Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Blood ; 138(22): 2173-2184, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34086880

RESUMEN

End-stage renal disease (ESRD) patients on chronic hemodialysis have repeated blood exposure to artificial surfaces that can trigger clot formation within the hemodialysis circuit. Dialyzer clotting can lead to anemia despite erythropoietin and iron supplementation. Unfractionated heparin prevents clotting during hemodialysis, but it is not tolerated by all patients. Although heparin-free dialysis is performed, intradialytic blood entrapment can be problematic. To address this issue, we performed a randomized, double-blind, phase 2 study comparing AB023, a unique antibody that binds factor XI (FXI) and blocks its activation by activated FXII, but not by thrombin, to placebo in 24 patients with ESRD undergoing heparin-free hemodialysis. Patients were randomized to receive a single predialysis dose of AB023 (0.25 or 0.5 mg/kg) or placebo in a 2:1 ratio, and safety and preliminary efficacy were compared with placebo and observations made prior to dosing within each treatment arm. AB023 administration was not associated with impaired hemostasis or other drug-related adverse events. Occlusive events requiring hemodialysis circuit exchange were less frequent and levels of thrombin-antithrombin complexes and C-reactive protein were lower after AB023 administration compared with data collected prior to dosing. AB023 also reduced potassium and iron entrapment in the dialyzers, consistent with less blood accumulation within the dialyzers. We conclude that despite the small sample size, inhibition of contact activation-induced coagulation with AB023 was well tolerated and reduced clotting within the dialyzer. This trial was registered at www.clinicaltrials.gov as #NCT03612856.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antitrombinas/uso terapéutico , Fallo Renal Crónico/terapia , Diálisis Renal/métodos , Adulto , Anticuerpos Monoclonales Humanizados/efectos adversos , Antitrombinas/efectos adversos , Método Doble Ciego , Factor XI/antagonistas & inhibidores , Femenino , Hemostasis/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Efecto Placebo , Diálisis Renal/efectos adversos , Trombosis/etiología , Trombosis/prevención & control
2.
Blood ; 135(9): 689-699, 2020 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-31977000

RESUMEN

Although thrombin is a key enzyme in the coagulation cascade and is required for both normal hemostasis and pathologic thrombogenesis, it also participates in its own negative feedback via activation of protein C, which downregulates thrombin generation by enzymatically inactivating factors Va and VIIIa. Our group and others have previously shown that thrombin's procoagulant and anticoagulant activities can be effectively disassociated to varying extents through site-directed mutagenesis. The thrombin mutant W215A/E217A (WE thrombin) has been one of the best characterized constructs with selective activity toward protein C. Although animal studies have demonstrated that WE thrombin acts as an anticoagulant through activated protein C (APC) generation, the observed limited systemic anticoagulation does not fully explain the antithrombotic potency of this or other thrombin mutants. AB002 (E-WE thrombin) is an investigational protein C activator thrombin analog in phase 2 clinical development (clinicaltrials.gov NCT03963895). Here, we demonstrate that this molecule is a potent enzyme that is able to rapidly interrupt arterial-type thrombus propagation at exceedingly low doses (<2 µg/kg, IV), yet without substantial systemic anticoagulation in baboons. We demonstrate that AB002 produces APC on platelet aggregates and competitively inhibits thrombin-activatable fibrinolysis inhibitor (carboxypeptidase B2) activation in vitro, which may contribute to the observed in vivo efficacy. We also describe its safety and activity in a phase 1 first-in-human clinical trial. Together, these results support further clinical evaluation of AB002 as a potentially safe and effective new approach for treating or preventing acute thrombotic and thromboembolic conditions. This trial was registered at www.clinicaltrials.gov as #NCT03453060.


Asunto(s)
Fibrinolíticos/farmacología , Proteína C/efectos de los fármacos , Trombina/análogos & derivados , Trombosis/prevención & control , Adulto , Animales , Método Doble Ciego , Humanos , Persona de Mediana Edad , Papio , Proteínas Recombinantes/farmacología
3.
Commun Med (Lond) ; 4(1): 153, 2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-39060370

RESUMEN

BACKGROUND: The protein C system regulates blood coagulation, inflammation, and vascular integrity. AB002 is an injectable protein C activating enzyme under investigation to safely prevent and treat thrombosis. In preclinical models, AB002 is antithrombotic, cytoprotective, and anti-inflammatory. Since prophylactic use of heparin is contraindicated during hemodialysis in some end-stage renal disease (ESRD) patients, we propose using AB002 as a short-acting alternative to safely limit blood loss due to clotting in the dialysis circuit. METHODS: This phase 2, randomized, double-blind, placebo-controlled, single-dose study evaluates the safety and tolerability of AB002 administered into the hemodialysis line of ESRD patients during hemodialysis at one study center in the United States (ClinicalTrials.gov: NCT03963895). In this study, 36 patients were sequentially enrolled into two cohorts and randomized to AB002 or placebo in a 2:1 ratio. In cohort 1, patients received 1.5 µg/kg AB002 (n = 12) or placebo (n = 6); in cohort 2, patients received 3 µg/kg AB002 (n = 12) or placebo (n = 6). Patients underwent five heparin-free hemodialysis sessions over 10 days and were dosed with AB002 or placebo during session four. RESULTS: Here we show that AB002 is safe and well-tolerated in ESRD patients, with no treatment-related adverse events. Clinically relevant bleeding did not occur in any patient, and the time to hemostasis at the vascular access sites is not affected by AB002. CONCLUSIONS: As far as we are aware, this proof-of-concept study is the first clinical trial assessing the therapeutic potential of protein C activation. The results herein support additional investigation of AB002 to safely prevent and treat thrombosis in at-risk populations.


Some people with kidney disease require hemodialysis, a process in which a machine filters the blood to remove waste products. The process of hemodialysis can trigger blood clotting in the hemodialysis circuit. Therefore, the blood-thinner heparin is commonly used to prevent blood from clotting. However, some patients cannot tolerate heparin. Here we describe a clinical trial in which we tested whether a drug called AB002 is safe and can reduce hemodialysis circuit clotting in people with permanent kidney disease (end-stage renal disease) undergoing hemodialysis. AB002 appears to be safe and well-tolerated, and we observed reduced clotting without any signs of increased bleeding. Further studies are required in more patients to determine whether AB002 can be used routinely during hemodialysis to safely prevent or treat blood clots.

4.
Methods Mol Biol ; 2598: 45-63, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36355284

RESUMEN

Cartilage resides under a low oxygen tension within articulating joints. The oxygen tension within cartilage of the knee joint has been measured to be between 2% and 5% oxygen. Although the literature has historically termed this level of oxygen as hypoxia, particularly when doing experiments in vitro in this range, this is actually the physiological oxygen tension experienced in vivo and is more accurately termed physioxia. In general, culture of chondrogenic cells under physioxia has demonstrated a donor-dependent beneficial effect on chondrogenesis, with an upregulation in cartilage genes (SOX9, COL2A1, ACAN) and matrix deposition (sulfated glycosaminoglycans (sGAGs), collagen II). Physioxia also reduces the expression of hypertrophic markers (COL10A1, MMP13). This chapter will outline the methods for the expansion and differentiation of chondrogenic cells under physioxia using oxygen-controlled incubators and glove box environments, with the typical assays used for qualitative and quantitative assessment of chondrogenesis.


Asunto(s)
Condrogénesis , Células Madre Mesenquimatosas , Condrocitos/metabolismo , Células Cultivadas , Diferenciación Celular/fisiología , Oxígeno/metabolismo
5.
Tissue Eng Part A ; 24(3-4): 264-274, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28474537

RESUMEN

INTRODUCTION: Biomaterial-based tissue engineering has not successfully reproduced the structural architecture or functional mechanical properties of native articular cartilage. In scaffold-free tissue engineering systems, cells secrete and organize the entire extracellular matrix over time in response to environmental signals such as oxygen level. In this study, we investigated the effect of oxygen on the formation of neocartilage from human-derived chondrogenic cells. MATERIALS AND METHODS: Articular chondrocytes (ACs) and articular cartilage progenitor cells (ACPs) derived from healthy human adults were guided toward cell condensation by centrifugation onto plate inserts that were uncoated or coated with either agarose or fibronectin. Neocartilage discs were cultured at hyperoxic (20%) or physioxic (5%) oxygen levels, and biochemical, biomechanical, and molecular analyses were used to compare the cartilage produced by ACs versus ACPs. RESULTS: Fibronectin-coated inserts proved optimal for growing cartilaginous discs from both cell types. In comparison with culture in hyperoxia, AC neocartilage cultured at physioxia exhibited a significant increase in chondrogenic gene expression, proteoglycan production, and mechanical properties with a concomitant decrease in collagen content. At both oxygen levels, ACP-derived neocartilage produced tissue with significantly enhanced mechanical properties and collagen content relative to AC-derived neocartilage. Both ACs and ACPs produced substantial collagen II and reduced levels of collagens I and X in physioxia relative to hyperoxia. Neocartilage from ACPs exhibited anisotropic organization characteristic of native cartilage with respect to collagen VI of the pericellular matrix when compared with AC-derived neocartilage; however, only ACs produced abundant surface-localized lubricin. DISCUSSION AND CONCLUSIONS: Guiding human-derived cells toward condensation and subsequent culture in physioxia promoted the articular cartilage tissue phenotype for ACs and ACPs. Unlike ACs, ACPs are clonable and highly expandable while retaining chondrogenicity. The ability to generate large tissues utilizing a scaffold-free approach from a single autologous progenitor cell may represent a promising source of neocartilage destined for cartilage repair.


Asunto(s)
Cartílago Articular/citología , Condrocitos/citología , Condrocitos/fisiología , Condrogénesis/fisiología , Células Cultivadas , Colágeno Tipo I/química , Fibronectinas/química , Humanos , Ingeniería de Tejidos/métodos , Andamios del Tejido/química
6.
Stem Cell Res Ther ; 7(1): 154, 2016 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-27765063

RESUMEN

BACKGROUND: Lowering oxygen from atmospheric level (hyperoxia) to the physiological level (physioxia) of articular cartilage promotes mesenchymal stem cell (MSC) chondrogenesis. However, the literature is equivocal regarding the benefits of physioxic culture on preventing hypertrophy of MSC-derived chondrocytes. Articular cartilage progenitors (ACPs) undergo chondrogenic differentiation with reduced hypertrophy marker expression in hyperoxia but have not been studied in physioxia. This study sought to delineate the effects of physioxic culture on both cell types undergoing chondrogenesis. METHODS: MSCs were isolated from human bone marrow aspirates and ACP clones were isolated from healthy human cartilage. Cells were differentiated in pellet culture in physioxia (2 % oxygen) or hyperoxia (20 % oxygen) over 14 days. Chondrogenesis was characterized by biochemical assays and gene and protein expression analysis. RESULTS: MSC preparations and ACP clones of high intrinsic chondrogenicity (termed high-GAG) produced abundant matrix in hyperoxia and physioxia. Poorly chondrogenic cells (low-GAG) demonstrated a significant fold-change matrix increase in physioxia. Both high-GAG and low-GAG groups of MSCs and ACPs significantly upregulated chondrogenic genes; however, only high-GAG groups had a concomitant decrease in hypertrophy-related genes. High-GAG MSCs upregulated many common hypoxia-responsive genes in physioxia while low-GAG cells downregulated most of these genes. In physioxia, high-GAG MSCs and ACPs produced comparable type II collagen but less type I collagen than those in hyperoxia. Type X collagen was detectable in some ACP pellets in hyperoxia but reduced or absent in physioxia. In contrast, type X collagen was detectable in all MSC preparations in hyperoxia and physioxia. CONCLUSIONS: MSC preparations and ACP clones had a wide range of chondrogenicity between donors. Physioxia significantly enhanced the chondrogenic potential of both ACPs and MSCs compared with hyperoxia, but the magnitude of response was inversely related to intrinsic chondrogenic potential. Discrepancies in the literature regarding MSC hypertrophy in physioxia can be explained by the use of low numbers of preparations of variable chondrogenicity. Physioxic differentiation of MSC preparations of high chondrogenicity significantly decreased hypertrophy-related genes but still produced type X collagen protein. Highly chondrogenic ACP clones had significantly lower hypertrophic gene levels, and there was little to no type X collagen protein in physioxia, emphasizing the potential advantage of these cells.


Asunto(s)
Condrogénesis/fisiología , Células Madre Mesenquimatosas/metabolismo , Oxígeno/metabolismo , Anciano , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/fisiología , Cartílago Articular/metabolismo , Cartílago Articular/fisiología , Diferenciación Celular/fisiología , Células Cultivadas , Condrocitos/metabolismo , Condrocitos/fisiología , Colágeno Tipo I/metabolismo , Colágeno Tipo II/metabolismo , Colágeno Tipo X/metabolismo , Femenino , Humanos , Hipertrofia/metabolismo , Hipertrofia/fisiopatología , Masculino , Células Madre Mesenquimatosas/fisiología , Persona de Mediana Edad
7.
J Orthop Res ; 33(11): 1561-70, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26174816

RESUMEN

The hypoxia-inducible factors HIF-1α and HIF-2α are important regulators of the chondrocyte phenotype but little is known about HIF-3α in cartilage. The objective of this study was to characterize HIF-3α (HIF3A) expression during chondrocyte differentiation in vitro and in native cartilage tissues. HIF3A, COL10A1, and MMP13 were quantified in mesenchymal stem cells (MSCs) and articular chondrocytes from healthy and osteoarthritic (OA) tissue in three-dimensional cultures and in human embryonic epiphyses and adult articular cartilage. HIF3A was found to have an inverse association with hypertrophic markers COL10A1 and MMP13 in chondrogenic cells and tissues. In healthy chondrocytes, HIF3A was induced by dexamethasone and increased during redifferentiation. By comparison, HIF3A expression was extremely low in chondrogenically differentiated MSCs expressing high levels of COL10A1 and MMP13. HIF3A was also lower in redifferentiated OA chondrocytes than in healthy chondrocytes. In human embryonic epiphyseal tissue, HIF3A expression was lowest in the hypertrophic zone. Distinct splice patterns were also found in embryonic cartilage when compared with adult articular cartilage and redifferentiated chondrocytes. These in vitro and in vivo findings suggest that HIF3A levels are indicative of the hypertrophic state of chondrogenic cells and one or more splice variants may be important regulators of the chondrocyte phenotype.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Condrocitos/citología , Condrogénesis , Células Madre Mesenquimatosas/fisiología , Proteínas Reguladoras de la Apoptosis , Cartílago Articular/embriología , Células Cultivadas , Condrocitos/metabolismo , Humanos , Osteoartritis/metabolismo , Fenotipo , Proteínas Represoras
8.
Arthritis Res Ther ; 15(4): R92, 2013 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-23965235

RESUMEN

INTRODUCTION: Hypoxia is considered to be a positive influence on the healthy chondrocyte phenotype and cartilage matrix formation. However, hypoxia-inducible factors (HIFs) have been implicated in the pathogenesis of osteoarthritis (OA). Thus, we assessed whether healthy and OA chondrocytes have distinct responses to oxygen, particularly with regard to hypertrophy and degradation during redifferentiation. METHODS: Monolayer-expanded healthy and OA chondrocytes were redifferentiated for 14 days in pellet cultures under standard (20% oxygen) or hypoxic (2% oxygen) conditions. Cartilage matrix gene expression, matrix quality and quantity, degradative enzyme expression and HIF expression were measured. RESULTS: In hypoxia, both healthy and OA chondrocytes had higher human collagen type II, α1 gene (COL2A1), and aggrecan (ACAN) expression and sulfated glycosaminoglycan (sGAG) accumulation, concomitant with lower human collagen type X, α1 gene (COL10A1), and human collagen type I, α1 gene (COL1A1), expression and collagen I extracellular accumulation. OA chondrocytes had significantly lower sGAGs/DNA than healthy chondrocytes, but only in high oxygen conditions. Hypoxia also caused significantly greater sGAG retention and hyaluronic acid synthase 2 (HAS2) expression by OA chondrocytes. Both healthy and OA chondrocytes had significantly lower expression of matrix metalloproteinases (MMPs) MMP1, MMP2, MMP3 and MMP13 in hypoxia and less active MMP2 enzyme, consistent with lower MMP14 expression. However, aggrecanase (ADAMTS4 and ADAMTS5) expression was significantly lowered by hypoxia only in healthy cells, and COL10A1 and MMP13 remained significantly higher in OA chondrocytes than in healthy chondrocytes in hypoxic conditions. HIF-1α and HIF-2α had similar expression profiles in healthy and OA cells, increasing to maximal levels early in hypoxia and decreasing over time. CONCLUSIONS: Hypoxic culture of human chondrocytes has long been acknowledged to result in increased matrix accumulation, but still little is known of its effects on catabolism. We show herein that the increased expression of matrix proteins, combined with decreased expression of numerous degradative enzymes by hypoxia, minimizes but does not abolish differences between redifferentiated healthy and OA chondrocytes. Hypoxia-induced HIF expression is associated with hypertrophic marker and degradative enzyme downregulation and increased measures of redifferentiation in both healthy and OA chondrocytes. Therefore, though HIFs may be involved in the pathogenesis of OA, conditions that promote HIF expression in vitro promote matrix accumulation and decrease degradation and hypertrophy, even in cells from OA joints.


Asunto(s)
Hipoxia de la Célula/fisiología , Condrocitos/citología , Condrocitos/metabolismo , Osteoartritis/metabolismo , Osteoartritis/patología , Western Blotting , Cartílago Articular/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Humanos , Hipertrofia/metabolismo , Hipertrofia/patología , Inmunohistoquímica , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
Cell Transplant ; 19(1): 29-42, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19878627

RESUMEN

Chondrogenesis of mesenchymal stem cells (MSCs) is typically induced when they are condensed into a single aggregate and exposed to transforming growth factor-beta (TGF-beta). Hypoxia, like aggregation and TGF-beta delivery, may be crucial for complete chondrogenesis. However, the pellet dimensions and associated self-induced oxygen gradients of current chondrogenic methods may limit the effectiveness of in vitro differentiation and subsequent therapeutic uses. Here we describe the use of embryoid body-forming technology to produce microscopic aggregates of human bone marrow MSCs (BM-MSCs) for chondrogenesis. The use of micropellets reduces the formation of gradients within the aggregates, resulting in a more homogeneous and controlled microenvironment. These micropellet cultures (approximately 170 cells/micropellet) as well as conventional pellet cultures (approximately 2 x 10(5) cells/pellet) were chondrogenically induced under 20% and 2% oxygen environments for 14 days. Compared to conventional pellets under both environments, micropellets differentiated under 2% O(2) showed significantly increased sulfated glycosaminoglycan (sGAG) production and more homogeneous distribution of proteoglycans and collagen II. Aggrecan and collagen II gene expressions were increased in pellet cultures differentiated under 2% O(2) relative to 20% O(2) pellets but 2% O(2) micropellets showed even greater increases in these genes, as well as increased SOX9. These results suggest a more advanced stage of chondrogenesis in the micropellets accompanied by more homogeneous differentiation. Thus, we present a new method for enhancing MSC chondrogenesis that reveals a unique relationship between oxygen tension and aggregate size. The inherent advantages of chondrogenic micropellets over a single macroscopic aggregate should allow for easy integration with a variety of cartilage engineering strategies.


Asunto(s)
Diferenciación Celular/fisiología , Condrocitos/metabolismo , Condrogénesis/fisiología , Hipoxia/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Agrecanos/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Agregación Celular/fisiología , Técnicas de Cultivo de Célula/métodos , Linaje de la Célula/fisiología , Proliferación Celular , Separación Celular/métodos , Células Cultivadas , Condrocitos/citología , Colágeno Tipo II/metabolismo , Glicosaminoglicanos/metabolismo , Humanos , Hipoxia/fisiopatología , Células Madre Mesenquimatosas/citología , Consumo de Oxígeno/fisiología , Esferoides Celulares/citología , Esferoides Celulares/metabolismo
10.
Tissue Eng Part C Methods ; 16(3): 407-15, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19622005

RESUMEN

In membrane bioreactors the cells are isolated from the bulk medium through a semipermeable membrane. This concept, which is analogous to how the circulatory system supplies solid tissues with nutrients, allows the maintenance of cells at much higher densities than is possible in traditional cultures. The membrane-based microbioreactor described herein is easy to operate, requiring only a pipette to load and harvest cells. A 10 microL culture volume was isolated from 1 mL of bulk medium through a semipermeable membrane having a molecular weight cutoff of 10 kDa. Here we describe the benefits regarding the retention of both cells and their secretions within this small culture volume using the following two model systems: hematopoietic stem cell expansion and mesenchymal stem cell-derived cartilage matrix accumulation.


Asunto(s)
Reactores Biológicos , Membranas Artificiales , Diferenciación Celular , Medios de Cultivo , Células Madre Hematopoyéticas/citología , Humanos
11.
Biomaterials ; 30(34): 6614-20, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19781762

RESUMEN

The presentation of proteins on surfaces is fundamental to numerous cell culture and tissue engineering applications. While a number of physisorption and cross-linking methods exist to facilitate this process, few avoid denaturation of proteins or allow control over protein orientation, both of which are critical to the functionality of many signal proteins and ligands. Often recombinant protein sequences include a poly-histidine tag to facilitate purification. We utilize this sequence to anchor proteins to biosurfaces via a peptide bonded to the surface which conjugates with the poly-histidine tag in the presence of zinc rather than nickel, which is more traditionally used to conjugate poly-histidine tags to surfaces. We demonstrate that this strategy enables the display of proteins on 2D and 3D surfaces without compromising protein function through direct cross-linking or physisorption.


Asunto(s)
Proteínas Portadoras/química , Péptidos/química , Secuencia de Aminoácidos , Fibronectinas/química , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Recombinantes/química
12.
Biomaterials ; 30(25): 4047-52, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19481255

RESUMEN

In vivo, stem cell factor (SCF) exists in both a bound and soluble isoform. It is believed that the bound form is more potent and fundamentally required for the maintenance of hematopoietic stem cells (HSCs). This theory is supported by the observation that steel-Dickie mice lacking the bound isoform of SCF are unable to maintain hematopoiesis and by the fact that bound SCF displayed on the surface of transgenic cells is better able to maintain c-kit activation than soluble SCF. Further work has shown that recombinant SCF molecules, which include a surface-binding domain, are more potent than their soluble equivalent. It is generally assumed that such an elegant approach is necessary to provide the correct molecular orientation and avoid the pitfalls of random cross-linking or the denaturation associated with the adsorption of proteins to surfaces. However, in this work we demonstrate that SCF physisorbed to tissue culture plastic (TCP) is not only bioactive, but more potent than the soluble equivalent. By contrast, cross-linking of SCF via free amines is shown to compromise its bioactivity. These observations demonstrate that simple surface modification solutions cannot be discounted and with the advent of low-cost pharmaceutical grade proteins, they should not be.


Asunto(s)
Proliferación Celular , Células Madre Hematopoyéticas/fisiología , Factor de Células Madre/metabolismo , Animales , Técnicas de Cultivo de Célula , Células Cultivadas , Medios de Cultivo Condicionados/metabolismo , Células Madre Hematopoyéticas/citología , Ratones , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Factor de Células Madre/genética , Propiedades de Superficie
13.
Tissue Eng Part C Methods ; 14(2): 97-105, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18544026

RESUMEN

In humans, self-endothelialization of synthetic grafts is severely limited, but a recent interesting idea is to attract endothelial progenitor cells (EPCs) from peripheral blood onto grafts via antibodies directed at proposed EPC markers. Results with anti-CD34 antibodies have shown some promise, but it is unclear whether CD34 is the best marker for cells with re-endothelializing potential. Much evidence points to kinase insert domain receptor (KDR) as an important indicator of endothelial potential if not a definitive marker. Because KDR is not an adhesion molecule (like CD34), we first demonstrated the ability to use adsorbed and protein G-oriented antibody to this receptor to capture flowing cells onto a solid surface. Using endothelial cells and smooth muscle cells, we show in a model system under low shear rates the ability to selectively capture cells by this receptor. Furthermore, our results indicate that concomitant flow of cells lacking the receptor does not affect the efficiency of capture of KDR(+) cells but that orienting the antibody significantly increases the efficiency of capture.


Asunto(s)
Antígenos CD34/química , Ingeniería de Tejidos/métodos , Animales , Antígenos CD34/biosíntesis , Plaquetas/metabolismo , Células Cultivadas , Endotelio Vascular/metabolismo , Humanos , Modelos Biológicos , Miocitos del Músculo Liso/metabolismo , Papio , Estructura Terciaria de Proteína , Propiedades de Superficie , Trombosis/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
14.
J Biomed Mater Res A ; 86(3): 804-12, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18041726

RESUMEN

Thrombosis and intimal hyperplasia limit the usefulness of small caliber vascular grafts. While some improvements have been reported for grafts seeded with mature endothelial cells (EC), the harvesting of ECs from autologous sources, for example, veins or adipose tissue, remains problematic. More recently, endothelial progenitor cells (EPCs) have been considered a promising source of ECs because EPCs can be readily isolated from whole blood then rapidly expanded in vitro. Additionally, EPCs are increasingly recognized to play important roles in hemostasis, angiogenesis, and arterial injury repair. However, the characterization of EPCs in relevant animal models remains poorly defined. Accordingly, we have characterized the isolation, growth, and functional characteristics of Baboon EPCs (BaEPCs) to evaluate their potential for an autologous cell source for tissue engineered vascular grafts. BaEPCs were successfully cultured from the peripheral blood with an average population doubling time of 1.17 +/- 0.43 days. While the BaEPCs were positive for typical EC markers of vWF, CD31, VE-cadherin, VEGF-R2, Thrombomodulin, and E-selectin, there was reduced eNOS expression. The BaEPCs cell body and actin filaments align in the direction of flow typical of mature ECs. Thus while the lack of eNOS expression is worthy of investigation, EPCs are an attractive cell source for tissue engineered vascular grafts and the baboon model has great potential for continuing evaluations of these cells.


Asunto(s)
Prótesis Vascular , Células Endoteliales/citología , Células Madre/citología , Ingeniería de Tejidos , Actinas/metabolismo , Animales , Coagulación Sanguínea , Western Blotting , Proliferación Celular , Separación Celular , Forma de la Célula , Células Cultivadas , Células Endoteliales/ultraestructura , Humanos , Inmunohistoquímica , Masculino , Papio , Fenotipo , Politetrafluoroetileno/metabolismo , Ratas , Reología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA