Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Front Bioeng Biotechnol ; 12: 1410863, 2024.
Article in English | MEDLINE | ID: mdl-38903186

ABSTRACT

Tissue-engineered vascular grafts (TEVGs) poised for regenerative applications are central to effective vascular repair, with their efficacy being significantly influenced by scaffold architecture and the strategic distribution of bioactive molecules either embedded within the scaffold or elicited from responsive tissues. Despite substantial advancements over recent decades, a thorough understanding of the critical cellular dynamics for clinical success remains to be fully elucidated. Graft failure, often ascribed to thrombogenesis, intimal hyperplasia, or calcification, is predominantly linked to improperly modulated inflammatory reactions. The orchestrated behavior of repopulating cells is crucial for both initial endothelialization and the subsequent differentiation of vascular wall stem cells into functional phenotypes. This necessitates the TEVG to provide an optimal milieu wherein immune cells can promote early angiogenesis and cell recruitment, all while averting persistent inflammation. In this study, we present an innovative TEVG designed to enhance cellular responses by integrating a physicochemical gradient through a multilayered structure utilizing synthetic (poly (ester urethane urea), PEUU) and natural polymers (Gelatin B), thereby modulating inflammatory reactions. The luminal surface is functionalized with a four-arm polyethylene glycol (P4A) to mitigate thrombogenesis, while the incorporation of adhesive peptides (RGD/SV) fosters the adhesion and maturation of functional endothelial cells. The resultant multilayered TEVG, with a diameter of 3.0 cm and a length of 11 cm, exhibits differential porosity along its layers and mechanical properties commensurate with those of native porcine carotid arteries. Analyses indicate high biocompatibility and low thrombogenicity while enabling luminal endothelialization and functional phenotypic behavior, thus limiting inflammation in in-vitro models. The vascular wall demonstrated low immunogenicity with an initial acute inflammatory phase, transitioning towards a pro-regenerative M2 macrophage-predominant phase. These findings underscore the potential of the designed TEVG in inducing favorable immunomodulatory and pro-regenerative environments, thus holding promise for future clinical applications in vascular tissue engineering.

2.
Front Bioeng Biotechnol ; 11: 1257778, 2023.
Article in English | MEDLINE | ID: mdl-37799814

ABSTRACT

Introduction: Thrombogenesis, a major cause of implantable cardiovascular device failure, can be addressed through the use of biodegradable polymers modified with anticoagulating moieties. This study introduces a novel polyester urethane urea (PEUU) functionalized with various anti-platelet deposition molecules for enhanced antiplatelet performance in regenerative cardiovascular devices. Methods: PEUU, synthesized from poly-caprolactone, 1,4-diisocyanatobutane, and putrescine, was chemically oxidized to introduce carboxyl groups, creating PEUU-COOH. This polymer was functionalized in situ with polyethyleneimine, 4-arm polyethylene glycol, seleno-L-cystine, heparin sodium, and fondaparinux. Functionalization was confirmed using Fourier-transformed infrared spectroscopy and X-ray photoelectron spectroscopy. Bio-compatibility and hemocompatibility were validated through metabolic activity and hemolysis assays. The anti-thrombotic activity was assessed using platelet aggregation, lactate dehydrogenase activation assays, and scanning electron microscopy surface imaging. The whole-blood clotting time quantification assay was employed to evaluate anticoagulation properties. Results: Results demonstrated high biocompatibility and hemocompatibility, with the most potent anti-thrombotic activity observed on pegylated surfaces. However, seleno-L-cystine and fondaparinux exhibited no anti-platelet activity. Discussion: The findings highlight the importance of balancing various factors and addressing challenges associated with different approaches when developing innovative surface modifications for cardiovascular devices.

3.
Polymers (Basel) ; 14(11)2022 May 24.
Article in English | MEDLINE | ID: mdl-35683808

ABSTRACT

Fabrication of scaffolds with hierarchical structures exhibiting the blood vessel topological and biochemical features of the native extracellular matrix that maintain long-term patency remains a major challenge. Within this context, scaffold assembly using biodegradable synthetic polymers (BSPs) via electrospinning had led to soft-tissue-resembling microstructures that allow cell infiltration. However, BSPs fail to exhibit the sufficient surface reactivity, limiting protein adsorption and/or cell adhesion and jeopardizing the overall graft performance. Here, we present a methodology for the fabrication of three-layered polycaprolactone (PCL)-based tubular structures with biochemical cues to improve protein adsorption and cell adhesion. For this purpose, PCL was backbone-oxidized (O-PCL) and cast over a photolithography-manufactured microgrooved mold to obtain a bioactive surface as demonstrated using a protein adsorption assay (BSA), Fourier transform infrared spectroscopy (FTIR) and calorimetric analyses. Then, two layers of PCL:gelatin (75:25 and 95:5 w/w), obtained using a novel single-desolvation method, were electrospun over the casted O-PCL to mimic a vascular wall with a physicochemical gradient to guide cell adhesion. Furthermore, tensile properties were shown to withstand the physiological mechanical stresses and strains. In vitro characterization, using L929 mouse fibroblasts, demonstrated that the multilayered scaffold is a suitable platform for cell infiltration and proliferation from the innermost to the outermost layer as is needed for vascular wall regeneration. Our work holds promise as a strategy for the low-cost manufacture of next-generation polymer-based hierarchical scaffolds with high bioactivity and resemblance of ECM's microstructure to accurately guide cell attachment and proliferation.

4.
Cells ; 11(6)2022 03 10.
Article in English | MEDLINE | ID: mdl-35326390

ABSTRACT

Tissue-engineered vascular grafts (TEVGs) are a promising alternative to treat vascular disease under complex hemodynamic conditions. However, despite efforts from the tissue engineering and regenerative medicine fields, the interactions between the material and the biological and hemodynamic environment are still to be understood, and optimization of the rational design of vascular grafts is an open challenge. This is of special importance as TEVGs not only have to overcome the surgical requirements upon implantation, they also need to withhold the inflammatory response and sustain remodeling of the tissue. This work aims to analyze and evaluate the bio-molecular interactions and hemodynamic phenomena between blood components, cells and materials that have been reported to be related to the failure of the TEVGs during the regeneration process once the initial stages of preimplantation have been resolved, in order to tailor and refine the needed criteria for the optimal design of TEVGs.


Subject(s)
Blood Vessel Prosthesis Implantation , Blood Vessel Prosthesis , Tissue Engineering
5.
Int J Mol Sci ; 23(3)2022 Jan 20.
Article in English | MEDLINE | ID: mdl-35163056

ABSTRACT

Currently available small diameter vascular grafts (<6 mm) present several long-term limitations, which has prevented their full clinical implementation. Computational modeling and simulation emerge as tools to study and optimize the rational design of small diameter tissue engineered vascular grafts (TEVG). This study aims to model the correlation between mechanical-hemodynamic-biochemical variables on protein adsorption over TEVG and their regenerative potential. To understand mechanical-hemodynamic variables, two-way Fluid-Structure Interaction (FSI) computational models of novel TEVGs were developed in ANSYS Fluent 2019R3® and ANSYS Transient Structural® software. Experimental pulsatile pressure was included as an UDF into the models. TEVG mechanical properties were obtained from tensile strength tests, under the ISO7198:2016, for novel TEVGs. Subsequently, a kinetic model, linked to previously obtained velocity profiles, of the protein-surface interaction between albumin and fibrinogen, and the intima layer of the TEVGs, was implemented in COMSOL Multiphysics 5.3®. TEVG wall properties appear critical to understand flow and protein adsorption under hemodynamic stimuli. In addition, the kinetic model under flow conditions revealed that size and concentration are the main parameters to trigger protein adsorption on TEVGs. The computational models provide a robust platform to study multiparametrically the performance of TEVGs in terms of protein adsorption and their regenerative potential.


Subject(s)
Blood Vessel Prosthesis , Extracellular Matrix/metabolism , Adsorption , Animals , Computer Simulation , Hemodynamics , Models, Anatomic , Models, Theoretical , Tensile Strength
6.
Cells ; 10(11)2021 11 12.
Article in English | MEDLINE | ID: mdl-34831361

ABSTRACT

Vascular grafts (VG) are medical devices intended to replace the function of a diseased vessel. Current approaches use non-biodegradable materials that struggle to maintain patency under complex hemodynamic conditions. Even with the current advances in tissue engineering and regenerative medicine with the tissue engineered vascular grafts (TEVGs), the cellular response is not yet close to mimicking the biological function of native vessels, and the understanding of the interactions between cells from the blood and the vascular wall with the material in operative conditions is much needed. These interactions change over time after the implantation of the graft. Here we aim to analyze the current knowledge in bio-molecular interactions between blood components, cells and materials that lead either to an early failure or to the stabilization of the vascular graft before the wall regeneration begins.


Subject(s)
Blood Vessel Prosthesis , Blood/immunology , Immunity , Animals , Biocompatible Materials/pharmacology , Blood Coagulation/drug effects , Humans , Treatment Failure
7.
Polymers (Basel) ; 11(3)2019 Mar 26.
Article in English | MEDLINE | ID: mdl-30960553

ABSTRACT

Decellularized extracellular matrices (dECMs) represent a promising alternative as a source of materials to develop scaffolds that closely mimic the native environment of cells. As a result, dECMs have attracted significant attention for their applications in regenerative medicine and tissue engineering. One such application is 3D bioprinting, in which dECMs can be used to prepare bioinks with the biomimicry attributes required for regeneration purposes. Formulating bioinks is, however, challenging, due to difficulties in assuring that the printed materials match the mechanical properties of the tissue which is to be regenerated. To tackle this issue, a number of strategies have been devised, including crosslinking methods, the addition of synthetic materials as excipients, and the use of synthetic matrices for casting. We are particularly interested in extrusion-based 3D bioprinting, mainly due to the ease of rapidly conducting tests for adjusting operating conditions such that the required rheological and mechanical properties are met when using it. Here, we propose a novel bioink that consists of an acid-based precipitation of a small intestinal submucosa (SIS) dECM. The formulated bioink also relies on photocrosslinking reactions to attempt to control gelation and ultimately the mechanical properties of the extruded material. Photoinitiation was explored with the aid of varying concentrations of riboflavin (RF). Manual extrusion and rheological flow tests confirmed the printability and shear-thinning behavior of all formulations. Photocrosslinking reactions, however, failed to promote a substantial increase in gelation, which was attributed to considerable entanglement of undigested collagen molecules. As a result, pendant amine groups thought to be involved in the photo-mediated reactions remain largely inaccessible. In silico computational fluid dynamics (CFD) simulations were implemented to determine shear stress values on the bioink along the exit of the printing nozzle. Moreover, we calculated a stability parameter as a means to estimate changes in the bioink stability during the extrusion process. Future studies should be directed toward assessing the role of temperature-induced gelation in the rheological properties of the bioink and the development of strategies to improve the efficiency of photocrosslinking processes.

8.
ACS Appl Bio Mater ; 2(9): 3706-3721, 2019 Sep 16.
Article in English | MEDLINE | ID: mdl-35021344

ABSTRACT

Vascular grafts are used as vascular access for hemodialysis, the most common renal replacement therapy to artificially clean blood waste after kidney malfunction. Despite that they are widely used in clinical practice, upon implantation, synthetic vasculars show complications such as thrombogenesis, reduced patency rates, low blood pressure, or even complete collapse. In this study, a C-shaped vascular graft was manufactured with small intestinal submucosa (SIS) and modified on the surface and the bulk of the material via conjugation of polyethylene glycol (PEG) to obtain a biocompatible and less thrombogenic vascular graft than the commercially available polytetrafluoroethylene (ePTFE) vascular grafts. Molecular weight and concentration of PEG molecules were systematically varied to gain insights into the underlying structure-function relationships. We analyzed the chemical, thermal, and mechanical properties of vascular grafts modified with 6 equiv of SIS-PEG 400 as well as cytotoxicity and in vitro platelet deposition. Immune response, patency rates, and extent of regeneration were also tested in vivo with the aid of swine animal models. Results showed that the conjugation levels achieved were sufficient to improve graft compliance, therefore approaching that of native vessels, while platelet deposition was altered leading to a 95% reduction compared with pristine SIS and 92% with respect to ePTFE. H&E staining on explanted samples corroborated SIS-PEG 400 biocompatibility and the ability to promote regeneration. The obtained results set solid foundations for the rational design and manufacture of a regenerative, small diameter vascular graft model and introduce an alternative to ePTFE vascular grafts for hemodialysis access.

9.
ASAIO J ; 64(2): 270-277, 2018.
Article in English | MEDLINE | ID: mdl-29470246

ABSTRACT

Synthetic vascular access for hemodialysis exhibits biological and mechanical material properties mismatch with the native vessels. These limitations prevent infiltration of endothelial cells and decrease grafts long-term patency, particularly in small diameter vessels. We aimed to design a curved structural reinforced small intestinal submucosa (SIS) vascular graft for hemodialysis access and to evaluate in a porcine animal model graft patency by Doppler ultrasonography, tissue remodeling by histology, and vascular wall Young's modulus after implantation by biaxial tensile test. Curved 4 mm inner diameter, 0.5 mm thickness, and 150 mm length SIS grafts were designed. Small intestinal submucosa vascular grafts were preliminary tested in vivo in a porcine animal model (n=3) constructing an arteriovenous fistula between the carotid artery and the jugular vein; GORE-TEX grafts were implanted as control. Small intestinal submucosa grafts remained patent 46 ± 7 days against the control, 30 ± 3 days. Histology showed thrombus formation on the lumen (80% to 100% surface area) of all explanted grafts. Small intestinal submucosa grafts exhibited neovascularization and endothelial cells alignment on the graft wall, indicating regeneration. Biaxial tensile tests demonstrated no significant differences in Young's moduli between SIS grafts (ECirc = 2.5 ± 1.0 MPa, ELong = 5.7 ± 2.6 MPa) and native artery (ECirc = 1.4 ± 0.8 MPa, ELong = 5.5 ± 1.1 MPa), indicating similar wall stiffness. This study proposes an innovative design of a tissue-engineered vascular graft for hemodialysis access that, besides its structural characteristics similar to those of current synthetic grafts, could enhance biological performance because of its composition.


Subject(s)
Blood Vessel Prosthesis , Renal Dialysis/instrumentation , Animals , Intestinal Mucosa/transplantation , Models, Animal , Swine
10.
J Tissue Eng Regen Med ; 11(5): 1427-1434, 2017 05.
Article in English | MEDLINE | ID: mdl-26220892

ABSTRACT

In small intestinal submucosa (SIS) scaffolds for functional tissue engineering, the impact of scaffold fabrication parameters on cellular response and tissue regeneration may relate to the mechanotransductory properties of the final arrangement of collagen fibres. We previously proved that two fabrication parameters, (a) preservation (P) or removal (R) of a dense collagen layer present in SIS, and (b) SIS in a final dehydrated (D) or hydrated (H) state, have an effect on the micromechanical environment of SIS. In a continuation of our studies, we herein hypothesized that these fabrication parameters also modulate early mechanotransduction in cells populating the scaffold. Mechanotransduction was investigated by seeding human umbilical vein endothelial cells (HUVECs) on scaffolds, exposing them to pulsatile shear stress (12 ± 4 dyne/cm2 ) for 1 h (n = 5) in a cone-and-plate shear system, and evaluating the expression of the mechanosensitive genes Pecam1 and Enos by immunofluorescence and qPCR. Expression of mechanosensitive genes was highest in PD grafts, followed by PH and RH grafts. The RD group had similar expression to that of unsheared control cells, suggesting that the RD combination potentially reduced mechanotransduction of shear to cells. We concluded that the two fabrication parameters studied, which modify SIS micromechanics, also potentially modulated the early shear-induced expression of mechanosensitive genes in seeded HUVECs. Our findings suggest that fabrication parameters influence the outcome of SIS as a therapeutic scaffold. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Mechanotransduction, Cellular , Nitric Oxide Synthase Type III/biosynthesis , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Stress, Mechanical , Tissue Scaffolds/chemistry , Animals , Intestinal Mucosa/cytology , Intestine, Small/cytology , Shear Strength , Swine , Tissue Engineering
11.
ASAIO J ; 61(5): 596-604, 2015.
Article in English | MEDLINE | ID: mdl-26098175

ABSTRACT

Small intestinal submucosa grafts for vascular regeneration have produced variable patency (0-100%) that has been concurrent with variability in fabrication techniques. We hypothesized that 1) preservation (P) or removal (R) of the stratum compactum layer of the intestine and 2) a dehydrated (D) or hydrated (H) state of the graft, affect early patency and tissue regeneration. We combined both parameters through a 2(2) factorial experimental design into four groups (PD, RD, PH, RH), and compared them in an in vivo early response predictive model (swine, ID 4.5 mm, 7d, n = 4). Patency, thrombogenicity, vascularization, fibroblast infiltration, macrophage polarization profile, endothelialization, and biaxial mechanics were assessed. PD grafts remained patent (4/4) but had scarce vascularization and fibroblast infiltration. RD and RH had extensive vascularization and fibroblast infiltration, however, RD had sustained patency (4/4) and the highest number of regeneration-associated phenotype macrophages (M2), whereas RH had lower patency (3/4) and less M2 macrophages. PH had a modest cellular infiltration, but the lowest patency (2/4) and a dominant adverse macrophage phenotype. Elasticity of R grafts evolved toward that of native carotids (particularly RD), while P grafts kept their initial stiffness. We concluded that fabrication parameters drastically affected early patency and regeneration, with RD providing the best results.


Subject(s)
Blood Vessel Prosthesis , Blood Vessels/physiology , Carotid Arteries/physiology , Jejunum/physiology , Regeneration , Vascular Patency , Animals , Bioprosthesis , Carotid Arteries/surgery , Graft Occlusion, Vascular , Intestinal Mucosa/physiology , Intestinal Mucosa/transplantation , Jejunum/transplantation , Models, Animal , Swine
12.
J Biomech ; 47(11): 2766-73, 2014 Aug 22.
Article in English | MEDLINE | ID: mdl-24877881

ABSTRACT

In small intestinal submucosa scaffolds for functional tissue engineering, the impact of scaffold fabrication parameters on success rate may be related to the mechanotransductory properties of the final microstructural organization of collagen fibers. We hypothesized that two fabrication parameters, 1) preservation (P) or removal (R) of a dense collagen layer present in SIS and 2) SIS in a final dehydrated (D) or hydrated (H) state, have an effect on scaffold void area, microstructural anisotropy (fiber alignment) and mechanical anisotropy (global mechanical compliance). We further integrated our experimental measurements in a constitutive model to explore final effects on the micromechanical environment inside the scaffold volume. Our results indicated that PH scaffolds might exhibit recurrent and large force fluctuations between layers (up to 195 pN), while fluctuations in RH scaffolds might be larger (up to 256 pN) but not as recurrent. In contrast, both PD and RD groups were estimated to produce scarcer and smaller fluctuations (not larger than 50 pN). We concluded that the hydration parameter strongly affects the micromechanics of SIS and that an adequate choice of fabrication parameters, assisted by the herein developed method, might leverage the use of SIS for functional tissue engineering applications, where forces at the cellular level are of concern in the guidance of new tissue formation.


Subject(s)
Intestinal Mucosa/physiology , Tissue Engineering/methods , Tissue Scaffolds , Animals , Anisotropy , Collagen/chemistry , Extracellular Matrix/chemistry , Intestine, Small/physiology , Stress, Mechanical , Swine
13.
Biotechnol Prog ; 29(6): 1565-72, 2013.
Article in English | MEDLINE | ID: mdl-23966236

ABSTRACT

Perfluorocarbon (PFC) emulsions can transport and release various gases based on concentration gradients. The objective of this study was to determine the possibility of carrying and delivering exogenous nitric oxide (NO) into the circulation by simply loading PFC emulsion with NO prior infusion. PFC was equilibrated with room air (PFC) or 300 ppm NO (PFC-NO) at atmospheric pressure. Isotonic saline solution was used as a volume control (Saline). PFC and PFC-NO were infused at a dose of 3.5 mL/kg in the hamster window chamber model. Blood chemistry, and systemic and microvascular hemodynamic response were measured. Infusion of PFC preloaded with NO reduced blood pressure, induced microvascular vasodilation and increased capillary perfusion; although these changes lasted less than 30 min post infusion. On the other hand, infusion of PFC (without NO) produced vasoconstriction; however, the vasoconstriction was followed by vasodilatation at 30 min post infusion. Plasma nitrite and nitrate increased 15 min after infusion of NO preloaded PFC compared with PFC, 60 min after infusion nitrite and nitrate were not different, and 90 min after infusion plasma S-nitrosothiols increased in both groups. Infusion of NO preloaded PFC resulted in acute vascular relaxation, where as infusion of PFC (without NO) produced vasoconstriction, potentially due to NO sequestration by the PFC micelles. The late effects of PFC infusion are due to NO redistribution and plasma S-nitrosothiols. Gas solubility in PFC can provide a tool to modulate plasma vasoactive NO forms availability and improve microcirculatory function and promote increased blood flow.


Subject(s)
Blood Pressure/drug effects , Fluorocarbons/administration & dosage , Nitric Oxide/administration & dosage , Vasoconstriction/drug effects , Animals , Cricetinae , Emulsions/administration & dosage , Fluorocarbons/chemistry , Isotonic Solutions , Mesocricetus , Microcirculation/drug effects , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Vasodilation/drug effects
14.
Biotechnol Prog ; 29(3): 796-807, 2013.
Article in English | MEDLINE | ID: mdl-23606592

ABSTRACT

Perfluorocarbon (PFC) emulsions used as artificial oxygen carriers lack colloid osmotic pressure (COP) and must be administered with colloid-based plasma expanders (PEs). Although PFC emulsions have been widely studied, there is limited information about PFC emulsion interaction with PEs and blood. Their interaction forms aggregates due to electrostatic and rheological phenomena, and change blood rheology and blood flow. This study analyzes the effects of the interaction between PFC emulsions with blood in the presence of clinically-used PEs. The rheological behavior of the mixtures was analyzed in vitro in parallel with in vivo analysis of blood flow in the microcirculation using intravital microscopy, when PEs were administered in a clinically relevant scenario. The interaction between the PFC emulsion and PE with blood produced PFC droplets and red blood cell (RBCs) aggregation and increased blood viscosity in a shear dependent fashion. The PFC droplets formed aggregates when mixed with PEs containing electrolytes, and the aggregation increased with the electrolyte concentration. Mixtures of PFC with PEs that produced PFC aggregates also induced RCBs aggregation when mixed with blood, increasing blood viscosity at low shear rates. The more viscous suspension at low shear rates produced a blunted blood flow velocity profile in vivo compared to nonaggregating mixtures of PFC and PEs. For the PEs evaluated, human serum albumin produced minimal to undetectable aggregation. PFC and PEs interaction with blood can affect sections of the microcirculation with low shear rates (e.g., arterioles, venules, and pulmonary circulation) when used in a clinical setting, because persistent aggregates could cause capillary occlusion, decreased perfusion, pulmonary emboli or focal ischemia.


Subject(s)
Blood Viscosity/drug effects , Colloids/pharmacology , Fluorocarbons/pharmacology , Plasma Substitutes/pharmacology , Animals , Blood Pressure/drug effects , Colloids/chemistry , Colloids/metabolism , Drug Interactions , Erythrocyte Aggregation/drug effects , Fluorocarbons/chemistry , Fluorocarbons/metabolism , Oxygen/metabolism , Plasma Substitutes/chemistry , Plasma Substitutes/metabolism , Rats , Rats, Sprague-Dawley
15.
ASAIO J ; 53(3): 257-62, 2007.
Article in English | MEDLINE | ID: mdl-17515713

ABSTRACT

Our objective was to study the effect of glutaraldehyde (GLU) concentration, heat, and photooxidation on mechanical properties and calcification of bovine pericardium grafts in an in vivo model. Fresh pericardia were treated as follows: 0.625% GLU for 7 days (standard); 0.625%, 1%, and 3% GLU at 4 degrees C for 20 days and 50 degrees C for additional 20 days; irradiation in cross-linking medium with metilene blue at 0 degrees C for 8 hours. Tissues were subjected to tensile mechanical tests (n = 76). Fixed patches were subcutaneously implanted in mice for 50 days (n = 16 per treatment). Calcification was assessed by atomic absorption spectrophotometry (n = 55) and von Kossa staining (n = 28). Analysis of variance and Tukey's test were used for statistical analysis. The 3% GLU and 3% GLU + heat treatments showed an enhancement of the mechanical properties above standard treatment. No significant difference was found in calcification between treatments. The 3% GLU treatment enhances the mechanical properties of the tissue above standard treatment without increasing calcification and without applying heat; therefore it is recommended for high-strength applications. Supplementary treatments to decrease calcification could be combined with this methodology to obtain a high-strength-low-calcification biomaterial for manufacturing of long-term cardiovascular grafts.


Subject(s)
Blood Vessel Prosthesis , Calcinosis/prevention & control , Fixatives , Glutaral , Pericardium/transplantation , Animals , Calcinosis/pathology , Cattle , Foreign-Body Reaction/pathology , Hot Temperature , Materials Testing , Mice , Oxidation-Reduction , Pericardium/physiology , Spectrophotometry , Subcutaneous Tissue , Tensile Strength
16.
Rev. colomb. radiol ; 17(3): 1972-1981, sept. 2006. ilus, tab, graf
Article in Spanish | LILACS | ID: lil-521541

ABSTRACT

Objetivos: Es necesario generar criterios de diagnóstico más sensibles para enfermedad carotídea; esto, basados en el riesgo que presentan, por un lado, el desprendimiento de placa y, por el otro, la activación y agregación plaquetaria, tema de este trabajo. Nuestro objetivo es encontrar un indicador de activación plaquetaria (LA) que permita evaluar el riesgo de evento cerebrovascular (ECV) asociado con activación y agregación en presencia de estenosis de la arteria carótida interna. Metodología: Se estudia la dinámica de la sangre en geometrías idealizadas de la bifurcación de la arteria carótida con modelos computacionales (MC). Se plantea la metodología experimental para estudiar activación plaquetaria debido a esfuerzos cortantes. Resultados: Se implementó un código que encuentra las líneas de trayectoria en flujo 3D y estado transiente. Los valores de pico sistólico de velocidad (PSV), justo en la estenosis, obtenidos con el MC son mayores que los medidos con eco Doppler y reportados en literatura, con lo que se hace evidente la necesidad de implementar modelos más reales. Conclusiones: Conocer la relación existente entre activación plaquetaria, magnitud de los esfuerzos cortantes y tiempo durante el que éstas residen en este régimen de esfuerzos será de especial interés, pues permitirá encontrar un LA sensible a cambios de la hemodinámica y al porcentaje de reducción de lumen que sirva para evaluar el riesgo de ECV debido a activación plaquetaria.


Subject(s)
Humans , Arteriosclerosis , Blood Platelets , Carotid Artery Diseases , Carotid Artery, Internal , Carotid Stenosis
17.
Am J Physiol Heart Circ Physiol ; 287(1): H320-30, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15210452

ABSTRACT

The oxygen transport capacity of fluorocarbons was investigated in the hamster chamber window model microcirculation to determine the rate at which oxygen is delivered to the tissue in conditions of extreme hemodilution [hematocrit (Hct) 11%]. Hydroxyethlyl starch (HES 200; 200 kDa molecular mass) was used as a plasma expander for two isovolemic hemodilutions performed with 10% HES 200 until a Hct of 65%. A third step reduced the Hct to 75% of baseline and was performed with either HES 200 or a 60% perfluorocarbon (PFC) emulsion. Comparisons of HES 200-only-hemodiluted animals versus 4.2 g/kg PFC emulsion-hemodiluted animals were made at 21% and 100% normobaric oxygen ventilation. It was found that systemic and microvascular oxygen delivery was 25% and 400% higher in the PFC animals compared with HES 200 animals, respectively, showing that PFCs deliver oxygen to the tissue when combined with hyperoxic ventilation in the present experiments, with no evidence of vasoconstriction or impaired microvascular function. Oxygen ventilation (100%) led to a positive base excess for the PFC group (5.5 +/- 2.5 mmol/l) versus a negative balance (-0.8 +/- 1.4 mmol/l) for the HES 200 group, suggesting that microvascular findings corresponded to systemic events.


Subject(s)
Fluorocarbons/pharmacology , Hemodilution , Oxygen Consumption , Oxygen/blood , Animals , Biological Availability , Blood Flow Velocity , Capillaries/anatomy & histology , Cardiac Output , Cricetinae , Erythrocytes/physiology , Gases/blood , Hemodilution/methods , Hemodynamics , Hydroxyethyl Starch Derivatives/pharmacology , Mesocricetus , Microcirculation , Oxygen/metabolism , Plasma Substitutes/pharmacology
18.
Am J Physiol Heart Circ Physiol ; 286(3): H878-83, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14630627

ABSTRACT

Oxygen phosphorescence quenching was used to measure tissue Po(2) of lymphatic vessels of 43.6 +/- 23.1 microm (mean +/- SD) diameter in tissue locations of the rat mesentery classified according to anatomic location. Lymph and adipose tissue Po(2) were 20.6 +/- 9.1 and 34.1 +/- 7.8 mmHg, respectively, with the difference being statistically significant. Rare microlymphatic vessels in connective tissue not surrounded by microvessels had a Po(2) of 0.8 +/- 0.2 mmHg, whereas the surrounding tissue Po(2) was 3.0 +/- 3.2 mmHg, with both values being significantly lower than those of adipose tissue. Lower of lymph fluid Po(2) relative to the surrounding tissue was also evident in paired measurements of Po(2) in the lymphatic vessels and perilymphatic adipose tissue, which was significantly lower than the Po(2) in paired adipose tissue. The Po(2) of the lymphatic fluid of the mesenteric microlymphatics is consistently lower than that of the surrounding adipose tissue by approximately 11 mmHg; therefore, lymph fluid has the lowest Po(2) of this tissue. The disparity between lymph and tissue Po(2) is attributed to the microlymphatic vessel wall and lymphocyte oxygen consumption.


Subject(s)
Lymph/metabolism , Lymphatic System/metabolism , Oxygen Consumption/physiology , Adipose Tissue/metabolism , Animals , Luminescent Measurements , Mesentery/metabolism , Microscopy/methods , Oxygen/metabolism , Rats , Rats, Sprague-Dawley
19.
Rev. Cuerpo Méd ; 14(2): 24-6, 1994. ilus
Article in Spanish | LILACS | ID: lil-176194

ABSTRACT

Esclerosis múltiple es una enfermedad infrecuente en latitudes similares al Perú, su inicio es raro en menores de 10 años y la presentación en crisis convulsivas es excepcional. Se describe un caso iniciado a los 8 años y se revisa la literatura al respecto.


Subject(s)
Multiple Sclerosis/diagnosis , Multiple Sclerosis/urine , Multiple Sclerosis/blood
20.
Rev. Cuerpo Méd ; 14(1): 17-8, 1992.
Article in Spanish | LILACS | ID: lil-176178

ABSTRACT

El reporte trata de una niña de 4 años con retraso mental y convulsiones, quien estuvo recibiendo durante 2 meses 130 mg/kg valproato de sodio ocasionándole anemia, plaquetopenia, leucopenia e hipoproteinemia. Al cabo de 2 semanas se disminuyó el valproato a 30 mg/kg y se observó la normalización de las alteraciones hematológicas presentadas.


Subject(s)
Humans , Female , Child, Preschool , Valproic Acid/adverse effects , Valproic Acid/therapeutic use , Hematologic Diseases/etiology , Pancytopenia/diagnosis , Pancytopenia/etiology , Rebound Effect
SELECTION OF CITATIONS
SEARCH DETAIL
...