Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Bioeng Biotechnol ; 9: 640419, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33718342

RESUMEN

Recently, we and others have illustrated that extracellular vesicles (EVs) have the potential to support hematopoietic stem and progenitor cell (HSPC) expansion; however, the mechanism and processes responsible for the intercellular communication by EVs are still unknown. In the current study, we investigate whether primary human bone marrow derived mesenchymal stromal cells (BMSC) EVs isolated from two different origins, fetal (fEV) and adult (aEV) tissue, can increase the relative low number of HSPCs found in umbilical cord blood (UCB) and which EV-derived components are responsible for ex vivo HSPC expansion. Interestingly, aEVs and to a lesser extent fEVs, showed supportive ex vivo expansion capacity of UCB-HSPCs. Taking advantage of the two BMSC sources with different supportive effects, we analyzed the EV cargo and investigated how gene expression is modulated in HSPCs after incubation with aEVs and fEVs. Proteomics analyses of the protein cargo composition of the supportive aEV vs. the less-supportive fEV identified 90% of the Top100 exosome proteins present in the ExoCarta database. Gene Ontology (GO) analyses illustrated that the proteins overrepresented in aEVs were annotated to oxidation-reduction process, mitochondrial ATP synthesis coupled proton transport, or protein folding. In contrast, the proteins overrepresented in fEVs were annotated to extracellular matrix organization positive regulation of cell migration or transforming growth factor beta receptor (TGFBR) signaling pathway. Small RNA sequencing identified different molecular signatures between aEVs and fEVs. Interestingly, the microRNA cluster miR-99b/let-7e/miR-125a, previously identified to increase the number of HSPCs by targeting multiple pro-apoptotic genes, was highly and significantly enriched in aEVs. Although we identified significant differences in the supportive effects of aEVs and fEVs, RNAseq analyses of the 24 h treated HSPCs indicated that a limited set of genes was differentially regulated when compared to cells that were treated with cytokines only. Together, our study provides novel insights into the complex biological role of EVs and illustrates that aEVs and fEVs differentially support ex vivo expansion capacity of UCB-HSPCs. Together opening new means for the application of EVs in the discovery of therapeutics for more efficient ex vivo HSPC expansion.

2.
FASEB J ; 34(4): 5435-5452, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32086861

RESUMEN

Osteolineage cell-derived extracellular vesicles (EVs) play a regulatory role in hematopoiesis and have been shown to promote the ex vivo expansion of human hematopoietic stem and progenitor cells (HSPCs). Here, we demonstrate that EVs from different human osteolineage sources do not have the same HSPC expansion promoting potential. Comparison of stimulatory and non-stimulatory osteolineage EVs by next-generation sequencing and mass spectrometry analyses revealed distinct microRNA and protein signatures identifying EV-derived candidate regulators of ex vivo HSPC expansion. Accordingly, the treatment of umbilical cord blood-derived CD34+ HSPCs with stimulatory EVs-altered HSPC transcriptome, including genes with known roles in cell proliferation. An integrative bioinformatics approach, which connects the HSPC gene expression data with the candidate cargo in stimulatory EVs, delineated the potentially targeted biological functions and pathways during hematopoietic cell expansion and development. In conclusion, our study gives novel insights into the complex biological role of EVs in osteolineage cell-HSPC crosstalk and promotes the utility of EVs and their cargo as therapeutic agents in regenerative medicine.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Vesículas Extracelulares/metabolismo , Hematopoyesis , Células Madre Hematopoyéticas/citología , Osteoblastos/citología , Antígenos CD34/metabolismo , Proliferación Celular , Células Cultivadas , Células Madre Hematopoyéticas/metabolismo , Humanos , Osteoblastos/metabolismo , Transcriptoma
3.
Tissue Eng Part A ; 24(3-4): 322-334, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28530157

RESUMEN

One of the ligaments most often damaged during sports-the anterior cruciate ligament (ACL)-has poor healing capacity. On damage, reconstructive surgery is performed to restore the mechanical stability of the knee and to reduce the inflammatory milieu otherwise present in the joint. A return to normal activities, however, takes between 9 and 12 months. Thus, strategies capable of improving ACL graft healing are needed. Embryonic development of tendon and ligament (T/L) is regulated by a crosstalk between different cell types. We hypothesized that terminally differentiated skeletal-derived cells such as osteoblasts, chondrocytes, and myoblasts modulate T/L healing. Using an indirect coculture system, we discovered that myoblast-secreted signals-but not osteoblasts, chondrocytes, or stromal-secreted signals-are capable of upregulating classical T/L markers such as scleraxis and tenomodulin on human hamstring tendon-derived cells (hTC), which contribute to ACL graft healing. Transcriptome analysis showed that coculturing hTC with myoblasts led to an upregulation of extracellular matrix (ECM) genes and resulted in enhanced ECM deposition. In vivo, using a rat model of ACL reconstruction showed that conditioned media derived from human muscle tissue accelerated femoral tunnel closure, a key step for autograft integration. Collectively, these results indicate that muscle-secreted signals can be used to improve ACL graft healing in a clinical setting where muscle remnants are often discarded.


Asunto(s)
Reconstrucción del Ligamento Cruzado Anterior/métodos , Ligamento Cruzado Anterior/citología , Cicatrización de Heridas/fisiología , Animales , Ligamento Cruzado Anterior/metabolismo , Línea Celular , Medios de Cultivo Condicionados , Matriz Extracelular/metabolismo , Humanos , Masculino , Ratones , Mioblastos/citología , Ratas , Ratas Sprague-Dawley , Tendones/citología
4.
J Tissue Eng Regen Med ; 11(4): 1077-1088, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-25758215

RESUMEN

Anterior cruciate ligament (ACL) reconstruction involves the replacement of the torn ligament with a new graft, often a hamstring tendon (HT). Described as similar, the ACL and HT have intrinsic differences related to their distinct anatomical locations. From a cellular perspective, identifying these differences represents a step forward in the search for new cues that enhance recovery after the reconstruction. The purpose of this study was to characterize the phenotype and multilineage potential of ACL- and HT-derived cells. ACL- and HT-derived cells were isolated from tissue harvest from patients undergoing total knee arthroplasty (TKA) or ACL reconstruction. In total, three ACL and three HT donors were investigated. Cell morphology, self-renewal potential (CFU-F), surface marker profiling, expression of tendon/ligament-related markers (PCR) and multilineage potential were analysed for both cell types; both had fibroblast-like morphology and low self-renewal potential. No differences in the expression of tendon/ligament-related genes or a selected set of surface markers were observed between the two cell types. However, differences in their multilineage potential were observed: while ACL-derived cells showed a high potential to differentiate into chondrocytes and adipocytes, but not osteoblasts, HT-derived cells showed poor potential to form adipocytes, chondrocytes and osteoblasts. Our results demonstrated that HT-derived cells have low multilineage potential compared to ACL-derived cells, further highlighting the need for extrinsic signals to fully restore the function of the ACL upon reconstruction. Copyright © 2015 John Wiley & Sons, Ltd.


Asunto(s)
Reconstrucción del Ligamento Cruzado Anterior/métodos , Ligamento Cruzado Anterior/citología , Ligamento Cruzado Anterior/cirugía , Tendones Isquiotibiales/citología , Anciano , Biomarcadores/metabolismo , Linaje de la Célula , Autorrenovación de las Células , Separación Celular , Colagenasas/metabolismo , Ensayo de Unidades Formadoras de Colonias , Femenino , Humanos , Masculino , Persona de Mediana Edad , Osteogénesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Adulto Joven
5.
SLAS Discov ; 22(1): 40-50, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27628690

RESUMEN

Modulating the bone morphogenetic protein 2 (BMP-2) and transforming growth factor-ß1 (TGF-ß1) signaling pathways is essential during tendon/ligament (T/L) healing. Unfortunately, growth factor delivery in situ is far from trivial and, in many cases, the necessary growth factors are not approved for clinical use. Here we used a BMP-2 and a TGF-ß1 reporter cell line to screen a library of 1280 Food and Drug Administration-approved small molecules and identify modulators of both signaling pathways. We identified four compounds capable of modulating BMP and TGF signaling on primary human tendon-derived cells (huTCs) and describe their effects on proliferation and differentiation of these cells.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , Transducción de Señal , Bibliotecas de Moléculas Pequeñas/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Colágeno/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Humanos , Ratones , Visón , Transporte de Proteínas/efectos de los fármacos , Reproducibilidad de los Resultados , Proteínas Smad/metabolismo , Tendones/citología
6.
BMC Biotechnol ; 16(1): 89, 2016 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-27955656

RESUMEN

BACKGROUND: The human body has an extensive capacity to regenerate bone tissue after trauma. However large defects such as long bone fractures of the lower limbs cannot be restored without intervention and often lead to nonunion. Therefore, the aim of the present study was to assess the pool and biological functions of human mesenchymal stromal cells (hMSCs) isolated from different bone marrow locations of the lower limbs and to identify novel strategies to prime the cells prior to their use in bone fracture healing. Following, bone marrow from the ilium, proximal femur, distal femur and proximal tibia was aspirated and the hMSCs isolated. Bone marrow type, volume, number of mononuclear cells/hMSCs and their self-renewal, multilineage potential, extracellular matrix (ECM) production and surface marker profiling were analyzed. Additionally, the cells were primed to accelerate bone fracture healing either by using acoustic stimulation or varying the initial hMSCs isolation conditions. RESULTS: We found that the more proximal the bone marrow aspiration location, the larger the bone marrow volume was, the higher the content in mononuclear cells/hMSCs and the higher the self-renewal and osteogenic differentiation potential of the isolated hMSCs were. Acoustic stimulation of bone marrow, as well as the isolation of hMSCs in the absence of fetal bovine serum, increased the osteogenic and ECM production potential of the cells, respectively. CONCLUSION: We showed that bone marrow properties change with the aspiration location, potentially explaining the differences in bone fracture healing between the tibia and the femur. Furthermore, we showed two new priming methods capable of enhancing bone fracture healing.


Asunto(s)
Regeneración Ósea/fisiología , Curación de Fractura/fisiología , Fracturas Óseas/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Terapia por Ultrasonido/métodos , Estimulación Acústica/métodos , Regeneración Ósea/efectos de la radiación , Terapia Combinada/métodos , Curación de Fractura/efectos de la radiación , Humanos , Células Madre Mesenquimatosas/clasificación , Resultado del Tratamiento
7.
Acta Biomater ; 36: 210-9, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26969523

RESUMEN

UNLABELLED: Articular cartilage lesions have a limited ability to heal by themselves. Yet, golden standard treatments for cartilage repair such as drilling, microfracture and mosaicplasty provide further damage and an unstable solution that degenerates into fibrocartilage in time. Articular cartilage presents a number of gradients in cell number and size along with structural gradients in extra cellular matrix (ECM) composition. Therefore, creating scaffolds that display a structural gradient can be an appealing strategy for cartilage tissue regeneration treatments. In the present study, a scaffold with an in-built discrete gradient in pore size was produced by additive manufacturing. Human mesenchymal stromal cells (hMSCs) were seeded within the gradient scaffolds and their proliferation, differentiation and ECM deposition was evaluated with respect to 2 non-gradient scaffolds. Glycosaminoglycan (GAG) deposition was significantly higher in gradient scaffolds and non-gradient scaffolds with the smallest pore size compared to non-gradient scaffolds with the largest pore size. A gradual increase of chondrogenic markers was observed within the gradient structures with decreasing pore size, which was also accompanied by an increasingly compact ECM formation. Therefore, scaffolds displaying a structural gradient in pore size seem to be a promising strategy to aid in the process of hMSC chondrogenic differentiation and could be considered for improved cartilage tissue regeneration applications. STATEMENT OF SIGNIFICANCE: We present the development of a novel hierarchical scaffold obtained by additive manufacturing. Structural hierarchy is obtained by changing pore size within the pore network characterizing the fabricated scaffolds and proves to be a functional element in the scaffold to influence adult stem cell differentiation in the chondrogenic lineage. Specifically, in regions of the scaffolds presenting smaller pores an increasing differentiation of stem cells toward the chondrogenic differentiation is displayed. Taking inspiration from the zonal organization of articular cartilage tissue, pore size gradients could, therefore, be considered as a new and important element in designing 3D scaffolds for regenerative medicine applications, in particular for all those tissues where gradient physical properties are present.


Asunto(s)
Diferenciación Celular , Condrogénesis , Células Madre Mesenquimatosas/metabolismo , Andamios del Tejido/química , Adulto , Células Cultivadas , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Porosidad
8.
J Cell Biochem ; 117(3): 684-93, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26308651

RESUMEN

Tendon and ligament (T/L) pathologies account for a significant portion of musculoskeletal injuries and disorders. Tissue engineering has emerged as a promising solution in the regeneration of both tissues. Specifically, the use of multipotent human mesenchymal stromal cells (hMSC) has shown great promise to serve as both a suitable cell source for tenogenic regeneration and a source of trophic factors to induce tenogenesis. Using four donor sets, we investigated the bidirectional paracrine tenogenic response between human hamstring tenocytes (hHT) and bone marrow-derived hMSC. Cell metabolic assays showed that only one hHT donor experienced sustained notable increases in cell metabolic activity during co-culture. Histological staining confirmed that co-culture induced elevated collagen protein levels in both cell types at varying time-points in two of four donor sets assessed. Gene expression analysis using qPCR showed the varied up-regulation of anabolic and catabolic markers involved in extracellular matrix maintenance for hMSC and hHT. Furthermore, analysis of hMSC/hHT co-culture secretome using a reporter cell line for TGF-ß, a potent inducer of tenogenesis, revealed a trend of higher TGF-ß bioactivity in hMSC secretome compared to hHT. Finally, hHT cytoskeletal immunostaining confirmed that both cell types released soluble factors capable of inducing favorable tenogenic morphology, comparable to control levels of soluble TGF-ß1. These results suggest a potential for TGF-ß-mediated signaling mechanism that is involved during the paracrine interplay between the two cell types that is reminiscent of T/L matrix remodeling/turnover. These findings have significant implications in the clinical use of hMSC for common T/L pathologies.


Asunto(s)
Matriz Extracelular/metabolismo , Células Madre Mesenquimatosas/fisiología , Tendones/citología , Comunicación Celular , Forma de la Célula , Células Cultivadas , Técnicas de Cocultivo , Colágeno/metabolismo , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Expresión Génica , Humanos , Medicina Regenerativa , Tendones/metabolismo , Ingeniería de Tejidos , Factor de Crecimiento Transformador beta1/metabolismo
9.
Front Physiol ; 4: 138, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23760194

RESUMEN

Systemic inflammatory response (SIR) comprises both direct effects of inflammatory mediators (IM) and indirect effects, such as secondary circulatory failure which results in tissue hypoxia (HOX). These two key components, SIR and HOX, cause multiple organ failure (MOF). Since HOX and IM occur and interact simultaneously in vivo, it is difficult to clarify their individual pathological impact. To eliminate this interaction, precision cut liver slices (PCLS) were used in this study aiming to dissect the effects of HOX and IM on mitochondrial function, integrity of cellular membrane, and the expression of genes associated with inflammation. HOX was induced by incubating PCLS or rat liver mitochondria at pO2 < 1% followed by reoxygenation (HOX/ROX model). Inflammatory injury was stimulated by incubating PCLS with IM (IM model). We found upregulation of inducible nitric oxide synthase (iNOS) expression only in the IM model, while heme oxygenase 1 (HO-1) expression was upregulated only in the HOX/ROX model. Elevated expression of interleukin 6 (IL-6) was found in both models reflecting converging pathways regulating the expression of this gene. Both models caused damage to hepatocytes resulting in the release of alanine aminotransferase (ALT). The leakage of aspartate aminotransferase (AST) was observed only during the hypoxic phase in the HOX/ROX model. The ROX phase of HOX, but not IM, drastically impaired mitochondrial electron supply via complex I and II. Additional experiments performed with isolated mitochondria showed that free iron, released during HOX, is likely a key prerequisite of mitochondrial dysfunction induced during the ROX phase. Our data suggests that mitochondrial dysfunction, previously observed in in vivo SIR-models, is the result of secondary circulatory failure inducing HOX rather than the result of a direct interaction of IM with liver cells.

10.
Biofabrication ; 5(2): 025003, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23443652

RESUMEN

Decellularized extracellular matrix (ECM) has recently gained a lot of interest as an instructive biomaterial for regenerative medicine applications. In this study, the ability of adult human mesenchymal stem cell (hMSC)-derived ECM to rescue the phenotype of osteoarthritic (OA) chondrocytes and to further stimulate the differentiation of healthy (HL) chondrocytes was evaluated. ECMs were prepared by decellularizing hMSCs cultured in basic medium (BM) and chondrogenic medium (CM). The obtained ECM was then combined with a polymeric solution of Poly (ε-caprolactone) (PCL) dissolved in 1, 1, 1, 3, 3, 3-hexafluoro-2-propanol (HFIP) and electrospun meshes were fabricated. Electrospun ECM scaffolds were characterized using scanning electron microscopy (SEM) and picrosirius red staining was used to confirm the presence of collagen. OA and HL chondrocytes were cultured on scaffolds containing hMSC ECM in BM or CM and compared to PCL electrospun scaffolds without ECM. Metabolic activity and chondrogenic gene expression were assessed by Alamar blue assay and quantitative PCR (qPCR) analysis, respectively. The ECM presence resulted in a significant difference in chondrocyte metabolic activity compared to PCL scaffolds alone. HL chondrocytes cultured for 21 days in chondrogenic medium on electrospun scaffolds containing hMSC ECM from BM showed a significant increase in collagen II and aggrecan expression compared to hMSC ECM from CM and PCL scaffolds without ECM incorporation. No significant influence of hMSC ECM presence on the chondrogenic signature of OA chondrocytes was found. The influence of decellularized hMSC ECM on HL chondrocytes suggests that hMSC-derived ECM scaffolds are promising candidates for cartilage tissue engineering applications.


Asunto(s)
Condrocitos/metabolismo , Matriz Extracelular/metabolismo , Células Madre Mesenquimatosas/metabolismo , Adulto , Anciano , Agrecanos/genética , Agrecanos/metabolismo , Cartílago/fisiología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Condrocitos/citología , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Medios de Cultivo Condicionados/farmacología , Matriz Extracelular/química , Matriz Extracelular/ultraestructura , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/citología , Poliésteres/química , Ingeniería de Tejidos , Andamios del Tejido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA