Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Biofabrication ; 16(2)2024 01 11.
Article in English | MEDLINE | ID: mdl-38128127

ABSTRACT

Insulin is an essential regulator of blood glucose homeostasis that is produced exclusively byßcells within the pancreatic islets of healthy individuals. In those affected by diabetes, immune inflammation, damage, and destruction of isletßcells leads to insulin deficiency and hyperglycemia. Current efforts to understand the mechanisms underlyingßcell damage in diabetes rely onin vitro-cultured cadaveric islets. However, isolation of these islets involves removal of crucial matrix and vasculature that supports islets in the intact pancreas. Unsurprisingly, these islets demonstrate reduced functionality over time in standard culture conditions, thereby limiting their value for understanding native islet biology. Leveraging a novel, vascularized micro-organ (VMO) approach, we have recapitulated elements of the native pancreas by incorporating isolated human islets within a three-dimensional matrix nourished by living, perfusable blood vessels. Importantly, these islets show long-term viability and maintain robust glucose-stimulated insulin responses. Furthermore, vessel-mediated delivery of immune cells to these tissues provides a model to assess islet-immune cell interactions and subsequent islet killing-key steps in type 1 diabetes pathogenesis. Together, these results establish the islet-VMO as a novel,ex vivoplatform for studying human islet biology in both health and disease.


Subject(s)
Diabetes Mellitus , Islets of Langerhans Transplantation , Islets of Langerhans , Humans , Insulin/metabolism , Diabetes Mellitus/metabolism , Glucose/metabolism
2.
bioRxiv ; 2023 Jul 14.
Article in English | MEDLINE | ID: mdl-37503139

ABSTRACT

Assessing B cell affinity to pathogen-specific antigens prior to or following exposure could facilitate the assessment of immune status. Current standard tools to assess antigen-specific B cell responses focus on equilibrium binding of the secreted antibody in serum. These methods are costly, time-consuming, and assess antibody affinity under zero-force. Recent findings indicate that force may influence BCR-antigen binding interactions and thus immune status. Here, we designed a simple laminar flow microfluidic chamber in which the antigen (hemagglutinin of influenza A) is bound to the chamber surface to assess antigen-specific BCR binding affinity of five hemagglutinin-specific hybridomas under 65- to 650-pN force range. Our results demonstrate that both increasing shear force and bound lifetime can be used to enrich antigen-specific high affinity B cells. The affinity of the membrane-bound BCR in the flow chamber correlates well with the affinity of the matched antibodies measured in solution. These findings demonstrate that a microfluidic strategy can rapidly assess BCR-antigen binding properties and identify antigen-specific high affinity B cells. This strategy has the potential to both assess functional immune status from peripheral B cells and be a cost-effective way of identifying individual B cells as antibody sources for a range of clinical applications.

3.
Lab Chip ; 23(13): 3050-3061, 2023 06 28.
Article in English | MEDLINE | ID: mdl-37278194

ABSTRACT

A feature of severe COVID-19 is the onset of an acute and intense systemic inflammatory response referred to as the "cytokine storm". The cytokine storm is characterized by high serum levels of inflammatory cytokines and the subsequent transport of inflammatory cells to damaging levels in vital organs (e.g., myocarditis). Immune trafficking and its effect on underlying tissues (e.g., myocardium) are challenging to observe at a high spatial and temporal resolution in mouse models. In this study, we created a vascularized organ-on-a-chip system to mimic cytokine storm-like conditions and tested the effectiveness of a novel multivalent selectin-targeting carbohydrate conjugate (composed of DS - dermatan sulfate and IkL - a selectin-binding peptide, termed DS-IkL) in blocking infiltration of polymorphonuclear leukocytes (PMN). Our data shows that cytokine storm-like conditions induce endothelial cells to produce additional inflammatory cytokines and facilitate infiltration of PMNs into tissue. Treatment of tissues with DS-IkL (60 µM) reduced PMN accumulation in the tissue by >50%. We then created cytokine storm-like conditions in a vascularized cardiac tissue-chip and found that PMN infiltration increases the spontaneous beating rate of the cardiac tissue, and this effect is eliminated by treatment with DS-IkL (60 µM). In summary, we demonstrate the utility of an organ-on-a-chip platform to mimic COVID-19 related cytokine storm and that blocking leukocyte infiltration with DS-IkL could be a viable strategy to mitigate associated cardiac complications.


Subject(s)
COVID-19 , Neutrophils , Mice , Animals , Cardiotoxicity , Endothelial Cells , Microphysiological Systems , Cytokines
4.
J Extracell Vesicles ; 12(4): e12323, 2023 04.
Article in English | MEDLINE | ID: mdl-37073802

ABSTRACT

Extracellular vesicles (EVs) influence a host of normal and pathophysiological processes in vivo. Compared to soluble mediators, EVs can traffic a wide range of proteins on their surface including extracellular matrix (ECM) binding proteins, and their large size (∼30-150 nm) limits diffusion. We isolated EVs from the MCF10 series-a model human cell line of breast cancer progression-and demonstrated increasing presence of laminin-binding integrins α3ß1 and α6ß1 on the EVs as the malignant potential of the MCF10 cells increased. Transport of the EVs within a microfluidic device under controlled physiological interstitial flow (0.15-0.75 µm/s) demonstrated that convection was the dominant mechanism of transport. Binding of the EVs to the ECM enhanced the spatial concentration and gradient, which was mitigated by blocking integrins α3ß1 and α6ß1. Our studies demonstrate that convection and ECM binding are the dominant mechanisms controlling EV interstitial transport and should be leveraged in nanotherapeutic design.


Subject(s)
Extracellular Vesicles , Laminin , Humans , Laminin/metabolism , Convection , Integrin alpha6beta1/metabolism , Extracellular Vesicles/metabolism , Integrin alpha3beta1/metabolism , Extracellular Matrix/metabolism
5.
J Immunol Methods ; 511: 113381, 2022 12.
Article in English | MEDLINE | ID: mdl-36341963

ABSTRACT

Although parallel plate flow chamber assays are widely performed, extraction of kinetic parameters is limited to specialized labs with mathematical expertise and customized video-microscopy tracking tools. The recent development of Trackmate has increased researcher accessibility to tracking particles in video-microscopy experiments; however, there is a lack of tools that analyze this tracking information. We report a software tool, compatible with Trackmate, that extracts Receptor Ligand Non-Equilibrium Kinetic (RLNEK) parameters from video-microscopy data. This software should be of particular interest to the community of researchers and scientists interrogating the target-specific binding and release of immune cells.


Subject(s)
Ligands
6.
Physiol Rep ; 10(18): e15407, 2022 09.
Article in English | MEDLINE | ID: mdl-36117385

ABSTRACT

Atrial fibrillation (AF) is the most common arrhythmia in the United States, affecting approximately 1 in 10 adults, and its prevalence is expected to rise as the population ages. Treatment options for AF are limited; moreover, the development of new treatments is hindered by limited (1) knowledge regarding human atrial electrophysiological endpoints (e.g., conduction velocity [CV]) and (2) accurate experimental models. Here, we measured the CV and refractory period, and subsequently calculated the conduction wavelength, in vivo (four subjects with AF and four controls), and ex vivo (atrial slices from human hearts). Then, we created an in vitro model of human atrial conduction using induced pluripotent stem (iPS) cells. This model consisted of iPS-derived human atrial cardiomyocytes plated onto a micropatterned linear 1D spiral design of Matrigel. The CV (34-41 cm/s) of the in vitro model was nearly five times faster than 2D controls (7-9 cm/s) and similar to in vivo (40-64 cm/s) and ex vivo (28-51 cm/s) measurements. Our iPS-derived in vitro model recapitulates key features of in vivo atrial conduction and may be a useful methodology to enhance our understanding of AF and model patient-specific disease.


Subject(s)
Atrial Fibrillation , Heart Conduction System , Adult , Heart Atria , Heart Rate , Humans
7.
Biomaterials ; 280: 121245, 2022 01.
Article in English | MEDLINE | ID: mdl-34810038

ABSTRACT

Bone marrow niches (endosteal and perivascular) play important roles in both normal bone marrow function and pathological processes such as cancer cell dormancy. Unraveling the mechanisms underlying these events in humans has been severely limited by models that cannot dissect dynamic events at the niche level. Utilizing microfluidic and stem cell technologies, we present a 3D in vitro model of human bone marrow that contains both the perivascular and endosteal niches, complete with dynamic, perfusable vascular networks. We demonstrate that our model can replicate in vivo bone marrow function, including maintenance and differentiation of CD34+ hematopoietic stem/progenitor cells, egress of neutrophils (CD66b+), and niche-specific responses to doxorubicin and granulocyte-colony stimulating factor. Our platform provides opportunities to accelerate current understanding of human bone marrow function and drug response with high spatial and temporal resolution.


Subject(s)
Bone Marrow , Lab-On-A-Chip Devices , Bone Marrow Cells , Bone and Bones , Cell Differentiation/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells , Humans , Stem Cell Niche
8.
Adv Drug Deliv Rev ; 175: 113798, 2021 08.
Article in English | MEDLINE | ID: mdl-34015419

ABSTRACT

Every year, cancer claims millions of lives around the globe. Unfortunately, model systems that accurately mimic human oncology - a requirement for the development of more effective therapies for these patients - remain elusive. Tumor development is an organ-specific process that involves modification of existing tissue features, recruitment of other cell types, and eventual metastasis to distant organs. Recently, tissue engineered microfluidic devices have emerged as a powerful in vitro tool to model human physiology and pathology with organ-specificity. These organ-on-chip platforms consist of cells cultured in 3D hydrogels and offer precise control over geometry, biological components, and physiochemical properties. Here, we review progress towards organ-specific microfluidic models of the primary and metastatic tumor microenvironments. Despite the field's infancy, these tumor-on-chip models have enabled discoveries about cancer immunobiology and response to therapy. Future work should focus on the development of autologous or multi-organ systems and inclusion of the immune system.


Subject(s)
Lab-On-A-Chip Devices , Neoplasm Metastasis/pathology , Neoplasms/pathology , Animals , Humans , Tissue Engineering , Tumor Microenvironment
9.
Annu Rev Biomed Eng ; 23: 141-167, 2021 07 13.
Article in English | MEDLINE | ID: mdl-33756087

ABSTRACT

Recreating human organ-level function in vitro is a rapidly evolving field that integrates tissue engineering, stem cell biology, and microfluidic technology to produce 3D organoids. A critical component of all organs is the vasculature. Herein, we discuss general strategies to create vascularized organoids, including common source materials, and survey previous work using vascularized organoids to recreate specific organ functions and simulate tumor progression. Vascularization is not only an essential component of individual organ function but also responsible for coupling the fate of all organs and their functions. While some success in coupling two or more organs together on a single platform has been demonstrated, we argue that the future of vascularized organoid technology lies in creating organoid systems complete with tissue-specific microvasculature and in coupling multiple organs through a dynamic vascular network to create systems that can respond to changing physiological conditions.


Subject(s)
Lab-On-A-Chip Devices , Organoids , Humans , Microfluidics , Stem Cells , Tissue Engineering
10.
Integr Biol (Camb) ; 12(9): 221-232, 2020 09 30.
Article in English | MEDLINE | ID: mdl-32930334

ABSTRACT

Tumor-infiltrating leukocytes, in particular macrophages, play an important role in tumor behavior and clinical outcome. The spectrum of macrophage subtypes ranges from antitumor 'M1'-type to protumor 'M2'-type macrophages. Tumor-associated macrophages (TAMs) typically display phenotypic features of both M1 and M2, and the population distribution is thought to be dynamic and evolves as the tumor progresses. However, our understanding of how TAMs impact the tumor microenvironment remains limited by the lack of appropriate 3D in vitro models that can capture cell-cell dynamics at high spatial and temporal resolution. Using our recently developed microphysiological 'tumor-on-a-chip' (TOC) device, we present here our findings on the impact of defined macrophage subsets on tumor behavior. The TOC device design contains three adjacent and connected chambers in which both the upper and lower chambers are loaded with tumor cells, whereas the central chamber contains a dynamic, perfused, living microvascular network. Introduction of human pancreatic or colorectal cancer cells together with M1-polarized macrophages significantly inhibited tumor growth and tumor-induced angiogenesis. Protein analysis and antibody-based neutralization studies confirmed that these effects were mediated through production of C-X-C motif chemokines (CXCL9), CXCL10 and CXCL11. By contrast, M2-macrophages mediated increased tumor cell migration into the vascularized chamber and did not inhibit tumor growth or angiogenesis. In fact, single-cell RNA sequencing showed that M2 macrophages further segregated endothelial cells into two distinct subsets, corresponding to static cells in vessels versus active cells involved in angiogenesis. The impact of M2 macrophages was mediated mostly by production of matrix metalloproteinase 7 and angiopoietin 2. In summary, our data demonstrate the utility of the TOC device to mechanistically probe biological questions in a 3D in vitro microenvironment.


Subject(s)
Disease Progression , Lab-On-A-Chip Devices , Macrophages/cytology , Neoplasms/pathology , Amino Acid Motifs , Animals , Cell Culture Techniques , Cell Line, Tumor , Endothelial Cells , Humans , In Vitro Techniques , Lymphocytes, Tumor-Infiltrating/cytology , Macrophage Activation , Macrophages/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neovascularization, Pathologic/pathology , Phenotype , RNA, Small Cytoplasmic/metabolism , RNA-Seq , Tumor Microenvironment , Tumor-Associated Macrophages , U937 Cells
11.
Lab Chip ; 20(16): 3036-3050, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32716448

ABSTRACT

Hypoxia, or low oxygen (O2) tension, is a central feature of important disease processes including wound healing and cancer. Subtle temporal and spatial variations (≤1% change) in the concentration of O2 can profoundly impact gene expression and cellular functions. Sodium sulfite reacts rapidly with O2 and can be used to lower the O2 concentrations in PDMS-based tissue culture systems without exposing the cell culture to the chemical reaction. By carefully considering the mass transfer and reaction kinetics of sodium sulfite and O2, we developed a flexible theoretical framework to design an experimental microfluidic system that provides fine spatial and temporal control of O2 tension. The framework packages the dimensions, fluid flow, reaction rates, concentrations, and material properties of the fluidic lines and device into dimensionless groups that facilitate scaling and design. We validated the theoretical results by experimentally measuring O2 tension throughout the experimental system using phosphorescence lifetime imaging. We then tested the system by examining the impact of hypoxia inducible factor-1α (HIF-1α) on the proliferation and migration of MDA-MB-231 breast cancer cells. Using this system, we demonstrate that mild constant hypoxia (≤4%) induces HIF-1α mediated functional changes in the tumor cells. Furthermore, slow (>12 hours), but not rapid (<1 hour), fluctuations in O2 tension impact HIF-1α mediated proliferation and migration. Our results provide a generalized framework for fine temporal and spatial control of O2 and emphasize the need to consider mild spatial and temporal changes in O2 tension as potentially important factors in disease processes such as cancer.


Subject(s)
Microfluidics , Oxygen , Cell Culture Techniques , Cell Hypoxia , Humans , Hypoxia
12.
Front Immunol ; 11: 614300, 2020.
Article in English | MEDLINE | ID: mdl-33643296

ABSTRACT

Colorectal cancer (CRC) is the third most common cancer and second leading cause of cancer-related death in the US. CRC frequently metastasizes to the liver and these patients have a particularly poor prognosis. The infiltration of immune cells into CRC tumors and liver metastases accurately predicts disease progression and patient survival. Despite the evident influence of immune cells in the CRC tumor microenvironment (TME), efforts to identify immunotherapies for CRC patients have been limited. Here, we argue that preclinical model systems that recapitulate key features of the tumor microenvironment-including tumor, stromal, and immune cells; the extracellular matrix; and the vasculature-are crucial for studies of immunity in the CRC TME and the utility of immunotherapies for CRC patients. We briefly review the discoveries, advantages, and disadvantages of current in vitro and in vivo model systems, including 2D cell culture models, 3D culture systems, murine models, and organ-on-a-chip technologies.


Subject(s)
Colorectal Neoplasms/immunology , Immunotherapy/methods , Organ Culture Techniques/methods , Tissue Engineering/methods , Tumor Microenvironment/immunology , Animals , Cells, Cultured , Disease Models, Animal , Disease Progression , Humans , Tumor Microenvironment/drug effects
13.
Tissue Eng Part C Methods ; 26(1): 44-55, 2020 01.
Article in English | MEDLINE | ID: mdl-31797733

ABSTRACT

Cancer remains a leading health threat in the United States, and cardiovascular drug toxicity is a primary cause to eliminate a drug from FDA approval. As a result, the demand to develop new anticancer drugs without cardiovascular toxicity is high. Human induced pluripotent stem (iPS) cell-derived tissue chips provide potentially a cost-effective preclinical drug testing platform, including potential avenues for personalized medicine. We have developed a three-dimensional microfluidic device that simultaneously cultures tumor cell spheroids with iPS-derived cardiomyocytes (iPS-CMs) and iPS-derived endothelial cells (iPS-EC). The iPS-derived cells include a GCaMP6 fluorescence reporter to allow real-time imaging to monitor intracellular calcium transients. The multiple-chambered tissue chip features electrodes for pacing of the cardiac tissue to assess cardiomyocyte function such as the maximum capture rate and conduction velocity. We measured the inhibition concentration (IC50) of the anticancer drugs, Doxorubicin (0.1 µM) and Oxaliplatin (4.2 µM), on the tissue chip loaded with colon cancer cells (SW620). We simultaneously evaluated the cardiotoxicity of these anticancer drugs by assessing the drug effect on the spontaneous beat frequency and conduction velocity of iPS-derived cardiac tissue. Consistent with in vivo observations, Doxorubicin reduced the spontaneous beating rate and maximum capture rate at or near the IC50 (0.04 and 0.22 µM, respectively), whereas the toxicity of Oxaliplatin was only observed at concentrations beyond the IC50 (33 and 9.9 µM, respectively). Our platform demonstrates the feasibility to simultaneously assess cardiac toxicity and antitumor effects of drugs and could be used to enhance personalized drug testing safety and efficacy. Impact statement Drug development using murine models for preclinical testing is no longer adequate nor acceptable both financially for the pharmaceutical industry as well as for generalized or personalized assessment of safety and efficacy. Innovative solutions using human cells and tissues provide exciting new opportunities. In this study, we report on the creation of a 3D microfluidic device that simultaneously cultures human tumor cell spheroids with cardiomyocytes and endothelial cells derived from the same induced pluripotent stem cell line. The platform provides the opportunity to assess efficacy of anticancer agents while simultaneously screening for potential cardiovascular toxicity in a format conducive for personalized medicine.


Subject(s)
Antineoplastic Agents/adverse effects , Cardiotoxicity/pathology , Colonic Neoplasms/pathology , Endothelial Cells/pathology , Induced Pluripotent Stem Cells/pathology , Lab-On-A-Chip Devices , Myocytes, Cardiac/pathology , Cardiotoxicity/etiology , Cell Differentiation , Colonic Neoplasms/drug therapy , Drug Evaluation, Preclinical , Endothelial Cells/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects
14.
PLoS One ; 13(12): e0209574, 2018.
Article in English | MEDLINE | ID: mdl-30571786

ABSTRACT

Microfluidic devices have been successfully used to recreate in vitro biological microenvironments, including disease states. However, one constant issue for replicating microenvironments is that atmospheric oxygen concentration (21% O2) does not mimic physiological values (often around 5% O2). We have created a microfluidic device that can control both the spatial and temporal variations in oxygen tensions that are characteristic of in vivo biology. Additionally, since the microcirculation is responsive to hypoxia, we used a 3D sprouting angiogenesis assay to confirm the biological relevance of the microfluidic platform. Our device consists of three parallel connected tissue chambers and an oxygen scavenger channel placed adjacent to these tissue chambers. Experimentally measured oxygen maps were constructed using phosphorescent lifetime imaging microscopy and compared with values from a computational model. The central chamber was loaded with endothelial and fibroblast cells to form a 3D vascular network. Four to six days later, fibroblasts were loaded into the side chambers, and a day later the oxygen scavenger (sodium sulfite) was flowed through the adjacent channel to induce a spatial and temporal oxygen gradient. Our results demonstrate that both constant chronic and intermittent hypoxia can bias vessel growth, with constant chronic hypoxia showing higher degrees of biased angiogenesis. Our simple design provides consistent control of spatial and temporal oxygen gradients in the tissue microenvironment and can be used to investigate important oxygen-dependent biological processes in conditions such as cancer and ischemic heart disease.


Subject(s)
Cell Hypoxia/physiology , Cellular Microenvironment/physiology , Microfluidic Analytical Techniques/instrumentation , Oxygen/metabolism , Cell Hypoxia/drug effects , Cell Line, Tumor , Cellular Microenvironment/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Sulfites/pharmacology
15.
Lab Chip ; 18(23): 3687-3702, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30393802

ABSTRACT

Most cancer treatment strategies target cell proliferation, angiogenesis, migration, and intravasation of tumor cells in an attempt to limit tumor growth and metastasis. An in vitro platform to assess tumor progression and drug sensitivity could provide avenues to enhance our understanding of tumor metastasis as well as precision medicine. We present a microfluidic platform that mimics biological mass transport near the arterial end of a capillary in the tumor microenvironment. A central feature is a quiescent perfused 3D microvascular network created prior to loading tumor cells or patient-derived tumor organoids in an adjacent compartment. The physiological delivery of nutrients and/or drugs to the tumor then occurs through the vascular network. We demonstrate the culture, growth, and treatment of tumor cell lines and patient-derived breast cancer organoids. The platform provides the opportunity to simultaneously and dynamically observe hallmark features of tumor progression including cell proliferation, angiogenesis, cell migration, and tumor cell intravasation. Additionally, primary breast tumor organoids are viable in the device for several weeks and induce robust sprouting angiogenesis. Finally, we demonstrate the feasibility of our platform for drug discovery and personalized medicine by analyzing the response to chemo- and anti-angiogenic therapy. Precision medicine-based cancer treatments can only be realized if individual tumors can be rapidly assessed for therapeutic sensitivity in a clinically relevant timeframe (⪅14 days). Our platform indicates that this goal can be achieved and provides compelling opportunities to advance precision medicine for cancer.


Subject(s)
Drug Screening Assays, Antitumor/instrumentation , Lab-On-A-Chip Devices , Organoids/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Equipment Design , Humans , Microvessels/drug effects , Neoplasm Invasiveness , Tumor Microenvironment/drug effects
16.
Angiogenesis ; 20(4): 493-504, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28608153

ABSTRACT

Convective transport can significantly distort spatial concentration gradients. Interstitial flow is ubiquitous throughout living tissue, but our understanding of how interstitial flow affects concentration gradients in biological processes is limited. Interstitial flow is of particular interest for angiogenesis because pathological and physiological angiogenesis is associated with altered interstitial flow, and both interstitial flow and morphogen gradients (e.g., vascular endothelial growth factor, VEGF) can potentially stimulate and guide new blood vessel growth. We designed an in vitro microfluidic platform to simulate 3D angiogenesis in a tissue microenvironment that precisely controls interstitial flow and spatial morphogen gradients. The microvascular tissue was developed from endothelial colony forming cell-derived endothelial cells extracted from cord blood and stromal fibroblasts in a fibrin extracellular matrix. Pressure in the microfluidic lines was manipulated to control the interstitial flow. A mathematical model of mass and momentum transport, and experimental studies with fluorescently labeled dextran were performed to validate the platform. Our data demonstrate that at physiological interstitial flow (0.1-10 µm/s), morphogen gradients were eliminated within hours, and angiogenesis demonstrated a striking bias in the opposite direction of interstitial flow. The interstitial flow-directed angiogenesis was dependent on the presence of VEGF, and the effect was mediated by αvß3 integrin. We conclude that under physiological conditions, growth factors such as VEGF and fluid forces work together to initiate and spatially guide angiogenesis.


Subject(s)
Extracellular Fluid/physiology , Neovascularization, Physiologic , Diffusion , Humans , Integrin alphaVbeta3/metabolism , Microfluidics , Neovascularization, Physiologic/drug effects , Vascular Endothelial Growth Factor A/pharmacology
17.
Tissue Eng Part C Methods ; 23(8): 474-484, 2017 08.
Article in English | MEDLINE | ID: mdl-28622076

ABSTRACT

Microphysiological systems (MPS), or "organ-on-a-chip" platforms, aim to recapitulate in vivo physiology using small-scale in vitro tissue models of human physiology. While significant efforts have been made to create vascularized tissues, most reports utilize primary endothelial cells that hinder reproducibility. In this study, we report the use of human induced pluripotent stem cell-derived endothelial cells (iPS-ECs) in developing three-dimensional (3D) microvascular networks. We established a CDH5-mCherry reporter iPS cell line, which expresses the vascular endothelial (VE)-cadherin fused to mCherry. The iPS-ECs demonstrate physiological functions characteristic of primary endothelial cells in a series of in vitro assays, including permeability, response to shear stress, and the expression of endothelial markers (CD31, von Willibrand factor, and endothelial nitric oxide synthase). The iPS-ECs form stable, perfusable microvessels over the course of 14 days when cultured within 3D microfluidic devices. We also demonstrate that inhibition of TGF-ß signaling improves vascular network formation by the iPS-ECs. We conclude that iPS-ECs can be a source of endothelial cells in MPS providing opportunities for human disease modeling and improving the reproducibility of 3D vascular networks.


Subject(s)
Cell Culture Techniques/methods , Endothelial Cells/cytology , Induced Pluripotent Stem Cells/cytology , Neovascularization, Physiologic , Angiogenesis Inhibitors/pharmacology , Antigens, CD/metabolism , Cadherins/metabolism , Cell Differentiation/drug effects , Cell Line , Cell Separation , Endothelial Cells/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Microfluidics , Neovascularization, Physiologic/drug effects , Phenotype , Shear Strength , Small Molecule Libraries/pharmacology , Transforming Growth Factor beta/pharmacology
18.
Integr Biol (Camb) ; 9(6): 539-547, 2017 06 19.
Article in English | MEDLINE | ID: mdl-28492659

ABSTRACT

Overexpression of HER2, a receptor tyrosine kinase of the ERBB family, in breast cancer is related to increased cancer progression and aggressiveness. A breast epithelial cell model with the single perturbation of HER2 overexpression is capable of replicating the increased aggressiveness of HER2 overexpressing cancers. In previous work, Wolf-Yadlin and colleagues (Wolf-Yadlin et al., Mol. Syst. Biol., 2006, 2) measured the proximal tyrosine phosphorylation dynamics of the parental and HER2 overexpressing cells (24H) in response to EGF. Here, we apply an ensemble clustering approach to dynamic phosphorylation measurements of the two cell models in order to identify signaling events that explain the increased migratory potential of HER2 overexpressing cells. The use of an ensemble approach for identifying relationships within a dataset and how these relationships change across datasets uncovers relationships that cannot be found by the direct comparison of dynamic responses in the two conditions. Of particular note is a drastic change in the clustering of SHC1 phosphorylation (on site Y349) from an EGFR-MAPK module in parental cells to a module consisting of an E-cadherin junction protein phosphorylation site, catenin delta-1 Y228, in HER2 overexpressing (24H) cells. Given the importance of E-cadherin junctions in healthy epithelial wound healing and migration, we chose to test the computationally-derived identification of altered cell junctions and CTNND1:SHC1 relationships. Our cell and molecular biology experiments demonstrate that SHC and CTNND1 interact in an EGF- and HER2-dependent manner and that the cell junctions are phenotypically affected by HER2, breaking down in response to EGF and yet avoiding apoptosis as a result of cell junction loss. The results suggest a mechanism by which HER2 alters the localization of the SHC-MAPK signaling axis and a phenotypic effect on cell junction integrity.


Subject(s)
Breast Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Antigens, CD , Breast Neoplasms/pathology , Cadherins/metabolism , Catenins/metabolism , Cell Line, Tumor , Disease Progression , Epidermal Growth Factor/metabolism , Female , Humans , Intercellular Junctions/metabolism , Microfluidic Analytical Techniques , Neoplasm Invasiveness , Phosphoproteins/metabolism , Protein Interaction Mapping , Proteomics , Signal Transduction , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , Delta Catenin
19.
PLoS One ; 12(3): e0173747, 2017.
Article in English | MEDLINE | ID: mdl-28282455

ABSTRACT

A growing body of evidence suggests that L-selectin ligands presented on circulating tumor cells facilitate metastasis by binding L-selectin presented on leukocytes. Commonly used methods for detecting L-selectin ligands on tissues, e.g., immunostaining, are performed under static, no-flow conditions. However, such analysis does not assay for functional L-selectin ligands, specifically those ligands that promote adhesion under shear flow conditions. Recently our lab developed a method, termed dynamic biochemical tissue analysis (DBTA), to detect functional selectin ligands in situ by probing tissues with L-selectin-coated microspheres under hemodynamic flow conditions. In this investigation, DBTA was used to probe human colon tissues for L-selectin ligand activity. The detection of L-selectin ligands using DBTA was highly specific. Furthermore, DBTA reproducibly detected functional L-selectin ligands on diseased, e.g., cancerous or inflamed, tissues but not on noncancerous tissues. In addition, DBTA revealed a heterogeneous distribution of functional L-selectin ligands on colon cancer tissues. Most notably, detection of L-selectin ligands by immunostaining using HECA-452 antibody only partially correlated with functional L-selectin ligands detected by DBTA. In summation, the results of this study demonstrate that DBTA detects functional selectin ligands to provide a unique characterization of pathological tissue.


Subject(s)
Biochemistry/methods , Colonic Neoplasms/metabolism , L-Selectin/metabolism , Adenocarcinoma, Mucinous/metabolism , Adenocarcinoma, Mucinous/pathology , Adenocarcinoma, Papillary/metabolism , Adenocarcinoma, Papillary/pathology , Carcinoma, Signet Ring Cell/metabolism , Carcinoma, Signet Ring Cell/pathology , Colonic Neoplasms/pathology , Formaldehyde , Glycoconjugates/analysis , Glycoconjugates/metabolism , Humans , Ligands , Microscopy, Fluorescence , Microspheres , Tissue Fixation/methods
20.
Lab Chip ; 15(17): 3521-9, 2015 Sep 07.
Article in English | MEDLINE | ID: mdl-26190172

ABSTRACT

Tissue engineering can potentially recreate in vivo cellular microenvironments in vitro for an array of applications such as biological inquiry and drug discovery. However, the majority of current in vitro systems still neglect many biological, chemical, and mechanical cues that are known to impact cellular functions such as proliferation, migration, and differentiation. To address this gap, we have developed a novel microfluidic device that precisely controls the spatial and temporal interactions between adjacent three-dimensional cellular environments. The device consists of four interconnected microtissue compartments (~0.1 mm(3)) arranged in a square. The top and bottom pairs of compartments can be sequentially loaded with discrete cellularized hydrogels creating the opportunity to investigate homotypic (left to right or x-direction) and heterotypic (top to bottom or y-direction) cell-cell communication. A controlled hydrostatic pressure difference across the tissue compartments in both x and y direction induces interstitial flow and modulates communication via soluble factors. To validate the biological significance of this novel platform, we examined the role of stromal cells in the process of vasculogenesis. Our device confirms previous observations that soluble mediators derived from normal human lung fibroblasts (NHLFs) are necessary to form a vascular network derived from endothelial colony forming cell-derived endothelial cells (ECFC-ECs). We conclude that this platform could be used to study important physiological and pathological processes that rely on homotypic and heterotypic cell-cell communication.


Subject(s)
Cell Communication/physiology , Cell Culture Techniques/instrumentation , Cellular Microenvironment/physiology , Extracellular Fluid/physiology , Microfluidic Analytical Techniques/instrumentation , Models, Biological , Tissue Engineering/instrumentation , Cell Culture Techniques/methods , Cells, Cultured , Equipment Design , Fibroblasts/cytology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...