Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 484(3): 656-661, 2017 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-28153723

RESUMEN

Trypsin is frequently used to dissociate mesenchymal stem cells (MSCs) for in vitro adhesion and chemotaxis assays. However, its potential impact on surface receptor degradation is poorly understood. The purpose of this study was to evaluate the effect of trypsin-EDTA exposure versus PBS-EDTA on MSC surface receptor integrity and function. Primary human MSCs were detached with PBS-EDTA alone, or Cell Dissociation Buffer followed by 30 s exposure to 0.05% w/v trypsin-EDTA (trace trypsin method, TT), or 0.25% w/v trypsin exposure for 2 or 5 min. Cells were characterized for surface integrity of ß1 integrin (CD29) and PDGF Receptor (PDGF-R), and assessed in vitro for adhesion to atelocollagen-coated surfaces and migration to PDGF-BB. PBS-EDTA detachment fully preserved receptor integrity but routinely detached only half of the adherent cells and led to cell aggregates that failed to adhere evenly across the Transwell migration insert. Both CD29 and PDGF-R were significantly degraded by 0.25% trypsin detachment for 2 or 5 min compared to the TT method or PBS-EDTA (p < 0.05). Cells migrated optimally to PDGF-BB when detached with the TT method (3.1-fold vs α-MEM, p = 0.01). Cells attached optimally to atelocollagen when detached using the TT method or PBS-EDTA (6- to 10-fold vs 0.25% trypsin, p < 0.01). CDB followed by trace trypsin-EDTA exposure is recommended over PBS-EDTA to produce a single-cell MSC suspension that preserves receptor integrity and more reproducible receptor-mediated responses.


Asunto(s)
Adhesión Celular/fisiología , Ensayos de Migración Celular/métodos , Quimiotaxis/fisiología , Ácido Edético/administración & dosificación , Células Madre Mesenquimatosas/fisiología , Tripsina/administración & dosificación , Adulto , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Quimiotaxis/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos
2.
J Mater Sci Mater Med ; 25(9): 2083-93, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24962985

RESUMEN

Porous structures destined for tissue engineering applications should ideally show controlled and narrow pore size distributions with fully interconnected pores. This study focuses on the development of novel poly(ε-caprolactone) (PCL) structures with fully connected pores of 84, 116, 141, and 162 µm average diameter, from melt blending of PCL with poly(ethylene oxide) (PEO) at the co-continuous composition, followed by static annealing and selective extraction of PEO. Our results demonstrate a low onset concentration for PEO continuity and a broad region of phase inversion. A novel in vitro assay was used to compare scaffold infiltration by 10-µm diameter polystyrene beads intended to mimic trypsinized human bone marrow stromal cells (hBMSCs). Beads showed a linear increase in the extent of scaffold infiltration with increasing pore size, whereas BMSCs infiltrated 162 and 141 µm pores, below which the cells aggregated and adhered near the seeding area with low infiltration into the porous device. While providing a baseline for non-aggregated systems, the beads closely mimic trypsinized cells at pore sizes equal to or larger than 141 µm, where optimal retention and distribution of hBMSCs are detected. A cytotoxicity assay using L929 cells showed that these scaffolds were cytocompatible and no cell necrosis was detected. This study shows that a melt blending approach produces porous PCL scaffolds of highly controlled pore size, narrow size distribution and complete interconnectivity, while the bead model system reveals the baseline potential for a homogeneous, non-aggregated distribution of hBMSCs at all penetration depths.


Asunto(s)
Poliésteres/química , Andamios del Tejido , Animales , Línea Celular , Humanos , Ratones , Microscopía Electrónica de Rastreo , Porosidad
3.
J Thromb Haemost ; 22(4): 1031-1045, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38135253

RESUMEN

BACKGROUND: Blood clots are living tissues that release inflammatory mediators including IL-8/CXCL8 and MCP-1/CCL2. A deeper understanding of blood clots is needed to develop new therapies for prothrombotic disease states and regenerative medicine. OBJECTIVES: To identify a common transcriptional shift in cultured blood clot leukocytes. METHODS: Differential gene expression of whole blood and cultured clots (4 hours at 37 °C) was assessed by RNA sequencing (RNAseq), reverse transcriptase-polymerase chain reaction, proteomics, and histology (23 diverse healthy human donors). Cultured clot serum bioactivity was tested in endothelial barrier functional assays. RESULTS: All cultured clots developed a polymorphonuclear myeloid-derived suppressor cell (PMN-MDSC) signature, including up-regulation of OLR1 (mRNA encoding lectin-like oxidized low-density lipoprotein receptor 1 [Lox-1]), IL-8/CXCL8, CXCL2, CCL2, IL10, IL1A, SPP1, TREM1, and DUSP4/MKP. Lipopolysaccharide enhanced PMN-MDSC gene expression and specifically induced a type II interferon response with IL-6 production. Lox-1 was specifically expressed by cultured clot CD15+ neutrophils. Cultured clot neutrophils, but not activated platelets, shed copious amounts of soluble Lox-1 (sLox-1) with a donor-dependent amplitude. sLox-1 shedding was enhanced by phorbol ester and suppressed by heparin and by beta-glycerol phosphate, a phosphatase inhibitor. Cultured clot serum significantly enhanced endothelial cell monolayer barrier function, consistent with a proresolving bioactivity. CONCLUSION: This study suggests that PMN-MDSC activation is part of the innate immune response to coagulation which may have a protective role in inflammation. The cultured blood clot is an innovative thrombus model that can be used to study both sterile and nonsterile inflammatory states and could be used as a personalized medicine tool for drug screening.


Asunto(s)
Células Supresoras de Origen Mieloide , Trombosis , Humanos , Interleucina-8 , Neutrófilos , Células Supresoras de Origen Mieloide/patología , Coagulación Sanguínea/fisiología , Trombosis/patología
4.
BMC Musculoskelet Disord ; 14: 27, 2013 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-23324433

RESUMEN

BACKGROUND: In this study we evaluated a novel approach to guide the bone marrow-driven articular cartilage repair response in skeletally aged rabbits. We hypothesized that dispersed chitosan particles implanted close to the bone marrow degrade in situ in a molecular mass-dependent manner, and attract more stromal cells to the site in aged rabbits compared to the blood clot in untreated controls. METHODS: Three microdrill hole defects, 1.4 mm diameter and 2 mm deep, were created in both knee trochlea of 30 month-old New Zealand White rabbits. Each of 3 isotonic chitosan solutions (150, 40, 10 kDa, 80% degree of deaceylation, with fluorescent chitosan tracer) was mixed with autologous rabbit whole blood, clotted with tissue factor to form cylindrical implants, and press-fit in drill holes in the left knee while contralateral holes received tissue factor or no treatment. At day 1 or day 21 post-operative, defects were analyzed by micro-computed tomography, histomorphometry and stereology for bone and soft tissue repair. RESULTS: All 3 implants filled the top of defects at day 1 and were partly degraded in situ at 21 days post-operative. All implants attracted neutrophils, osteoclasts and abundant bone marrow-derived stromal cells, stimulated bone resorption followed by new woven bone repair (bone remodeling) and promoted repair tissue-bone integration. 150 kDa chitosan implant was less degraded, and elicited more apoptotic neutrophils and bone resorption than 10 kDa chitosan implant. Drilled controls elicited a poorly integrated fibrous or fibrocartilaginous tissue. CONCLUSIONS: Pre-solidified implants elicit stromal cells and vigorous bone plate remodeling through a phase involving neutrophil chemotaxis. Pre-solidified chitosan implants are tunable by molecular mass, and could be beneficial for augmented marrow stimulation therapy if the recruited stromal cells can progress to bone and cartilage repair.


Asunto(s)
Materiales Biocompatibles , Resorción Ósea/metabolismo , Enfermedades de los Cartílagos/tratamiento farmacológico , Cartílago Articular/efectos de los fármacos , Quimiotaxis , Quitosano/farmacología , Matriz Extracelular/metabolismo , Articulación de la Rodilla/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Regeneración/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Animales , Coagulación Sanguínea , Resorción Ósea/patología , Enfermedades de los Cartílagos/metabolismo , Enfermedades de los Cartílagos/patología , Cartílago Articular/metabolismo , Cartílago Articular/patología , Cartílago Articular/cirugía , Quitosano/administración & dosificación , Quitosano/química , Implantes de Medicamentos , Femenino , Articulación de la Rodilla/metabolismo , Articulación de la Rodilla/patología , Articulación de la Rodilla/cirugía , Masculino , Modelos Animales , Peso Molecular , Neutrófilos/metabolismo , Neutrófilos/patología , Conejos , Células del Estroma/metabolismo , Células del Estroma/patología , Tromboplastina/farmacología , Factores de Tiempo , Microtomografía por Rayos X
5.
J Knee Surg ; 26(5): 347-55, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23529699

RESUMEN

Microfracture (MFX) is a cartilage repair technique that depends on cell migration from marrow-rich trabecular bone cavities into the cartilage lesion. This study tested the hypothesis that MFX awls with distinct geometry generate different hole shapes and variable bone marrow access in condyles with Grade III to IV lesions. Lateral and medial condyles from total knee arthroplasty (N = 24 male and female patients, 66 ± 9 years) were systematically microfractured ex vivo to 2 and 4 mm deep and the bone holes analyzed by micro-computed tomography. Subchondral bone in lesional condyles showed different degrees of sclerosis up to 2 mm deep ("porous," sclerotic, extremely dense). MFX holes ranged from 1.1 to 2.0 mm in diameter, and retained the awl shape with evidence of slight bone elastic rebound and bone compaction lining the holes that were increased by wider awl diameter and deeper MFX. Marrow access was significantly diminished by sclerosis for all three awls, with an average marrow access varying from 70% (nonlesional bone) to 40% (extremely dense bone). This study revealed that subchondral bone sclerosis can reach a critical limit beyond which MFX creates bone compaction and fissures instead of marrow access.


Asunto(s)
Artroplastia Subcondral , Fémur/patología , Fémur/cirugía , Anciano , Densidad Ósea , Femenino , Humanos , Imagenología Tridimensional , Masculino , Osteoartritis de la Rodilla/patología , Porosidad , Esclerosis , Microtomografía por Rayos X
6.
Molecules ; 18(1): 1015-35, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23322067

RESUMEN

UNLABELLED: Chitosan is a biocompatible polysaccharide composed of glucosamine and N-acetylglucosamine. The polymer has a unique behavior of fluctuating between soluble chains at pH 6 and insoluble microparticles at pH 7. The purpose of this study was to test the hypothesis that chitosan structure, solubility state, and serum influence the rate of cell uptake. Chitosans with 80% and 95% degree of deacetylation (medium and low viscosity) were tagged with rhodamine and analyzed for particle size, media solubility, and uptake by HEK293 epithelial cells using live confocal microscopy and flow cytometry. In media pH 7.4 with or without 10% serum, chitosans fully precipitated into 0.5 to 1.4 µm diameter microparticles with a slight negative charge. During 24 h of culture in serum-free medium, chitosan particles remained extracellular. In cultures with serum, particles were taken up into intracellular vesicles in a serum dose-dependent manner. Opsonization of chitosan with serum, or replacement of serum by epidermal growth factor (EGF) failed to mediate serum-free chitosan particle uptake. Serum stimulated cells to acidify the media, partly by lactate generation. Media acidified to pH 6.5 by 7 mM lactate maintained 50% of chitosan in the soluble fraction, and led to minor uniform serum-free uptake in small vesicles. CONCLUSION: Media acidification mediates minor in vitro uptake of non-biofouled soluble chitosan chains, while serum-biofouled insoluble chitosan microparticles require sustained serum exposure to generate energy required for macropinocytosis.


Asunto(s)
Quitosano/metabolismo , Ácido Láctico/metabolismo , Suero/fisiología , Conformación de Carbohidratos , Quitosano/química , Medios de Cultivo , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Luz , Microscopía Confocal , Microscopía Fluorescente , Tamaño de la Partícula , Rodaminas/química , Rodaminas/metabolismo , Dispersión de Radiación , Solubilidad , Espectroscopía Infrarroja por Transformada de Fourier
7.
J Knee Surg ; 25(2): 85-97, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22928426

RESUMEN

In the knee joint, the purpose of the cartilage-bone interface is to maintain structural integrity of the osteochondral unit during walking, kneeling, pivoting, and jumping--during which tensile, compressive, and shear forces are transmitted from the viscoelastic articular cartilage layer to the much stiffer mineralized end of the long bone. Mature articular cartilage is integrated with subchondral bone through a approximately 20 to approximately 250 microm thick layer of calcified cartilage. Inside the calcified cartilage layer, perpendicular chondrocyte-derived collagen type II fibers become structurally cemented to collagen type I osteoid deposited by osteoblasts. The mature mineralization front is delineated by a thin approximately 5 microm undulating tidemark structure that forms at the base of articular cartilage. Growth plate cartilage is anchored to epiphyseal bone, sometimes via a thin layer of calcified cartilage and tidemark, while the hypertrophic edge does not form a tidemark and undergoes continual vascular invasion and endochondral ossification (EO) until skeletal maturity upon which the growth plates are fully resorbed and replaced by bone. In this review, the formation of the cartilage-bone interface during skeletal development and cartilage repair, and its structure and composition are presented. Animal models and human anatomical studies show that the tidemark is a dynamic structure that forms within a purely collagen type II-positive and collagen type I-negative hyaline cartilage matrix. Cartilage repair strategies that elicit fibrocartilage, a mixture of collagen type I and type II, are predicted to show little tidemark/calcified cartilage regeneration and to develop a less stable repair tissue-bone interface. The tidemark can be regenerated through a bone marrow-driven growth process of EO near the articular surface.


Asunto(s)
Desarrollo Óseo , Cartílago Articular/crecimiento & desarrollo , Articulación de la Rodilla/crecimiento & desarrollo , Animales , Artroplastia Subcondral , Huesos/anatomía & histología , Cartílago Articular/anatomía & histología , Humanos , Articulación de la Rodilla/anatomía & histología
8.
Bioact Mater ; 10: 430-442, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34901558

RESUMEN

This study tested whether osseous integration into poly (ε-caprolactone) (PCL) bioplastic scaffolds with fully-interconnecting 155 ± 8 µm pores is enhanced by an adhesive, non-inflammatory 99% degree of deacetylation (DDA) chitosan coating (99-PCL), or further incorporation of pro-inflammatory 83% DDA chitosan microparticles (83-99-PCL) to accelerate angiogenesis. New Zealand White rabbit osteochondral knee defects were press-fit with PCL, 99-PCL, 83-99-PCL, or allowed to bleed (drill-only). Between day 1 and 6 weeks of repair, drill-only defects repaired by endochondral ossification, with an 8-fold higher bone volume fraction (BVF) versus initial defects, compared to a 2-fold (99-PCL), 1.1-fold (PCL), or 0.4-fold (83-99-PCL) change in BVF. Hematoma innate immune cells swarmed to 83-99-PCL, elicited angiogenesis throughout the pores and induced slight bone resorption. PCL and 99-PCL pores were variably filled with cartilage or avascular mesenchyme near the bone plate, or angiogenic mesenchyme into which repairing trabecular bone infiltrated up to 1 mm deep. More repair cartilage covered the 99-PCL scaffold (65%) than PCL (18%) or 83-99-PCL (0%) (p < 0.005). We report the novel finding that non-inflammatory chitosan coatings promoted cartilage infiltration into and over a bioplastic scaffold, and were compatible with trabecular bone integration. This study also revealed that in vitro osteogenesis assays have limited ability to predict osseous integration into porous scaffolds, because (1) in vivo, woven bone integrates from the leading edge of regenerating trabecular bone and not from mesenchymal cells adhering to scaffold surfaces, and (2) bioactive coatings that attract inflammatory cells induce bone resorption.

9.
PLoS One ; 17(9): e0274420, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36107941

RESUMEN

UDP-glucose dehydrogenase (UGDH) generates essential precursors of hyaluronic acid (HA) synthesis, however mechanisms regulating its activity are unclear. We used enzyme histostaining and quantitative image analysis to test whether cytokines that stimulate HA synthesis upregulate UGDH activity. Fibroblast-like synoviocytes (FLS, from N = 6 human donors with knee pain) were cultured, freeze-thawed, and incubated for 1 hour with UDP-glucose, NAD+ and nitroblue tetrazolium (NBT) which allows UGDH to generate NADH, and NADH to reduce NBT to a blue stain. Compared to serum-free medium, FLS treated with PDGF showed 3-fold higher UGDH activity and 6-fold higher HA release, but IL-1beta/TGF-beta1 induced 27-fold higher HA release without enhancing UGDH activity. In selected proliferating cells, UGDH activity was lost in the cytosol, but preserved in the nucleus. Cell-free assays led us to discover that diaphorase, a cytosolic enzyme, or glutathione reductase, a nuclear enzyme, was necessary and sufficient for NADH to reduce NBT to a blue formazan dye in a 1-hour timeframe. Primary synovial fibroblasts and transformed A549 fibroblasts showed constitutive diaphorase/GR staining activity that varied according to supplied NADH levels, with relatively stronger UGDH and diaphorase activity in A549 cells. Unilateral knee injury in New Zealand White rabbits (N = 3) stimulated a coordinated increase in synovial membrane UGDH and diaphorase activity, but higher synovial fluid HA in only 2 out of 3 injured joints. UGDH activity (but not diaphorase) was abolished by N-ethyl maleimide, and inhibited by peroxide or UDP-xylose. Our results do not support the hypothesis that UGDH is a rate-liming enzyme for HA synthesis under catabolic inflammatory conditions that can oxidize and inactivate the UGDH active site cysteine. Our novel data suggest a model where UGDH activity is controlled by a redox switch, where intracellular peroxide inactivates, and high glutathione and diaphorase promote UGDH activity by maintaining the active site cysteine in a reduced state, and by recycling NAD+ from NADH.


Asunto(s)
Sinoviocitos , Animales , Cisteína/metabolismo , Fibroblastos/metabolismo , Formazáns , Glucosa/farmacología , Glucosa Deshidrogenasas/metabolismo , Glutatión/metabolismo , Glutatión Reductasa/metabolismo , Humanos , Ácido Hialurónico/metabolismo , Ácido Hialurónico/farmacología , Maleimidas , NAD/metabolismo , Nitroazul de Tetrazolio , Oxidación-Reducción , Peróxidos , Conejos , Sinoviocitos/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Uridina Difosfato/metabolismo , Uridina Difosfato Glucosa Deshidrogenasa/química , Uridina Difosfato Glucosa Deshidrogenasa/metabolismo , Xilosa
10.
Biochem Biophys Res Commun ; 405(4): 538-44, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21256824

RESUMEN

Alternatively activated macrophages have been implicated in the therapeutic activity of biodegradable chitosan on wound healing, however, the mechanisms of phenotypic differentiation are still unclear.In vitro, macrophages stimulated with high doses of chitosan (≥ 500 µg/mL) were reported to produce low-level markers associated with alternative activation (arginase-1) as well as classical activation (nitric oxide), and to undergo apoptosis. In this study, we tested the hypothesis that 40 kDa biodegradable chitosan (5-500 µg/mL) is sufficient to polarize mouse bone marrow-derived macrophages (BMDM) in vitro to an alternatively activated phenotype. Control cultures were stimulated with IL-4 (alternative activation), IFN-γ/LPS (classical activation), 1 µm diameter latex beads (phagocytosis), or left untreated. After 48 h of in vitro exposure, BMDM phagocytosed fluorescent chitosan particles or latex beads, and remained viable and metabolically active, although some cells detached with increasing chitosan and latex bead dosage. Arginase-1 was over 100-fold more strongly induced by IL-4 than by chitosan, which induced only sporadic and weak arginase-1 activity over untreated BMDM, and no nitric oxide. IFN-γ/LPS stimulated nitric oxide production and arginase-1 activity and high concentrations of inflammatory cytokines (IL-6, IL-1ß, TNF-α, MIP-1α/MIP-1ß), while latex beads stimulated nitric oxide and not arginase-1 activity. Chitosan or latex bead exposure, but not IL-4, tended to promote the release of several chemokines (MIP-1α/ß, GM-CSF, RANTES, IL-1ß), while all treatments promoted MCP-1 release. These data show that chitosan phagocytosis is not sufficient to polarize BMDM to the alternative or the classical pathway, suggesting that biodegradable chitosan elicits alternatively activated macrophages in vivo through indirect mechanisms.


Asunto(s)
Quimiocinas/metabolismo , Quitosano/farmacología , Activación de Macrófagos , Macrófagos/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Animales , Arginasa/metabolismo , Medios de Contraste/farmacología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Microesferas
11.
Cartilage ; 12(2): 237-250, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-30569762

RESUMEN

OBJECTIVE: Previous studies have shown that intrinsic behavior of subchondral bone marrow stem cells (BMSCs) is influenced by donors and locations. To understand the variability in cartilage repair outcomes following bone marrow stimulation, we tested the hypothesis that in vivo cartilage repair correlates with in vitro biological properties of BMSCs using a rabbit model. METHODS: Full-thickness cartilage defects were created in the trochlea and condyle in one knee of skeletally mature New Zealand White rabbits (n = 8) followed by microdrilling. Three-week repair tissues were analyzed by macroscopic International Cartilage Repair Society (ICRS) scores, O'Driscoll histological scores, and Safranin-O (Saf-O) and type-II collagen (Coll-II) % stain. BMSCs isolated from contralateral knees were assessed for cell yield, surface marker expression, CFU-f, %Saf-O, and %Coll-II in pellet culture followed by correlation analyses with the above cartilage repair responses. RESULTS: In vivo cartilage repair scores showed strong, positive correlation with cell number, clonogenic, chondrogenic, and matrix production (Coll-II, GAG) potential of in vitro TGF-ßIII stimulated BMSC cultures. Trochlear repair showed clear evidence of donor dependency and strong correlation was observed for interdonor variation in repair and the above in vitro properties of trochlear BMSCs. Correlation analyses indicated that donor- and location-dependent variability observed in cartilage repair can be attributed to variation in the properties of BMSCs in underlying subchondral bone. CONCLUSION: Variation in cell number, clonogenic, chondrogenic, and matrix production potential of BMSCs correlated with repair response observed in vivo and appear to be responsible for interanimal variability as well as location-dependent repair.


Asunto(s)
Médula Ósea , Matriz Ósea/citología , Cartílago Articular/citología , Condrogénesis/fisiología , Células Madre Mesenquimatosas/fisiología , Animales , Artroplastia Subcondral , Matriz Ósea/cirugía , Huesos , Cartílago Articular/cirugía , Recuento de Células , Células Cultivadas , Colágeno Tipo II/metabolismo , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/metabolismo , Miembro Posterior , Trasplante de Células Madre Mesenquimatosas , Conejos
12.
Crit Care Explor ; 3(3): e0374, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33786450

RESUMEN

OBJECTIVES: Since the beginning of the coronavirus disease 2019 pandemic, hundreds of thousands of patients have been treated in ICUs across the globe. The severe acute respiratory syndrome-associated coronavirus 2 virus enters cells via the angiotensin-converting enzyme 2 receptor and activates several distinct inflammatory pathways, resulting in hematologic abnormalities and dysfunction in respiratory, cardiac, gastrointestinal renal, endocrine, dermatologic, and neurologic systems. This review summarizes the current state of research in coronavirus disease 2019 pathophysiology within the context of potential organ-based disease mechanisms and opportunities for translational research. DATA SOURCES: Investigators from the Research Section of the Society of Critical Care Medicine were selected based on expertise in specific organ systems and research focus. Data were obtained from searches conducted in Medline via the PubMed portal, Directory of Open Access Journals, Excerpta Medica database, Latin American and Caribbean Health Sciences Literature, and Web of Science from an initial search from December 2019 to October 15, 2020, with a revised search to February 3, 2021. The medRxiv, Research Square, and clinical trial registries preprint servers also were searched to limit publication bias. STUDY SELECTION: Content experts selected studies that included mechanism-based relevance to the severe acute respiratory syndrome-associated coronavirus 2 virus or coronavirus disease 2019 disease. DATA EXTRACTION: Not applicable. DATA SYNTHESIS: Not applicable. CONCLUSIONS: Efforts to improve the care of critically ill coronavirus disease 2019 patients should be centered on understanding how severe acute respiratory syndrome-associated coronavirus 2 infection affects organ function. This review articulates specific targets for further research.

13.
Connect Tissue Res ; 51(3): 216-23, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20053130

RESUMEN

Primary chondrocytes cultured in agarose can escape the gel, accumulate at the interface between agarose and the culture medium, and form an outgrowing tissue. These outgrowths can appear as voluminous cartilage-like nodules that have never been previously investigated. In the present study, bovine articular chondrocytes from three age groups (fetal, young adult, aged) were seeded and cultured in agarose to test the hypothesis that hyaline-like cartilage outgrowths develop at the interface by appositional growth, in an age-dependant manner. Macroscopic appearance, cell content, cell division, cytoskeletal morphology, and extracellular matrix (ECM) composition were analyzed. Fetal chondrocytes produced a fibrous interfacial tissue while aged chondrocytes produced ECM-poor cell clusters. In contrast young adult chondrocytes produced large cartilaginous outgrowths, rich in proteoglycan and collagen II, where cells in the central region displayed a chondrocyte morphology. Cell proliferation was confined to the peripheral edge of these outgrowths, where elongated cell morphology, cell-cell contacts, and cell extensions toward the culture medium were seen. Thus these voluminous cartilaginous outgrowths formed in an appositional growth process and only for donor chondrocytes from young adult animals. This system offers an interesting ability to proliferate chondrocytes in a manner that results in a chondrocyte morphology and a cartilaginous ECM in central regions of the outgrowing tissue. It also provides an in vitro model system to study neocartilage appositional growth.


Asunto(s)
Cartílago Articular/citología , Condrocitos/citología , Envejecimiento/fisiología , Animales , Cartílago Articular/crecimiento & desarrollo , Bovinos , Recuento de Células , Movimiento Celular/fisiología , Proliferación Celular , Células Cultivadas , Condrocitos/fisiología , Citoesqueleto/fisiología , Citoesqueleto/ultraestructura , ADN/análisis , ADN/biosíntesis , Matriz Extracelular/metabolismo , Feto/citología , Geles , Antígeno Ki-67/metabolismo , Masculino , Sefarosa , Ingeniería de Tejidos
14.
J Tissue Eng Regen Med ; 13(4): 599-611, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30706995

RESUMEN

Bone-marrow stimulation (BMS) improves knee-joint function but elicits incomplete repair. Liquid chitosan (CS)-glycerol phosphate/blood clots have been shown to improve BMS-based cartilage repair. Platelet-rich-plasma (PRP)-a rich source of growth factors and cytokines-improves recruitment and chondrogenic potential of subchondral mesenchymal stem cells. We hypothesised that repair response in a rabbit chronic-defect model will improve when freeze-dried CS/PRP is used to augment BMS. Bilateral trochlear defects created in New Zealand white rabbits were allowed to progress to a chronic stage over 4 weeks. Chronic defects were debrided and treated by BMS in second surgery, then augmented with PRP (BMS + PRP) or freeze-dried CS/PRP implants (BMS + CS/PRP). The quality of 8-week repair tissue was assessed by macroscopic, histological, and micro computed tomography (Micro-CT) analysis. ICRS macroscopic scores indicated fibrocartilaginous or fibrous repair in control defects that were improved in the BMS + CS/PRP group. An overall improvement in repair in BMS + CS/PRP group was further confirmed by higher O'Driscoll scores, %Saf-O and %Coll-II values. Micro-CT analysis of subchondral bone indicated ongoing remodelling with repair still underway. Quality and quantity of cartilage repair was improved when freeze-dried CS/PRP implants were used to augment BMS in a chronic defect model.


Asunto(s)
Médula Ósea/patología , Cartílago Articular/patología , Quitosano/farmacología , Liofilización , Inyecciones , Plasma Rico en Plaquetas/química , Prótesis e Implantes , Cicatrización de Heridas , Animales , Médula Ósea/efectos de los fármacos , Remodelación Ósea , Cartílago Articular/diagnóstico por imagen , Cartílago Articular/efectos de los fármacos , Cartílago Articular/cirugía , Modelos Animales de Enfermedad , Femenino , Inflamación/patología , Proyectos Piloto , Implantación de Prótesis , Conejos , Cicatrización de Heridas/efectos de los fármacos
15.
Biomed Mater Eng ; 30(4): 349-364, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31476140

RESUMEN

BACKGROUND: Platelet-rich plasma (PRP) has been used to solubilize freeze-dried chitosan (CS) formulations to form injectable implants for tissue repair. OBJECTIVE: To determine whether the in vitro performance of the formulations depends on the type of PRP preparation used to solubilize CS. METHODS: Formulations containing 1% (w/v) CS with varying degrees of deacetylation (DDA 80.5-84.8%) and number average molar mass (Mn 32-55 kDa), 1% (w/v) trehalose and 42.2 mM calcium chloride were freeze-dried. Seven different PRP preparations were used to solubilize the formulations. Controls were recalcified PRP. RESULTS: CS solubilization was achieved with all PRP preparations. CS-PRP formulations were less runny than their corresponding PRP controls. All CS-PRP formulations had a clotting time below 9 minutes, assessed by thromboelastography, while the leukocyte-rich PRP controls took longer to coagulate (>32 min), and the leukocyte-reduced PRP controls did not coagulate in this dynamic assay. In glass culture tubes, all PRP controls clotted, expressed serum and retracted (43-82% clot mass lost) significantly more than CS-PRP clots (no mass lost). CS dispersion was homogenous within CS-PRP clots. CONCLUSIONS: In vitro performance of the CS-PRP formulations was comparable for all types of PRPs assessed.


Asunto(s)
Materiales Biocompatibles/química , Quitosano/química , Plasma Rico en Plaquetas/química , Materiales Biocompatibles/administración & dosificación , Coagulación Sanguínea , Quitosano/administración & dosificación , Liofilización , Humanos , Inyecciones , Masculino , Prótesis e Implantes , Solubilidad , Cicatrización de Heridas
16.
Colloids Surf B Biointerfaces ; 175: 596-605, 2019 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-30580150

RESUMEN

Biomaterials are frequently evaluated for pro-coagulant activity but usually in the presence of microparticles (MPs), cell-derived vesicles in blood plasma whose phospholipid surfaces allow coagulation factors to set up as functional assemblies. We tested the hypothesis that synthetic anionic surfaces can catalyze burst thrombin activation in human blood plasma in the absence of MPs. In a thromboelastography (TEG) assay with plastic sample cups and pins, recalcified human citrated platelet-poor plasma spontaneously burst-coagulated but with an unpredictable clotting time whereas plasma depleted of MPs by ultracentrifugation failed to coagulate. Coagulation of MP-depleted plasma was restored in a dose-dependent manner by glass microbeads, hydroxyapatite nanoparticles (HA NPs), and carboxylic acid-containing anionic nanocoatings of TEG cups and pins (coated by glow-discharge plasma-polymerized ethylene containing oxygen, L-PPE:O with 4.4 and 6.8 atomic % [COOH]). Glass beads lost their pro-coagulant activity in MP-depleted plasma after their surfaces were nanocoated with hydrophobic plasma-polymerized hexamethyl disiloxane (PP-HMDSO). In FXII-depleted MP-depleted plasma, glass microbeads failed to induce coagulation, however, FXIa was sufficient to induce coagulation in a dose-dependent manner, with no effect of glass beads. These data suggest that anionic surfaces of crystalline, organic, and amorphous solid synthetic materials catalyze explosive thrombin generation in MP-depleted plasma by activating the FXII-dependent intrinsic contact pathway. The data also show that microparticles are pro-coagulant surfaces whose activity has been largely overlooked in many coagulation studies to-date. These results suggest a possible mechanism by which anionic biomaterial surfaces induce bone healing by contact osteogenesis, through fibrin clot formation in the absence of platelet activation.


Asunto(s)
Materiales Biocompatibles/farmacología , Coagulación Sanguínea/efectos de los fármacos , Micropartículas Derivadas de Células/química , Durapatita/farmacología , Nanopartículas/química , Siloxanos/farmacología , Aniones , Materiales Biocompatibles/química , Plaquetas/citología , Plaquetas/efectos de los fármacos , Durapatita/química , Factor XII/metabolismo , Vidrio/química , Humanos , Membranas Artificiales , Siloxanos/química , Propiedades de Superficie , Tromboelastografía , Trombina/metabolismo
17.
Anal Biochem ; 378(2): 158-65, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18457651

RESUMEN

We report an innovative at-line method to monitor concentration of bioactive antibody (i.e., antibody with conserved antigen-binding activity) secreted during bioreactor culture by the use of surface plasmon resonance (SPR)-based biosensor technology. In a first series of experiments, conditions for SPR-based measurements were validated off-line by monitoring bioactive antibody concentration in conditioned medium from 500-ml baffled flask hybridoma cell cultures. A fully automated experimental setup in which the SPR-based biosensor was harnessed to a bioreactor was then used at-line to monitor the concentration of bioactive antibody produced in a 3.5-L bioreactor. Quantitative SPR measurements performed both at-line and off-line were in excellent agreement with quantitative Western blotting followed by densitometry analyses. Thus, our experimental study confirms that SPR biosensors can be applied to at-line quantification of correctly folded proteins that are secreted by cells cultured in a bioreactor. Our experimental approach represents a novel and robust analytical strategy to be applied to the control and optimization of the production of bioactive secreted proteins.


Asunto(s)
Anticuerpos Monoclonales/análisis , Reactores Biológicos , Epítopos/análisis , Resonancia por Plasmón de Superficie/métodos , Animales , Técnicas Biosensibles , Western Blotting , Línea Celular , Densitometría , Hibridomas , Ligandos , Masculino , Antígeno Prostático Específico/metabolismo , Reproducibilidad de los Resultados
18.
Cartilage ; 9(4): 378-390, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29156978

RESUMEN

OBJECTIVE: Bone marrow stimulation procedures initiate repair by fracturing or drilling subchondral bone at base of cartilaginous defect. Earlier studies have shown that defect location and animal age affect cartilage repair outcome, suggesting a strong influence of structural and biological characteristics of subchondral bone. Here, we analyzed comprehensive biological characteristics of bone marrow progenitor cells (BMPCs) in subchondral bone of young and old rabbit condyle and trochlea. We tested the hypothesis that in vitro biological properties of BMPCs are influenced by location, age of donor and method of their isolation. DESIGN: In vitro biological properties, including cell yield, colony-forming unit fibroblasts (CFU-f), surface marker expression, and differentiation potential were determined. Comparisons were carried out between trochlea versus condyle and epiphyseal versus metaphyseal bone using old ( N = 5) and young animal knees ( N = 8) to generate collagenase and explant-derived BMPC cultures. RESULTS: CFU-f, cell yield, expression of stem cell markers, and osteogenic differentiation were significantly superior for younger animals. Trochlear subchondral bone yielded the most progenitors with the highest clonogenic potential and cartilaginous matrix expression. Trochlear collagenase-derived BMPCs had higher clonogenic capacity than explant-derived ones. Epiphyseal cells generated a larger chondrogenic pellet mass than metaphyseal-derived BMPCs. All older pellet cultures and one non-responder young rabbit failed to accumulate glycosaminoglycans (GAGs). CONCLUSION: Taken together, these results suggest that properties intrinsic to subchondral progenitors could significantly influence cartilage repair potential, and could partly explain variability in cartilage repair outcomes using same cartilage repair approach.


Asunto(s)
Células de la Médula Ósea/fisiología , Huesos/citología , Células Madre/fisiología , Cúbito/citología , Animales , Condrogénesis , Ensayo de Unidades Formadoras de Colonias , Osteogénesis , Conejos
19.
Stem Cells Dev ; 16(3): 489-502, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17610379

RESUMEN

The mesenchymal stem cell (MSC) is a critical element in tissue repair and regeneration. Its ability to differentiate into multiple connective tissue cell types and to self-renew has made it a prime candidate in regenerative medicine strategies. Currently, the environmental cues responsible for in situ recruitment and control of MSC distribution at repair sites are not entirely revealed and in particular the role of extracellular matrix (ECM) proteins as motogenic factors has not been studied. Here we have used a standardized transmembrane chemotaxis assay to assess the chemotactic and haptotactic potential of fibronectin, vitronectin, and collagen type 1 on MSCs from both rabbit and human origin. The use of both cell types was based in part on the widespread use of rabbit models for musculoskeletal-related tissue engineering and repair models and their unknown correspondence to human in terms of MSC migration. The optimized assay yielded a greatly increased chemotactic response toward known factors such as platelet-derived growth factor-BB (PDGF)-BB compared to previous studies. Our primary finding was that all three ECM proteins tested (fibronectin, vitronectin, and collagen I) induced significant motogenic activity, in both soluble and insoluble forms, for both rabbit and human MSCs. These results suggest that ECM proteins could play roles as significant as cytokines in the recruitment of pluripotential repair cells wound and tissue repair sites. Furthermore, designed ECM coatings of scaffolds or implants could provide a new tool to control both cell influx and outflux from the scaffold post-implantation. Finally, the similarity of motogenic behavior of both rabbit and human cells suggests the rabbit is a reliable model for assessing MSC recruitment in repair and regeneration strategies.


Asunto(s)
Células de la Médula Ósea/fisiología , Movimiento Celular/fisiología , Colágeno Tipo I/metabolismo , Fibronectinas/metabolismo , Mesodermo/citología , Células Madre/fisiología , Vitronectina/metabolismo , Animales , Becaplermina , Bioensayo , Células de la Médula Ósea/citología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Humanos , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas c-sis , Conejos , Células Madre/citología
20.
Cartilage ; 8(4): 417-431, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28934884

RESUMEN

Objective This study tested the hypothesis that presolidified chitosan-blood implants are retained in subchondral bone channels perforated in critical-size sheep cartilage defects, and promote bone repair and hyaline-like cartilage resurfacing versus blood implant. Design Cartilage defects (10 × 10 mm) with 3 bone channels (1 drill, 2 Jamshidi biopsy, 2 mm diameter), and 6 small microfracture holes were created bilaterally in n = 11 sheep knee medial condyles. In one knee, 10 kDa chitosan-NaCl/blood implant (presolidified using recombinant factor VIIa or tissue factor), was inserted into each drill and Jamshidi hole. Contralateral knee defects received presolidified whole blood clot. Repair tissues were assessed histologically, biochemically, biomechanically, and by micro-computed tomography after 1 day ( n = 1) and 6 months ( n = 10). Results Day 1 defects showed a 60% loss of subchondral bone plate volume fraction along with extensive subchondral hematoma. Chitosan implant was resident at day 1, but had no effect on any subsequent repair parameter compared with blood implant controls. At 6 months, bone defects exhibited remodeling and hypomineralized bone repair and were partly resurfaced with tissues containing collagen type II and scant collagen type I, 2-fold lower glycosaminoglycan and fibril modulus, and 4.5-fold higher permeability compared with intact cartilage. Microdrill holes elicited higher histological ICRS-II overall assessment scores than Jamshidi holes (50% vs. 30%, P = 0.041). Jamshidi biopsy holes provoked sporadic osteonecrosis in n = 3 debrided condyles. Conclusions Ten kilodalton chitosan was insufficient to improve repair. Microdrilling is a feasible subchondral marrow stimulation surgical approach with the potential to elicit poroelastic tissues with at least half the compressive modulus as intact articular cartilage.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA