Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Adv ; 9(1): eade1067, 2023 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-36598982

RESUMEN

Perianal fistulas (PAFs) represent a severe complication of Crohn's disease (CD). Despite the advent of biologic and small-molecule therapeutics for luminal disease, PAFs in CD (CD-PAF) are relatively resistant to treatment, with less than 50% responding to any therapy. We report an injectable, biodegradable, mechanically fragmented nanofiber-hydrogel composite (mfNHC) loaded with adipose-derived stem cells (ADSCs) for the treatment of fistulas in a rat model of CD-PAF. The ADSC-loaded mfNHC results in a higher degree of healing when compared to surgical treatment of fistulas, which is a standard treatment. The volume of fistulas treated with mfNHC is decreased sixfold compared to the surgical treatment control. Molecular studies reveal that utilization of mfNHC reduced local inflammation and improved tissue regeneration. This study demonstrates that ADSC-loaded mfNHC is a promising therapy for CD-PAF, and warrants further studies to advance mfNHC toward clinical translation.

2.
Small ; 18(36): e2202309, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35948487

RESUMEN

Functional microgels are preferred stem cell carriers due to the ease of delivery through minimally invasive injection and seamless integration with the surrounding host tissue. A biostimulatory nanofiber-hydrogel composite (NHC) has been previously developed through covalently crosslinking a hyaluronic acid hydrogel network with surface-functionalized poly (ε-caprolactone) nanofiber fragments. The NHC mimics the microarchitecture of native soft tissue matrix, showing enhanced cell infiltration, immunomodulation, and proangiogenic properties. Here, injectability of the pre-formed NHC is improved by mechanical fragmentation, making it into micro-fragmented NHC (mfNHC) in a granular gel form as a stem cell carrier to deliver mesenchymal stem cells (MSCs) for soft tissue remodeling. The mfNHC shows a similar storage modulus but a significantly reduced injection force, as compared with the corresponding bulk NHC. When injected subcutaneously in a rat model, mfNHC-MSC constructs initiate an elevated level of host macrophage infiltration, more pro-regenerative polarization, and subsequently, improved angiogenesis and adipogenesis response when compared to mfNHC alone. A similar trend of host cell infiltration and pro-angiogenic response is detected in a swine model with a larger volume injection. These results suggest a strong potential for use of the mfNHC as an injectable carrier for cell delivery and soft tissue remodeling.


Asunto(s)
Células Madre Mesenquimatosas , Nanofibras , Animales , Ácido Hialurónico , Hidrogeles , Inyecciones , Células Madre Mesenquimatosas/fisiología , Ratas , Porcinos , Ingeniería de Tejidos/métodos
3.
J Funct Biomater ; 11(1)2020 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-31963629

RESUMEN

Matrix-assisted chondrocyte transplantation (MACT) is of great interest for the treatment of patients with cartilage lesions. However, the roles of the matrix properties in modulating cartilage tissue integration during MACT recovery have not been fully understood. The objective of this study was to uncover the effects of substrate mechanics on the integration of implanted chondrocyte-laden hydrogels with native cartilage tissues. To this end, agarose hydrogels with Young's moduli ranging from 0.49 kPa (0.5%, w/v) to 23.08 kPa (10%) were prepared and incorporated into an in vitro human cartilage explant model. The hydrogel-cartilage composites were cultivated for up to 12 weeks and harvested for evaluation via scanning electron microscopy, histology, and a push-through test. Our results demonstrated that integration strength at the hydrogel-cartilage interface in the 1.0% (0.93 kPa) and 2.5% (3.30 kPa) agarose groups significantly increased over time, whereas hydrogels with higher stiffness (>8.78 kPa) led to poor integration with articular cartilage. Extensive sprouting of extracellular matrix in the interfacial regions was only observed in the 0.5% to 2.5% agarose groups. Collectively, our findings suggest that while neocartilage development and its integration with native cartilage are modulated by substrate elasticity, an optimal Young's modulus (3.30 kPa) possessed by agarose hydrogels is identified such that superior quality of tissue integration is achieved without compromising tissue properties of implanted constructs.

4.
Methods Mol Biol ; 2045: 107-117, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30484146

RESUMEN

Multipotent mesenchymal stem cells (MSCs) are an attractive candidate for regeneration of damaged cells, tissues, and organs. Due to limited availabilities, MSC populations must be rapidly expanded to satisfy clinical needs. However, senescence attributed to extensive in vitro expansion compromises the regenerative and therapeutic potential of MSCs. In this chapter, we describe a step-by-step protocol that aims to induce adipogenic and osteogenic differentiation of in vitro aged human MSCs and highlight noteworthy issues that may arise during the process.


Asunto(s)
Adipocitos/citología , Adipogénesis , Diferenciación Celular/efectos de los fármacos , Senescencia Celular/fisiología , Células Madre Mesenquimatosas/citología , Osteocitos/citología , Osteogénesis , Adipocitos/metabolismo , Adipogénesis/efectos de los fármacos , Células de la Médula Ósea/citología , Proliferación Celular , Células Cultivadas , Citometría de Flujo , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Osteocitos/metabolismo , Osteogénesis/efectos de los fármacos , Flujo de Trabajo
5.
Regen Ther ; 9: 120-122, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30525083

RESUMEN

Multipotent mesenchymal stem cells (MSCs) represent a great candidate for various clinical applications including regenerative medicine. However, aging both in vivo and in vitro can significantly compromise MSC characteristics and performance. This paper highlights current thoughts on senescence-induced damage to MSCs that should be considered prior to their use for regeneration of different cells, tissues or organs.

6.
Stem Cell Res Ther ; 9(1): 131, 2018 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-29751774

RESUMEN

BACKGROUND: Adult mesenchymal stem cells (MSCs) hold great promise for regenerative medicine because of their self-renewal, multipotency, and trophic and immunosuppressive effects. Due to the rareness and high heterogeneity of freshly isolated MSCs, extensive in-vitro passage is required to expand their populations prior to clinical use; however, senescence usually accompanies and can potentially affect MSC characteristics and functionality. Therefore, a thorough characterization of the variations in phenotype and differentiation potential of in-vitro aging MSCs must be sought. METHODS: Human bone marrow-derived MSCs were passaged in vitro and cultivated with either DMEM-based or αMEM-based expansion media. Cells were prepared for subculture every 10 days up to passage 8 and were analyzed for cell morphology, proliferative capacity, and surface marker expression at the end of each passage. The gene expression profile and adipogenic and osteogenic differentiation capability of MSCs at early (passage 4) and late (passage 8) passages were also evaluated. RESULTS: In-vitro aging MSCs gradually lost the typical fibroblast-like spindle shape, leading to elevated morphological abnormality and inhomogeneity. While the DMEM-based expansion medium better facilitated MSC proliferation in the early passages, the cell population doubling rate reduced over time in both DMEM and αMEM groups. CD146 expression decreased with increasing passage number only when MSCs were cultured under the DMEM-based condition. Senescence also resulted in MSCs with genetic instability, which was further regulated by the medium recipe. Regardless of the expansion condition, MSCs at both passages 4 and 8 could differentiate into adipocyte-like cells whereas osteogenesis of aged MSCs was significantly compromised. For osteogenic induction, use of the αMEM-based expansion medium yielded longer osteogenesis and better quality. CONCLUSIONS: Human MSCs subjected to extensive in-vitro passage can undergo morphological, phenotypic, and genetic changes. These properties are also modulated by the medium composition employed to expand the cell populations. In addition, adipogenic potential may be better preserved over osteogenesis in aged MSCs, suggesting that MSCs at early passages must be used for osteogenic differentiation. The current study presents valuable information for future basic science research and clinical applications leading to the development of novel MSC-based therapeutic strategies for different diseases.


Asunto(s)
Envejecimiento , Células Madre Mesenquimatosas/metabolismo , Adulto , Diferenciación Celular , Proliferación Celular , Humanos , Masculino , Fenotipo , Adulto Joven
7.
J Biomed Mater Res A ; 105(3): 824-833, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27798956

RESUMEN

Mechanotransduction in neural cells involves multiple signaling pathways that are not fully understood. Differences in lineage and maturation state are suggested causes for conflicting reports on neural cell mechanosensitivity. To optimize matrices for use in stem cell therapy treatments transplanting human induced pluripotent stem cell derived neural stem cells (hNSC) into lesions after spinal cord injury, the effects of Young's Modulus changes on hNSC behavior must be understood. The present study utilizes polyethylene glycol hydrogels containing a continuous gradient in Young's modulus to examine changes in the Young's Modulus of the culture substrate on hNSC neurite extension and neural differentiation. Changes in the Young's Modulus of the polyethylene glycol hydrogels was found to affect neurite extension and cellular organization on the matrices. hNSC cultured on 907 Pa hydrogels were found to extend longer neurites than hNSC cultured on other tested Young's Moduli hydrogels. The gene expression of ß tubulin III and microtubule-associated protein 2 in hNSC was affected by changes in the Young's Modulus of the hydrogel. The combinatory method approach used in the present study demonstrates that hNSC are mechanosensitive and the matrix Young's Modulus should be a design consideration for hNSC transplant applications. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 824-833, 2017.


Asunto(s)
Diferenciación Celular , Hidrogeles/química , Células Madre Pluripotentes Inducidas/metabolismo , Células-Madre Neurales/metabolismo , Neuritas/metabolismo , Polietilenglicoles/química , Línea Celular , Módulo de Elasticidad , Humanos , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología
8.
Acta Biomater ; 21: 55-62, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25931018

RESUMEN

Stem cell therapies, which aim to restore neurological function after central nervous system injury, have shown increased efficacy when a tissue engineering matrix is implanted with cells compared to implantation of the cells alone. However, much work still needs to be done to characterize materials that can be used to facilitate and direct the differentiation of implanted cells. In the current study, polyethylene glycol hydrogels functionalized with continuous Ile-Lys-Val-Ala-Val (IKVAV) concentration gradients were fabricated and utilized to systematically study and optimize the adhesive conditions for neural differentiation of mouse embryonic stem cells in two- and three-dimensional environments. The results suggest that 570 µM and 60 µM are the optimal IKVAV concentrations for 2D and 3D neural differentiation, respectively, to maximize mRNA expression of neuron-specific markers and neurite extension while minimizing apoptotic activities in cultured cells compared to those exposed to higher IKVAV concentrations. The combinatorial approach presented in this work demonstrates that hydrogels functionalized with bioactive peptides provide a defined and tunable platform that can be employed to characterize and improve culture conditions for superior survival, maturation and integration of implanted cells, leading to enhanced restoration of neurological function for those receiving stem cell therapies after traumatic brain and spinal cord injuries.


Asunto(s)
Adhesión Celular , Diferenciación Celular , Células Madre Embrionarias/citología , Hidrogeles , Oligopéptidos/química , Animales , Adhesión Bacteriana , Ratones , Microscopía Electrónica de Rastreo , Peso Molecular
9.
Ann Biomed Eng ; 42(4): 716-26, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24362632

RESUMEN

Successful integration of engineered constructs with host tissues is crucial for cartilage repair, yet achieving it remains challenging. A collagen I-based fibrous capsule characterized by increased cell density and decreased glycosaminoglycan deposition usually forms at the periphery of tissue-engineered cartilage. The current study aimed to evaluate the effects of a solid fibrous capsule on construct integration with native articular cartilage. To this end, capsule-containing (CC) and capsule-free (CF) constructs were grown by culturing chondrocyte-seeded scaffolds with insulin-like growth factor-1 and transforming growth factor-ß1, respectively, in a wavy-walled bioreactor that imparts hydrodynamic forces for 4 weeks. The ability of harvested constructs to integrate with native cartilage was determined using a cartilage explant model. Our results revealed that adhesive stress between native cartilage and the CC constructs was 57% higher than that in the CF group, potentially due to the absence of glycosaminoglycans and increased cell density in the capsule region and deposition of denser and thicker collagen fibrils at the integration site. The present work demonstrates that the fibrous capsule can effectively enhance early integration of engineered and native cartilage tissues and thus suggests the need to include the capsule as a variable in the development of cartilage tissue engineering strategies.


Asunto(s)
Cartílago Articular/crecimiento & desarrollo , Condrocitos/citología , Colágeno Tipo I , Ingeniería de Tejidos , Adhesividad , Animales , Reactores Biológicos , Bovinos , Células Cultivadas , Andamios del Tejido
10.
Tissue Eng Part A ; 19(21-22): 2349-60, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23672482

RESUMEN

Successful tissue-engineering strategies for cartilage repair must maximize the efficacy of chondrocytes within their limited life span. To that end, the combination of exogenous growth factors with mechanical stimuli holds promise for development of clinically relevant cartilage tissue substitutes. The current study aimed to determine whether incorporation of transient exposure to growth factors into a hydrodynamic bioreactor system can improve the functional maturation of tissue-engineered cartilage. Chondrocyte-seeded polyglycolic acid scaffolds were cultivated within a wavy-walled bioreactor that imparts fluid flow-induced shear stress for 4 weeks. Constructs were nourished with 100 ng/mL insulin-like growth factor-1 (IGF-1) or 10 ng/mL transforming growth factor-ß1 (TGF-ß1) either for the first 15 days of the culture (transient) or throughout the entire cultivation (continuous). Transiently treated constructs were found to exhibit better functional properties than continuously nourished constructs. The limited development of engineered tissues continuously stimulated by IGF-1 or TGF-ß1 was related to massive growth factor leftovers in the environments that downregulated the expression of the associated receptors. Treatment with TGF-ß1 eliminated the formation of a fibrous capsule at the construct periphery possibly through suppression of Smad3 phosphorylation, yielding constructs with greater homogeneity. Furthermore, TGF-ß1 reversely regulated Smad2 and Smad3 pathways in articular chondrocytes under hydrodynamic stimuli partially via Smad7. Collectively, transient exposure to growth factors is likely to maintain chondrocyte homeostasis, and thus promotes their anabolic activities under hydrodynamic stimuli. The present work suggests that robust hydrodynamically engineered neocartilage with a reduced fibrotic response and enhanced tissue homogeneity can be achieved through optimization of growth factor supplementation protocols and potentially through manipulation of intracellular signals such as Smad.


Asunto(s)
Cartílago/citología , Cartílago/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/farmacología , Ingeniería de Tejidos/métodos , Factor de Crecimiento Transformador beta1/farmacología , Animales , Reactores Biológicos , Bovinos , Células Cultivadas , Condrocitos/citología , Condrocitos/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Hidrodinámica , Inmunohistoquímica
11.
Stem Cells Transl Med ; 1(11): 843-54, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23197696

RESUMEN

Controlled differentiation of mesenchymal stem cells (MSCs) into the chondrogenic lineage is crucial for in vitro generation of neocartilage, yet achieving it remains challenging. Traditional protocols for MSC differentiation using exogenous inductive molecules, such as transforming growth factor-ß, fall short in meeting the needs of clinical applications because they yield differentiated cells that exhibit hypertrophic characteristics and subsequently facilitate endochondral bone formation. The objective of the current study was to deliver endogenous inductive factors from juvenile articular chondrocytes to bone marrow-derived MSCs to drive MSC chondrogenic differentiation through cocultivation of the two cell types in the absence of direct physical contact and exogenous stimulators. An initial chondrocyte/MSC ratio of 63:1 was identified as the appropriate proportion of the two cell populations to ensure that coculture-driven MSC-differentiated (CDMD) cells replicated the cellular morphology, behavior, and phenotype of articular chondrocytes. In a three-dimensional agarose system, CDMD cells were further shown to develop into robust neocartilage structurally and mechanically stronger than chondrocyte-laden constructs and with reduced hypertrophic potential. Although MSCs tended to lose the ability to express CD44, an important regulator in cartilage biology, during the coculture induction, CDMD cells regained this function in the three-dimensional tissue cultivation. The present work establishes a chondrocyte/MSC coculture model that serves as a template to better understand chondrocyte-driven MSC differentiation and provides insights for improved strategies to develop clinically relevant cartilage tissue replacements.


Asunto(s)
Cartílago Articular/crecimiento & desarrollo , Condrocitos/fisiología , Condrogénesis , Células Madre Mesenquimatosas/metabolismo , Molécula de Adhesión Celular del Leucocito Activado/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Bovinos , Diferenciación Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Receptores de Hialuranos/metabolismo , Regeneración , Técnicas de Cultivo de Tejidos
12.
Tissue Eng Part A ; 17(15-16): 2025-35, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21457088

RESUMEN

Achievement of viable engineered tissues through in vitro cultivation in bioreactor systems requires a thorough understanding of the complex interplay between hydrodynamic forces and biochemical cues such as serum. To this end, chondrocyte-seeded constructs were cultured under continuous fluid-induced shear forces with reduced serum content (0%-2%, v/v), which was partially or completely replaced by a potential substitute, insulin-transferrin-selenium, to minimize deleterious effects associated with the use of culture media containing high levels of serum (10%-20%). Low-serum cultures yielded constructs with similar biochemical properties to those cultivated with high-serum supplements, whereas the serum-free constructs exhibited poor cell proliferation, insufficient extracellular matrix production, and rapid degradation of and/or shear-induced damage to polyglycolic acid scaffolds. A fibrous outer capsule typically observed in hydrodynamic cultures and characterized by increased cell density and decreased (virtually none) glycosaminoglycan deposition was eliminated when serum concentration was equal to or <0.2% in the presence of hydrodynamic stimuli. Our findings suggest that serum is a requirement in insulin-transferrin-selenium-supplemented cultures in order for constructs to exhibit improved properties in response to hydrodynamic forces, and that mechanical and biochemical stimuli may synergistically modulate tissue properties and morphology through shear-responsive signals.


Asunto(s)
Cartílago/fisiología , Hidrodinámica , Insulina/farmacología , Selenio/farmacología , Suero/metabolismo , Ingeniería de Tejidos/métodos , Transferrina/farmacología , Animales , Biomasa , Reactores Biológicos , Cartílago/efectos de los fármacos , Bovinos , Recuento de Células , Proliferación Celular/efectos de los fármacos , Condrocitos/citología , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Colágeno/metabolismo , Medios de Cultivo/química , Glicosaminoglicanos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...