698 results on '"Griffith, Linda G."'
Search Results
152. A process engineering approach to increase organoid yield
- Author
-
Arora, Natasha, primary, Alsous, Jasmin Imran, additional, Guggenheim, Jacob W., additional, Mak, Michael, additional, Munera, Jorge, additional, Wells, James M., additional, Kamm, Roger D., additional, Asada, H. Harry, additional, Shvartsman, Stanislav Y., additional, and Griffith, Linda G., additional
- Published
- 2017
- Full Text
- View/download PDF
153. Modification of proteolytic activity matrix analysis (PrAMA) to measure ADAM10 and ADAM17 sheddase activities in cell and tissue lysates
- Author
-
Yoneyama, Toshie, primary, Gorry, Michael, additional, Miller, Miles A, additional, Gaither-Davis, Autumn, additional, Lin, Yan, additional, Moss, Marcia L., additional, Griffith, Linda G., additional, Lauffenburger, Douglas A., additional, Stabile, Laura P., additional, Herman, James G., additional, and Vujanovic, Nikola L., additional
- Published
- 2017
- Full Text
- View/download PDF
154. Multi-functional scaling methodology for translational pharmacokinetic and pharmacodynamic applications using integrated microphysiological systems (MPS)
- Author
-
Maass, Christian, primary, Stokes, Cynthia L., additional, Griffith, Linda G., additional, and Cirit, Murat, additional
- Published
- 2017
- Full Text
- View/download PDF
155. Marrow-Derived Stem Cell Motility in 3D Synthetic Scaffold Is Governed by Geometry Along With Adhesivity and Stiffness
- Author
-
Massachusetts Institute of Technology. Center for Gynepathology Research, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Biology, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Materials Science and Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Peyton, Shelly R., Kalcioglu, Zeynep Ilke, Cohen, Joshua C., Runkle, Anne P., Van Vliet, Krystyn J., Lauffenburger, Douglas A., Griffith, Linda G., Van Vliet, Krystyn J, Lauffenburger, Douglas A, Griffith, Linda G, Massachusetts Institute of Technology. Center for Gynepathology Research, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Biology, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Materials Science and Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Peyton, Shelly R., Kalcioglu, Zeynep Ilke, Cohen, Joshua C., Runkle, Anne P., Van Vliet, Krystyn J., Lauffenburger, Douglas A., Griffith, Linda G., Van Vliet, Krystyn J, Lauffenburger, Douglas A, and Griffith, Linda G
- Abstract
Author Manuscript 2012 May 21., Design of 3D scaffolds that can facilitate proper survival, proliferation, and differentiation of progenitor cells is a challenge for clinical applications involving large connective tissue defects. Cell migration within such scaffolds is a critical process governing tissue integration. Here, we examine effects of scaffold pore diameter, in concert with matrix stiffness and adhesivity, as independently tunable parameters that govern marrow-derived stem cell motility. We adopted an “inverse opal” processing technique to create synthetic scaffolds by crosslinking poly(ethylene glycol) at different densities (controlling matrix elastic moduli or stiffness) and small doses of a heterobifunctional monomer (controlling matrix adhesivity) around templating beads of different radii. As pore diameter was varied from 7 to 17 µm (i.e., from significantly smaller than the spherical cell diameter to approximately cell diameter), it displayed a profound effect on migration of these stem cells—including the degree to which motility was sensitive to changes in matrix stiffness and adhesivity. Surprisingly, the highest probability for substantive cell movement through pores was observed for an intermediate pore diameter, rather than the largest pore diameter, which exceeded cell diameter. The relationships between migration speed, displacement, and total path length were found to depend strongly on pore diameter. We attribute this dependence to convolution of pore diameter and void chamber diameter, yielding different geometric environments experienced by the cells within. Bioeng. 2011; 108:1181–1193, (National Institute of General Medical Sciences (U.S.) (NRSA Fellowship GM083472), National Institutes of Health (U.S.) (National Institute of General Medical Sciences (U.S.) Cell Migration Consortium Grant GM064346), National Science Foundation (U.S.) (CAREER CBET-0644846)
- Published
- 2012
156. Co-regulation of primary mouse hepatocyte viability and function by oxygen and matrix
- Author
-
Inman, S. Walker, Rusyn, Ivan, Griffith, Linda G., Buck, Lorenna D., Massachusetts Institute of Technology. Department of Biological Engineering, Buck, Lorenna D., Inman, S. Walker, and Griffith, Linda G.
- Abstract
Although oxygen and extracellular matrix cues both influence differentiation state and metabolic function of primary rat and human hepatocytes, relatively little is known about how these factors together regulate behaviors of primary mouse hepatocytes in culture. To determine the effects of pericellular oxygen tension on hepatocellular function, we employed two methods of altering oxygen concentration in the local cellular microenvironment of cells cultured in the presence or absence of an extracellular matrix (Matrigel) supplement. By systematically altering medium depth and gas phase oxygen tension, we created multiple oxygen regimes (hypoxic, normoxic, and hyperoxic) and measured the local oxygen concentrations in the pericellular environment using custom-designed oxygen microprobes. From these measurements of oxygen concentrations, we derived values of oxygen consumption rates under a spectrum of environmental contexts, thus providing the first reported estimates of these values for primary mouse hepatocytes. Oxygen tension and matrix microenvironment were found to synergistically regulate hepatocellular survival and function as assessed using quantitative image analysis for cells stained with vital dyes, and assessment of secretion of albumin. Hepatocellular viability was affected only at strongly hypoxic conditions. Surprisingly, albumin secretion rates were greatest at a moderately supra-physiological oxygen concentration, and this effect was mitigated at still greater supra-physiological concentrations. Matrigel enhanced the effects of oxygen on retention of function. This study underscores the importance of carefully controlling cell density, medium depth, and gas phase oxygen, as the effects of these parameters on local pericellular oxygen tension and subsequent hepatocellular function are profound., National Institutes of Health (U.S.) (Grant P50-GM068762-08), National Institutes of Health (U.S.) (Grant R01-EB010246-04), National Institutes of Health (U.S.) (Grant R01-ES015241), National Institutes of Health (U.S.) (Grant P30-ES002109)
- Published
- 2013
157. Sortase Mediated Ligation to Covalently Tether Bioactive Proteins to 2D and 3D Hydrogel Systems
- Author
-
Renggli Kasper, Maurissen Thomas L, Cambria Elena, Imperiali Barbara, and Griffith Linda G
- Subjects
technology, industry, and agriculture ,macromolecular substances - Abstract
Synthetic extracellular matrices are widely used in regenerative medicine and as tools in building in vitro physiological culture models. Synthetic hydrogels display advantageous physical properties but are challenging to modify with large peptides or proteins. Here a facile mild enzymatic tethering approach is presented. Sortase mediated ligation was used to conjugate human epidermal growth factor fused to a GGG ligation motif (GGG EGF) to pre formed PEG hydrogels or PEG macromers containing the sortase LPRTG substrate. The reversibility of the sortase reaction was then exploited to cleave tethered EGF from the hydrogels for analysis. Analyses of the reaction supernatant and the post ligation of pre formed hydrogels showed that the amount of tethered EGF increases with increasing LPRTG in the hydrogel or GGG EGF in the supernatant. Sortase tethered EGF was biologically active as demonstrated by stimulation of DNA synthesis in a 2D culture of primary human hepatocytes and endometrial epithelial cells. In addition results with the PEG macromers demonstrate that EGF integrates into 3D PEG hydrogels and that the EGF is released from crosslinked PEG EGF macromers upon sortase reaction on the gels. Furthermore the tethering of several EGF molecules to single PEG stars macromers allows a local clustering effect for growth factor presentation to cells. The simplicity specificity and reversibility of sortase mediated ligation and cleavage reactions make it an attrac tive approach for modification of hydrogel systems.
- Published
- 2015
- Full Text
- View/download PDF
158. Engineering Helical Modular Polypeptide-Based Hydrogels as Synthetic Extracellular Matrices for Cell Culture
- Author
-
He, Hongkun, Sofman, Marianna, Wang, Alex J-S, Ahrens, Caroline C., Wang, Wade, Griffith, Linda G., and Hammond, Paula T.
- Abstract
Expanding the toolkit of modular and functional synthetic material systems for biomimetic extracellular matrices (ECMs) is needed for achieving more predictable and characterizable cell culture. In the present study, we engineered a synthetic hydrogel system incorporating poly(γ-propargyl-l-glutamate) (PPLG), an N-carboxy anhydride polypeptide with a unique α-helical secondary structure. PPLG macromers were cross-linked into poly(ethylene glycol) (PEG) networks to form hybrid polypeptide-PEG hydrogels. We compared the properties of PPLG-PEG to systems where the PPLG macromers were replaced with 8-arm PEG or poly(γ-propargyl-d,l-glutamate) (PPDLG), which has a flexible random-coil conformation. We evaluated each hydrogel system as synthetic ECMs for two-dimensional (2D) endothelial cell culture. Cells on PPLG-PEG displayed superior attachment and spreading at comparable adhesion ligand incorporation concentrations, demonstrating the unique benefit of combining the more rigid and hydrophobic α-helical PPLG within the more flexible and hydrophilic PEG matrix. The modular PPLG macromer is a promising building block for developing other types of PPLG-based hydrogels with favorable and tunable properties.
- Published
- 2020
- Full Text
- View/download PDF
159. High-Throughput Mutiplexed Protease Activity Measurement Using a Droplet Based Microfluidic Platform with Picoinjector
- Author
-
chia hung chen, Miller, Miles A., Aniruddh Sarkar, Beste, Michael T., Lauffenburger, Douglas A., Griffith, Linda G., Jongyoon Han, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology. Research Laboratory of Electronics, Miller, Miles Aaron, Sarkar, Aniruddh, Beste, Michael T., Lauffenburger, Douglas A., Griffith, Linda G., and Han, Jongyoon
- Abstract
In this study, we integrated several components, including a droplet generator, a pico-injector, and an analytical inference technique, Proteolytic Activity Matrix Analysis (PrAMA), to create a platform for assessing multiple specific protease activity assays with minimal liquid handling and sample-requirement for personal medicine analysis. The microfluidic platform enables the direct measurements of protease enzyme activity, which is more physiologically informative than the standard measurements to determine the enzyme concentration alone. By tracking hundreds of picoliter droplets containing biological samples mixed with unique FRET-based protease substrates and inhibitors, the assay simultaneously infers multiple specific protease activities with minimal (, National Science Foundation (U.S.). Graduate Research Fellowship, National Institutes of Health (U.S.) (CDP Center Grant P50-GM68762), National Institutes of Health (U.S.) (Grant R01-GM081336), National Cancer Institute (U.S.) (Grant U54-CA112967), United States. Defense Advanced Research Projects Agency (Cipher Program)
- Published
- 2012
160. Genetically engineering self-organization of human pluripotent stem cells into a liver bud-like tissue using Gata6
- Author
-
MIT Synthetic Biology Center, Massachusetts Institute of Technology. Department of Biological Engineering, MIT Emergent Behaviors of Integrated Cellular Systems Center, Guye, Patrick, Ebrahimkhani, Mohammad Reza, Kipniss, Nathan, Velazquez, Jeremy J., Schoenfeld, Eldi, Kiani, Samira, Griffith, Linda G., Weiss, Ron, MIT Synthetic Biology Center, Massachusetts Institute of Technology. Department of Biological Engineering, MIT Emergent Behaviors of Integrated Cellular Systems Center, Guye, Patrick, Ebrahimkhani, Mohammad Reza, Kipniss, Nathan, Velazquez, Jeremy J., Schoenfeld, Eldi, Kiani, Samira, Griffith, Linda G., and Weiss, Ron
- Abstract
Human induced pluripotent stem cells (hiPSCs) have potential for personalized and regenerative medicine. While most of the methods using these cells have focused on deriving homogenous populations of specialized cells, there has been modest success in producing hiPSC-derived organotypic tissues or organoids. Here we present a novel approach for generating and then co-differentiating hiPSC-derived progenitors. With a genetically engineered pulse of GATA-binding protein 6 (GATA6) expression, we initiate rapid emergence of all three germ layers as a complex function of GATA6 expression levels and tissue context. Within 2 weeks we obtain a complex tissue that recapitulates early developmental processes and exhibits a liver bud-like phenotype, including haematopoietic and stromal cells as well as a neuronal niche. Collectively, our approach demonstrates derivation of complex tissues from hiPSCs using a single autologous hiPSCs as source and generates a range of stromal cells that co-develop with parenchymal cells to form tissues., National Science Foundation (U.S.). Emergent Behaviors of Integrated Cellular Systems (NSF CBET-0939511), Synthetic Biology Engineering Research Center, National Institutes of Health (U.S.) (P50GM098792), Ernst Schering Foundation, Swiss National Science Foundation
- Published
- 2016
161. Photopatterning of hydrogel scaffolds coupled to filter materials using stereolithography for perfused 3D culture of hepatocytes
- Author
-
Massachusetts Institute of Technology. Center for Gynepathology Research, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Materials Science and Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Neiman, Jaclyn A. Shepard, Chan, Vincent, Rhoads, Mary G., Raredon, Micha Sam Brickman, Velazquez, Jeremy J., Dyer, Rachel Lee, Hammond, Paula T., Griffith, Linda G., Raman, Ritu, Bashir, Rashid, Massachusetts Institute of Technology. Center for Gynepathology Research, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Materials Science and Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Neiman, Jaclyn A. Shepard, Chan, Vincent, Rhoads, Mary G., Raredon, Micha Sam Brickman, Velazquez, Jeremy J., Dyer, Rachel Lee, Hammond, Paula T., Griffith, Linda G., Raman, Ritu, and Bashir, Rashid
- Abstract
In vitro models that recapitulate the liver's structural and functional complexity could prolong hepatocellular viability and function to improve platforms for drug toxicity studies and understanding liver pathophysiology. Here, stereolithography (SLA) was employed to fabricate hydrogel scaffolds with open channels designed for post-seeding and perfused culture of primary hepatocytes that form 3D structures in a bioreactor. Photopolymerizable polyethylene glycol-based hydrogels were fabricated coupled to chemically activated, commercially available filters (polycarbonate and polyvinylidene fluoride) using a chemistry that permitted cell viability, and was robust enough to withstand perfused culture of up to 1 µL/s for at least 7 days. SLA energy dose, photoinitiator concentrations, and pretreatment conditions were screened to determine conditions that maximized cell viability and hydrogel bonding to the filter. Multiple open channel geometries were readily achieved, and included ellipses and rectangles. Rectangular open channels employed for subsequent studies had final dimensions on the order of 350 µm by 850 µm. Cell seeding densities and flow rates that promoted cell viability were determined. Perfused culture of primary hepatocytes in hydrogel scaffolds in the presence of soluble epidermal growth factor (EGF) prolonged the maintenance of albumin production throughout the 7-day culture relative to 2D controls. This technique of bonding hydrogel scaffolds can be employed to fabricate soft scaffolds for a number of bioreactor configurations and applications., National Institutes of Health (U.S.). National Center for Advancing Translational Sciences (5UH2TR000496-02), National Science Foundation (U.S.). Emergent Behaviors of Integrated Cellular Systems, National Science Foundation (U.S.). Integrative Graduate Education and Research Traineeship (Grant 0965918), United States. Defense Advanced Research Projects Agency (BAA-11-73 Microphysiological Systems W911NF-12-2-0039), National Science Foundation (U.S.). Graduate Research Fellowship (Grant DGE-1144245), Massachusetts Institute of Technology. Center for Environmental Health Sciences (National Institutes of Health (U.S.) P30-ES002109)
- Published
- 2016
162. Proteolytic Activity Matrix Analysis (PrAMA) for simultaneous multiple protease activities
- Author
-
Miller, Aaron, Barkal, Layla J., Jeng, Karen, Herrlich, Andreas, Moss, Marcia L., Griffith, Linda G., Lauffenburger, Douglas A., Massachusetts Institute of Technology. Department of Biological Engineering, Whitehead Institute for Biomedical Research, Lauffenburger, Douglas A., Aaron, Miles, Barkal, Layla J., Jeng, Karen, Herrlich, Andreas, and Griffith, Linda G.
- Abstract
Matrix metalloproteinases (MMPs) and A Disintegrin and Metalloproteinases (ADAMs) are two related protease families that play key roles in matrix remodeling and growth factor ligand shedding. Directly ascertaining the proteolytic activities of particular MMPs and ADAMs in physiological environments in a non-invasive, real-time, multiplex manner remains a challenge. This work describes Proteolytic Activity Matrix Analysis (PrAMA), an integrated experimental measurement and mathematical analysis framework for simultaneously determining the activities of particular enzymes in complex mixtures of MMPs and ADAMs. The PrAMA method interprets dynamic signals from panels of moderately specific FRET-based polypeptide protease substrates to deduce a profile of specific MMP and ADAM proteolytic activities. Deconvolution of signals from complex mixtures of proteases is accomplished using prior data on individual MMP/ADAM cleavage signatures for the substrate panel measured with purified enzymes. We first validate PrAMA inference using a compendium of roughly 4000 measurements involving known mixtures of purified enzymes and substrates, and then demonstrate application to the live-cell response of wildtype, ADAM10−/−, and ADAM17−/− fibroblasts to phorbol ester and ionomycin stimulation. Results indicate PrAMA can distinguish closely related enzymes from each other with high accuracy, even in the presence of unknown background proteolytic activity. PrAMA offers a valuable tool for applications ranging from live-cell in vitro assays to high-throughput inhibitor screening with complex enzyme mixtures. Moreover, our approach may extend to other families of proteases, such as caspases and cathepsins, that also can lack highly-specific substrates., Andrew and Edna Viterbi Fellowship in Computational Biology, National Science Foundation (U.S.). Graduate Research Fellowship Program, National Science Foundation (U.S.) (grant 1R01EB010246-01), National Science Foundation (U.S.) (grant 5R01GM081336-02)
- Published
- 2010
163. Integration of Semisynthetic Biomaterials and Nanofabrication to Optimize 3D Liver Cultures
- Author
-
Renggli Kasper, Raredon Micha Sam B, Hammond Paula T, Imperiali Barbara, and Griffith Linda G
- Abstract
A major challenge in the in vitro organogenesis field is to create microscale tissues with intercalated vascular networks similar challenges exist in virtually every tissue type as blood and lymph networks must productively coexist while remaining physically separated by the intervening tissue cells. We address the important challenge of how to create a scaffold that provides both physical and chemical cues to organize liver derived cells to create such integrated vascular networks within a hepatic soft tissue. We use existing 3D microperfused liver models but dramatically expand the ability to create fine tissue architecture by exploiting a very recent scaffold microfabrication approach (projection microstereolithography) to fabricate complex three dimensional microstructures with very high resolution. Cells including hepatocytes in the liver undergo different interactions e.g. with integrins and the local extra cellular matric of the neighboring cells to generate a basal phenotype. Using bioconjugation techniques we functionalize our 3D printed scaffolds with integrin binding peptides and growth factors to render them biologically active. Our approach allows to create the kinds of biomaterials technologies i.e. integration of high resolution scaffold fabrication with practical functionalized materials that will enable entirely new kinds of in vitro tissues to be constructed.
- Published
- 2014
- Full Text
- View/download PDF
164. Design Principles for SuCESsFul Biosensors: Specific Fluorophore/Analyte Binding and Minimization of Fluorophore/Scaffold Interactions
- Author
-
de Picciotto, Seymour, primary, Dickson, Paige M., additional, Traxlmayr, Michael W., additional, Marques, Bryan S., additional, Socher, Elke, additional, Zhao, Sixing, additional, Cheung, Stephanie, additional, Kiefer, Jonathan D., additional, Wand, A. Joshua, additional, Griffith, Linda G., additional, Imperiali, Barbara, additional, and Wittrup, K. Dane, additional
- Published
- 2016
- Full Text
- View/download PDF
165. Modeling Therapeutic Antibody–Small Molecule Drug-Drug Interactions Using a Three-Dimensional Perfusable Human Liver Coculture Platform
- Author
-
Long, Thomas J., primary, Cosgrove, Patrick A., additional, Dunn, Robert T., additional, Stolz, Donna B., additional, Hamadeh, Hisham, additional, Afshari, Cynthia, additional, McBride, Helen, additional, and Griffith, Linda G., additional
- Published
- 2016
- Full Text
- View/download PDF
166. Abstract A41: Elucidating candidate biomarkers of breast cancer progression and dormancy using a 3D model of metastasis
- Author
-
Clark, Amanda M., primary, Wheeler, Sarah E., additional, Young, Carissa L., additional, Pillai, Venkateswaran C., additional, Stolz, Donna B., additional, Lauffenburger, Douglas A., additional, Venkataramanan, Raman, additional, Griffith, Linda G., additional, and Wells, Alan, additional
- Published
- 2016
- Full Text
- View/download PDF
167. Development of Physiologically Relevant Methods for Assessing Inhibition of Glycoprotein Virulence Factors by Small Molecules in Human Pathogen Campylobacter jejuni
- Author
-
Zamora, Cristina Y., primary, De Schutter, Joris W., additional, Chen, Kelly W., additional, Griffith, Linda G., additional, and Imperiali, Barbara, additional
- Published
- 2016
- Full Text
- View/download PDF
168. Abstract PR11: Elucidating candidate biomarkers of breast cancer progression and dormancy using a 3D model of metastasis
- Author
-
Clark, Amanda M., primary, Wheeler, Sarah E., additional, Young, Carissa L., additional, Pillai, Venkateswaran C., additional, Stolz, Donna B., additional, Lauffenburger, Douglas A., additional, Venkataramanan, Raman, additional, Griffith, Linda G., additional, and Wells, Alan, additional
- Published
- 2016
- Full Text
- View/download PDF
169. Reduced Proteolytic Shedding of Receptor Tyrosine Kinases Is a Post-Translational Mechanism of Kinase Inhibitor Resistance
- Author
-
Miller, Miles A., primary, Oudin, Madeleine J., additional, Sullivan, Ryan J., additional, Wang, Stephanie J., additional, Meyer, Aaron S., additional, Im, Hyungsoon, additional, Frederick, Dennie T., additional, Tadros, Jenny, additional, Griffith, Linda G., additional, Lee, Hakho, additional, Weissleder, Ralph, additional, Flaherty, Keith T., additional, Gertler, Frank B., additional, and Lauffenburger, Douglas A., additional
- Published
- 2016
- Full Text
- View/download PDF
170. A mouse-human phase 1 co-clinical trial of a protease-activated fluorescent probe for imaging cancer
- Author
-
Whitley, Melodi Javid, primary, Cardona, Diana M., additional, Lazarides, Alexander L., additional, Spasojevic, Ivan, additional, Ferrer, Jorge M., additional, Cahill, Joan, additional, Lee, Chang-Lung, additional, Snuderl, Matija, additional, Blazer, Dan G., additional, Hwang, E. Shelley, additional, Greenup, Rachel A., additional, Mosca, Paul J., additional, Mito, Jeffrey K., additional, Cuneo, Kyle C., additional, Larrier, Nicole A., additional, O’Reilly, Erin K., additional, Riedel, Richard F., additional, Eward, William C., additional, Strasfeld, David B., additional, Fukumura, Dai, additional, Jain, Rakesh K., additional, Lee, W. David, additional, Griffith, Linda G., additional, Bawendi, Moungi G., additional, Kirsch, David G., additional, and Brigman, Brian E., additional
- Published
- 2016
- Full Text
- View/download PDF
171. Genetically engineering self-organization of human pluripotent stem cells into a liver bud-like tissue using Gata6
- Author
-
Guye, Patrick, primary, Ebrahimkhani, Mohammad R., additional, Kipniss, Nathan, additional, Velazquez, Jeremy J., additional, Schoenfeld, Eldi, additional, Kiani, Samira, additional, Griffith, Linda G., additional, and Weiss, Ron, additional
- Published
- 2016
- Full Text
- View/download PDF
172. The influence of tethered epidermal growth factor on connective tissue progenitor colony formation
- Author
-
Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Marcantonio, Nicholas A., Au, Ada, Griffith, Linda G., Boehm, Cynthia A., Rozic, Richard J., Wells, Alan, Muschler, George F., Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Marcantonio, Nicholas A., Au, Ada, Griffith, Linda G., Boehm, Cynthia A., Rozic, Richard J., Wells, Alan, and Muschler, George F.
- Abstract
Strategies to combine aspirated marrow cells with scaffolds to treat connective tissue defects are gaining increasing clinical attention and use. In situations such as large defects where initial survival and proliferation of transplanted connective tissue progenitors (CTPs) are limiting, therapeutic outcomes might be improved by using the scaffold to deliver growth factors that promote the early stages of cell function in the graft. Signaling by the epidermal growth factor receptor (EGFR) plays a role in cell survival and has been implicated in bone development and homeostasis. Providing epidermal growth factor (EGF) in a scaffold-tethered format may sustain local delivery and shift EGFR signaling to pro-survival modes compared to soluble ligand. We therefore examined the effect of tethered EGF on osteogenic colony formation from human bone marrow aspirates in the context of three different adhesion environments using a total of 39 donors. We found that tethered EGF, but not soluble EGF, increased the numbers of colonies formed regardless of adhesion background, and that tethered EGF did not impair early stages of osteogenic differentiation., National Institute of General Medical Sciences (U.S.) (Grant NIH RO1 AR42997), National Institute of General Medical Sciences (U.S.) (Grant NIH RO1 AG024980), National Institute of General Medical Sciences (U.S.) (Grant NIH RO1 GM59870), National Institute of General Medical Sciences (U.S.) (Grant NIH DE019523)
- Published
- 2015
173. Approaches to in vitro tissue regeneration with application for human disease modeling and drug development
- Author
-
Massachusetts Institute of Technology. Center for Gynepathology Research, Massachusetts Institute of Technology. Department of Biological Engineering, Ebrahimkhani, Mohammad Reza, Young, Carissa L., Lauffenburger, Douglas A., Griffith, Linda G., Borenstein, Jeffrey T., Massachusetts Institute of Technology. Center for Gynepathology Research, Massachusetts Institute of Technology. Department of Biological Engineering, Ebrahimkhani, Mohammad Reza, Young, Carissa L., Lauffenburger, Douglas A., Griffith, Linda G., and Borenstein, Jeffrey T.
- Abstract
Reliable in vitro human disease models that capture the complexity of in vivo tissue behaviors are crucial to gain mechanistic insights into human disease and enable the development of treatments that are effective across broad patient populations. The integration of stem cell technologies, tissue engineering, emerging biomaterials strategies and microfabrication processes, as well as computational and systems biology approaches, is enabling new tools to generate reliable in vitro systems to study the molecular basis of human disease and facilitate drug development. In this review, we discuss these recently developed tools and emphasize opportunities and challenges involved in combining these technologies toward regenerative science., National Institute for Biomedical Imaging and Bioengineering (U.S.) (Grant 5R01EB010246-02), National Center for Advancing Translational Sciences (U.S.) (Grant 1UH2TR000496), United States. Defense Advanced Research Projects Agency (Cooperative Agreement W911NF-12-2-0039)
- Published
- 2015
174. Human mesenchymal stem cells/multipotent stromal cells consume accumulated autophagosomes early in differentiation
- Author
-
Massachusetts Institute of Technology. Department of Biological Engineering, Griffith, Linda G., Nuschke, Austin, Rodrigues, Melanie, Stolz, Donna B, Chu, Charleen T, Wells, Alan, Massachusetts Institute of Technology. Department of Biological Engineering, Griffith, Linda G., Nuschke, Austin, Rodrigues, Melanie, Stolz, Donna B, Chu, Charleen T, and Wells, Alan
- Abstract
Introduction Bone marrow mesenchymal stem cells/multipotent stromal cells (MSCs) are recruited to sites of injury and subsequently support regeneration through differentiation or paracrine activity. During periods of stress such as wound site implant or differentiation, MSCs are subjected to a variety of stressors that might activate pathways to improve cell survival and generate energy. In this study, we monitored MSC autophagy in response to the process of differentiation. Methods MSC autophagosome structures were observed by using transmission electron microscopy and a tandem green fluorescent protein-red fluorescent protein autophagic flux reporter to monitor the mammalian microtubule-associated protein-1 light chain 3 (LC3) turnover in real time. MSCs were differentiated by using standard osteogenic and adipogenic media, and autophagy was examined during short-term and long-term differentiation via immunoblots for LC3I and II. Autophagy was modulated during differentiation by using rapamycin and bafilomycin treatments to disrupt the autophagosome balance during the early stages of the differentiation process, and differentiation was monitored in the long term by using Von Kossa and Oil Red O staining as well as quantitative polymerase chain reaction analysis of typical differentiation markers. Results We found that undifferentiated MSCs showed an accumulation of a large number of undegraded autophagic vacuoles, with little autophagic turnover. Stimulation of autophagy with rapamycin led to rapid degradation of these autophagosomes and greatly increased rough endoplasmic reticulum size. Upon induction of osteogenic differentiation, MSC expression of LC3II, a common autophagosome marker, was lost within 12 hours, consistent with increased turnover. However, during adipogenic differentiation, drug treatment to alter the autophagosome balance during early differentiation led to changes in differentiation efficiency, with inhibited adipocyte formation following rapa, National Institute of General Medical Sciences (U.S.) (Grant GM069668), National Institute of Dental and Craniofacial Research (U.S.) (Grant DE019523), National Institute on Aging (Grant AG026389)
- Published
- 2015
175. Bioreactor technologies to support liver function in vitro
- Author
-
Massachusetts Institute of Technology. Center for Gynepathology Research, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Materials Science and Engineering, Ebrahimkhani, Mohammad Reza, Neiman, Jaclyn A. Shepard, Raredon, Micha Sam Brickman, Griffith, Linda G., Hughes, David J., Massachusetts Institute of Technology. Center for Gynepathology Research, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Materials Science and Engineering, Ebrahimkhani, Mohammad Reza, Neiman, Jaclyn A. Shepard, Raredon, Micha Sam Brickman, Griffith, Linda G., and Hughes, David J.
- Abstract
Liver is a central nexus integrating metabolic and immunologic homeostasis in the human body, and the direct or indirect target of most molecular therapeutics. A wide spectrum of therapeutic and technological needs drives efforts to capture liver physiology and pathophysiology in vitro, ranging from prediction of metabolism and toxicity of small molecule drugs, to understanding off-target effects of proteins, nucleic acid therapies, and targeted therapeutics, to serving as disease models for drug development. Here we provide perspective on the evolving landscape of bioreactor-based models to meet old and new challenges in drug discovery and development, emphasizing design challenges in maintaining long-term liver-specific function and how emerging technologies in biomaterials and microdevices are providing new experimental models., National Institutes of Health (U.S.) (R01 EB010246), National Institutes of Health (U.S.) (P50-GM068762-08), National Institutes of Health (U.S.) (R01-ES015241), National Institutes of Health (U.S.) (P30-ES002109), 5UH2TR000496-02, National Science Foundation (U.S.). Emergent Behaviors of Integrated Cellular Systems (CBET-0939511), United States. Defense Advanced Research Projects Agency. Microphysiological Systems Program (W911NF-12-2-0039)
- Published
- 2015
176. Surface Tethered Epidermal Growth Factor Protects Proliferating and Differentiating Multipotential Stromal Cells from FasL-Induced Apoptosis
- Author
-
Massachusetts Institute of Technology. Department of Biological Engineering, Stockdale, Linda, Griffith, Linda G., Rodrigues, Melanie, Blair, Harry, Wells, Alan, Massachusetts Institute of Technology. Department of Biological Engineering, Stockdale, Linda, Griffith, Linda G., Rodrigues, Melanie, Blair, Harry, and Wells, Alan
- Abstract
Multipotential stromal cells or mesenchymal stem cells (MSCs) have been proposed as aids in regenerating bone and adipose tissues, as these cells form osteoblasts and adipocytes. A major obstacle to this use of MSC is the initial loss of cells postimplantation. This cell death in part is due to ubiquitous nonspecific inflammatory cytokines such as FasL generated in the implant site. Our group previously found that soluble epidermal growth factor (sEGF) promotes MSC expansion. Furthermore, tethering EGF (tEGF) onto a two-dimensional surface altered MSC responses, by restricting epidermal growth factor receptor (EGFR) to the cell surface, causing sustained activation of EGFR, and promoting survival from FasL-induced death. sEGF by causing internalization of EGFR does not support MSC survival. However, for tEGF to be useful in bone regeneration, it needs to allow for MSC differentiation into osteoblasts while also protecting emerging osteoblasts from apoptosis. tEGF did not block induced differentiation of MSCs into osteoblasts, or adipocytes, a common default MSC-differentiation pathway. MSC-derived preosteoblasts showed increased Fas levels and became more susceptible to FasL-induced death, which tEGF prevented. Differentiating adipocytes underwent a reduction in Fas expression and became resistant to FasL-induced death, with tEGF having no further survival effect. tEGF protected undifferentiated MSC from combined insults of FasL, serum deprivation, and physiologic hypoxia. Additionally, tEGF was dominant in the face of sEGF to protect MSC from FasL-induced death. Our results suggest that MSCs and differentiating osteoblasts need protective signals to survive in the inflammatory wound milieu and that tEGF can serve this function., National Institute of General Medical Sciences (U.S.) (GM069668), National Institute of Dental and Craniofacial Research (U.S.) (DE019523)
- Published
- 2015
177. Co-regulation of primary mouse hepatocyte viability and function by oxygen and matrix
- Author
-
Massachusetts Institute of Technology. Department of Biological Engineering, Buck, Lorenna D., Inman, S. Walker, Griffith, Linda G., Rusyn, Ivan, Massachusetts Institute of Technology. Department of Biological Engineering, Buck, Lorenna D., Inman, S. Walker, Griffith, Linda G., and Rusyn, Ivan
- Abstract
Although oxygen and extracellular matrix cues both influence differentiation state and metabolic function of primary rat and human hepatocytes, relatively little is known about how these factors together regulate behaviors of primary mouse hepatocytes in culture. To determine the effects of pericellular oxygen tension on hepatocellular function, we employed two methods of altering oxygen concentration in the local cellular microenvironment of cells cultured in the presence or absence of an extracellular matrix (Matrigel) supplement. By systematically altering medium depth and gas phase oxygen tension, we created multiple oxygen regimes (hypoxic, normoxic, and hyperoxic) and measured the local oxygen concentrations in the pericellular environment using custom-designed oxygen microprobes. From these measurements of oxygen concentrations, we derived values of oxygen consumption rates under a spectrum of environmental contexts, thus providing the first reported estimates of these values for primary mouse hepatocytes. Oxygen tension and matrix microenvironment were found to synergistically regulate hepatocellular survival and function as assessed using quantitative image analysis for cells stained with vital dyes, and assessment of secretion of albumin. Hepatocellular viability was affected only at strongly hypoxic conditions. Surprisingly, albumin secretion rates were greatest at a moderately supra-physiological oxygen concentration, and this effect was mitigated at still greater supra-physiological concentrations. Matrigel enhanced the effects of oxygen on retention of function. This study underscores the importance of carefully controlling cell density, medium depth, and gas phase oxygen, as the effects of these parameters on local pericellular oxygen tension and subsequent hepatocellular function are profound., National Institutes of Health (U.S.) (Grant P50-GM068762-08), National Institutes of Health (U.S.) (Grant R01-EB010246-04), National Institutes of Health (U.S.) (Grant R01-ES015241), National Institutes of Health (U.S.) (Grant P30-ES002109)
- Published
- 2015
178. Synergistic effects of tethered growth factors and adhesion ligands on DNA synthesis and function of primary hepatocytes cultured on soft synthetic hydrogels
- Author
-
Massachusetts Institute of Technology. Center for Environmental Health Sciences, Massachusetts Institute of Technology. Center for Gynepathology Research, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Mehta, Geeta, Williams, Courtney M., Alvarez, Luis, Kamm, Roger Dale, Griffith, Linda G., Lesniewski, Martha, Massachusetts Institute of Technology. Center for Environmental Health Sciences, Massachusetts Institute of Technology. Center for Gynepathology Research, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Mehta, Geeta, Williams, Courtney M., Alvarez, Luis, Kamm, Roger Dale, Griffith, Linda G., and Lesniewski, Martha
- Abstract
The composition, presentation, and spatial orientation of extracellular matrix molecules and growth factors are key regulators of cell behavior. Here, we used self-assembling peptide nanofiber gels as a modular scaffold to investigate how fibronectin-derived adhesion ligands and different modes of epidermal growth factor (EGF) presentation synergistically regulate multiple facets of primary rat hepatocyte behavior in the context of a soft gel. In the presence of soluble EGF, inclusion of dimeric RGD and the heparin binding domain from fibronectin (HB) increased hepatocyte aggregation, spreading, and metabolic function compared to unmodified gels or gels modified with a single motif, but unlike rigid substrates, gels failed to induce DNA synthesis. Tethered EGF dramatically stimulated cell aggregation and spreading under all adhesive ligand conditions and also preserved metabolic function. Surprisingly, tethered EGF elicited DNA synthesis on gels with RGD and HB. Phenotypic differences between soluble and tethered EGF stimulation of cells on peptide gels are correlated with differences in expression and phosphorylation the EGF receptor and its heterodimerization partner ErbB2, and activation of the downstream signaling node ERK1/2. These modular matrices reveal new facets of hepatocellular biology in culture and may be more broadly useful in culture of other soft tissues., United States. Army, Hertz Foundation (Graduate Fellowship), National Institute for Biomedical Imaging and Bioengineering (U.S.) (R01EB003805), National Institute of Dental and Craniofacial Research (U.S.) (R01DE019523), Massachusetts Institute of Technology. Center for Environmental Health Sciences (National Institute of Environmental Health Sciences P30ES002109), Massachusetts Institute of Technology. Center for Environmental Health Sciences (National Institute of Environmental Health Sciences R01ES015241), Armed Forces Institute of Regenerative Medicine
- Published
- 2015
179. The Dormancy Dilemma: Quiescence versus Balanced Proliferation
- Author
-
Massachusetts Institute of Technology. Department of Biological Engineering, Griffith, Linda G., Wells, Alan, Wells, Jakob Z., Taylor, Donald P., Massachusetts Institute of Technology. Department of Biological Engineering, Griffith, Linda G., Wells, Alan, Wells, Jakob Z., and Taylor, Donald P.
- Abstract
Metastatic dissemination with subsequent clinical outgrowth leads to the greatest part of morbidity and mortality from most solid tumors. Even more daunting is that many of these metastatic deposits silently lie undetected, recurring years to decades after primary tumor extirpation by surgery or radiation (termed metastatic dormancy). As primary tumors are frequently curable, a critical focus now turns to preventing the lethal emergence from metastatic dormancy. Current carcinoma treatments include adjuvant therapy intended to kill the cryptic metastatic tumor cells. Because such standard therapies mainly kill cycling cells, this approach carries an implicit assumption that metastatic cells are in the mitogenic cycle. Thus, the pivotal question arises as to whether clinically occult micrometastases survive in a state of balanced proliferation and death, or whether these cells undergo at least long periods of quiescence marked by cell-cycle arrest. The treatment implications are thus obvious—if the carcinoma cells are cycling then therapies should target cycling cells, whereas if cells are quiescent then therapies should either maintain dormancy or be toxic to dormant cells. Because this distinction is paramount to rational therapeutic development and administration, we investigated whether quiescence or balanced proliferation is the most likely etiology underlying metastatic dormancy. We recently published a computer simulation study that determined that balanced proliferation is not the likely driving force and that quiescence most likely participates in metastatic dormancy. As such, a greater emphasis on developing diagnostics and therapeutics for quiescent carcinomas is needed., National Institutes of Health (U.S.). National Center for Advancing Translational Sciences (Grant UH2TR000496)
- Published
- 2015
180. Targeting autocrine HB-EGF signaling with specific ADAM12 inhibition using recombinant ADAM12 prodomain
- Author
-
Massachusetts Institute of Technology. Department of Biological Engineering, Koch Institute for Integrative Cancer Research at MIT, Miller, Miles Aaron, Meyer, Aaron Samuel, Griffith, Linda G., Lauffenburger, Douglas A., Moss, Marcia L., Powell, Gary, Petrovich, Robert, Edwards, Lori, Massachusetts Institute of Technology. Department of Biological Engineering, Koch Institute for Integrative Cancer Research at MIT, Miller, Miles Aaron, Meyer, Aaron Samuel, Griffith, Linda G., Lauffenburger, Douglas A., Moss, Marcia L., Powell, Gary, Petrovich, Robert, and Edwards, Lori
- Abstract
Dysregulation of ErbB-family signaling underlies numerous pathologies and has been therapeutically targeted through inhibiting ErbB-receptors themselves or their cognate ligands. For the latter, “decoy” antibodies have been developed to sequester ligands including heparin-binding epidermal growth factor (HB-EGF); however, demonstrating sufficient efficacy has been difficult. Here, we hypothesized that this strategy depends on properties such as ligand-receptor binding affinity, which varies widely across the known ErbB-family ligands. Guided by computational modeling, we found that high-affinity ligands such as HB-EGF are more difficult to target with decoy antibodies compared to low-affinity ligands such as amphiregulin (AREG). To address this issue, we developed an alternative method for inhibiting HB-EGF activity by targeting its cleavage from the cell surface. In a model of the invasive disease endometriosis, we identified A Disintegrin and Metalloproteinase 12 (ADAM12) as a protease implicated in HB-EGF shedding. We designed a specific inhibitor of ADAM12 based on its recombinant prodomain (PA12), which selectively inhibits ADAM12 but not ADAM10 or ADAM17. In endometriotic cells, PA12 significantly reduced HB-EGF shedding and resultant cellular migration. Overall, specific inhibition of ligand shedding represents a possible alternative to decoy antibodies, especially for ligands such as HB-EGF that exhibit high binding affinity and localized signaling., National Institutes of Health (U.S.) (Grant R01-CA096504), National Institutes of Health (U.S.) (Grant U54-CA112967)
- Published
- 2015
181. Controlling multipotent stromal cell migration by integrating “course-graining” materials and “fine-tuning” small molecules via decision tree signal-response modeling
- Author
-
Massachusetts Institute of Technology. Department of Biological Engineering, Wu, Shan, Griffith, Linda G., Lauffenburger, Douglas A., Wells, Alan, Massachusetts Institute of Technology. Department of Biological Engineering, Wu, Shan, Griffith, Linda G., Lauffenburger, Douglas A., and Wells, Alan
- Abstract
Biomimetic scaffolds have been proposed as a means to facilitate tissue regeneration by multi-potent stromal cells (MSCs). Effective scaffold colonization requires a control of multiple MSC responses including survival, proliferation, differentiation, and migration. As MSC migration is relatively unstudied in this context, we present here a multi-level approach to its understanding and control, integratively tuning cell speed and directional persistence to achieve maximal mean free path (MFP) of migration. This approach employs data-driven computational modeling to ascertain small molecule drug treatments that can enhance MFP on a given materials substratum. Using poly(methyl methacrylate)-graft-poly(ethylene oxide) polymer surfaces tethered with epidermal growth factor (tEGF) and systematically adsorbed with fibronectin, vitronectin, or collagen-I to present hTERT-immortalized human MSCs with growth factor and extracellular matrix cues, we measured cell motility properties along with signaling activities of EGFR, ERK, Akt, and FAK on 19 different substrate conditions. Speed was consistent on collagen/tEGF substrates, but low associated directional persistence limited MFP. Decision tree modeling successfully predicted that ERK inhibition should enhance MFP on collagen/tEGF substrates by increasing persistence. Thus, we demonstrated a two-tiered approach to control MSC migration: materials-based “coarse-graining” complemented by small molecule “fine-tuning”., National Institutes of Health (U.S.) (NIH grant R01-DE019523), National Institutes of Health (U.S.) (NIH Cell Migration Consortium U54-GM064346), National Institutes of Health (U.S.) (NIH grant R01-GM018336), National Institutes of Health (U.S.) (NIH grant R01-DE019523)
- Published
- 2015
182. Evaluation of Osteoconductive Scaffolds in the Canine Femoral Multi-Defect Model
- Author
-
Massachusetts Institute of Technology. Department of Biological Engineering, Stockdale, Linda, Griffith, Linda G., Luangphakdy, Viviane, Walker, Esteban, Shinohara, Kentaro, Pan, Hui, Hefferan, Theresa, Bauer, Thomas W., Saini, Sunil, Dadsetan, Mahrokh, Runge, M. Brett, Vasanji, Amit, Yaszemski, Michael, Muschler, George F., Massachusetts Institute of Technology. Department of Biological Engineering, Stockdale, Linda, Griffith, Linda G., Luangphakdy, Viviane, Walker, Esteban, Shinohara, Kentaro, Pan, Hui, Hefferan, Theresa, Bauer, Thomas W., Saini, Sunil, Dadsetan, Mahrokh, Runge, M. Brett, Vasanji, Amit, Yaszemski, Michael, and Muschler, George F.
- Abstract
Treatment of large segmental bone defects remains an unsolved clinical challenge, despite a wide array of existing bone graft materials. This project was designed to rapidly assess and compare promising biodegradable osteoconductive scaffolds for use in the systematic development of new bone regeneration methodologies that combine scaffolds, sources of osteogenic cells, and bioactive scaffold modifications. Promising biomaterials and scaffold fabrication methods were identified in laboratories at Rutgers, MIT, Integra Life Sciences, and Mayo Clinic. Scaffolds were fabricated from various materials, including poly(L-lactide-co-glycolide) (PLGA), poly(L-lactide-co-ɛ-caprolactone) (PLCL), tyrosine-derived polycarbonate (TyrPC), and poly(propylene fumarate) (PPF). Highly porous three-dimensional (3D) scaffolds were fabricated by 3D printing, laser stereolithography, or solvent casting followed by porogen leaching. The canine femoral multi-defect model was used to systematically compare scaffold performance and enable selection of the most promising substrate(s) on which to add cell sourcing options and bioactive surface modifications. Mineralized cancellous allograft (MCA) was used to provide a comparative reference to the current clinical standard for osteoconductive scaffolds. Percent bone volume within the defect was assessed 4 weeks after implantation using both MicroCT and limited histomorphometry. Bone formed at the periphery of all scaffolds with varying levels of radial ingrowth. MCA produced a rapid and advanced stage of bone formation and remodeling throughout the defect in 4 weeks, greatly exceeding the performance of all polymer scaffolds. Two scaffold constructs, TyrPC[subscript PL]/TCP and PPF4[subscript SLA]/HA[subscript PLGA Dip], proved to be significantly better than alternative PLGA and PLCL scaffolds, justifying further development. MCA remains the current standard for osteoconductive scaffolds., United States. Army Medical Research and Materiel Command (Armed Forces Institute of Regenerative Medicine), United States. Office of Naval Research, United States. Air Force. Office of the Surgeon General, United States. Navy, National Institutes of Health (U.S.), United States. Veterans Administration, Cleveland Clinic Foundation
- Published
- 2015
183. Folding artificial mucosa with cell-laden hydrogels guided by mechanics models.
- Author
-
Hon Fai Chan, Ruike Zhao, Parada, German A., Hu Meng, Leong, Kam W., Griffith, Linda G., and Xuanhe Zhao
- Subjects
RESPIRATORY organs ,GASTROINTESTINAL system ,GENITOURINARY organs ,MUCOUS membranes ,HYDROGELS - Abstract
The surfaces of many hollow or tubular tissues/organs in our respiratory, gastrointestinal, and urogenital tracts are covered by mucosa with folded patterns. The patterns are induced by mechanical instability of the mucosa under compression due to constrained growth. Recapitulating this folding process in vitro will facilitate the understanding and engineering of mucosa in various tissues/organs. However, scant attention has been paid to address the challenge of reproducing mucosal folding. Here we mimic the mucosal folding process using a cell-laden hydrogel film attached to a prestretched tough-hydrogel substrate. The cell-laden hydrogel constitutes a human epithelial cell lining on stromal component to recapitulate the physiological feature of a mucosa. Relaxation of the prestretched tough-hydrogel substrate applies compressive strains on the cellladen hydrogel film, which undergoes mechanical instability and evolves into morphological patterns. We predict the conditions for mucosal folding as well as themorphology of and strain in the folded artificial mucosa using a combination of theory and simulation. The work not only provides a simple method to fold artificial mucosa but also demonstrates a paradigm in tissue engineering via harnessing mechanical instabilities guided by quantitative mechanics models. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
184. Uncharged Helical Modular Polypeptide Hydrogels for Cellular Scaffolds
- Author
-
Ahrens, Caroline C., primary, Welch, M. Elizabeth, additional, Griffith, Linda G., additional, and Hammond, Paula T., additional
- Published
- 2015
- Full Text
- View/download PDF
185. Targeting autocrine HB-EGF signaling with specific ADAM12 inhibition using recombinant ADAM12 prodomain
- Author
-
Miller, Miles A., primary, Moss, Marcia L., additional, Powell, Gary, additional, Petrovich, Robert, additional, Edwards, Lori, additional, Meyer, Aaron S., additional, Griffith, Linda G., additional, and Lauffenburger, Douglas A., additional
- Published
- 2015
- Full Text
- View/download PDF
186. Abstract SY36-03: Intraoperative molecular imaging with protease-activated fluorescent imaging agents
- Author
-
Whitley, Melodi J., primary, Cardona, Diana M., additional, Blazer, Dan G., additional, Hwang, Shelley, additional, Greenup, Rachel A., additional, Mosca, Paul J., additional, Cahill, Joan, additional, Mito, Jeffrey K., additional, Cuneo, Kyle C., additional, Larrier, Nicole, additional, O'Reilly, Erin, additional, Spasojevic, Ivan, additional, Riedel, Richard F., additional, Eward, William C., additional, Griffith, Linda G., additional, Bawendi, Moungi G., additional, Ferrer, Jorge, additional, Strasfeld, David B., additional, Lee, W. David, additional, Brigman, Brian, additional, and Kirsch, David G., additional
- Published
- 2015
- Full Text
- View/download PDF
187. Covalent Modification of Synthetic Hydrogels with Bioactive Proteins via Sortase-Mediated Ligation
- Author
-
Cambria, Elena, primary, Renggli, Kasper, additional, Ahrens, Caroline C., additional, Cook, Christi D., additional, Kroll, Carsten, additional, Krueger, Andrew T., additional, Imperiali, Barbara, additional, and Griffith, Linda G., additional
- Published
- 2015
- Full Text
- View/download PDF
188. Tethering of Epidermal Growth Factor (EGF) to Beta Tricalcium Phosphate (βTCP) via Fusion to a High Affinity, Multimeric βTCP-Binding Peptide: Effects on Human Multipotent Stromal Cells/Connective Tissue Progenitors
- Author
-
Alvarez, Luis M., primary, Rivera, Jaime J., additional, Stockdale, Linda, additional, Saini, Sunil, additional, Lee, Richard T., additional, and Griffith, Linda G., additional
- Published
- 2015
- Full Text
- View/download PDF
189. Abstract P1-07-01: Modeling breast cancer dormancy and re-emergence
- Author
-
Clark, Amanda M, primary, Wheeler, Sarah E, additional, Taylor, Donald P, additional, Young, Carissa L, additional, Pillai, Venkateswaran C, additional, Stolz, Donna B, additional, Venkataramanan, Raman, additional, Lauffenburger, Douglas A, additional, Griffith, Linda G, additional, and Wells, Alan, additional
- Published
- 2015
- Full Text
- View/download PDF
190. Metabolite Profiling and Pharmacokinetic Evaluation of Hydrocortisone in a Perfused Three-Dimensional Human Liver Bioreactor
- Author
-
Sarkar, Ujjal, primary, Rivera-Burgos, Dinelia, additional, Large, Emma M., additional, Hughes, David J., additional, Ravindra, Kodihalli C., additional, Dyer, Rachel L., additional, Ebrahimkhani, Mohammad R., additional, Wishnok, John S., additional, Griffith, Linda G., additional, and Tannenbaum, Steven R., additional
- Published
- 2015
- Full Text
- View/download PDF
191. Photopatterning of hydrogel scaffolds coupled to filter materials using stereolithography for perfused 3D culture of hepatocytes
- Author
-
Neiman, Jaclyn A. Shepard, primary, Raman, Ritu, additional, Chan, Vincent, additional, Rhoads, Mary G., additional, Raredon, Micha Sam B., additional, Velazquez, Jeremy J., additional, Dyer, Rachel L., additional, Bashir, Rashid, additional, Hammond, Paula T., additional, and Griffith, Linda G., additional
- Published
- 2015
- Full Text
- View/download PDF
192. Regenerating the cell resistance of micromolded PEG hydrogels
- Author
-
Heath, Daniel E., primary, Sharif, Abdul Rahim Mohamed, additional, Ng, Chee Ping, additional, Rhoads, Mary G., additional, Griffith, Linda G., additional, Hammond, Paula T., additional, and Chan-Park, Mary B., additional
- Published
- 2015
- Full Text
- View/download PDF
193. Lipids promote survival, proliferation, and maintenance of differentiation of rat liver sinusoidal endothelial cells in vitro
- Author
-
Massachusetts Institute of Technology. Department of Biological Engineering, Griffith, Linda G., Hang, Ta-Chun, Lauffenburger, Douglas A., Stolz, Donna B., Massachusetts Institute of Technology. Department of Biological Engineering, Griffith, Linda G., Hang, Ta-Chun, Lauffenburger, Douglas A., and Stolz, Donna B.
- Abstract
Primary rat liver sinusoidal endothelial cells (LSEC) are difficult to maintain in a differentiated state in culture for scientific studies or technological applications. Relatively little is known about molecular regulatory processes that affect LSEC differentiation because of this inability to maintain cellular viability and proper phenotypic characteristics for extended times in vitro, given that LSEC typically undergo death and detachment around 48–72 h even when treated with VEGF. We demonstrate that particular lipid supplements added to serum-free, VEGF-containing medium increase primary rat liver LSEC viability and maintain differentiation. Addition of a defined lipid combination, or even oleic acid (OA) alone, promotes LSEC survival beyond 72 h and proliferation to confluency. Moreover, assessment of LSEC cultures for endocytic function, CD32b surface expression, and exhibition of fenestrae showed that these differentiation characteristics were maintained when lipids were included in the medium. With respect to the underlying regulatory pathways, we found lipid supplement-enhanced phosphatidylinositol 3-kinase and MAPK signaling to be critical for ensuring LSEC function in a temporally dependent manner. Inhibition of Akt activity before 72 h prevents growth of SEC, whereas MEK inhibition past 72 h prevents survival and proliferation. Our findings indicate that OA and lipids modulate Akt/PKB signaling early in culture to mediate survival, followed by a switch to a dependence on ERK signaling pathways to maintain viability and induce proliferation after 72 h. We conclude that free fatty acids can support maintenance of liver LSEC cultures in vitro; key regulatory pathways involved include early Akt signaling followed by ERK signaling., National Science Foundation (U.S.) (Grant EFRI-0735997), National Institutes of Health (U.S.) (Grant R01 GM069668)
- Published
- 2014
194. In vitro models for liver toxicity testing
- Author
-
Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Griffith, Linda G., Soldatow, Valerie Y., LeCluyse, Edward L., Rusyn, Ivan, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Griffith, Linda G., Soldatow, Valerie Y., LeCluyse, Edward L., and Rusyn, Ivan
- Abstract
Over the years, various liver-derived in vitro model systems have been developed to enable investigation of the potential adverse effects of chemicals and drugs. Liver tissue slices, isolated microsomes, perfused liver, immortalized cell lines, and primary hepatocytes have been used extensively. Immortalized cell lines and primary isolated liver cells are currently the most widely used in vitro models for liver toxicity testing. Limited throughput, loss of viability, and decreases in liver-specific functionality and gene expression are common shortcomings of these models. Recent developments in the field of in vitro hepatotoxicity include three-dimensional tissue constructs and bioartificial livers, co-cultures of various cell types with hepatocytes, and differentiation of stem cells into hepatic lineage-like cells. In an attempt to provide a more physiological environment for cultured liver cells, some of the novel cell culture systems incorporate fluid flow, micro-circulation, and other forms of organotypic microenvironments. Co-cultures aim to preserve liver-specific morphology and functionality beyond those provided by cultures of pure parenchymal cells. Stem cells, both embryonic- and adult tissue-derived, may provide a limitless supply of hepatocytes from multiple individuals to improve reproducibility and enable testing of the individual-specific toxicity. This review describes various traditional and novel in vitro liver models and provides a perspective on the challenges and opportunities afforded by each individual test system., National Institutes of Health (U.S.) (P42 ES005948), National Institutes of Health (U.S.) (R01 ES01524)
- Published
- 2014
195. Perfused multiwell plate for 3D liver tissue engineering
- Author
-
Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Domansky, Karel, Inman, Samuel Walker, Serdy, James George, Dash, Ajit, Griffith, Linda G., Lim, Matthew H. M., Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Domansky, Karel, Inman, Samuel Walker, Serdy, James George, Dash, Ajit, Griffith, Linda G., and Lim, Matthew H. M.
- Abstract
In vitro models that capture the complexity of in vivo tissue and organ behaviors in a scalable and easy-to-use format are desirable for drug discovery. To address this, we have developed a bioreactor that fosters maintenance of 3D tissue cultures under constant perfusion and we have integrated multiple bioreactors into an array in a multiwell plate format. All bioreactors are fluidically isolated from each other. Each bioreactor in the array contains a scaffold that supports formation of hundreds of 3D microscale tissue units. The tissue units are perfused with cell culture medium circulated within the bioreactor by integrated pneumatic diaphragm micropumps. Electronic controls for the pumps are kept outside the incubator and connected to the perfused multiwell by pneumatic lines. The docking design and open-well bioreactor layout make handling perfused multiwell plates similar to using standard multiwell tissue culture plates. A model of oxygen consumption and transport in the circulating culture medium was used to predict appropriate operating parameters for primary liver cultures. Oxygen concentrations at key locations in the system were then measured as a function of flow rate and time after initiation of culture to determine oxygen consumption rates. After seven days of culture, tissue formed from cells seeded in the perfused multiwell reactor remained functionally viable as assessed by immunostaining for hepatocyte and liver sinusoidal endothelial cell (LSEC) phenotypic markers., National Institute of Environmental Health Sciences (grant number 5P30ES002109-30), National Institutes of Health (U.S.) (NIH grant number 5R01ES015241), DuPont MIT Alliance, Pfizer Inc., National Science Foundation (U.S.) (NSF grant number EEC-9843342)
- Published
- 2014
196. Production of Reactive Oxygen Species by Multipotent Stromal Cells/Mesenchymal Stem Cells Upon Exposure to Fas Ligand
- Author
-
Massachusetts Institute of Technology. Department of Biological Engineering, Griffith, Linda G., Rodrigues, Melanie, Turner, Omari, Stolz, Donna, Wells, Alan, Massachusetts Institute of Technology. Department of Biological Engineering, Griffith, Linda G., Rodrigues, Melanie, Turner, Omari, Stolz, Donna, and Wells, Alan
- Abstract
Multipotent stromal cells (MSCs) can be differentiated into osteoblasts and chondrocytes, making these cells candidates to regenerate cranio-facial injuries and lesions in long bones. A major problem with cell replacement therapy, however, is the loss of transplanted MSCs at the site of graft. Reactive oxygen species (ROS) and nonspecific inflammation generated at the ischemic site have been hypothesized to lead to MSCs loss; studies in vitro show MSCs dying both in the presence of ROS or cytokines like FasL. We questioned whether MSCs themselves may be the source of these death inducers, specifically whether MSCs produce ROS under cytokine challenge. On treating MSCs with FasL, we observed increased ROS production within 2 h, leading to apoptotic death after 6 h of exposure to the cytokine. N-acetyl cysteine, an antioxidant, is able to protect MSCs from FasL-induced ROS production and subsequent ROS-dependent apoptosis, though the MSCs eventually succumb to ROS-independent death signaling. Epidermal growth factor (EGF), a cell survival factor, is able to protect cells from FasL-induced ROS production initially; however, the protective effect wanes with continued FasL exposure. In parallel, FasL induces upregulation of the uncoupling protein UCP2, the main uncoupling protein in MSCs, which is not abrogated by EGF; however, the production of ROS is followed by a delayed apoptotic cell death despite moderation by UCP2. FasL-induced ROS activates the stress-induced MAPK pathways JNK and p38MAPK as well as ERK, along with the activation of Bad, a proapoptotic protein, and suppression of survivin, an antiapoptotic protein; the latter two key modulators of the mitochondrial death pathway. FasL by itself also activates its canonical extrinsic death pathway noted by a time-dependent degradation of c-FLIP and activation of caspase 8. These data suggest that MSCs participate in their own demise due to nonspecific inflammation, holding implications for replacement therapies., National Institute of General Medical Sciences (U.S.) (GM069668), National Institute of Dental and Craniofacial Research (U.S.) (DE019523)
- Published
- 2014
197. Micromachined Bioreactor for in Vitro Cell Self-Assembly and 3D Tissue Formation
- Author
-
Domansky, Karel, primary, Sivaraman, Anand, additional, and Griffith, Linda G., additional
- Full Text
- View/download PDF
198. Preparation and Use of Tethered Ligands as Biomaterials and Tools for Cell Biology
- Author
-
Sofia, Susan J., primary, Kuhl, Philip R., additional, and Griffith, Linda G., additional
- Full Text
- View/download PDF
199. Micromachined Bioreactor for in Vitro Cell Self-Assembly and 3D Tissue Formation
- Author
-
Domansky, Karel, primary, Sivaraman, Anand, additional, and Griffith, Linda G., additional
- Full Text
- View/download PDF
200. Human Vascular Tissue Models Formed from Human Induced Pluripotent Stem Cell Derived Endothelial Cells
- Author
-
Belair, David G., primary, Whisler, Jordan A., additional, Valdez, Jorge, additional, Velazquez, Jeremy, additional, Molenda, James A., additional, Vickerman, Vernella, additional, Lewis, Rachel, additional, Daigh, Christine, additional, Hansen, Tyler D., additional, Mann, David A., additional, Thomson, James A., additional, Griffith, Linda G., additional, Kamm, Roger D., additional, Schwartz, Michael P., additional, and Murphy, William L., additional
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.