Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Chem Phys ; 159(14)2023 Oct 14.
Article in English | MEDLINE | ID: mdl-37811828

ABSTRACT

Metal-reducing bacteria have adapted the ability to respire extracellular solid surfaces instead of soluble oxidants. This process requires an electron transport pathway that spans from the inner membrane, across the periplasm, through the outer membrane, and to an external surface. Multiheme cytochromes are the primary machinery for moving electrons through this pathway. Recent studies show that the chiral-induced spin selectivity (CISS) effect is observable in some of these proteins extracted from the model metal-reducing bacteria, Shewanella oneidensis MR-1. It was hypothesized that the CISS effect facilitates efficient electron transport in these proteins by coupling electron velocity to spin, thus reducing the probability of backscattering. However, these studies focused exclusively on the cell surface electron conduits, and thus, CISS has not been investigated in upstream electron transfer components such as the membrane-associated MtrA, or periplasmic proteins such as small tetraheme cytochrome (STC). By using conductive probe atomic force microscopy measurements of protein monolayers adsorbed onto ferromagnetic substrates, we show that electron transport is spin selective in both MtrA and STC. Moreover, we have determined the spin polarization of MtrA to be ∼77% and STC to be ∼35%. This disparity in spin polarizations could indicate that spin selectivity is length dependent in heme proteins, given that MtrA is approximately two times longer than STC. Most significantly, our study indicates that spin-dependent interactions affect the entire extracellular electron transport pathway.


Subject(s)
Electrons , Periplasm , Electron Transport , Oxidation-Reduction , Periplasm/metabolism , Metals , Bacteria/metabolism , Bacterial Proteins/metabolism , Bacterial Outer Membrane Proteins/metabolism
2.
Bioeng Transl Med ; 8(3): e10495, 2023 May.
Article in English | MEDLINE | ID: mdl-37206238

ABSTRACT

Macro-encapsulation systems for delivery of cellular therapeutics in diabetes treatment offer major advantages such as device retrievability and high cell packing density. However, microtissue aggregation and absence of vasculature have been implicated in the inadequate transfer of nutrients and oxygen to the transplanted cellular grafts. Herein, we develop a hydrogel-based macrodevice to encapsulate therapeutic microtissues positioned in homogeneous spatial distribution to mitigate their aggregation while concurrently supporting an organized intra-device network of vascular-inductive cells. Termed Waffle-inspired Interlocking Macro-encapsulation (WIM) device, this platform comprises two modules with complementary topography features that fit together in a lock-and-key configuration. The waffle-inspired grid-like micropattern of the "lock" component effectively entraps insulin-secreting microtissues in controlled locations while the interlocking design places them in a co-planar spatial arrangement with close proximity to vascular-inductive cells. The WIM device co-laden with INS-1E microtissues and human umbilical vascular endothelial cells (HUVECs) maintains desirable cellular viability in vitro with the encapsulated microtissues retaining their glucose-responsive insulin secretion while embedded HUVECs express pro-angiogenic markers. Furthermore, a subcutaneously implanted alginate-coated WIM device encapsulating primary rat islets achieves blood glucose control for 2 weeks in chemically induced diabetic mice. Overall, this macrodevice design lays foundation for a cell delivery platform, which has the potential to facilitate nutrients and oxygen transport to therapeutic grafts and thereby might lead to improved disease management outcome.

3.
ACS Omega ; 7(24): 21337-21345, 2022 Jun 21.
Article in English | MEDLINE | ID: mdl-35755371

ABSTRACT

Gadolinium chelates are employed worldwide today as clinical contrast agents for magnetic resonance imaging. Until now, the commonly used linear contrast agents based on the rare-earth element gadolinium have been considered safe and well-tolerated. Recently, concerns regarding this type of contrast agent have been reported, which is why there is an urgent need to develop the next generation of stable contrast agents with enhanced spin-lattice relaxation, as measured by improved T 1 relaxivity at lower doses. Here, we show that by the integration of gadolinium ions in cerium oxide nanoparticles, a stable crystalline 5 nm sized nanoparticulate system with a homogeneous gadolinium ion distribution is obtained. These cerium oxide nanoparticles with entrapped gadolinium deliver strong T 1 relaxivity per gadolinium ion (T 1 relaxivity, r 1 = 12.0 mM-1 s-1) with the potential to act as scavengers of reactive oxygen species (ROS). The presence of Ce3+ sites and oxygen vacancies at the surface plays a critical role in providing the antioxidant properties. The characterization of radial distribution of Ce3+ and Ce4+ oxidation states indicated a higher concentration of Ce3+ at the nanoparticle surfaces. Additionally, we investigated the ROS-scavenging capabilities of pure gadolinium-containing cerium oxide nanoparticles by bioluminescent imaging in vivo, where inhibitory effects on ROS activity are shown.

4.
Macromol Biosci ; 22(7): e2100472, 2022 07.
Article in English | MEDLINE | ID: mdl-35261175

ABSTRACT

Environmental accumulation of non-degradable polystyrene (PS) microparticles from plastic waste poses potential adverse impact on marine life and human health. Herein, microparticles from a degradable PS analogue (dePS) are formulated and their immuno-modulatory characteristics are comprehensively evaluated. Both dePS copolymer and microparticles are chemically degradable under accelerated hydrolytic condition. In vitro studies show that dePS microparticles are non-toxic to three immortalized cell lines. While dePS microparticles do not induce macrophage polarization in vitro, dePS microparticles induce in vivo upregulation of both pro-inflammatory and anti-inflammatory biomarkers in immuno-competent mice, suggesting the coexistence of mixed phenotypes of macrophages in the host immune response to these microparticles. Interestingly, on day 7 following subcutaneous in mice, dePS microparticles induce a lower level of several immuno-modulatory biomarkers (matrix metallo-proteinases (MMPs), tumor necrosis factor (TNF-α), and arginase activity) compared to that of reference poly(lactic-co-glycolic acid) microparticles. Remarkably, compared to PS microparticles, dePS microparticles exhibit similar in vitro and in vivo bioactivity while acquiring additional chemical degradability. Overall, this study gains new insights into the host immune response to dePS microparticles and suggests that this dePS analogue might be explored as an alternative material choice for biomedical and consumer care applications.


Subject(s)
Macrophages , Polystyrenes , Animals , Humans , Immunity , Macrophages/metabolism , Mice , Polylactic Acid-Polyglycolic Acid Copolymer/metabolism , Polystyrenes/pharmacology , Tumor Necrosis Factor-alpha/metabolism
5.
Acta Biomater ; 117: 167-179, 2020 11.
Article in English | MEDLINE | ID: mdl-32977069

ABSTRACT

Systemic drug administration has conventionally been prescribed to alleviate persistent local inflammation which is prevalent in chronic diseases. However, this approach is associated with drug-induced toxicity, particularly when the dosage exceeds that necessitated by pathological conditions of diseased tissues. Herein, we developed a modular hybrid hydrogel which could be triggered to release an anti-inflammatory drug upon exposure to elevated protease activity associated with inflammatory diseases. Modular design of the hybrid hydrogel enabled independent optimization of its protease-cleavable and drug-loaded subdomains to facilitate hydrogel formation, cleavability by matrix-metalloprotease-9 (MMP-9), and tuning drug release rate. In vitro study demonstrated the protease-triggered enhancement of drug release from the hybrid hydrogel system for effective inhibition of TNF-α production by pro-inflammatory macrophages and suggested its potential to mitigate drug-induced cytotoxicity. Using non-invasive imaging to monitor the activity of reactive oxygen species in biomaterial-induced host response, we confirmed that the hybrid hydrogel and its constituent materials did not induce adverse immune response after 5 days following their subcutaneous injection in immuno-competent mice. We subsequently incorporated this hybrid hydrogel onto a commercial wound dressing which could release the drug upon exposure to MMP-9. Together, our findings suggested that this hybrid hydrogel might be a versatile platform for on-demand drug delivery via either injectable or topical application to modulate inflammation in chronic diseases.


Subject(s)
Hydrogels , Tumor Necrosis Factor-alpha , Animals , Drug Delivery Systems , Drug Liberation , Hydrogels/pharmacology , Macrophages , Mice , Peptide Hydrolases
6.
Acta Biomater ; 97: 260-271, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31404714

ABSTRACT

Transplantation of immuno-isolated islets is a promising strategy to restore insulin-secreting function in patients with Type 1 diabetes. However, the clinical translation of this treatment approach remains elusive due to the loss of islet viability resulting from hypoxia at the avascular transplantation site. To address this challenge, we designed non-spherical islet-like microtissues and investigated the effect of their geometries on cellular viability. Insulin-secreting microtissues with different shapes were fabricated by assembly of monodispersed rat insulinoma beta cells on micromolded nonadhesive hydrogels. Our study quantitatively demonstrated that toroid microtissues exhibited enhanced cellular viability and metabolic activity compared to rod and spheroid microtissues with the same volume. At a similar level of cellular viability, toroid geometry facilitated efficient packing of more cells into each microtissue than rod and spheroid geometries. In addition, toroid microtissues maintained the characteristic glucose-responsive insulin secretion of rat insulinoma beta cells. Furthermore, toroid microtissues preserved their geometry and structural integrity following their microencapsulation in immuno-isolatory alginate hydrogel. Our study suggests that adopting toroid geometry in designing therapeutic microtissues potentially reduces mass loss of cellular grafts and thereby may improve the performance of transplanted islets towards a clinically viable cure for Type 1 diabetes. STATEMENT OF SIGNIFICANCE: Transplantation of therapeutic cells is a promising strategy for the treatment of a wide range of hormone or protein-deficiency diseases. However, the clinical application of this approach is hindered by the loss of cell viability and function at the avascular transplantation site. To address this challenge, we fabricated hydrogel-encapsulated islet-like microtissues with non-spheroidal geometry and optimal surface-to-volume ratio. This study demonstrated that the viability of therapeutic cells can be significantly increased solely by redesigning the microtissue configuration without requiring any additional biochemical or operational accessories. This study suggests that the adoption of toroid geometry provides a possible avenue to improve the long-term survival of transplanted therapeutic cells and expedite the translation of cell-based therapy towards clinical application.


Subject(s)
Cells, Immobilized/cytology , Hydrogels/chemistry , Islets of Langerhans/metabolism , Animals , Capsules , Cell Line, Tumor , Cell Survival , Cells, Immobilized/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/therapy , Islets of Langerhans/cytology , Rats
7.
Methods Enzymol ; 568: 219-53, 2016.
Article in English | MEDLINE | ID: mdl-26795473

ABSTRACT

The discovery of the causative link between keratin mutations and a growing number of human diseases opened the way for a better understanding of the function of the whole intermediate filament families of cytoskeleton proteins. This chapter describes analytical approaches to identification and interpretation of the consequences of keratin mutations, from the clinical and diagnostic level to cells in tissue culture. Intermediate filament pathologies can be accurately diagnosed from skin biopsies and DNA samples. The Human Intermediate Filament Database collates reported mutations in intermediate filament genes and their diseases, and can help clinicians to establish accurate diagnoses, leading to disease stratification for genetic counseling, optimal care delivery, and future mutation-aligned new therapies. Looking at the best-studied keratinopathy, epidermolysis bullosa simplex, the generation of cell lines mimicking keratinopathies is described, in which tagged mutant keratins facilitate live-cell imaging to make use of today's powerful enhanced light microscopy modalities. Cell stress assays such as cell spreading and cell migration in scratch wound assays can interrogate the consequences of the compromised cytoskeletal network. Application of extrinsic stresses, such as heat, osmotic, or mechanical stress, can enhance the differentiation of mutant keratin cells from wild-type cells. To bring the experiments to the next level, 3D organotypic human cultures can be generated, and even grafted onto the backs of immunodeficient mice for greater in vivo relevance. While development of these assays has focused on mutant K5/K14 cells, the approaches are often applicable to mutations in other intermediate filaments, reinforcing fundamental commonalities in spite of diverse clinical pathologies.


Subject(s)
Epidermis/metabolism , Intermediate Filaments/metabolism , Keratins/metabolism , Animals , Epidermolysis Bullosa/genetics , Epidermolysis Bullosa/metabolism , Humans , Intermediate Filaments/genetics , Keratins/genetics , Mutation
8.
Adv Mater ; 25(44): 6385-91, 2013 Nov 26.
Article in English | MEDLINE | ID: mdl-23996513

ABSTRACT

Incorporating graphene oxide inside GelMA hydrogels enhances their mechanical properties and reduces UV-induced cell damage while preserving their favorable characteristics for 3D cell encapsulation. NIH-3T3 fibroblasts encapsulated in GO-GelMA microgels demonstrate excellent cellular viability, proliferation, spreading, and alignment. GO reinforcement combined with a multi-stacking approach offers a facile engineering strategy for the construction of complex artificial tissues.


Subject(s)
Gelatin/chemistry , Graphite/chemistry , Hydrogels/chemistry , Animals , Cell Survival , Mice , Microscopy , NIH 3T3 Cells , Oxides/chemistry , Porosity , Tissue Engineering , Ultraviolet Rays
9.
ACS Nano ; 7(8): 6758-66, 2013 Aug 27.
Article in English | MEDLINE | ID: mdl-23834678

ABSTRACT

A glucose-responsive closed-loop insulin delivery system represents the ideal treatment of type 1 diabetes mellitus. In this study, we develop uniform injectable microgels for controlled glucose-responsive release of insulin. Monodisperse microgels (256 ± 18 µm), consisting of a pH-responsive chitosan matrix, enzyme nanocapsules, and recombinant human insulin, were fabricated through a one-step electrospray procedure. Glucose-specific enzymes were covalently encapsulated into the nanocapsules to improve enzymatic stability by protecting from denaturation and immunogenicity as well as to minimize loss due to diffusion from the matrix. The microgel system swelled when subjected to hyperglycemic conditions, as a result of the enzymatic conversion of glucose into gluconic acid and protonation of the chitosan network. Acting as a self-regulating valve system, microgels were adjusted to release insulin at basal release rates under normoglycemic conditions and at higher rates under hyperglycemic conditions. Finally, we demonstrated that these microgels with enzyme nanocapsules facilitate insulin release and result in a reduction of blood glucose levels in a mouse model of type 1 diabetes.


Subject(s)
Drug Carriers/chemistry , Drug Delivery Systems , Enzymes/chemistry , Glucose/chemistry , Insulin/administration & dosage , Animals , Blood Glucose/analysis , Catalysis , Chitosan/chemistry , Disease Models, Animal , Gels , Gluconates/chemistry , Humans , Hydrogen-Ion Concentration , Hyperglycemia/drug therapy , Insulin/chemistry , Mice , Microscopy, Electron, Transmission , Nanocapsules/chemistry , Recombinant Proteins/chemistry , Time Factors
10.
Biomaterials ; 34(23): 5792-801, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23660251

ABSTRACT

Immuno-isolation of islets has the potential to enable the replacement of pancreatic function in diabetic patients. However, host response to the encapsulated islets frequently leads to fibrotic overgrowth with subsequent impairment of the transplanted grafts. Here, we identified and incorporated anti-inflammatory agents into islet-containing microcapsules to address this challenge. In vivo subcutaneous screening of 16 small molecule anti-inflammatory drugs was performed to identify promising compounds that could minimize the formation of fibrotic cell layers. Using parallel non-invasive fluorescent and bioluminescent imaging, we identified dexamethasone and curcumin as the most effective drugs in inhibiting the activities of inflammatory proteases and reactive oxygen species in the host response to subcutaneously injected biomaterials. Next, we demonstrated that co-encapsulating curcumin with pancreatic rat islets in alginate microcapsules reduced fibrotic overgrowth and improved glycemic control in a mouse model of chemically-induced type I diabetes. These results showed that localized administration of anti-inflammatory drug can improve the longevity of encapsulated islets and may facilitate the translation of this technology toward a long-term cure for type I diabetes.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Capsules/chemistry , Diabetes Mellitus, Experimental/therapy , Islets of Langerhans Transplantation , Islets of Langerhans/immunology , Animals , Anti-Inflammatory Agents/pharmacology , Cathepsins/metabolism , Diabetes Mellitus, Experimental/drug therapy , Fibrosis , Islets of Langerhans/drug effects , Lactic Acid/chemistry , Male , Mice , Mice, Inbred C57BL , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use
11.
ACS Nano ; 7(5): 4194-201, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23638642

ABSTRACT

Diabetes mellitus, a disorder of glucose regulation, is a global burden affecting 366 million people across the world. An artificial "closed-loop" system able to mimic pancreas activity and release insulin in response to glucose level changes has the potential to improve patient compliance and health. Herein we develop a glucose-mediated release strategy for the self-regulated delivery of insulin using an injectable and acid-degradable polymeric network. Formed by electrostatic interaction between oppositely charged dextran nanoparticles loaded with insulin and glucose-specific enzymes, the nanocomposite-based porous architecture can be dissociated and subsequently release insulin in a hyperglycemic state through the catalytic conversion of glucose into gluconic acid. In vitro insulin release can be modulated in a pulsatile profile in response to glucose concentrations. In vivo studies validated that these formulations provided improved glucose control in type 1 diabetic mice subcutaneously administered with a degradable nano-network. A single injection of the developed nano-network facilitated stabilization of the blood glucose levels in the normoglycemic state (<200 mg/dL) for up to 10 days.


Subject(s)
Drug Carriers/administration & dosage , Drug Carriers/chemistry , Glucose/metabolism , Insulin/metabolism , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Alginates/chemistry , Animals , Biocompatible Materials/administration & dosage , Biocompatible Materials/chemistry , Chitosan/chemistry , Dextrans/chemistry , Diabetes Mellitus, Type 1/drug therapy , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Humans , Injections , Insulin/chemistry , Insulin/therapeutic use , Male , Mice , Mice, Inbred C57BL
12.
Adv Healthc Mater ; 2(5): 667-72, 2013 May.
Article in English | MEDLINE | ID: mdl-23208618

ABSTRACT

Islets microencapsulation holds great promise to treat type 1 diabetes. Currently used alginate microcapsules often have islets protruding outside capsules, leading to inadequate immuno-protection. A novel design of microcapsules with core-shell structures using a two-fluid co-axial electro-jetting is reported. Improved encapsulation and diabetes correction is achieved in a single step by simply confining the islets in the core region of the capsules.


Subject(s)
Alginates/chemistry , Capsules/chemical synthesis , Diabetes Mellitus, Experimental/surgery , Hydrogels/administration & dosage , Hydrogels/chemical synthesis , Islets of Langerhans Transplantation/methods , Tissue Scaffolds , Animals , Biocompatible Materials/chemical synthesis , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cells, Cultured , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/pathology , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Male , Mice , Mice, Inbred C57BL , Porosity , Streptozocin , Treatment Outcome
13.
Proc Natl Acad Sci U S A ; 109(52): 21444-9, 2012 Dec 26.
Article in English | MEDLINE | ID: mdl-23236189

ABSTRACT

The treatment of diseased vasculature remains challenging, in part because of the difficulty in implanting drug-eluting devices without subjecting vessels to damaging mechanical forces. Implanting materials using adhesive forces could overcome this challenge, but materials have previously not been shown to durably adhere to intact endothelium under blood flow. Marine mussels secrete strong underwater adhesives that have been mimicked in synthetic systems. Here we develop a drug-eluting bioadhesive gel that can be locally and durably glued onto the inside surface of blood vessels. In a mouse model of atherosclerosis, inflamed plaques treated with steroid-eluting adhesive gels had reduced macrophage content and developed protective fibrous caps covering the plaque core. Treatment also lowered plasma cytokine levels and biomarkers of inflammation in the plaque. The drug-eluting devices developed here provide a general strategy for implanting therapeutics in the vasculature using adhesive forces and could potentially be used to stabilize rupture-prone plaques.


Subject(s)
Adhesives/chemistry , Blood Vessels/pathology , Dexamethasone/therapeutic use , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/pathology , Adhesiveness/drug effects , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Arteries/drug effects , Arteries/pathology , Blood Vessels/drug effects , Catechols/chemistry , Dexamethasone/pharmacology , Drug Delivery Systems , Female , Gels/chemistry , Human Umbilical Vein Endothelial Cells/drug effects , Implants, Experimental , Inflammation/pathology , Mice , Mice, Inbred C57BL , Solubility , Stress, Mechanical , Stress, Physiological/drug effects
14.
Biomacromolecules ; 13(10): 3031-8, 2012 Oct 08.
Article in English | MEDLINE | ID: mdl-22920794

ABSTRACT

Synthetic scaffolds are crucial to applications in regenerative medicine; however, the foreign body response can impede regeneration and may lead to failure of the implant. Herein we report the development of a tissue engineering scaffold that allows attachment and proliferation of regenerating cells while reducing the foreign body response by localized delivery of an anti-inflammatory agent. Electrospun fibers composed of poly(l-lactic) acid (PLLA) and poly(ε-caprolactone) (PCL) were prepared with and without the steroid anti-inflammatory drug, dexamethasone. Analysis of subcutaneous implants demonstrated that the PLLA fibers encapsulating dexamethasone evoked a less severe inflammatory response than the other fibers examined. They also displayed a controlled release of dexamethasone over a period of time conducive to tissue regeneration and allowed human mesenchymal stem cells to adhere to and proliferate on them in vitro. These observations demonstrate their potential as a building block for tissue engineering scaffolds.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Biocompatible Materials/chemistry , Dexamethasone/administration & dosage , Dexamethasone/pharmacology , Foreign-Body Reaction/prevention & control , Mesenchymal Stem Cells/cytology , Anti-Inflammatory Agents/therapeutic use , Biocompatible Materials/chemical synthesis , Cell Adhesion , Cell Proliferation , Cell Survival , Dexamethasone/therapeutic use , Foreign-Body Reaction/drug therapy , Humans , Lactic Acid/chemistry , Mesenchymal Stem Cells/drug effects , Particle Size , Polyesters/chemistry , Polymers/chemistry , Surface Properties , Temperature , Tissue Engineering/methods
15.
Annu Rev Phys Chem ; 63: 541-69, 2012.
Article in English | MEDLINE | ID: mdl-22404587

ABSTRACT

The recent literature on photochemical and photoelectrochemical reductions of CO(2) is reviewed. The different methods of achieving light absorption, electron-hole separation, and electrochemical reduction of CO(2) are considered. Energy gap matching for reduction of CO(2) to different products, including CO, formic acid, and methanol, is used to identify the most promising systems. Different approaches to lowering overpotentials and achieving high chemical selectivities by employing catalysts are described and compared.

16.
Biomaterials ; 32(19): 4464-70, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21429573

ABSTRACT

In general, biomaterials induce a non-specific host response when implanted in the body. This reaction has the potential to interfere with the function of the implanted materials. One method for controlling the host response is through local, controlled-release of anti-inflammatory agents. Herein, we investigate the spatial and temporal effects of an anti-inflammatory drug on the cellular dynamics of the innate immune response to subcutaneously implanted poly(lactic-co-glycolic) microparticles. Noninvasive fluorescence imaging was used to investigate the influence of dexamethasone drug loading and release kinetics on the local and systemic inhibition of inflammatory cellular activities. Temporal monitoring of host response showed that inhibition of inflammatory proteases in the early phase was correlated with decreased cellular infiltration in the later phase of the foreign body response. We believe that using controlled-release anti-inflammatory platforms to modulate early cellular dynamics will be useful in reducing the foreign body response to implanted biomaterials and medical devices.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Delayed-Action Preparations , Inflammation/drug therapy , Prostheses and Implants/adverse effects , Animals , Anti-Inflammatory Agents/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Dexamethasone/administration & dosage , Dexamethasone/chemistry , Dexamethasone/therapeutic use , Drug Carriers/chemistry , Drug Carriers/metabolism , Foreign-Body Reaction/drug therapy , Lactic Acid/chemistry , Lactic Acid/immunology , Male , Materials Testing , Mice , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer
17.
Biomaterials ; 32(7): 1796-801, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21146868

ABSTRACT

The non-specific host response to implanted biomaterials is often a key challenge of medical device design. To evaluate biocompatibility, measuring the release of reactive oxygen species (ROS) produced by inflammatory cells in response to biomaterial surfaces is a well-established method. However, the detection of ROS in response to materials implanted in vivo has not yet been demonstrated. Here, we develop a bioluminescence whole animal imaging approach to observe ROS released in response to subcutaneously-implanted materials in live animals. We compared the real-time generation of ROS in response to two representative materials, polystyrene and alginate, over the course of 28 days. High levels of ROS were observed near polystyrene, but not alginate implants, and persisted throughout the course of 28 days. Histological analysis revealed that high levels of ROS correlated not only with the presence of phagocytic cells at early timepoints, but also fibrosis at later timepoints, suggesting that ROS may be involved in both the acute and chronic phase of the foreign body response. These data are the first in vivo demonstration of ROS generation in response to implanted materials, and describe a novel technique to evaluate the host response.


Subject(s)
Biocompatible Materials/adverse effects , Prostheses and Implants/adverse effects , Reactive Oxygen Species/metabolism , Animals , Cells, Cultured , Free Radicals/metabolism , Immunohistochemistry , Male , Mice
18.
PLoS One ; 5(4): e10032, 2010 Apr 06.
Article in English | MEDLINE | ID: mdl-20386609

ABSTRACT

BACKGROUND: Many materials are unsuitable for medical use because of poor biocompatibility. Recently, advances in the high throughput synthesis of biomaterials has significantly increased the number of potential biomaterials, however current biocompatibility analysis methods are slow and require histological analysis. METHODOLOGY/PRINCIPAL FINDINGS: Here we develop rapid, non-invasive methods for in vivo quantification of the inflammatory response to implanted biomaterials. Materials were placed subcutaneously in an array format and monitored for host responses as per ISO 10993-6: 2001. Host cell activity in response to these materials was imaged kinetically, in vivo using fluorescent whole animal imaging. Data captured using whole animal imaging displayed similar temporal trends in cellular recruitment of phagocytes to the biomaterials compared to histological analysis. CONCLUSIONS/SIGNIFICANCE: Histological analysis similarity validates this technique as a novel, rapid approach for screening biocompatibility of implanted materials. Through this technique there exists the possibility to rapidly screen large libraries of polymers in vivo.


Subject(s)
Biocompatible Materials , Diagnostic Imaging/methods , Fluorescence , Materials Testing/methods , Animals , Macrophage Activation , Mice , Models, Animal , Peptide Hydrolases/metabolism , Phagocytes
19.
Biomaterials ; 30(36): 6896-902, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19800116

ABSTRACT

Cell encapsulation has been broadly investigated as a technology to provide immunoprotection for transplanted endocrine cells. Here we develop a new fabrication method that allows for rapid, homogenous microencapsulation of insulin-secreting cells with varying microscale geometries and asymmetrically modified surfaces. Micromolding systems were developed using polypropylene mesh, and the material/surface properties associated with efficient encapsulation were identified. Cells encapsulated using these methods maintain desirable viability and preserve their ability to proliferate and secrete insulin in a glucose-responsive manner. This new cell encapsulation approach enables a practical route to an inexpensive and convenient process for the generation of cell-laden microcapsules without requiring any specialized equipment or microfabrication process.


Subject(s)
Biocompatible Materials/chemistry , Capsules/chemistry , Hydrogels/chemistry , Insulin-Secreting Cells , Microtechnology , Alginates/chemistry , Animals , Cell Line , Cell Proliferation , Cell Survival , Glucose/metabolism , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Materials Testing , Microtechnology/instrumentation , Microtechnology/methods , Rats , Surface Properties
20.
Small ; 5(13): 1575-81, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19296563

ABSTRACT

Degradable microparticles have broad utility as vehicles for drug delivery and form the basis of several therapies approved by the US Food and Drug Administration. Conventional emulsion-based methods of manufacturing produce particles with a wide range of diameters (and thus kinetics of release) in each batch. This paper describes the fabrication of monodisperse, drug-loaded microparticles from biodegradable polymers using the microfluidic flow-focusing (FF) devices and the drug-delivery properties of those particles. Particles are engineered with defined sizes, ranging from 10 microm to 50 microm. These particles are nearly monodisperse (polydispersity index = 3.9%). A model amphiphilic drug (bupivacaine) is incorporated within the biodegradable matrix of the particles. Kinetic analysis shows that the release of the drug from these monodisperse particles is slower than that from conventional methods of the same average size but a broader distribution of sizes and, most importantly, exhibit a significantly lower initial burst than that observed with conventional particles. The difference in the initial kinetics of drug release is attributed to the uniform distribution of the drug inside the particles generated using the microfluidic methods. These results demonstrate the utility of microfluidic FF for the generation of homogenous systems of particles for the delivery of drugs.


Subject(s)
Colloids/chemistry , Delayed-Action Preparations/chemistry , Lactic Acid/chemistry , Microfluidics/instrumentation , Microfluidics/methods , Polyglycolic Acid/chemistry , Absorption , Equipment Design , Equipment Failure Analysis , Materials Testing , Microspheres , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer
SELECTION OF CITATIONS
SEARCH DETAIL
...