Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Transpl Int ; 36: 11077, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37908676

RESUMO

Islet delivery devices (IDDs) offer potential benefits for islet transplantation and stem cell-based replacement in type 1 diabetes. Little is known about patient preferences regarding islet delivery device characteristics and implantation strategies. Patient preferences for IDDs and implantation strategies remain understudied. We invited patients, parents and caregivers to fill in an online questionnaire regarding IDDs. An online survey gathered responses from 809 type 1 diabetes patients and 47 caregivers. We also assessed diabetes distress in a subgroup of 412 patients. A significant majority (97%) expressed willingness to receive an IDD. Preferred IDD attributes included a 3.5 cm diameter for 37.7% of respondents, while when provided with all options, 30.4% found dimensions unimportant. Respondents were open to approximately 4 implants, each with a 5 cm incision. Many favored a device functioning for 12 months (33.4%) or 24 months (24.8%). Younger participants (16-30) were more inclined to accept a 6 months functional duration (p < 0.001). Functional duration outweighed implant quantity and size (p < 0.001) in device importance. This emphasizes patients' willingness to accommodate burdens related to IDD features and implantation methods, crucial for designing future beta cell replacement strategies.


Assuntos
Diabetes Mellitus Tipo 1 , Transplante das Ilhotas Pancreáticas , Humanos , Diabetes Mellitus Tipo 1/cirurgia , Transplante das Ilhotas Pancreáticas/métodos , Preferência do Paciente
2.
J Mater Sci Mater Med ; 29(11): 174, 2018 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-30413974

RESUMO

Extrahepatic transplantation of islets of Langerhans could aid in better survival of islets after transplantation. When islets are transfused into the liver 60-70% of them are lost immediately after transplantation. An important factor for a successful extrahepatic transplantation is a well-vascularized tissue surrounding the implant. There are many strategies known for enhancing vessel formation such as adding cells with endothelial potential, the combination with angiogenic factors and / or applying surface topography at the exposed surface of the device. Previously we developed porous, micropatterned membranes which can be applied as a lid for an islet encapsulation device and we showed that the surface topography induces human umbilical vein endothelial cell (HUVEC) alignment and interconnection. This was achieved without the addition of hydrogels, often used in angiogenesis assays. In this work, we went one step further towards clinical implementation of the device by combining this micropatterned lid with Mesenchymal Stem Cells (MSCs) to facilitate prevascularization in vivo. As for HUVECs, the micropatterned membranes induced MSC alignment and organization in vitro, an important contributor to vessel formation, whereas in vivo (subcutaneous rat model) they contributed to improved implant prevascularization. In fact, the combination of MSCs seeded on the micropatterned membrane induced the highest vessel formation score in 80% of the sections.


Assuntos
Composição de Medicamentos , Ilhotas Pancreáticas/crescimento & desenvolvimento , Membranas Artificiais , Células-Tronco Mesenquimais , Alicerces Teciduais , Células Endoteliais da Veia Umbilical Humana , Humanos , Ilhotas Pancreáticas/irrigação sanguínea , Neovascularização Fisiológica
3.
Tissue Eng Part C Methods ; 24(11): 628-636, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30306836

RESUMO

IMPACT STATEMENT: This research deals with finding a proper bioengineering strategy for the creation of improved ß-cell replacement therapy in type 1 diabetes. It specifically deals with the microenvironment of ß-cells and its relationship to their endocrine function.


Assuntos
Colágeno Tipo IV/metabolismo , Fibronectinas/metabolismo , Secreção de Insulina , Insulina/metabolismo , Insulinoma/metabolismo , Neoplasias Pancreáticas/metabolismo , Animais , Colágeno Tipo IV/química , Fibronectinas/química , Insulinoma/patologia , Neoplasias Pancreáticas/patologia , Impressão Tridimensional , Ratos , Células Tumorais Cultivadas
4.
J Biomed Mater Res A ; 105(9): 2533-2542, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28470672

RESUMO

The liver as transplantation site for pancreatic islets is associated with significant loss of islets, which can be prevented by grafting in a prevascularized, subcutaneous scaffold. Supporting vascularization of a scaffold to limit the period of ischemia is challenging and was developed here by applying liposomes for controlled release of angiogenic factors. The angiogenic capacity of platelet-derived growth factor, vascular endothelial growth factor, acidic fibroblast growth factor (aFGF), and basic FGF were compared in a tube formation assay. Furthermore, the release kinetics of different liposome compositions were tested. aFGF and L-α-phosphatidylcholine/cholesterol liposomes were selected to support vascularization. Two dosages of aFGF-liposomes (0.5 and 1.0 µg aFGF per injection) were administered weekly for a month after which islets were transplanted. We observed enhanced efficacy in the immediate post-transplant period compared to the untreated scaffolds. However, on the long-term, glucose levels of the aFGF treated animals started to increase to diabetic levels. These results suggest that injections with aFGF liposomes do improve vascularization and the immediate restoration of blood glucose levels but does not facilitate the long-term survival of islets. Our data emphasize the need for long-term studies to evaluate potential beneficial and adverse effects of vascularization protocols of scaffolds. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2533-2542, 2017.


Assuntos
Glicemia/metabolismo , Transplante das Ilhotas Pancreáticas , Neovascularização Fisiológica , Tela Subcutânea/irrigação sanguínea , Alicerces Teciduais/química , Animais , Preparações de Ação Retardada , Teste de Tolerância a Glucose , Células Endoteliais da Veia Umbilical Humana , Humanos , Insulina/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/administração & dosagem , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Ilhotas Pancreáticas/patologia , Lipossomos , Masculino , Camundongos Nus , Neovascularização Fisiológica/efeitos dos fármacos , Ratos Sprague-Dawley , Fatores de Tempo
5.
Ann Surg ; 266(1): 149-157, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-27429018

RESUMO

OBJECTIVE: We aim on developing a polymeric ectopic scaffold in a readily accessible site under the skin. SUMMARY BACKGROUND DATA: The liver as transplantation site for pancreatic islets is associated with significant loss of islets. Several extrahepatic sites were tested in experimental animals, but many have practical limitations in the clinical setting and do not have the benefit of easy accessibility. METHODS AND RESULTS: Functional survival of rat islets was tested during 7 days of culture in the presence of poly(D,L-lactide-co-ε-caprolactone) (PDLLCL), poly(ethylene oxide terephthalate)/polybutylene terephthalate (PEOT/PBT) block copolymer, and polysulfone. Tissue responses were studied in vivo after subcutaneous implantation in rats. Culture on PEOT/PBT and polysulfone profoundly disturbed function of islets, and induced severe tissue responses in vivo. Modification of their hydrophilicity did not change the suitability of the polymers. PDLLCL was the only polymer that promoted functional survival of rat islets in vitro and was associated with minor tissue reactions after 28 days. Rat islets were transplanted in the PDLLCL scaffold in a diabetic rat model. Before islet seeding, the scaffold was allowed to engraft for 28 days to allow the tissue response to dampen and to allow blood vessel growth into the device. Islet transplantation into the scaffold resulted in normoglycemia within 3 days and for the duration of the study period of 16 weeks. CONCLUSIONS: In conclusion, we found that some polymers such as PEOT/PBT and polysulfone interfere with islet function. PDLLCL is a suitable polymer to create an artificial islet transplantation site under the skin and supports islet survival.


Assuntos
Diabetes Mellitus Experimental/cirurgia , Transplante das Ilhotas Pancreáticas/métodos , Polímeros , Alicerces Teciduais , Animais , Materiais Biocompatíveis , Técnicas de Cultura de Células , Sobrevivência Celular , Poliésteres , Polietilenoglicóis , Sulfonas
6.
Macromol Biosci ; 16(10): 1524-1532, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27440382

RESUMO

Cell-laden micrometer-sized hydrogels (microgels) hold great promise for improving high throughput ex-vivo drug screening and engineering biomimetic tissues. Microfluidics is a powerful tool to produce microgels. However, only a limited amount of biomaterials have been reported to be compatible with on-chip microgel formation. Moreover, these biomaterials are often associated with mechanical instability, cytotoxicity, and cellular senescence. To resolve this challenge, dextran-tyramine has been explored as a novel biomaterial for on-chip microgel formation. In particular, dextran-tyramine is compared with two commonly used biomaterials, namely, polyethylene-glycol diacrylate (PEGDA) and alginate, which crosslink through enzymatic reaction, UV polymerization, and ionic interaction, respectively. Human mesenchymal stem cells (hMSCs) encapsulated in dextran-tyramine microgels demonstrate significantly higher (95%) survival as compared to alginate (81%) and PEGDA (69%). Long-term cell cultures demonstrate that hMSCs in PEGDA microgels become senescent after 7 d. Alginate microgels dissolve within 7 d due to Ca2+ loss. In contrast, dextran-tyramine based microgels remain stable, sustain hMSCs metabolic activity, and permit for single-cell level analysis for at least 28 d of culture. In conclusion, enzymatically crosslinking dextran-tyramine conjugates represent a novel biomaterial class for the on-chip production of cell-laden microgels, which possesses unique advantages as compared to the commonly used UV and ionic crosslinking biomaterials.


Assuntos
Alginatos/química , Reagentes de Ligações Cruzadas/química , Dispositivos Lab-On-A-Chip , Células-Tronco Mesenquimais/metabolismo , Técnicas Analíticas Microfluídicas/métodos , Polietilenoglicóis/química , Técnicas de Cultura de Células/métodos , Células Cultivadas , Células Imobilizadas/citologia , Células Imobilizadas/metabolismo , Géis , Ácido Glucurônico/química , Ácidos Hexurônicos/química , Humanos , Células-Tronco Mesenquimais/citologia , Fatores de Tempo
7.
Adv Healthc Mater ; 5(13): 1606-16, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27113576

RESUMO

Although regarded as a promising treatment for type 1 diabetes, clinical islet transplantation in the portal vein is still hindered by a low transplantation outcome. Alternative transplantation sites have been proposed, but the survival of extra-hepatically transplanted islets of Langerhans critically depends on quick revascularization after engraftment. This study aims at developing a new 3D scaffold platform that can actively boost vascularization and may find an application for extra-hepatic islet transplantation. The construct consists of a 3D ring-shaped polycaprolactone (PCL) scaffold with heparinized surface to electrostatically bind vascular endothelial growth factor (VEGF), surrounding a hydrogel core for islets encapsulation. Heparin immobilization improves the amount of VEGF retained by the construct, up to 3.6 fold, compared to untreated PCL scaffolds. In a chicken chorioallanthoic membrane model, VEGF immobilized on the construct enhances angiogenesis in close proximity and on the surface of the scaffolds. After 7 days, islets encapsulated in the alginate core show functional response to glucose stimuli comparable to free-floating islets. Thus, the developed platform has the potential to support rapid vascularization and islet endocrine function.


Assuntos
Alginatos/química , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Neovascularização Fisiológica/efeitos dos fármacos , Poliésteres/química , Alicerces Teciduais/química , Fator A de Crescimento do Endotélio Vascular , Animais , Embrião de Galinha , Ácido Glucurônico/química , Heparina/química , Ácidos Hexurônicos/química , Humanos , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/metabolismo , Fator A de Crescimento do Endotélio Vascular/química , Fator A de Crescimento do Endotélio Vascular/farmacologia
8.
Tissue Eng Part C Methods ; 22(6): 534-42, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27056242

RESUMO

The noninvasive and longitudinal imaging of cells or cell aggregates in large optically scattering scaffolds is still a largely unresolved problem in tissue engineering. In this work, we investigated the potential of near-infrared (NIR) photoacoustic (PA) tomography imaging to address this issue. We used clinically relevant sizes of highly light scattering polyethersulfone multibore(®) hollow fiber scaffolds seeded with cells. Since cells have little optical absorption at NIR wavelengths, we studied labeling of cells with absorbers. Four NIR labels were examined for their suitability based on absorption characteristics, resistance to bleaching, and influence on cell viability. On the basis of these criteria, carbon nanoparticles proved most suitable in a variety of cells. For PA imaging, we used a research setup, based on computed tomography geometry. As proof of principle, using this imager we monitored the distribution and clustering of labeled rat insulinoma beta cell aggregates in the scaffolds. This was performed for the duration of 1 week in a nondestructive manner. The results were validated using fluorescence imaging, histology, and light microscopy imaging. Based on our findings, we conclude that PA tomography is a powerful tool for the nondestructive imaging of cells in optically scattering tissue-engineered scaffolds.


Assuntos
Condrócitos/citologia , Tecnologia de Fibra Óptica/métodos , Processamento de Imagem Assistida por Computador/métodos , Insulinoma/patologia , Imagem Óptica/métodos , Engenharia Tecidual/métodos , Animais , Bovinos , Agregação Celular , Sobrevivência Celular , Células Cultivadas , Ratos , Alicerces Teciduais
9.
Tissue Eng Part A ; 22(3-4): 375-85, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26871862

RESUMO

While subcutaneous tissue has been proposed as a clinically relevant site for pancreatic islet transplantation, a major issue of concern remains, which is its poor vascular state. In an effort to overcome this limitation, we present an efficient and reproducible method to form human composite islets (CIs) with proangiogenic cell types in a controlled manner using nonadherent agarose microwell templates. In this study, we assessed the three-dimensional structure, function, and angiogenic potential of human CIs with human mesenchymal stromal cells (hMSCs), with or without human umbilical vein endothelial cells (HUVECs), and preconditioned hMSCs (PC-hMSCs) in EGM-2 under shear stress. Distinct cellular rearrangements could be observed in CIs, but islet functionality was maintained. In vitro angiogenesis assays found significantly enhanced sprout formation in case of CIs. In particular, the number of sprouts emanating from CIs with PC-hMSCs was significantly increased compared to other conditions. Subsequent in vivo assessment confirmed the proangiogenic potential of CIs. However, in contrast to our in vitro angiogenesis assays, CIs with hMSCs and HUVECs exhibited a higher in vivo angiogenic potential compared to control islets or islets combined with hMSCs or PC-hMSCs. These findings highlight the importance and necessity of verifying in vitro studies with in vivo models to reliably predict, in this case, revascularization outcomes. Regardless, we demonstrate here the therapeutic potential of CIs with proangiogenic support cells to enhance islet revascularization at a clinically relevant, although poorly vascularized, transplantation site.


Assuntos
Células Endoteliais da Veia Umbilical Humana/citologia , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/citologia , Células-Tronco Mesenquimais/citologia , Neovascularização Fisiológica , Técnicas de Cocultura , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Ilhotas Pancreáticas/metabolismo , Células-Tronco Mesenquimais/metabolismo
10.
J Cell Mol Med ; 19(8): 1836-46, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25782016

RESUMO

Clinical islet transplantation is a promising treatment for patients with type 1 diabetes. However, pancreatic islets vary in size and shape affecting their survival and function after transplantation because of mass transport limitations. To reduce diffusion restrictions and improve islet cell survival, the generation of islets with optimal dimensions by dispersion followed by reassembly of islet cells, can help limit the length of diffusion pathways. This study describes a microwell platform that supports the controlled and reproducible production of three-dimensional pancreatic cell clusters of human donor islets. We observed that primary human islet cell aggregates with a diameter of 100-150 µm consisting of about 1000 cells best resembled intact pancreatic islets as they showed low apoptotic cell death (<2%), comparable glucose-responsiveness and increasing PDX1, MAFA and INSULIN gene expression with increasing aggregate size. The re-associated human islet cells showed an a-typical core shell configuration with beta cells predominantly on the outside unlike human islets, which became more randomized after implantation similar to native human islets. After transplantation of these islet cell aggregates under the kidney capsule of immunodeficient mice, human C-peptide was detected in the serum indicating that beta cells retained their endocrine function similar to human islets. The agarose microwell platform was shown to be an easy and very reproducible method to aggregate pancreatic islet cells with high accuracy providing a reliable tool to study cell-cell interactions between insuloma and/or primary islet cells.


Assuntos
Glucose/farmacologia , Ilhotas Pancreáticas/citologia , Animais , Agregação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Tamanho Celular , Sobrevivência Celular , Células Cultivadas , Feminino , Humanos , Insulinoma/patologia , Masculino , Camundongos SCID , Pessoa de Meia-Idade , Reprodutibilidade dos Testes
11.
J Control Release ; 205: 181-9, 2015 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-25637703

RESUMO

Two linear poly(amido amine)s, pCABOL and pCHIS, prepared by polyaddition of cystamine bisacrylamide (C) with 4-aminobutanol (ABOL) or histamine (HIS), were explored to form alternating multilayer thin films with DNA to obtain functionalized materials with transfection capacity in 2D and 3D. Therefore, COS-7 cells were cultured on top of multilayer films formed by layer-by-layer dipcoating of these polymers with GFP-encoded pDNA, and the effect of the number of layers and cell seeding density on the transfection efficiency was evaluated. Multilayer films with pCABOL were found to be superior to pCHIS in facilitating transfection, which was attributed to higher incorporation of pDNA and release of the transfection agent. High amounts of transfected cells were obtained on pCABOL films, correlating proportionally over a wide range with seeding density. Optimal transfection efficiency was obtained with pCABOL films composed of 10 bilayers. Further increase in the number of bilayers only marginally increased transfection efficiency. Using the optimal multilayer and cell seeding conditions, pCABOL multilayers were fabricated on poly(ε-caprolactone) (PCL), heparinized PCL (PCL-HEP), and poly(lactic acid) (PLA) disks as examples of common biomedical supports. The multilayers were found to completely mask the properties of the original substrates, with significant improvement in cell adhesion, which is especially pronounced for PCL and PLA disks. With all these substrates, transfection efficiency was found to be in the range of 25-50% transfected cells. The pCABOL/pDNA multilayer films can also conveniently add transfection capability to 3D scaffolds. Significant improvement in cell adhesion was observed after multilayer coating of 3D-plotted fibers of PCL (with and without an additional covalent heparin layer), especially for the PCL scaffold without heparin layer and transfection was observed on both 3D PCL and PCL-HEP scaffolds. These results show that layer-by-layer dip-coating of pCABOL with functional DNA is an easy and inexpensive method to introduce transfection capability to biomaterials of any nature and shape, which can be beneficially used in various biomedical and tissue engineering applications.


Assuntos
Acrilamidas/química , Amino Álcoois/química , Cistamina/química , Histamina/química , Plasmídeos/metabolismo , Alicerces Teciduais , Transfecção/métodos , Animais , Células COS , Adesão Celular , Técnicas de Cultura de Células , Chlorocebus aethiops , Cistamina/análogos & derivados , Regulação da Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Heparina/química , Histamina/análogos & derivados , Ácido Láctico/química , Plasmídeos/química , Plasmídeos/genética , Poliaminas/química , Poliésteres/química , Polímeros/química
12.
PLoS One ; 9(8): e105171, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25133500

RESUMO

The main challenge in hepatic tissue engineering is the fast dedifferentiation of primary hepatocytes in vitro. One successful approach to maintain hepatocyte phenotype on the longer term is the cultivation of cells as aggregates. This paper demonstrates the use of an agarose micro-well chip for the high throughput generation of hepatocyte aggregates, uniform in size. In our study we observed that aggregation of hepatocytes had a beneficial effect on the expression of certain hepatocyte specific markers. Moreover we observed that the beneficial effect was dependent on the aggregate dimensions, indicating that aggregate parameters should be carefully considered. In a second part of the study, the selected aggregates were immobilized by encapsulation in methacrylamide-modified gelatin. Phenotype evaluations revealed that a stable hepatocyte phenotype could be maintained during 21 days when encapsulated in the hydrogel. In conclusion we have demonstrated the beneficial use of micro-well chips for hepatocyte aggregation and the size-dependent effects on hepatocyte phenotype. We also pointed out that methacrylamide-modified gelatin is suitable for the encapsulation of these aggregates.


Assuntos
Técnicas de Cultura de Células , Hepatócitos/citologia , Fígado/citologia , Engenharia Tecidual/métodos , Animais , Sobrevivência Celular/fisiologia , Células Cultivadas , Células Hep G2 , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato , Camundongos , Reação em Cadeia da Polimerase em Tempo Real
13.
PLoS One ; 8(10): e78148, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24167603

RESUMO

Intrahepatic transplantation of donor islets of Langerhans is a promising therapy for patients with type 1 diabetes. It is of critical importance to accurately monitor islet quality before transplantation, which is currently done by standard histological methods that are performed off-line and require extensive sample preparation. As an alternative, we propose Raman spectroscopy which is a non-destructive and label-free technique that allows continuous real-time monitoring of the tissue to study biological changes as they occur. By performing Raman spectroscopic measurements on purified insulin and glucagon, we showed that the 520 cm(-1) band assigned to disulfide bridges in insulin, and the 1552 cm(-1) band assigned to tryptophan in glucagon are mutually exclusive and could therefore be used as indirect markers for the label-free distinction between both hormones. High-resolution hyperspectral Raman imaging for these bands showed the distribution of disulfide bridges and tryptophan at sub-micrometer scale, which correlated with the location of insulin and glucagon as revealed by conventional immunohistochemistry. As a measure for this correlation, quantitative analysis was performed comparing the Raman images with the fluorescence images, resulting in Dice coefficients (ranging between 0 and 1) of 0.36 for insulin and 0.19 for glucagon. Although the use of separate microscope systems with different spatial resolution and the use of indirect Raman markers cause some image mismatch, our findings indicate that Raman bands for disulfide bridges and tryptophan can be used as distinctive markers for the label-free detection of insulin and glucagon in human islets of Langerhans.


Assuntos
Glucagon/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Análise Espectral Raman/métodos , Animais , Linhagem Celular Tumoral , Glucagon/análise , Humanos , Insulina/análise , Ilhotas Pancreáticas/citologia , Camundongos , Ratos
14.
Tissue Eng Part A ; 19(15-16): 1817-28, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23530894

RESUMO

Human mesenchymal stromal cells (hMSCs) offer great potential for bone tissue engineering applications, but their in vivo performance remains limited. Preconditioning of these cells with small molecules to improve their differentiation before implantation, or incorporation of growth factors are possible solutions. Insulin-like growth factor-1 (IGF-1) is one of the most abundant growth factors in bone, involved in growth, development, and metabolism, but its effects on hMSCs are still subject of debate. Here we examined the effects of IGF-1 on proliferation and differentiation of hMSCs in vitro and we found that serum abolished the effects of IGF-1. Only in the absence of serum, IGF-1 increased proliferation, alkaline phosphatase expression, and osteogenic gene expression of hMSCs. Furthermore, we examined synergistic effects of bone morphogenetic protein-2 (BMP-2) and IGF-1 and, although IGF-1 enhanced BMP-2-induced mineralization, IGF-1 only slightly affected in vivo bone formation.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/farmacologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/ultraestrutura , Microscopia Eletroquímica de Varredura
15.
J Tissue Eng Regen Med ; 5(3): 180-90, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20718035

RESUMO

For more than a decade, human mesenchymal stem cells (hMSCs) have been used in bone tissue-engineering research. More recently some of the focus in this field has shifted towards the use of embryonic stem cells. While it is well known that hMSCs are able to form bone when implanted subcutaneously in immune-deficient mice, the osteogenic potential of embryonic stem cells has been mainly assessed in vitro. Therefore, we performed a series of studies to compare the in vitro and in vivo osteogenic capacities of human and mouse embryonic stem cells to those of hMSCs. Embryonic and mesenchymal stem cells showed all characteristic signs of osteogenic differentiation in vitro when cultured in osteogenic medium, including the deposition of a mineralized matrix and expression of genes involved in osteogenic differentiation. As such, based on the in vitro results, osteogenic ES cells could not be discriminated from osteogenic hMSCs. Nevertheless, although osteogenic hMSCs formed bone upon implantation, osteogenic cells derived from both human and mouse embryonic stem cells did not form functional bone, indicated by absence of osteocytes, bone marrow and lamellar bone. Although embryonic stem cells show all signs of osteogenic differentiation in vitro, it appears that, in contrast to mesenchymal stem cells, they do not possess the ability to form bone in vivo when a similar culture method and osteogenic differentiation protocol was applied.


Assuntos
Células da Medula Óssea/citologia , Células-Tronco Embrionárias/citologia , Células-Tronco Mesenquimais/citologia , Osteócitos/citologia , Osteogênese/fisiologia , Fosfatase Alcalina/metabolismo , Animais , Calcificação Fisiológica/efeitos dos fármacos , Fosfatos de Cálcio/farmacologia , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/ultraestrutura , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/enzimologia , Camundongos , Osteócitos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Análise Espectral Raman
16.
Int J Nanomedicine ; 2(2): 241-52, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17722552

RESUMO

Micro- and nanospheres composed of biodegradable polymers show promise as versatile devices for the controlled delivery of biopharmaceuticals. Whereas important properties such as drug release profiles, biocompatibility, and (bio)degradability have been determined for many types of biodegradable particles, information about particle degradation inside phagocytic cells is usually lacking. Here, we report the use of confocal Raman microscopy to obtain chemical information about cross-linked dextran hydrogel microspheres and amphiphilic poly(ethylene glycol)-terephthalate/poly(butylene terephthalate) (PEGT/PBT) microspheres inside RAW 264.7 macrophage phagosomes. Using quantitative Raman microspectroscopy, we show that the dextran concentration inside phagocytosed dextran microspheres decreases with cell incubation time. In contrast to dextran microspheres, we did not observe PEGT/PBT microsphere degradation after 1 week of internalization by macrophages, confirming previous studies showing that dextran microsphere degradation proceeds faster than PEGT/PBT degradation. Raman microscopy further showed the conversion of macrophages to lipid-laden foam cells upon prolonged incubation with both types of microspheres, suggesting that a cellular inflammatory response is induced by these biomaterials in cell culture. Our results exemplify the power of Raman microscopy to characterize microsphere degradation in cells and offer exciting prospects for this technique as a noninvasive, label-free optical tool in biomaterials histology and tissue engineering.


Assuntos
Dextranos/farmacocinética , Macrófagos/citologia , Macrófagos/metabolismo , Fagocitose/fisiologia , Poliésteres/farmacocinética , Polietilenoglicóis/farmacocinética , Análise Espectral Raman/métodos , Animais , Linhagem Celular , Hidrogéis/farmacocinética , Camundongos , Microesferas
17.
J Gerontol A Biol Sci Med Sci ; 62(1): 34-40, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17301035

RESUMO

Although it has been established that premature aging trichothiodystrophy (TTD) mice display typical signs of osteoporosis, exact changes in physicochemical properties of these mice have not been elucidated. We used confocal Raman microscopy and histology to study femora of TTD mice. We measured femora isolated from xeroderma pigmentosum group A (XPA)/TTD double mutant mice to establish that Raman microscopy can be applied to measure differences in bone composition. Raman data from XPA/TTD mice showed remarkable changes in bone mineral composition. Moreover, we observed a severe form of osteoporosis, with strongly reduced cortical bone thickness. We used Raman microscopy to analyze bone composition in eight wild-type and eight TTD animals, and observed decreased levels of phosphate and carbonate in the cortex of femora isolated from TTD mice. In contrast, the bands representing the bone protein matrix were not affected in these mice.


Assuntos
Senilidade Prematura , Carbonatos/análise , Colágeno/análise , Fêmur/química , Doenças do Cabelo/metabolismo , Osteoporose , Fosfatos/análise , Senilidade Prematura/etiologia , Senilidade Prematura/metabolismo , Senilidade Prematura/patologia , Animais , Densidade Óssea , Matriz Óssea/química , Modelos Animais de Doenças , Fêmur/patologia , Cabelo/anormalidades , Doenças do Cabelo/complicações , Doenças do Cabelo/genética , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Osteoporose/complicações , Osteoporose/metabolismo , Osteoporose/patologia , Análise Espectral Raman , Xeroderma Pigmentoso/complicações , Xeroderma Pigmentoso/metabolismo , Xeroderma Pigmentoso/patologia
18.
J Am Chem Soc ; 126(41): 13226-7, 2004 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-15479068

RESUMO

Understanding the degradation behavior of polymeric microspheres is crucial for the successful application of such devices in controlled drug delivery. The degradation mechanism of poly(lactic-co-glycolic acid) (PLGA) microspheres inside phagocytic cells is not known, but different models for degradation in aqueous solution have been proposed. We have used confocal Raman spectroscopy and imaging to study the intracellular degradation of PLGA microspheres inside individual macrophages. Our results show that ingested microspheres degrade in a heterogeneous manner, with a more rapid degradation in the center. Comparison of Raman spectra from degrading beads with those of uningested beads reveals that ester hydrolysis occurs throughout the phagocytosed microspheres, with a selective loss of glycolic acid units. Furthermore, we show that PLGA degradation is a cell-mediated process, possibly caused by the low pH of the phagosome and/or the presence of hydrolytic enzymes. In conclusion, we have demonstrated that the chemical composition of degrading polymers inside cells can be probed by Raman spectral imaging. This technique will expand the capabilities of investigating biomaterial degradation in vivo.


Assuntos
Materiais Biocompatíveis/farmacocinética , Glicolatos/farmacocinética , Macrófagos/metabolismo , Animais , Materiais Biocompatíveis/química , Linhagem Celular , Glicolatos/química , Humanos , Ácido Láctico , Camundongos , Microscopia Confocal , Microscopia Eletrônica de Varredura , Microesferas , Fagocitose , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Análise Espectral Raman
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA