Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Lab Chip ; 23(18): 3906-3935, 2023 09 13.
Article in English | MEDLINE | ID: mdl-37592893

ABSTRACT

Over the past 15 years, the field of oncology research has witnessed significant progress in the development of new cell culture models, such as tumor-on-chip (ToC) systems. In this comprehensive overview, we present a multidisciplinary perspective by bringing together physicists, biologists, clinicians, and experts from pharmaceutical companies to highlight the current state of ToC research, its unique features, and the challenges it faces. To offer readers a clear and quantitative understanding of the ToC field, we conducted an extensive systematic analysis of more than 300 publications related to ToC from 2005 to 2022. ToC offer key advantages over other in vitro models by enabling precise control over various parameters. These parameters include the properties of the extracellular matrix, mechanical forces exerted on cells, the physico-chemical environment, cell composition, and the architecture of the tumor microenvironment. Such fine control allows ToC to closely replicate the complex microenvironment and interactions within tumors, facilitating the study of cancer progression and therapeutic responses in a highly representative manner. Importantly, by incorporating patient-derived cells or tumor xenografts, ToC models have demonstrated promising results in terms of clinical validation. We also examined the potential of ToC for pharmaceutical industries in which ToC adoption is expected to occur gradually. Looking ahead, given the high failure rate of clinical trials and the increasing emphasis on the 3Rs principles (replacement, reduction, refinement of animal experimentation), ToC models hold immense potential for cancer research. In the next decade, data generated from ToC models could potentially be employed for discovering new therapeutic targets, contributing to regulatory purposes, refining preclinical drug testing and reducing reliance on animal models.


Subject(s)
Cell Culture Techniques , Neoplasms , Humans , Animals , Drug Industry , Extracellular Matrix , Tumor Microenvironment , Neoplasms/drug therapy
2.
Microsyst Nanoeng ; 7: 4, 2021.
Article in English | MEDLINE | ID: mdl-33456784

ABSTRACT

The vascular network of the circulatory system plays a vital role in maintaining homeostasis in the human body. In this paper, a novel modular microfluidic system with a vertical two-layered configuration is developed to generate large-scale perfused microvascular networks in vitro. The two-layer polydimethylsiloxane (PDMS) configuration allows the tissue chambers and medium channels not only to be designed and fabricated independently but also to be aligned and bonded accordingly. This method can produce a modular microfluidic system that has high flexibility and scalability to design an integrated platform with multiple perfused vascularized tissues with high densities. The medium channel was designed with a rhombic shape and fabricated to be semiclosed to form a capillary burst valve in the vertical direction, serving as the interface between the medium channels and tissue chambers. Angiogenesis and anastomosis at the vertical interface were successfully achieved by using different combinations of tissue chambers and medium channels. Various large-scale microvascular networks were generated and quantified in terms of vessel length and density. Minimal leakage of the perfused 70-kDa FITC-dextran confirmed the lumenization of the microvascular networks and the formation of tight vertical interconnections between the microvascular networks and medium channels in different structural layers. This platform enables the culturing of interconnected, large-scale perfused vascularized tissue networks with high density and scalability for a wide range of multiorgan-on-a-chip applications, including basic biological studies and drug screening.

3.
Nat Commun ; 11(1): 5400, 2020 10 26.
Article in English | MEDLINE | ID: mdl-33106502

ABSTRACT

Slug (SNAI2), a member of the well-conserved Snail family of transcription factors, has multiple developmental roles, including in epithelial-to-mesenchymal transition (EMT). Here, we show that Slug is critical for the pathological angiogenesis needed to sustain tumor growth, and transiently necessary for normal developmental angiogenesis. We find that Slug upregulation in angiogenic endothelial cells (EC) regulates an EMT-like suite of target genes, and suppresses Dll4-Notch signaling thereby promoting VEGFR2 expression. Both EC-specific Slug re-expression and reduced Notch signaling, either by γ-secretase inhibition or loss of Dll4, rescue retinal angiogenesis in SlugKO mice. Conversely, inhibition of VEGF signaling prevents excessive angiogenic sprouting of Slug overexpressing EC. Finally, endothelial Slug (but not Snail) is activated by the pro-angiogenic factor SDF1α via its canonical receptor CXCR4 and the MAP kinase ERK5. Altogether, our data support a critical role for Slug in determining the angiogenic response during development and disease.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Calcium-Binding Proteins/metabolism , Endothelial Cells/metabolism , Neovascularization, Pathologic/metabolism , Snail Family Transcription Factors/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Calcium-Binding Proteins/genetics , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Snail Family Transcription Factors/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
4.
Toxicology ; 445: 152601, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32980478

ABSTRACT

Angiogenesis is a complex process that is required for development and tissue regeneration and it may be affected by many pathological conditions. Chemicals and drugs can impact formation and maintenance of the vascular networks; these effects may be both desirable (e.g., anti-cancer drugs) or unwanted (e.g., side effects of drugs). A number of in vivo and in vitro models exist for studies of angiogenesis and endothelial cell function, including organ-on-a-chip microphysiological systems. An arrayed organ-on-a-chip platform on a 96-well plate footprint that incorporates perfused microvessels, with and without tumors, was recently developed and it was shown that survival of the surrounding tissue was dependent on delivery of nutrients through the vessels. Here we describe a technology transfer of this complex microphysiological model between laboratories and demonstrate that reproducibility and robustness of these tissue chip-enabled experiments depend primarily on the source of the endothelial cells. The model was highly reproducible between laboratories and was used to demonstrate the advantages of the perfusable vascular networks for drug safety evaluation. As a proof-of-concept, we tested Fluorouracil (1-1,000 µM), Vincristine (1-1,000 nM), and Sorafenib (0.1-100 µM), in the perfusable and non-perfusable micro-organs, and in a colon cancer-containing micro-tumor model. Tissue chip experiments were compared to the traditional monolayer cultures of endothelial or tumor cells. These studies showed that human in vitro vascularized micro-organ and micro-tumor models are reproducible organ-on-a-chip platforms for studies of anticancer drugs. The data from the 3D models confirmed advantages of the physiological environment as compared to 2D cell cultures. We demonstrated how these models can be translated into practice by verifying that the endothelial cell source and passage are critical elements for establishing a perfusable model.


Subject(s)
Antineoplastic Agents/therapeutic use , Human Umbilical Vein Endothelial Cells/drug effects , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/methods , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Antineoplastic Agents/pharmacology , Cell Culture Techniques , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/pathology , HCT116 Cells , Human Umbilical Vein Endothelial Cells/physiology , Humans , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Organ Culture Techniques , Reproducibility of Results
5.
Stem Cell Reports ; 12(3): 451-460, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30745035

ABSTRACT

In the CNS, perivascular cells ("pericytes") associate with endothelial cells to mediate the formation of tight junctions essential to the function of the blood-brain barrier (BBB). The BBB protects the CNS by regulating the flow of nutrients and toxins into and out of the brain. BBB dysfunction has been implicated in the progression of Alzheimer's disease (AD), but the role of pericytes in BBB dysfunction in AD is not well understood. In the developing embryo, CNS pericytes originate from two sources: mesoderm and neural crest. In this study, we report two protocols using mesoderm or neural crest intermediates, to generate brain-specific pericyte-like cells from induced pluripotent stem cell (iPSC) lines created from healthy and AD patients. iPSC-derived pericytes display stable expression of pericyte surface markers and brain-specific genes and are functionally capable of increasing vascular tube formation and endothelial barrier properties.


Subject(s)
Blood-Brain Barrier/physiology , Induced Pluripotent Stem Cells/physiology , Mesoderm/physiology , Neural Crest/physiology , Pericytes/physiology , Pluripotent Stem Cells/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Biomarkers/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/metabolism , Brain/pathology , Brain/physiology , Humans , Mesoderm/metabolism , Mesoderm/pathology , Neural Crest/metabolism , Neural Crest/pathology , Pericytes/metabolism , Pericytes/pathology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology , Tight Junctions/metabolism , Tight Junctions/pathology , Tight Junctions/physiology
6.
Lab Chip ; 18(15): 2167-2177, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29931005

ABSTRACT

Autonomous and self-powered micropumps are in critical demand for versatile cell- and tissue-based applications as well as for low-cost point-of-care testing (POCT) in microfluidics fields. The hydrostatic pressure-driven passive micropumps are simple and widely used, but they cannot maintain steady and continuous flow for long periods of time. Here, we propose a hydrostatic pressure-driven passive micropump enhanced with siphon-based autofill function, which can realize the autonomous and continuous perfusion with well-controlled steady flow over an extended time without electric power consumption. The characterization results reveal that both the cycle number in one refilling loop and the siphon diameter will affect the refilling time. Furthermore, this micropump also enables multiplexed medium delivery under either the same or different flow conditions with high flexibility. The system was validated using an in vitro vasculogenesis model over the course of several days. Most importantly, the device can consistently provide steady medium perfusion for up to 5 days at a predefined hydrostatic pressure drop without the need for supplemental medium changes. We believe that this hydrostatic pressure-driven passive micropump will become a critical module for a broad range of sophisticated microfluidic operations and applications.


Subject(s)
Lab-On-A-Chip Devices , Cell Line , Equipment Design , Finite Element Analysis , Humans , Hydrostatic Pressure , Neovascularization, Physiologic
7.
Methods Mol Biol ; 1612: 325-344, 2017.
Article in English | MEDLINE | ID: mdl-28634954

ABSTRACT

This protocol describes detailed practical procedures for generating 3D intact and perfusable microvascular network that connects to microfluidic channels without appreciable leakage. This advanced 3D microvascular network model incorporates different stages of vascular development including vasculogenesis, endothelial cell (EC) lining, sprouting angiogenesis, and anastomosis in sequential order. The capillary network is first induced via vasculogenesis in a middle tissue chamber and then EC linings along the microfluidic channel on either side serve as artery and vein. The anastomosis is then induced by sprouting angiogenesis to facilitate tight interconnection between the artery/vein and the capillary network. This versatile device design and its robust construction methodology establish a physiological microcirculation transport model of interconnected perfused vessels from artery to vascularized tissue to vein.


Subject(s)
Capillaries/cytology , Endothelial Cells/cytology , Microfluidics/methods , Cells, Cultured , Humans , Lab-On-A-Chip Devices , Microfluidics/instrumentation , Models, Biological , Tissue Engineering/instrumentation , Tissue Engineering/methods
8.
Lab Chip ; 17(3): 511-520, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28092382

ABSTRACT

There is a growing awareness that complex 3-dimensional (3D) organs are not well represented by monolayers of a single cell type - the standard format for many drug screens. To address this deficiency, and with the goal of improving screens so that drugs with good efficacy and low toxicity can be identified, microphysiological systems (MPS) are being developed that better capture the complexity of in vivo physiology. We have previously described an organ-on-a-chip platform that incorporates perfused microvessels, such that survival of the surrounding tissue is entirely dependent on delivery of nutrients through the vessels. Here we describe an arrayed version of the platform that incorporates multiple vascularized micro-organs (VMOs) on a 96-well plate. Each VMO is independently-addressable and flow through the micro-organ is driven by hydrostatic pressure. The platform is easy to use, requires no external pumps or valves, and is highly reproducible. As a proof-of-concept we have created arrayed vascularized micro tumors (VMTs) and used these in a blinded screen to assay a small library of compounds, including FDA-approved anti-cancer drugs, and successfully identified both anti-angiogenic and anti-tumor drugs. This 3D platform is suitable for efficacy/toxicity screening against multiple tissues in a more physiological environment than previously possible.


Subject(s)
Cell Culture Techniques/instrumentation , Drug Evaluation, Preclinical/instrumentation , Microfluidic Analytical Techniques/instrumentation , Tissue Array Analysis/instrumentation , Antineoplastic Agents/pharmacology , Drug Evaluation, Preclinical/methods , Equipment Design , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasms, Experimental/metabolism , Neovascularization, Pathologic/metabolism , Tissue Array Analysis/methods
9.
Acta Biomater ; 43: 122-138, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27475528

ABSTRACT

UNLABELLED: Human neural stem/progenitor cells (hNSPCs) are good candidates for treating central nervous system (CNS) trauma since they secrete beneficial trophic factors and differentiate into mature CNS cells; however, many cells die after transplantation. This cell death can be ameliorated by inclusion of a biomaterial scaffold, making identification of optimal scaffolds for hNSPCs a critical research focus. We investigated the properties of fibrin-based scaffolds and their effects on hNSPCs and found that fibrin generated from salmon fibrinogen and thrombin stimulates greater hNSPC proliferation than mammalian fibrin. Fibrin scaffolds degrade over the course of a few days in vivo, so we sought to develop a novel scaffold that would retain the beneficial properties of fibrin but degrade more slowly to provide longer support for hNSPCs. We found combination scaffolds of salmon fibrin with interpenetrating networks (IPNs) of hyaluronic acid (HA) with and without laminin polymerize more effectively than fibrin alone and generate compliant hydrogels matching the physical properties of brain tissue. Furthermore, combination scaffolds support hNSPC proliferation and differentiation while significantly attenuating the cell-mediated degradation seen with fibrin alone. HNSPCs express two fibrinogen-binding integrins, αVß1 and α5ß1, and several laminin binding integrins (α7ß1, α6ß1, α3ß1) that can mediate interaction with the scaffold. Lastly, to test the ability of scaffolds to support vascularization, we analyzed human cord blood-derived endothelial cells alone and in co-culture with hNSPCs and found enhanced vessel formation and complexity in co-cultures within combination scaffolds. Overall, combination scaffolds of fibrin, HA, and laminin are excellent biomaterials for hNSPCs. STATEMENT OF SIGNIFICANCE: Interest has increased recently in the development of biomaterials as neural stem cell transplantation scaffolds to treat central nervous system (CNS) injury since scaffolds improve survival and integration of transplanted cells. We report here on a novel combination scaffold composed of fibrin, hyaluronic acid, and laminin to support human neural stem/progenitor cell (hNSPC) function. This combined biomaterial scaffold has appropriate physical properties for hNSPCs and the CNS, supports hNSPC proliferation and differentiation, and attenuates rapid cell-mediated scaffold degradation. The hNSPCs and scaffold components synergistically encourage new vessel formation from human endothelial cells. This work marks the first report of a combination scaffold supporting human neural and vascular cells to encourage vasculogenesis, and sets a benchmark for biomaterials to treat CNS injury.


Subject(s)
Blood Vessels/physiology , Fibrin/pharmacology , Hyaluronic Acid/pharmacology , Laminin/pharmacology , Neural Stem Cells/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Blood Vessels/drug effects , Cattle , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Integrins/metabolism , Neovascularization, Physiologic/drug effects , Neural Stem Cells/drug effects , Polymerization/drug effects , Salmon
10.
Sci Rep ; 6: 31589, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27549930

ABSTRACT

There is a growing interest in developing microphysiological systems that can be used to model both normal and pathological human organs in vitro. This "organs-on-chips" approach aims to capture key structural and physiological characteristics of the target tissue. Here we describe in vitro vascularized microtumors (VMTs). This "tumor-on-a-chip" platform incorporates human tumor and stromal cells that grow in a 3D extracellular matrix and that depend for survival on nutrient delivery through living, perfused microvessels. Both colorectal and breast cancer cells grow vigorously in the platform and respond to standard-of-care therapies, showing reduced growth and/or regression. Vascular-targeting agents with different mechanisms of action can also be distinguished, and we find that drugs targeting only VEGFRs (Apatinib and Vandetanib) are not effective, whereas drugs that target VEGFRs, PDGFR and Tie2 (Linifanib and Cabozantinib) do regress the vasculature. Tumors in the VMT show strong metabolic heterogeneity when imaged using NADH Fluorescent Lifetime Imaging Microscopy and, compared to their surrounding stroma, many show a higher free/bound NADH ratio consistent with their known preference for aerobic glycolysis. The VMT platform provides a unique model for studying vascularized solid tumors in vitro.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Breast Neoplasms , Colorectal Neoplasms , Microfluidic Analytical Techniques , Models, Biological , Neovascularization, Pathologic , Breast Neoplasms/blood supply , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Humans , MCF-7 Cells , Male , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology
11.
Lab Chip ; 16(5): 868-876, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26879519

ABSTRACT

Coculturing multiple cell types together in 3-dimensional (3D) cultures better mimics the in vivo microphysiological environment, and has become widely adopted in recent years with the development of organ-on-chip systems. However, a bottleneck in set-up of these devices arises as a result of the delivery of the gel into the microfluidic chip being sensitive to pressure fluctuations, making gel confinement at a specific region challenging, especially when manual operation is performed. In this paper, we present a novel design of an on-chip regulator module with pressure-releasing safety microvalves that can facilitate stable gel delivery into designated microchannel regions while maintaining well-controlled, non-bursting gel interfaces. This pressure regulator design can be integrated into different microfluidic chip designs and is compatible with a wide variety of gel injection apparatuses operated automatically or manually at different flow rates. The sensitivity and working range of this pressure regulator can be adjusted by changing the width of its pressure releasing safety microvalve design. The effectiveness of the design is validated by its incorporation into a microfluidic platform we have developed for generating 3D vascularized micro-organs (VMOs). Reproducible gel loading is demonstrated for both an automatic syringe pump and a manually-operated micropipettor. This design allows for rapid and reproducible loading of hydrogels into microfluidic devices without the risk of bursting gel-air interfaces.


Subject(s)
Endothelial Progenitor Cells/metabolism , Hydrogels/chemistry , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Cells, Cultured , Cells, Immobilized/cytology , Cells, Immobilized/metabolism , Endothelial Progenitor Cells/cytology , Humans
12.
Lab Chip ; 16(2): 282-90, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26616908

ABSTRACT

This paper reports a method for generating an intact and perfusable microvascular network that connects to microfluidic channels without appreciable leakage. This platform incorporates different stages of vascular development including vasculogenesis, endothelial cell (EC) lining, sprouting angiogenesis, and anastomosis in sequential order. After formation of a capillary network inside the tissue chamber via vasculogenesis, the adjacent microfluidic channels are lined with a monolayer of ECs, which then serve as the high-pressure input ("artery") and low pressure output ("vein") conduits. To promote a tight interconnection between the artery/vein and the capillary network, sprouting angiogenesis is induced, which promotes anastomosis of the vasculature inside the tissue chamber with the EC lining along the microfluidic channels. Flow of fluorescent microparticles confirms the perfusability of the lumenized microvascular network, and minimal leakage of 70 kDa FITC-dextran confirms physiologic tightness of the EC junctions and completeness of the interconnections between artery/vein and the capillary network. This versatile device design and its robust construction methodology establish a physiological transport model of interconnected perfused vessels from artery to vascularized tissue to vein. The system has utility in a wide range of organ-on-a-chip applications as it enables the physiological vascular interconnection of multiple on-chip tissue constructs that can serve as disease models for drug screening.


Subject(s)
Endothelial Cells/cytology , Microfluidic Analytical Techniques , Microvessels , Neovascularization, Physiologic , Tissue Engineering , Anastomosis, Surgical , Cells, Cultured , Humans , Microfluidic Analytical Techniques/instrumentation
SELECTION OF CITATIONS
SEARCH DETAIL
...