Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
1.
Infect Dis Ther ; 10(2): 1015-1022, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33432534

ABSTRACT

INTRODUCTION: The gold standard for COVID-19 diagnosis is currently a real-time reverse transcriptase polymerase chain reaction (RT-PCR) to detect SARS-CoV-2. This is most commonly performed on respiratory secretions obtained via a nasopharyngeal swab. Due to supply chain limitations and high demand worldwide because of the COVID-19 pandemic, access to commercial nasopharyngeal swabs has not been assured. 3D printing methods have been used to meet the shortfall. For longer-term considerations, 3D printing may not compare well with injection molding as a production method due to the challenging scalability and greater production costs of 3D printing. METHODS: To secure sufficient nasopharyngeal swab availability for our national healthcare system, we designed a novel injection molded nasopharyngeal swab (the IM2 swab). We performed a clinical diagnostic study comparing the IM2 swab to the Copan FLOQSwab. Forty patients with a known diagnosis of COVID-19 and 10 healthy controls were recruited. Paired nasopharyngeal swabs were obtained from the same nostril of each participant and tested for SARS-CoV-2 by RT-PCR. RESULTS: When compared to the Copan FLOQswab, results from the IM2 swab displayed excellent overall agreement and positive percent agreement of 96.0% and 94.9%, respectively. There was no significant difference in mean RT-PCR cycle threshold values for the ORF1ab (28.05 vs. 28.03, p = 0.97) and E-gene (29.72 vs. 29.37, p = 0.64) targets, respectively. We did not observe any significant adverse events and there was no significant difference in patient-reported pain. CONCLUSION: In summary, the IM2 nasopharyngeal swab is a clinically safe, highly accurate option to commercial nasopharyngeal swabs.

2.
Sci Rep ; 8(1): 3937, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29500447

ABSTRACT

Tissue development, regeneration, or de-novo tissue engineering in-vitro, are based on reciprocal cell-niche interactions. Early tissue formation mechanisms, however, remain largely unknown given complex in-vivo multifactoriality, and limited tools to effectively characterize and correlate specific micro-scaled bio-mechanical interplay. We developed a unique model system, based on decellularized porcine cardiac extracellular matrices (pcECMs)-as representative natural soft-tissue biomaterial-to study a spectrum of common cell-niche interactions. Model monocultures and 1:1 co-cultures on the pcECM of human umbilical vein endothelial cells (HUVECs) and human mesenchymal stem cells (hMSCs) were mechano-biologically characterized using macro- (Instron), and micro- (AFM) mechanical testing, histology, SEM and molecular biology aspects using RT-PCR arrays. The obtained data was analyzed using developed statistics, principal component and gene-set analyses tools. Our results indicated biomechanical cell-type dependency, bi-modal elasticity distributions at the micron cell-ECM interaction level, and corresponding differing gene expression profiles. We further show that hMSCs remodel the ECM, HUVECs enable ECM tissue-specific recognition, and their co-cultures synergistically contribute to tissue integration-mimicking conserved developmental pathways. We also suggest novel quantifiable measures as indicators of tissue assembly and integration. This work may benefit basic and translational research in materials science, developmental biology, tissue engineering, regenerative medicine and cancer biomechanics.


Subject(s)
Cell Lineage , Biomechanical Phenomena , Cell Differentiation , Coculture Techniques , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Extracellular Matrix/metabolism , Gene Expression Profiling , Human Umbilical Vein Endothelial Cells , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Microscopy, Atomic Force , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Tissue Engineering/methods
3.
Bioeng Transl Med ; 2(2): 156-169, 2017 06.
Article in English | MEDLINE | ID: mdl-29313029

ABSTRACT

Cardiovascular disease is a major cause of morbidity and mortality, especially in developed countries. Most academic research efforts in cardiovascular disease management focus on pharmacological interventions, or are concerned with discovering new disease markers for diagnosis and monitoring. Nonpharmacological interventions with therapeutic devices, conversely, are driven largely by novel materials and device design. Examples of such devices include coronary stents, heart valves, ventricular assist devices, and occluders for septal defects. Until recently, development of such devices remained largely with medical device companies. We trace the materials evolution story in two of these devices (stents and occluders), while also highlighting academic contributions, including our own, to the evolution story. Specifically, it addresses not only our successes, but also the challenges facing the translatability of concepts generated via academic research.

4.
Acta Biomater ; 50: 220-233, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27956366

ABSTRACT

Injectable scaffolds for cardiac tissue regeneration are a promising therapeutic approach for progressive heart failure following myocardial infarction (MI). Their major advantage lies in their delivery modality that is considered minimally invasive due to their direct injection into the myocardium. Biomaterials comprising such scaffolds should mimic the cardiac tissue in terms of composition, structure, mechanical support, and most importantly, bioactivity. Nonetheless, natural biomaterial-based gels may suffer from limited mechanical strength, which often fail to provide the long-term support required by the heart for contraction and relaxation. Here we present newly-developed injectable scaffolds, which are based on solubilized decellularized porcine cardiac extracellular matrix (pcECM) cross-linked with genipin alone or engineered with different amounts of chitosan to better control the gel's mechanical properties while still leveraging the ECM biological activity. We demonstrate that these new biohybrid materials are naturally remodeled by mesenchymal stem cells, while supporting high viabilities and affecting cell morphology and organization. They exhibit neither in vitro nor in vivo immunogenicity. Most importantly, their application in treating acute and long term chronic MI in rat models clearly demonstrates the significant therapeutic potential of these gels in the long-term (12weeks post MI). The pcECM-based gels enable not only preservation, but also improvement in cardiac function eight weeks post treatment, as measured using echocardiography as well as hemodynamics. Infiltration of progenitor cells into the gels highlights the possible biological remodeling properties of the ECM-based platform. STATEMENT OF SIGNIFICANCE: This work describes the development of new injectable scaffolds for cardiac tissue regeneration that are based on solubilized porcine cardiac extracellular matrix (ECM), combined with natural biomaterials: genipin, and chitosan. The design of such scaffolds aims at leveraging the natural bioactivity and unique structure of cardiac ECM, while overcoming its limited mechanical strength, which may fail to provide the long-term support required for heart contraction and relaxation. Here, we present a biocompatible gel-platform with custom-tailored mechanical properties that significantly improve cardiac function when injected into rat hearts following acute and chronic myocardial infarction. We clearly demonstrate the substantial therapeutic potential of these scaffolds, which not only preserved heart functions but also alleviated MI damage, even after the formation of a mature scar tissue.


Subject(s)
Extracellular Matrix/chemistry , Hydrogels , Myocardial Infarction/therapy , Myocardium/metabolism , Tissue Scaffolds/chemistry , Animals , Cell Line , Chitosan/chemistry , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Iridoids/chemistry , Male , Mesenchymal Stem Cells/metabolism , Mice , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardium/pathology , Rats , Rats, Wistar
5.
Acta Biomater ; 44: 209-220, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27545814

ABSTRACT

OBJECTIVE: To evaluate the regenerative capacity of non-supplemented and bioactive patches made of decellularized porcine cardiac extracellular matrix (pcECM) and characterize the biological key factors involved in possible cardiac function (CF) restoration following acute and 8weeks chronic MI. BACKGROUND: pcECM is a key natural biomaterial that can affect cardiac regeneration following myocardial infarction (MI), through mechanisms, which are still not clearly understood. METHODS: Wistar rats underwent MI and received pcECM patch (pcECM-P) treatment in either acute or chronic inflammatory phases. Treated, sham operated (no MI), and control (MI without treatment) animals, were compared through echocardiography, hemodynamics, pathological evaluation and analyses of various mRNA and protein level markers. RESULTS: Our results show that in both acute and long-term chronic MI models, pcECM promotes significant cardiac function improvement, which is correlated to progenitor (GATA4(+), c-kit(+)) and myocyte (MYLC(+), TRPI(+)) recruitment. Interestingly, recruited progenitors, isolated using laser capture microdissection (LCM), expressed both early and late cardiomyocyte (CM) differentiation markers, suggesting differentiation towards the CM lineage. Recruited CM-like cells organized in a partially striated and immature muscle fiber arrangement that presented connexin43 -a crucial mediator of cardiac electrical conductivity. Concomitantly, pcECM was rapidly vascularized, and induced a constructive remodeling process as indicated by increased M2/M1 macrophage phenotypic ratio and pathological evaluation. CONCLUSIONS: Acellular pcECM patch implants alone, i.e., without added biologics, are bioactive, and exert potent efficacy, stimulating biological regenerative processes that cooperatively lead to a cardiac progenitor-based restoration of function, even after scar tissue had already formed. STATEMENT OF SIGNIFICANCE: MI ('heart attack') remains the leading cause of heart failure and death in developed-countries. Restoration of cardiac function requires active turnover of damaged heart contracting cells (CM), however, CM endogenous regeneration is not efficient and is a matter of controversy. We show that a bioactive biomaterial alone-decellularized heart tissue (pcECM)-without added cells or growth factors, can elicit a complex regenerative response even after irreversible scarring. The pcECM patch induces macrophage polarization towards constructive remodeling and cardiomyocyte progenitor cell (GATA4(+), c-kit(+)) recruitment (evidenced at both mRNA and protein levels) resulting in de novo immature striated-like muscle patterns (MLC(+), TrpI(+), connexin43(+)). We, therefore, suggest this bioactive pcECM can model cardiac regeneration, and serve as a candidate for fast-track clinical application.


Subject(s)
Cicatrix/pathology , Extracellular Matrix/metabolism , Myocardium/metabolism , Regeneration , Stem Cells/cytology , Animals , Cell Count , Hemodynamics , Implants, Experimental , Macrophages/pathology , Male , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Neovascularization, Physiologic , Rats, Wistar , Sus scrofa
6.
ACS Nano ; 10(5): 5362-73, 2016 05 24.
Article in English | MEDLINE | ID: mdl-27148792

ABSTRACT

Recently, Ag-Ag2S hybrid nanostructures have attracted a great deal of attention due to their enhanced chemical and thermal stability, in addition to their morphology- and composition-dependent tunable local surface plasmon resonances. Although Ag-Ag2S nanostructures can be synthesized via sulfidation of as-prepared anisotropic Ag nanoparticles, this process is poorly understood, often leading to materials with anomalous compositions, sizes, and shapes and, consequently, optical properties. In this work, we use theory and experiment to investigate the structural and plasmonic evolution of Ag-Ag2S nanoprisms during the sulfidation of Ag precursors. The previously observed red-shifted extinction of the Ag-Ag2S hybrid nanoprism as sulfidation occurs contradicts theoretical predictions, indicating that the reaction does not just occur at the prism tips as previously speculated. Our experiments show that sulfidation can induce either blue or red shifts in the extinction of the dipole plasmon mode, depending on reaction conditions. By elucidating the correlation with the final structure and morphology of the synthesized Ag-Ag2S nanoprisms, we find that, depending on the reaction conditions, sulfidation occurs on the prism tips and/or the (111) surfaces, leading to a core(Ag)-anisotropic shell(Ag2S) prism nanostructure. Additionally, we demonstrate that the direction of the shift in the dipole plasmon is a function of the relative amounts of Ag2S at the prism tips and Ag2S shell thickness around the prism.

7.
Colloids Surf B Biointerfaces ; 132: 253-63, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26052108

ABSTRACT

A straight forward strategy of heparin surface grafting employs a terminal reactive-aldehyde group introduced through nitrous acid depolymerization. An advanced method that allows simultaneously monitoring of both heparin molar mass and monomer/aldehyde ratio by size exclusion chromatography, multi-angle laser light scattering and UV-absorbance (SEC-MALLS-UV) has been developed to improve upon heparin surface grafting. Advancements over older methods allow quantitative characterization by direct (aldehyde absorbance) and indirect (Schiff-based absorbance) evaluation of terminal functional aldehydes. The indirect quantitation of functional aldehydes through labeling with aniline (and the formation of a Schiff-base) allows independent quantitation of both polymer mass and terminal functional groups with the applicable UV mass extinction coefficients determined. The protocol was subsequently used to synthesize an optimized heparin-aldehyde that had minimal polydispersity (PDI<2) and high reaction yields (yield >60% by mass). The 8 kDa weight averaged molar mass heparin-aldehyde was then grafted on polycaprolactone (PCL), a common implant material. This optimized heparin-aldehyde retained its antithrombin activity, assessed in freshly drawn blood or surface immobilized on PCL films. Anticoagulant activity was equal to or better than the 24 kDa unmodified heparin it was fragmented from.


Subject(s)
Aldehydes/chemistry , Biocompatible Materials , Heparin/analysis , Polyesters/chemistry , Spectrophotometry, Ultraviolet/methods , Heparin/chemistry , Schiff Bases/chemistry , Surface Properties
8.
J Mech Behav Biomed Mater ; 36: 143-60, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24846584

ABSTRACT

Patent Ductus Arteriosus (PDA) is a cardiovascular defect that occurs in 1 out of every 2000 births, and if left untreated, may lead to severe cardiovascular problems. Current options for occluding utilize meta scaffolds with polymer fabric, and are permanent. The purpose of this study was to develop a fully degradable occluder for the closure of PDA, that can be deployed percutaneously without open-heart surgery. For percutaneous deployment, both elasticity and sufficient mechanical strength are required of the device components. As this combination of properties is not achievable with currently-available homo- or copolymers, blends of biodegradable poly(ε-caprolactone) (PCL) and poly(L-lactide-co-ε-caprolactone) (PLC) with various compositions were studied as the potential material for the PDA occlusion device. Microstructures of this blend were characterized by differential scanning calorimetry (DSC) and tensile tests. DSC results demonstrated the immiscibility between PCL and its copolymer PLC. Furthermore, the mechanical properties, i.e. elastic modulus and strain recovery, of the blends could be largely tailored by changing the continuous phase component. Based on the thermo-mechanical tests, suitable blends were selected to fabricate a prototype of PDA occluder and its in vitro performance, in term of device recovery (from its sheathed configuration), biodegradation rate and blood compatibility, was evaluated. The current results indicate that the device is able to recover elastically from a sheath within 2-3min for deployment; the device starts to disintegrate within 5-6 months, and the materials have no adverse effects on the platelet and leucocyte components of the blood. Biocompatibility implantation studies of the device showed acceptable tissue response. Finally, an artificial PDA conduit was created in a pig model, and the device deployment was tested from a sheath: the device recovered within 2-3min of unsheathing and fully sealed the conduit, the device remains stable and is completely covered by tissue at 1 month follow up. Thus, a novel prototype for PDA occlusion that is fully degradable has been developed to overcome the limitations of the currently used metal/fabric devices.


Subject(s)
Biocompatible Materials/chemical synthesis , Ductus Arteriosus, Patent/therapy , Polyesters/chemistry , Septal Occluder Device , Animals , Biocompatible Materials/adverse effects , Compressive Strength , Ductus Arteriosus, Patent/diagnostic imaging , Elastic Modulus , Equipment Design , Equipment Failure Analysis , Feasibility Studies , Hardness , Polyesters/therapeutic use , Rabbits , Radiography , Tensile Strength , Treatment Outcome
9.
J Pharm Sci ; 103(5): 1400-10, 2014 May.
Article in English | MEDLINE | ID: mdl-24590596

ABSTRACT

In this work, we focused on the development and investigation of controlled release matrices for a novel cardiotherapeutic peptide, cenderitide (CD-NP) that has shown to be useful for control of ventricular remodeling. To circumvent the hydrophilicity disparity between CD-NP and hydrophobic polymer matrix, a cosolvent system (water/dichloromethane) was selected for investigation. The effect of emulsification conditions, addition of poly(ethylene glycol) (PEG) and its copolymer on the release mechanism and profile were investigated. To verify the retention of bioactivity of entrapped CD-NP in different formulations, the generation of 3',5' cyclic guanosine monophospate (cGMP) and the inhibition of human cardiac fibroblast (HCF) were evaluated. The results showed that neat poly(ε-caprolactone) matrices carried out via two distinct emulsification conditions had either an unacceptably high burst or incomplete release of CD-NP; and the addition of PEG and its copolymer obtained intermediate profiles. Our confocal laser scanning microscopy and surface morphological investigations showed that the copolymer excipient was superior in playing stabilizer role by colocalizing and redistributing peptide throughout the matrix, making the release less sensitive to emulsification conditions. Furthermore, the released CD-NP is able to generate the cGMP and inhibit the HCF proliferation. Our investigations showed that CD-NP-loaded platforms can be a feasible option to provide sustained antifibrotic moderation of fibrotic scar formation and be potentially used to alleviate the adverse effects of cardiac remodeling.


Subject(s)
Cardiovascular Diseases/drug therapy , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacology , Natriuretic Peptides/chemistry , Natriuretic Peptides/pharmacology , Snake Venoms/chemistry , Snake Venoms/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Chemistry, Pharmaceutical/methods , Cyclic GMP/metabolism , Emulsions/chemistry , Excipients/chemistry , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Methylene Chloride/chemistry , Polyesters/chemistry , Polyethylene Glycols/chemistry , Polymers/chemistry , Polymers/pharmacology , Ventricular Remodeling/drug effects , Water/chemistry
10.
Biomatter ; 4: e28238, 2014.
Article in English | MEDLINE | ID: mdl-24553126

ABSTRACT

Human umbilical vein endothelial cells (HUVECs) were successfully entrapped in polyethylene oxide (PEO) core /polycaprolactone (PCL) shell electrospun fibers thus creating a "bioactive fiber." The viability and release of biomolecules from the entrapped cells in the bioactive fibers were characterized. A key modification to the core solution was the inclusion of 50% fetal bovine serum (FBS), which improved cell viability substantially. The fluorescein diacetate (FDA) staining revealed that the entrapped cells were intact and viable immediately after the electrospinning process. A long-term cell viability assay using AlamarBlue® showed that cells were viable for over two weeks. Secreted Interleukin-8 (IL-8) was monitored as a candidate released protein, which can also act as an indicator of HUVEC stress. These results demonstrated that HUVECs could be entrapped within the electrospun scaffold with the potential of controllable cell deposition and the creation of a bioactive fibrous scaffold with extended functionality.


Subject(s)
Biocompatible Materials/chemistry , Human Umbilical Vein Endothelial Cells , Tissue Scaffolds/chemistry , Cell Survival/drug effects , Cells, Immobilized , Humans , Interleukin-8/metabolism , Polyesters/chemistry , Polyethylene Glycols/chemistry , Porosity , Solutions/chemistry , Tissue Engineering/methods
11.
ACS Nano ; 8(1): 419-29, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24392729

ABSTRACT

Therapeutic nanomedicine has concentrated mostly on anticancer therapy by making use of the nanosize for targeted therapy. Such nanocarriers are not expected to have sustained release of the bioactive molecule beyond a few days. There are other conditions where patients can benefit from sustained duration of action following a single instillation, but achieving this has been difficult in nanosized carriers. An important prerequisite for sustained delivery over several months is to have sufficiently high drug loading, without disruption or changes to the shape of the nanocarriers. Here we report on successful development of a drug-encapsulated nanocarrier for reducing intraocular pressure in a diseased nonhuman primate model and explain why it has been possible to achieve sustained action in vivo. The drug is a prostaglandin derivative, latanoprost, while the carrier is a nanosized unilamellar vesicle. The mechanistic details of this unique drug-nanocarrier combination were elucidated by isothermal titration calorimetry. We show, using Cryo-TEM and dynamic light scattering, that the spherical shape of the liposomes is conserved even at the highest loading of latanoprost and that specific molecular interactions between the drug and the lipid are the reasons behind improved stability and sustained release. The in vivo results clearly attest to sustained efficacy of lowering the intraocular pressure for 120 days, making this an excellent candidate to be the first truly sustained-release nanomedicine product. The mechanistic details we have uncovered should enable development of similar systems for other conditions where sustained release from nanocarriers is desired.


Subject(s)
Chemistry, Pharmaceutical , Drug Carriers , Glaucoma/drug therapy , Nanomedicine , Animals , Calorimetry , Delayed-Action Preparations , Macaca fascicularis
12.
ACS Appl Mater Interfaces ; 6(3): 1652-61, 2014 Feb 12.
Article in English | MEDLINE | ID: mdl-24405311

ABSTRACT

Engineered scaffold surface provides stem cells with vital cues that could determine the eventual fate of stem cells. In this work, biodegradable poly(L-lactide-co-ε-caprolactone) (PLCL) scaffold conjugated with Notch agonist-Jagged-1(JAG) peptide (2.1 kDa) was prepared to initiate myogenic differentiation of human mesenchymal stem cells (hMSCs). The scaffold surface was activated with oxygen plasma and acrylic acid was engrafted via UV polymerization to form a surface bearing carboxylic groups. JAG peptide was subsequently immobilized onto the carboxylated scaffold surface. Surface chemistry and topography were examined using attenuated total reflection Fourier transform infrared, X-ray photoelectron spectroscopy, and atomic force microscopy. Quantitative real time polymerase chain reaction analysis revealed activation of the Notch pathway; furthermore, several specific markers associated with myogenic but not osteogenic differentiation were shown to be up-regulated in hMSCs cultured on the engineered surface. The pro-myocardial effect of surface bound JAG peptide was further affirmed via immunodetection of the distinct myocardial marker, cardiac troponin T. Collectively, our results suggest that PLCL conjugated JAG peptide is a viable strategy to enhance the functional potential of scaffolds to be used as a bioengineered cardiac patch in myocardial infarction repair.


Subject(s)
Biocompatible Materials/pharmacology , Calcium-Binding Proteins/pharmacology , Cell Differentiation/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , Membrane Proteins/pharmacology , Mesenchymal Stem Cells/cytology , Muscle Development/drug effects , Receptors, Notch/agonists , Tissue Scaffolds/chemistry , Cell Differentiation/genetics , Cells, Cultured , Free Radicals/analysis , Gene Expression Regulation/drug effects , Humans , Jagged-1 Protein , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Microscopy, Atomic Force , Muscle Development/genetics , Peptides/pharmacology , Photoelectron Spectroscopy , Polyesters/pharmacology , Serrate-Jagged Proteins , Signal Transduction/drug effects , Spectroscopy, Fourier Transform Infrared , Surface Properties
13.
PLoS One ; 8(7): e68346, 2013.
Article in English | MEDLINE | ID: mdl-23861890

ABSTRACT

Cenderitide, also known as CD-NP, is a designer peptide developed by combining native mammalian c-type natriuretic peptide (CNP) and the C-terminus isolated from the dendroapis natriuretic peptide (DNP) of the venom from the green mamba. In early studies, intravenous and subcutaneous infusion of cenderitide was reported to reduce left ventricular (LV) mass and ameliorate cardiac remodelling. In this work, biodegradable polymeric films encapsulating CD-NP were developed and were investigated for their in vitro release and degradation characteristics. Subsequently, the bioactivity of released peptide and its effects on human cardiac fibroblast (HCF) were explored. We achieved sustained release from three films with low, intermediate and high release profiles for 30 days. Moreover, the bioactivity of released peptide was verified from the elevated production of cyclic guanosine monophospate (cGMP). The CD-NP released from films was able to inhibit the proliferation of hypertrophic HCF as well as suppress DNA synthesis in HCF. Furthermore, the sustained delivery from films showed comparable or superior suppressive actions on hypertrophic HCF compared to daily infusion of CD-NP. The results suggest that these films could be used to inhibit fibrosis and reduce cardiac remodelling via local delivery as cardiac patches.


Subject(s)
Cardiotonic Agents/pharmacology , Delayed-Action Preparations/chemistry , Fibroblasts/drug effects , Natriuretic Peptides/pharmacology , Polyesters/chemistry , Snake Venoms/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cyclic GMP/biosynthesis , Cyclic GMP/metabolism , DNA/antagonists & inhibitors , DNA/biosynthesis , Drug Compounding , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Kinetics , Methylene Chloride/chemistry , Transdermal Patch
14.
J Mater Sci Mater Med ; 24(8): 2013-27, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23670603

ABSTRACT

Mechanical properties of collagen films are less than ideal for biomaterial development towards musculoskeletal repair or cardiovascular applications. Herein, we present a collagen-cellulose composite film (CCCF) compared against swine small intestine submucosa in regards to mechanical properties, cell growth, and histological analysis. CCCF was additionally characterized by FE-SEM, NMR, mass spectrometry, and Raman Microscopy to elucidate its physical structure, collagen-cellulose composition, and structure activity relationships. Mechanical properties of the CCCF were tested in both wet and dry environments, with anisotropic stress-strain curves that mimicked soft-tissue. Mesenchymal stem cells, human umbilical vein endothelial cells, and human coronary artery smooth muscle cells were able to proliferate on the collagen films with specific cell orientation. Mesenchymal stem cells had a higher proliferation index and were able to infiltrate CCCF to a higher degree than small intestine submucosa. With the underlying biological properties, we present a collagen-cellulose composite film towards forthcoming biomaterial-related applications.


Subject(s)
Cellulose/chemistry , Collagen/chemistry , Connective Tissue , Membranes, Artificial , Mesenchymal Stem Cells/physiology , Animals , Biomimetic Materials/chemical synthesis , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cell Polarity/drug effects , Cell Polarity/physiology , Cells, Cultured , Cellulose/pharmacology , Collagen/pharmacology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/physiology , Humans , Materials Testing , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Nanofibers/chemistry , Tissue Engineering/instrumentation , Tissue Engineering/methods , Tissue Scaffolds/chemistry
15.
Nano Lett ; 13(4): 1616-21, 2013 Apr 10.
Article in English | MEDLINE | ID: mdl-23484520

ABSTRACT

We show that Rhodamine 6G (R6G), patterned by dip-pen nanolithography on graphene, can be used to locally n-dope it in a controlled fashion. In addition, we study the transport and assembly properties of R6G on graphene and show that in general the π-π stacking between the aromatic components of R6G and the underlying graphene drives the assembly of these molecules onto the underlying substrate. However, two distinct transport and assembly behaviors, dependent upon the presence or absence of R6G dimers, have been identified. In particular, at high concentrations of R6G on the tip, dimers are transferred to the substrate and form contiguous and stable lines, while at low concentrations, the R6G is transferred as monomers and forms patchy, unstable, and relatively ill-defined features. Finally, Kelvin probe force microscopy experiments show that the local electrostatic potential of the graphene changes as function of modification with R6G; this behavior is consistent with local molecular doping, highlighting a path for controlling the electronic properties of graphene with nanoscale resolution.


Subject(s)
Graphite/chemistry , Rhodamines/chemistry , Microscopy, Atomic Force , Spectrum Analysis, Raman , Static Electricity , Surface Properties
16.
Small ; 9(13): 2260-5, 2013 Jul 08.
Article in English | MEDLINE | ID: mdl-23362212

ABSTRACT

A gold nanotip array platform with a combination of ultrasensitive electrochemical sensing and spectroscopic monitoring capability is reported. Adenosine triphosphate is detected down to 1 pM according to the impedance changes in response to aptamer-specific binding. Furthermore, the local molecular information can be monitored at the individual plasmonic nanotips, and hence provide the capability for a better understanding of complex biological processes.


Subject(s)
Electrochemical Techniques/instrumentation , Gold/chemistry , Nanotechnology/instrumentation , Spectrum Analysis, Raman , Adenosine Triphosphate/analysis , DNA/chemistry , Dimethylpolysiloxanes/chemistry , Methylene Blue/chemistry
17.
Tissue Eng Part C Methods ; 19(8): 620-30, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23265414

ABSTRACT

The clinical success of tissue-engineered constructs commonly requires mechanical properties that closely mimic those of the human tissue. Determining the viscoelastic properties of such biomaterials and the factors governing their failure profiles, however, has proven challenging, although collecting extensive data regarding their tensile behavior is straightforward. The easily calculated Young's modulus remains the most reported mechanical measure, regardless of its limitations, even though single-relaxation-time (SRT) models can provide much more information, which remain scarce due to a lack of manageable tools for implementing these models. We developed an easy-to-use algorithm for applying the Zener SRT model and determining the elastic moduli, viscosity, and failure profiles of materials under different mechanical tests in a user-independent manner. The algorithm was validated on the data resulting from tensile tests on native and decellularized porcine cardiac tissue, previously suggested as a promising scaffold material for cardiac tissue engineering. This analysis yields new and more accurate measurements such as the elastic moduli and viscosity, the model's relaxation time, and information on the factors governing the materials' failure profiles. These measurements indicate that the viscoelasticity and strength of the decellularized acellular extracellular matrix (ECM) are similar to those of native tissue, although its elasticity and apparent viscosity are higher. Nonetheless, reseeding and culturing the ECM with mesenchymal stem cells was shown to partially restore the mechanical properties lost after decellularization. We propose this algorithm as a platform for soft-tissue analysis that can provide comparable and unbiased measures for characterizing viscoelastic biomaterials commonly used in tissue engineering.


Subject(s)
Elastic Modulus , Extracellular Matrix/chemistry , Models, Biological , Myocardium/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Humans , Swine , Viscosity
18.
Macromol Biosci ; 13(3): 299-310, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23233197

ABSTRACT

Direct laser machining and electrospinning are utilized to obtain a bi-layered hybrid scaffold with hierarchical topographical features to mimic extracellular matrix-like microenvironment of cells. Adult bone marrow derived human mesenchymal stem cells (hMSCs) are cultured in vitro in these hybrid scaffolds, and cell orientation, proliferation, viability, and differentiation are evaluated. The results show that this novel hybrid scaffold not only supports cell growth like traditional scaffolds, but also elicits positive responses from the cells, like lineage commitment and alignment, which are essential features of future scaffolds.


Subject(s)
Biomimetic Materials/chemical synthesis , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Polyesters/chemical synthesis , Tissue Engineering/methods , Adolescent , Biomarkers/metabolism , Biomimetic Materials/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Electrochemical Techniques , Extracellular Matrix/chemistry , Humans , Lasers , Male , Mesenchymal Stem Cells/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , Polyesters/pharmacology , Tissue Scaffolds
19.
Tissue Eng Part A ; 19(9-10): 1155-64, 2013 May.
Article in English | MEDLINE | ID: mdl-23216214

ABSTRACT

In most tissue engineering applications, understanding the factors affecting the growth dynamics of coculture systems is crucial for directing the population toward a desirable regenerative process. Yet, no comprehensive analysis method exists to quantify coculture population dynamics, let alone, a unifying model addressing the "environmental" factors influencing cell growth, all together. Here we suggest a modification of the Lotka-Volterra model to analyze the population dynamics of cocultured cells and predict their growth profiles for tissue engineering applications. This model, commonly used to describe the population dynamics of a prey and predator sharing a closed ecological niche, was found to fit our empirical data on cocultures of endothelial cells (ECs) and mesenchymal stem cells (MSCs) that have been widely investigated for their regenerative potential. Applying this model to cocultures of this sort allows us to quantify the effect that culturing conditions have on the way cell growth is affected by the same cells or by the other cells in the coculture. We found that in most cases, EC growth was inhibited by the same cells but promoted by MSCs. The principles resulting from this analysis can be used in various applications to guide the population toward a desired direction while shedding new light on the fundamental interactions between ECs and MSCs. Similar results were also demonstrated on complex substrates made from decellularized porcine cardiac extracellular matrix, where growth occurred only after coculturing ECs and MSCs together. Finally, this unique implementation of the Lotka-Volterra model may also be regarded as a roadmap for using such models with other potentially regenerative cocultures in various applications.


Subject(s)
Endothelial Cells/cytology , Mesenchymal Stem Cells/cytology , Models, Theoretical , Cell Survival , Cells, Cultured , Humans , Tissue Engineering
20.
Adv Healthc Mater ; 1(4): 513-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-23184785

ABSTRACT

Recent interest in the use of human hair keratins as a biomaterial has grown, fuelled by improvements in keratin extraction methods and better understanding of keratin bioactivity. The use of keratins as a bioactive coating for in vitro cell culture studies is an attractive proposition. In this light, the surface adsorption of human hair keratins onto tissue culture polystyrene surfaces has been investigated. Keratin density, nano-topography and hydrophobicity of keratin coated surfaces were characterized. To understand the cellular influence of these coated surfaces, murine L929 fibroblasts were cultured on them and evaluated for cytotoxicity, proliferation, metabolic activity and detachment behaviors compared to collagen type 1 coated surfaces. Keratins were deposited up to a density of 650 ng/cm(2) when a coating concentration of 80 µg/ml or higher was used. The surface features formed by adsorbed keratins also changed in a coating concentration dependent manner. These surfaces improved L929 mouse fibroblast adhesion and proliferation in comparison to uncoated and collagen type 1 coated tissue culture polystyrene. Furthermore, the expression of fibronectin was accelerated on surfaces coated with solutions of higher keratin concentrations. These results suggest that human hair keratins can be used as a viable surface coating material to enhance substrate compliance for culturing cells.


Subject(s)
Fibroblasts/drug effects , Fibroblasts/physiology , Hair/chemistry , Keratins/chemistry , Keratins/pharmacology , Keratins/pharmacokinetics , Nanostructures/chemistry , Adsorption , Animals , Binding Sites , Cell Line , Fibroblasts/ultrastructure , Humans , Mice , Protein Binding , Surface Properties/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...