56 results on '"Griffith, Linda G."'
Search Results
2. The nuclear receptor THRB facilitates differentiation of human PSCs into more mature hepatocytes
- Author
-
Ma, Haiting, de Zwaan, Esmée, Guo, Yang Eric, Cejas, Paloma, Thiru, Prathapan, van de Bunt, Martijn, Jeppesen, Jacob F., Syamala, Sudeepa, Dall’Agnese, Alessandra, Abraham, Brian J., Fu, Dongdong, Garrett-Engele, Carrie, Lee, Tony, Long, Henry W, Griffith, Linda G., Young, Richard A., and Jaenisch, Rudolf
- Subjects
Mice ,Hepatocytes ,Genetics ,Animals ,Humans ,Receptors, Cytoplasmic and Nuclear ,Molecular Medicine ,Cell Differentiation ,Cell Biology ,Regulatory Sequences, Nucleic Acid ,Polymorphism, Single Nucleotide ,Article ,Chromatin - Abstract
To understand the mechanisms regulating the in vitro maturation of hPSC-derived hepatocytes, we developed a 3D differentiation system and compared gene regulatory elements in human primary hepatocytes with those in hPSC-hepatocytes that were differentiated in 2D or 3D conditions by RNA-seq, ATAC-seq, and H3K27Ac ChIP-seq. Regulome comparisons showed a reduced enrichment of thyroid receptor THRB motifs in accessible chromatin and active enhancers without a reduced transcription of THRB. The addition of thyroid hormone T3 increased the binding of THRB to the CYP3A4 proximal enhancer, restored the super-enhancer status and gene expression of NFIC, and reduced the expression of AFP. The resultant hPSC-hepatocytes showed gene expression, epigenetic status, and super-enhancer landscape closer to primary hepatocytes and activated regulatory regions including non-coding SNPs associated with liver-related diseases. Transplanting the hPSC-hepatocytes resulted in the engraftment of human hepatocytes into the mouse liver without disrupting normal liver histology. This work implicates the environmental factor-nuclear receptor axis in regulating the maturation of hPSC-hepatocytes.
- Published
- 2022
- Full Text
- View/download PDF
3. Biology-inspired microphysiological systems to advance patient benefit and animal welfare in drug development
- Author
-
Marx, Uwe, Akabane, Takafumi, Andersson, Tommy B., Baker, Elizabeth, Beilmann, Mario, Beken, Sonja, Brendler-Schwaab, Susanne, Cirit, Murat, David, Rhiannon, Dehne, Eva-Maria, Durieux, Isabell, Ewart, Lorna, Fitzpatrick, Suzanne C., Frey, Olivier, Fuchs, Florian, Griffith, Linda G., Hamilton, Geraldine A., Hartung, Thomas, Hoeng, Julia, Hogberg, Helena, Hughes, David J., Ingber, Donald E., Iskandar, Anita, Kanamori, Toshiyuki, Kojima, Hajime, Kuehnl, Jochen, Leist, Marcel, Li, Bo, Loskill, Peter, Mendrick, Donna L., Neumann, Thomas, Pallocca, Giorgia, Rusyn, Ivan, Smirnova, Lena, Steger-Hartmann, Thomas, Tagle, Danilo A., Tonevitsky, Alexander, Tsyb, Sergej, Trapecar, Martin, van de Water, Bob, van den Eijnden-van Raaij, Janny, Vulto, Paul, Watanabe, Kengo, Wolf, Armin, Zhou, Xiaobing, Roth, Adrian, and Publica
- Subjects
Drug Development ,Drug Industry ,Lab-On-A-Chip Devices ,ddc:570 ,Drug Evaluation, Preclinical ,Animals ,Humans ,Animal Testing Alternatives ,Animal Welfare ,Models, Biological ,Article - Abstract
The first microfluidic microphysiological systems (MPS) entered the academic scene more than 15 years ago and were considered an enabling technology to human (patho)biology in vitro and, therefore, provide alternative approaches to laboratory animals in pharmaceutical drug development and academic research. Nowadays, the field generates more than a thousand scientific publications per year. Despite the MPS hype in academia and by platform providers, which says this technology is about to reshape the entire in vitro culture landscape in basic and applied research, MPS approaches have neither been widely adopted by the pharmaceutical industry yet nor reached regulated drug authorization processes at all.Here, 46 leading experts from all stakeholders - academia, MPS supplier industry, pharmaceutical and consumer products industries, and leading regulatory agencies - worldwide have analyzed existing challenges and hurdles along the MPS-based assay life cycle in a second workshop of this kind in June 2019. They identified that the level of qualification of MPS-based assays for a given context of use and a communication gap between stakeholders are the major challenges for industrial adoption by end-users. Finally, a regulatory acceptance dilemma exists against that background. This t4 report elaborates on these findings in detail and summarizes solutions how to overcome the roadblocks. It provides recommendations and a roadmap towards regulatory accepted MPS-based models and assays for patients' benefit and further laboratory animal reduction in drug development. Finally, experts highlighted the potential of MPS-based human disease models to feedback into laboratory animal replacement in basic life science research. published
- Published
- 2020
4. OrgaQuant: Intestinal Organoid Localization and Quantification Using Deep Convolutional Neural Networks
- Author
-
Kassis, Timothy, Hernandez-Gordillo, Victor, Langer, Ronit, and Griffith, Linda G.
- Subjects
Artificial neural network ,business.industry ,Computer science ,Intestinal organoids ,Pattern recognition ,Convolutional neural network ,Pipeline (software) ,Object detection ,In vitro ,Cell culture ,Native tissue ,Extracellular ,Organoid ,Artificial intelligence ,business - Abstract
Organoid cultures are proving to be powerful in vitro models that closely mimic the cellular constituents of their native tissue. The organoids are typically expanded and cultured in a 3D environment using either naturally derived or synthetic extracellular matrices. Assessing the morphology and growth characteristics of these cultures has been difficult due to the many imaging artifacts that accompany the corresponding images. Unlike single cell cultures, there are no reliable segmentation techniques that allow for the localization and quantification of organoids in their 3D culture environment. Here we describe OrgaQuant, a deep convolutional neural network implementation that can locate and quantify the size distribution of intestinal organoids in brightfield images. OrgaQuant is an end-to-end trained neural network that requires no parameter tweaking, thus it can be fully automated to analyze thousands of images with no user intervention. To develop OrgaQuant we created a unique dataset of manually annotated intestinal organoid images and trained an object detection pipeline using TensorFlow. We have made the dataset, trained model and inference scripts publically available along with detailed usage instructions.
- Published
- 2018
- Full Text
- View/download PDF
5. Metabolite profiling and pharmacokinetic evaluation of hydrocortisone in a perfused 3D human liver bioreactor
- Author
-
Sarkar, Ujjal, Rivera-Burgos, Dinelia, Large, Emma M., Hughes, David J., Kodihalli, Ravindra, Dyer, Rachel Lee, Ebrahimkhani, Mohammad Reza, Wishnok, John S., Griffith, Linda G., Tannenbaum, Steven Robert, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Chemistry, Massachusetts Institute of Technology. Department of Mechanical Engineering, Sarkar, Ujjal, Rivera-Burgos, Dinelia, Kodihalli, Ravindra, Dyer, Rachel Lee, Ebrahimkhani, Mohammad Reza, Wishnok, John S., Griffith, Linda G., and Tannenbaum, Steven Robert
- Abstract
Endotoxin lipopolysaccharide (LPS) is known to cause liver injury primarily involving inflammatory cells such as Kupffer cells, but few in vitro culture models are applicable for investigation of inflammatory effects on drug metabolism. We have developed a 3D human microphysiological hepatocyte-Kupffer-cell coculture system and evaluated the anti-inflammatory effect of glucocorticoids on liver cultures. LPS was introduced to the cultures to elicit an inflammatory response and assessed by the release of pro-inflammatory cytokines, IL6 and TNFα. A sensitive and specific RP-UHPLC-QTOF-MS method was used to evaluate hydrocortisone disappearance and metabolism at near physiological levels. For this, the systems were dosed with 100 nM hydrocortisone and circulated for two days; hydrocortisone was depleted to approximately 30 nM, with first-order kinetics. Phase I metabolites, including tetrahydrocortisone and dihydrocortisol, accounted for 8-10 % of the loss, and 45-52 % was phase II metabolites, including glucuronides of tetrahydrocortisol and tetrahydrocortisone. Pharmacokinetic parameters, i.e., half-life (t1/2), rate of elimination (kel), clearance (CL), and area under the curve (AUC), were 23.03 h, 0.03 h-1, 6.6x10-5 L. h-1 and 1.03 mg/L*h respectively. The ability of the bioreactor to predict the in vivo clearance of hydrocortisone was characterized and the obtained intrinsic clearance values correlated with human data. This system offers a physiologically-relevant tool for investigating hepatic function in an inflamed liver. Endotoxin lipopolysaccharide (LPS) is known to cause liver injury primarily involving inflammatory cells such as Kupffer cells, but few in vitro culture models are applicable for investigation of inflammatory effects on drug metabolism. We have developed a 3D human microphysiological hepatocyte-Kupffer-cell coculture system and evaluated the anti-inflammatory effect of glucocorticoids on liver cultures. LPS was introduced to the cultures to elicit an inflammatory response and assessed by the release of pro-inflammatory cytokines, IL6 and TNFα. A sensitive and specific RP-UHPLC-QTOF-MS method was used to evaluate hydrocortisone disappearance and metabolism at near physiological levels. For this, the systems were dosed with 100 nM hydrocortisone and circulated for two days; hydrocortisone was depleted to approximately 30 nM, with first-order kinetics. Phase I metabolites, including tetrahydrocortisone and dihydrocortisol, accounted for 8-10 % of the loss, and 45-52 % was phase II metabolites, including glucuronides of tetrahydrocortisol and tetrahydrocortisone. Pharmacokinetic parameters, i.e., half-life (t[subscript 1/2]), rate of elimination (k[subscript el]), clearance (CL), and area under the curve (AUC), were 23.03 h, 0.03 h[superscript -1], 6.6x10[superscript -5] L. h-1 and 1.03 mg/L*h respectively. The ability of the bioreactor to predict the in vivo clearance of hydrocortisone was characterized and the obtained intrinsic clearance values correlated with human data. This system offers a physiologically-relevant tool for investigating hepatic function in an inflamed liver., United States. Defense Advanced Research Projects Agency (DARPA-BAA-11-73 Microphysiological Systems W911NF-12-2-0039), National Institutes of Health (U.S.) (5-UH2-TR000496), Massachusetts Institute of Technology. Center for Environmental Health Sciences (P30-ES002109)
- Published
- 2015
6. Photopatterning of hydrogel scaffolds coupled to filter materials using stereolithography for perfused 3D culture of hepatocytes
- Author
-
Neiman, Jaclyn A. Shepard, Raman, Ritu, Chan, Vincent, Rhoads, Mary G., Velazquez, Jeremy J., Bashir, Rashid, Hammond, Paula T., Griffith, Linda G., Raredon, Micha Sam Brickman, Dyer, Rachel Lee, 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., and Griffith, Linda G.
- Subjects
technology, industry, and agriculture - 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
- 2014
7. 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
8. 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
9. 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
10. 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
11. 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
12. Liver ‘organ on a chip’
- Author
-
Linda G. Griffith, Colin H. Beckwitt, D. Lansing Taylor, Alan Wells, Sarah E Wheeler, Amanda M. Clark, Donna B. Stolz, Massachusetts Institute of Technology. Department of Biological Engineering, and Griffith, Linda G
- Subjects
0301 basic medicine ,Tissue Engineering ,Microfluidics ,Cell Biology ,Human physiology ,Biology ,Models, Biological ,Organ-on-a-chip ,Article ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Liver ,Tissue engineering ,Lab-On-A-Chip Devices ,030220 oncology & carcinogenesis ,Liver tissue ,Hepatocytes ,Animals ,Humans ,Homeostasis - Abstract
© 2017 The liver plays critical roles in both homeostasis and pathology. It is the major site of drug metabolism in the body and, as such, a common target for drug-induced toxicity and is susceptible to a wide range of diseases. In contrast to other solid organs, the liver possesses the unique ability to regenerate. The physiological importance and plasticity of this organ make it a crucial system of study to better understand human physiology, disease, and response to exogenous compounds. These aspects have impelled many to develop liver tissue systems for study in isolation outside the body. Herein, we discuss these biologically engineered organoids and microphysiological systems. Keywords: Microphysiologic systems; Organoids; 3D culture systems, National Institutes of Health (U.S.) (Grant UH3TR000496), National Institutes of Health (U.S.) (Grant UH3TR000503)
- Published
- 2018
- Full Text
- View/download PDF
13. ADAM10 Sheddase Activity is a Potential Lung-Cancer Biomarker
- Author
-
Linda G. Griffith, Michael C. Gorry, Miles A. Miller, Nikola L. Vujanovic, Autumn Gaither-Davis, Marcia L. Moss, Lazar Vujanovic, Yan Lin, James G. Herman, Andrea Sobo-Vujanovic, Douglas A. Lauffenburger, Laura P. Stabile, Toshie Yoneyama, Massachusetts Institute of Technology. Biotechnology Process Engineering Center, 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 Mechanical Engineering, Miller, Miles Aaron, Griffith, Linda G, and Lauffenburger, Douglas A
- Subjects
0301 basic medicine ,Sheddase activity ,ADAM10 ,Cell ,Tumor tissue ,03 medical and health sciences ,0302 clinical medicine ,Lysate ,Blood exosomes ,Medicine ,Lung cancer ,ADAM17 ,Cancer biomarker ,business.industry ,Cancer ,Sheddase ,Fluorogenic peptide substrate ,medicine.disease ,Microvesicles ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Proteolytic activity matrix analysis ,030220 oncology & carcinogenesis ,Cancer research ,Biomarker (medicine) ,Cancer biomarkers ,business ,Research Paper - Abstract
Background: Increases in expression of ADAM10 and ADAM17 genes and proteins are inconsistently found in cancer lesions, and are not validated as clinically useful biomarkers. The enzyme-specific proteolytic activities, which are solely mediated by the active mature enzymes, directly reflect enzyme cellular functions and might be superior biomarkers than the enzyme gene or protein expressions, which comprise the inactive proenzymes and active and inactivated mature enzymes. Methods: Using a recent modification of the proteolytic activity matrix analysis (PrAMA) measuring specific enzyme activities in cell and tissue lysates, we examined the specific sheddase activities of ADAM10 (ADAM10sa) and ADAM17 (ADAM17sa) in human non-small cell lung-carcinoma (NSCLC) cell lines, patient primary tumors and blood exosomes, and the noncancerous counterparts. Results: NSCLC cell lines and patient tumors and exosomes consistently showed significant increases of ADAM10sa relative to their normal, inflammatory and/or benign-tumor controls. Additionally, stage IA-IIB NSCLC primary tumors of patients who died of the disease exhibited greater increases of ADAM10sa than those of patients who survived 5 years following diagnosis and surgery. In contrast, NSCLC cell lines and patient tumors and exosomes did not display increases of ADAM17sa. Conclusions: This study is the first to investigate enzyme-specific proteolytic activities as potential cancer biomarkers. It provides a proof-of-concept that ADAM10sa could be a biomarker for NSCLC early detection and outcome prediction. To ascertain that ADAM10sa is a useful cancer biomarker, further robust clinical validation studies are needed., National Institutes of Health (U.S.) (R01 CA96504)
- Published
- 2018
- Full Text
- View/download PDF
14. On-demand dissolution of modular, synthetic extracellular matrix reveals local epithelial-stromal communication networks
- Author
-
Douglas A. Lauffenburger, Forest M. White, Linda G. Griffith, Kasper Renggli, Manu P. Kumar, Daniel A. Rothenberg, Linda Stockdale, Elizabeth A. Gordon, Jorge Valdez, Alex J. Wang, Christi D. Cook, Alexander Brown, Caroline C. Ahrens, Massachusetts Institute of Technology. Biotechnology Process Engineering Center, 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 Mechanical Engineering, Valdez Macias, Jorge Luis, Cook, Christi Dionne, Wang, Alex J-S, Brown, Alexander Thomas, Kumar, Manu Prajapati, Stockdale, Linda, Rothenberg, Daniel Abram, Renggli-Frey, Kasper, Gordon, Elizabeth A, Lauffenburger, Douglas A, White, Forest M, and Griffith, Linda G
- Subjects
0301 basic medicine ,Materials science ,Stromal cell ,medicine.medical_treatment ,Interleukin-1beta ,Biophysics ,Bioengineering ,Cell Communication ,02 engineering and technology ,Epithelial-Stromal Communication ,Hydrogel, Polyethylene Glycol Dimethacrylate ,Article ,Biomaterials ,Extracellular matrix ,03 medical and health sciences ,Paracrine signalling ,Bacterial Proteins ,Sortase ,Cell Line, Tumor ,medicine ,Humans ,Amino Acid Sequence ,Epithelial Cells ,Aminoacyltransferases ,021001 nanoscience & nanotechnology ,Coculture Techniques ,Extracellular Matrix ,Cell biology ,Cysteine Endopeptidases ,Kinetics ,030104 developmental biology ,Cytokine ,Solubility ,Biochemistry ,Mechanics of Materials ,Sortase A ,Self-healing hydrogels ,Ceramics and Composites ,Intercellular Signaling Peptides and Proteins ,Inflammation Mediators ,Stromal Cells ,Peptides ,0210 nano-technology - Abstract
Methods to parse paracrine epithelial-stromal communication networks are a vital need in drug development, as disruption of these networks underlies diseases ranging from cancer to endometriosis. Here, we describe a modular, synthetic, and dissolvable extracellular matrix (MSD-ECM) hydrogel that fosters functional 3D epithelial-stromal co-culture, and that can be dissolved on-demand to recover cells and paracrine signaling proteins intact for subsequent analysis. Specifically, synthetic polymer hydrogels, modified with cell-interacting adhesion motifs and crosslinked with peptides that include a substrate for cell-mediated proteolytic remodeling, can be rapidly dissolved by an engineered version of the microbial transpeptidase Sortase A (SrtA) if the crosslinking peptide includes a SrtA substrate motif and a soluble second substrate. SrtA-mediated dissolution affected only 1 of 31 cytokines and growth factors assayed, whereas standard protease degradation methods destroyed about half of these same molecules. Using co-encapsulated endometrial epithelial and stromal cells as one model system, we show that the dynamic cytokine and growth factor response of co-cultures to an inflammatory cue is richer and more nuanced when measured from SrtA-dissolved gel microenvironments than from the culture supernate. This system employs accessible, reproducible reagents and facile protocols; hence, has potential as a tool in identifying and validating therapeutic targets in complex diseases., National Institutes of Health (U.S.) (R01EB010246), National Institutes of Health (U.S.) (UH2TR000496), Institute for Collaborative Biotechnologies (W911NF-09-0001), National Institutes of Health (U.S.) (T32GM008334), United States. Defense Advanced Research Projects Agency. Microphysiological Systems Program (W911NF-12-2-0039), John and Karinne Begg Fund, Begg New Horizon Fund for Undergraduate Research at MIT, Massachusetts Institute of Technology. Biophysical Instrumentation Facility, Manton Foundation, Ludwig Postdoctoral Fellowship for Cancer Research, Swiss National Science Foundation (Postdoctoral Fellowship)
- Published
- 2017
- Full Text
- View/download PDF
15. Multi-functional scaling methodology for translational pharmacokinetic and pharmacodynamic applications using integrated microphysiological systems (MPS)
- Author
-
Murat Cirit, Cynthia L. Stokes, Linda G. Griffith, Christian Maass, Massachusetts Institute of Technology. Biotechnology Process Engineering Center, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Maass, Christian Alexander, Griffith, Linda G, and Cirit, Murat
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Computer science ,MathematicsofComputing_NUMERICALANALYSIS ,Cell Culture Techniques ,Drug Evaluation, Preclinical ,Biophysics ,Pharmacology ,Kidney ,Models, Biological ,Biochemistry ,Article ,Translational Research, Biomedical ,03 medical and health sciences ,Pharmacokinetics ,In vivo ,Animals ,Humans ,Intestinal Mucosa ,skin and connective tissue diseases ,Scaling ,Dose-Response Relationship, Drug ,nutritional and metabolic diseases ,Kidney metabolism ,Intestines ,030104 developmental biology ,Liver ,Scale (social sciences) ,Pharmacodynamics ,Systems design ,Short exposure ,Biological system - Abstract
Microphysiological systems (MPS) provide relevant physiological environments in vitro for studies of pharmacokinetics, pharmacodynamics and biological mechanisms for translational research. Designing multi-MPS platforms is essential to study multi-organ systems. Typical design approaches, including direct and allometric scaling, scale each MPS individually and are based on relative sizes not function. This study's aim was to develop a new multi-functional scaling approach for integrated multi-MPS platform design for specific applications. We developed an optimization approach using mechanistic modeling and specification of an objective that considered multiple MPS functions, e.g., drug absorption and metabolism, simultaneously to identify system design parameters. This approach informed the design of two hypothetical multi-MPS platforms consisting of gut and liver (multi-MPS platform I) and gut, liver and kidney (multi-MPS platform II) to recapitulate in vivo drug exposures in vitro. This allows establishment of clinically relevant drug exposure-response relationships, a prerequisite for efficacy and toxicology assessment. Design parameters resulting from multi-functional scaling were compared to designs based on direct and allometric scaling. Human plasma time-concentration profiles of eight drugs were used to inform the designs, and profiles of an additional five drugs were calculated to test the designed platforms on an independent set. Multi-functional scaling yielded exposure times in good agreement with in vivo data, while direct and allometric scaling approaches resulted in short exposure durations. Multi-functional scaling allows appropriate scaling from in vivo to in vitro of multi-MPS platforms, and in the cases studied provides designs that better mimic in vivo exposures than standard MPS scaling methods., United States. Defense Advanced Research Projects Agency. Microphysiological Systems Program (W911NF-12-2-0039), National Institutes of Health (U.S.). Microphysiological Systems Program (4-UH3-TR000496-03)
- Published
- 2017
- Full Text
- View/download PDF
16. Quantitative Assessment of Population Variability in Hepatic Drug Metabolism Using a Perfused Three-Dimensional Human Liver Microphysiological System
- Author
-
CL Stokes, David J. Hughes, Tomasz Kostrzewski, Linda G. Griffith, Nikolaos Tsamandouras, Murat Cirit, Massachusetts Institute of Technology. Biotechnology Process Engineering Center, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Tsamandouras, Nikolaos, Griffith, Linda G, and Cirit, Murat
- Subjects
0301 basic medicine ,Metabolite ,Population ,Context (language use) ,Pharmacology ,Biology ,030226 pharmacology & pharmacy ,Metabolism, Transport, and Pharmacogenomics ,Tissue Culture Techniques ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cytochrome P-450 Enzyme System ,Pharmacokinetics ,In vivo ,Lactate dehydrogenase ,medicine ,Humans ,Tissue Distribution ,education ,Serum Albumin ,Cryopreservation ,education.field_of_study ,L-Lactate Dehydrogenase ,3. Good health ,Perfusion ,Phenotype ,030104 developmental biology ,Liver ,Pharmaceutical Preparations ,chemistry ,Phenacetin ,Hepatocytes ,Molecular Medicine ,Drug metabolism ,medicine.drug - Abstract
In this work, we first describe the population variability in hepatic drug metabolism using cryopreserved hepatocytes from five different donors cultured in a perfused three-dimensional human liver microphysiological system, and then show how the resulting data can be integrated with a modeling and simulation framework to accomplish in vitro-in vivo translation. For each donor, metabolic depletion profiles of six compounds (phenacetin, diclofenac, lidocaine, ibuprofen, propranolol, and prednisolone) were measured, along with metabolite formation, mRNA levels of 90 metabolism-related genes, and markers of functional viability [lactate dehydrogenase (LDH) release, albumin, and urea production]. Drug depletion data were analyzed with mixed-effects modeling. Substantial interdonor variability was observed with respect to gene expression levels, drug metabolism, and other measured hepatocyte functions. Specifically, interdonor variability in intrinsic metabolic clearance ranged from 24.1% for phenacetin to 66.8% for propranolol (expressed as coefficient of variation). Albumin, urea, LDH, and cytochrome P450 mRNA levels were identified as significant predictors of in vitro metabolic clearance. Predicted clearance values from the liver microphysiological system were correlated with the observed in vivo values. A population physiologically based pharmacokinetic model was developed for lidocaine to illustrate the translation of the in vitro output to the observed pharmacokinetic variability in vivo. Stochastic simulations with this model successfully predicted the observed clinical concentration-time profiles and the associated population variability. This is the first study of population variability in drug metabolism in the context of a microphysiological system and has important implications for the use of these systems during the drug development process., United States. Defense Advanced Research Projects Agency. Microphysiological Systems Program (Grant W911NF-12-2-0039), National Institutes of Health (U.S.). Microphysiological Systems Program (Grant 4-UH3-TR000496-03)
- Published
- 2016
- Full Text
- View/download PDF
17. Establishing quasi-steady state operations of microphysiological systems (MPS) using tissue-specific metabolic dependencies
- Author
-
Jorge Valdez, Cynthia L. Stokes, Matthew Dallas, Emily Geishecker, Michael Shockley, Linda G. Griffith, Matthew E. LaBarge, Murat Cirit, Christian Maass, Massachusetts Institute of Technology. Biotechnology Process Engineering Center, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Maass, Christian Alexander, LaBarge, Matthew E, Shockley, Michael J, Valdez Macias, Jorge Luis, Geishecker, Emily R, Griffith, Linda G, and Cirit, Murat
- Subjects
0301 basic medicine ,Biochemical Phenomena ,Computer science ,Science ,Systems biology ,Induced Pluripotent Stem Cells ,Cell Culture Techniques ,Computational biology ,Article ,03 medical and health sciences ,Nutrient ,Humans ,Tissue specific ,Computer Simulation ,Myocytes, Cardiac ,Lactic Acid ,Cells, Cultured ,Ecosystem ,Multidisciplinary ,Extramural ,Systems Biology ,Microfluidic Analytical Techniques ,In vitro ,Culture Media ,Intestines ,Systems Integration ,Glucose ,030104 developmental biology ,Liver ,Organ Specificity ,Microtechnology ,Medicine ,Monitoring glucose ,Caco-2 Cells ,Energy Metabolism ,HT29 Cells - Abstract
Microphysiological systems (MPS), consisting of tissue constructs, biomaterials, and culture media, aim to recapitulate relevant organ functions in vitro. MPS components are housed in fluidic hardware with operational protocols, such as periodic complete media replacement. Such batch-like operations provide relevant nutrients and remove waste products but also reset cell-secreted mediators (e.g. cytokines, hormones) and potentially limit exposure to drugs (and metabolites). While each component plays an essential role for tissue functionality, MPS-specific nutrient needs are not yet well-characterized nor utilized to operate MPSs at more physiologically-relevant conditions. MPS-specific nutrient needs for gut (immortalized cancer cells), liver (human primary hepatocytes) and cardiac (iPSC-derived cardiomyocytes) MPSs were experimentally quantified. In a long-term study of the gut MPS (10 days), this knowledge was used to design operational protocols to maintain glucose and lactate at desired levels. This quasi-steady state operation was experimentally validated by monitoring glucose and lactate as well as MPS functionality. In a theoretical study, nutrient needs of an integrated multi-MPS platform (gut, liver, cardiac MPSs) were computationally simulated to identify long-term quasi-steady state operations. This integrative experimental and computational approach demonstrates the utilization of quantitative multi-scale characterization of MPSs and incorporating MPS-specific information to establish more physiologically-relevant experimental operations., United States. Defense Advanced Research Projects Agency. Microphysiological Systems Program (W911NF-12-2-0039), National Institutes of Health (U.S.) (U24TR001951), National Institutes of Health (U.S.). Microphysiological Systems Program (4-UH3-TR000496-03)
- Published
- 2018
- Full Text
- View/download PDF
18. Quantitative Systems Pharmacology Approaches Applied to Microphysiological Systems (MPS): Data Interpretation and Multi-MPS Integration
- Author
-
Emma M. Large, Ujjal Sarkar, John S. Wishnok, Nicholas A. Cilfone, CL Stokes, Douglas A. Lauffenburger, Linda G. Griffith, Jiajie Yu, Steven R. Tannenbaum, Murat Cirit, David J. Hughes, 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 Chemistry, Massachusetts Institute of Technology. School of Engineering, Yu, Jiajie, Cilfone, Nicholas A., Sarkar, Ujjal, Wishnok, John S, Tannenbaum, Steven R, Lauffenburger, Douglas A, Griffith, Linda G, and Cirit, Murat
- Subjects
Engineering ,congenital, hereditary, and neonatal diseases and abnormalities ,Drug discovery ,business.industry ,Data interpretation ,nutritional and metabolic diseases ,Computational biology ,Original Articles ,computer.software_genre ,Interactome ,3. Good health ,Pharmacological Concepts ,Modeling and Simulation ,Pharmacology (medical) ,Data mining ,business ,skin and connective tissue diseases ,computer ,Systems pharmacology - Abstract
Our goal in developing Microphysiological Systems (MPS) technology is to provide an improved approach for more predictive preclinical drug discovery via a highly integrated experimental/computational paradigm. Success will require quantitative characterization of MPSs and mechanistic analysis of experimental findings sufficient to translate resulting insights from in vitro to in vivo. We describe herein a systems pharmacology approach to MPS development and utilization that incorporates more mechanistic detail than traditional pharmacokinetic/pharmacodynamic (PK/PD) models. A series of studies illustrates diverse facets of our approach. First, we demonstrate two case studies: a PK data analysis and an inflammation response––focused on a single MPS, the liver/immune MPS. Building on the single MPS modeling, a theoretical investigation of a four-MPS interactome then provides a quantitative way to consider several pharmacological concepts such as absorption, distribution, metabolism, and excretion in the design of multi-MPS interactome operation and experiments., United States. Defense Advanced Research Projects Agency. Microphysiological Systems Program (W911NF-12-2-0039), National Institutes of Health (U.S.) Microphysiological Systems Program (4-UH3-TR000496-03), Massachusetts Institute of Technology. Center for Environmental Health Sciences (NIEHS Grant P30-ES002109)
- Published
- 2015
19. PiFlow: A Biocompatible Low-Cost Programmable Dynamic Flow Pumping System Utilizing a Raspberry Pi Zero and Commercial Piezoelectric Pumps
- Author
-
Timothy Kassis, Chloe J. W. Yang, Linda G. Griffith, Paola M. Perez, Luis R. Soenksen, David L. Trumper, Massachusetts Institute of Technology. Biotechnology Process Engineering Center, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Massachusetts Institute of Technology. Research Laboratory of Electronics, Kassis, Timothy, Perez, Paola M., Yang, Chloe J., Soenksen Martinez, Luis Ruben, Trumper, David L, and Griffith, Linda G
- Subjects
0301 basic medicine ,Materials science ,Computer science ,Biomedical Engineering ,Peristaltic pump ,Micromixer ,Industrial and Manufacturing Engineering ,Raspberry pi ,03 medical and health sciences ,Fluid dynamics ,lcsh:Science (General) ,Instrumentation ,Civil and Structural Engineering ,business.industry ,Mechanical Engineering ,Electrical engineering ,Zero (complex analysis) ,Biocompatible material ,Piezoelectricity ,Volumetric flow rate ,Microcontroller ,030104 developmental biology ,Flow (mathematics) ,business ,Computer hardware ,lcsh:Q1-390 - Abstract
With the rise of research utilizing microphysiological systems (MPSs), the need for tools that enable the physiological mimicking of the relevant cellular environment is vital. The limited ability to reproduce crucial features of the microenvironment, such as surrounding fluid flow and dynamic changes in biochemical stimuli, severely limits the types of experiments that can be carried out. Current equipment to achieve this, such as syringe and peristaltic pumps, is expensive, large, difficult to program and has limited potential for scalability. Here, we present a new pumping platform that is open-source, low-cost, modular, scalable, fully-programmable and easy to assemble that can be incorporated into cell culture systems to better recapitulate physiological environments. By controlling two commercially available piezoelectric pumps using a Raspberry Pi Zero microcontroller, the system is capable of producing arbitrary dynamic flow profiles with reliable flow rates ranging from 1 to 3000 µL/min as specified by an easily programmable Python-based script. We validated the accuracy of the flow rates, the use of time-varying profiles, and the practicality of the system by creating repeatable dynamic concentration profiles using a 3D-printed static micromixer., United States. Defense Advanced Research Projects Agency. Microphysiological Systems Program (W911NF-12-2-0039)
- Published
- 2017
- Full Text
- View/download PDF
20. Integrated gut/liver microphysiological systems elucidates inflammatory inter-tissue crosstalk
- Author
-
Wen L.K. Chen, Collin Edington, Emily Suter, Jiajie Yu, Jeremy J. Velazquez, Jason G. Velazquez, Michael Shockley, Emma M. Large, Raman Venkataramanan, David J. Hughes, Cynthia L. Stokes, David L. Trumper, Rebecca L. Carrier, Murat Cirit, Linda G. Griffith, Douglas A. Lauffenburger, Institute for Medical Engineering and Science, Harvard University--MIT Division of Health Sciences and Technology, 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, Chen, Wen Li, Edington, Collin D, Suter, Emily C, Yu, Jiajie, Velazquez, Jeremy J., Velazquez, Jason G, Shockley, Michael J, Trumper, David L, Carrier, Rebecca, Cirit, Murat, Griffith, Linda G, and Lauffenburger, Douglas A
- Subjects
0301 basic medicine ,Cell signaling ,Chemokine ,Colon ,Kupffer Cells ,medicine.medical_treatment ,Bioengineering ,Inflammation ,Cell Communication ,Biology ,Applied Microbiology and Biotechnology ,Article ,Systems Biotechnology ,sepsis ,03 medical and health sciences ,gut‐liver interaction ,Downregulation and upregulation ,Lab-On-A-Chip Devices ,medicine ,Humans ,Immunologic Factors ,organ‐on‐a‐chip ,microphysiological system ,Cells, Cultured ,Immunoassay ,Miniaturization ,CXCR3 ligands ,FGF19 ,Articles ,Equipment Design ,Coculture Techniques ,Cell biology ,Equipment Failure Analysis ,Systems Integration ,Crosstalk (biology) ,030104 developmental biology ,Cytokine ,Liver ,Immunology ,Hepatocytes ,biology.protein ,Cytokines ,CXCL9 ,Caco-2 Cells ,medicine.symptom ,Biotechnology - Abstract
A capability for analyzing complex cellular communication among tissues is important in drug discovery and development, and in vitro technologies for doing so are required for human applications. A prominent instance is communication between the gut and the liver, whereby perturbations of one tissue can influence behavior of the other. Here, we present a study on human gut-liver tissue interactions under normal and inflammatory contexts, via an integrative multi-organ platform comprising human liver (hepatocytes and Kupffer cells), and intestinal (enterocytes, goblet cells, and dendritic cells) models. Our results demonstrated long-term (>2 weeks) maintenance of intestinal (e.g., barrier integrity) and hepatic (e.g., albumin) functions in baseline interaction. Gene expression data comparing liver in interaction with gut, versus isolation, revealed modulation of bile acid metabolism. Intestinal FGF19 secretion and associated inhibition of hepatic CYP7A1 expression provided evidence of physiologically relevant gut-liver crosstalk. Moreover, significant non-linear modulation of cytokine responses was observed under inflammatory gut-liver interaction; for example, production of CXCR3 ligands (CXCL9,10,11) was synergistically enhanced. RNA-seq analysis revealed significant upregulation of IFNα/β/γ signaling during inflammatory gut-liver crosstalk, with these pathways implicated in the synergistic CXCR3 chemokine production. Exacerbated inflammatory response in gut-liver interaction also negatively affected tissue-specific functions (e.g., liver metabolism). These findings illustrate how an integrated multi-tissue platform can generate insights useful for understanding complex pathophysiological processes such as inflammatory organ crosstalk., National Institutes of Health (U.S.) (grant UH3TR00069), United States. Defense Advanced Research Projects Agency (grant Microphysiological Systems Program (W911NF-12-2-00))
- Published
- 2017
21. Integrated Gut and Liver Microphysiological Systems for Quantitative In Vitro Pharmacokinetic Studies
- Author
-
Linda G. Griffith, Nikolaos Tsamandouras, Cynthia L. Stokes, Wen Li Kelly Chen, Murat Cirit, Collin Edington, Massachusetts Institute of Technology. Department of Biological Engineering, Tsamandouras, Nikolaos, Chen, Wen Li, Edington, Collin D, Griffith, Linda G, and Cirit, Murat
- Subjects
0301 basic medicine ,Diclofenac ,Hydrocortisone ,Pharmacology toxicology ,Pharmaceutical Science ,02 engineering and technology ,Computational biology ,Biology ,Pharmacology ,In Vitro Techniques ,Article ,03 medical and health sciences ,Pharmacokinetics ,medicine ,Humans ,Intestinal Mucosa ,Intestinal permeability ,Robustness (evolution) ,021001 nanoscience & nanotechnology ,medicine.disease ,In vitro ,030104 developmental biology ,Drug development ,Liver ,0210 nano-technology ,Drug metabolism ,Oral retinoid - Abstract
Investigation of the pharmacokinetics (PK) of a compound is of significant importance during the early stages of drug development, and therefore several in vitro systems are routinely employed for this purpose. However, the need for more physiologically realistic in vitro models has recently fueled the emerging field of tissue-engineered 3D cultures, also referred to as organs-on-chips, or microphysiological systems (MPSs). We have developed a novel fluidic platform that interconnects multiple MPSs, allowing PK studies in multi-organ in vitro systems along with the collection of high-content quantitative data. This platform was employed here to integrate a gut and a liver MPS together in continuous communication, and investigate simultaneously different PK processes taking place after oral drug administration in humans (e.g., intestinal permeability, hepatic metabolism). Measurement of tissue-specific phenotypic metrics indicated that gut and liver MPSs can be fluidically coupled with circulating common medium without compromising their functionality. The PK of diclofenac and hydrocortisone was investigated under different experimental perturbations, and results illustrate the robustness of this integrated system for quantitative PK studies. Mechanistic model-based analysis of the obtained data allowed the derivation of the intrinsic parameters (e.g., permeability, metabolic clearance) associated with the PK processes taking place in each MPS. Although these processes were not substantially affected by the gut-liver interaction, our results indicate that inter-MPS communication can have a modulating effect (hepatic metabolism upregulation). We envision that our integrative approach, which combines multi-cellular tissue models, multi-MPS platforms, and quantitative mechanistic modeling, will have broad applicability in pre-clinical drug development., United States. Defense Advanced Research Projects Agency (Grant W911NF-12-2-0039), National Institutes of Health (U.S.) (Grant 4-UH3-TR000496-03)
- Published
- 2017
22. Modification of proteolytic activity matrix analysis (PrAMA) to measure ADAM10 and ADAM17 sheddase activities in cell and tissue lysates
- Author
-
Douglas A. Lauffenburger, Toshie Yoneyama, James G. Herman, Autumn Gaither-Davis, Laura P. Stabile, Marcia L. Moss, Linda G. Griffith, Michael C. Gorry, Yan Lin, Miles A. Miller, Nikola L. Vujanovic, Massachusetts Institute of Technology. Biotechnology Process Engineering Center, 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 Mechanical Engineering, Miller, Miles Aaron, Griffith, Linda G, and Lauffenburger, Douglas A
- Subjects
Sheddase activity ,ADAM10 ,Cell ,030232 urology & nephrology ,030204 cardiovascular system & hematology ,Matrix metalloproteinase ,Gene knockout ,03 medical and health sciences ,0302 clinical medicine ,Fluorogenic peptide substrates ,lysate ,medicine ,Multiplex ,Cell lysate ,chemistry.chemical_classification ,ADAM17 ,Cancer biomarker ,Chemistry ,Transfection ,Protease inhibitors ,Sheddase ,Tissues ,medicine.anatomical_structure ,Enzyme ,Gene restoration ,Oncology ,Biochemistry ,Proteolytic activity matrix analysis ,Cancer biomarkers ,Research Paper ,Gene silence - Abstract
Increases in expression of ADAM10 and ADAM17 genes and proteins have been evaluated, but not validated as cancer biomarkers. Specific enzyme activities better reflect enzyme cellular functions, and might be better biomarkers than enzyme genes or proteins. However, no high throughput assay is available to test this possibility. Recent studies have developed the high throughput real-time proteolytic activity matrix analysis (PrAMA) that integrates the enzymatic processing of multiple enzyme substrates with mathematical-modeling computation. The original PrAMA measures with significant accuracy the activities of individual metalloproteinases expressed on live cells. To make the biomarker assay usable in clinical practice, we modified PrAMA by testing enzymatic activities in cell and tissue lysates supplemented with broad-spectrum non-MP enzyme inhibitors, and by maximizing the assay specificity using systematic mathematical-modeling analyses. The modified PrAMA accurately measured the absence and decreases of ADAM10 sheddase activity (ADAM10sa) and ADAM17sa in ADAM10-/- and ADAM17-/- mouse embryonic fibroblasts (MEFs), and ADAM10- and ADAM17-siRNA transfected human cancer cells, respectively. It also measured the restoration and inhibition of ADAM10sa in ADAM10-cDNA-transfected ADAM10-/- MEFs and GI254023X-treated human cancer cell and tissue lysates, respectively. Additionally, the modified PrAMA simultaneously quantified with significant accuracy ADAM10sa and ADAM17sa in multiple human tumor specimens, and showed the essential characteristics of a robust high throughput multiplex assay that could be broadly used in biomarker studies. Selectively measuring specific enzyme activities, this new clinically applicable assay is potentially superior to the standard protein- and gene-expression assays that do not distinguish active and inactive enzyme forms.
- Published
- 2017
23. Human Vascular Tissue Models Formed from Human Induced Pluripotent Stem Cell Derived Endothelial Cells
- Author
-
Jeremy J. Velazquez, Roger D. Kamm, Christine A. Daigh, James A. Thomson, Rachel L. Lewis, James A. Molenda, Tyler D. Hansen, Linda G. Griffith, William L. Murphy, Jordan A. Whisler, Vernella Vickerman, David A. Mann, David G. Belair, Michael P. Schwartz, Jorge Valdez, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Whisler, Jordan Ari, Kamm, Roger Dale, Valdez Macias, Jorge Luis, Velazquez, Jeremy J., and Griffith, Linda G.
- Subjects
Vascular Endothelial Growth Factor A ,Cancer Research ,Matrigel ,Tumor Necrosis Factor-alpha ,Angiogenesis ,Cellular differentiation ,Induced Pluripotent Stem Cells ,Endothelial Cells ,Gene Expression Regulation, Developmental ,Neovascularization, Physiologic ,Cell Differentiation ,Cell Biology ,Fibroblasts ,Biology ,Article ,Cell biology ,Endothelial stem cell ,Vascular endothelial growth factor A ,Blood Vessels ,Humans ,CD146 ,Stem cell ,Induced pluripotent stem cell - Abstract
Here we describe a strategy to model blood vessel development using a well-defined induced pluripotent stem cell-derived endothelial cell type (iPSC-EC) cultured within engineered platforms that mimic the 3D microenvironment. The iPSC-ECs used here were first characterized by expression of endothelial markers and functional properties that included VEGF responsiveness, TNF-α-induced upregulation of cell adhesion molecules (MCAM/CD146; ICAM1/CD54), thrombin-dependent barrier function, shear stress-induced alignment, and 2D and 3D capillary-like network formation in Matrigel. The iPSC-ECs also formed 3D vascular networks in a variety of engineering contexts, yielded perfusable, interconnected lumen when co-cultured with primary human fibroblasts, and aligned with flow in microfluidics devices. iPSC-EC function during tubule network formation, barrier formation, and sprouting was consistent with that of primary ECs, and the results suggest a VEGF-independent mechanism for sprouting, which is relevant to therapeutic anti-angiogenesis strategies. Our combined results demonstrate the feasibility of using a well-defined, stable source of iPSC-ECs to model blood vessel formation within a variety of contexts using standard in vitro formats., National Institutes of Health (U.S.) (NIH 1UH2 TR000506-01), National Institutes of Health (U.S.) (3UH2 TR000506-02S1), National Institutes of Health (U.S.) (T32 HL007936-12), National Institutes of Health (U.S.) (RO1 HL093282), National Institutes of Health (U.S.) (R21 EB016381-01)
- Published
- 2014
- Full Text
- View/download PDF
24. Engineering liver
- Author
-
Linda G. Griffith, Donna B. Stolz, Alan Wells, Massachusetts Institute of Technology. Department of Biological Engineering, and Griffith, Linda G.
- Subjects
Disease Models, Animal ,Liver ,Hepatology ,Chemistry ,Liver Diseases ,Systems Biology ,Animals ,Humans ,Bioengineering ,Computer Simulation ,Article - Abstract
Interest in “engineering liver” arises from multiple communities: therapeutic replacement; mechanistic models of human processes; and drug safety and efficacy studies. An explosion of micro- and nanofabrication, biomaterials, microfluidic, and other technologies potentially affords unprecedented opportunity to create microphysiological models of the human liver, but engineering design principles for how to deploy these tools effectively toward specific applications, including how to define the essential constraints of any given application (available sources of cells, acceptable cost, and user-friendliness), are still emerging. Arguably less appreciated is the parallel growth in computational systems biology approaches toward these same problems—particularly in parsing complex disease processes from clinical material, building models of response networks, and in how to interpret the growing compendium of data on drug efficacy and toxicology in patient populations. Here, we provide insight into how the complementary paths of engineering liver—experimental and computational—are beginning to interplay toward greater illumination of human disease states and technologies for drug development., National Institutes of Health (U.S.) (UH2TR000496), National Institutes of Health (U.S.) (R01-EB 010246), National Institutes of Health (U.S.) (R01-ES015241), National Institutes of Health (U.S.) (P30-ES002109)
- Published
- 2014
- Full Text
- View/download PDF
25. Tailoring Chimeric Ligands for Studying and Biasing ErbB Receptor Family Interactions
- Author
-
Andrew T. Krueger, Barbara Imperiali, Linda G. Griffith, Carsten Kroll, Edgar Sanchez, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Biology, Massachusetts Institute of Technology. Department of Chemistry, Griffith, Linda G., Imperiali, Barbara, Krueger, Andrew T., Kroll, Carsten, and Sanchez, Edgar
- Subjects
Models, Molecular ,Receptor, ErbB-2 ,Stereochemistry ,Chemistry ,Ligand ,Motility ,Mesenchymal Stem Cells ,Biological activity ,General Chemistry ,General Medicine ,Ligands ,Article ,Catalysis ,Bivalent (genetics) ,law.invention ,Cell biology ,ErbB Receptors ,Tetrazine ,chemistry.chemical_compound ,Epidermal growth factor ,law ,Recombinant DNA ,Humans ,Receptor - Abstract
Described is the development and application of a versatile semisynthetic strategy, based on a combination of sortase-mediated coupling and tetrazine ligation chemistry, which can be exploited for the efficient incorporation of tunable functionality into chimeric recombinant proteins. To demonstrate the scope of the method, the assembly of a set of bivalent ligands, which integrate members of the epidermal growth factor (EGF) ligand family, is described. By using a series of bivalent EGFs with variable intraligand spacing, the differences in structure were correlated with the ability to bias signaling in the ErbB receptor family in a cell motility assay. Biasing away from EGFR-HER2 dimerization with a bivalent EGF was observed to reduce cell motility in an intraligand distance-dependent fashion, thus demonstrating the utility of the approach for acutely perturbing receptor-mediated cell signaling pathways., National Cancer Institute (U.S.). Integrative Cancer Biology Program (Grant U54-CA112967), National Institutes of Health (U.S.) (R01DE019523-13), Massachusetts Institute of Technology. Computational and Systems Biology Program. MIT-Merck Postdoctoral Fellowship, Swiss National Science Foundation (Postdoctoral Fellowship), National Institute of Environmental Health Sciences (Training Grant in Environmental Toxicology 5-T32-ES007020)
- Published
- 2014
- Full Text
- View/download PDF
26. A liver microphysiological system of tumor cell dormancy and inflammatory responsiveness is affected by scaffold properties
- Author
-
Linda G. Griffith, Carissa L. Young, Douglas A. Lauffenburger, Linda Stockdale, D. B. Stolz, Amanda M. Clark, J. Shepard Neiman, Alan Wells, Sarah E Wheeler, Raman Venkataramanan, Wenchen Zhao, Massachusetts Institute of Technology. Biotechnology Process Engineering Center, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Biology, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Clark, Abigail M, Young, Carissa L., Stockdale, Linda, Zhao, Weian, Lauffenburger, Douglas A, and Griffith, Linda G
- Subjects
0301 basic medicine ,Cell Survival ,Microfluidics ,Cell Culture Techniques ,Biomedical Engineering ,Antineoplastic Agents ,Breast Neoplasms ,Bioengineering ,Biology ,Biochemistry ,Article ,Metastasis ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Cluster Analysis ,Humans ,Immunoassay ,Tissue Scaffolds ,Fibrinogen ,Hydrogels ,General Chemistry ,medicine.disease ,Primary tumor ,Phenotype ,Metastatic breast cancer ,030104 developmental biology ,Cell culture ,alpha 1-Antitrypsin ,Immunology ,Hepatocytes ,Cancer research ,Cytokines ,Intercellular Signaling Peptides and Proteins ,Polystyrenes ,Dormancy ,Female ,Chemokines ,Signal transduction ,Ex vivo ,Signal Transduction - Abstract
Distant metastasis is the major cause of breast cancer-related mortality, commonly emerging clinically after 5 or more years of seeming 'cure' of the primary tumor, indicating a quiescent dormancy. The lack of relevant accessible model systems for metastasis that recreate this latent stage has hindered our understanding of the molecular basis and the development of therapies against these lethal outgrowths. We previously reported on the development of an all-human 3D ex vivo hepatic microphysiological system that reproduces several features of liver physiology and enables spontaneous dormancy in a subpopulation of breast cancer cells. However, we observed that the dormant cells were localized primarily within the 3D tissue, while the proliferative cells were in contact with the polystyrene scaffold. As matrix stiffness is known to drive inflammatory and malignant behaviors, we explored the occurrence of spontaneous tumor dormancy and inflammatory phenotype. The microphysiological system was retrofitted with PEGDa-SynKRGD hydrogel scaffolding, which is softer and differs in the interface with the tissue. The microphysiological system incorporated donor-matched primary human hepatocytes and non-parenchymal cells (NPCs), with MDA-MB-231 breast cancer cells. Hepatic tissue in hydrogel scaffolds secreted lower levels of pro-inflammatory analytes, and was more responsive to inflammatory stimuli. The proportion of tumor cells entering dormancy was markedly increased in the hydrogel-supported tissue compared to polystyrene. Interestingly, an unexpected differential response of dormant cells to varying chemotherapeutic doses was identified, which if reflective of patient pathophysiology, has important implications for patient dosing regimens. These findings highlight the metastatic microphysiological system fitted with hydrogel scaffolds as a critical tool in the assessment and development of therapeutic strategies to target dormant metastatic breast cancer.
- Published
- 2016
27. Modeling Therapeutic Antibody-Small Molecule Drug-Drug Interactions Using a Three-Dimensional Perfusable Human Liver Coculture Platform
- Author
-
Linda G. Griffith, Thomas Joseph Long, Hisham K. Hamadeh, Donna B. Stolz, Helen J. McBride, Patrick A Cosgrove, Cynthia A. Afshari, Robert T. Dunn, Massachusetts Institute of Technology. Biotechnology Process Engineering Center, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Long, Thomas J, and Griffith, Linda G
- Subjects
0301 basic medicine ,Kupffer Cells ,Pharmaceutical Science ,Pharmacology ,Antibodies, Monoclonal, Humanized ,Small Molecule Libraries ,03 medical and health sciences ,chemistry.chemical_compound ,Tocilizumab ,medicine ,Cytochrome P-450 CYP3A ,Humans ,Drug Interactions ,Interleukin 6 ,Receptor ,Cells, Cultured ,Inflammation ,biology ,CYP3A4 ,Interleukin-6 ,Area under the curve ,Antibodies, Monoclonal ,Articles ,In vitro ,Coculture Techniques ,030104 developmental biology ,medicine.anatomical_structure ,C-Reactive Protein ,chemistry ,Liver ,Cell culture ,Hepatocyte ,biology.protein ,Hepatocytes ,Half-Life - Abstract
Traditional in vitro human liver cell culture models lose key hepatic functions such as metabolic activity during short-term culture. Advanced three-dimensional (3D) liver coculture platforms offer the potential for extended hepatocyte functionality and allow for the study of more complex biologic interactions, which can improve and refine human drug safety evaluations. Here, we use a perfusion flow 3D microreactor platform for the coculture of cryopreserved primary human hepatocytes and Kupffer cells to study the regulation of cytochrome P450 3A4 isoform (CYP3A4) activity by chronic interleukin 6 (IL-6)-mediated inflammation over 2 weeks. Hepatocyte cultures remained stable over 2 weeks, with consistent albumin production and basal IL-6 levels. Direct IL-6 stimulation that mimics an inflammatory state induced a dose-dependent suppression of CYP3A4 activity, an increase in C-reactive protein (CRP) secretion, and a decrease in shed soluble interleukin-6 receptor (IL-6R) levels, indicating expected hepatic IL-6 bioactivity. Tocilizumab, an anti-IL- 6R monoclonal antibody used to treat rheumatoid arthritis, has been demonstrated clinically to impact small molecule drug pharmacokinetics by modulating cytochrome P450 enzyme activities, an effect not observed in traditional hepatic cultures. We have now recapitulated the clinical observation in a 3D bioreactor system. Tocilizumab was shown to desuppress CYP3A4 activity while reducing the CRP concentration after 72 hours in the continued presence of IL-6. This change in CYP3A4 activity decreased the half-life and area under the curve up to the last measurable concentration (AUClast) of the small molecule CYP3A4 substrate simvastatin hydroxy acid, measured before and after tocilizumab treatment. We conclude that next-generation in vitro liver culture platforms are well suited for these types of long-term treatment studies and show promise for improved drug safety assessment., United States. Defense Advanced Research Projects Agency (W911NF-12-2-0039), National Institutes of Health (U.S.) (UH3TR000496)
- Published
- 2016
28. A mouse-human phase 1 co-clinical trial of a protease-activated fluorescent probe for imaging cancer
- Author
-
William C. Eward, Rachel A. Greenup, Chang-Lung Lee, Rakesh K. Jain, Matija Snuderl, Moungi G. Bawendi, E. Shelley Hwang, Ivan Spasojevic, W. David Lee, Melodi Javid Whitley, Dan G. Blazer, Alexander L. Lazarides, Joan Cahill, Erin K. O'Reilly, Paul J. Mosca, Richard F. Riedel, David G. Kirsch, Jeffrey K. Mito, Linda G. Griffith, Kyle C. Cuneo, David B. Strasfeld, Jorge Ferrer, Dai Fukumura, Brian E. Brigman, Diana M. Cardona, Nicole A. Larrier, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Chemistry, Griffith, Linda G, and Bawendi, Moungi G
- Subjects
Diagnostic Imaging ,0301 basic medicine ,Biodistribution ,Pathology ,medicine.medical_specialty ,Breast Neoplasms ,Biology ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,In vivo ,Neoplasms ,Adjuvant therapy ,medicine ,Animals ,Humans ,Tissue Distribution ,Fluorescent Dyes ,Soft tissue sarcoma ,Cancer ,Sarcoma ,General Medicine ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,030220 oncology & carcinogenesis ,Injections, Intravenous ,Metabolome ,Female ,Ex vivo ,Peptide Hydrolases - Abstract
Local recurrence is a common cause of treatment failure for patients with solid tumors. Intraoperative detection of microscopic residual cancer in the tumor bed could be used to decrease the risk of a positive surgical margin, reduce rates of reexcision, and tailor adjuvant therapy. We used a protease-activated fluorescent imaging probe, LUM015, to detect cancer in vivo in a mouse model of soft tissue sarcoma (STS) and ex vivo in a first-in-human phase 1 clinical trial. In mice, intravenous injection of LUM015 labeled tumor cells, and residual fluorescence within the tumor bed predicted local recurrence. In 15 patients with STS or breast cancer, intravenous injection of LUM015 before surgery was well tolerated. Imaging of resected human tissues showed that fluorescence from tumor was significantly higher than fluorescence from normal tissues. LUM015 biodistribution, pharmacokinetic profiles, and metabolism were similar in mouse and human subjects. Tissue concentrations of LUM015 and its metabolites, including fluorescently labeled lysine, demonstrated that LUM015 is selectively distributed to tumors where it is activated by proteases. Experiments in mice with a constitutively active PEGylated fluorescent imaging probe support a model where tumor-selective probe distribution is a determinant of increased fluorescence in cancer. These co-clinical studies suggest that the tumor specificity of protease-activated imaging probes, such as LUM015, is dependent on both biodistribution and enzyme activity. Our first-in-human data support future clinical trials of LUM015 and other protease-sensitive probes.
- Published
- 2016
- Full Text
- View/download PDF
29. Production of Reactive Oxygen Species by Multipotent Stromal Cells/Mesenchymal Stem Cells upon Exposure to Fas Ligand
- Author
-
Linda G. Griffith, Omari Turner, Alan Wells, Melanie Rodrigues, Donna B. Stolz, Massachusetts Institute of Technology. Department of Biological Engineering, and Griffith, Linda G.
- Subjects
MAPK/ERK pathway ,Fas Ligand Protein ,Stromal cell ,medicine.medical_treatment ,Cell Culture Techniques ,Biomedical Engineering ,lcsh:Medicine ,Apoptosis ,Biology ,Caspase 8 ,p38 Mitogen-Activated Protein Kinases ,Article ,Fas ligand ,Superoxides ,Survivin ,medicine ,Humans ,Membrane Potential, Mitochondrial ,Transplantation ,lcsh:R ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Cell Biology ,Recombinant Proteins ,Cell biology ,Oxidative Stress ,Cytokine ,Reactive Oxygen Species ,Signal Transduction - 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
- 2012
- Full Text
- View/download PDF
30. Controlling multipotent stromal cell migration by integrating 'course-graining' materials and 'fine-tuning' small molecules via decision tree signal-response modeling
- Author
-
Linda G. Griffith, Alan Wells, Douglas A. Lauffenburger, Shan Wu, Massachusetts Institute of Technology. Department of Biological Engineering, Wu, Shan, Griffith, Linda G., and Lauffenburger, Douglas A.
- Subjects
Stromal cell ,Materials science ,Biophysics ,Biocompatible Materials ,Context (language use) ,Bioengineering ,Models, Biological ,Article ,Cell Line ,Extracellular matrix ,Biomaterials ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Epidermal growth factor ,Humans ,Computer Simulation ,Cell migration ,030304 developmental biology ,0303 health sciences ,biology ,Multipotent Stem Cells ,Decision Trees ,Mesenchymal stem cell ,Computational modeling ,3. Good health ,Cell biology ,Fibronectin ,Multipotent Stem Cell ,Mechanics of Materials ,030220 oncology & carcinogenesis ,biology.protein ,Ceramics and Composites ,Mesenchymal stem cells ,Biomedical engineering - 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)
- Published
- 2011
- Full Text
- View/download PDF
31. Synergistic effects of tethered growth factors and adhesion ligands on DNA synthesis and function of primary hepatocytes cultured on soft synthetic hydrogels
- Author
-
Linda G. Griffith, Geeta Mehta, Roger D. Kamm, Courtney M. Williams, Luis M. Alvarez, Martha Lesniewski, 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, and Griffith, Linda G.
- Subjects
Male ,Cell Survival ,Biophysics ,Biocompatible Materials ,Bioengineering ,Article ,Biomaterials ,Extracellular matrix ,Epidermal growth factor ,Animals ,Humans ,Immunoprecipitation ,Phosphorylation ,Cells, Cultured ,Epidermal Growth Factor ,biology ,DNA synthesis ,Hydrogels ,DNA ,Cell aggregation ,Fibronectins ,Rats ,ErbB Receptors ,Fibronectin ,Microscopy, Fluorescence ,Biochemistry ,Mechanics of Materials ,Hepatocytes ,Ceramics and Composites ,biology.protein ,Signal transduction ,Signal Transduction ,Binding domain - 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
- 2010
- Full Text
- View/download PDF
32. Multipathway Kinase Signatures of Multipotent Stromal Cells Are Predictive for Osteogenic Differentiation
- Author
-
Manu O. Platt, Douglas A. Lauffenburger, Catera L. Wilder, Linda G. Griffith, Alan Wells, Massachusetts Institute of Technology. Department of Biological Engineering, Lauffenburger, Douglas A., Platt, Manu O., Wilder, Catera L., and Griffith, Linda G.
- Subjects
Cell signaling ,Stromal cell ,Mitogen-Activated Protein Kinase 3 ,Cellular differentiation ,Mesenchymal stem cell ,Molecular Medicine ,Cell Biology ,Cell fate determination ,Signal transduction ,Biology ,Stem cell ,Developmental Biology ,Cell biology - Abstract
Bone marrow-derived multipotent stromal cells (MSCs) offer great promise for regenerating tissue. Although certain transcription factors have been identified in association with tendency toward particular MSC differentiation phenotypes, the regulatory network of key receptor-mediated signaling pathways activated by extracellular ligands that induce various differentiation responses remains poorly understood. Attempts to predict differentiation fate tendencies from individual pathways in isolation are problematic due to the complex pathway interactions inherent in signaling networks. Accordingly, we have undertaken a multivariate systems approach integrating experimental measurement of multiple kinase pathway activities and osteogenic differentiation in MSCs, together with computational analysis to elucidate quantitative combinations of kinase signals predictive of cell behavior across diverse contexts. In particular, for culture on polymeric biomaterial surfaces presenting tethered epidermal growth factor, type I collagen, neither, or both, we have found that a partial least-squares regression model yields successful prediction of phenotypic behavior on the basis of two principal components comprising the weighted sums of eight intracellular phosphoproteins: phospho-epidermal growth factor receptor, phospho-Akt, phospho-extracellular signal-related kinase 1/2, phospho-heat shock protein 27, phospho-c-Jun, phospho-glycogen synthase kinase 3α/β, phospho-p38, and phospho-signal transducer and activator of transcription 3. This combination provides the strongest predictive capability for 21-day differentiated phenotype status when calculated from day-7 signal measurements; day-4 and day-14 signal measurements are also significantly predictive, indicating a broad time frame during MSC osteogenesis wherein multiple pathways and states of the kinase signaling network are quantitatively integrated to regulate gene expression, cell processes, and ultimately, cell fate. STEM CELLS 2009;27:2804–2814, National Institutes of Health (U.S.) (Grant NIH R01-GM059870-07), National Institutes of Health (U.S.) (Grant R01 DE019523- 10), United Negro College Fund ((UNCF)/Merck Postdoctoral Fellowship), Georgia Institute of Technology (Facilitating Academic Careers in Engineering and Sciences Fellowship)
- Published
- 2009
- Full Text
- View/download PDF
33. Targeting autocrine HB-EGF signaling with specific ADAM12 inhibition using recombinant ADAM12 prodomain
- Author
-
Aaron S. Meyer, Gary Powell, Douglas A. Lauffenburger, Marcia L. Moss, Linda G. Griffith, Miles A. Miller, Lori L. Edwards, Robert M. Petrovich, 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., and Lauffenburger, Douglas A.
- Subjects
Heparin-binding EGF-like growth factor ,ADAM10 ,Endometriosis ,ADAM12 Protein ,Biology ,Ligands ,Amphiregulin ,Models, Biological ,Article ,Cell Line ,03 medical and health sciences ,Endometrium ,0302 clinical medicine ,Epidermal growth factor ,Cell Movement ,Humans ,Enzyme Inhibitors ,Autocrine signalling ,030304 developmental biology ,0303 health sciences ,Metalloproteinase ,Multidisciplinary ,Antibodies, Monoclonal ,Membrane Proteins ,Molecular biology ,Recombinant Proteins ,3. Good health ,Cell biology ,ADAM Proteins ,Autocrine Communication ,030220 oncology & carcinogenesis ,Female ,Signal transduction ,Decoy ,Heparin-binding EGF-like Growth Factor ,Protein Binding ,Signal Transduction - 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
- Full Text
- View/download PDF
34. Human mesenchymal stem cells/multipotent stromal cells consume accumulated autophagosomes early in differentiation
- Author
-
Austin Nuschke, Charleen T. Chu, Melanie Rodrigues, Donna B. Stolz, Linda G. Griffith, Alan Wells, Massachusetts Institute of Technology. Department of Biological Engineering, and Griffith, Linda G.
- Subjects
Autophagosome ,Cellular differentiation ,Medicine (miscellaneous) ,Clinical uses of mesenchymal stem cells ,Biology ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,03 medical and health sciences ,0302 clinical medicine ,Phagosomes ,Autophagy ,Humans ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,Research ,Mesenchymal stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell Biology ,3. Good health ,Cell biology ,Endothelial stem cell ,Adipogenesis ,030220 oncology & carcinogenesis ,Molecular Medicine ,Adult stem cell - 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 rapamycin treatment and accelerated fat accumulation following autophagosome blockade by bafilomycin. Conclusions Our findings suggest that MSCs exist in a state of arrested autophagy with high autophagosome accumulation and are poised to rapidly undergo autophagic degradation. This phenotype is highly sensitive, and a balance of autophagy appears to be key in efficient MSC differentiation and function, as evidenced by our results implicating autophagic flux in early osteogenesis and adipogenesis., 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
- 2014
35. A microphysiological system model of therapy for liver micrometastases
- Author
-
Donald P. Taylor, Linda G. Griffith, Carissa L. Young, Donna B. Stolz, Transon V. Nguyen, Douglas A. Lauffenburger, Alan Wells, Sarah E Wheeler, Amanda M. Clark, Raman Venkataramanan, Jeffrey T. Borenstein, Rachelle Prantil-Baun, Venkateswaran C. Pillai, Massachusetts Institute of Technology. Department of Biological Engineering, Young, Carissa L., Lauffenburger, Douglas A., and Griffith, Linda G.
- Subjects
Time Factors ,medicine.medical_treatment ,Cell Culture Techniques ,Antineoplastic Agents ,Breast Neoplasms ,Biology ,Fibrinogen ,Models, Biological ,Article ,General Biochemistry, Genetics and Molecular Biology ,Metastasis ,Paracrine signalling ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,Insulin ,Liver Neoplasms ,Micrometastasis ,Cancer ,medicine.disease ,Crosstalk (biology) ,Liver ,Immunology ,Hepatocytes ,Cancer research ,Female ,Drug Screening Assays, Antitumor ,medicine.drug ,Hormone - Abstract
Metastasis accounts for almost 90% of cancer-associated mortality. The effectiveness of cancer therapeutics is limited by the protective microenvironment of the metastatic niche and consequently these disseminated tumors remain incurable. Metastatic disease progression continues to be poorly understood due to the lack of appropriate model systems. To address this gap in understanding, we propose an all-human microphysiological system that facilitates the investigation of cancer behavior in the liver metastatic niche. This existing LiverChip is a 3D-system modeling the hepatic niche; it incorporates a full complement of human parenchymal and non-parenchymal cells and effectively recapitulates micrometastases. Moreover, this system allows real-time monitoring of micrometastasis and assessment of human-specific signaling. It is being utilized to further our understanding of the efficacy of chemotherapeutics by examining the activity of established and novel agents on micrometastases under conditions replicating diurnal variations in hormones, nutrients and mild inflammatory states using programmable microdispensers. These inputs affect the cues that govern tumor cell responses. Three critical signaling groups are targeted: the glucose/insulin responses, the stress hormone cortisol and the gut microbiome in relation to inflammatory cues. Currently, the system sustains functioning hepatocytes for a minimum of 15 days; confirmed by monitoring hepatic function (urea, α-1-antitrypsin, fibrinogen, and cytochrome P450) and injury (AST and ALT). Breast cancer cell lines effectively integrate into the hepatic niche without detectable disruption to tissue, and preliminary evidence suggests growth attenuation amongst a subpopulation of breast cancer cells. xMAP technology combined with systems biology modeling are also employed to evaluate cellular crosstalk and illustrate communication networks in the early microenvironment of micrometastases. This model is anticipated to identify new therapeutic strategies for metastasis by elucidating the paracrine effects between the hepatic and metastatic cells, while concurrently evaluating agent efficacy for metastasis, metabolism and tolerability., National Institutes of Health (U.S.) (Grant 1UH2TR000496-01), United States. Defense Advanced Research Projects Agency. Microphysiological Systems Program (W911NF-12-2-0039)
- Published
- 2014
36. Approaches to in vitro tissue regeneration with application for human disease modeling and drug development
- Author
-
Linda G. Griffith, Jeffrey T. Borenstein, Carissa L. Young, Douglas A. Lauffenburger, Mohammad R. Ebrahimkhani, 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., and Griffith, Linda G.
- Subjects
Pharmacology ,Tissue Engineering ,Drug discovery ,Extramural ,business.industry ,Guided Tissue Regeneration ,Regeneration (biology) ,Systems biology ,Computational biology ,Biology ,Article ,Biotechnology ,Disease Models, Animal ,Human disease ,Drug development ,Tissue engineering ,Drug Discovery ,Animals ,Humans ,Regeneration ,business - 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
- 2014
37. Bioreactor technologies to support liver function in vitro
- Author
-
Micha Sam Brickman Raredon, Mohammad R. Ebrahimkhani, David Hughes, Linda G. Griffith, Jaclyn A. Shepard Neiman, 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, and Griffith, Linda G.
- Subjects
Organ Culture Technique ,Emerging technologies ,business.industry ,Drug discovery ,Cell Culture Techniques ,Pharmaceutical Science ,Computational biology ,Biology ,Article ,Biotechnology ,Bioreactors ,Organ Culture Techniques ,Liver ,Drug development ,Hepatocytes ,Bioreactor ,Animals ,Humans ,Liver function ,business ,Drug toxicity - 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
- 2014
38. Molecular network analysis of endometriosis reveals a role for c-Jun-regulated macrophage activation
- Author
-
Nicole Pfäffle-Doyle, Linda G. Griffith, S.N. Morris, Douglas A. Lauffenburger, Keith B. Isaacson, Emily A. Prentice, Michael T. Beste, Massachusetts Institute of Technology. Center for Gynepathology Research, Massachusetts Institute of Technology. Department of Biological Engineering, Beste, Michael T., Pfaffle-Doyle, Nicole, Prentice, Emily A., Lauffenburger, Douglas A., Isaacson, Keith B., and Griffith, Linda G.
- Subjects
Adult ,Proto-Oncogene Proteins c-jun ,medicine.medical_treatment ,Gene regulatory network ,Endometriosis ,Inflammation ,Biology ,Article ,Young Adult ,medicine ,Macrophage ,Humans ,Gene Regulatory Networks ,Regulation of gene expression ,Kinase ,Genome, Human ,Biopsy, Needle ,Reproducibility of Results ,General Medicine ,Macrophage Activation ,Middle Aged ,medicine.disease ,Human genetics ,Cytokine ,Gene Expression Regulation ,Immunology ,Multivariate Analysis ,Macrophages, Peritoneal ,Trans-Activators ,Cytokines ,Female ,medicine.symptom ,Biomarkers - Abstract
Clinical management of endometriosis is limited by the complex relationship between symptom severity, heterogeneous surgical presentation, and variability in clinical outcomes. As a complement to visual classification schemes, molecular profiles of disease activity may improve risk stratification to better inform treatment decisions and identify new approaches to targeted treatment. We use a network analysis of information flow within and between inflammatory cells to discern consensus behaviors characterizing patient subpopulations. Unsupervised multivariate analysis of cytokine profiles quantified by multiplex immunoassays identified a subset of patients with a shared “consensus signature” of 13 elevated cytokines that was associated with common clinical features of endometriosis, but was not observed among patient subpopulations defined by morphologic presentation alone. Enrichment analysis of consensus markers reinforced the primacy of peritoneal macrophage infiltration and activation, which was demonstrably elevated in ex vivo cultures. Although familiar targets of the nuclear factor κB family emerged among overrepresented transcriptional binding sites for consensus markers, our analysis provides evidence for an unexpected contribution from c-Jun, c-Fos, and AP-1 effectors of mitogen-associated kinase signaling. Their crucial involvement in propagation of macrophage-driven inflammatory networks was confirmed via targeted inhibition of upstream kinases. Collectively, these analyses suggest a clinically relevant inflammatory network that may serve as an objective measure for guiding treatment decisions for endometriosis management, and in the future may provide a mechanistic endpoint for assessing efficacy of new agents aimed at curtailing inflammatory mechanisms that drive disease progression., National Institutes of Health (U.S.) (R01EB10246), Singapore-MIT Alliance, United States. Army Research Office (Institute for Collaborative Biotechnologies W911NF-09-001), John D. and Catherine T. MacArthur Foundation
- Published
- 2014
39. Equilibrium and dynamic design principles for binding molecules engineered for reagentless biosensors
- Author
-
Barbara Imperiali, Seymour de Picciotto, Linda G. Griffith, K. Dane Wittrup, 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 Chemistry, Koch Institute for Integrative Cancer Research at MIT, De Picciotto, Seymour, Imperiali, Barbara, Griffith, Linda G, and Wittrup, Karl Dane
- Subjects
Fluorophore ,Chemistry ,Biophysics ,technology, industry, and agriculture ,Nanotechnology ,Cell Biology ,macromolecular substances ,Biosensing Techniques ,Ligand (biochemistry) ,Ligands ,Biochemistry ,Signal ,Models, Biological ,Article ,Dissociation constant ,chemistry.chemical_compound ,Kinetics ,Förster resonance energy transfer ,Reaction rate constant ,Genetic Engineering ,Molecular Biology ,Biosensor ,Equilibrium constant - Abstract
Reagentless biosensors rely on the interaction of a binding partner and its target to generate a change in fluorescent signal using an environment-sensitive fluorophore or Förster resonance energy transfer. Binding affinity can exert a significant influence on both the equilibrium and the dynamic response characteristics of such a biosensor. We here develop a kinetic model for the dynamic performance of a reagentless biosensor. Using a sinusoidal signal for ligand concentration, our findings suggest that it is optimal to use a binding moiety whose equilibrium dissociation constant matches that of the average predicted input signal, while maximizing both the association rate constant and the dissociation rate constant at the necessary ratio to create the desired equilibrium constant. Although practical limitations constrain the attainment of these objectives, the derivation of these design principles provides guidance for improved reagentless biosensor performance and metrics for quality standards in the development of biosensors. These concepts are broadly relevant to reagentless biosensor modalities., National Cancer Institute (U.S.). Integrative Cancer Biology Program (Grant 1 U54 CA112967), National Institutes of Health (U.S.) (R01 EB 010246)
- Published
- 2014
40. An Engineered Bivalent Neuregulin Protects Against Doxorubicin-Induced Cardiotoxicity With Reduced Proneoplastic Potential
- Author
-
Ashwin C. Murthy, Luis M. Alvarez, Joshua R. Wortzel, Joseph Gannon, Jessica F. Hawkins, Steven M. Jay, Linda G. Griffith, Calum A. MacRae, Matthew L. Steinhauser, Richard T. Lee, Massachusetts Institute of Technology. Center for Gynepathology Research, Massachusetts Institute of Technology. Department of Biological Engineering, Jay, Steven M., Alvarez, Luis M., and Griffith, Linda G.
- Subjects
Cardiotoxicity ,biology ,Anthracycline ,Pharmacology ,Physiology (medical) ,Cancer cell ,biology.protein ,medicine ,Cancer research ,Neuregulin ,Doxorubicin ,ERBB3 ,Neuregulin 1 ,Cardiology and Cardiovascular Medicine ,Receptor ,medicine.drug - Abstract
Background—Doxorubicin (DOXO) is an effective anthracycline chemotherapeutic, but its use is limited by cumulative dose-dependent cardiotoxicity. Neuregulin-1β is an ErbB receptor family ligand that is effective against DOXO-induced cardiomyopathy in experimental models but is also proneoplastic. We previously showed that an engineered bivalent neuregulin-1β (NN) has reduced proneoplastic potential in comparison with the epidermal growth factor–like domain of neuregulin-1β (NRG), an effect mediated by receptor biasing toward ErbB3 homotypic interactions uncommonly formed by native neuregulin-1β. Here, we hypothesized that a newly formulated, covalent NN would be cardioprotective with reduced proneoplastic effects in comparison with NRG. Methods and Results—NN was expressed as a maltose-binding protein fusion in Escherichia coli. As established previously, NN stimulated antineoplastic or cytostatic signaling and phenotype in cancer cells, whereas NRG stimulated proneoplastic signaling and phenotype. In neonatal rat cardiomyocytes, NN and NRG induced similar downstream signaling. NN, like NRG, attenuated the double-stranded DNA breaks associated with DOXO exposure in neonatal rat cardiomyocytes and human cardiomyocytes derived from induced pluripotent stem cells. NN treatment significantly attenuated DOXO-induced decrease in fractional shortening as measured by blinded echocardiography in mice in a chronic cardiomyopathy model (57.7±0.6% versus 50.9±2.6%, P=0.004), whereas native NRG had no significant effect (49.4±3.7% versus 50.9±2.6%, P=0.813). Conclusions—NN is a cardioprotective agent that promotes cardiomyocyte survival and improves cardiac function in DOXO-induced cardiotoxicity. Given the reduced proneoplastic potential of NN versus NRG, NN has translational potential for cardioprotection in patients with cancer receiving anthracyclines., United States. Dept. of Defense (Congressionally Directed Medical Research Programs, Breast Cancer Research Program (W81XWH-11-1-0035)), National Institutes of Health (U.S.) (grant HL112905), National Institutes of Health (U.S.) (grant AG032977), National Institutes of Health (U.S.) (grant DE019523), National Institutes of Health (U.S.) (grant DK090147), National Institutes of Health (U.S.) (grant U54-CA112967), National Institutes of Health (U.S.) (grant EB003805), United States. Dept. of Defense (Congressionally Directed Medical Research Programs, Orthopedic Research Program Career Development Award (W81XWH-11-1-0821)), Sarnoff Cardiovascular Research Foundation (fellowship), Hertz Foundation (Fellowship), Harvard Stem Cell Institute
- Published
- 2013
41. Challenges in using cultured primary rodent hepatocytes or cell lines to study hepatic HDL receptor SR-BI regulation by its cytoplasmic adaptor PDZK1
- Author
-
Monty Krieger, Olivier Kocher, Kosuke Tsukamoto, Linda G. Griffith, Lorenna D. Buck, Walker Inman, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Biology, Tsukamoto, Kosuke, Krieger, Monty, Buck, Lorenna Dianne, Griffith, Linda G., and Inman, Samuel Walker
- Subjects
Male ,Cytoplasm ,Time Factors ,Gene Expression ,lcsh:Medicine ,Cardiovascular ,Biochemistry ,Hepatitis ,Mice ,Molecular Cell Biology ,Gene expression ,lcsh:Science ,Mice, Knockout ,0303 health sciences ,Multidisciplinary ,Liver Diseases ,030302 biochemistry & molecular biology ,Intracellular Signaling Peptides and Proteins ,Transfection ,Scavenger Receptors, Class B ,Lipids ,Hepatitis C ,3. Good health ,Cell biology ,medicine.anatomical_structure ,Liver ,Hepatocyte ,Knockout mouse ,Medicine ,Infectious diseases ,Membranes and Sorting ,Cholesterol Esters ,Cellular Types ,Research Article ,Biotechnology ,Gastroenterology and Hepatology ,Viral diseases ,Biology ,Cell Line ,03 medical and health sciences ,In vivo ,medicine ,Animals ,Humans ,030304 developmental biology ,Tissue Engineering ,Infectious Hepatitis ,HEK 293 cells ,lcsh:R ,Membrane Proteins ,Proteins ,Molecular biology ,Mice, Inbred C57BL ,Membrane protein ,Cell culture ,Hepatocytes ,lcsh:Q - Abstract
Background: PDZK1 is a four PDZ-domain containing cytoplasmic protein that binds to a variety of membrane proteins via their C-termini and can influence the abundance, localization and/or function of its target proteins. One of these targets in hepatocytes in vivo is the HDL receptor SR-BI. Normal hepatic expression of SR-BI protein requires PDZK1, National Institutes of Health (U.S.) (Grant HL052212), National Institutes of Health (U.S.) (Grant HL066105), National Institutes of Health (U.S.) (Grant ES015241), National Institutes of Health (U.S.) (Grant GM068762)
- Published
- 2013
42. In vitro models for liver toxicity testing
- Author
-
Linda G. Griffith, Valerie Y. Soldatow, Ivan Rusyn, Edward L. LeCluyse, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, and Griffith, Linda G.
- Subjects
Cell type ,Pathology ,medicine.medical_specialty ,business.industry ,Health, Toxicology and Mutagenesis ,Toxicology ,Embryonic stem cell ,In vitro ,Article ,Cell biology ,Cell culture ,Toxicity ,Microsome ,Medicine ,Stem cell ,business ,Immortalised cell line - 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
- 2012
43. Helix versus coil polypeptide macromers: gel networks with decoupled stiffness and permeability
- Author
-
Linda G. Griffith, Shannon M. Morey, Abigail M. Oelker, Paula T. Hammond, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Chemistry, Oelker, Abigail M., Morey, Shannon M., Griffith, Linda G., and Hammond, Paula T.
- Subjects
chemistry.chemical_classification ,Circular dichroism ,Materials science ,technology, industry, and agriculture ,General Chemistry ,Polymer ,Condensed Matter Physics ,Random coil ,Article ,chemistry.chemical_compound ,Crystallography ,Monomer ,chemistry ,Polymerization ,Polymer chemistry ,Self-healing hydrogels ,Ethylene glycol ,Protein secondary structure - Abstract
As a platform for investigating the individual effects of substrate stiffness, permeability, and ligand density on cellular behavior, we developed a set of hydrogels with stiffness tuned by polymer backbone rigidity, independent of cross-link density and concentration. Previous studies report that poly(propargyl-L-glutamate) (PPLG), synthesized by ring-opening polymerization of the N-carboxy anhydride of γ-propargyl-L-glutamate (γpLglu), adopts a rigid a-helix conformation: we hypothesized that a random copolymer (PPDLG) with equal amounts of γpLglu and γ-propargyl-D-glutamate (γpDglu) monomers would exhibit a more flexible random coil conformation. The resulting macromers exhibited narrow molecular weight distributions (PDI = 1.15) and were grafted with ethylene glycol groups using a highly efficient “click” azide/alkyne cycloaddition reaction with average grafting efficiency of 97% for PPLG and 85% for PPDLG. The polypeptide secondary structure, characterized via circular dichroism spectroscopy, FTIR spectroscopy, and dynamic light scattering, is indeed dependent upon monomer chirality: PPLG exhibits an α-helix conformation while PPDLG adopts a random coil conformation. Hydrogel networks produced by cross-linking either helical or random coil polypeptides with poly(ethylene glycol) (PEG) were analyzed for amount of swelling, gelation efficiency, and permeability to a model protein. In addition, the elastic modulus of helical and coil polypeptide gels was determined by AFM indentation in fluid. Importantly, we found that helical and coil polypeptide gels exhibited similar swelling and permeability but different stiffnesses, which correspond to predictions from the theory of semi-flexible chains., National Institutes of Health (U.S.) (R01 EB10246), National Science Foundation (U.S.). Emergent Behaviors of Integrated Cellular Systems
- Published
- 2012
44. Multilayer thin-film coatings capable of extended programmable drug release: application to human mesenchymal stem cell differentiation
- Author
-
Jinkee Hong, Kookheon Char, Paula T. Hammond, Byeong Su Kim, Linda G. Griffith, Luis M. Alvarez, Nisarg J. Shah, Younghyun Cho, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Department of Mechanical Engineering, Koch Institute for Integrative Cancer Research at MIT, Hong, Jinkee, Alvarez, Luis M., Shah, Nisarg J., Kim, Byeong-Su, Griffith, Linda G., and Hammond, Paula T.
- Subjects
Transplantation ,Cell therapy ,Chemistry ,Cell culture ,Layer by layer ,Mesenchymal stem cell ,Pharmaceutical Science ,Nanotechnology ,Mesenchymal stem cell differentiation ,Stem cell ,Controlled release ,Article - Abstract
The promise of cellular therapy lies in healing damaged tissues and organs in vivo as well as generating tissue constructs in vitro for subsequent transplantation. Postnatal stem cells are ideally suited for cellular therapies due to their pluripotency and the ease with which they can be cultured on functionalized substrates. Creating environments to control and successfully drive their differentiation toward a lineage of choice is one of the most important challenges of current cell-based engineering strategies. In recent years, a variety of biomaterials platforms have been prepared for stem cell cultures, primarily to provide efficient delivery of growth or survival factors to cells and a conductive microenvironment for their growth. Here, we demonstrate that repeating tetralayer structures composed of biocompatible poly(methacrylic acid), poly(acrylamide), and poly(ethylene oxide)-block-poly(ε-caprolactone) micelles arrayed in layer-by-layer films can serve as a payload region for dexamethasone delivery to human mesenchymal stem cells (MSCs). This architecture can induce MSC differentiation into osteoblasts in a dose-dependent manner. The amount of Dex loaded in the films is controlled by varying the deposition conditions and the film thickness. Release of Dex is tuned by changing the amount of covalent cross-linking of multilayers via thermal treatments. The multilayer architecture including payload and cell-adhesion region introduced here are well suited for extended cell culture thus affording the important and protective effect of both Dex release and immobilization. These films may find applications in the local delivery of immobilized therapeutics for biomedical applications, as they can be deposited on a wide range of substrates with different shapes, sizes, and composition., Singapore-MIT Alliance for Research and Technology, Hertz Foundation
- Published
- 2012
45. Surface Tethered Epidermal Growth Factor Protects Proliferating and Differentiating Multipotential Stromal Cells from FasL-Induced Apoptosis
- Author
-
Linda Stockdale, Melanie Rodrigues, Harry C. Blair, Linda G. Griffith, Alan Wells, Massachusetts Institute of Technology. Department of Biological Engineering, Stockdale, Linda, and Griffith, Linda G.
- Subjects
Programmed cell death ,medicine.medical_specialty ,Stromal cell ,Fas Ligand Protein ,Cellular differentiation ,Apoptosis ,Biocompatible Materials ,Bone Marrow Cells ,Biology ,Fas ligand ,Article ,Cell Line ,Epidermal growth factor ,Internal medicine ,medicine ,Adipocytes ,Humans ,Cell Proliferation ,Osteoblasts ,Epidermal Growth Factor ,Mesenchymal stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell Biology ,Cell biology ,Enzyme Activation ,ErbB Receptors ,Endocrinology ,Molecular Medicine ,Stem cell ,Developmental Biology - 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
- 2012
46. Lipids promote survival, proliferation, and maintenance of differentiation of rat liver sinusoidal endothelial cells in vitro
- Author
-
Linda G. Griffith, Douglas A. Lauffenburger, Donna B. Stolz, Ta-Chun Hang, Massachusetts Institute of Technology. Department of Biological Engineering, Griffith, Linda G., Hang, Ta-Chun, and Lauffenburger, Douglas A.
- Subjects
Male ,Programmed cell death ,Physiology ,Liver cytology ,Cell Survival ,MAP Kinase Signaling System ,Cellular differentiation ,Morpholines ,MAP Kinase Kinase 2 ,MAP Kinase Kinase 1 ,Biology ,Fatty Acids, Nonesterified ,Culture Media, Serum-Free ,chemistry.chemical_compound ,Phosphatidylinositol 3-Kinases ,Physiology (medical) ,Proliferating Cell Nuclear Antigen ,Animals ,Phosphatidylinositol ,Phosphorylation ,Protein kinase B ,Cell Proliferation ,Phosphoinositide-3 Kinase Inhibitors ,Mitogen-Activated Protein Kinase 1 ,Confluency ,Mitogen-Activated Protein Kinase 3 ,Hepatology ,Cell Death ,Cell growth ,Receptors, IgG ,Gastroenterology ,Diphenylamine ,Endothelial Cells ,Cell Differentiation ,Endocytosis ,Rats, Inbred F344 ,Cell biology ,Rats ,Platelet Endothelial Cell Adhesion Molecule-1 ,Liver and Biliary Tract ,chemistry ,Bromodeoxyuridine ,Liver ,Chromones ,Benzamides ,Microscopy, Electron, Scanning ,Proto-Oncogene Proteins c-akt ,Oleic Acid - 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
- 2011
47. ADAM9 inhibition increases membrane activity of ADAM10 and controls α-secretase processing of amyloid precursor protein
- Author
-
Stefan F. Lichtenthaler, Qing-Xiang Amy Sang, Taheera Ferdous, Robert M. Petrovich, Bart De Strooper, Marcia L. Moss, Ina Tesseur, Gary Powell, Colin Dingwall, Jörg W. Bartsch, Bin Qi, Douglas A. Lauffenburger, Julia Li Zhong, Uwe Schlomann, Linda G. Griffith, Miles A. Miller, Mara Taverna, Lori L. Edwards, Pei Zhou, Massachusetts Institute of Technology. Department of Biological Engineering, Miller, Miles Aaron, Griffith, Linda G., and Lauffenburger, Douglas A.
- Subjects
chemistry [ADAM Proteins] ,ADAM10 protein, human ,ADAM10 ,Protein Array Analysis ,ADAM9 protein, human ,Biochemistry ,metabolism [Cell Membrane] ,antagonists & inhibitors [Membrane Proteins] ,ADAM10 Protein ,Amyloid beta-Protein Precursor ,Epidermal growth factor ,Cell Line, Tumor ,metabolism [Amyloid beta-Protein Precursor] ,Membrane activity ,Amyloid precursor protein ,Humans ,chemistry [Membrane Proteins] ,Protease Inhibitors ,Molecular Biology ,enzymology [Cell Membrane] ,biology ,Cell Membrane ,P3 peptide ,Membrane Proteins ,Cell Biology ,ADAM Proteins ,metabolism [Amyloid Precursor Protein Secretases] ,Peptide Fragments ,Protein Structure, Tertiary ,metabolism [ADAM Proteins] ,ddc:540 ,pharmacology [Peptide Fragments] ,Enzymology ,Biocatalysis ,biology.protein ,drug effects [Cell Membrane] ,drug effects [Biocatalysis] ,Amyloid Precursor Protein Secretases ,pharmacology [Protease Inhibitors] ,ADAM9 ,Amyloid precursor protein secretase ,metabolism [Membrane Proteins] ,antagonists & inhibitors [ADAM Proteins] - Abstract
Prodomains of A disintegrin and metalloproteinase (ADAM) metallopeptidases can act as highly specific intra- and intermolecular inhibitors of ADAM catalytic activity. The mouse ADAM9 prodomain (proA9; amino acids 24–204), expressed and characterized from Escherichia coli, is a competitive inhibitor of human ADAM9 catalytic/disintegrin domain with an overall inhibition constant of 280 ± 34 nm and high specificity toward ADAM9. In SY5Y neuroblastoma cells overexpressing amyloid precursor protein, proA9 treatment reduces the amount of endogenous ADAM10 enzyme in the medium while increasing membrane-bound ADAM10, as shown both by Western and activity assays with selective fluorescent peptide substrates using proteolytic activity matrix analysis. An increase in membrane-bound ADAM10 generates higher levels of soluble amyloid precursor protein α in the medium, whereas soluble amyloid precursor protein β levels are decreased, demonstrating that inhibition of ADAM9 increases α-secretase activity on the cell membrane. Quantification of physiological ADAM10 substrates by a proteomic approach revealed that substrates, such as epidermal growth factor (EGF), HER2, osteoactivin, and CD40-ligand, are increased in the medium of BT474 breast tumor cells that were incubated with proA9, demonstrating that the regulation of ADAM10 by ADAM9 applies for many ADAM10 substrates. Taken together, our results demonstrate that ADAM10 activity is regulated by inhibition of ADAM9, and this regulation may be used to control shedding of amyloid precursor protein by enhancing α-secretase activity, a key regulatory step in the etiology of Alzheimer disease., National Institutes of Health (U.S.) (Grant R01EB010246), National Institutes of Health (U.S.) (Grant R01GM081336), Heptagon Fund (London, England), Cancer Research UK, Whitehead Foundation, Duke University. School of Medicine (Bridge Funding Program), Germany. Bundesministerium für Bildung und Forschung, China (National Fellowship from the Chinese Scholarship Council), Florida State University
- Published
- 2011
- Full Text
- View/download PDF
48. Enhancing Protease Activity Assay in Droplet-Based Microfluidics Using a Biomolecule Concentrator
- Author
-
Linda G. Griffith, Sung Jae Kim, Miles A. Miller, Aniruddh Sarkar, Jongyoon Han, Yong Ak Song, Douglas A. Lauffenburger, Chia-Hung Chen, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Electrical Engineering and Computer Science, Goyal, Chia-Hung, Sarkar, Aniruddh, Song, Yong-Ak, Miller, Miles Aaron, Kim, Sung Jae, Griffith, Linda G., Lauffenburger, Douglas A., and Han, Jongyoon
- Subjects
medicine.medical_treatment ,Microfluidics ,Nanotechnology ,Concentrator ,Biochemistry ,Sensitivity and Specificity ,Catalysis ,Article ,Sample volume ,Mice ,Colloid and Surface Chemistry ,medicine ,Animals ,Cells, Cultured ,chemistry.chemical_classification ,Protease ,Extramural ,Biomolecule ,General Chemistry ,Fibroblasts ,Microfluidic Analytical Techniques ,Matrix Metalloproteinases ,chemistry ,Droplet-based microfluidics ,Microreactor - Abstract
We introduce an integrated microfluidic device consisting of a biomolecule concentrator and a microdroplet generator, which enhances the limited sensitivity of low-abundance enzyme assays by concentrating biomolecules before encapsulating them into droplet microreactors. We used this platform to detect ultralow levels of matrix metalloproteinases (MMPs) from diluted cellular supernatant and showed that it significantly (~10-fold) reduced the time required to complete the assay and the sample volume used., National Institutes of Health (U.S.) (Grant GM68762), National Institutes of Health (U.S.) (Grant U54-CA112967), National Institutes of Health (U.S.) (Grant R01-EB010246), National Institutes of Health (U.S.) (Grant R01-GM081336), National Science Foundation (U.S.) (Graduate Fellowship), United States. Defense Advanced Research Projects Agency (Cipher Program)
- Published
- 2011
49. Marrow-Derived Stem Cell Motility in 3D Synthetic Scaffold Is Governed by Geometry Along With Adhesivity and Stiffness
- Author
-
Shelly R. Peyton, Douglas A. Lauffenburger, Anne P. Runkle, Joshua C. Cohen, Krystyn J. Van Vliet, Z. Ilke Kalcioglu, Linda G. Griffith, 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., and Griffith, Linda G.
- Subjects
Scaffold ,Materials science ,Motility ,Stiffness ,Bioengineering ,Nanotechnology ,Cell migration ,Hematopoietic Stem Cells ,Applied Microbiology and Biotechnology ,Article ,chemistry.chemical_compound ,chemistry ,Cell Movement ,Biophysics ,medicine ,Cell Adhesion ,Humans ,Progenitor cell ,medicine.symptom ,Cell adhesion ,Elastic modulus ,Ethylene glycol ,Biotechnology ,Cell Line, Transformed - 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
- 2010
50. Growth factor regulation of proliferation and survival of multipotential stromal cells
- Author
-
Melanie Rodrigues, Alan Wells, Linda G. Griffith, Massachusetts Institute of Technology. Department of Biological Engineering, and Griffith, Linda G.
- Subjects
Vascular Endothelial Growth Factor A ,Stromal cell ,Cell Survival ,medicine.medical_treatment ,Cellular differentiation ,Cell- and Tissue-Based Therapy ,Medicine (miscellaneous) ,Motility ,Review ,Biology ,Mesenchymal Stem Cell Transplantation ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Transforming Growth Factor beta ,medicine ,Humans ,Regeneration ,Progenitor cell ,Cell Proliferation ,Platelet-Derived Growth Factor ,Epidermal Growth Factor ,Hepatocyte Growth Factor ,Cell growth ,Growth factor ,Mesenchymal stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell Biology ,Cell biology ,Fibroblast Growth Factors ,Wnt Proteins ,Intercellular Signaling Peptides and Proteins ,Molecular Medicine ,Stem cell - Abstract
Multipotential stromal cells (MSCs) have been touted to provide an alternative to conservative procedures of therapy, be it heart transplants, bone reconstruction, kidney grafts, or skin, neuronal and cartilage repair. A wide gap exists, however, between the number of MSCs that can be obtained from the donor site and the number of MSCs needed for implantation to regenerate tissue. Standard methods of MSC expansion being followed in laboratories are not fully suitable due to time and age-related constraints for autologous therapies, and transplant issues leave questions for allogenic therapies. Beyond these issues of sufficient numbers, there also exists a problem of MSC survival at the graft. Experiments in small animals have shown that MSCs do not persist well in the graft environment. Either there is no incorporation into the host tissue, or, if there is incorporation, most of the cells are lost within a month. The use of growth and other trophic factors may be helpful in counteracting these twin issues of MSC expansion and death. Growth factors are known to influence cell proliferation, motility, survival and morphogenesis. In the case of MSCs, it would be beneficial that the growth factor does not induce differentiation at an early stage since the number of early-differentiating progenitors would be very low. The present review looks at the effect of and downstream signaling of various growth factors on proliferation and survival in MSCs.
- Published
- 2010
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.