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










Publication year range
1.
ACS Biomater Sci Eng ; 8(11): 4629-4633, 2022 11 14.
Article in English | MEDLINE | ID: mdl-35679122

ABSTRACT

Many academic researchers are familiar with the technical challenges inherent in creating patient-specific, tissue-engineered therapeutics for clinical applications. However, for the potential of these products to mature into clinical practice and the profound possibilities to be realized, developers must apply not only technical expertise but also a comprehensive translational strategy─making a product marketable, scalable, manufacturable, and approvable by regulators, while providing genuine advantages to patients. Here, we provide a brief overview of three crucial steps for successful translation: adoption of a "translational mindset," consideration of three categories of core challenges (economic, regulatory, and manufacturing), and detailed planning at the earliest stages of development.


Subject(s)
Cell- and Tissue-Based Therapy , Tissue Engineering , Humans
2.
Nat Protoc ; 14(8): 2598, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30770888

ABSTRACT

The version of this paper originally published contained an incorrect unit abbreviation in Step 21: "0.20 g/mL" should have been "0.20 mg/mL." In addition, the first sentence in Step 33 should have read "Use a second pipette with a cut-off pipette tip to add Matrigel to the center well," instead of "Use a second pipette to cut off the tip of the pipette and add Matrigel to the center well." These errors have been corrected in the PDF and HTML versions of the protocol.

3.
Nat Protoc ; 13(11): 2601-2614, 2018 11.
Article in English | MEDLINE | ID: mdl-30349048

ABSTRACT

Live imaging of stem cells and their support cells can be used to visualize cellular dynamics and fluctuations of intracellular signals, proteins, and organelles in order to better understand stem cell behavior in the niche. We describe a simple protocol for imaging stem cells in the Drosophila ovary that improves on alternative protocols in that flies of any age can be used, dissection is simplified because the epithelial sheath that surrounds each ovariole need not be removed, and ovarioles are imaged in a closed chamber with a large volume of medium that buffers oxygen, pH, and temperature. We also describe how to construct the imaging chamber, which can be easily modified and used to image other tissues and non-adherent cells. Imaging is limited by follicle cells moving out of the germarium in culture around the time of egg chamber budding; however, the epithelial sheath delays this abnormal cell migration. This protocol requires an hour to prepare the ovarioles, followed by half an hour on the confocal microscope to locate germaria and set z limits. Successful imaging time depends on germarial morphology at the time of dissection, but we suggest 10-11 h to encompass all specimens.


Subject(s)
Drosophila melanogaster/cytology , Equipment Design , Ovary/cytology , Stem Cells/cytology , Time-Lapse Imaging/instrumentation , Animals , Cell Division , Cell Movement , Cell Tracking/methods , Collagen/chemistry , Culture Media/chemistry , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Drug Combinations , Female , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Laminin/chemistry , Oocytes/cytology , Oocytes/growth & development , Oocytes/metabolism , Oogenesis/physiology , Ovary/growth & development , Ovary/metabolism , Proteoglycans/chemistry , Silicon/chemistry , Stem Cells/metabolism , Time-Lapse Imaging/methods
4.
Nat Cell Biol ; 19(5): 433-444, 2017 May.
Article in English | MEDLINE | ID: mdl-28414313

ABSTRACT

Adult stem cells provide a renewable source of differentiated cells for a wide variety of tissues and generally give rise to multiple cell types. Basic principles of stem cell organization and regulation underlying this behaviour are emerging. Local niche signals maintain stem cells, while different sets of signals act outside the niche to diversify initially equivalent stem cell progeny. Here we show that Drosophila ovarian follicle stem cells (FSCs) produced two distinct cell types directly. This cell fate choice was determined by the anterior-posterior position of an FSC and by the magnitude of spatially graded Wnt pathway activity. These findings reveal a paradigm of immediate diversification of stem cell derivatives according to stem cell position within a larger population, guided by a graded niche signal. We also found that FSCs strongly resemble mammalian intestinal stem cells in many aspects of their organization, including population asymmetry and dynamic heterogeneity.


Subject(s)
Adult Stem Cells/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Ovarian Follicle/metabolism , Stem Cell Niche , Wnt Signaling Pathway , Animals , Animals, Genetically Modified , Cell Lineage , Cell Movement , Cell Proliferation , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Female , Genotype , Ovarian Follicle/cytology , Phenotype , Time Factors
5.
Exp Cell Res ; 320(1): 79-91, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24113575

ABSTRACT

Electrical signals have been implied in many biological mechanisms, including wound healing, which has been associated with transient electrical currents not present in intact skin. One method to generate electrical signals similar to those naturally occurring in wounds is by supplementation of galvanic particles dispersed in a cream or gel. We constructed a three-layered model of skin consisting of human dermal fibroblasts in hydrogel (mimic of dermis), a hydrogel barrier layer (mimic of epidermis) and galvanic microparticles in hydrogel (mimic of a cream containing galvanic particles applied to skin). Using this model, we investigated the effects of the properties and amounts of Cu/Zn galvanic particles on adult human dermal fibroblasts in terms of the speed of wound closing and gene expression. The collected data suggest that the effects on wound closing are due to the ROS-mediated enhancement of fibroblast migration, which is in turn mediated by the BMP/SMAD signaling pathway. These results imply that topical low-grade electric currents via microparticles could enhance wound healing.


Subject(s)
Cell Movement , Fibroblasts/cytology , Fibroblasts/metabolism , Models, Biological , Reactive Oxygen Species/metabolism , Skin/cytology , Wound Healing , Adult , Female , Galvanic Skin Response , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate , Particle Size , Surface Properties
6.
Article in English | MEDLINE | ID: mdl-24110015

ABSTRACT

The goal of this study was to simulate in vitro the spontaneous electrical wave activity associated with retinal development and investigate if such biometrically designed signals can enhance differentiation of mouse retinal progenitor cells (mRPC). To this end, we cultured cells on an electroconductive transplantable polymer, polypyrrole (PPy) and measured gene expression and morphology of the cells. Custom-made 8-well cell culture chambers were designed to accommodate PPy deposited onto indium tin oxide-coated (ITO) glass slides, with precise control of the PPy film thickness. mRPCs were isolated from post-natal day 1 (P1) green fluorescent protein positive (GFP+) mice, expanded, seeded onto PPY films, allowed to adhere for 24 hours, and then subjected to electrical stimulation (100 µA pulse trains, 5 s in duration, once per minute) for 4 days. Cultured cells and non-stimulated controls were processed for immunostaining and confocal analysis, and for RNA extraction and quantitative PCR. Stimulated cells expressed significantly higher levels of the early photoreceptor marker cone-rod homebox (CRX, the earliest known marker of photoreceptor identity), and protein kinase-C (PKC), and significantly lower levels of the glial fibrillary acidic protein (GFAP). Consistently, stimulated cells developed pronounced neuronal morphologies with significantly longer dendritic processes and larger cell bodies than non-stimulated controls. Taken together, the experimental evidence shows that the application of an electrical stimulation designed based on retinal development can be implemented to direct and enhance retinal differentiation of mRPCs, suggesting a role for biomimetic electrical stimulation in directing progenitor cells toward neural fates.


Subject(s)
Biocompatible Materials/chemistry , Electric Stimulation , Polymers/chemistry , Pyrroles/chemistry , Retina/cytology , Stem Cells/cytology , Animals , Cell Differentiation , Cell Size , Cells, Cultured , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Polymerase Chain Reaction , Protein Kinase C/genetics , Protein Kinase C/metabolism , Stem Cells/metabolism , Tin Compounds/chemistry , Trans-Activators/genetics , Trans-Activators/metabolism
7.
Article in English | MEDLINE | ID: mdl-24111023

ABSTRACT

Electrical activity is abundant in early retinal development, and electrical stimulation has been shown to modulate embryonic stem cell differentiation towards a neuronal fate. The goal of this study was to simulate in vitro retinal developmental electrical activity to drive changes in mouse retinal progenitor cell (mRPC) gene expression and morphology. We designed a biomimetic electrical stimulation protocol based on spontaneous waves present during retinal development, and applied it to retinal progenitor cells (RPCs) over 3 days of culture. Analysis of protein localization and calcium dynamics, indicate that mRPCs undergo functional neuronal maturation. Our findings suggest that this type of electrical stimulation may be utilized for application in neural tissue engineering and open possibilities for understanding mechanisms guiding active electric membrane development and functional organization during early retinogenesis.


Subject(s)
Biomimetic Materials , Electric Stimulation/instrumentation , Retina/cytology , Stem Cells/physiology , Tissue Engineering/instrumentation , Tissue Engineering/methods , Animals , Calcium Signaling , Cell Differentiation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism
8.
Article in English | MEDLINE | ID: mdl-24111161

ABSTRACT

Cardiac tissue engineering aims to create functional tissue constructs that can reestablish the structure and function of injured myocardium. Although bioreactors have facilitated the engineering of cardiac patches of clinically relevant size in vitro, a major drawback remains the transportation of the engineered tissues from a production facility to a medical operation facility while maintaining tissue viability and preventing contamination. Furthermore, after implantation, most of the cells are endangered by hypoxic conditions that exist before vascular flow is established. We developed a portable device that provides the perfusion and electrical stimulation necessary to engineer cardiac tissue in vitro, and to transport it to the site where it will be implantated. The micropump-powered perfusion apparatus may additionally function as an extracorporeal active pumping system providing nutrients and oxygen supply to the graft post-implantation. Such a system, through perfusion of oxygenated media and bioactive molecules (e.g. growth factors), could transiently support the tissue construct until it connects to the host vasculature and heart muscle, after which it could be taken away or let biodegrade.


Subject(s)
Bioreactors , Heart/physiology , Perfusion , Tissue Engineering/instrumentation , Animals , Collagen/chemistry , Electric Stimulation , Electrodes , Equipment Design , Models, Theoretical , Oxygen/analysis , Rats, Sprague-Dawley , Reproducibility of Results , Tissue Scaffolds
9.
Biotechnol Adv ; 31(7): 1020-31, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23531529

ABSTRACT

Stem cells hold promise to revolutionize modern medicine by the development of new therapies, disease models and drug screening systems. Standard cell culture systems have limited biological relevance because they do not recapitulate the complex 3-dimensional interactions and biophysical cues that characterize the in vivo environment. In this review, we discuss the current advances in engineering stem cell environments using novel biomaterials and bioreactor technologies. We also reflect on the challenges the field is currently facing with regard to the translation of stem cell based therapies into the clinic.


Subject(s)
Bioreactors , Cell Engineering , Stem Cell Niche , Stem Cells , Animals , High-Throughput Screening Assays , Humans , Mice , Models, Biological , Regenerative Medicine
10.
J Tissue Eng Regen Med ; 6(10): e12-23, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22170772

ABSTRACT

Maintenance of normal myocardial function depends intimately on synchronous tissue contraction, driven by electrical activation and on adequate nutrient perfusion in support thereof. Bioreactors have been used to mimic aspects of these factors in vitro to engineer cardiac tissue but, due to design limitations, previous bioreactor systems have yet to simultaneously support nutrient perfusion, electrical stimulation and unconstrained (i.e. not isometric) tissue contraction. To the best of our knowledge, the bioreactor system described herein is the first to integrate these three key factors in concert. We present the design of our bioreactor and characterize its capability in integrated experimental and mathematical modelling studies. We then cultured cardiac cells obtained from neonatal rats in porous, channelled elastomer scaffolds with the simultaneous application of perfusion and electrical stimulation, with controls excluding either one or both of these two conditions. After 8 days of culture, constructs grown with simultaneous perfusion and electrical stimulation exhibited substantially improved functional properties, as evidenced by a significant increase in contraction amplitude (0.23 ± 0.10% vs 0.14 ± 0.05%, 0.13 ± 0.08% or 0.09 ± 0.02% in control constructs grown without stimulation, without perfusion, or either stimulation or perfusion, respectively). Consistently, these constructs had significantly improved DNA contents, cell distribution throughout the scaffold thickness, cardiac protein expression, cell morphology and overall tissue organization compared to control groups. Thus, the simultaneous application of medium perfusion and electrical conditioning enabled by the use of the novel bioreactor system may accelerate the generation of fully functional, clinically sized cardiac tissue constructs.


Subject(s)
Biomimetic Materials , Bioreactors , Myocardial Contraction , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Tissue Engineering , Tissue Scaffolds , Animals , Electric Stimulation , Muscle Proteins/biosynthesis , Myocardium/cytology , Myocytes, Cardiac/cytology , Porosity , Rats , Rats, Sprague-Dawley
11.
J Tissue Eng Regen Med ; 5(6): e115-25, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21604379

ABSTRACT

In vitro application of pulsatile electrical stimulation to neonatal rat cardiomyocytes cultured on polymer scaffolds has been shown to improve the functional assembly of cells into contractile engineered cardiac tissues. However, to date, the conditions of electrical stimulation have not been optimized. We have systematically varied the electrode material, amplitude and frequency of stimulation to determine the conditions that are optimal for cardiac tissue engineering. Carbon electrodes, exhibiting the highest charge-injection capacity and producing cardiac tissues with the best structural and contractile properties, were thus used in tissue engineering studies. Engineered cardiac tissues stimulated at 3 V/cm amplitude and 3 Hz frequency had the highest tissue density, the highest concentrations of cardiac troponin-I and connexin-43 and the best-developed contractile behaviour. These findings contribute to defining bioreactor design specifications and electrical stimulation regime for cardiac tissue engineering.


Subject(s)
Heart/physiology , Tissue Engineering/methods , Animals , Bioreactors , Electric Stimulation , Electrodes , Models, Biological , Myocardium/ultrastructure , Rats , Rats, Sprague-Dawley
12.
Annu Rev Biomed Eng ; 13: 245-67, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21568715

ABSTRACT

The idea of extending the lifetime of our organs is as old as humankind, fueled by major advances in organ transplantation, novel drugs, and medical devices. However, true regeneration of human tissue has become increasingly plausible only in recent years. The human heart has always been a focus of such efforts, given its notorious inability to repair itself following injury or disease. We discuss here the emerging bioengineering approaches to regeneration of heart muscle as a paradigm for regenerative medicine. Our focus is on biologically inspired strategies for heart regeneration, knowledge gained thus far about how to make a "perfect" heart graft, and the challenges that remain to be addressed for tissue-engineered heart regeneration to become a clinical reality. We emphasize the need for interdisciplinary research and training, as recent progress in the field is largely being made at the interfaces between cardiology, stem cell science, and bioengineering.


Subject(s)
Heart/physiology , Myocardium/cytology , Regeneration/physiology , Regenerative Medicine , Tissue Scaffolds/chemistry , Animals , Bioengineering , Coronary Vessels/cytology , Hematopoietic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Models, Animal , Myocardium/immunology , Organ Culture Techniques/methods , Stem Cells/metabolism , Tissue Engineering/methods , Transplants/trends
13.
Biotechnol Prog ; 26(5): 1382-90, 2010.
Article in English | MEDLINE | ID: mdl-20945492

ABSTRACT

We investigated the effects of the initial stiffness of a three-dimensional elastomer scaffold--highly porous poly(glycerol sebacate)--on functional assembly of cardiomyocytes cultured with perfusion for 8 days. The polymer elasticity varied with the extent of polymer cross-links, resulting in three different stiffness groups, with compressive modulus of 2.35 ± 0.03 (low), 5.28 ± 0.36 (medium), and 5.99 ± 0.40 (high) kPa. Laminin coating improved the efficiency of cell seeding (from 59 ± 15 to 90 ± 21%), resulting in markedly increased final cell density, construct contractility, and matrix deposition, likely because of enhanced cell interaction and spreading on scaffold surfaces. Compact tissue was formed in the low and medium stiffness groups, but not in the high stiffness group. In particular, the low stiffness group exhibited the greatest contraction amplitude in response to electric field pacing, and had the highest compressive modulus at the end of culture. A mathematical model was developed to establish a correlation between the contractile amplitude and the cell distribution within the scaffold. Taken together, our findings suggest that the contractile function of engineered cardiac constructs positively correlates with low compressive stiffness of the scaffold.


Subject(s)
Tissue Engineering/methods , Tissue Scaffolds , Animals , Models, Theoretical , Myocytes, Cardiac/cytology , Rats , Rats, Sprague-Dawley
14.
Lab Chip ; 10(6): 692-700, 2010 Mar 21.
Article in English | MEDLINE | ID: mdl-20221556

ABSTRACT

We present a microscale cell culture system with an interdigitated microarray of excimer-laser-ablated indium tin oxide electrodes for electrical stimulation of cultured cells. The system has been characterized in a range of geometeries and stimulation regimes via electrochemical impedance spectroscopy and used to culture primary cardiomyocytes and human adipose derived stem cells. Over 6 days of culture with electrical stimulation (2 ms duration, 1 Hz, 180 microm wide electrodes with 200 microm spacing), both cell types exhibited enhanced proliferation, elongation and alignment, and adipose derived stem cells exhibited higher numbers of Connexin-43-composed gap junctions.


Subject(s)
Bioreactors , Cell Culture Techniques/instrumentation , Electric Stimulation/instrumentation , Electrodes , Microfluidics/instrumentation , Myocytes, Cardiac/physiology , Animals , Cells, Cultured , Equipment Design , Equipment Failure Analysis , Rats , Surface Properties
15.
Tissue Eng Part B Rev ; 16(2): 169-87, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19698068

ABSTRACT

Cardiac tissue engineering aims to create functional tissue constructs that can reestablish the structure and function of injured myocardium. Engineered constructs can also serve as high-fidelity models for studies of cardiac development and disease. In a general case, the biological potential of the cell-the actual "tissue engineer"-is mobilized by providing highly controllable three-dimensional environments that can mediate cell differentiation and functional assembly. For cardiac regeneration, some of the key requirements that need to be met are the selection of a human cell source, establishment of cardiac tissue matrix, electromechanical cell coupling, robust and stable contractile function, and functional vascularization. We review here the potential and challenges of cardiac tissue engineering for developing therapies that could prevent or reverse heart failure.


Subject(s)
Heart/physiology , Tissue Engineering/methods , Animals , Electric Stimulation/methods , Heart Transplantation/methods , Heart Transplantation/physiology , Humans , Models, Biological , Myocardium/cytology , Organ Culture Techniques , Perfusion/methods , Tissue Engineering/standards
16.
Article in English | MEDLINE | ID: mdl-19964171

ABSTRACT

In vivo, direct current electric fields are present during embryonic development and wound healing. In vitro, direct current (DC) electric fields induce directional cell migration and elongation. For the first time, we demonstrate that cultured human adipose tissue-derived stem cells (hASCs) respond to the presence of direct-current electric fields. Cells were stimulated for 2-4 hours with DC electric fields of 6 V/cm that were similar to those encountered in vivo post-injury. Upon stimulation, hASCs were observed to elongate and align perpendicularly to the applied electric field, disassemble gap junctions, and upregulate the expression of genes for connexin-43, thrombomodulin, vascular endothelial growth factor, and fibroblast growth factor. In separate related studies, human epicardial fat-derived stem cells (heASCs) were also observed to align and elongate. It is interesting that the morphological and phenotypic characteristics of mesenchymal stem cells derived both from liposuction aspirates and from cardiac fat can be modulated by direct current electric fields. In further studies, we will quantify the effects of the electrical fields in the context of wound healing.


Subject(s)
Adipocytes/cytology , Adipocytes/physiology , Stem Cells/cytology , Stem Cells/physiology , Adipocytes/radiation effects , Cell Differentiation/radiation effects , Cell Polarity/radiation effects , Cell Size/radiation effects , Cells, Cultured , Dose-Response Relationship, Radiation , Humans , Radiation Dosage , Stem Cells/radiation effects
17.
Exp Cell Res ; 315(20): 3611-9, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19720058

ABSTRACT

Exogenous electric fields have been implied in cardiac differentiation of mouse embryonic stem cells and the generation of reactive oxygen species (ROS). In this work, we explored the effects of electrical field stimulation on ROS generation and cardiogenesis in embryoid bodies (EBs) derived from human embryonic stem cells (hESC, line H13), using a custom-built electrical stimulation bioreactor. Electrical properties of the bioreactor system were characterized by electrochemical impedance spectroscopy (EIS) and analysis of electrical currents. The effects of the electrode material (stainless steel, titanium-nitride-coated titanium, titanium), length of stimulus (1 and 90 s) and age of EBs at the onset of electrical stimulation (4 and 8 days) were investigated with respect to ROS generation. The amplitude of the applied electrical field was 1 V/mm. The highest rate of ROS generation was observed for stainless steel electrodes, for signal duration of 90 s and for 4-day-old EBs. Notably, comparable ROS generation was achieved by incubation of EBs with 1 nM H(2)O(2). Cardiac differentiation in these EBs was evidenced by spontaneous contractions, expression of troponin T and its sarcomeric organization. These results imply that electrical stimulation plays a role in cardiac differentiation of hESCs, through mechanisms associated with the intracellular generation of ROS.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Bioreactors , Cell Culture Techniques , Cell Survival , Electric Stimulation , Electricity , Electrodes , Embryonic Stem Cells/metabolism , Fluoresceins/metabolism , Humans , Hydrogen Peroxide/pharmacology , Microscopy, Fluorescence , Myocardial Contraction , Sarcomeres/metabolism , Stainless Steel/chemistry , Titanium/chemistry , Troponin T/metabolism
18.
Nat Protoc ; 4(2): 155-73, 2009.
Article in English | MEDLINE | ID: mdl-19180087

ABSTRACT

We describe a protocol for tissue engineering of synchronously contractile cardiac constructs by culturing cardiac cells with the application of pulsatile electrical fields designed to mimic those present in the native heart. Tissue culture is conducted in a customized chamber built to allow for cultivation of (i) engineered three-dimensional (3D) cardiac tissue constructs, (ii) cell monolayers on flat substrates or (iii) cells on patterned substrates. This also allows for analysis of the individual and interactive effects of pulsatile electrical field stimulation and substrate topography on cell differentiation and assembly. The protocol is designed to allow for delivery of predictable electrical field stimuli to cells, monitoring environmental parameters, and assessment of cell and tissue responses. The duration of the protocol is 5 d for two-dimensional cultures and 10 d for 3D cultures.


Subject(s)
Myocardium/cytology , Tissue Engineering/instrumentation , Tissue Engineering/methods , Animals , Cattle , Electrophysiology , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Myocytes, Cardiac/physiology , Rats
19.
Methods Mol Med ; 140: 291-307, 2007.
Article in English | MEDLINE | ID: mdl-18085215

ABSTRACT

Heart disease is a leading cause of death in western society. Despite the success of heart transplantation, a chronic shortage of donor organs, along with the associated immunological complications of this approach, demands that alternative treatments be found. One such option is to repair, rather than replace, the heart with engineered cardiac tissue. Multiple studies have shown that to attain functional tissue, assembly signaling cues must be recapitulated in vitro. In their native environment, cardiomyocytes are directed to beat in synchrony by propagation of pacing current through the tissue. Recently, we have shown that electrical stimulation directs neonatal cardiomyocytes to assemble into native-like tissue in vitro. This chapter provides detailed methods we have employed in taking this "biomimetic" approach. After an initial discussion on how electric field stimulation can influence cell behavior, we examine the practical aspects of cardiac tissue engineering with electrical stimulation, such as electrode selection and cell seeding protocols, and conclude with what we feel are the remaining challenges to be overcome.


Subject(s)
Electric Stimulation , Heart , Tissue Engineering/methods , Animals , Animals, Newborn , Bioartificial Organs , Bioreactors , Cell Culture Techniques , Cells, Cultured , Heart/anatomy & histology , Heart/physiology , Humans , Myocardial Contraction/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley
20.
Article in English | MEDLINE | ID: mdl-17946862

ABSTRACT

Electrical stimulation has been shown to improve functional assembly of cardiomyocytes in vitro for cardiac tissue engineering. The goal of this study was to assess the conditions of electrical stimulation with respect to the electrode geometry, material properties and charge-transfer characteristics at the electrode-electrolyte interface. We compared various biocompatible materials, including nanoporous carbon, stainless steel, titanium and titanium nitride, for use in cardiac tissue engineering bioreactors. The faradaic and non-faradaic charge transfer mechanisms were assessed by electrochemical impedance spectroscopy (EIS), studying current injection characteristics, and examining surface properties of electrodes with scanning electron microscopy. Carbon electrodes were found to have the best current injection characteristics. However, these electrodes require careful handling because of their limited mechanical strength. The efficacy of various electrodes for use in 2-D and 3-D cardiac tissue engineering systems with neonatal rat cardiomyocytes is being determined by assessing cell viability, amplitude of contractions, excitation thresholds, maximum capture rate, and tissue morphology.


Subject(s)
Action Potentials/physiology , Cell Culture Techniques/instrumentation , Electric Stimulation/instrumentation , Heart Conduction System/physiology , Microelectrodes , Myocytes, Cardiac/physiology , Tissue Engineering/instrumentation , Animals , Animals, Newborn , Equipment Design , Equipment Failure Analysis , Rats , Tissue Engineering/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...