Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Lab Chip ; 24(5): 1293-1306, 2024 02 27.
Article in English | MEDLINE | ID: mdl-38230512

ABSTRACT

Microphysiological systems (MPS), also known as miniaturized physiological environments, have been engineered to create and study functional tissue units capable of replicating organ-level responses in specific contexts. The MPS has the potential to provide insights about the safety, characterization, and effectiveness of medical products that are different and complementary to insights gained from traditional testing systems, which can help facilitate the transition of potential medical products from preclinical phases to clinical trials, and eventually to market. While many MPS are versatile and can be used in various applications, most of the current applications have primarily focused on drug discovery and testing. Yet, there is a limited amount of research available that demonstrates the use of MPS in assessing biological products such as cellular and gene therapies. This review paper aims to address this gap by discussing recent technical advancements in MPS and their potential for assessing biological products. We further discuss the challenges and considerations involved in successful translation of MPS into mainstream product testing.


Subject(s)
Biological Products , Microphysiological Systems , Drug Discovery
2.
Adv Biol (Weinh) ; : e2300094, 2023 Jul 06.
Article in English | MEDLINE | ID: mdl-37409400

ABSTRACT

Mesenchymal stromal cells (MSCs) continue to be proposed for clinical investigation to treat myriad diseases given their purported potential to stimulate endogenous regenerative processes, such as angiogenesis. However, MSC functional heterogeneity has hindered clinical success and still poses a substantial manufacturing challenge from a product quality control perspective. Here, a quantitative bioassay based on an enhanced-throughput is described, microphysiological system (MPS) to measure the specific bioactivity of MSCs to stimulate angiogenesis as a potential measure of MSC potency. Using this novel bioassay, MSCs derived from multiple donors at different passages are co-cultured with human umbilical vein endothelial cells and exhibit significant heterogeneity in angiogenic potency between donors and cell passage. Depending on donor source and cellular passage number, MSCs varied in their ability to stimulate tip cell dominant or stalk cell dominant phenotypes in angiogenic sprout morphology which correlated with expression levels of hepatocyte growth factor (HGF). These findings suggest that MSC angiogenic bioactivity may be considered as a possible potency attribute in MSC quality control strategies. Development of a reliable and functionally relevant potency assay for measuring clinically relevant potency attributes of MSCs will help to improve consistency in quality and thereby, accelerate clinical development of these cell-based products.

3.
Exp Biol Med (Maywood) ; 248(22): 2001-2019, 2023 Nov.
Article in English | MEDLINE | ID: mdl-38166397

ABSTRACT

There is a need for better predictive models of the human immune system to evaluate safety and efficacy of immunomodulatory drugs and biologics for successful product development and regulatory approvals. Current in vitro models, which are often tested in two-dimensional (2D) tissue culture polystyrene, and preclinical animal models fail to fully recapitulate the function and physiology of the human immune system. Microphysiological systems (MPSs) that can model key microenvironment cues of the human immune system, as well as of specific organs and tissues, may be able to recapitulate specific features of the in vivo inflammatory response. This minireview provides an overview of MPS for modeling lymphatic tissues, immunity at tissue interfaces, inflammatory diseases, and the inflammatory tumor microenvironment in vitro and ex vivo. Broadly, these systems have utility in modeling how certain immunotherapies function in vivo, how dysfunctional immune responses can propagate diseases, and how our immune system can combat pathogens.


Subject(s)
Immunotherapy , Microphysiological Systems , Animals , Humans , Tumor Microenvironment
4.
Biomaterials ; 290: 121826, 2022 11.
Article in English | MEDLINE | ID: mdl-36201944

ABSTRACT

Mesenchymal stromal cells (MSCs) continue to be proposed for use in clinical trials to treat various diseases due to their therapeutic potential to pleiotropically influence endogenous regenerative processes, such as vasculogenesis. However, the functional heterogeneity of MSCs has hampered their clinical success and poses a significant manufacturing challenge with respect to MSC quality control. Here, we evaluated and qualified a quantitative bioassay based on an enhanced-throughput, microphysiological system to measure the specific paracrine bioactivity of MSCs to stimulate vasculogenesis as a measure of MSC potency. Using this novel bioassay, MSCs derived from multiple donors at different passages were co-cultured with human umbilical vein endothelial cells (HUVECs) and exhibited significant heterogeneity in vasculogenic potency between donors and cell passage. Using our microphysiological system (MPS)-based platform, we demonstrated that variations in MSC vasculogenic bioactivity were maintained when assayed across laboratories and operators. The differences in MSC vasculogenic bioactivity were also correlated with the baseline expression of several genes involved in vasculogenesis (hepatocyte growth factor (HGF), angiopoietin-1 (ANGPT)) or the production of matricellular proteins (fibronectin (FN), insulin-like growth factor-binding protein 7 (IGFBP7)). These findings emphasize the significant functional heterogeneity of MSCs in vasculogenic bioactivity and suggest that changes in baseline gene expression of vasculogenic or matricellular protein genes during manufacturing may affect this bioactivity. The development of a reliable and functionally relevant potency assay for measuring the specific vasculogenic bioactivity of manufactured MSCs will help to reliably assure their quality when used in appropriate clinical trials.


Subject(s)
Mesenchymal Stem Cells , Humans , Mesenchymal Stem Cells/metabolism , Coculture Techniques , Cell Differentiation , Human Umbilical Vein Endothelial Cells/metabolism , Biological Assay , Cells, Cultured , Cell Proliferation
5.
ALTEX ; 39(2): 297­314, 2022.
Article in English | MEDLINE | ID: mdl-35064273

ABSTRACT

Complex in vitro models (CIVM) offer the potential to improve pharmaceutical clinical drug attrition due to safety and/ or efficacy concerns. For this technology to have an impact, the establishment of robust characterization and qualifi­cation plans constructed around specific contexts of use (COU) is required. This article covers the output from a workshop between the Food and Drug Administration (FDA) and Innovation and Quality Microphysiological Systems (IQ MPS) Affiliate. The intent of the workshop was to understand how CIVM technologies are currently being applied by pharma­ceutical companies during drug development and are being tested at the FDA through various case studies in order to identify hurdles (real or perceived) to the adoption of microphysiological systems (MPS) technologies, and to address evaluation/qualification pathways for these technologies. Output from the workshop includes the alignment on a working definition of MPS, a detailed description of the eleven CIVM case studies presented at the workshop, in-depth analysis, and key take aways from breakout sessions on ADME (absorption, distribution, metabolism, and excretion), pharmacology, and safety that covered topics such as qualification and performance criteria, species differences and concordance, and how industry can overcome barriers to regulatory submission of CIVM data. In conclusion, IQ MPS Affiliate and FDA scientists were able to build a general consensus on the need for animal CIVMs for preclinical species to better determine species concordance. Furthermore, there was acceptance that CIVM technologies for use in ADME, pharmacology and safety assessment will require qualification, which will vary depending on the specific COU.


Subject(s)
Animal Testing Alternatives , Lab-On-A-Chip Devices , Animals , Drug Evaluation, Preclinical , Drug Industry , Pharmaceutical Preparations/metabolism , United States , United States Food and Drug Administration
6.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Article in English | MEDLINE | ID: mdl-34446555

ABSTRACT

Mesenchymal stromal cells (MSCs) are increasingly combined with biomaterials to enhance their therapeutic properties, including their immunosuppressive function. However, clinical trials utilizing MSCs with or without biomaterials have shown limited success, potentially due to their functional heterogeneity across different donors and among different subpopulations of cells. Here, we evaluated the immunosuppressive capacity, as measured by the ability to reduce T-cell proliferation and activation, of interferon-gamma (IFN-γ)-licensed MSCs from multiple donors on fibrin and collagen hydrogels, the two most commonly utilized biomaterials in combination with MSCs in clinical trials worldwide according to ClinicalTrials.gov Variations in the immunosuppressive capacity between IFN-γ-licensed MSC donors on the biomaterials correlated with the magnitude of indoleamine-2,3-dioxygenase activity. Immunosuppressive capacity of the IFN-γ-licensed MSCs depended on the αV/α5 integrins when cultured on fibrin and on the α2/ß1 integrins when cultured on collagen. While all tested MSCs were nearly 100% positive for these integrins, sorted MSCs that expressed higher levels of αV/α5 integrins demonstrated greater immunosuppressive capacity with IFN-γ licensing than MSCs that expressed lower levels of these integrins on fibrin. These findings were equivalent for MSCs sorted based on the α2/ß1 integrins on collagen. These results demonstrate the importance of integrin engagement to IFN-γ licensed MSC immunosuppressive capacity and that IFN-γ-licensed MSC subpopulations of varying immunosuppressive capacity can be identified by the magnitude of integrin expression specific to each biomaterial.


Subject(s)
Collagen/metabolism , Fibrin/metabolism , Immunosuppression Therapy , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Integrin alpha2beta1/metabolism , Interferon-gamma/pharmacology , Mesenchymal Stem Cells/cytology , Antiviral Agents/pharmacology , Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Collagen/chemistry , Fibrin/chemistry , Humans , Hydrogels/chemistry , Hydrogels/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism
7.
Exp Biol Med (Maywood) ; 246(16): 1845-1856, 2021 08.
Article in English | MEDLINE | ID: mdl-34250847

ABSTRACT

There are a growing number of globally approved products and clinical trials utilizing autologous and allogeneic therapeutic cells for applications in regenerative medicine and immunotherapies. However, there is a need to develop rapid and cost-effective methods for manufacturing therapeutically effective cells. Furthermore, the resulting manufactured cells may exhibit heterogeneities that result in mixed therapeutic outcomes. Engineering approaches that can provide distinct microenvironmental cues to these cells may be able to enhance the growth and characterization of these cell products. This mini-review describes strategies to potentially enhance the expansion of therapeutic cells with biomaterials and bioreactors, as well as to characterize the cell products with microphysiological systems. These systems can provide distinct cues to maintain the quality attributes of the cells and evaluate their function in physiologically relevant conditions.


Subject(s)
Biocompatible Materials , Bioreactors , Regenerative Medicine , Tissue Engineering , Biocompatible Materials/metabolism , Cell- and Tissue-Based Therapy/methods , Drug Development/methods , Humans , Tissue Engineering/methods
8.
EBioMedicine ; 42: 408-419, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30902740

ABSTRACT

BACKGROUND: Anti-angiogenic treatment failure is often attributed to drug resistance, unsuccessful drug delivery, and tumor heterogeneity. Recent studies have speculated that anti-angiogenic treatments may fail due to characteristics inherent to tumor-associated blood vessels. Tumor-associated blood vessels are phenotypically different from their normal counterparts, having defective or permeable endothelial monolayers, abnormal sprouts, and abnormal vessel hierarchy. Therefore, to predict the efficacy of anti-angiogenic therapies in an individual patient, in vitro models that mirror individual patient's tumor vascular biology and response to anti-angiogenic treatment are needed. METHODS: We used a microfluidic in vitro organotypic model to create patient-specific biomimetic blood vessels from primary patient-specific tumor endothelial cells (TEnCs) and normal endothelial cells (NEnC). We assessed number of sprouts and vessel organization via microscopy imaging and image analysis. We characterized NEnC and TEnC vessel secretions via multiplex bead-based ELISA. FINDINGS: Using this model, we found that TEnC vessels exhibited more angiogenic sprouts than NEnC vessels. We also found a more disorganized and gap-filled endothelial monolayer. NEnCs and TEnC vessels exhibited heterogeneous functional drug responses across the five patients screened, as described in the clinic. INTERPRETATION: Our model recapitulated hallmarks of TEnCs and NEnCs found in vivo and captured the functional and structural differences between TEnC and NEnC vessels. This model enables a platform for therapeutic drug screening and assessing patient-specific responses with great potential to inform personalized medicine approaches. FUNDING: NIH grants R01 EB010039, R33 CA225281, R01CA186134 University of Wisconsin Carbone Cancer Center (CA014520), and University of Wisconsin Hematology training grant T32 HL07899.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Models, Biological , Neovascularization, Pathologic , Carcinoma, Renal Cell/drug therapy , Cell Adhesion , Cell Line, Tumor , Cell Movement , Endothelial Cells/metabolism , Humans , Immunophenotyping , Kidney Neoplasms/drug therapy , Molecular Imaging , Neovascularization, Pathologic/drug therapy , Phenotype
9.
Stem Cells Transl Med ; 7(9): 664-675, 2018 09.
Article in English | MEDLINE | ID: mdl-30084545

ABSTRACT

Multipotent stromal cells (MSCs) are an attractive cell source for bone and cartilage tissue repair strategies. However, the functional heterogeneity of MSCs derived from different donors and manufacturing conditions has limited clinical translation, emphasizing the need for improved methods to assess MSC chondrogenic capacity. We used functionally relevant morphological profiling to dynamically monitor emergent morphological phenotypes of chondrogenically induced MSC aggregates to identify morphological features indicative of MSC chondrogenesis. Toward this goal, we characterized the morphology of chondrogenically stimulated MSC aggregates from eight different human cell-lines at multiple passages and demonstrated that MSC aggregates exhibited unique morphological dynamics that were both cell line- and passage-dependent. This variation in 3D morphology was shown to be informative of long-term MSC chondrogenesis based on multiple quantitative functional assays. We found that the specific morphological features of spheroid area, radius, minimum feret diameter, and minor axis length to be strongly correlated with MSC chondrogenic synthetic activity but not gene expression as early as day 4 in 3D culture. Our high-throughput, nondestructive approach could potentially serve as a tool to identify MSC lines with desired chondrogenic capacity toward improving manufacturing strategies for MSC-based cellular products for cartilage tissue repair. Stem Cells Translational Medicine 2018;1-12.


Subject(s)
Mesenchymal Stem Cells/metabolism , Spheroids, Cellular/metabolism , Transcriptome , Bone Marrow Cells/cytology , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Chondrogenesis , Cluster Analysis , Humans , Mesenchymal Stem Cells/cytology , Phenotype , Principal Component Analysis , Spheroids, Cellular/cytology
10.
Trends Biotechnol ; 36(1): 105-118, 2018 01.
Article in English | MEDLINE | ID: mdl-29126572

ABSTRACT

Heterogeneity in cell function has presented a significant hurdle to the successful clinical translation of many cellular therapies. Current techniques for assessing cell quality and the effects of microenvironmental cues and manufacturing processes on cell behavior often inadequately address heterogeneity due to issues such as population versus single-cell measurements and the therapeutic relevance and throughput/robustness of the assay. Due to the well-established relationship between morphology and cellular function, morphological profiling has become increasingly utilized to better understand functional heterogeneity and its impact on therapeutic development. In this review, we introduce an emerging field we term functionally-relevant morphological profiling with great potential to improve our understanding of cellular heterogeneity through discovering novel quality attributes, optimizing manufacturing, and screening drugs/biomaterials.


Subject(s)
Cell Shape , Cytological Techniques/methods , Image Processing, Computer-Assisted/methods , Microscopy/methods , Optical Imaging/methods , Cell Culture Techniques/methods , Cell- and Tissue-Based Therapy/methods , Humans , Technology, Pharmaceutical/methods
11.
SLAS Technol ; 22(6): 646-661, 2017 12.
Article in English | MEDLINE | ID: mdl-28825968

ABSTRACT

Multipotent stromal cells (MSCs, often called mesenchymal stem cells) have garnered significant attention within the field of regenerative medicine because of their purported ability to differentiate down musculoskeletal lineages. Given the inherent heterogeneity of MSC populations, recent studies have suggested that cell morphology may be indicative of MSC differentiation potential. Toward improving current methods and developing simple yet effective approaches for the morphological evaluation of MSCs, we combined passive pumping microfluidic technology with high-dimensional morphological characterization to produce robust tools for standardized high-throughput analysis. Using ultraviolet (UV) light as a modality for reproducible polystyrene substrate modification, we show that MSCs seeded on microfluidic straight channel devices incorporating UV-exposed substrates exhibited morphological changes that responded accordingly to the degree of substrate modification. Substrate modification also effected greater morphological changes in MSCs seeded at a lower rather than higher density within microfluidic channels. Despite largely comparable trends in morphology, MSCs seeded in microscale as opposed to traditional macroscale platforms displayed much higher sensitivity to changes in substrate properties. In summary, we adapted and qualified microfluidic cell culture platforms comprising simple straight channel arrays as a viable and robust tool for high-throughput quantitative morphological analysis to study cell-material interactions.


Subject(s)
Cytological Techniques/methods , Lab-On-A-Chip Devices , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Microfluidics/methods , Polystyrenes , Cells, Cultured , Humans , Ultraviolet Rays
12.
Methods Mol Biol ; 1458: 59-69, 2016.
Article in English | MEDLINE | ID: mdl-27581014

ABSTRACT

Microscale 3D in vitro systems have attracted significant interest as tools for cancer research because the microscale systems offer better organization of the cellular microenvironment and enhance throughput of the systems by lowering costs and reducing the amount of reagents and cells. Lumens (i.e., tubular structures) are ubiquitous in vivo being present in blood vessels, mammary ducts, prostate ducts, and the lymphatic system. Lumen structures of varying size and geometry are involved in key normal and disease processes including morphogenesis, angiogenesis, cancer development, and drug delivery. Therefore, there is a need for practical methods that create various lumen structures having different size and geometries to investigate how cells in the lumen structure respond to certain microenvironmental conditions during cancer development and progression. Here, we present a method to create multiple three-dimensional (3D) luminal structures, where parameters, such as size, geometry, and distance, can easily be controlled using simple poly-dimethylsiloxane (PDMS) micro-molds.


Subject(s)
Microfluidics , Neoplasms/pathology , Tumor Microenvironment , Biomimetics/methods , Cell Culture Techniques , Epithelial Cells , Humans , In Vitro Techniques , Microfluidics/instrumentation , Microfluidics/methods
13.
Pharmacol Ther ; 165: 79-92, 2016 09.
Article in English | MEDLINE | ID: mdl-27218886

ABSTRACT

Personalized cancer therapy focuses on characterizing the relevant phenotypes of the patient, as well as the patient's tumor, to predict the most effective cancer therapy. Historically, these methods have not proven predictive in regards to predicting therapeutic response. Emerging culture platforms are designed to better recapitulate the in vivo environment, thus, there is renewed interest in integrating patient samples into in vitro cancer models to assess therapeutic response. Successful examples of translating in vitro response to clinical relevance are limited due to issues with patient sample acquisition, variability and culture. We will review traditional and emerging in vitro models for personalized medicine, focusing on the technologies, microenvironmental components, and readouts utilized. We will then offer our perspective on how to apply a framework derived from toxicology and ecology towards designing improved personalized in vitro models of cancer. The framework serves as a tool for identifying optimal readouts and culture conditions, thus maximizing the information gained from each patient sample.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Screening Assays, Antitumor/methods , Neoplasms/drug therapy , Precision Medicine/methods , Antineoplastic Agents/adverse effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Separation/methods , Drug Resistance, Neoplasm , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Patient Selection , Predictive Value of Tests , Primary Cell Culture , Signal Transduction/drug effects , Treatment Outcome , Tumor Cells, Cultured , Tumor Microenvironment
14.
J Biomol Screen ; 21(1): 65-73, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26420788

ABSTRACT

Botulinum neurotoxin (BoNT) is the most lethal naturally produced neurotoxin. Due to the extreme toxicity, BoNTs are implicated in bioterrorism, while the specific mechanism of action and long-lasting effect was found to be medically applicable in treating various neurological disorders. Therefore, for both public and patient safety, a highly sensitive, physiologic, and specific assay is needed. In this paper, we show a method for achieving a highly sensitive cell-based assay for BoNT/A detection using the motor neuron-like continuous cell line NG108-15. To achieve high sensitivity, we performed a media optimization study evaluating three commercially available neural supplements in combination with retinoic acid, purmorphamine, transforming growth factor ß1 (TGFß1), and ganglioside GT1b. We found nonlinear combinatorial effects on BoNT/A detection sensitivity, achieving an EC50 of 7.4 U ± 1.5 SD (or ~7.9 pM). The achieved detection sensitivity is comparable to that of assays that used primary and stem cell-derived neurons as well as the mouse lethality assay.


Subject(s)
Biological Assay/methods , Botulinum Toxins, Type A/toxicity , Culture Media/metabolism , Motor Neurons/drug effects , Neurotoxins/toxicity , Animals , Cell Line, Tumor , Embryonic Stem Cells/drug effects , Gangliosides/metabolism , Mice , Morpholines/metabolism , Purines/metabolism , Rats , Sensitivity and Specificity , Transforming Growth Factor beta1/metabolism , Tretinoin/metabolism
15.
Adv Healthc Mater ; 5(2): 198-204, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26610188

ABSTRACT

In vitro biomimetic modeling of physio-logical structures bridges the gap between 2D in vitro culture and animal models. Lumens (tubular structures) are ubiquitous in vivo, being present in blood vessels, mammary ducts, and the lymphatic system. A method 'LumeNEXT' is presented here that allows the fabrication of 3D embedded lumens where size, structure, distance, and configuration can be controlled using standard poly-dimethylsiloxane micromolding methods.


Subject(s)
Extracellular Matrix/metabolism , Gels/chemistry , Tissue Engineering/methods , Animals , Cell Shape , Human Umbilical Vein Endothelial Cells/cytology , Humans , Magnetics , Rats
16.
BMC Cancer ; 15: 12, 2015 Jan 21.
Article in English | MEDLINE | ID: mdl-25605670

ABSTRACT

BACKGROUND: Ductal carcinoma in situ (DCIS) is a non-invasive form of breast cancer that is thought to be a precursor to most invasive and metastatic breast cancers. Understanding the mechanisms regulating the invasive transition of DCIS is critical in order to better understand how some types of DCIS become invasive. While significant insights have been gained using traditional in vivo and in vitro models, existing models do not adequately recapitulate key structure and functions of human DCIS well. In addition, existing models are time-consuming and costly, limiting their use in routine screens. Here, we present a microscale DCIS model that recapitulates key structures and functions of human DCIS, while enhancing the throughput capability of the system to simultaneously screen numerous molecules and drugs. METHODS: Our microscale DCIS model is prepared in two steps. First, viscous finger patterning is used to generate mammary epithelial cell-lined lumens through extracellular matrix hydrogels. Next, DCIS cells are added to fill the mammary ducts to create a DCIS-like structure. For coculture experiments, human mammary fibroblasts (HMF) are added to the two side channels connected to the center channel containing DCIS. To validate the invasive transition of the DCIS model, the invasion of cancer cells and the loss of cell-cell junctions are then examined. A student t-test is conducted for statistical analysis. RESULTS: We demonstrate that our DCIS model faithfully recapitulates key structures and functions of human mammary DCIS and can be employed to study the mechanisms involved in the invasive progression of DCIS. First, the formation of cell-cell junctions and cell polarity in the normal mammary duct, and the structure of the DCIS model are characterized. Second, coculture with HMF is shown to induce the invasion of DCIS. Third, multiple endpoint analyses are demonstrated to validate the invasion. CONCLUSIONS: We have developed and characterized a novel in vitro model of normal and DCIS-inflicted mammary ducts with 3D lumen structures. These models will enable researchers to investigate the role of microenvironmental factors on the invasion of DCIS in more in vivo-like conditions.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , In Vitro Techniques/methods , Models, Biological , Cell Line, Tumor , Coculture Techniques , Female , Humans , In Vitro Techniques/instrumentation , Microfluidics , Neoplasm Invasiveness
17.
FASEB J ; 28(11): 4583-90, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25077562

ABSTRACT

Advances in tissue engineering and microtechnology have enabled researchers to more easily generate in vitro tissue models that mimic the tissue geometry and spatial organization found in vivo (e.g., vessel or mammary duct models with tubular structures). However, the widespread adoption of these models for biological studies has been slow, in part due to the lack of direct comparisons between existing 2-dimensional and 3-dimensional cell culture models and new organotypic models that better replicate tissue structure. Using previously developed vessel and mammary duct models with 3-dimensional lumen structures, we have begun to explore this question. In a direct comparison between these next generation organotypic models and more traditional methods, we observed differences in the levels of several secreted growth factors and cytokines. In addition, endothelial vessel geometry profoundly affects the phenotypic behavior of carcinoma cells, suggesting that more traditional in vitro assays may not capture in vivo events. Here, we seek to review and add to the increasing evidence supporting the hypothesis that using cell culture models with more relevant tissue structure influences cell fate and behavior, potentially increasing the relevance of biological findings.


Subject(s)
Blood Vessels/cytology , Cell Differentiation/physiology , Cell Culture Techniques/methods , Cytokines/biosynthesis , Endothelial Cells/cytology , Humans , Models, Biological , Tissue Engineering/methods
18.
PLoS One ; 8(12): e81689, 2013.
Article in English | MEDLINE | ID: mdl-24349113

ABSTRACT

Here, we describe an engineering approach to quantitatively compare migration, morphologies, and adhesion for tumorigenic human fibrosarcoma cells (HT-1080s) and primary human dermal fibroblasts (hDFs) with the aim of identifying distinguishing properties of the transformed phenotype. Relative adhesiveness was quantified using self-assembled monolayer (SAM) arrays and proteolytic 3-dimensional (3D) migration was investigated using matrix metalloproteinase (MMP)-degradable poly(ethylene glycol) (PEG) hydrogels ("synthetic extracellular matrix" or "synthetic ECM"). In synthetic ECM, hDFs were characterized by vinculin-containing features on the tips of protrusions, multipolar morphologies, and organized actomyosin filaments. In contrast, HT-1080s were characterized by diffuse vinculin expression, pronounced ß1-integrin on the tips of protrusions, a cortically-organized F-actin cytoskeleton, and quantitatively more rounded morphologies, decreased adhesiveness, and increased directional motility compared to hDFs. Further, HT-1080s were characterized by contractility-dependent motility, pronounced blebbing, and cortical contraction waves or constriction rings, while quantified 3D motility was similar in matrices with a wide range of biochemical and biophysical properties (including collagen) despite substantial morphological changes. While HT-1080s were distinct from hDFs for each of the 2D and 3D properties investigated, several features were similar to WM239a melanoma cells, including rounded, proteolytic migration modes, cortical F-actin organization, and prominent uropod-like structures enriched with ß1-integrin, F-actin, and melanoma cell adhesion molecule (MCAM/CD146/MUC18). Importantly, many of the features observed for HT-1080s were analogous to cellular changes induced by transformation, including cell rounding, a disorganized F-actin cytoskeleton, altered organization of focal adhesion proteins, and a weakly adherent phenotype. Based on our results, we propose that HT-1080s migrate in synthetic ECM with functional properties that are a direct consequence of their transformed phenotype.


Subject(s)
Cell Movement/genetics , Cell Transformation, Neoplastic , Fibroblasts/pathology , Phenotype , Actins/genetics , Actins/metabolism , CD146 Antigen/genetics , CD146 Antigen/metabolism , Cell Adhesion , Cell Culture Techniques , Cell Line, Tumor , Extracellular Matrix/chemistry , Fibroblasts/metabolism , Gene Expression , Humans , Hydrogels , Integrin beta1/genetics , Integrin beta1/metabolism , Matrix Metalloproteinases/chemistry , Molecular Mimicry , Primary Cell Culture , Vinculin/genetics , Vinculin/metabolism
19.
PLoS One ; 7(10): e46685, 2012.
Article in English | MEDLINE | ID: mdl-23056402

ABSTRACT

Stromal fibroblasts actively participate in normal mammary gland homeostasis and in breast carcinoma growth and progression by secreting paracrine factors; however, little is known about the identity of paracrine mediators in individual patients. The purpose of this study was to characterize paracrine signaling pathways between breast carcinoma cells and breast carcinoma-associated fibroblasts (CAF) or normal mammary fibroblasts (NF), respectively. CAF and NF were isolated from breast carcinoma tissue samples and adjacent normal mammary gland tissue of 28 patients. The fibroblasts were grown in 3D collagen gel co-culture with T47D human breast carcinoma cells and T47D cell growth was measured. CAF stimulated T47D cell growth to a significantly greater degree than NF. We detected a considerable inter-individual heterogeneity of paracrine interactions but identified FGF2, HB-EGF, heparanase-1 and SDF1 as factors that were consistently responsible for the activity of carcinoma-associated fibroblasts. CAF from low-grade but not high-grade carcinomas required insulin-like growth factor 1 and transforming growth factor beta 1 to stimulate carcinoma growth. Paradoxically, blocking of membrane-type 1 matrix metalloprotease stimulated T47D cell growth in co-culture with NF. The results were largely mirrored by treating the fibroblasts with siRNA oligonucleotides prior to co-culture, implicating the fibroblasts as principal production site for the secreted mediators. In summary, we identify a paracrine signaling network with inter-individual commonalities and differences. These findings have significant implications for the design of stroma-targeted therapies.


Subject(s)
Breast Neoplasms/metabolism , Fibroblasts/metabolism , Breast , Breast Neoplasms/genetics , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Female , Humans , Paracrine Communication/genetics , Paracrine Communication/physiology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...