Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
EMBO J ; 34(8): 987-1008, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25766254

ABSTRACT

Derived from any somatic cell type and possessing unlimited self-renewal and differentiation potential, induced pluripotent stem cells (iPSCs) are poised to revolutionize stem cell biology and regenerative medicine research, bringing unprecedented opportunities for treating debilitating human diseases. To overcome the limitations associated with safety, efficiency, and scalability of traditional iPSC derivation, expansion, and differentiation protocols, biomaterials have recently been considered. Beyond addressing these limitations, the integration of biomaterials with existing iPSC culture platforms could offer additional opportunities to better probe the biology and control the behavior of iPSCs or their progeny in vitro and in vivo. Herein, we discuss the impact of biomaterials on the iPSC field, from derivation to tissue regeneration and modeling. Although still exploratory, we envision the emerging combination of biomaterials and iPSCs will be critical in the successful application of iPSCs and their progeny for research and clinical translation.


Subject(s)
Biocompatible Materials/therapeutic use , Induced Pluripotent Stem Cells/cytology , Regeneration , Cellular Reprogramming , Gene Expression Regulation , Genetic Therapy/methods , Humans , Induced Pluripotent Stem Cells/transplantation , Stem Cell Research , Stem Cell Transplantation/methods , Tissue Engineering/methods
2.
Int J Biol Macromol ; 277(Pt 3): 134453, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39098691

ABSTRACT

Chitosan possesses electron-rich amino (-NH2) and hydroxyl (-OH) moieties which can anchor with transition metal ions during synthesis. Herein, chitosan was employed as an additive to prepare bismuth ferrite (BFO) via hydrothermal approach. The characterization studies revealed that adding chitosan during BFO synthesis leads to the creation of more oxygen vacancies. The performance of chitosan modified BFO (CMB) was evaluated as peroxymonosulfate (PMS) activator for ciprofloxacin (CIP) removal. Apparently, the addition of 10 wt% chitosan during BFO synthesis (CMB-10) resulted in 1.7 times increase of performance compared to the pristine BFO. Increasing the catalyst loading and PMS dosage resulted in positive effect with 5.7 and 1.9 times rate enhancement, respectively. The CMB-10 exhibited tolerance against pH variation, water matrix, and interfering species. The scavenging experiments indicated that singlet oxygen (1O2), superoxide radicals (O2•-) and sulfate radicals (SO4•-) played a major role in CIP degradation. These reactive oxygen species were generated from PMS activation via Fe3+/Fe2+ and Bi5+/Bi3+ coupling, and oxygen vacancies on the catalyst surface. The CIP degradation pathways were also elucidated based on the detected CIP intermediates. Overall, this study provides insights into the use of chitosan to prepare sustainable materials for pollutants removal via PMS activation.


Subject(s)
Anti-Bacterial Agents , Bismuth , Chitosan , Ferric Compounds , Peroxides , Chitosan/chemistry , Bismuth/chemistry , Catalysis , Ferric Compounds/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Peroxides/chemistry , Water Pollutants, Chemical/chemistry , Ciprofloxacin/chemistry , Ciprofloxacin/pharmacology , Water Purification/methods , Hydrogen-Ion Concentration
3.
Biomacromolecules ; 14(11): 3808-19, 2013 Nov 11.
Article in English | MEDLINE | ID: mdl-24093583

ABSTRACT

Synthetic hydrogels containing covalently integrated soft and deformable drug depots capable of releasing therapeutic molecules in response to mechanical forces are attractive candidates for the treatment of degenerated tissues that are normally load bearing. Herein, radically cross-linkable block copolymer micelles (xBCM) assembled from an amphiphilic block copolymer consisting of hydrophilic poly(acrylic acid) (PAA) partially modified with 2-hydroxyethyl acrylate, and hydrophobic poly(n-butyl acryclate) (PnBA) were employed as the drug depots and the microscopic cross-linkers for the preparation of hyaluronic acid (HA)-based, hydrogels. HA hydrogels containing covalently integrated micelles (HAxBCM) were prepared by radical polymerization of glycidyl methacrylate (GMA)-modified HA (HAGMA) in the presence of xBCMs. When micelles prepared from the parent PAA-b-PnBA without any polymerizable double bonds were used, hydrogels containing physically entrapped micelles (HApBCM) were obtained. The addition of xBCMs to a HAGMA precursor solution accelerated the gelation kinetics and altered the hydrogel mechanical properties. The resultant HAxBCM gels exhibit an elastic modulus of 847 ± 43 Pa and a compressive modulus of 9.2 ± 0.7 kPa. Diffusion analysis of Nile Red (NR)-labeled xBCMs employing fluorescence correlation spectroscopy confirmed the covalent immobilization of xBCMs in HA networks. Covalent integration of dexamethasone (DEX)-loaded xBCMs in HA gels significantly reduced the initial burst release and provided sustained release over a prolonged period. Importantly, DEX release from HAxBCM gels was accelerated by intermittently applied external compression in a strain-dependent manner. Culturing macrophages in the presence of DEX-releasing HAxBCM gels significantly reduced cellular production of inflammatory cytokines. Incorporating mechano-responsive modules in synthetic matrices offers a novel strategy to harvest mechanical stress present in the healing wounds to initiate tissue repair.


Subject(s)
Dexamethasone/chemistry , Hyaluronic Acid/chemistry , Hydrogels/chemistry , Inflammation/drug therapy , Animals , Cell Line , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Dexamethasone/pharmacology , Hydrogels/chemical synthesis , Macrophages/drug effects , Macrophages/metabolism , Mice , Micelles , Models, Molecular , Molecular Structure , Particle Size , Polymers/chemistry , Surface Properties
4.
Soft Matter ; 9(3): 665-673, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23505396

ABSTRACT

Natural resilin, the rubber-like protein that exists in specialized compartments of most arthropods, possesses excellent mechanical properties such as low stiffness, high resilience and effective energy storage. Recombinantly-engineered resilin-like polypeptides (RLPs) that possess the favorable attributes of native resilin would be attractive candidates for the modular design of biomaterials for engineering mechanically active tissues. Based on our previous success in creating a novel RLP-based hydrogel and demonstrating useful mechanical and cell-adhesive properties, we have produced a suite of new RLP-based constructs, each equipped with 12 repeats of the putative resilin consensus sequence and a single, distinct biologically active domain. This approach allows independent control over the concentrations of cell-binding, MMP-sensitive, and polysaccharide-sequestration domains in hydrogels comprising mixtures of the various RLPs. The high purity, molecular weight and correct compositions of each new polypeptide have been confirmed via high performance liquid chromatography (HPLC), sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE), matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS), and amino acid analysis. These RLP-based polypeptides exhibit largely random-coil conformation, both in solution and in the cross-linked hydrogels, as indicated by circular dichroic and infrared spectroscopic analyses. Hydrogels of various compositions, with a range of elastic moduli (1kPa to 25kPa) can be produced from these polypeptides, and the activity of the cell-binding and matrix metalloproteinase (MMP) sensitive domains was confirmed. Tris(hydroxymethyl phosphine) cross-linked RLP hydrogels were able to maintain their mechanical integrity as well as the viability of encapsulated primary human mesenchymal stem cells (MSCs). These results validate the promising properties of these RLP-based elastomeric biomaterials.

5.
J Mol Med (Berl) ; 99(5): 663-671, 2021 05.
Article in English | MEDLINE | ID: mdl-33398468

ABSTRACT

Mesenchymal stem cells (MSCs) are promising candidates for the development of cell-based drug delivery systems for autoimmune inflammatory diseases, such as multiple sclerosis (MS). Here, we investigated the effect of Ro-31-8425, an ATP-competitive kinase inhibitor, on the therapeutic properties of MSCs. Upon a simple pretreatment procedure, MSCs spontaneously took up and then gradually released significant amounts of Ro-31-8425. Ro-31-8425 (free or released by MSCs) suppressed the proliferation of CD4+ T cells in vitro following polyclonal and antigen-specific stimulation. Systemic administration of Ro-31-8425-loaded MSCs ameliorated the clinical course of experimental autoimmune encephalomyelitis (EAE), a murine model of MS, displaying a stronger suppressive effect on EAE than control MSCs or free Ro-31-8425. Ro-31-8425-MSC administration resulted in sustained levels of Ro-31-8425 in the serum of EAE mice, modulating immune cell trafficking and the autoimmune response during EAE. Collectively, these results identify MSC-based drug delivery as a potential therapeutic strategy for the treatment of autoimmune diseases. KEY MESSAGES: MSCs can spontaneously take up the ATP-competitive kinase inhibitor Ro-31-8425. Ro-31-8425-loaded MSCs gradually release Ro-31-8425 and exhibit sustained suppression of T cells. Ro-31-8425-loaded MSCs have more sustained serum levels of Ro-31-8425 than free Ro-31-8425. Ro-31-8425-loaded MSCs are more effective than MSCs and free Ro-31-8425 for EAE therapy.


Subject(s)
Drug Delivery Systems/methods , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Enzyme Inhibitors/administration & dosage , Indoles/administration & dosage , Maleimides/administration & dosage , Mesenchymal Stem Cells/drug effects , Multiple Sclerosis/drug therapy , Transplantation, Heterologous/methods , Animals , Cell Proliferation/drug effects , Drug Liberation , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/immunology , Enzyme Inhibitors/blood , Female , Humans , Immunity/drug effects , Indoles/blood , Maleimides/blood , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Multiple Sclerosis/blood , Multiple Sclerosis/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tissue Distribution , Treatment Outcome
6.
Chem Commun (Camb) ; 55(62): 9128-9131, 2019 Jul 30.
Article in English | MEDLINE | ID: mdl-31298674

ABSTRACT

We report a simple route for high-density flower-like NiCo2O4 nanosheets with ultrahigh mass loading of 9.2 mg cm-2 on nickel foam to achieve superior capacitance. The as-assembled asymmetric supercapacitor yields a high energy density of 87.4 W h kg-1 and an excellent cycle life with 93.2% capacitance retention after 11 000 cycles.

7.
Nat Biomed Eng ; 3(8): 621-631, 2019 08.
Article in English | MEDLINE | ID: mdl-31391590

ABSTRACT

The precision of the delivery of therapeutics to the desired injection site by syringes and hollow needles typically depends on the operator. Here, we introduce a highly sensitive, completely mechanical and cost-effective injector for targeting tissue reliably and precisely. As the operator pushes the syringe plunger, the injector senses the loss-of-resistance on encountering a softer tissue or a cavity, stops advancing the needle and delivers the payload. We demonstrate that the injector can reliably deliver liquids to the suprachoroidal space-a challenging injection site that provides access to the back of the eye-for a wide range of eye sizes, scleral thicknesses and intraocular pressures, and target sites relevant for epidural injections, subcutaneous injections and intraperitoneal access. The design of this simple and effective injector can be adapted for a broad variety of clinical applications.


Subject(s)
Drug Delivery Systems/instrumentation , Drug Delivery Systems/methods , Injections/instrumentation , Injections/methods , Animals , Drug Delivery Systems/adverse effects , Equipment Design/instrumentation , Equipment Design/methods , Eye/pathology , Humans , Infusion Pumps/adverse effects , Injections/adverse effects , Injections, Epidural/instrumentation , Injections, Epidural/methods , Injections, Intraperitoneal/instrumentation , Injections, Intraperitoneal/methods , Injections, Subcutaneous/instrumentation , Injections, Subcutaneous/methods , Needles , Rabbits , Syringes , Wounds and Injuries
8.
Biomaterials ; 154: 60-73, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29120819

ABSTRACT

Current ISC culture systems face significant challenges such as animal-derived or undefined matrix compositions, batch-to-batch variability (e.g. Matrigel-based organoid culture), and complexity of assaying cell aggregates such as organoids which renders the research and clinical translation of ISCs challenging. Here, through screening for suitable ECM components, we report a defined, collagen based monolayer culture system that supports the growth of mouse and human intestinal epithelial cells (IECs) enriched for an Lgr5+ population comparable or higher to the levels found in a standard Matrigel-based organoid culture. The system, referred to as the Bolstering Lgr5 Transformational (BLT) Sandwich culture, comprises a collagen IV-coated porous substrate and a collagen I gel overlay which sandwich an IEC monolayer in between. The distinct collagen cues synergistically regulate IEC attachment, proliferation, and Lgr5 expression through maximizing the engagement of distinct cell surface adhesion receptors (i.e. integrin α2ß1, integrin ß4) and cell polarity. Further, we apply our BLT Sandwich system to identify that the addition of a bone morphogenetic protein (BMP) receptor inhibitor (LDN-193189) improves the expansion of Lgr5-GFP+ cells from mouse small intestinal crypts by nearly 2.5-fold. Notably, the BLT Sandwich culture is capable of expanding human-derived IECs with higher LGR5 mRNA levels than conventional Matrigel culture, providing superior expansion of human LGR5+ ISCs. Considering the key roles Lgr5+ ISCs play in intestinal epithelial homeostasis and regeneration, we envision that our BLT Sandwich culture system holds great potential for understanding and manipulating ISC biology in vitro (e.g. for modeling ISC-mediated gut diseases) or for expanding a large number of ISCs for clinical utility (e.g. for stem cell therapy).


Subject(s)
Cell Culture Techniques/methods , Extracellular Matrix/metabolism , Intestines/cytology , Stem Cells/cytology , Animals , Cell Proliferation/drug effects , Coated Materials, Biocompatible/pharmacology , Collagen/pharmacology , Collagen Type IV/pharmacology , Drug Combinations , Epithelial Cells/cytology , Extracellular Matrix/drug effects , Green Fluorescent Proteins/metabolism , Humans , Laminin/pharmacology , Mice, Inbred C57BL , Proteoglycans/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptors, G-Protein-Coupled/metabolism , Stem Cells/drug effects
9.
EMBO Mol Med ; 9(5): 558-570, 2017 05.
Article in English | MEDLINE | ID: mdl-28283650

ABSTRACT

Both the incidence and prevalence of inflammatory bowel disease (IBD) is increasing globally; in the industrialized world up to 0.5% of the population are affected and around 4.2 million individuals suffer from IBD in Europe and North America combined. Successful engraftment in experimental colitis models suggests that intestinal stem cell transplantation could constitute a novel treatment strategy to re-establish mucosal barrier function in patients with severe disease. Intestinal stem cells can be grown in vitro in organoid structures, though only a fraction of the cells contained are stem cells with regenerative capabilities. Hence, techniques to enrich stem cell populations are being pursued through the development of multiple two-dimensional and three-dimensional culture protocols, as well as co-culture techniques and multiple growth medium compositions. Moreover, research in support matrices allowing for efficient clinical application is in progress. In vitro culture is accomplished by modulating the signaling pathways fundamental for the stem cell niche with a suitable culture matrix to provide additional contact-dependent stimuli and structural support. The aim of this review was to discuss medium compositions and support matrices for optimal intestinal stem cell culture, as well as potential modifications to advance clinical use in IBD.


Subject(s)
Cell Culture Techniques/methods , Culture Media/metabolism , Inflammatory Bowel Diseases/therapy , Intestines/cytology , Stem Cell Transplantation , Stem Cells/cytology , Tissue Scaffolds , Animals , Bone Morphogenetic Proteins/metabolism , Culture Media/chemistry , Dinoprostone/metabolism , Epidermal Growth Factor/metabolism , Humans , Inflammatory Bowel Diseases/metabolism , Intestines/physiology , Receptors, Notch/metabolism , Regeneration , Regenerative Medicine/methods , Signal Transduction , Stem Cell Transplantation/methods , Stem Cells/metabolism , Tissue Scaffolds/chemistry , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway
10.
Stem Cell Reports ; 6(6): 926-939, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27264972

ABSTRACT

Mesenchymal stromal cells (MSCs) are promising therapeutic candidates given their potent immunomodulatory and anti-inflammatory secretome. However, controlling the MSC secretome post-transplantation is considered a major challenge that hinders their clinical efficacy. To address this, we used a microparticle-based engineering approach to non-genetically modulate pro-inflammatory pathways in human MSCs (hMSCs) under simulated inflammatory conditions. Here we show that microparticles loaded with TPCA-1, a small-molecule NF-κB inhibitor, when delivered to hMSCs can attenuate secretion of pro-inflammatory factors for at least 6 days in vitro. Conditioned medium (CM) derived from TPCA-1-loaded hMSCs also showed reduced ability to attract human monocytes and prevented differentiation of human cardiac fibroblasts to myofibroblasts, compared with CM from untreated or TPCA-1-preconditioned hMSCs. Thus, we provide a broadly applicable bioengineering solution to facilitate intracellular sustained release of agents that modulate signaling. We propose that this approach could be harnessed to improve control over MSC secretome post-transplantation, especially to prevent adverse remodeling post-myocardial infarction.


Subject(s)
Amides/pharmacology , Bone Marrow Cells/drug effects , Delayed-Action Preparations , Drug Carriers , Mesenchymal Stem Cells/drug effects , NF-kappa B/antagonists & inhibitors , Thiophenes/pharmacology , Amides/chemistry , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation , Chemical Engineering/methods , Coculture Techniques , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/pharmacology , Drug Compounding , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression , Humans , Inflammation , Lactic Acid/chemistry , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Models, Biological , Monocytes/cytology , Monocytes/drug effects , NF-kappa B/genetics , NF-kappa B/metabolism , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Primary Cell Culture , Thiophenes/chemistry , Tumor Necrosis Factor-alpha/pharmacology
11.
Adv Healthc Mater ; 5(2): 266-75, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26632334

ABSTRACT

The outstanding elasticity, excellent resilience at high-frequency, and hydrophilic capacity of natural resilin have motivated investigations of recombinant resilin-based biomaterials as a new class of bio-elastomers in the engineering of mechanically active tissues. Accordingly, here the comprehensive characterization of modular resilin-like polypeptide (RLP) hydrogels is presented and their suitability as a novel biomaterial for in vivo applications is introduced. Oscillatory rheology confirmed that a full suite of the RLPs can be rapidly cross-linked upon addition of the tris(hydroxymethyl phosphine) cross-linker, achieving similar in situ shear storage moduli (20 k ± 3.5 Pa) across various material compositions. Uniaxial stress relaxation tensile testing of hydrated RLP hydrogels under cyclic loading and unloading showed negligible stress reduction and hysteresis, superior reversible extensibility, and high resilience with Young's moduli of 30 ± 7.4 kPa. RLP hydrogels containing MMP-sensitive domains are susceptible to enzymatic degradation by matrix metalloproteinase-1 (MMP-1). Cell culture studies revealed that RLP-based hydrogels supported the attachment and spreading (2D) of human mesenchymal stem cells and did not activate cultured macrophages. Subcutaneous transplantation of RLP hydrogels in a rat model, which to our knowledge is the first such reported in vivo analysis of RLP-based hydrogels, illustrated that these materials do not elicit a significant inflammatory response, suggesting their potential as materials for tissue engineering applications with targets of mechanically demanding tissues such as vocal fold and cardiovascular tissues.


Subject(s)
Elastomers/pharmacology , Insect Proteins/pharmacology , Recombinant Proteins/pharmacology , Regenerative Medicine/methods , Animals , Drosophila , Humans , Hydrogels/pharmacology , Immunohistochemistry , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , RAW 264.7 Cells , Rats, Sprague-Dawley , Subcutaneous Tissue/drug effects , Tensile Strength/drug effects
12.
Sci Rep ; 6: 30263, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27457881

ABSTRACT

Pre-treatment or priming of mesenchymal stem cells (MSC) prior to transplantation can significantly augment the immunosuppressive effect of MSC-based therapies. In this study, we screened a library of 1402 FDA-approved bioactive compounds to prime MSC. We identified tetrandrine as a potential hit that activates the secretion of prostaglandin E2 (PGE2), a potent immunosuppressive agent, by MSC. Tetrandrine increased MSC PGE2 secretion through the NF-κB/COX-2 signaling pathway. When co-cultured with mouse macrophages (RAW264.7), tetrandrine-primed MSC attenuated the level of TNF-α secreted by RAW264.7. Furthermore, systemic transplantation of primed MSC into a mouse ear skin inflammation model significantly reduced the level of TNF-α in the inflamed ear, compared to unprimed cells. Screening of small molecules to pre-condition cells prior to transplantation represents a promising strategy to boost the therapeutic potential of cell therapy.


Subject(s)
Benzylisoquinolines/pharmacology , Immunosuppressive Agents/pharmacology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/drug effects , Animals , Cyclooxygenase 2/immunology , Humans , Immunomodulation/drug effects , Mass Screening , Mesenchymal Stem Cells/immunology , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/immunology , RAW 264.7 Cells , Signal Transduction/drug effects , Signal Transduction/immunology , Small Molecule Libraries
13.
Cell Rep ; 10(8): 1261-1268, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25732817

ABSTRACT

Poor homing of systemically infused cells to disease sites may limit the success of exogenous cell-based therapy. In this study, we screened 9,000 signal-transduction modulators to identify hits that increase mesenchymal stromal cell (MSC) surface expression of homing ligands that bind to intercellular adhesion molecule 1 (ICAM-1), such as CD11a. Pretreatment of MSCs with Ro-31-8425, an identified hit from this screen, increased MSC firm adhesion to an ICAM-1-coated substrate in vitro and enabled targeted delivery of systemically administered MSCs to inflamed sites in vivo in a CD11a- (and other ICAM-1-binding domains)-dependent manner. This resulted in a heightened anti-inflammatory response. This represents a new strategy for engineering cell homing to enhance therapeutic efficacy and validates CD11a and ICAM-1 as potential targets. Altogether, this multi-step screening process may significantly improve clinical outcomes of cell-based therapies.


Subject(s)
Mesenchymal Stem Cells/cytology , Small Molecule Libraries/chemistry , Animals , CD11a Antigen/genetics , CD11a Antigen/metabolism , Cell Adhesion/drug effects , Cell Line , Cell Movement , High-Throughput Screening Assays , Humans , Indoles/chemistry , Indoles/pharmacology , Inflammation/chemically induced , Inflammation/pathology , Inflammation/therapy , Intercellular Adhesion Molecule-1/chemistry , Intercellular Adhesion Molecule-1/metabolism , Lipopolysaccharides/toxicity , Maleimides/chemistry , Maleimides/pharmacology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Up-Regulation
14.
Tissue Eng Part A ; 20(13-14): 1922-34, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24456068

ABSTRACT

Vocal fold disorders affect 3-9% of the U.S. population. Tissue engineering offers an alternative strategy for vocal fold repair. Successful engineering of vocal fold tissues requires a strategic combination of therapeutic cells, biomimetic scaffolds, and physiologically relevant mechanical and biochemical factors. Specifically, we aim to create a vocal fold-like microenvironment to coax stem cells to adopt the phenotype of vocal fold fibroblasts (VFFs). Herein, high frequency vibratory stimulations and soluble connective tissue growth factor (CTGF) were sequentially introduced to mesenchymal stem cells (MSCs) cultured on a poly(ɛ-caprolactone) (PCL)-derived microfibrous scaffold for a total of 6 days. The initial 3-day vibratory culture resulted in an increased production of hyaluronic acids (HA), tenascin-C (TNC), decorin (DCN), and matrix metalloproteinase-1 (MMP1). The subsequent 3-day CTGF treatment further enhanced the cellular production of TNC and DCN, whereas CTGF treatment alone without the vibratory preconditioning significantly promoted the synthesis of collagen I (Col 1) and sulfated glycosaminoglycans (sGAGs). The highest level of MMP1, TNC, Col III, and DCN production was found for cells being exposed to the combined vibration and CTGF treatment. Noteworthy, the vibration and CTGF elicited a differential stimulatory effect on elastin (ELN), HA synthase 1 (HAS1), and fibroblast-specific protein-1 (FSP-1). The mitogenic activity of CTGF was only elicited in naïve cells without the vibratory preconditioning. The combined treatment had profound, but opposite effects on mitogen-activated protein kinase (MAPK) pathways, Erk1/2 and p38, and the Erk1/2 pathway was critical for the observed mechano-biochemical responses. Collectively, vibratory stresses and CTGF signals cooperatively coaxed MSCs toward a VFF-like phenotype and accelerated the synthesis and remodeling of vocal fold matrices.


Subject(s)
Connective Tissue Growth Factor/pharmacology , Mesenchymal Stem Cells/cytology , Vibration , Biomechanical Phenomena/drug effects , Bioreactors , Blotting, Western , Butadienes/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Extracellular Matrix Proteins/drug effects , Extracellular Matrix Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/enzymology , Nitriles/pharmacology , Time Factors
15.
J Vis Exp ; (90): e51594, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-25145349

ABSTRACT

In vitro engineering of mechanically active tissues requires the presentation of physiologically relevant mechanical conditions to cultured cells. To emulate the dynamic environment of vocal folds, a novel vocal fold bioreactor capable of producing vibratory stimulations at fundamental phonation frequencies is constructed and characterized. The device is composed of a function generator, a power amplifier, a speaker selector and parallel vibration chambers. Individual vibration chambers are created by sandwiching a custom-made silicone membrane between a pair of acrylic blocks. The silicone membrane not only serves as the bottom of the chamber but also provides a mechanism for securing the cell-laden scaffold. Vibration signals, generated by a speaker mounted underneath the bottom acrylic block, are transmitted to the membrane aerodynamically by the oscillating air. Eight identical vibration modules, fixed on two stationary metal bars, are housed in an anti-humidity chamber for long-term operation in a cell culture incubator. The vibration characteristics of the vocal fold bioreactor are analyzed non-destructively using a Laser Doppler Vibrometer (LDV). The utility of the dynamic culture device is demonstrated by culturing cellular constructs in the presence of 200-Hz sinusoidal vibrations with a mid-membrane displacement of 40 µm. Mesenchymal stem cells cultured in the bioreactor respond to the vibratory signals by altering the synthesis and degradation of vocal fold-relevant, extracellular matrix components. The novel bioreactor system presented herein offers an excellent in vitro platform for studying vibration-induced mechanotransduction and for the engineering of functional vocal fold tissues.


Subject(s)
Bioreactors , Tissue Engineering/methods , Vocal Cords , Biomimetic Materials/chemistry , Cell Culture Techniques/methods , Equipment Design , Humans , Mesenchymal Stem Cells/cytology , Polyesters/chemistry , Silicones/chemistry , Vibration
16.
Tissue Eng Part A ; 19(15-16): 1862-78, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23516973

ABSTRACT

We are interested in the in vitro engineering of artificial vocal fold tissues via the strategic combination of multipotent mesenchymal stem cells (MSCs), physiologically relevant mechanical stimulations, and biomimetic artificial matrices. We have constructed a vocal fold bioreactor that is capable of imposing vibratory stimulations on the cultured cells at human phonation frequencies. Separately, fibrous poly (ɛ-caprolactone) (PCL) scaffolds emulating the ligamentous structure of the vocal fold were prepared by electrospinning, were incorporated in the vocal fold bioreactor, and were driven into a wave-like motion in an axisymmetrical fashion by the oscillating air. MSC-laden PCL scaffolds were subjected to vibrations at 200 Hz with a normal center displacement of ∼40 µm for a total of 7 days. A continuous (CT) or a 1 h-on-1 h-off (OF) regime with a total dynamic culture time of 12 h per day was applied. The dynamic loading did not cause any physiological trauma to the cells. Immunohistotochemical staining revealed the reinforcement of the actin filament and the enhancement of α5ß1 integrin expression under selected dynamic culture conditions. Cellular expression of essential vocal fold extracellular matrix components, such as elastin, hyaluronic acid, and matrix metalloproteinase-1, was significantly elevated as compared with the static controls, and the OF regime is more conducive to matrix production than the CT vibration mode. Analyses of genes of typical fibroblast hallmarks (tenascin-C, collagen III, and procollagen I) as well as markers for MSC differentiation into nonfibroblastic lineages confirmed MSCs' adaptation of fibroblastic behaviors. Overall, the high-frequency vibratory stimulation, when combined with a synthetic fibrous scaffold, serves as a potent modulator of MSC functions. The novel bioreactor system presented here, as a versatile, yet well-controlled model, offers an in vitro platform for understanding vibration-induced mechanotransduction and for engineering of functional vocal fold tissues.


Subject(s)
Mesenchymal Stem Cells/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Bioreactors , Cell Proliferation , Cell Survival/physiology , Cells, Cultured , Humans , Immunohistochemistry , Integrin alpha5beta1/metabolism
17.
Acta Biomater ; 9(9): 8232-44, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23770222

ABSTRACT

Block copolymers of poly(ethylene glycol) and poly(ε-caprolactone) (PCL) with chemically addressable functional groups were synthesized and characterized. Ring-opening polymerization of ε-caprolactone (CL) and 1,4,8-trioxaspiro-[4,6]-9-undecanone (TSU) using α-methoxy, ω-hydroxyl poly(ethylene glycol) as the initiator afforded a copolymer with cyclic ketals being randomly distributed in the hydrophobic PCL block. At an initiator/catalyst molar ratio of 10/1 and a TSU/CL weight ratio of 1/4, a ketal-carrying copolymer (ECT2-CK) with Mn of 52 kDa and a ketal content of 15 mol.% was obtained. Quantitative side-chain deacetalization revealed the reactive ketones without noticeable polymer degradation. In our study, 10 mol.% of cyclic ketals were deprotected and the ketone-containing copolymer was designated as ECT2-CO. Reaction of ECT2-CO with 2-(2-(aminooxy)acetoxy)-ethyl acrylate gave rise to an acrylated product (ECT2-AC) containing an estimated 3-5 acrylate groups per chain. UV-initiated radical polymerization of ECT2-AC in dichloromethane resulted in a crosslinked network (xECT2-AC). Thermal and morphological analyses employing differential scanning calorimetry and atomic force microscopy operated in PeakForce Tapping mode revealed the semicrystalline nature of the network, which contained stiff crystalline lamellae dispersed in a softer amorphous interstitial. Macroscopic and nanoscale mechanical characterizations showed that ECT2-CK exhibited a significantly lower modulus than PCL of a similar molecular weight. Whereas ECT2-CK undergoes a plastic deformation with a distinct yield point and a cold-drawing region, xECT2-AC exhibits a compliant, elastomeric deformation with a Young's modulus of 0.5±0.1 MPa at 37°C. When properly processed, the crosslinked network exhibited shape-memory behaviors, with shape fixity and shape recovery values close to 1 and a shape recovery time of less than 4s at 37°C. In vitro studies showed that xECT2-AC films did not induce any cytotoxic effects on the cultured mesenchymal stem cells. The crosslinkable polyester copolymers can be potentially used as tissue engineering scaffolds and minimally invasive medical devices.


Subject(s)
Acrylates/chemistry , Elastomers/chemical synthesis , Elastomers/toxicity , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Polyesters/chemical synthesis , Polyesters/toxicity , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/toxicity , Acrylates/radiation effects , Biocompatible Materials/chemical synthesis , Biocompatible Materials/radiation effects , Biocompatible Materials/toxicity , Cell Survival/drug effects , Cells, Cultured , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/radiation effects , Crystallization/methods , Elastic Modulus/radiation effects , Hardness/radiation effects , Humans , Light , Materials Testing , Polyesters/radiation effects , Polyethylene Glycols/radiation effects , Tensile Strength/radiation effects
18.
MRS Commun ; 2(2): 31-39, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-25250199

ABSTRACT

Load-bearing, mechanically active tissues are routinely subjected to non-linear mechanical deformations. Consequently, these tissues exhibit complex mechanical properties and unique tissue organizations. Successful engineering of mechanically active tissues relies on the integration of the mechanical sensing mechanism found in the native tissues into polymeric scaffolds. Intelligent biomaterials that closely mimic the structural organizations and multi-scale responsiveness of the natural extracellular matrices (ECM), when strategically combined with multipotent cells and dynamic culture devices that generate physiologically relevant physical forces, will lead to the creation of artificial tissues that are mechanically robust and biologically functional.

19.
Tissue Eng Part A ; 17(21-22): 2773-85, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21689062

ABSTRACT

Controlled differentiation of multi-potent mesenchymal stem cells (MSCs) into vocal fold-specific, fibroblast-like cells in vitro is an attractive strategy for vocal fold repair and regeneration. The goal of the current study was to define experimental parameters that can be used to control the initial fibroblastic differentiation of MSCs in vitro. To this end, connective tissue growth factor (CTGF) and micro-structured, fibrous scaffolds based on poly(glycerol sebacate) (PGS) and poly(ɛ-caprolactone) (PCL) were used to create a three-dimensional, connective tissue-like microenvironment. MSCs readily attached to and elongated along the microfibers, adopting a spindle-shaped morphology during the initial 3 days of preculture in an MSC maintenance medium. The cell-laden scaffolds were subsequently cultivated in a conditioned medium containing CTGF and ascorbic acids for up to 21 days. Cell morphology, proliferation, and differentiation were analyzed collectively by quantitative PCR analyses, and biochemical and immunocytochemical assays. F-actin staining showed that MSCs maintained their fibroblastic morphology during the 3 weeks of culture. The addition of CTGF to the constructs resulted in an enhanced cell proliferation, elevated expression of fibroblast-specific protein-1, and decreased expression of mesenchymal surface epitopes without markedly triggering chondrogenesis, osteogenesis, adipogenesis, or apoptosis. At the mRNA level, CTGF supplement resulted in a decreased expression of collagen I and tissue inhibitor of metalloproteinase 1, but an increased expression of decorin and hyaluronic acid synthesase 3. At the protein level, collagen I, collagen III, sulfated glycosaminoglycan, and elastin productivity was higher in the conditioned PGS-PCL culture than in the normal culture. These findings collectively demonstrate that the fibrous mesh, when combined with defined biochemical cues, is capable of fostering MSC fibroblastic differentiation in vitro.


Subject(s)
Cell Survival/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Tissue Scaffolds/chemistry , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/physiology , Cells, Cultured , Connective Tissue Growth Factor , Humans , Immunohistochemistry , Tissue Engineering/methods
SELECTION OF CITATIONS
SEARCH DETAIL