Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biomater Adv ; 159: 213805, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38457904

RESUMEN

Bone defects may occur in different sizes and shapes due to trauma, infections, and cancer resection. Autografts are still considered the primary treatment choice for bone regeneration. However, they are hard to source and often create donor-site morbidity. Injectable microgels have attracted much attention in tissue engineering and regenerative medicine due to their ability to replace inert implants with a minimally invasive delivery. Here, we developed novel cell-laden bioprinted gelatin methacrylate (GelMA) injectable microgels, with controllable shapes and sizes that can be controllably mineralized on the nanoscale, while stimulating the response of cells embedded within the matrix. The injectable microgels were mineralized using a calcium and phosphate-rich medium that resulted in nanoscale crystalline hydroxyapatite deposition and increased stiffness within the crosslinked matrix of bioprinted GelMA microparticles. Next, we studied the effect of mineralization in osteocytes, a key bone homeostasis regulator. Viability stains showed that osteocytes were maintained at 98 % viability after mineralization with elevated expression of sclerostin in mineralized compared to non-mineralized microgels, showing that mineralization can effectively enhances osteocyte maturation. Based on our findings, bioprinted mineralized GelMA microgels appear to be an efficient material to approximate the bone microarchitecture and composition with desirable control of sample injectability and polymerization. These bone-like bioprinted mineralized biomaterials are exciting platforms for potential minimally invasive translational methods in bone regenerative therapies.


Asunto(s)
Gelatina , Microgeles , Gelatina/farmacología , Gelatina/química , Materiales Biocompatibles , Metacrilatos/química
2.
bioRxiv ; 2023 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-37873385

RESUMEN

Bone defects may occur in different sizes and shapes due to trauma, infections, and cancer resection. Autografts are still considered the primary treatment choice for bone regeneration. However, they are hard to source and often create donor-site morbidity. Injectable microgels have attracted much attention in tissue engineering and regenerative medicine due to their ability to replace inert implants with a minimally invasive delivery. Here, we developed novel cell-laden bioprinted gelatin methacrylate (GelMA) injectable microgels, with controllable shapes and sizes that can be controllably mineralized on the nanoscale, while stimulating the response of cells embedded within the matrix. The injectable microgels were mineralized using a calcium and phosphate-rich medium that resulted in nanoscale crystalline hydroxyapatite deposition and increased stiffness within the crosslinked matrix of bioprinted GelMA microparticles. Next, we studied the effect of mineralization in osteocytes, a key bone homeostasis regulator. Viability stains showed that osteocytes were maintained at 98% viability after mineralization with elevated expression of sclerostin in mineralized compared to non-mineralized microgels, indicating that mineralization effectively enhances osteocyte maturation. Based on our findings, bioprinted mineralized GelMA microgels appear to be an efficient material to approximate the bone microarchitecture and composition with desirable control of sample injectability and polymerization. These bone-like bioprinted mineralized biomaterials are exciting platforms for potential minimally invasive translational methods in bone regenerative therapies.

3.
Materials (Basel) ; 16(17)2023 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-37687480

RESUMEN

The tribological properties and preosteoblast behavior of an RF magnetron-sputtered amorphous carbon coating on a Si (100) substrate were evaluated. The graphite target power was varied from 200 to 500 W to obtain various coating structures. The amorphous nature of the coatings was confirmed via Raman analysis. The contact angle also increased from 58º to 103º, which confirmed the transformation of the a-C surface from a hydrophilic to hydrophobic nature with an increasing graphite target power. A minimum wear rate of about 4.73 × 10-8 mm3/N*mm was obtained for an a-C coating deposited at a 300 W target power. The 300 W and 400 W target power coatings possessed good tribological properties, and the 500 W coating possessed better cell viability and adhesion on the substrate. The results suggest that the microstructure, wettability, tribological behavior and biocompatibility of the a-C coating were highly dependent on the target power of the graphite. A Finite Element Analysis (FEA) showed a considerable increase in the Von Mises stress as the mesh size decreased. Considering both the cell viability and tribological properties, the 400 W target power coating was identified to have the best tribological property as well as biocompatibility.

4.
Adv Healthc Mater ; 12(22): e2202840, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37219011

RESUMEN

Successful integration of cell-laden tissue constructs with host vasculature depends on the presence of functional capillaries to provide oxygen and nutrients to the embedded cells. However, diffusion limitations of cell-laden biomaterials challenge regeneration of large tissue defects that require bulk-delivery of hydrogels and cells. Herein, a strategy to bioprint geometrically controlled, endothelial and stem-cell laden microgels in high-throughput is introduced, allowing these cells to form mature and functional pericyte-supported vascular capillaries in vitro, and then injecting these pre-vascularized constructs minimally invasively in-vivo. It is demonstrated that this approach offers both desired scalability for translational applications as well as unprecedented levels of control over multiple microgel parameters to design spatially-tailored microenvironments for better scaffold functionality and vasculature formation. As a proof-of-concept, the regenerative capacity of the bioprinted pre-vascularized microgels is compared with that of cell-laden monolithic hydrogels of the same cellular and matrix composition in hard-to-heal defects in vivo. The results demonstrate that the bioprinted microgels have faster and higher connective tissue formation, more vessels per area, and widespread presence of functional chimeric (human and murine) vascular capillaries across regenerated sites. The proposed strategy, therefore, addresses a significant issue in regenerative medicine, demonstrating a superior potential to facilitate translational regenerative efforts.


Asunto(s)
Bioimpresión , Microgeles , Ratones , Humanos , Animales , Ingeniería de Tejidos/métodos , Bioimpresión/métodos , Materiales Biocompatibles , Hidrogeles , Andamios del Tejido , Impresión Tridimensional
5.
Adv Healthc Mater ; 12(11): e2200976, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36808718

RESUMEN

Bone autografts remain the gold standard for bone grafting surgeries despite having increased donor site morbidity and limited availability. Bone morphogenetic protein-loaded grafts represent another successful commercial alternative. However, the therapeutic use of recombinant growth factors has been associated with significant adverse clinical outcomes. This highlights the need to develop biomaterials that closely approximate the structure and composition of bone autografts, which are inherently osteoinductive and biologically active with embedded living cells, without the need for added supplements. Here, injectable growth factor-free bone-like tissue constructs are developed, that closely approximate the cellular, structural, and chemical composition of bone autografts. It is demonstrated that these micro-constructs are inherently osteogenic, and demonstrate the ability to stimulate mineralized tissue formation and regenerate bone in critical-sized defects in-vivo. Furthermore, the mechanisms that allow human mesenchymal stem cells (hMSCs) to be highly osteogenic in these constructs, despite the lack of osteoinductive supplements, are assessed, whereby Yes activated protein (YAP) nuclear localization and adenosine signaling appear to regulate osteogenic cell differentiation. The findings represent a step toward a new class of minimally invasive, injectable, and inherently osteoinductive scaffolds, which are regenerative by virtue of their ability to mimic the tissue cellular and extracellular microenvironment, thus showing promise for clinical applications in regenerative engineering.


Asunto(s)
Microgeles , Humanos , Regeneración Ósea/fisiología , Osteogénesis/fisiología , Huesos , Materiales Biocompatibles/química , Diferenciación Celular/fisiología , Ingeniería de Tejidos , Andamios del Tejido/química
6.
Clin Oral Investig ; 27(3): 1215-1225, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36287273

RESUMEN

OBJECTIVES: To develop a 3D-printed, microparticulate hydrogel supplemented with dentin matrix molecules (DMM) as a novel regenerative strategy for dental pulp capping. MATERIALS AND METHODS: Gelatin methacryloyl microgels (7% w/v) mixed with varying concentrations of DMM were printed using a digital light projection 3D printer and lyophilized for 2 days. The release profile of the DMM-loaded microgels was measured using a bicinchoninic acid assay. Next, dental pulp exposure defects were created in maxillary first molars of Wistar rats. The exposures were randomly capped with (1) inert material - negative control, (2) microgels, (3) microgels + DMM 500 µg/ml, (4) microgels + DMM 1000 µg/ml, (5) microgels + platelet-derived growth factor (PDGF 10 ng/ml), or (6) MTA (n = 15/group). After 4 weeks, animals were euthanized, and treated molars were harvested and then processed to evaluate hard tissue deposition, pulp tissue organization, and blood vessel density. RESULTS: All the specimens from groups treated with microgel + 500 µg/ml, microgel + 1000 µg/ml, microgel + PDGF, and MTA showed the formation of organized pulp tissue, tertiary dentin, newly formed tubular and atubular dentin, and new blood vessel formation. Dentin bridge formation was greater and pulp necrosis was less in the microgel + DMM groups compared to MTA. CONCLUSIONS: The 3D-printed photocurable microgels doped with DMM exhibited favorable cellular and inflammatory pulp responses, and significantly more tertiary dentin deposition. CLINICAL RELEVANCE: 3D-printed microgel with DMM is a promising biomaterial for dentin and dental pulp regeneration in pulp capping procedures.


Asunto(s)
Dentina Secundaria , Microgeles , Materiales de Recubrimiento Pulpar y Pulpectomía , Ratas , Animales , Pulpa Dental , Compuestos de Calcio/uso terapéutico , Recubrimiento de la Pulpa Dental/métodos , Materiales Biocompatibles , Silicatos/uso terapéutico , Ratas Wistar , Regeneración , Impresión Tridimensional , Combinación de Medicamentos , Óxidos/uso terapéutico
8.
ACS Appl Mater Interfaces ; 14(36): 40522-40534, 2022 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-36036800

RESUMEN

The mechanism by which stromal cells fill voids in injured tissue remains a fundamental question in regenerative medicine. While it is well-established that fibroblasts fill voids by depositing extracellular matrix (ECM) proteins as they migrate toward the wound site, little is known about their ability to adopt an epithelial-like purse-string behavior. To investigate fibroblast behavior during gap closure, we created an artificial wound with a large void space. We discovered that fibroblasts could form a free-standing bridge over deep microvoids, closing the void via purse-string contraction, a mechanism previously thought to be unique to epithelial wound closure. The findings also revealed that myosin II mediated contractility and intercellular adherent junctions were required for the closure of the fibroblast gap in our fabricated three-dimensional artificial wound. To fulfill their repair function under the specific microenvironmental conditions of wounds, fibroblasts appeared to acquire the structural features of epithelial cells, namely, contractile actin bundles that span over multiple cells along the boundary. These findings shed light on a novel mechanism by which stromal cells bridge the 3D gap during physiological processes such as morphogenesis and wound healing.


Asunto(s)
Actinas , Cicatrización de Heridas , Actinas/metabolismo , Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Miosina Tipo II , Cicatrización de Heridas/fisiología
9.
bioRxiv ; 2022 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-35411345

RESUMEN

Pericytes stabilize blood vessels and promote vascular barrier function. However, vessels subjected to pro-inflammatory conditions have impaired barrier function, which has been suggested to potentially expose perivascular cells to SARS-CoV-2. To test this hypothesis, we engineered pericyte-supported vascular capillaries on-a-chip, and determined that the extravasation and binding of spike protein (S1) on perivascular cells of inflamed vessels to be significantly higher that in healthy controls, indicating a potential target to understand COVID-19 vascular complications.

10.
J Mater Chem B ; 9(46): 9583-9593, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34779469

RESUMEN

Conventional biomaterials developed for bone regeneration fail to fully recapitulate the nanoscale structural organization and complex composition of the native bone microenvironment. Therefore, despite promoting osteogenic differentiation of stem cells, they fall short of providing the structural, biochemical, and mechanical stimuli necessary to drive osteogenesis for bone regeneration and function. To address this, we have recently developed a novel strategy to engineer bone-like tissue using a biomimetic approach to achieve rapid and controlled nanoscale mineralization of a cell-laden matrix in the presence of osteopontin, a non-collagenous protein, and a supersaturated solution of calcium and phosphate medium. Here, we build on this approach to engineer bone regeneration scaffolds comprising methacrylated gelatin (GelMA) hydrogels incorporated with calcium citrate core-shell microparticles as a sustained and reliable source of calcium ions for in situ mineralization. We demonstrate successful biomineralization of GelMA hydrogels by embedded calcium carbonate-calcium citrate core-shell microparticles with the resultant mineral chemistry, structure, and organization reminiscent of that of native bone. The biomimetic mineralization was further shown to promote osteogenic differentiation of encapsulated human mesenchymal stem cells even in the absence of other exogenous osteogenic induction factors. Ultimately, by combining the superior biological response engendered by biomimetic mineralization with the intrinsic tissue engineering advantages offered by GelMA, such as biocompatibility, biodegradability, and printability, we envision that our system offers great potential for bone regeneration efforts.


Asunto(s)
Gelatina/química , Hidrogeles/química , Células Madre Mesenquimatosas/fisiología , Metacrilatos/química , Carbonato de Calcio , Citrato de Calcio , Diferenciación Celular , Supervivencia Celular , Humanos , Osteogénesis , Tamaño de la Partícula
11.
Acta Biomater ; 127: 180-192, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33823326

RESUMEN

Successful bone healing in severe trauma depends on early revascularization to restore oxygen, nutrient, growth factor, and progenitor cell supply to the injury. Therapeutic angiogenesis strategies have therefore been investigated to promote revascularization following severe bone injuries; however, results have been inconsistent. This is the first study investigating the effects of dual angiogenic growth factors (VEGF and PDGF) with low-dose bone morphogenetic protein-2 (BMP-2; 2.5 µg) on bone healing in a clinically challenging composite bone-muscle injury model. Our hydrogel-based delivery systems demonstrated a more than 90% protein entrapment efficiency and a controlled simultaneous release of three growth factors over 28 days. Co-stimulation of microvascular fragment constructs with VEGF and PDGF promoted vascular network formation in vitro compared to VEGF or PDGF alone. In an in vivo model of segmental bone and volumetric muscle loss injury, combined VEGF (5 µg) and PDGF (7.5 µg or 15 µg) delivery with a low dose of BMP-2 significantly enhanced regeneration of vascularized bone compared to BMP-2 treatment alone. Notably, the regenerated bone mechanics reached ~60% of intact bone, a value that was previously only achieved by delivery of high-dose BMP-2 (10 µg) in this injury model. Overall, sustained delivery of VEGF, PDFG, and BMP-2 is a promising strategy to promote functional vascularized bone tissue regeneration following severe composite musculoskeletal injury. Although this study is conducted in a clinically relevant composite injury model in rats using a simultaneous release strategy, future studies are necessary to test the regenerative potential of spatiotemporally controlled delivery of triple growth factors on bone healing using large animal models. STATEMENT OF SIGNIFICANCE: Volumetric muscle loss combined with delayed union or non-union bone defect causes deleterious effects on bone regeneration even with the supplementation of bone morphogenetic protein-2 (BMP-2). In this study, the controlled delivery of dual angiogenic growth factors (vascular endothelial growth factor [VEGF] + Platelet-derived growth factor [PDGF]) increases vascular growth in vitro. Co-delivering VEGF+PDGF significantly increase the bone formation efficacy of low-dose BMP-2 and improves the mechanics of regenerated bone in a challenging composite bone-muscle injury model.


Asunto(s)
Proteína Morfogenética Ósea 2/farmacología , Regeneración Ósea , Sistema Musculoesquelético/lesiones , Animales , Huesos , Hidrogeles/farmacología , Osteogénesis , Factor de Crecimiento Derivado de Plaquetas/farmacología , Ratas , Factor A de Crecimiento Endotelial Vascular/farmacología
12.
J Tissue Eng Regen Med ; 15(3): 219-231, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33434398

RESUMEN

Adequate vascularization of scaffolds is a prerequisite for successful repair and regeneration of lost and damaged tissues. It has been suggested that the maturity of engineered vascular capillaries, which is largely determined by the presence of functional perivascular mural cells (or pericytes), plays a vital role in maintaining vessel integrity during tissue repair and regeneration. Here, we investigated the role of pericyte-supported-engineered capillaries in regenerating bone in a critical-size rat calvarial defect model. Prior to implantation, human umbilical vein endothelial cells and human bone marrow stromal cells (hBMSCs) were cocultured in a collagen hydrogel to induce endothelial cell morphogenesis into microcapillaries and hBMSC differentiation into pericytes. Upon implantation into the calvarial bone defects (8 mm), the prevascularized hydrogels showed better bone formation than either untreated controls or defects treated with autologous bone grafts (positive control). Bone formation parameters such as bone volume, coverage area, and vascularity were significantly better in the prevascularized hydrogel group than in the autologous bone group. Our results demonstrate that tissue constructs engineered with pericyte-supported vascular capillaries may approximate the regenerative capacity of autologous bone, despite the absence of osteoinductive or vasculogenic growth factors.


Asunto(s)
Células Inmovilizadas , Hidrogeles , Células Madre Mesenquimatosas , Cráneo , Animales , Células Inmovilizadas/metabolismo , Células Inmovilizadas/patología , Células Inmovilizadas/trasplante , Xenoinjertos , Humanos , Hidrogeles/química , Hidrogeles/farmacología , Masculino , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratas , Ratas Desnudas , Cráneo/irrigación sanguínea , Cráneo/lesiones , Cráneo/metabolismo , Cráneo/patología
13.
J Struct Biol ; 212(3): 107636, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33039511

RESUMEN

Bone mineralization is a highly specific and dynamic nanoscale process that has been studied extensively from a structural, chemical, and biological standpoint. Bone tissue, therefore, may be defined by the interplay of its intricately mineralized matrix and the cells that regulate its biological function. However, the far majority of engineered bone model systems and bone replacement materials have been unable to replicate this key characteristic of bone tissue; that is, the ability of cells to be gradually and rapidly embedded in a three-dimensional (3D) heavily calcified matrix material. Here we review the characteristics that define the bone matrix from a nanostructural perspective. We then revisit the benefits and challenges of existing model systems and engineered bone replacement materials, and discuss recent efforts to replicate the biological, cellular, mechanical, and materials characteristics of bone tissue on the nano- to microscale. We pay particular attention to a recently proposed method developed by our group, which seeks to replicate key aspects of the entrapment of bone cells within a mineralized matrix with precisions down to the level of individual nano-crystallites, inclusive of the bone vasculature, and osteogenic differentiation process. In summary, this paper discusses existing and emerging evidence pointing towards future developments bridging the gap between the fields of biomineralization, structural biology, stem cells, and tissue engineering, which we believe will hold the key to engineer truly functional bone-like tissue in the laboratory.


Asunto(s)
Huesos/efectos de los fármacos , Calcificación Fisiológica/efectos de los fármacos , Hidrogeles/farmacología , Nanoestructuras/química , Diferenciación Celular/efectos de los fármacos , Humanos , Osteogénesis/efectos de los fármacos , Ingeniería de Tejidos/métodos
14.
Adv Mater ; 32(36): e2001736, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32700332

RESUMEN

Biomaterial scaffolds have served as the foundation of tissue engineering and regenerative medicine. However, scaffold systems are often difficult to scale in size or shape in order to fit defect-specific dimensions, and thus provide only limited spatiotemporal control of therapeutic delivery and host tissue responses. Here, a lithography-based 3D printing strategy is used to fabricate a novel miniaturized modular microcage scaffold system, which can be assembled and scaled manually with ease. Scalability is based on an intuitive concept of stacking modules, like conventional toy interlocking plastic blocks, allowing for literally thousands of potential geometric configurations, and without the need for specialized equipment. Moreover, the modular hollow-microcage design allows each unit to be loaded with biologic cargo of different compositions, thus enabling controllable and easy patterning of therapeutics within the material in 3D. In summary, the concept of miniaturized microcage designs with such straight-forward assembly and scalability, as well as controllable loading properties, is a flexible platform that can be extended to a wide range of materials for improved biological performance.


Asunto(s)
Microgeles , Impresión Tridimensional , Ingeniería de Tejidos , Andamios del Tejido/química
15.
Acta Biomater ; 114: 63-75, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32688092

RESUMEN

The objective of this study was to investigate the controlled release of two growth factors (BMP-2 and VEGF) as a treatment strategy for bone healing in clinically challenging composite injuries, consisting of a femoral segmental bone defect and volumetric muscle loss. This is the first investigation of dual growth factor delivery in a composite injury model using an injectable delivery system consisting of heparin microparticles and alginate gel. The loading efficiency of growth factors into these biomaterials was found to be >90%, revealing a strong affinity of VEGF and BMP-2 to heparin and alginate. The system could achieve simultaneous or tunable release of VEGF and BMP-2 by varying the loading strategy. Single growth factor delivery (VEGF or BMP-2 alone) significantly enhanced vascular growth in vitro. However, no synergistic effect was observed for dual growth factor (BMP-2 + VEGF) delivery in vitro. Effective bone healing was achieved in all treatment groups (BMP-2, simultaneous or tunable delivery of BMP-2 and VEGF) in the composite injury model. The mechanics of the regenerated bone reached a maximum strength of ~52% of intact bone with tunable delivery of VEGF and BMP-2. Overall, simultaneous or tunable co-delivery of low-dose BMP-2 and VEGF failed to fully restore the mechanics of bone in this injury model. Given the severity of the composite injury, VEGF alone may not be sufficient to establish mature and stable blood vessels when compared with previous studies co-delivering BMP-2+VEGF enhanced bone tissue regeneration. Hence, future studies are warranted to develop an alternative treatment strategy focusing on better control over growth factor dose, spatiotemporal delivery, and additional growth factors to regenerate fully functional bone tissue. STATEMENT OF SIGNIFICANCE: We have developed an injectable delivery system consisting of heparin microparticles and an alginate hydrogel that is capable of delivering multiple growth factors in a tunable manner. We used this delivery system to deliver BMP-2 and VEGF in a rodent model of composite bone-muscle injury that mimics clinical type III open fractures. An advanced treatment strategy is necessary for these injuries in order to avoid the negative side effects of high doses of growth factors and because it has been shown that the addition of a muscle injury in this model attenuates the bone regenerative effect of BMP-2. This is the first study to test the effects of dual growth factor delivery (BMP-2/VEGF) on bone healing in a composite bone-muscle injury model and is expected to open up new directions in protein delivery for regenerative medicine.


Asunto(s)
Proteína Morfogenética Ósea 2 , Regeneración Ósea , Materiales Biocompatibles , Huesos , Hidrogeles , Músculos
16.
Adv Healthc Mater ; 8(17): e1900593, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31304685

RESUMEN

Current cell-based therapies administered after myocardial infarction (MI) show limited efficacy due to subpar cell retention in a dynamically beating heart. In particular, cardiac patches generally provide a cursory level of cell attachment due to the lack of an adequate microenvironment. From this perspective, decellularized cell-derived ECM (CDM) is attractive in its recapitulation of a natural biophysical environment for cells. Unfortunately, its weak physical property renders it difficult to retain in its original form, limiting its full potential. Here, a novel strategy to peel CDM off from its underlying substrate is proposed. By physically stamping it onto a polyvinyl alcohol hydrogel, the resulting stretchable extracellular matrix (ECM) membrane preserves the natural microenvironment of CDM, thereby conferring a biological interface to a viscoelastic membrane. Its various mechanical and biological properties are characterized and its capacity to improve cardiomyocyte functionality is demonstrated. Finally, evidence of enhanced stem cell delivery using the stretchable ECM membrane is presented, which leads to improved cardiac remodeling in a rat MI model. A new class of material based on natural CDM is envisioned for the enhanced delivery of cells and growth factors that have a known affinity with ECM.


Asunto(s)
Sistema Cardiovascular/patología , Matriz Extracelular/química , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Infarto del Miocardio/terapia , Animales , Apoptosis , Sistema Cardiovascular/diagnóstico por imagen , Sistema Cardiovascular/fisiopatología , Fibroblastos/citología , Fibrosis , Humanos , Macrófagos/metabolismo , Membranas , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/metabolismo , Alcohol Polivinílico/química , Ratas Sprague-Dawley , Resistencia a la Tracción , Remodelación Ventricular
17.
Sci Rep ; 9(1): 2463, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30792420

RESUMEN

Hydrogels have been developed and applied to various biomedical applications due to their biocompatibility. However, understanding of modulation between cells to hydrogel interface is still unclear, and parameters to explain the interaction are not sophisticated enough. In this report, we studied the effect of polymer chain flexibility on cell adhesion to various hydrogel constructs of collagen and fibrin gels. Specifically, novel method of semi-flexible model-based analysis confirmed that chain flexibility mediated microstructure of the hydrogels is a critical factor for cell adhesion on their surfaces. The proposed analysis showed possibility of more accurate prediction of biocompatibility of hydrogels, and it should be considered as one of the important criteria for polymer design and selections for enhancing both biocompatibility and biofunctionality.


Asunto(s)
Materiales Biocompatibles/química , Células Endoteliales de la Vena Umbilical Humana/citología , Hidrogeles/química , Animales , Adhesión Celular , Colágeno/química , Módulo de Elasticidad , Fibrina/química , Humanos
18.
Adv Healthc Mater ; 8(1): e1801000, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30398700

RESUMEN

Growth factors (GFs) are signaling molecules that direct cell development by providing biochemical cues for stem cell proliferation, migration, and differentiation. GFs play a key role in tissue regeneration, but one major limitation of GF-based therapies is dosage-related adverse effects. Additionally, the clinical applications and efficacy of GFs are significantly affected by the efficiency of delivery systems and other pharmacokinetic factors. Hence, it is crucial to design delivery systems that provide optimal activity, stability, and tunable delivery for GFs. Understanding the physicochemical properties of the GFs and the biomaterials utilized for the development of biomimetic GF delivery systems is critical for GF-based regeneration. Many different delivery systems have been developed to achieve tunable delivery kinetics for single or multiple GFs. The identification of ideal biomaterials with tunable properties for spatiotemporal delivery of GFs is still challenging. This review characterizes the types, properties, and functions of GFs, the materials science of widely used biomaterials, and various GF loading strategies to comprehensively summarize the current delivery systems for tunable spatiotemporal delivery of GFs aimed for tissue regeneration applications. This review concludes by discussing fundamental design principles for GF delivery vehicles based on the interactive physicochemical properties of the proteins and biomaterials.


Asunto(s)
Sistemas de Liberación de Medicamentos , Péptidos y Proteínas de Señalización Intercelular/administración & dosificación , Ciencia de los Materiales , Medicina Regenerativa , Ingeniería de Tejidos , Animales , Materiales Biocompatibles/química , Humanos , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico
19.
Carbohydr Polym ; 202: 488-496, 2018 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-30287027

RESUMEN

Alginate (Alg) hydrogels, the most popular natural biomaterials, mimic the extracellular matrix (ECM) microenvironment and offer potential biomedical applications. Despite their excellent properties such as biocompatibility, hydrophilicity and ionic crosslinking, the absence of an intrinsic cell adhesion domain lessens their cell-carrier applications in tissue engineering. Herein, we suggest a three-dimensional (3D) cell culture system by integrating Alg hydrogel and fibroblast-derived matrix (FDM). FDM including cell-adhesion motifs, signaling, and physico-mechanical cues is prepared by the decellularization process by avoiding unfavorable chemical reactions. This cues-integrated-biomaterials (CiB) 3D platform shows increased cell viability, proliferation, chondrogenic and osteogenic differentiation of human bone-marrow-derived mesenchymal stem cells (hMSCs) in situ. The results show that the Alg/FDM hydrogel (CiB) matrix provides an excellent microenvironment for cell adhesion and can control the differentiation of hMSCs into specific lineages. Thus, these results suggest the potential applications of the Alg/FDM hydrogel matrix as a viable 3D culture system for tissue regeneration.


Asunto(s)
Alginatos/farmacología , Materiales Biocompatibles/farmacología , Hidrogeles/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Alginatos/química , Animales , Materiales Biocompatibles/química , Adhesión Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Humanos , Hidrogeles/química , Ratones , Células 3T3 NIH , Tamaño de la Partícula , Propiedades de Superficie , Ingeniería de Tejidos
20.
Sci Rep ; 8(1): 7263, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29740005

RESUMEN

The mechanism of efflux is a tour-de-force in the bacterial armoury that has thwarted the development of novel antibiotics. We report the discovery of a novel chemical series with potent antibacterial properties that was engineered to overcome efflux liability. Compounds liable to efflux specifically via the Resistance Nodulation and cell Division (RND) pump, AcrAB-TolC were chosen for a hit to lead progression. Using structure-based design, the compounds were optimised to lose their binding to the efflux pump, thereby making them potent on wild-type bacteria. We discovered these compounds to be pro-drugs that require activation in E. coli by specific bacterial nitroreductases NfsA and NfsB. Hit to lead chemistry led to the generation of compounds that were potent on wild-type and multi-drug resistant clinical isolates of E. coli, Shigella spp., and Salmonella spp. These compounds are bactericidal and efficacious in a mouse thigh infection model.


Asunto(s)
Antibacterianos/química , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Proteínas de Escherichia coli/química , Profármacos/química , Tiofenos/química , Animales , Antibacterianos/síntesis química , Antibacterianos/farmacología , División Celular/efectos de los fármacos , Farmacorresistencia Bacteriana Múltiple/genética , Proteínas de Escherichia coli/efectos de los fármacos , Humanos , Ratones , Pruebas de Sensibilidad Microbiana , Profármacos/síntesis química , Profármacos/farmacología , Conformación Proteica/efectos de los fármacos , Salmonella/química , Salmonella/efectos de los fármacos , Salmonella/patogenicidad , Shigella/química , Shigella/efectos de los fármacos , Shigella/patogenicidad , Tiofenos/síntesis química , Tiofenos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA