
Abstract
The fields of tissue engineering and disease modeling have become increasingly cognizant of the need to create complex and mature structures in vitro to adequately mimic the in vivo niche. Specifically for neural applications, human brain cortical organoids (COs) require highly stratified neurons and glial cells to generate synaptic functions, and to date, most efforts achieve only fetal functionality at best. Moreover, COs are usually avascular, inducing the development of necrotic cores, which can limit growth, development, and maturation. Recent efforts have attempted to vascularize cortical and other organoid types. In this review, we will outline the components of a fully vascularized CO as they relate to neocortical development in vivo. These components address challenges in recapitulating neurovascular tissue patterning, biomechanical properties, and functionality with the goal of mirroring the quality of organoid vascularization only achieved with an in vivo host. We will provide a comprehensive summary of the current progress made in each one of these categories, highlighting advances in vascularization technologies and areas still under investigation.
Collapse
Affiliation(s)
- Erin LaMontagne
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
| | - Alysson R. Muotri
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, United States
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Adam J. Engler
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| |
Collapse
|
21
|
Zhang S, Wan Z, Pavlou G, Zhong AX, Xu L, Kamm RD. Interstitial flow promotes the formation of functional microvascular networks in vitro through upregulation of matrix metalloproteinase-2. ADVANCED FUNCTIONAL MATERIALS 2022; 32:2206767. [PMID: 36569597 PMCID: PMC9783342 DOI: 10.1002/adfm.202206767] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Indexed: 05/02/2023]
Abstract
Self-organized microvascular networks (MVNs) have become key to the development of many microphysiological models. However, the self-organizing nature of this process combined with variations between types or batches of endothelial cells (ECs) often lead to inconsistency or failure to form functional MVNs. Since interstitial flow (IF) has been reported to play a beneficial role in angiogenesis, vasculogenesis, and 3D capillary morphogenesis, we systematically investigated the role IF plays during neovessel formation in a customized single channel microfluidic chip for which IF has been fully characterized. Compared to static conditions, MVNs formed under IF have higher vessel density and diameters and greater network perfusability. Through a series of inhibitory experiments, we demonstrated that IF treatment improves vasculogenesis by ECs through upregulation of matrix metalloproteinase-2 (MMP-2). We then successfully implemented a novel strategy involving the interplay between IF and MMP-2 inhibitor to regulate morphological parameters of the self-organized MVNs, with vascular permeability and perfusability well maintained. The revealed mechanism and proposed methodology were further validated with a brain MVN model. Our findings and methods have the potential to be widely utilized to boost the development of various organotypic MVNs and could be incorporated into related bioengineering applications where perfusable vasculature is desired.
Collapse
Affiliation(s)
- Shun Zhang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Zhengpeng Wan
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Georgios Pavlou
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Amy X Zhong
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Liling Xu
- Ragon institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - Roger D Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
22
|
Zhang S, Kan EL, Kamm RD. Integrating functional vasculature into organoid culture: A biomechanical perspective. APL Bioeng 2022; 6:030401. [DOI: 10.1063/5.0097967] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/27/2022] [Indexed: 12/30/2022] Open
Affiliation(s)
- Shun Zhang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Ellen L. Kan
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Roger D. Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
23
|
Sayer S, Zandrini T, Markovic M, Van Hoorick J, Van Vlierberghe S, Baudis S, Holnthoner W, Ovsianikov A. Guiding cell migration in 3D with high-resolution photografting. Sci Rep 2022; 12:8626. [PMID: 35606455 PMCID: PMC9126875 DOI: 10.1038/s41598-022-11612-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 03/24/2022] [Indexed: 11/09/2022] Open
Abstract
Multi-photon lithography (MPL) has proven to be a suitable tool to precisely control the microenvironment of cells in terms of the biochemical and biophysical properties of the hydrogel matrix. In this work, we present a novel method, based on multi-photon photografting of 4,4′-diazido-2,2′-stilbenedisulfonic acid (DSSA), and its capabilities to induce cell alignment, directional cell migration and endothelial sprouting in a gelatin-based hydrogel matrix. DSSA-photografting allows for the fabrication of complex patterns at a high-resolution and is a biocompatible, universally applicable and straightforward process that is comparably fast. We have demonstrated the preferential orientation of human adipose-derived stem cells (hASCs) in response to a photografted pattern. Co-culture spheroids of hASCs and human umbilical vein endothelial cells (HUVECs) have been utilized to study the directional migration of hASCs into the modified regions. Subsequently, we have highlighted the dependence of endothelial sprouting on the presence of hASCs and demonstrated the potential of photografting to control the direction of the sprouts. MPL-induced DSSA-photografting has been established as a promising method to selectively alter the microenvironment of cells.
Collapse
Affiliation(s)
- Simon Sayer
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, TU Wien, Vienna, Austria.,Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Vienna, Austria
| | - Tommaso Zandrini
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, TU Wien, Vienna, Austria.,Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Vienna, Austria
| | - Marica Markovic
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, TU Wien, Vienna, Austria.,Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Vienna, Austria
| | - Jasper Van Hoorick
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Sandra Van Vlierberghe
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Stefan Baudis
- Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Vienna, Austria.,Polymer Chemistry and Technology Group, Institute of Applied Synthetic Chemistry, TU Wien, Vienna, Austria
| | - Wolfgang Holnthoner
- Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Vienna, Austria.,Ludwig-Boltzmann-Institute for Traumatology, The Research Centre in Cooperation with AUVA, Vienna, Austria
| | - Aleksandr Ovsianikov
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, TU Wien, Vienna, Austria. .,Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Vienna, Austria.
| |
Collapse
|
24
|
ABSTRACTS (BY NUMBER). Tissue Eng Part A 2022. [DOI: 10.1089/ten.tea.2022.29025.abstracts] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
25
|
Methods for vascularization and perfusion of tissue organoids. Mamm Genome 2022; 33:437-450. [PMID: 35333952 DOI: 10.1007/s00335-022-09951-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 03/10/2022] [Indexed: 12/17/2022]
Abstract
Tissue organoids or "mini organs" can be invaluable tools for understanding health and disease biology, modeling tissue dynamics, or screening potential drug candidates. Effective vascularization of these models is critical for truly representing the in vivo tissue environment. Not only is the formation of a vascular network, and ultimately a microcirculation, essential for proper distribution and exchange of oxygen and nutrients throughout larger organoids, but vascular cells dynamically communicate with other cells to modulate overall tissue behavior. Additionally, interstitial fluid flow, mediated by a perfused microvasculature, can have profound influences on tissue biology. Thus, a truly functionally and biologically relevant organoid requires a vasculature. Here, we review existing strategies for fabricating and incorporating vascular elements and perfusion within tissue organoids.
Collapse
|
26
|
Inverting angiogenesis with interstitial flow and chemokine matrix-binding affinity. Sci Rep 2022; 12:4237. [PMID: 35273299 PMCID: PMC8913640 DOI: 10.1038/s41598-022-08186-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 02/15/2022] [Indexed: 11/30/2022] Open
Abstract
The molecular signaling pathways that orchestrate angiogenesis have been widely studied, but the role of biophysical cues has received less attention. Interstitial flow is unavoidable in vivo, and has been shown to dramatically change the neovascular patterns, but the mechanisms by which flow regulates angiogenesis remain poorly understood. Here, we study the complex interactions between interstitial flow and the affinity for matrix binding of different chemokine isoforms. Using a computational model, we find that changing the matrix affinity of the chemokine isoform can invert the effect of interstitial flow on angiogenesis—from preferential growth in the direction of the flow when the chemokine is initially matrix-bound to preferential flow against the flow when it is unbound. Although fluid forces signal endothelial cells directly, our data suggests a mechanism for the inversion based on biotransport arguments only, and offers a potential explanation for experimental results in which interstitial flow produced preferential vessel growth with and against the flow. Our results point to a particularly intricate effect of interstitial flow on angiogenesis in the tumor microenvironment, where the vessel network geometry and the interstitial flow patterns are complex.
Collapse
|
27
|
Seymour AJ, Westerfield AD, Cornelius VC, Skylar-Scott MA, Heilshorn SC. Bioprinted microvasculature: progressing from structure to function. Biofabrication 2022; 14:10.1088/1758-5090/ac4fb5. [PMID: 35086069 PMCID: PMC8988885 DOI: 10.1088/1758-5090/ac4fb5] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/27/2022] [Indexed: 11/12/2022]
Abstract
Three-dimensional (3D) bioprinting seeks to unlock the rapid generation of complex tissue constructs, but long-standing challenges with efficientin vitromicrovascularization must be solved before this can become a reality. Microvasculature is particularly challenging to biofabricate due to the presence of a hollow lumen, a hierarchically branched network topology, and a complex signaling milieu. All of these characteristics are required for proper microvascular-and, thus, tissue-function. While several techniques have been developed to address distinct portions of this microvascularization challenge, no single approach is capable of simultaneously recreating all three microvascular characteristics. In this review, we present a three-part framework that proposes integration of existing techniques to generate mature microvascular constructs. First, extrusion-based 3D bioprinting creates a mesoscale foundation of hollow, endothelialized channels. Second, biochemical and biophysical cues induce endothelial sprouting to create a capillary-mimetic network. Third, the construct is conditioned to enhance network maturity. Across all three of these stages, we highlight the potential for extrusion-based bioprinting to become a central technique for engineering hierarchical microvasculature. We envision that the successful biofabrication of functionally engineered microvasculature will address a critical need in tissue engineering, and propel further advances in regenerative medicine andex vivohuman tissue modeling.
Collapse
Affiliation(s)
- Alexis J. Seymour
- Department of Bioengineering, Stanford University, 443 Via Ortega, Shriram Center Room 119, Stanford, CA 94305, USA
| | - Ashley D. Westerfield
- Department of Bioengineering, Stanford University, 443 Via Ortega, Shriram Center Room 119, Stanford, CA 94305, USA
| | - Vincent C. Cornelius
- Department of Bioengineering, Stanford University, 443 Via Ortega, Shriram Center Room 119, Stanford, CA 94305, USA
| | - Mark A. Skylar-Scott
- Department of Bioengineering, Stanford University, 443 Via Ortega, Shriram Center Room 119, Stanford, CA 94305, USA
| | - Sarah C. Heilshorn
- Department of Materials Science & Engineering, Stanford University, 476 Lomita Mall, McCullough Room 246, Stanford, CA 94305, USA
| |
Collapse
|
28
|
Mykuliak A, Yrjänäinen A, Mäki AJ, Gebraad A, Lampela E, Kääriäinen M, Pakarinen TK, Kallio P, Miettinen S, Vuorenpää H. Vasculogenic Potency of Bone Marrow- and Adipose Tissue-Derived Mesenchymal Stem/Stromal Cells Results in Differing Vascular Network Phenotypes in a Microfluidic Chip. Front Bioeng Biotechnol 2022; 10:764237. [PMID: 35211462 PMCID: PMC8861308 DOI: 10.3389/fbioe.2022.764237] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/11/2022] [Indexed: 12/27/2022] Open
Abstract
The vasculature is an essential, physiological element in virtually all human tissues. Formation of perfusable vasculature is therefore crucial for reliable tissue modeling. Three-dimensional vascular networks can be formed through the co-culture of endothelial cells (ECs) with stromal cells embedded in hydrogel. Mesenchymal stem/stromal cells (MSCs) derived from bone marrow (BMSCs) and adipose tissue (ASCs) are an attractive choice as stromal cells due to their natural perivascular localization and ability to support formation of mature and stable microvessels in vitro. So far, BMSCs and ASCs have been compared as vasculature-supporting cells in static cultures. In this study, BMSCs and ASCs were co-cultured with endothelial cells in a fibrin hydrogel in a perfusable microfluidic chip. We demonstrated that using MSCs of different origin resulted in vascular networks with distinct phenotypes. Both types of MSCs supported formation of mature and interconnected microvascular networks-on-a-chip. However, BMSCs induced formation of fully perfusable microvasculature with larger vessel area and length whereas ASCs resulted in partially perfusable microvascular networks. Immunostainings revealed that BMSCs outperformed ASCs in pericytic characteristics. Moreover, co-culture with BMSCs resulted in significantly higher expression levels of endothelial and pericyte-specific genes, as well as genes involved in vasculature maturation. Overall, our study provides valuable knowledge on the properties of MSCs as vasculature-supporting cells and highlights the importance of choosing the application-specific stromal cell source for vascularized organotypic models.
Collapse
Affiliation(s)
- Anastasiia Mykuliak
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Alma Yrjänäinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Antti-Juhana Mäki
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Arjen Gebraad
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Ella Lampela
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Minna Kääriäinen
- Department of Plastic and Reconstructive Surgery, Tampere University Hospital, Tampere, Finland
| | | | - Pasi Kallio
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Susanna Miettinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Hanna Vuorenpää
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
29
|
Angiogenesis and Functional Vessel Formation Induced by Interstitial Flow and Vascular Endothelial Growth Factor Using a Microfluidic Chip. MICROMACHINES 2022; 13:mi13020225. [PMID: 35208349 PMCID: PMC8876009 DOI: 10.3390/mi13020225] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 02/07/2023]
Abstract
Angiogenesis occurs during both physiological and pathological processes. In this study, a microfluidic chip for the development of angiogenesis was utilized to assess angiogenic sprouting and functional vessel formation. We also found that vascular endothelial growth factor (VEGF) was a determinant of the initiation of vascular sprouts, while the direction of these sprouts was greatly influenced by interstitial flow. Isoforms of VEGF such as VEGF121, VEGF165, and VEGF189 displayed different angiogenic properties on the chip as assessed by sprout length and number, vessel perfusion, and connectivity. VEGF165 had the highest capacity to induce vascular sprouting among the three isoforms assessed and furthermore, also induced functional vessel formation. This chip could be used to analyze the effect of different angiogenic factors and drugs, as well as to explore the mechanism of angiogenesis induced by such factors.
Collapse
|
30
|
Miller B, Sewell-Loftin MK. Mechanoregulation of Vascular Endothelial Growth Factor Receptor 2 in Angiogenesis. Front Cardiovasc Med 2022; 8:804934. [PMID: 35087885 PMCID: PMC8787114 DOI: 10.3389/fcvm.2021.804934] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/10/2021] [Indexed: 12/17/2022] Open
Abstract
The endothelial cells that compose the vascular system in the body display a wide range of mechanotransductive behaviors and responses to biomechanical stimuli, which act in concert to control overall blood vessel structure and function. Such mechanosensitive activities allow blood vessels to constrict, dilate, grow, or remodel as needed during development as well as normal physiological functions, and the same processes can be dysregulated in various disease states. Mechanotransduction represents cellular responses to mechanical forces, translating such factors into chemical or electrical signals which alter the activation of various cell signaling pathways. Understanding how biomechanical forces drive vascular growth in healthy and diseased tissues could create new therapeutic strategies that would either enhance or halt these processes to assist with treatments of different diseases. In the cardiovascular system, new blood vessel formation from preexisting vasculature, in a process known as angiogenesis, is driven by vascular endothelial growth factor (VEGF) binding to VEGF receptor 2 (VEGFR-2) which promotes blood vessel development. However, physical forces such as shear stress, matrix stiffness, and interstitial flow are also major drivers and effectors of angiogenesis, and new research suggests that mechanical forces may regulate VEGFR-2 phosphorylation. In fact, VEGFR-2 activation has been linked to known mechanobiological agents including ERK/MAPK, c-Src, Rho/ROCK, and YAP/TAZ. In vascular disease states, endothelial cells can be subjected to altered mechanical stimuli which affect the pathways that control angiogenesis. Both normalizing and arresting angiogenesis associated with tumor growth have been strategies for anti-cancer treatments. In the field of regenerative medicine, harnessing biomechanical regulation of angiogenesis could enhance vascularization strategies for treating a variety of cardiovascular diseases, including ischemia or permit development of novel tissue engineering scaffolds. This review will focus on the impact of VEGFR-2 mechanosignaling in endothelial cells (ECs) and its interaction with other mechanotransductive pathways, as well as presenting a discussion on the relationship between VEGFR-2 activation and biomechanical forces in the extracellular matrix (ECM) that can help treat diseases with dysfunctional vascular growth.
Collapse
Affiliation(s)
- Bronte Miller
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mary Kathryn Sewell-Loftin
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
31
|
Zhang X, Karim M, Hasan MM, Hooper J, Wahab R, Roy S, Al-Hilal TA. Cancer-on-a-Chip: Models for Studying Metastasis. Cancers (Basel) 2022; 14:648. [PMID: 35158914 PMCID: PMC8833392 DOI: 10.3390/cancers14030648] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
The microfluidic-based cancer-on-a-chip models work as a powerful tool to study the tumor microenvironment and its role in metastasis. The models recapitulate and systematically simplify the in vitro tumor microenvironment. This enables the study of a metastatic process in unprecedented detail. This review examines the development of cancer-on-a-chip microfluidic platforms at the invasion/intravasation, extravasation, and angiogenesis steps over the last three years. The on-chip modeling of mechanical cues involved in the metastasis cascade are also discussed. Finally, the popular design of microfluidic chip models for each step are discussed along with the challenges and perspectives of cancer-on-a-chip models.
Collapse
Affiliation(s)
- Xiaojun Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79968, USA; (X.Z.); (M.K.); (M.M.H.); (R.W.)
- Department of Biological Sciences, College of Science, University of Texas at El Paso, El Paso, TX 79968, USA; (J.H.); (S.R.)
| | - Mazharul Karim
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79968, USA; (X.Z.); (M.K.); (M.M.H.); (R.W.)
- Department of Environmental Science & Engineering, College of Science, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Md Mahedi Hasan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79968, USA; (X.Z.); (M.K.); (M.M.H.); (R.W.)
- Department of Environmental Science & Engineering, College of Science, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Jacob Hooper
- Department of Biological Sciences, College of Science, University of Texas at El Paso, El Paso, TX 79968, USA; (J.H.); (S.R.)
| | - Riajul Wahab
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79968, USA; (X.Z.); (M.K.); (M.M.H.); (R.W.)
| | - Sourav Roy
- Department of Biological Sciences, College of Science, University of Texas at El Paso, El Paso, TX 79968, USA; (J.H.); (S.R.)
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Taslim A. Al-Hilal
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79968, USA; (X.Z.); (M.K.); (M.M.H.); (R.W.)
- Department of Biological Sciences, College of Science, University of Texas at El Paso, El Paso, TX 79968, USA; (J.H.); (S.R.)
- Department of Environmental Science & Engineering, College of Science, University of Texas at El Paso, El Paso, TX 79968, USA
| |
Collapse
|
32
|
Lee GH, Huang SA, Aw WY, Rathod M, Cho C, Ligler FS, Polacheck WJ. Multilayer microfluidic platform for the study of luminal, transmural, and interstitial flow. Biofabrication 2022; 14:10.1088/1758-5090/ac48e5. [PMID: 34991082 PMCID: PMC8867496 DOI: 10.1088/1758-5090/ac48e5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 01/06/2022] [Indexed: 01/27/2023]
Abstract
Efficient delivery of oxygen and nutrients to tissues requires an intricate balance of blood, lymphatic, and interstitial fluid pressures (IFPs), and gradients in fluid pressure drive the flow of blood, lymph, and interstitial fluid through tissues. While specific fluid mechanical stimuli, such as wall shear stress, have been shown to modulate cellular signaling pathways along with gene and protein expression patterns, an understanding of the key signals imparted by flowing fluid and how these signals are integrated across multiple cells and cell types in native tissues is incomplete due to limitations with current assays. Here, we introduce a multi-layer microfluidic platform (MμLTI-Flow) that enables the culture of engineered blood and lymphatic microvessels and independent control of blood, lymphatic, and IFPs. Using optical microscopy methods to measure fluid velocity for applied input pressures, we demonstrate varying rates of interstitial fluid flow as a function of blood, lymphatic, and interstitial pressure, consistent with computational fluid dynamics (CFD) models. The resulting microfluidic and computational platforms will provide for analysis of key fluid mechanical parameters and cellular mechanisms that contribute to diseases in which fluid imbalances play a role in progression, including lymphedema and solid cancer.
Collapse
Affiliation(s)
- Gi-hun Lee
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University
| | - Stephanie A. Huang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University
| | - Wen Y. Aw
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University
| | - Mitesh Rathod
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University
| | - Crescentia Cho
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University
| | - Frances S. Ligler
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University
| | - William J. Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University,Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill,McAllister Heart Institute, University of North Carolina at Chapel Hill
| |
Collapse
|
33
|
Winkelman MA, Kim DY, Kakarla S, Grath A, Silvia N, Dai G. Interstitial flow enhances the formation, connectivity, and function of 3D brain microvascular networks generated within a microfluidic device. LAB ON A CHIP 2021; 22:170-192. [PMID: 34881385 PMCID: PMC9257897 DOI: 10.1039/d1lc00605c] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The bulk flow of interstitial fluid through tissue is an important factor in human biology, including the development of brain microvascular networks (MVNs) with the blood-brain barrier (BBB). Bioengineering perfused, functional brain MVNs has great potential for modeling neurovascular diseases and drug delivery. However, most in vitro models of brain MVNs do not implement interstitial flow during the generation of microvessels. Using a microfluidic device (MFD), we cultured primary human brain endothelial cells (BECs), pericytes, and astrocytes within a 3D fibrin matrix with (flow) and without (static) interstitial flow. We found that the bulk flow of interstitial fluid was beneficial for both BEC angiogenesis and vasculogenesis. Brain MVNs cultured under flow conditions achieved anastomosis and were perfusable, whereas those under static conditions lacked connectivity and the ability to be perfused. Compared to static culture, microvessels developed in flow culture exhibited an enhanced vessel area, branch length and diameter, connectivity, and longevity. Although there was no change in pericyte coverage of microvessels, a slight increase in astrocyte coverage was observed under flow conditions. In addition, the immunofluorescence intensity of basal lamina proteins, collagen IV and laminin, was nearly doubled in flow culture. Lastly, the barrier function of brain microvessels was enhanced under flow conditions, as demonstrated by decreased dextran permeability. Taken together, these results highlighted the importance of interstitial flow in the in vitro generation of perfused brain MVNs with characteristics similar to those of the human BBB.
Collapse
Affiliation(s)
- Max A Winkelman
- Department of Bioengineering, Northeastern University, 805 Columbus Ave, ISEC 224, Boston, MA, 02115, USA.
| | - Diana Y Kim
- Department of Bioengineering, Northeastern University, 805 Columbus Ave, ISEC 224, Boston, MA, 02115, USA.
| | - Shravani Kakarla
- Department of Bioengineering, Northeastern University, 805 Columbus Ave, ISEC 224, Boston, MA, 02115, USA.
| | - Alexander Grath
- Department of Bioengineering, Northeastern University, 805 Columbus Ave, ISEC 224, Boston, MA, 02115, USA.
| | - Nathaniel Silvia
- Department of Bioengineering, Northeastern University, 805 Columbus Ave, ISEC 224, Boston, MA, 02115, USA.
| | - Guohao Dai
- Department of Bioengineering, Northeastern University, 805 Columbus Ave, ISEC 224, Boston, MA, 02115, USA.
| |
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: 28] [Impact Index Per Article: 9.3] [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
|
Dessalles CA, Ramón-Lozano C, Babataheri A, Barakat AI. Luminal flow actuation generates coupled shear and strain in a microvessel-on-chip. Biofabrication 2021; 14. [PMID: 34592728 DOI: 10.1088/1758-5090/ac2baa] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022]
Abstract
In the microvasculature, blood flow-derived forces are key regulators of vascular structure and function. Consequently, the development of hydrogel-based microvessel-on-chip systems that strive to mimic thein vivocellular organization and mechanical environment has received great attention in recent years. However, despite intensive efforts, current microvessel-on-chip systems suffer from several limitations, most notably failure to produce physiologically relevant wall strain levels. In this study, a novel microvessel-on-chip based on the templating technique and using luminal flow actuation to generate physiologically relevant levels of wall shear stress and circumferential stretch is presented. Normal forces induced by the luminal pressure compress the surrounding soft collagen hydrogel, dilate the channel, and create large circumferential strain. The fluid pressure gradient in the system drives flow forward and generates realistic pulsatile wall shear stresses. Rigorous characterization of the system reveals the crucial role played by the poroelastic behavior of the hydrogel in determining the magnitudes of the wall shear stress and strain. The experimental measurements are combined with an analytical model of flow in both the lumen and the porous hydrogel to provide an exceptionally versatile user manual for an application-based choice of parameters in microvessels-on-chip. This unique strategy of flow actuation adds a dimension to the capabilities of microvessel-on-chip systems and provides a more general framework for improving hydrogel-basedin vitroengineered platforms.
Collapse
Affiliation(s)
- Claire A Dessalles
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, 91120 Palaiseau, France
| | - Clara Ramón-Lozano
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, 91120 Palaiseau, France
| | - Avin Babataheri
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, 91120 Palaiseau, France
| | - Abdul I Barakat
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, 91120 Palaiseau, France
| |
Collapse
|
36
|
Dessalles CA, Leclech C, Castagnino A, Barakat AI. Integration of substrate- and flow-derived stresses in endothelial cell mechanobiology. Commun Biol 2021; 4:764. [PMID: 34155305 PMCID: PMC8217569 DOI: 10.1038/s42003-021-02285-w] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 06/02/2021] [Indexed: 02/05/2023] Open
Abstract
Endothelial cells (ECs) lining all blood vessels are subjected to large mechanical stresses that regulate their structure and function in health and disease. Here, we review EC responses to substrate-derived biophysical cues, namely topography, curvature, and stiffness, as well as to flow-derived stresses, notably shear stress, pressure, and tensile stresses. Because these mechanical cues in vivo are coupled and are exerted simultaneously on ECs, we also review the effects of multiple cues and describe burgeoning in vitro approaches for elucidating how ECs integrate and interpret various mechanical stimuli. We conclude by highlighting key open questions and upcoming challenges in the field of EC mechanobiology.
Collapse
Affiliation(s)
- Claire A Dessalles
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, Palaiseau, France
| | - Claire Leclech
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, Palaiseau, France
| | - Alessia Castagnino
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, Palaiseau, France
| | - Abdul I Barakat
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, Palaiseau, France.
| |
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: 4.3] [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
|
Zilberman A, Cornelison RC. Microphysiological models of the central nervous system with fluid flow. Brain Res Bull 2021; 174:72-83. [PMID: 34029679 DOI: 10.1016/j.brainresbull.2021.05.015] [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: 01/13/2021] [Revised: 05/08/2021] [Accepted: 05/17/2021] [Indexed: 12/11/2022]
Abstract
There are over 1,000 described neurological and neurodegenerative disorders affecting nearly 100 million Americans - roughly one third of the U.S. population. Collectively, treatment of neurological conditions is estimated to cost $800 billion every year. Lowering this societal burden will require developing better model systems in which to study these diverse disorders. Microphysiological systems are promising tools for modeling healthy and diseased neural tissues to study mechanisms and treatment of neuropathology. One major benefit of microphysiological systems is the ability to incorporate biophysical forces, namely the forces derived from biological fluid flow. Fluid flow in the central nervous system (CNS) is a complex but important element of physiology, and pathologies as diverse as traumatic or ischemic injury, cancer, neurodegenerative disease, and natural aging have all been found to alter flow pathways. In this review, we summarize recent advances in three-dimensional microphysiological systems for studying the biology and therapy of CNS disorders and highlight the ability and growing need to incorporate biological fluid flow in these miniaturized model systems.
Collapse
Affiliation(s)
- Aleeza Zilberman
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, United States
| | - R Chase Cornelison
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, United States.
| |
Collapse
|
39
|
Rothbauer M, Bachmann BE, Eilenberger C, Kratz SR, Spitz S, Höll G, Ertl P. A Decade of Organs-on-a-Chip Emulating Human Physiology at the Microscale: A Critical Status Report on Progress in Toxicology and Pharmacology. MICROMACHINES 2021; 12:470. [PMID: 33919242 PMCID: PMC8143089 DOI: 10.3390/mi12050470] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 12/22/2022]
Abstract
Organ-on-a-chip technology has the potential to accelerate pharmaceutical drug development, improve the clinical translation of basic research, and provide personalized intervention strategies. In the last decade, big pharma has engaged in many academic research cooperations to develop organ-on-a-chip systems for future drug discoveries. Although most organ-on-a-chip systems present proof-of-concept studies, miniaturized organ systems still need to demonstrate translational relevance and predictive power in clinical and pharmaceutical settings. This review explores whether microfluidic technology succeeded in paving the way for developing physiologically relevant human in vitro models for pharmacology and toxicology in biomedical research within the last decade. Individual organ-on-a-chip systems are discussed, focusing on relevant applications and highlighting their ability to tackle current challenges in pharmacological research.
Collapse
Affiliation(s)
- Mario Rothbauer
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria; (B.E.M.B.); (C.E.); (S.R.A.K.); (S.S.); (G.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Karl Chiari Lab for Orthopaedic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Währinger Gürtel 18-22, 1090 Vienna, Austria
| | - Barbara E.M. Bachmann
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria; (B.E.M.B.); (C.E.); (S.R.A.K.); (S.S.); (G.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Allgemeine Unfallversicherungsanstalt (AUVA) Research Centre, Donaueschingenstraße 13, 1200 Vienna, Austria
| | - Christoph Eilenberger
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria; (B.E.M.B.); (C.E.); (S.R.A.K.); (S.S.); (G.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Sebastian R.A. Kratz
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria; (B.E.M.B.); (C.E.); (S.R.A.K.); (S.S.); (G.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Drug Delivery and 3R-Models Group, Buchmann Institute for Molecular Life Sciences & Institute for Pharmaceutical Technology, Goethe University Frankfurt Am Main, 60438 Frankfurt, Germany
| | - Sarah Spitz
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria; (B.E.M.B.); (C.E.); (S.R.A.K.); (S.S.); (G.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Gregor Höll
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria; (B.E.M.B.); (C.E.); (S.R.A.K.); (S.S.); (G.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Peter Ertl
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria; (B.E.M.B.); (C.E.); (S.R.A.K.); (S.S.); (G.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
40
|
Tronolone JJ, Jain A. Engineering new microvascular networks on-chip: ingredients, assembly, and best practices. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2007199. [PMID: 33994903 PMCID: PMC8114943 DOI: 10.1002/adfm.202007199] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Indexed: 05/23/2023]
Abstract
Tissue engineered grafts show great potential as regenerative implants for diseased or injured tissues within the human body. However, these grafts suffer from poor nutrient perfusion and waste transport, thus decreasing their viability post-transplantation. Graft vascularization is therefore a major area of focus within tissue engineering because biologically relevant conduits for nutrient and oxygen perfusion can improve viability post-implantation. Many researchers utilize microphysiological systems as testing platforms for potential grafts due to an ability to integrate vascular networks as well as biological characteristics such as fluid perfusion, 3D architecture, compartmentalization of tissue-specific materials, and biophysical and biochemical cues. While many methods of vascularizing these systems exist, microvascular self-assembly has great potential for bench-to-clinic translation as it relies on naturally occurring physiological events. In this review, we highlight the past decade of literature and critically discuss the most important and tunable components yielding a self-assembled vascular network on chip: endothelial cell source, tissue-specific supporting cells, biomaterial scaffolds, biochemical cues, and biophysical forces. This article discusses the bioengineered systems of angiogenesis, vasculogenesis, and lymphangiogenesis, and includes a brief overview of multicellular systems. We conclude with future avenues of research to guide the next generation of vascularized microfluidic models and future tissue engineered grafts.
Collapse
Affiliation(s)
- James J Tronolone
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Abhishek Jain
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77808, USA
| |
Collapse
|
41
|
Brassard-Jollive N, Monnot C, Muller L, Germain S. In vitro 3D Systems to Model Tumor Angiogenesis and Interactions With Stromal Cells. Front Cell Dev Biol 2020; 8:594903. [PMID: 33224956 PMCID: PMC7674638 DOI: 10.3389/fcell.2020.594903] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/05/2020] [Indexed: 12/15/2022] Open
Abstract
In vitro 3D culture systems provide promising tools for screening novel therapies and understanding drug resistance mechanisms in cancer because they are adapted for high throughput analysis. One of the main current challenges is to reproducibly culture patient samples containing cancer and stromal cells to faithfully recapitulate tumor microenvironment and move toward efficient personalized medicine. Tumors are composed of heterogeneous cell populations and characterized by chaotic vascularization in a remodeled microenvironment. Indeed, tumor angiogenesis occurs in a complex stroma containing immune cells and cancer-associated fibroblasts that secrete important amounts of cytokines, growth factors, extracellular vesicles, and extracellular matrix (ECM). This process leads to the formation of inflated, tortuous, and permeable capillaries that display deficient basement membrane (BM) and perivascular coverage. These abnormal capillaries affect responses to anti-cancer therapies such as anti-angiogenic, radio-, and immunotherapies. Current pre-clinical models are limited for investigating interactions between tumor cells and vascularization during tumor progression as well as mechanisms that lead to drug resistance. In vitro approaches developed for vascularization are either the result of engineered cell lining or based on physiological processes including vasculogenesis and sprouting angiogenesis. They allow investigation of paracrine and direct interactions between endothelial and tumor and/or stromal cells, as well as impact of biochemical and biophysical cues of the microenvironment, using either natural matrix components or functionalized synthetic hydrogels. In addition, microfluidic devices provide access to modeling the impact of shear stress and interstitial flow and growth factor gradients. In this review, we will describe the state of the art co-culture models of vascularized micro-tumors in order to study tumor progression and metastatic dissemination including intravasation and/or extravasation processes.
Collapse
Affiliation(s)
- Noémie Brassard-Jollive
- Center for Interdisciplinary Research in Biology, College de France, CNRS UMR 7241, INSERM U1050, PSL Research University, Paris, France.,Sorbonne Université, Collège Doctoral, Paris, France
| | - Catherine Monnot
- Center for Interdisciplinary Research in Biology, College de France, CNRS UMR 7241, INSERM U1050, PSL Research University, Paris, France
| | - Laurent Muller
- Center for Interdisciplinary Research in Biology, College de France, CNRS UMR 7241, INSERM U1050, PSL Research University, Paris, France
| | - Stéphane Germain
- Center for Interdisciplinary Research in Biology, College de France, CNRS UMR 7241, INSERM U1050, PSL Research University, Paris, France
| |
Collapse
|
42
|
Douglas SA, Haase K, Kamm RD, Platt MO. Cysteine cathepsins are altered by flow within an engineered in vitro microvascular niche. APL Bioeng 2020; 4:046102. [PMID: 33195960 PMCID: PMC7644274 DOI: 10.1063/5.0023342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/16/2020] [Indexed: 12/31/2022] Open
Abstract
Throughout the process of vascular growth and remodeling, the extracellular matrix (ECM) concurrently undergoes significant changes due to proteolytic activity—regulated by both endothelial and surrounding stromal cells. The role of matrix metalloproteinases has been well-studied in the context of vascular remodeling, but other proteases, such as cysteine cathepsins, could also facilitate ECM remodeling. To investigate cathepsin-mediated proteolysis in vascular ECM remodeling, and to understand the role of shear flow in this process, in vitro microvessels were cultured in previously designed microfluidic chips and assessed by immunostaining, zymography, and western blotting. Primary human vessels (HUVECs and fibroblasts) were conditioned by continuous fluid flow and/or small molecule inhibitors to probe cathepsin expression and activity. Luminal flow (in contrast to static culture) decreases the activity of cathepsins in microvessel systems, despite a total protein increase, due to a concurrent increase in the endogenous inhibitor cystatin C. Observations also demonstrate that cathepsins mostly co-localize with fibroblasts, and that fibrin (the hydrogel substrate) may stabilize cathepsin activity in the system. Inhibitor studies suggest that control over cathepsin-mediated ECM remodeling could contribute to improved maintenance of in vitro microvascular networks; however, further investigation is required. Understanding the role of cathepsin activity in in vitro microvessels and other engineered tissues will be important for future regenerative medicine applications.
Collapse
Affiliation(s)
- Simone A Douglas
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, USA
| | | | - Roger D Kamm
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Manu O Platt
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, USA
| |
Collapse
|
43
|
Sewell-Loftin MK, Katz JB, George SC, Longmore GD. Micro-strains in the extracellular matrix induce angiogenesis. LAB ON A CHIP 2020; 20:2776-2787. [PMID: 32614340 PMCID: PMC7659465 DOI: 10.1039/d0lc00145g] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
An improved understanding of biomechanical factors that control tumor development, including angiogenesis, could explain why few of the promising treatment strategies discovered via in vitro models translate well into in vivo or clinical studies. The ability to manipulate and in real-time study the multiple independent biomechanical properties on cellular activity has been limited, primarily due to limitations in traditional in vitro platforms or the inability to manipulate such factors in vivo. We present a novel microfluidic platform that mimics the vascularized tumor microenvironment with independent control of interstitial flow and mechanical strain. The microtissue platform design isolates mechanically-stimulated angiogenesis in the tumor microenvironment, by manipulating interstitial flow to eliminate soluble factors that could drive blood vessel growth. Our studies demonstrate that enhanced mechanical strain induced by cancer-associated fibroblasts (CAFs) promotes angiogenesis in microvasculature models, even when preventing diffusion of soluble factors to the growing vasculature. Moreover, small but significant decreases in micro-strains induced by inhibited CAFs were sufficient to reduce angiogenesis. Ultimately, we believe this platform represents a significant advancement in the ability to investigate biomechanical signals while controlling for biochemical signals, with a potential to be utilized in fields beyond cancer research.
Collapse
Affiliation(s)
- Mary Kathryn Sewell-Loftin
- Department of Biomedical Engineering, Wallace Tumor Institute, University of Alabama at Birmingham, 1824 6th Avenue South, Room 630A, Birmingham, AL 35294, USA.
| | | | | | | |
Collapse
|
44
|
Sano H, Watanabe M, Yamashita T, Tanishita K, Sudo R. Control of vessel diameters mediated by flow-induced outward vascular remodeling in vitro. Biofabrication 2020; 12:045008. [DOI: 10.1088/1758-5090/ab9316] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
45
|
Hickman JJ, Huh D, Kamm RD. Microphysiological systems. APL Bioeng 2019; 3:040401. [PMID: 31673671 PMCID: PMC6819166 DOI: 10.1063/1.5130170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022] Open
Affiliation(s)
- James J Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, Florida 32826, USA
| | - Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Roger D Kamm
- Department of Biological Engineering and Department of Mechanical Engineering, Massachusetts Institute of Technology, 500 Technology Square, MIT Building, Room NE47-321, Cambridge, Massachusetts 02139, USA
| |
Collapse
|