Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(14)2022 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-35887286

RESUMEN

Introduction: Three-dimensional bioprinting can be considered as an advancement of the classical tissue engineering concept. For bioprinting, cells have to be dispersed in hydrogels. Recently, a novel semi-synthetic thiolene hydrogel system based on norbornene-functionalized gelatin (GelNB) and thiolated gelatin (GelS) was described that resulted in the photoclick hydrogel GelNB/GelS. In this study, we evaluated the printability and biocompatibility of this hydrogel system towards adipose-tissue-derived mesenchymal stem cells (ASCs). Methods: GelNB/GelS was synthesized with three different crosslinking densities (low, medium and high), resulting in different mechanical properties with moduli of elasticity between 206 Pa and 1383 Pa. These hydrogels were tested for their biocompatibility towards ASCs in terms of their viability, proliferation and differentiation. The extrusion-based bioprinting of ASCs in GelNB/GelS-high was performed to manufacture three-dimensional cubic constructs. Results: All three hydrogels supported the viability, proliferation and chondrogenic differentiation of ASCs to a similar extent. The adipogenic differentiation of ASCs was better supported by the softer hydrogel (GelNB/GelS-low), whereas the osteogenic differentiation was more pronounced in the harder hydrogel (GelNB/GelS-high), indicating that the differentiation fate of ASCs can be influenced via the adaption of the mechanical properties of the GelNB/GelS system. After the ex vivo chondrogenic differentiation and subcutaneous implantation of the bioprinted construct into immunocompromised mice, the production of negatively charged sulfated proteoglycans could be observed with only minimal inflammatory signs in the implanted material. Conclusions: Our results indicate that the GelNB/GelS hydrogels are very well suited for the bioprinting of ASCs and may represent attractive hydrogels for subsequent in vivo tissue engineering applications.


Asunto(s)
Bioimpresión , Células Madre Mesenquimatosas , Animales , Bioimpresión/métodos , Gelatina , Hidrogeles , Ratones , Norbornanos , Osteogénesis , Impresión Tridimensional , Compuestos de Sulfhidrilo , Ingeniería de Tejidos/métodos , Andamios del Tejido
2.
Biol Chem ; 402(6): 693-702, 2021 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-33544464

RESUMEN

Mesenchymal stem cells (MSCs) play an important role in tissue engineering applications aiming at the regeneration or substitution of damaged tissues. In this context, off-the-shelf allogeneic MSCs would represent an attractive universal cell source. However, immune rejection is a major limitation for the clinical use of allogeneic MSCs. Immune rejection is mediated by the expression of major histocompatibility complexes (MHC)-I and -II on the donor cells. In this study, we eliminated MHC-I and/or MHC-II expression in human MSCs by using the CRISPR/Cas9 technology and investigated the effect of the individual or combined knockout of MHC-I and MHC-II on MSC survival after transplantation into immunocompetent mice. Elimination of MHC-I and/or MHC-II expression did not affect mesenchymal marker gene expression, viability, proliferation and the differentiation potential of MSCs in vitro. However, cell survival of transplanted MSCs was significantly elevated in MHC-I and MHC-II deficient MSCs. A direct side-by-side comparison does not reveal any significant difference in the immunogenicity of MHC-I and MHC-II knockout MSCs. Moreover, double knockout of MHC-I and MHC-II did not further increase in vivo cell survival of transplanted MSCs. Our results demonstrate that knockout of MHC-I and/or MHC-II represents an effective strategy to prevent immune rejection of allogeneic MSCs.


Asunto(s)
Complejo Mayor de Histocompatibilidad/inmunología , Células Madre Mesenquimatosas/inmunología , Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/inmunología , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Citometría de Flujo , Edición Génica , Humanos , Complejo Mayor de Histocompatibilidad/genética , Células Madre Mesenquimatosas/citología
3.
Biotechnol Bioeng ; 117(12): 3902-3911, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32749669

RESUMEN

Bioprinting can be considered as a progression of the classical tissue engineering approach, in which cells are randomly seeded into scaffolds. Bioprinting offers the advantage that cells can be placed with high spatial fidelity within three-dimensional tissue constructs. A decisive factor to be addressed for bioprinting approaches of artificial tissues is that almost all tissues of the human body depend on a functioning vascular system for the supply of oxygen and nutrients. In this study, we have generated cuboid prevascularized bone tissue constructs by bioprinting human adipose-derived mesenchymal stem cells (ASCs) and human umbilical vein endothelial cells (HUVECs) by extrusion-based bioprinting and drop-on-demand (DoD) bioprinting, respectively. The computer-generated print design could be verified in vitro after printing. After subcutaneous implantation of bioprinted constructs in immunodeficient mice, blood vessel formation with human microvessels of different calibers could be detected arising from bioprinted HUVECs and stabilization of human blood vessels by mouse pericytes was observed. In addition, bioprinted ASCs were able to synthesize a calcified bone matrix as an indicator of ectopic bone formation. These results indicate that the combined bioprinting of ASCs and HUVECs represents a promising strategy to produce prevascularized artificial bone tissue for prospective applications in the treatment of critical-sized bone defects.


Asunto(s)
Bioimpresión , Trasplante Óseo , Huesos , Células Madre Mesenquimatosas , Neovascularización Fisiológica , Ingeniería de Tejidos , Animales , Huesos/irrigación sanguínea , Huesos/metabolismo , Xenoinjertos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones SCID , Impresión Tridimensional , Andamios del Tejido
4.
J Cell Biochem ; 118(7): 1756-1763, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-27958650

RESUMEN

Vascularization is essential for bone development, fracture healing, and bone tissue engineering. We have previously described that coculture of primary human osteoblasts (hOBs) and human umbilical vein endothelial cells (HUVECs) improves differentiation of both cell types. Investigating the role of microRNAs (miRNAs) in this system, we found that miR-126 is highly upregulated in hOBs following coculturing with HUVECs. In this study we performed miR-126 gain-of-function and loss-of-function experiments in hOBs followed by microarray analysis in order to identify targets of miR-126. The transcript cluster IDs were sieved by applying cut-off criteria and by selecting transcripts which were upregulated following miR-126 downregulation and vice versa. The calmodulin regulated spectrin associated protein 1 (CAMSAP1) mRNA was confirmed to be differentially regulated by miR-126. Using the luciferase reporter assay it was demonstrated that CAMSAP1 is directly targeted by miR-126. In this study, we show that miR-126 and CAMSAP1 directly interact in hOBs. This finding has potential implications for tissue engineering applications. J. Cell. Biochem. 118: 1756-1763, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
MicroARNs/genética , MicroARNs/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Osteoblastos/metabolismo , ARN Mensajero/metabolismo , Remodelación Ósea/genética , Remodelación Ósea/fisiología , Citoesqueleto/metabolismo , Matriz Extracelular/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ingeniería de Tejidos
5.
J Cell Biochem ; 117(8): 1869-79, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26754918

RESUMEN

Vascularization is important for bone development, fracture healing and engineering of artificial bone tissue. In the context of bone tissue engineering, it was shown that coimplantation of human primary umbilical vein endothelial cells (HUVECs) and human osteoblasts (hOBs) results in the formation of functional blood vessels and enhanced bone regeneration. Implanted endothelial cells do not only contribute to blood vessel formation, but also support proliferation, cell survival and osteogenic differentiation of coimplanted hOBs. These effects are partially mediated by direct heterotypic cell contacts. In a previous report we could show that cocultivated hOBs strongly increase the expression of genes involved in extracellular matrix (ECM) formation in HUVECs, suggesting that ECM may be involved in the intercellular communication between hOBs and HUVECs. The present study aimed at investigating whether comparable changes occur in hOBs. We therefore performed a microarray analysis of hOBs cultivated in direct contact with HUVECs, revealing 1,004 differentially expressed genes. The differentially expressed genes could be assigned to the functional clusters ECM, proliferation, apoptosis and osteogenic differentiation. The microarray data could be confirmed by performing quantitative real time RT-PCR on selected genes. Furthermore, we could show that the ECM produced by HUVECs increased the expression of the osteogenic differentiation marker alkaline phosphatase (ALP) in hOBs. In summary, our data demonstrate that HUVECs provoke complex changes in gene expression patterns in cocultivated hOBs and that ECM plays and important role in this interaction. J. Cell. Biochem. 117: 1869-1879, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Apoptosis , Comunicación Celular , Diferenciación Celular , Matriz Extracelular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Osteoblastos/metabolismo , Técnicas de Cocultivo , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Osteoblastos/citología
6.
Microvasc Res ; 105: 85-92, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26827661

RESUMEN

Postnatal vasculogenesis is mediated by mobilization of endothelial progenitor cells (EPCs) from bone marrow and homing to ischemic tissues. This feature emphasizes this cell type for cell-based therapies aiming at the improvement of neovascularization in tissue engineering applications and regenerative medicine. In animal models, it was demonstrated that implantation of EPCs from cord blood (cbEPCs) led to the formation of a complex functional neovasculature, whereas EPCs isolated from adult peripheral blood (pbEPCs) showed a limited vasculogenic potential, which may be attributed to age-related dysfunction. Recently, it was demonstrated that activation of hypoxia-inducible factor-1α (Hif-1α) improves cell functions of progenitor cells of mesenchymal and endothelial origin. Thus, we hypothesized that overexpression of Hif-1α may improve the vasculogenesis-related phenotype of pbEPCs. In the present study, we overexpressed Hif-1α in pbEPCs and cbEPCs by using recombinant adenoviruses and investigated effects on stem cell- and vasculogenesis-related cell parameters. Overexpression of Hif-1α enhanced proliferation, invasion, cell survival and in vitro capillary sprout formation of both EPC populations. Migration was increased in cbEPCs upon Hif-1α overexpression, but not in pbEPCs. Cellular senescence was decreased in pbEPCs, while remained in cbEPCs, which showed, as expected, intrinsically a dramatically lower senescent phenotype in relation to pbEPCs. Similarly, the colony-formation capacity was much higher in cbEPCs in comparison to pbEPCs and was further increased by Hif-1α overexpression, whereas Hif-1α transduction exerted no significant influence on colony formation of pbEPCs. In summary, our experiments illustrated multifarious effects of Hif-1α overexpression on stem cell and vasculogenic parameters. Therefore, Hif-1α overexpression may represent a therapeutic option to improve cellular functions of adult as well as postnatal EPCs.


Asunto(s)
Células Progenitoras Endoteliales/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neovascularización Fisiológica , Factores de Edad , Apoptosis , Movimiento Celular , Proliferación Celular , Células Cultivadas , Senescencia Celular , Sangre Fetal/citología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Fenotipo , Transducción de Señal , Factores de Tiempo , Transfección , Regulación hacia Arriba
7.
Biol Chem ; 396(3): 245-52, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25473802

RESUMEN

Vascularization plays an important role in tissue engineering applications. It is known that implantation of differentiated endothelial cells or endothelial progenitor cells (EPCs) from cord blood (cbEPCs) gives rise to the formation of a complex functional neovasculature, whereas EPCs isolated from peripheral blood (pbEPCs) have a limited capability to form blood vessels upon implantation. MicroRNA-126 (miR-126) has been shown to have pro-angiogenic effects in vivo. In this study, we investigated whether modulation of miR-126 expression in pbEPCs may alter their angiogenic properties. Gain of function and loss of function experiments revealed that miR-126 has anti-angiogenic effects in pbEPCs. Overexpression of miR-126 resulted in decreased proliferation, migration, invasion and tube formation, while inhibition of miR-126 induced the opposite effects. However, modulation of miR-126 expression did not influence apoptotic susceptibility of pbEPCs. This study provides evidence that inhibition of miR-126 improves angiogenesis-related growth parameters in pbEPCs and may represent a therapeutic option to ameliorate the angiogenic and vasculogenic properties of pbEPCs.


Asunto(s)
Células Progenitoras Endoteliales/metabolismo , MicroARNs/metabolismo , Neovascularización Fisiológica/genética , Adulto , Animales , Apoptosis/genética , Movimiento Celular/genética , Proliferación Celular , Células Progenitoras Endoteliales/citología , Humanos , Ratas
8.
Biol Chem ; 396(3): 253-60, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25536665

RESUMEN

Vascularization is essential in bone tissue engineering and recent research has focused on interactions between osteoblasts (hOBs) and endothelial cells (ECs). It was shown that cocultivation increases the stability of osteoblastic alkaline phosphatase (ALP) mRNA. We investigated the mechanisms behind this observation, focusing on mRNA binding proteins. Using a luciferase reporter assay, we found that the 3'-untranslated region (UTR) of ALP mRNA is necessary for human umbilical vein endothelial cells (HUVEC)-mediated stabilization of osteoblastic ALP mRNA. Using pulldown experiments and nanoflow-HPLC mass spectrometry, vimentin was identified to bind to the 3'-UTR of ALP mRNA. Validation was performed by Western blotting. Functional experiments inhibiting intermediate filaments with iminodipropionitrile and specific inhibition of vimentin by siRNA transfection showed reduced levels of ALP mRNA and protein. Therefore, ALP mRNA binds to and is stabilized by vimentin. This data add to the understanding of intracellular trafficking of ALP mRNA, its function, and have possible implications in tissue engineering applications.


Asunto(s)
Fosfatasa Alcalina/genética , Filamentos Intermedios/metabolismo , Osteoblastos/enzimología , Estabilidad del ARN , Vimentina/metabolismo , Regiones no Traducidas 3'/genética , Fosfatasa Alcalina/metabolismo , Biotina/metabolismo , Cromatografía Líquida de Alta Presión , Pruebas de Enzimas , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Luciferasas/metabolismo , Espectrometría de Masas , Nanotecnología , Unión Proteica , ARN Interferente Pequeño/metabolismo
9.
Biol Chem ; 396(1): 61-70, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25205714

RESUMEN

Adequate vascularization is an essential requirement for bone development, fracture healing and bone tissue engineering. We have previously described the coculture of primary human osteoblasts (hOBs) and human endothelial cells (HUVECs), designed to investigate the interactions between these cells. In this system, we showed that cocultivation of these two cell types leads to a downregulation of platelet-derived growth factor receptor-α (PDGFR-α) in hOBs, which was a consequence of reduced mRNA stability. In the current study we investigated the possible involvement of microRNAs in this process. Firstly, we performed a microarray analysis of osteoblastic miRNAs following cocultivation with HUVECs, revealing an upregulation of miR-126. This result was confirmed by RT-qPCR, and we observed that the increase is dependent on direct cell-to-cell contacts. Gain-of-function and loss-of-function experiments showed that miR-126 is a negative regulator of PDGFR-α mRNA. Additionally, migration of hOBs was inhibited by miR-126 overexpression and stimulated by miR-126 inhibition. Addition of PDGFR-α blocking antibody to hOB culture also inhibited hOB migration. There was no effect of miR-126 modulation on osteoblast proliferation, apoptosis rate or differentiation. In conclusion, we report that the miR-126/PDGFR-α system regulates the migratory behavior of human osteoblasts, without exerting effects on cell survival and differentiation.


Asunto(s)
MicroARNs/metabolismo , Osteoblastos/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Movimiento Celular , Proliferación Celular , Humanos , Osteoblastos/citología
10.
Microvasc Res ; 98: 40-7, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25497270

RESUMEN

Neovascularization is crucial for fracture healing and plays an important role in long-time graft survival in tissue engineering applications. Endothelial progenitor cells (EPCs) can be isolated from peripheral blood avoiding donor site morbidity, which makes them attractive for autologous cell-based engineering of neovessels. However, contradictory results are published concerning the vasculogenic potential of this cell type. We could previously show that implanted human endothelial vein cells (HUVECs) gave rise to the formation of a complex functional human neovasculature in a heterotopic (subcutaneous) as well as in an orthotopic (calvarial defect) model of severe combined immunodeficiency (SCID) mice, where vessel formation could even be increased by coimplanting mesenchymal stem cells (MSCs) functioning as perivascular cells. In this study, we investigated whether coimplantation of MSCs which have been predifferentiated in vitro into SMCs (SMC-MSCs) may enable pbEPCs to form blood vessels upon implantation and, if this would be the case, whether the resulting enhanced vascularization may support bone regeneration. For this purpose, pbEPCs and SMC-MSCs were mono- or cocultured in collagen matrices and seeded into scaffolds consisting of decalcified processed bovine cancellous bone (PBCB, Tutobone). Neovascularization and osteogenesis were evaluated using a calvarial bone defect-model in SCID mice. Our experiments could show that the missing vasculogenic potential of pbEPCs is not rescued by coimplantation of SMCs derived from MSCs predifferentiated along the vascular smooth muscle lineage. However, implantation of both cell types alone, or in combination induced an angiogenic response, which correlated in a positive manner with bone formation within the implants.


Asunto(s)
Regeneración Ósea , Células Progenitoras Endoteliales/citología , Neovascularización Patológica , Osteogénesis/fisiología , Animales , Huesos/patología , Bovinos , Diferenciación Celular , Linaje de la Célula , Técnicas de Cocultivo , Células Endoteliales/fisiología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Células Madre Mesenquimatosas/citología , Ratones , Ratones SCID , Músculo Liso/citología , Neovascularización Fisiológica , Esferoides Celulares/metabolismo , Ingeniería de Tejidos/métodos
11.
J Surg Res ; 198(1): 50-6, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26026854

RESUMEN

BACKGROUND: Neovascularization plays an important role in tissue engineering applications. In animal models, it was demonstrated that implantation of endothelial progenitor cells (EPCs) from cord blood led to the formation of a complex functional neovasculature, whereas EPCs isolated from peripheral blood (pbEPCs) showed a limited vasculogenic potential, which may be attributed to age-related dysfunction. Growth differentiation factor 11 (GDF11) was recently identified as a rejuvenation factor, which was able to reverse age-related dysfunction of stem cells. Therefore, we hypothesized that GDF11 may improve the vasculogenesis-related phenotype of pbEPCs. MATERIALS AND METHODS: pbEPCs were isolated from adult peripheral blood. Transforming growth factor (TGF)-ß type-I receptor expression was analyzed by immunostaining. pbEPCs were treated with recombinant GDF11 for various time periods. Thereafter, phosphorylation of Smad2/Smad3, adhesion, proliferation, cell survival, migration, and in vitro sprout formation was investigated. RESULTS: pbEPCs express the TGF-ß type-I receptors ALK4 and ALK5, but not ALK7. Treatment of pbEPCs with recombinant GDF11 resulted in activation of the Smad2/Smad3 pathway and in increased migration, which was inhibited by the TGF-ß1 superfamily type-I activin receptor-like kinase inhibitor SB431542, demonstrating that the TGF-ß receptor-Smad2/Smad3 pathway is involved in GDF11 induced migration. Moreover, in vitro sprout formation was increased as well by GDF11 treatment. However, other parameters such as adherence, proliferation, and apoptosis were not affected by GDF11. CONCLUSIONS: This study provides evidence that GDF11 improves vasculogenesis-related growth parameters in pbEPCs and may represent a therapeutic option to ameliorate the angiogenic and vasculogenic properties of pbEPCs.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Movimiento Celular , Células Progenitoras Endoteliales/fisiología , Factores de Diferenciación de Crecimiento/fisiología , Células Cultivadas , Humanos , Neovascularización Fisiológica , Factor A de Crecimiento Endotelial Vascular/fisiología
12.
J Cell Biochem ; 114(7): 1584-94, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23334902

RESUMEN

The most promising strategies in bone engineering have concentrated on providing sufficient vascularization to support the newly forming tissue. In this context, recent research in the field has focused on studying the complex interactions between bone forming and endothelial cells. Our previous work has demonstrated that direct contact cocultivation of human umbilical vein endothelial cells (HUVECs) with primary human osteoblasts (hOBs) induces the osteogenic phenotype and survival of hOBs. In order to investigate the mechanisms that lead to this effect, we performed microarray gene expression profiling on HUVECs following cocultivation with hOBs. Our data reveal profound transcriptomic changes that are dependent on direct cell contact between these cell populations. Pathway analysis using the MetaCore™ platform and literature research suggested a striking upregulation of transcripts related to extracellular matrix and cell-matrix interactions. Upregulation of a number of major angiogenetic factors confirms previous observations that HUVECs enter a proangiogenic state upon cocultivation with osteoblasts. Interestingly, the downregulated transcripts clustered predominantly around cell cycle-related processes. The microarray data were confirmed by quantitative real-time RT-PCR on selected genes. Taken together, this study provides a platform for further inquiries in complex interactions between endothelial cells and osteoblasts.


Asunto(s)
Técnicas de Cocultivo/métodos , Matriz Extracelular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Células Cultivadas , Perfilación de la Expresión Génica , Humanos , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Ingeniería de Tejidos
13.
Cells ; 12(4)2023 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-36831313

RESUMEN

Spheroids, organoids, or cell-laden droplets are often used as building blocks for bioprinting, but so far little is known about the spatio-temporal cellular interactions subsequent to printing. We used a drop-on-demand bioprinting approach to study the biological interactions of such building blocks in dimensions of micrometers. Highly-density droplets (approximately 700 cells in 10 nL) of multiple cell types were patterned in a 3D hydrogel matrix with a precision of up to 70 µm. The patterns were used to investigate interactions of endothelial cells (HUVECs) and adipose-derived mesenchymal stem cells (ASCs), which are related to vascularization. We demonstrated that a gap of 200 µm between HUVEC and ASC aggregates led to decreased sprouting of HUVECs towards ASCs and increased growth from ASCs towards HUVECs. For mixed aggregates containing both cell types, cellular interconnections of ASCs with lengths of up to approximately 800 µm and inhibition of HUVEC sprouting were observed. When ASCs were differentiated into smooth muscle cells (dASCs), separate HUVEC aggregates displayed decreased sprouting towards dASCs, whereas no cellular interconnections nor inhibition of HUVEC sprouting were detected for mixed dASCs/HUVEC aggregates. These findings demonstrate that our approach could be applied to investigate cell-cell interactions of different cell types in 3D co-cultures.


Asunto(s)
Bioimpresión , Células Madre Mesenquimatosas , Humanos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Bioimpresión/métodos , Células Madre Mesenquimatosas/metabolismo , Comunicación Celular , Hidrogeles/farmacología
14.
Clin Oral Investig ; 16(3): 879-87, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21584693

RESUMEN

Enterococcus faecalis (E. faecalis) is a Gram-positive bacterium, mostly recovered from root-filled teeth with persistent periapical lesions. Bacterial contamination of root canals inevitably results in interaction between E. faecalis and periapical tissues during the dynamic process of periapical inflammation. This study investigated the impact of heat-inactivated endodontic E. faecalis on the proliferation and the differentiation of ovine osteoblast-like cells, in an attempt to elucidate its putative enhanced pathogenicity mechanisms. Therefore, two different concentrations of a heat-inactivated endodontic E. faecalis isolate (2 × 10(6) or 2 × 10(8) CFU/ml) were incubated with ovine osteoblast-like cells for 7 and 14 days, respectively. Cells without antigen served as control. The effects of antigen on cell growth were evaluated by a proliferation assay (EZ4U). Furthermore, the assessment of alkaline phosphatase (ALP) activity, calcium deposition, and osteocalcin (OCN) gene expression through quantitative real-time PCR determined the degree of osteogenic cell differentiation. Scanning electron microscopy (SEM) was also performed to detect alterations in cell morphology. Interestingly, although highly concentrated E. faecalis increased cellular reproduction after 14 days, ALP activity and OCN gene expression decreased in an antigen concentration-dependent and incubation time-independent way. SEM images revealed E. faecalis adhesion on cells, a fact that might contribute to its virulence. These results suggest that E. faecalis stimulated cell multiplication, whereas it likely restrained cell differentiation of ovine osteoblast-like cells. In conclusion, the presence of E. faecalis in root canals may negatively affect periapical new bone formation, and thus, the healing of periapical lesions.


Asunto(s)
Enterococcus faecalis/fisiología , Osteoblastos/microbiología , Periodontitis Periapical/microbiología , Virulencia , Fosfatasa Alcalina/biosíntesis , Fosfatasa Alcalina/genética , Animales , Antígenos Bacterianos/fisiología , Adhesión Bacteriana , Calcio/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Recuento de Colonia Microbiana , ADN Bacteriano/análisis , Cavidad Pulpar/microbiología , Humanos , Análisis de los Mínimos Cuadrados , Osteoblastos/citología , Osteoblastos/metabolismo , Osteocalcina/biosíntesis , Osteocalcina/genética , Oveja Doméstica , Calcificación de Dientes
15.
J Mech Behav Biomed Mater ; 130: 105219, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35413680

RESUMEN

The generation of artificial human tissue by 3D-bioprinting has expanded significantly as a clinically relevant research topic in recent years. However, to produce a complex and viable tissue, in-depth biological understanding and advanced printing techniques are required with a high number of process parameters. Here, we systematically evaluate the process parameters relevant for a hybrid bioprinting process based on fused-deposition modeling (FDM) of thermoplastic material and microextrusion of a cell-laden hydrogel. First, we investigated the effect of the printing temperature of polycaprolactone (PCL), on the junction strength between individual fused filaments and on the viability of immortalized mesenchymal stem cells (iMSC) in the surrounding alginate-gelatin-hydrogel. It was found that a printing temperature of 140 °C and bonds with an angle of 90° between the filaments provided a good compromise between bonding strength of the filaments and the viability of the surrounding cells. Using these process parameters obtained from individual fused filaments, we then printed cubic test structures with a volume of 10 × 10 × 10 mm3 with different designs of infill patterns. The variations in mechanical strength of these cubes were measured for scaffolds made of PCL-only as well as for hydrogel-filled PCL scaffolds printed by alternating hybrid bioprinting of PCL and hydrogel, layer by layer. The bare scaffolds showed a compressive modulus of up to 6 MPa, close to human hard tissue, that decreased to about 4 MPa when PCL was printed together with hydrogel. The scaffold design suited best for hybrid printing was incubated with cell-laden hydrogel and showed no degradation of its mechanical strength for up to 28 days.


Asunto(s)
Bioimpresión , Alginatos , Bioimpresión/métodos , Gelatina , Humanos , Hidrogeles , Poliésteres , Impresión Tridimensional , Ingeniería de Tejidos/métodos , Andamios del Tejido/química
16.
Nat Methods ; 5(5): 439-45, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18391960

RESUMEN

The complexity of the angiogenic cascade limits cellular approaches to studying angiogenic endothelial cells (ECs). In turn, in vivo assays do not allow the analysis of the distinct cellular behavior of ECs during angiogenesis. Here we show that ECs can be grafted as spheroids into a matrix to give rise to a complex three-dimensional network of human neovessels in mice. The grafted vasculature matures and is connected to the mouse circulation. The assay is highly versatile and facilitates numerous applications including studies of the effects of different cytokines on angiogenesis. Modifications make it possible to study human lymphangiogenic processes in vivo. EC spheroids can also be coimplanted with other cell types for tissue engineering purposes.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Endoteliales/citología , Neovascularización Fisiológica/fisiología , Esferoides Celulares/citología , Animales , Comunicación Celular , Células Endoteliales/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Ratones , Ingeniería de Tejidos , Factor A de Crecimiento Endotelial Vascular/farmacología
17.
Cells ; 10(7)2021 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-34359919

RESUMEN

Bone is a highly vascularized tissue, and its development, maturation, remodeling, and regeneration are dependent on a tight regulation of blood vessel supply. This condition also has to be taken into consideration in the context of the development of artificial tissue substitutes. In classic tissue engineering, bone-forming cells such as primary osteoblasts or mesenchymal stem cells are introduced into suitable scaffolds and implanted in order to treat critical-size bone defects. However, such tissue substitutes are initially avascular. Because of the occurrence of hypoxic conditions, especially in larger tissue substitutes, this leads to the death of the implanted cells. Therefore, it is necessary to devise vascularization strategies aiming at fast and efficient vascularization of implanted artificial tissues. In this review article, we present and discuss the current vascularization strategies in bone tissue engineering. These are based on the use of angiogenic growth factors, the co-implantation of blood vessel forming cells, the ex vivo microfabrication of blood vessels by means of bioprinting, and surgical methods for creating surgically transferable composite tissues.


Asunto(s)
Huesos/irrigación sanguínea , Neovascularización Fisiológica , Ingeniería de Tejidos , Bioimpresión , Células Endoteliales/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo
18.
PLoS One ; 16(3): e0238330, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33661950

RESUMEN

The homogeneity of the genetically modified single-cells is a necessity for many applications such as cell line development, gene therapy, and tissue engineering and in particular for regenerative medical applications. The lack of tools to effectively isolate and characterize CRISPR/Cas9 engineered cells is considered as a significant bottleneck in these applications. Especially the incompatibility of protein detection technologies to confirm protein expression changes without a preconditional large-scale clonal expansion creates a gridlock in many applications. To ameliorate the characterization of engineered cells, we propose an improved workflow, including single-cell printing/isolation technology based on fluorescent properties with high yield, a genomic edit screen (Surveyor assay), mRNA RT-PCR assessing altered gene expression, and a versatile protein detection tool called emulsion-coupling to deliver a high-content, unified single-cell workflow. The workflow was exemplified by engineering and functionally validating RANKL knockout immortalized mesenchymal stem cells showing bone formation capacity of these cells. The resulting workflow is economical, without the requirement of large-scale clonal expansions of the cells with overall cloning efficiency above 30% of CRISPR/Cas9 edited cells. Nevertheless, as the single-cell clones are comprehensively characterized at an early, highly parallel phase of the development of cells including DNA, RNA, and protein levels, the workflow delivers a higher number of successfully edited cells for further characterization, lowering the chance of late failures in the development process.


Asunto(s)
Bioimpresión/métodos , Clonación Molecular/métodos , Técnicas de Inactivación de Genes/métodos , Células Madre Mesenquimatosas/metabolismo , Ligando RANK/genética , Análisis de la Célula Individual/métodos , Sistemas CRISPR-Cas , Diferenciación Celular , Línea Celular , Humanos , Células Madre Mesenquimatosas/citología , Osteoblastos/citología , Osteoblastos/metabolismo , Flujo de Trabajo
19.
Cells Tissues Organs ; 192(1): 28-38, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20090302

RESUMEN

Platelet-derived growth factor receptor (PDGFR) signaling plays an important role in osteoblast function. Inhibition of PDGFR activity leads to a suppression of osteoblast proliferation, whereas mineralized matrix production is enhanced. In previous experiments, we showed that co-cultivation of human primary endothelial cells and human primary osteoblasts (hOBs) leads to a cell contact-dependent downregulation of PDGFR-alpha expression in the osteoblasts. In this study, we investigated this effect in more detail, revealing that human umbilical vein endothelial cell (HUVEC)-mediated PDGFR-alpha downregulation is dependent on time and cell number. This effect was specific to endothelial cells and was not observed when hOBs were co-cultured with human primary chondrocytes or fibroblasts. Likewise, HUVEC-mediated suppression of PDGFR-alpha expression was only seen in hOBs and mesenchymal stem cells but not in immortalized osteoblastic cell lines. Functional inhibition of gap junctional communication between HUVECs and hOBs by 18alpha-glycyrrhetinic acid had no effect on HUVEC-mediated PDGFR-alpha downregulation, whereas inhibition of p38 mitogen-activated protein kinase (MAPK) prevented the HUVEC-mediated reduction in osteoblastic PDGFR-alpha expression. To delineate the molecular mechanism underlying the PDGFR-alpha downregulation, we examined the effect of HUVEC co-cultivation on osteoblastic PDGFR-alpha promoter activity as well as mRNA stability. Co-cultivation of HUVECs with hOBs significantly shortened the half-life of osteoblastic PDGFR-alpha mRNA, but did not decrease its promoter activity. In summary, our data show that PDGFR-alpha is downregulated in hOBs by co-cultivation with human primary endothelial cells through a p38 MAPK-dependent post-transcriptional mechanism.


Asunto(s)
Células Endoteliales/enzimología , Osteoblastos/enzimología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Regulación hacia Abajo , Células Endoteliales/citología , Humanos , Osteoblastos/citología , ARN Mensajero/metabolismo , Transducción de Señal , Transcripción Genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
Sci Rep ; 10(1): 11071, 2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32632214

RESUMEN

The last two decades saw the establishment of three-dimensional (3D) cell cultures as an acknowledged tool to investigate cell behaviour in a tissue-like environment. Cells growing in spheroids differentiate and develop different characteristics in comparison to their two-dimensionally grown counterparts and are hence seen to exhibit a more in vivo-like phenotype. However, generating, treating and analysing spheroids in high quantities remains labour intensive and therefore limits its applicability in drugs and compound research. Here we present a fully automated pipetting robot that is able to (a) seed hanging drops from single cell suspensions, (b) treat the spheroids formed in these hanging drops with drugs and (c) analyse the viability of the spheroids by an image-based deep learning based convolutional neuronal network (CNN). The model is trained to classify between 'unaffected', 'mildly affected' and 'affected' spheroids after drug exposure. All corresponding spheroids are initially analysed by viability flow cytometry analysis to build a labelled training set for the CNN to subsequently reduce the number of misclassifications. Hence, this approach allows to efficiently examine the efficacy of drug combinatorics or new compounds in 3D cell culture. Additionally, it may provide a valuable instrument to screen for new and individualized systemic therapeutic strategies in second and third line treatment of solid malignancies using patient derived primary cells.


Asunto(s)
Aprendizaje Profundo , Ensayos de Selección de Medicamentos Antitumorales , Neoplasias/patología , Redes Neurales de la Computación , Preparaciones Farmacéuticas/metabolismo , Esferoides Celulares/patología , Supervivencia Celular , Estudios de Factibilidad , Humanos , Neoplasias/tratamiento farmacológico , Preparaciones Farmacéuticas/análisis , Esferoides Celulares/efectos de los fármacos , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA