Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Tissue Eng Part A ; 30(13-14): 333-341, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38126301

ABSTRACT

Tissues on a chip are sophisticated three-dimensional (3D) in vitro microphysiological systems designed to replicate human tissue conditions within dynamic physicochemical environments. However, the current fabrication methods for tissue spheroids on a chip require multiple parts and manual processing steps, including the deposition of spheroids onto prefabricated "chips." These challenges also lead to limitations regarding scalability and reproducibility. To overcome these challenges, we employed 3D printing techniques to automate the fabrication process of tissue spheroids on a chip. This allowed the simultaneous high-throughput printing of human liver spheroids and their surrounding polymeric flow chamber "chips" containing inner channels in a single step. The fabricated liver tissue spheroids on a liver-on-a-chip (LOC) were subsequently subjected to dynamic culturing by a peristaltic pump, enabling assessment of cell viability and metabolic activities. The 3D printed liver spheroids within the printed chips demonstrated high cell viability (>80%), increased spheroid size, and consistent adenosine triphosphate (ATP) activity and albumin production for up to 14 days. Furthermore, we conducted a study on the effects of acetaminophen (APAP), a nonsteroidal anti-inflammatory drug, on the LOC. Comparative analysis revealed a substantial decline in cell viability (<40%), diminished ATP activity, and reduced spheroid size after 7 days of culture within the APAP-treated LOC group, compared to the nontreated groups. These results underscore the potential of 3D bioprinted tissue chips as an advanced in vitro model that holds promise for accurately studying in vivo biological processes, including the assessment of tissue response to administered drugs, in a high-throughput manner.


Subject(s)
Bioprinting , Lab-On-A-Chip Devices , Liver , Printing, Three-Dimensional , Spheroids, Cellular , Humans , Spheroids, Cellular/drug effects , Spheroids, Cellular/cytology , Liver/drug effects , Liver/cytology , Bioprinting/methods , Acetaminophen/pharmacology , Drug Evaluation, Preclinical , Cell Survival/drug effects
2.
Biodes Manuf ; 5(1): 43-63, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35223131

ABSTRACT

The fields of regenerative medicine and tissue engineering offer new therapeutic options to restore, maintain or improve tissue function following disease or injury. To maximize the biological function of a tissue-engineered clinical product, specific conditions must be maintained within a bioreactor to allow the maturation of the product in preparation for implantation. Specifically, the bioreactor should be designed to mimic the mechanical, electrochemical and biochemical environment that the product will be exposed to in vivo. Real-time monitoring of the functional capacity of tissue-engineered products during manufacturing is a critical component of the quality management process. The present review provides a brief overview of bioreactor engineering considerations. In addition, strategies for bioreactor automation, in-line product monitoring and quality assurance are discussed.

3.
Biofabrication ; 12(2): 025017, 2020 02 26.
Article in English | MEDLINE | ID: mdl-32101533

ABSTRACT

Current practices in drug development have led to therapeutic compounds being approved for widespread use in humans, only to be later withdrawn due to unanticipated toxicity. These occurrences are largely the result of erroneous data generated by in vivo and in vitro preclinical models that do not accurately recapitulate human physiology. Herein, a human primary cell- and stem cell-derived 3D organoid technology is employed to screen a panel of drugs that were recalled from market by the FDA. The platform is comprised of multiple tissue organoid types that remain viable for at least 28 days, in vitro. For many of these compounds, the 3D organoid system was able to demonstrate toxicity. Furthermore, organoids exposed to non-toxic compounds remained viable at clinically relevant doses. Additional experiments were performed on integrated multi-organoid systems containing liver, cardiac, lung, vascular, testis, colon, and brain. These integrated systems proved to maintain viability and expressed functional biomarkers, long-term. Examples are provided that demonstrate how multi-organoid 'body-on-a-chip' systems may be used to model the interdependent metabolism and downstream effects of drugs across multiple tissues in a single platform. Such 3D in vitro systems represent a more physiologically relevant model for drug screening and will likely reduce the cost and failure rate associated with the approval of new drugs.


Subject(s)
Cell Culture Techniques/methods , Organoids/physiology , Pharmaceutical Preparations/metabolism , Astemizole/pharmacology , Capecitabine/pharmacology , Cell Culture Techniques/instrumentation , Cell Survival/drug effects , Cells, Cultured , Heart Rate/drug effects , Humans , Lab-On-A-Chip Devices , Liver/cytology , Liver/drug effects , Liver/metabolism , Myocardium/cytology , Myocardium/metabolism , Organoids/cytology , Organoids/drug effects , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism
4.
FASEB J ; 33(11): 12435-12446, 2019 11.
Article in English | MEDLINE | ID: mdl-31419161

ABSTRACT

Fibrosis is an underlying cause of cirrhosis and hepatic failure resulting in end stage liver disease with limited pharmacological options. The beneficial effects of relaxin peptide treatment were demonstrated in clinically relevant animal models of liver fibrosis. However, the use of relaxin is problematic because of a short half-life. The aim of this study was to test the therapeutic effects of recently identified small molecule agonists of the human relaxin receptor, relaxin family peptide receptor 1 (RXFP1). The lead compound of this series, ML290, was selected based on its effects on the expression of fibrosis-related genes in primary human stellate cells. RNA sequencing analysis of TGF-ß1-activated LX-2 cells showed that ML290 treatment primarily affected extracellular matrix remodeling and cytokine signaling, with expression profiles indicating an antifibrotic effect of ML290. ML290 treatment in human liver organoids with LPS-induced fibrotic phenotype resulted in a significant reduction of type I collagen. The pharmacokinetics of ML290 in mice demonstrated its high stability in vivo, as evidenced by the sustained concentrations of compound in the liver. In mice expressing human RXFP1 gene treated with carbon tetrachloride, ML290 significantly reduced collagen content, α-smooth muscle actin expression, and cell proliferation around portal ducts. In conclusion, ML290 demonstrated antifibrotic effects in liver fibrosis.-Kaftanovskaya, E. M., Ng, H. H., Soula, M., Rivas, B., Myhr, C., Ho, B. A., Cervantes, B. A., Shupe, T. D., Devarasetty, M., Hu, X., Xu, X., Patnaik, S., Wilson, K. J., Barnaeva, E., Ferrer, M., Southall, N. T., Marugan, J. J., Bishop, C. E., Agoulnik, I. U., Agoulnik, A. I. Therapeutic effects of a small molecule agonist of the relaxin receptor ML290 in liver fibrosis.


Subject(s)
Carbon Tetrachloride Poisoning/drug therapy , Cell Proliferation/drug effects , Liver Cirrhosis/drug therapy , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, Peptide/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Carbon Tetrachloride Poisoning/genetics , Cell Line, Transformed , Cell Proliferation/genetics , Cytokines/genetics , Cytokines/metabolism , Humans , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Mice , Mice, Transgenic , Organoids/metabolism , Organoids/pathology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/genetics , Receptors, Peptide/metabolism , Signal Transduction/genetics
5.
Stem Cell Res Ther ; 9(1): 304, 2018 11 08.
Article in English | MEDLINE | ID: mdl-30409188

ABSTRACT

BACKGROUND: Autologous urothelial cells are often obtained via bladder biopsy to generate the bio-engineered urethra or bladder, while urine-derived stem cells (USC) can be obtained by a non-invasive approach. The objective of this study is to develop an optimal strategy for urothelium with permeability barrier properties using human USC which could be used for tissue repair in the urinary tract system. METHODS: USC were harvested from six healthy adult individuals. To optimize urothelial differentiation, five different differentiation methods were studied. The induced cells were assessed for gene and protein expression markers of urothelial cells via RT-PCR, Western blotting, and immunofluorescent staining. Barrier function and ultrastructure of the tight junction were assessed with permeability assays and transmission electron microscopy (TEM). Induced cells were both cultured on trans-well membranes and small intestinal submucosa, then investigated under histology analysis. RESULTS: Differentiated USC expressed significantly higher levels of urothelial-specific transcripts and proteins (Uroplakin III and Ia), epithelial cell markers (CK20 and AE1/AE3), and tight junction markers (ZO-1, ZO-2, E-cadherin, and Cingulin) in a time-dependent manner, compared to non-induced USC. In vitro assays using fluorescent dye demonstrated a significant reduction in permeability of differentiated USC. In addition, transmission electron microscopy confirmed appropriate ultrastructure of urothelium differentiated from USC, including tight junction formation between neighboring cells, which was similar to positive controls. Furthermore, multilayered urothelial tissues formed 2 weeks after USC were differentiated on intestine submucosal matrix. CONCLUSION: The present study illustrates an optimal strategy for the generation of differentiated urothelium from stem cells isolated from the urine. The induced urothelium is phenotypically and functionally like native urothelium and has proposed uses in in vivo urological tissue repair or in vitro urethra or bladder modeling.


Subject(s)
Cell Differentiation , Stem Cells/cytology , Urinary Tract/metabolism , Urine/cytology , Urothelium/physiology , Adult , Animals , Biomarkers/metabolism , Cell Membrane Permeability , Cell Proliferation , Cell Shape , Collagen/metabolism , Humans , Male , Middle Aged , Stem Cells/ultrastructure , Swine , Tight Junctions/metabolism , Tight Junctions/ultrastructure
6.
Stem Cells Transl Med ; 7(8): 564-568, 2018 08.
Article in English | MEDLINE | ID: mdl-30009571

ABSTRACT

Regenerative medicine is poised to become a significant industry within the medical field. As such, the development of strategies and technologies for standardized and automated regenerative medicine clinical manufacturing has become a priority. An industry-driven roadmap toward industrial scale clinical manufacturing was developed over a 3-year period by a consortium of companies with significant investment in the field of regenerative medicine. Additionally, this same group identified critical roadblocks that stand in the way of advanced, large-scale regenerative medicine clinical manufacturing. This perspective article details efforts to reach a consensus among industry stakeholders on the shortest pathway for providing access to regenerative medicine therapies for those in need, both within the United States and around the world. Stem Cells Translational Medicine 2018;7:564-568.


Subject(s)
Bioreactors , Regenerative Medicine , Automation , Biocompatible Materials/chemistry , Bioreactors/standards , Culture Media/chemistry , Humans , Industry , Quality Control , Regenerative Medicine/standards , Tissue Engineering
7.
Front Public Health ; 6: 103, 2018.
Article in English | MEDLINE | ID: mdl-29755963

ABSTRACT

INTRODUCTION: Environmental toxins, such as lead and other heavy metals, pesticides, and other compounds, represent a significant health concern within the USA and around the world. Even in the twenty-first century, a plethora of cities and towns in the U.S. have suffered from exposures to lead in drinking water or other heavy metals in food or the earth, while there is a high possibility of further places to suffer such exposures in the near future. METHODS: We employed bioengineered 3D human liver and cardiac organoids to screen a panel of environmental toxins (lead, mercury, thallium, and glyphosate), and charted the response of the organoids to these compounds. Liver and cardiac organoids were exposed to lead (10 µM-10 mM), mercury (200 nM-200 µM), thallium (10 nM-10 µM), or glyphosate (25 µM-25 mM) for a duration of 48 h. The impacts of toxin exposure were then assessed by LIVE/DEAD viability and cytotoxicity staining, measuring ATP activity and determining IC50 values, and determining changes in cardiac organoid beating activity. RESULTS: As expected, all of the toxins induced toxicity in the organoids. Both ATP and LIVE/DEAD assays showed toxicity in both liver and cardiac organoids. In particular, thallium was the most toxic, with IC50 values of 13.5 and 1.35 µM in liver and cardiac organoids, respectively. Conversely, glyphosate was the least toxic of the four compounds, with IC50 values of 10.53 and 10.85 mM in liver and cardiac organoids, respectively. Additionally, toxins had a negative influence on cardiac organoid beating activity as well. Thallium resulting in the most significant decreases in beating rate, followed by mercury, then glyphosate, and finally, lead. These results suggest that the 3D organoids have significant utility to be deployed in additional toxicity screening applications, and future development of treatments to mitigate exposures. CONCLUSION: 3D organoids have significant utility to be deployed in additional toxicity screening applications, such as future development of treatments to mitigate exposures, drug screening, and environmental toxin detection.

8.
Sci Rep ; 7(1): 8837, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28821762

ABSTRACT

Many drugs have progressed through preclinical and clinical trials and have been available - for years in some cases - before being recalled by the FDA for unanticipated toxicity in humans. One reason for such poor translation from drug candidate to successful use is a lack of model systems that accurately recapitulate normal tissue function of human organs and their response to drug compounds. Moreover, tissues in the body do not exist in isolation, but reside in a highly integrated and dynamically interactive environment, in which actions in one tissue can affect other downstream tissues. Few engineered model systems, including the growing variety of organoid and organ-on-a-chip platforms, have so far reflected the interactive nature of the human body. To address this challenge, we have developed an assortment of bioengineered tissue organoids and tissue constructs that are integrated in a closed circulatory perfusion system, facilitating inter-organ responses. We describe a three-tissue organ-on-a-chip system, comprised of liver, heart, and lung, and highlight examples of inter-organ responses to drug administration. We observe drug responses that depend on inter-tissue interaction, illustrating the value of multiple tissue integration for in vitro study of both the efficacy of and side effects associated with candidate drugs.


Subject(s)
Lab-On-A-Chip Devices , Tissue Array Analysis , Drug Discovery/methods , Equipment Design , Heart , Humans , Liver/drug effects , Liver/metabolism , Lung/drug effects , Lung/metabolism , Microfluidics/instrumentation , Microfluidics/methods , Organoids/drug effects , Organoids/metabolism , Tissue Array Analysis/instrumentation , Tissue Array Analysis/methods
9.
Biosensors (Basel) ; 7(3)2017 Jun 23.
Article in English | MEDLINE | ID: mdl-28644395

ABSTRACT

Organoid and organ-on-a-chip technologies are rapidly advancing towards deployment for drug and toxicology screening applications. Liver and cardiac toxicities account for the majority of drug candidate failures in human trials. Liver toxicity generally produces liver cell death, while cardiac toxicity causes adverse changes in heart beat kinetics. In traditional 2D cultures, beating kinetics can be measured by electrode arrays, but in some 3D constructs, quantifying beating kinetics can be more challenging. For example, real time measurements of calcium flux or contractile forces are possible, yet rather complex. In this communication article, we demonstrate a simple sensing system based on software code that optically analyzes video capture files of beating cardiac organoids, translates these files in representations of moving pixels, and quantifies pixel movement activity over time to generate beat kinetic plots. We demonstrate this system using bioengineered cardiac organoids under baseline and drug conditions. This technology offers a non-invasive, low-cost, and incredibly simple method for tracking and quantifying beating behavior in cardiac organoids and organ-on-a-chip systems for drug and toxicology screening.


Subject(s)
Biosensing Techniques/methods , Drug-Related Side Effects and Adverse Reactions , Myocytes, Cardiac/drug effects , Organoids/drug effects , Biosensing Techniques/instrumentation , Calcium/metabolism , Heart/drug effects , Humans , Liver/drug effects
10.
Proc Natl Acad Sci U S A ; 114(12): E2293-E2302, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28265064

ABSTRACT

Organ-on-a-chip systems are miniaturized microfluidic 3D human tissue and organ models designed to recapitulate the important biological and physiological parameters of their in vivo counterparts. They have recently emerged as a viable platform for personalized medicine and drug screening. These in vitro models, featuring biomimetic compositions, architectures, and functions, are expected to replace the conventional planar, static cell cultures and bridge the gap between the currently used preclinical animal models and the human body. Multiple organoid models may be further connected together through the microfluidics in a similar manner in which they are arranged in vivo, providing the capability to analyze multiorgan interactions. Although a wide variety of human organ-on-a-chip models have been created, there are limited efforts on the integration of multisensor systems. However, in situ continual measuring is critical in precise assessment of the microenvironment parameters and the dynamic responses of the organs to pharmaceutical compounds over extended periods of time. In addition, automated and noninvasive capability is strongly desired for long-term monitoring. Here, we report a fully integrated modular physical, biochemical, and optical sensing platform through a fluidics-routing breadboard, which operates organ-on-a-chip units in a continual, dynamic, and automated manner. We believe that this platform technology has paved a potential avenue to promote the performance of current organ-on-a-chip models in drug screening by integrating a multitude of real-time sensors to achieve automated in situ monitoring of biophysical and biochemical parameters.


Subject(s)
Automation/methods , Biosensing Techniques/methods , Drug Evaluation, Preclinical/methods , Organoids/physiology , Automation/instrumentation , Biosensing Techniques/instrumentation , Drug Evaluation, Preclinical/instrumentation , Heart/physiology , Humans , Liver/chemistry , Liver/physiology , Microfluidics , Models, Biological , Myocardium , Organoids/chemistry , Organoids/drug effects
11.
Biomaterials ; 110: 45-59, 2016 12.
Article in English | MEDLINE | ID: mdl-27710832

ABSTRACT

Engineering cardiac tissues and organ models remains a great challenge due to the hierarchical structure of the native myocardium. The need of integrating blood vessels brings additional complexity, limiting the available approaches that are suitable to produce integrated cardiovascular organoids. In this work we propose a novel hybrid strategy based on 3D bioprinting, to fabricate endothelialized myocardium. Enabled by the use of our composite bioink, endothelial cells directly bioprinted within microfibrous hydrogel scaffolds gradually migrated towards the peripheries of the microfibers to form a layer of confluent endothelium. Together with controlled anisotropy, this 3D endothelial bed was then seeded with cardiomyocytes to generate aligned myocardium capable of spontaneous and synchronous contraction. We further embedded the organoids into a specially designed microfluidic perfusion bioreactor to complete the endothelialized-myocardium-on-a-chip platform for cardiovascular toxicity evaluation. Finally, we demonstrated that such a technique could be translated to human cardiomyocytes derived from induced pluripotent stem cells to construct endothelialized human myocardium. We believe that our method for generation of endothelialized organoids fabricated through an innovative 3D bioprinting technology may find widespread applications in regenerative medicine, drug screening, and potentially disease modeling.


Subject(s)
Bioprinting/methods , Endothelial Cells , Myocardium , Organoids/growth & development , Printing, Three-Dimensional , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Drug Evaluation, Preclinical , Endothelial Cells/chemistry , Endothelial Cells/cytology , Humans , Hydrogels/chemistry , Microfibrils/chemistry , Myocytes, Cardiac/chemistry , Myocytes, Cardiac/metabolism , Organoids/chemistry , Organoids/metabolism , Regenerative Medicine
12.
Drug Discov Today ; 21(9): 1399-1411, 2016 09.
Article in English | MEDLINE | ID: mdl-27422270

ABSTRACT

In recent years, advances in tissue engineering and microfabrication technologies have enabled rapid growth in the areas of in vitro organoid development as well as organoid-on-a-chip platforms. These 3D model systems often are able to mimic human physiology more accurately than traditional 2D cultures and animal models. In this review, we describe the progress that has been made to generate organ-on-a-chip platforms and, more recently, more complex multi-organoid body-on-a-chip platforms and their applications. Importantly, these systems have the potential to dramatically impact biomedical applications in the areas of drug development, drug and toxicology screening, disease modeling, and the emerging area of personalized precision medicine.


Subject(s)
Drug Evaluation, Preclinical/methods , Organoids , Humans , Models, Biological , Tissue Engineering
13.
J Vis Exp ; (110): e53606, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27166839

ABSTRACT

Bioprinting has emerged as a versatile biofabrication approach for creating tissue engineered organ constructs. These constructs have potential use as organ replacements for implantation in patients, and also, when created on a smaller size scale as model "organoids" that can be used in in vitro systems for drug and toxicology screening. Despite development of a wide variety of bioprinting devices, application of bioprinting technology can be limited by the availability of materials that both expedite bioprinting procedures and support cell viability and function by providing tissue-specific cues. Here we describe a versatile hyaluronic acid (HA) and gelatin-based hydrogel system comprised of a multi-crosslinker, 2-stage crosslinking protocol, which can provide tissue specific biochemical signals and mimic the mechanical properties of in vivo tissues. Biochemical factors are provided by incorporating tissue-derived extracellular matrix materials, which include potent growth factors. Tissue mechanical properties are controlled combinations of PEG-based crosslinkers with varying molecular weights, geometries (linear or multi-arm), and functional groups to yield extrudable bioinks and final construct shear stiffness values over a wide range (100 Pa to 20 kPa). Using these parameters, hydrogel bioinks were used to bioprint primary liver spheroids in a liver-specific bioink to create in vitro liver constructs with high cell viability and measurable functional albumin and urea output. This methodology provides a general framework that can be adapted for future customization of hydrogels for biofabrication of a wide range of tissue construct types.


Subject(s)
Bioprinting/methods , Hydrogel, Polyethylene Glycol Dimethacrylate , Tissue Scaffolds , Bioprinting/instrumentation , Cell Survival , Extracellular Matrix , Gelatin/chemistry , Humans , Hyaluronic Acid/chemistry , Hydrogels/chemistry , Polyethylene Glycols/chemistry , Tissue Engineering/methods
14.
Biofabrication ; 8(1): 014101, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26756674

ABSTRACT

The inadequacy of animal models in correctly predicting drug and biothreat agent toxicity in humans has resulted in a pressing need for in vitro models that can recreate the in vivo scenario. One of the most important organs in the assessment of drug toxicity is liver. Here, we report the development of a liver-on-a-chip platform for long-term culture of three-dimensional (3D) human HepG2/C3A spheroids for drug toxicity assessment. The bioreactor design allowed for in situ monitoring of the culture environment by enabling direct access to the hepatic construct during the experiment without compromising the platform operation. The engineered bioreactor could be interfaced with a bioprinter to fabricate 3D hepatic constructs of spheroids encapsulated within photocrosslinkable gelatin methacryloyl (GelMA) hydrogel. The engineered hepatic construct remained functional during the 30 days culture period as assessed by monitoring the secretion rates of albumin, alpha-1 antitrypsin, transferrin, and ceruloplasmin, as well as immunostaining for the hepatocyte markers, cytokeratin 18, MRP2 bile canalicular protein and tight junction protein ZO-1. Treatment with 15 mM acetaminophen induced a toxic response in the hepatic construct that was similar to published studies on animal and other in vitro models, thus providing a proof-of-concept demonstration of the utility of this liver-on-a-chip platform for toxicity assessment.


Subject(s)
Biological Assay/instrumentation , Chemical and Drug Induced Liver Injury/etiology , Lab-On-A-Chip Devices , Liver, Artificial , Printing, Three-Dimensional/instrumentation , Toxicity Tests/instrumentation , Chemical and Drug Induced Liver Injury/pathology , Equipment Design , Equipment Failure Analysis , Hep G2 Cells , Humans , Organ Culture Techniques/instrumentation , Spheroids, Cellular/drug effects
15.
J Mech Behav Biomed Mater ; 55: 87-103, 2015 Mar.
Article in English | MEDLINE | ID: mdl-26569044

ABSTRACT

Tissue engineering and cell based liver therapies have utilized primary hepatocytes with limited success due to the failure of hepatocytes to maintain their phenotype in vitro. In order to overcome this challenge, hyaluronic acid (HA) cell culture substrates were formulated to closely mimic the composition and stiffness of the normal liver cellular microenvironment. The stiffness of the substrate was modulated by adjusting HA hydrogel crosslinking. Additionally, the repertoire of bioactive molecules within the HA substrate was bolstered by supplementation with normal liver extracellular matrix (ECM). Primary human hepatocyte viability and phenotype were determined over a narrow physiologically relevant range of substrate stiffnesses from 600 to 4600Pa in both the presence and absence of liver ECM. Cell attachment, viability, and organization of the actin cytoskeleton improved with increased stiffness up to 4600Pa. These differences were not evident in earlier time points or substrates containing only HA. However, gene expression for the hepatocyte markers hepatocyte nuclear factor 4 alpha (HNF4α) and albumin significantly decreased on the 4600Pa stiffness at day 7 indicating that cells may not have maintained their phenotype long-term at this stiffness. Function, as measured by albumin secretion, varied with both stiffness and time in culture and peaked at day 7 at the 1200Pa stiffness, slightly below the stiffness of normal liver ECM at 3000Pa. Overall, gel stiffness affected primary human hepatocyte cell adhesion, functional marker expression, and morphological characteristics dependent on both the presence of liver ECM in gel substrates and time in culture.


Subject(s)
Extracellular Matrix/metabolism , Hepatocytes/cytology , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Liver/cytology , Mechanical Phenomena , Tissue Engineering , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Biomarkers/metabolism , Cell Adhesion/drug effects , Cell Survival/drug effects , Cell Transplantation , Cytoplasm/drug effects , Cytoplasm/enzymology , Gene Expression Regulation/drug effects , Glycosaminoglycans/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Hydrogels/chemistry , Protein Kinases/metabolism , Rats , Solubility , rho GTP-Binding Proteins/metabolism
16.
Acta Biomater ; 25: 24-34, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26210285

ABSTRACT

Advancement of bioprinting technology is limited by the availability of materials that both facilitate bioprinting logistics as well as support cell viability and function by providing tissue-specific cues. Herein we describe a modular hyaluronic acid (HA) and gelatin-based hydrogel toolbox comprised of a 2-crosslinker, 2-stage polymerization technique, and the capability to provide tissue specific biochemically and mechanically accurate signals to cells within biofabricated tissue constructs. First, we prepared and characterized several tissue-derived decellularized extracellular matrix-based solutions, which contain complex combinations of growth factors, collagens, glycosaminoglycans, and elastin. These solutions can be incorporated into bioinks to provide the important biochemical cues of different tissue types. Second, we employed combinations of PEG-based crosslinkers with varying molecular weights, geometries (linear, 4-arm, and 8-arm), and functional groups to yield hydrogel bioinks that supported extrusion bioprinting and the capability to achieve final construct shear stiffness values ranging from approximately 100 Pa to 20 kPa. Lastly, we integrated these hydrogel bioinks with a 3-D bioprinting platform, and validated their use by bioprinting primary liver spheroids in a liver-specific bioink to create in vitro liver constructs with high cell viability and measurable functional albumin and urea output. This hydrogel bioink system has the potential to be a versatile tool for biofabrication of a wide range of tissue construct types. STATEMENT OF SIGNIFICANCE: Biochemical and mechanical factors both have important implications in guiding the behavior of cells in vivo, yet both realms are rarely considered together in the context of biofabrication in vitro tissue construct models. We describe a modular hydrogel system that (1) facilitates extrusion bioprinting of cell-laden hydrogels, (2) incorporates tissue-specific factors derived from decellularized tissue extracellular matrix, thus mimicking biochemical tissue profile, and (3) allows control over mechanical properties to mimic the tissue stiffness. We believe that employing this technology to attend to both the biochemical and mechanical profiles of tissues, will allow us to more accurately recapitulate the in vivo environment of tissues while creating functional 3-D in vitro tissue constructs that can be used as disease models, personalized medicine, and in vitro drug and toxicology screening systems.


Subject(s)
Bioprinting/methods , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Albumins/metabolism , Animals , Biomechanical Phenomena/drug effects , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Humans , Polyethylene Glycols/chemistry , Rheology/drug effects , Solutions , Spheroids, Cellular/cytology , Spheroids, Cellular/drug effects , Sus scrofa , Tissue Survival/drug effects , Urea/metabolism
18.
Biomaterials ; 40: 72-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25433603

ABSTRACT

Donor shortage remains a continued challenge in liver transplantation. Recent advances in tissue engineering have provided the possibility of creating functional liver tissues as an alternative to donor organ transplantation. Small bioengineered liver constructs have been developed, however a major challenge in achieving functional bioengineered liver in vivo is the establishment of a functional vasculature within the scaffolds. Our overall goal is to bioengineer intact livers, suitable for transplantation, using acellular porcine liver scaffolds. We developed an effective method for reestablishing the vascular network within decellularized liver scaffolds by conjugating anti-endothelial cell antibodies to maximize coverage of the vessel walls with endothelial cells. This procedure resulted in uniform endothelial attachment throughout the liver vasculature extending to the capillary bed of the liver scaffold and greatly reduced platelet adhesion upon blood perfusion in vitro. The re-endothelialized livers, when transplanted to recipient pigs, were able to withstand physiological blood flow and maintained for up to 24 h. This study demonstrates, for the first time, that vascularized bioengineered livers, of clinically relevant size, can be transplanted and maintained in vivo, and represents the first step towards generating engineered livers for transplantation to patients with end-stage liver failure.


Subject(s)
Bioengineering , Endothelium, Vascular/physiology , Liver Transplantation , Liver/blood supply , Liver/physiology , Animals , Female , Liver/ultrastructure , Prosthesis Implantation , Sus scrofa , Tissue Scaffolds , Vascular Patency
19.
Am J Pathol ; 183(2): 558-65, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23747949

ABSTRACT

Liver disease affects millions of patients each year. The field of regenerative medicine promises alternative therapeutic approaches, including the potential to bioengineer replacement hepatic tissue. One approach combines cells with acellular scaffolds derived from animal tissue. The goal of this study was to scale up our rodent liver decellularization method to livers of a clinically relevant size. Porcine livers were cannulated via the hepatic artery, then perfused with PBS, followed by successive Triton X-100 and SDS solutions in saline buffer. After several days of rinsing, decellularized liver samples were histologically analyzed. In addition, biopsy specimens of decellularized scaffolds were seeded with hepatoblastoma cells for cytotoxicity testing or implanted s.c. into rodents to investigate scaffold immunogenicity. Histological staining confirmed cellular clearance from pig livers, with removal of nuclei and cytoskeletal components and widespread preservation of structural extracellular molecules. Scanning electron microscopy confirmed preservation of an intact liver capsule, a porous acellular lattice structure with intact vessels and striated basement membrane. Liver scaffolds supported cells over 21 days, and no increased immune response was seen with either allogeneic (rat-into-rat) or xenogeneic (pig-into-rat) transplants over 28 days, compared with sham-operated on controls. These studies demonstrate that successful decellularization of the porcine liver could be achieved with protocols developed for rat livers, yielding nonimmunogenic scaffolds for future hepatic bioengineering studies.


Subject(s)
Liver/cytology , Regenerative Medicine/methods , Tissue Engineering/methods , Tissue Scaffolds , Animals , Liver/immunology , Liver Transplantation/immunology , Male , Rats , Rats, Inbred F344 , Sus scrofa , Swine , Transplantation, Heterologous
20.
Gut ; 62(5): 774-86, 2013 May.
Article in English | MEDLINE | ID: mdl-22267591

ABSTRACT

This review illustrates promising regenerative medicine technologies that are being developed for the treatment of gastrointestinal diseases. The main strategies under validation to bioengineer or regenerate liver, pancreas, or parts of the digestive tract are twofold: engineering of progenitor cells and seeding of cells on supporting scaffold material. In the first case, stem cells are initially expanded under standard tissue culture conditions. Thereafter, these cells may either be delivered directly to the tissue or organ of interest, or they may be loaded onto a synthetic or natural three-dimensional scaffold that is capable of enhancing cell viability and function. The new construct harbouring the cells usually undergoes a maturation phase within a bioreactor. Within the bioreactor, cells are conditioned to adopt a phenotype similar to that displayed in the native organ. The specific nature of the scaffold within the bioreactor is critical for the development of this high-function phenotype. Efforts to bioengineer or regenerate gastrointestinal tract, liver and pancreas have yielded promising results and have demonstrated the immense potential of regenerative medicine. However, a myriad of technical hurdles must be overcome before transplantable, engineered organs become a reality.


Subject(s)
Gastrointestinal Diseases/surgery , Regenerative Medicine , Stem Cell Transplantation , Tissue Engineering/methods , Bioengineering/trends , Gastrointestinal Diseases/pathology , Humans , Intestinal Diseases/therapy , Liver Failure/surgery , Liver Regeneration , Liver Transplantation/methods , Organ Transplantation , Pancreatic Diseases/surgery , Regenerative Medicine/trends , Tissue Scaffolds
SELECTION OF CITATIONS
SEARCH DETAIL
...