Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biomacromolecules ; 22(2): 275-288, 2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33332959

RESUMEN

Vascularizing printed tissues is a critical challenge in bioprinting. While protein-based hydrogel bioinks have been successfully used to bioprint microvasculature, their compositions are ill-defined and subject to batch variation. Few studies have focused on engineering proangiogenic bioinks with defined properties to direct endogenous microvascular network formation after printing. Here, a peptide-functionalized alginate hydrogel bioink with defined mechanical, rheological, and biochemical properties is developed for direct bioprinting of microvascularized tissues. An integrin-binding peptide (RGD) and a vascular endothelial growth factor-mimetic peptide with a protease-sensitive linker are conjugated onto a biodegradable alginate to synergistically promote vascular morphogenesis and capillary-scale endothelial tube formation. Partial ionic crosslinking before printing converts the otherwise unprintable hydrogel into a viscoelastic bioink with excellent printability and cytocompatibility. We use the bioink to fabricate a compartmentalized vascularized tissue construct, wherein we observe pericyte-endothelial cell colocalization and angiogenic sprouting across a tissue interface, accompanied by deposition of fibronectin and collagen in vascular and tissue components, respectively. This study provides a tunable and translational "off-the-shelf" hydrogel bioink with defined composition for vascularized bioprinting.


Asunto(s)
Bioimpresión , Hidrogeles , Microvasos , Impresión Tridimensional , Ingeniería de Tejidos , Andamios del Tejido , Factor A de Crecimiento Endotelial Vascular
2.
Ann Plast Surg ; 80(6S Suppl 6): S410-S417, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29746273

RESUMEN

BACKGROUND: Biomedical devices are implanted into mammalian soft tissues to improve, monitor, or restore form or function. The utility of these implants is limited by the subsequent foreign body response (FBR), beginning with inflammation and terminating in a collagen envelope around the device, known as the capsule. This capsule then can contract and distort the shape of the device or limit its effectiveness in interacting with the surrounding host tissues. In the current study, we investigated the effect of therapeutic collagen-coated silicone discs in a rat model of the FBR. METHODS: A 3-dimensional printed mold was used to fabricate collagen-coated silicone discs incorporating 3 therapeutic agents: colchicine, a function-blocking antibody against interleukin 8 (IL-8) receptor B, and a powerful anti-inflammatory steroid, dexamethasone. Discs were implanted submuscularly into a well-characterized rat model of the FBR and evaluated for inflammatory response, fibrotic development, and cytokine release. RESULTS: Coated silicone discs exhibited reduced collagen deposition and little to no foreign body giant cells at the host-silicone interface when compared with the silicone-only group. Therapeutic hydrogels demonstrate a significant decrease in cellular infiltration into the coatings over the 2-week time point in contrast to therapeutic-free hydrogel coatings. Cytokine analysis revealed significant differences between therapeutic-free and therapeutic-containing coatings when compared with silicone-only controls. Levels of IL-1ß, IL-6, monocyte chemotactic protein 1, and macrophage inflammatory protein 3α were affected 48 hours after implantation, while differences in IL-18, growth-regulated oncogene/keratinocyte chemoattractant, and macrophage inflammatory protein 3α were observed 1 week after implantation. CONCLUSIONS: By utilizing the host's innate immune response, our engineered hydrogel coatings delivered therapeutic moieties directly to the implant microenvironment, thus delaying the FBR up to 2 weeks.


Asunto(s)
Antiinflamatorios/uso terapéutico , Colágeno/uso terapéutico , Reacción a Cuerpo Extraño/prevención & control , Hidrogeles/uso terapéutico , Prótesis e Implantes/efectos adversos , Animales , Biomarcadores/metabolismo , Citocinas/metabolismo , Reacción a Cuerpo Extraño/diagnóstico , Reacción a Cuerpo Extraño/inmunología , Reacción a Cuerpo Extraño/patología , Ratas , Ratas Sprague-Dawley , Siliconas/efectos adversos , Resultado del Tratamiento
3.
Am J Physiol Cell Physiol ; 313(3): C314-C326, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28701358

RESUMEN

Connexin-based therapeutics have shown the potential for therapeutic efficacy in improving wound healing. Our previous work demonstrated that the connexin43 (Cx43) mimetic peptide juxtamembrane 2 (JM2) reduced the acute inflammatory response to a submuscular implant model by inhibiting purinergic signaling. Given the prospective application in improving tissue-engineered construct tolerance that these results indicated, we sought to determine the mechanism of action for JM2 in the present study. Using confocal microscopy, a gap-FRAP cell communication assay, and an ethidium bromide uptake assay of hemichannel function we found that the peptide reduced cell surface Cx43 levels, Cx43 gap junction (GJ) size, GJ communication, and hemichannel activity. JM2 is based on the sequence of the Cx43 microtubule binding domain, and microtubules have a confirmed role in intracellular trafficking of Cx43 vesicles. Therefore, we tested the effect of JM2 on Cx43-microtubule interaction and microtubule polymerization. We found that JM2 enhanced Cx43-microtubule interaction and that microtubule polymerization was significantly enhanced. Taken together, these data suggest that JM2 inhibits trafficking of Cx43 to the cell surface by promoting irrelevant microtubule polymerization and thereby reduces the number of hemichannels in the plasma membrane available to participate in proinflammatory purinergic signaling. Importantly, this work indicates that JM2 may have therapeutic value in the treatment of proliferative diseases such as cancer. We conclude that the targeted action of JM2 on Cx43 channels may improve the tolerance of implanted tissue-engineered constructs against the innate inflammatory response.


Asunto(s)
Antiinflamatorios/inmunología , Antiinflamatorios/farmacología , Conexina 43/inmunología , Microtúbulos/efectos de los fármacos , Microtúbulos/inmunología , Péptidos/farmacología , Conexina 43/antagonistas & inhibidores , Células HeLa , Humanos , Péptidos/síntesis química , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/inmunología
4.
Microsc Microanal ; 23(4): 859-871, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28712382

RESUMEN

Vascular stenosis, the abnormal narrowing of blood vessels, arises from defective developmental processes or atherosclerosis-related adult pathologies. Stenosis triggers a series of adaptive cellular responses that induces adverse remodeling, which can progress to partial or complete vessel occlusion with numerous fatal outcomes. Despite its severity, the cellular interactions and biophysical cues that regulate this pathological progression are poorly understood. Here, we report the design and fabrication of a three-dimensional (3D) in vitro system to model vascular stenosis so that specific cellular interactions and responses to hemodynamic stimuli can be investigated. Tubular cellularized constructs (cytotubes) were produced, using a collagen casting system, to generate a stenotic arterial model. Fabrication methods were developed to create cytotubes containing co-cultured vascular cells, where cell viability, distribution, morphology, and contraction were examined. Fibroblasts, bone marrow primary cells, smooth muscle cells (SMCs), and endothelial cells (ECs) remained viable during culture and developed location- and time-dependent morphologies. We found cytotube contraction to depend on cellular composition, where SMC-EC co-cultures adopted intermediate contractile phenotypes between SMC- and EC-only cytotubes. Our fabrication approach and the resulting artery model can serve as an in vitro 3D culture system to investigate vascular pathogenesis and promote the tissue engineering field.


Asunto(s)
Constricción Patológica/patología , Modelos Teóricos , Enfermedades Vasculares/patología , Enfermedades Vasculares/fisiopatología , Animales , Comunicación Celular , Células Endoteliales/fisiología , Fibroblastos/fisiología , Miocitos del Músculo Liso/fisiología , Ratas , Ingeniería de Tejidos/métodos
5.
Ann Plast Surg ; 74(3): 371-5, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25668498

RESUMEN

To evaluate the anastomotic potential of prevascular tissue constructs generated from scaffold-free self-assembly of human endothelial and fibroblast cells, tissue constructs were implanted into athymic mice and immune-competent rats. Analysis of xenografts placed into hind limb muscle defects showed vascular anastomotic activity by 3 days after implantation and persisting for 2 weeks. Integration of the implanted prevascular tissue constructs with the host circulatory system was evident from presence of red blood cells in the implant as early as 3 days after implantation. Additionally, analysis of 3-day xenografts in the rat model showed activation of skeletal muscle satellite cells based on Pax-7 and MyoD expressions. We conclude that prevascular tissue constructs generated from scaffold-free self-assembly of human endothelial and fibroblast cells are a promising tool to provide both vascular supply and satellite cell activation toward the resolution of skeletal muscle injury.


Asunto(s)
Regeneración Tisular Dirigida/métodos , Músculo Esquelético/lesiones , Neovascularización Fisiológica , Traumatismos de los Tejidos Blandos/cirugía , Andamios del Tejido , Animales , Células Cultivadas , Humanos , Masculino , Ratones , Ratones Desnudos , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/patología , Músculo Esquelético/fisiología , Ratas , Ratas Sprague-Dawley , Células Satélite del Músculo Esquelético/patología , Células Satélite del Músculo Esquelético/fisiología , Traumatismos de los Tejidos Blandos/patología , Traumatismos de los Tejidos Blandos/fisiopatología , Resultado del Tratamiento , Cicatrización de Heridas
6.
Dev Biol ; 374(2): 345-56, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23261934

RESUMEN

Fibrous development of the extracellular matrix (ECM) of cardiac valves is necessary for proper heart function. Pathological remodeling of valve ECM is observed in both pediatric and adult cardiac disorders. It is well established that intracardiac hemodynamics play a significant role in the morphogenesis of cardiovascular tissues. However, the mechanisms that transduce mechanical forces into morphogenetic processes are not well understood. Here, we report the development of a three-dimensional, in vitro culture system that allows for culture of embryonic valve tissue under specific pulsatile flow conditions. This system was used to investigate the role that fluid flow plays in fibrous ECM expression during valve formation and to test the underlying cellular mechanisms that regulate this mechanotransduction. When cultured under pulsatile flow, developing valve tissues upregulated fibrous ECM expression at both the transcript and protein levels in comparison to no-flow controls. Flow-cultured valve tissues also underwent morphological development, as cushions elongated into leaflet-like structures that were absent in no-flow controls. Furthermore, rhoA, a member of the cytoskeletal actin-regulating GTPase family of proteins, was upregulated and activated by flow culture. Inhibition of the downstream rhoA effector kinase, ROCK, blocked flow-driven fibrous ECM accumulation and tissue stiffening, while the addition of lysophosphatidic acid (LPA), a rhoA activator, stimulated fibrous ECM deposition and tissue stiffening. These results support a prominent role for the rhoA pathway in the mechanotransduction of hemodynamic forces during fibrous remodeling of developing valve tissue. Our results also point to a potential link between regulation of the actinomyosin cytoskeleton and fibrous ECM synthesis in cardiovascular tissues.


Asunto(s)
Proteínas Aviares/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Válvulas Cardíacas/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Amidas/farmacología , Animales , Proteínas Aviares/genética , Western Blotting , Embrión de Pollo , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/genética , Perfilación de la Expresión Génica , Válvulas Cardíacas/embriología , Lisofosfolípidos/farmacología , Mecanotransducción Celular/genética , Mecanotransducción Celular/fisiología , Microscopía de Fuerza Atómica , Microscopía Confocal , Piridinas/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estrés Mecánico , Tenascina/genética , Factores de Tiempo , Técnicas de Cultivo de Tejidos/métodos , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/genética
7.
Ann Plast Surg ; 73(4): 451-60, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25046663

RESUMEN

Clinicians and investigators have been implanting biomedical devices into patients and experimental animals for centuries. There is a characteristic complex inflammatory response to the presence of the biomedical device with diverse cell signaling, followed by migration of fibroblasts to the implant surface and the eventual walling off of the implant in a collagen capsule. If the device is to interact with the surrounding tissues, the collagen envelope will eventually incapacitate the device or myofibroblasts can cause capsular contracture with resulting distortion, migration, or firmness. This review analyzes the various tactics used in the past to modify or control capsule formation with suggestions for future investigative approaches.


Asunto(s)
Reacción a Cuerpo Extraño/prevención & control , Prótesis e Implantes/efectos adversos , Implantes Absorbibles , Materiales Biocompatibles , Implantes de Mama/efectos adversos , Reacción a Cuerpo Extraño/etiología , Reacción a Cuerpo Extraño/inmunología , Humanos , Contractura Capsular en Implantes/etiología , Contractura Capsular en Implantes/inmunología , Contractura Capsular en Implantes/prevención & control , Implantación de Prótesis/métodos
8.
Methods Mol Biol ; 2367: 235-247, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32789778

RESUMEN

With the realization that mechanical forces mediate many biological processes and contribute to disease progression, researchers are focusing on developing new methods to understand the role of mechanotransduction in biological systems. Despite recent advances in stretching devices that analyze the effects of mechanical strain in vitro, there are still possibilities to develop new equipment. For example, many of these devices tend be expensive, whereas few have been designed to assess the effects of mechanical strain driven by the extracellular matrix (ECM) to epithelial cell monolayers and to cell-cell adhesion. In this chapter, we introduce a cost-efficient, user-friendly, 3D-printed stretching device that can be used to test the effects of mechanical strain on cultured epithelial cells. Evaluation of the device using speckle-tracking shows homogeneous strain distribution along the horizontal plane of membranes at 2.5% and 5% strains, supporting the reliability of the device. Since cell-cell junctions are mechanosensitive protein complexes, we hereby used this device to examine effects on cell-cell adhesion. For this, we used colon epithelial Caco2 cell monolayers that well-differentiate in culture and form mature adherens junctions. Subjecting Caco2 cells to 2.5% and 5% strain using our device resulted in significant reduction in the localization of the core adherens junction component E-cadherin at areas of cell-cell contact and its increased translocation to the cytoplasm, which in agreement with other methodologies showing that increased ECM-driven strain negatively affects cell-cell adhesion. In summary, we here present a new, cost-effective, homemade device that can be reliably used to examine effects of mechanical strain on epithelial cell monolayers and cell-cell adhesion, in vitro.


Asunto(s)
Camillas , Uniones Adherentes , Células CACO-2 , Cadherinas , Adhesión Celular , Células Epiteliales , Humanos , Mecanotransducción Celular , Impresión Tridimensional , Reproducibilidad de los Resultados
9.
IEEE Open J Eng Med Biol ; 2: 187-197, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34734189

RESUMEN

GOAL: The impact of hyperthermia (HT) method on tumor drug uptake with thermosensitive liposomes (TSL) is not well understood. METHODS: We created realistic three-dimensional (3-D) computer models that simulate TSL-encapsulated doxorubicin (TSL-DOX) delivery in mouse tumors with three HT methods (thermistor probe (T), laser (L) and water bath (WB), at 15 min and 60 min HT duration), with corroborating in vivo studies. RESULTS: Average computer model-predicted tumor drug concentrations (µg/g) were 8.8(T, 15 min), 21.0(T, 60 min), 14.1(L, 15 min), 25.2(L, 60 min), 9.4(WB, 15 min), and 8.7(WB, 60 min). Tumor fluorescence was increased by 2.6 × (T) and 1.6 × (L) when HT duration was extended from 15 to 60 min (p < 0.05), with no increase for WB HT. Pharmacokinetic analysis confirmed that water bath HT causes rapid depletion of encapsulated TSL-DOX in systemic circulation due to the large heated tissue volume. CONCLUSIONS: Untargeted large volume HT causes poor tumor drug uptake from TSL.

10.
Am Surg ; 76(1): 15-9, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20135933

RESUMEN

Skin flora immobilization technology is similar in efficacy to Iodine-Povidone in healthy volunteers. We did a prospective study in a university clinic with 60 healthy volunteers. Right inguinal skin area on healthy volunteers was used to compare the antimicrobial properties of cyanoacrylate sealant (FloraSeal, Adhesion Biomedical, Wyomissing, PA) versus standard surgical preparation Povidone-iodine (Betadine, Purdue Productions, Stamford, CT). Bacterial counts were measured at different time intervals: 15 minutes, 4 hours, and 24 hours. Bacterial colony forming units were compared between Povidone-iodine and cyanoacrylate sealant. The absolute log reduction was 5.568 for Povidone-iodine (7 absolute CFU); 5.028 for cyanoacrylate (59 absolute CFU); and 5.568 for Povidone-iodine and cyanoacrylate combined (21 absolute CFU). Cyanoacrylate was able to sustain a reduction on bacterial counts at 4 hours and 24 hours of more than 99.8 per cent as compared with the control group. Cyanoacrylate microbial sealant successfully reduces bacterial counts on normal healthy skin. The results were similar to Povidone-iodine alone. We believe this technology may be an excellent means of mitigating incisional surgical site infection by reducing the risk of contamination by skin flora and warrants further testing.


Asunto(s)
Antiinfecciosos Locales/farmacología , Cianoacrilatos/farmacología , Control de Infecciones , Piel/microbiología , Adulto , Recuento de Colonia Microbiana , Femenino , Humanos , Masculino , Povidona Yodada/farmacología , Estudios Prospectivos
11.
Ann Plast Surg ; 64(6): 794-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20407365

RESUMEN

Current use of prosthetic meshes and implants for myofascial reconstruction has been associated with infectious complications, long-term failure, and dissatisfying cosmetic results. Our laboratory has developed a small animal model for ventral hernia repair, which uses progenitor cells isolated from a skeletal muscle biopsy. In the model, progenitor cells are expanded in vitro, seeded onto a nonimmunogenic, novel aligned scaffold of bovine collagen and placed into the defect as a living adjuvant to the innate repair mechanism. The purpose of the current investigation is to examine the feasibility of translating our current model to humans. As a necessary first step we present our study on the efficacy of isolating satellite cells from 9 human donor biopsies. We were able to successfully translate our progenitor cell isolation and culture protocols to a human model with some modifications. Specifically, we have isolated human satellite muscle cells, expanded them in culture, and manipulated these cells to differentiate into myotubes in vitro. Immunohistochemical analysis allowed the characterization of distinct progenitor cell cycle stages and quantification of approximate cell number. Furthermore, isolated cells were tracked via cytoplasmic nanocrystal labeling and observed using confocal microscopy.


Asunto(s)
Trasplante de Células/métodos , Músculo Esquelético/fisiología , Neovascularización Fisiológica/fisiología , Regeneración/fisiología , Células Satélite del Músculo Esquelético/trasplante , Adulto , Anciano , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Femenino , Humanos , Inmunohistoquímica , Masculino , Microscopía Confocal/métodos , Persona de Mediana Edad , Músculo Esquelético/cirugía , Ratas , Procedimientos de Cirugía Plástica/métodos , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/ultraestructura , Sensibilidad y Especificidad , Células Madre/patología , Células Madre/ultraestructura , Ingeniería de Tejidos , Recolección de Tejidos y Órganos , Adulto Joven
12.
Annu Int Conf IEEE Eng Med Biol Soc ; 2020: 5021-5024, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-33019114

RESUMEN

Thermosensitive liposomes (TSL) are nanoparticles that can encapsulate therapeutic drugs, and release those drugs when exposed to hyperthermic temperatures (>40 °C). Combined with localized hyperthermia, TSL enable focused drug delivery. In this study, we created a three-dimensional (3D) computer model for simulating delivery with TSL-encapsulated doxorubicin (TSL-Dox) to mouse tumors. A mouse hind limb was scanned by a 3D scanner and the resulting geometry was imported into finite element modeling software, with a virtual tumor added. Then, heating by a surface probe was simulated. Further, a drug delivery model was coupled to the heat transfer model to simulate drug delivery kinetics. For comparison, experimental studies in gel phantoms and in vivo fluorescence imaging studies in mice carrying lung tumor xenografts were performed. We report the tissue temperature profile, drug concentration profile and compare the experimental studies with the computer model. The thermistor produced very localized heating that resulted in highest drug delivery to regions near the probe. The average tumor temperature was 38.2˚C (range 34.4-43.4˚C), and produced an average tumor drug concentration of 11.8 µg/g (0.3-28.1 µg/g) after 15 min heating, and 25.6 µg/g (0.3-52 µg/g), after 60 min heating. The computer model reproduced the temperature profile compared to phantom experiments (mean error 0.71 °C, range 0.59-1.25 °C), as well as drug delivery profile as compared to in vivo studies. Our results suggest feasibility of using this approach to model drug delivery in preclinical studies with accurate model geometry.


Asunto(s)
Hipertermia Inducida , Liposomas , Animales , Antibióticos Antineoplásicos/uso terapéutico , Simulación por Computador , Sistemas de Liberación de Medicamentos , Ratones
13.
Sci Adv ; 6(28): eaaz5894, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32923589

RESUMEN

Biologically active ligands (e.g., RGDS from fibronectin) play critical roles in the development of chemically defined biomaterials. However, recent decades have shown only limited progress in discovering novel extracellular matrix-protein-derived ligands for translational applications. Through motif analysis of evolutionarily conserved RGD-containing regions in laminin (LM) and peptide-functionalized hydrogel microarray screening, we identified a peptide (a1) that showed superior supports for endothelial cell (EC) functions. Mechanistic studies attributed the results to the capacity of a1 engaging both LM- and Fn-binding integrins. RNA sequencing of ECs in a1-functionalized hydrogels showed ~60% similarities with Matrigel in "vasculature development" gene ontology terms. Vasculogenesis assays revealed the capacity of a1-formulated hydrogels to improve EC network formation. Injectable alginates functionalized with a1 and MMPQK (a vascular endothelial growth factor-mimetic peptide with a matrix metalloproteinase-degradable linker) increased blood perfusion and functional recovery over decellularized extracellular matrix and (RGDS + MMPQK)-functionalized hydrogels in an ischemic hindlimb model, illustrating the power of this approach.


Asunto(s)
Hidrogeles , Factor A de Crecimiento Endotelial Vascular , Animales , Secuencia Conservada , Matriz Extracelular , Ligandos , Péptidos/farmacología
14.
Biores Open Access ; 8(1): 1-15, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30637179

RESUMEN

Vascularization remains a substantial limitation to the viability of engineered tissue. By comparing in vivo vascularization dynamics of a self-assembled prevascular endothelial-fibroblast model to avascular grafts, we explore the vascularization rate limitations in implants at early time intervals, during which tissue hypoxia begins to affect cell viability. Scaffold-free prevascular endothelial-fibroblast constructs (SPECs) may serve as a modular and reshapable vascular bed in replacement tissues. SPECs, fibroblast-only spheroids (FOS), and silicone implants were implanted in 54 Sprague Dawley rats and harvested at 6, 12, and 24 h (n = 5 per time point and implant type). We hypothesized that the primary endothelial networks of the SPECs allow earlier anastomosis and increased vessel formation in the interior of the implant compared to FOS and silicone implants within a 24 h window. All constructs were encapsulated by an endothelial lining at 6 h postimplantation and SPEC internal cords inosculated with the host vascular network by this time point. SPECs had a significantly higher microvascular area fraction and branch/junction density of penetrating cords at 6-12 h compared with other constructs. In addition, SPECs demonstrated perivascular cell recruitment, lumen formation, and network remodeling consistent with vessel maturation at 12-24 h; however, these implants were poorly perfused within our observation window, suggesting poor lumen patency. FOS vascular characteristics (microvessel area and penetrating cord density) increased within the 12-24 h period to represent those of the SPEC implants, suggesting a 12 h latency in host response to avascular grafts compared to prevascular grafts. Knowledge of this temporal advantage in in vitro prevascular network self-assembly as well as an understanding of the current limitations of SPEC engraftment builds on our theoretical temporal model of tissue graft vascularization and suggests a crucial time window, during which technological improvements and vascular therapy can improve engineered tissue survival.

15.
Trends Biotechnol ; 26(4): 173-80, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18295916

RESUMEN

Fibrotic scars deposited during skin wound healing can cause disfiguration and loss of dermal function. Scar differentiation involves inputs from multiple cell types in a predictable and overlapping sequence of cellular events that includes inflammation, migration/proliferation and extracellular matrix deposition. Research into the molecular mechanisms underpinning these processes in embryonic and adult wounds has contributed to the development of a growing number of novel therapeutic approaches for improving scar appearance. This review discusses some of these emerging strategies for shifting the balance of healing from scarring to regeneration in the context of non-pathological wounds. Particular focus is given to potential therapies based on transforming growth factor (TGF)-beta signaling and recent unexpected findings involving targeting of gap junctional connexins. Lessons learned in promoting scarless healing of cutaneous injuries might provide a basis for regenerative healing in other scenarios, such as spinal cord rupture or myocardial infarction.


Asunto(s)
Cicatriz/fisiopatología , Cicatriz/terapia , Piel/patología , Cicatrización de Heridas/fisiología , Adulto , Cicatriz/patología , Colágeno/inmunología , Colágeno/metabolismo , Conexinas/genética , Conexinas/metabolismo , Conexinas/uso terapéutico , Embrión de Mamíferos/inmunología , Embrión de Mamíferos/fisiopatología , Matriz Extracelular/inmunología , Matriz Extracelular/patología , Feto/inmunología , Feto/fisiopatología , Humanos , Inflamación/complicaciones , Inflamación/embriología , Inflamación/inmunología , Inflamación/fisiopatología , Ingeniería de Proteínas , Piel/metabolismo , Piel/fisiopatología , Factor de Crecimiento Transformador beta/agonistas , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/uso terapéutico , Cicatrización de Heridas/efectos de los fármacos
16.
Biomaterials ; 29(14): 2203-16, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18289664

RESUMEN

It is well established that the process of neovascularization or neoangiogenesis is coupled to the development and maturation of bone. Bone marrow stromal cells (BMSCs) or mesenchymal stem cells (MSCs) comprise a heterogeneous population of cells that can be differentiated in vitro into both mesenchymal and non-mesenchymal cell lineages. When both rat BMSCs and quail proepicardia (PEs) were seeded onto a three-dimensional (3-D) tubular scaffold engineered from aligned collagen type I strands and co-cultured in osteogenic media, the maturation and co-differentiation into osteoblastic and vascular cell lineages were observed. In addition, these cells produced abundant mineralized extracellular matrix materials and vessel-like structures. BMSCs were seeded at a density of 2 x 10(6)cells/15 mm tube and cultured in basal media for 3 days. Subsequently, on day 3, PEs were seeded onto the same tubes and the co-culture was continued for another 3, 6 or 9 days either in basal or in osteogenic media. Differentiated cells were subjected to immunohistochemical, cytochemical and biochemical analyses. Phenotypic induction was analyzed at mRNA level by reverse transcriptase quantitative polymerase chain reaction (RT-qPCR). Immunolocalization of key osteogenic and vasculogenic lineage specific markers were examined using confocal scanning laser microscopy. In osteogenic tube cultures, both early and late osteogenic markers were observed and were reminiscent of in vivo expression pattern. Alkaline phosphatase activity and calcium content significantly increased over the observed period of time in osteogenic medium. Abundant interlacing fascicles of QCPN, QH1, isolectin and alpha-smooth muscle actin (alpha-SMA) positive cells were observed in these tube cultures. These cells formed extensive arborizations of nascent capillary-like structures and were seen amidst the developing osteoblasts in osteogenic cultures. The 3-D culture system not only generated de novo vessel-like structures but also augmented the maturation and differentiation of BMSCs into osteoblasts. Thus, this novel co-culture system provides a useful in vitro model to investigate the functional role and effects of neovascularization in the proliferation, differentiation and maturation of BMSC derived osteoblasts.


Asunto(s)
Células de la Médula Ósea/citología , Osteogénesis/fisiología , Pericardio/citología , Células del Estroma/citología , Ingeniería de Tejidos/métodos , Fosfatasa Alcalina/análisis , Fosfatasa Alcalina/metabolismo , Animales , Calcio/análisis , Calcio/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular , Células Cultivadas , Técnicas de Cocultivo , Colágeno Tipo I/química , Inmunohistoquímica , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Codorniz , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Células del Estroma/metabolismo , Factores de Tiempo
17.
J Biomed Mater Res A ; 106(1): 86-94, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28869704

RESUMEN

Collagen has received considerable attention as a biomaterial for tissue engineering because of its low immunogenicity, controllable biodegradation, and ability to influence cell growth and proliferation. Frequently, collagen scaffolds require crosslinking to improve mechanical strength, requiring agents like glutaraldehyde that have high residual cytotoxicity. A novel method for extracting residual glutaraldehyde from crosslinked collagen films with supercritical carbon dioxide (CO2 ) is presented. CO2 is a nontoxic, nonflammable substance that is relatively inert and can be used to process biomaterials at mild pressures and physiologic temperatures. In this work, it was first determined that type I collagen is chemically compatible with both liquid and supercritical CO2 . Treated collagen showed minimal changes in physicochemical properties as determined by differential scanning calorimetry, gel electrophoresis, and circular dichroism. CO2 was subsequently used to extract residual glutaraldehyde from crosslinked collagen films. Glutaraldehyde concentration was reduced by over 95%, from over 20 ppm before treatment to about 1 ppm, in only 1 h. CO2 treatment caused negligible alteration of thermal stability but did significantly increase film stiffness and tensile strength. However, these changes were minor compared to heat-based removal of glutaraldehyde. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 86-94, 2018.


Asunto(s)
Plásticos Biodegradables/química , Dióxido de Carbono/química , Colágeno Tipo I/química , Reactivos de Enlaces Cruzados/química , Glutaral/química , Ensayo de Materiales , Peso Molecular , Estructura Secundaria de Proteína , Resistencia a la Tracción , Ingeniería de Tejidos
18.
Med Eng Phys ; 53: 39-48, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29396019

RESUMEN

Collagen hydrogels have been used ubiquitously as engineering biomaterials with a biphasic network of fibrillar collagen and aqueous-filled voids that contribute to a complex, compressible, and nonlinear mechanical behavior - not well captured within the infinitesimal strain theory. In this study, type-I collagen, processed from a bovine corium, was fabricated into disks at 2, 3, and 4% (w/w) and exposed to 0, 105, 106, and 107 microjoules of ultraviolet light or enzymatic degradation via matrix metalloproteinase-2. Fully hydrated gels were subjected to unconfined, aqueous, compression testing with experimental data modeled within a continuum mechanics framework by employing the uncommon Blatz-Ko material model for porous elastic materials and a nonlinear form of the Poisson's ratio. From the Generalized form, the Special Blatz-Ko, compressible Neo-Hookean, and incompressible Mooney-Rivlin models were derived and the best-fit material parameters reported for each. The average root-mean-squared (RMS) error for the General (RMS = 0.13 ±â€¯0.07) and Special Blatz-Ko (RMS = 0.13 ±â€¯0.07) were lower than the Neo-Hookean (RMS = 0.23 ±â€¯0.10) and Mooney-Rivlin (RMS = 0.18 ±â€¯0.08) models. We conclude that, with a single fitted-parameter, the Special Blatz-Ko sufficiently captured the salient features of collagen hydrogel compression over most examined formulations and treatments.


Asunto(s)
Colágeno Tipo I/química , Fuerza Compresiva , Hidrogeles/química , Modelos Moleculares , Animales , Bovinos , Colágeno Tipo I/metabolismo , Ratas
19.
J Vis Exp ; (133)2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29658916

RESUMEN

Kidney transplantation is now a mainstream therapy for end-stage renal disease. However, with approximately 96,000 people on the waiting list and only one-fourth of these patients achieving transplantation, there is a dire need for alternatives for those with failing organs. In order to decrease the harmful consequences of dialysis along with the overall healthcare costs it incurs, active investigation is ongoing in search of alternative solutions to organ transplantation. Implantable tissue-engineered renal cellular constructs are one such feasible approach to replacing lost renal functionality. Here, described for the first time, is the microdissection of murine kidneys for isolation of living corticomedullary renal segments. These segments are capable of rapid incorporation within scaffold-free endothelial-fibroblast constructs which may enable rapid connection with host vasculature once implanted. Adult mouse kidneys were procured from living donors, followed by stereoscope microdissection to obtain renal segments 200 - 300 µm in diameter. Multiple renal constructs were fabricated using primary renal segments harvested from only one kidney. This method demonstrates a procedure which could salvage functional renal tissue from organs that would otherwise be discarded.


Asunto(s)
Trasplante de Riñón/métodos , Riñón/patología , Microdisección/métodos , Ingeniería de Tejidos/métodos , Animales , Femenino , Humanos , Masculino , Ratones
20.
Tissue Eng ; 13(11): 2781-90, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17880268

RESUMEN

Loss of skeletal muscle profoundly affects the health and well-being of patients, and there currently is no way to replace lost muscle. We believe that a key step in the development of a prosthesis for reconstruction of dysfunctional muscular tissue is the ability to reconstitute the in vivo-like 3-dimensional (3D) organization of skeletal muscle in vitro with isolated satellite cells. In our present proof of principle studies, we have successfully constructed a multilayered culture of skeletal muscle cells, derived from neonatal satellite cells, that are distributed in a 3D pattern of organization that mimics many of the features of intact tissue. These multilayered cultures are composed of elongated multinucleated myotubes that are MyoD positive. Histological studies indicate that the multiple layers of myotubes can be distinguished. Expression of muscle-specific markers such as myosin heavy chain, dystrophin, integrin alpha-7, alpha-enolase, and beta-enolase was detected using real-time reverse transcriptase polymerase chain reaction at levels near adult values. Physiological measurements of the engineered skeletal muscle showed that they tetanize and display physiologic force length behavior, although developed force per cross-sectional area was below that of native rat skeletal muscle.


Asunto(s)
Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo , Ingeniería de Tejidos , Animales , Animales Recién Nacidos , Técnicas de Cultivo de Célula , Separación Celular , Supervivencia Celular/efectos de los fármacos , Colágeno Tipo I/metabolismo , Medios de Cultivo , Fluoresceína-5-Isotiocianato/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Colorantes Fluorescentes/metabolismo , Geles , Concentración de Iones de Hidrógeno , Inmunohistoquímica , Contracción Isométrica , Microscopía Confocal , Contracción Muscular , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Proteína MioD/metabolismo , Faloidina/metabolismo , ARN Mensajero/análisis , Ratas , Células Satélite del Músculo Esquelético/fisiología , Especificidad por Sustrato , Temperatura , Factores de Tiempo , Azul de Tripano/metabolismo , Azul de Tripano/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA