Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Exp Dermatol ; 31(6): 906-917, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35119146

RESUMO

Androgenetic alopecia (AGA) is a prevalent hair loss condition in males that develops due to the influence of androgens and genetic predisposition. With the aim of elucidating genes involved in AGA pathogenesis, we modelled AGA with three-dimensional culture of keratinocyte-surrounded dermal papilla (DP) cells. We co-cultured immortalised balding and non-balding human DP cells (DPCs) derived from male AGA patients with epidermal keratinocyte (NHEK) using multi-interfacial polyelectrolyte complexation technique. We observed up-regulated mitochondria-related gene expression in balding compared with non-balding DP aggregates which indicated altered mitochondria metabolism. Further observation of significantly reduced electron transport chain complex activity (complexes I, IV and V), ATP levels and ability to uptake metabolites for ATP generation demonstrated compromised mitochondria function in balding DPC. Balding DP was also found to be under significantly higher oxidative stress than non-balding DP. Our experiments suggest that application of antioxidants lowers oxidative stress levels and improves metabolite uptake in balding DPC. We postulate that the observed up-regulation of mitochondria-related genes in balding DP aggregates resulted from an over-compensatory effort to rescue decreased mitochondrial function in balding DP through the attempted production of new functional mitochondria. In all, our three-dimensional co-culturing revealed mitochondrial dysfunction in balding DPC, suggesting a metabolic component in the aetiology of AGA.


Assuntos
Alopecia , Androgênios , Trifosfato de Adenosina/metabolismo , Alopecia/patologia , Androgênios/metabolismo , Folículo Piloso/metabolismo , Humanos , Queratinócitos/metabolismo , Masculino , Mitocôndrias/metabolismo
3.
J Biomed Mater Res A ; 109(5): 745-753, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-32677207

RESUMO

Acellular scaffolds from decellularized donor organs are showing promising clinical results in tissue and organ repair and regeneration. A successful decellularization process is determined by (a) its capability to decellularize complete organs of large animals, (b) retention of the extracellular matrix (ECM) structures and morphologies, and (c) minimal loss of ECM proteins. In this study, porcine esophagi were perfused in full thickness with 0.25% w/v sodium dodecyl sulfate at perfusion rates 0.1-0.2 ml/min for up to 5 days. Decellularized tissues were characterized for their residual DNA, histological staining for their matrix structures, immunohistochemical staining for collagen type IV and laminin, and scanning electron microscopy for structural integrity. Our results showed that full thickness esophageal tissues treated using the horizontal perfusion setup were decellularized with good structural and biochemical integrity in the ECM. Residual DNA content in decellularized tissues was found to be 36 ± 12 ng/mg of tissues (n = 6) which was significantly lower than that of native tissues (p = .00022). Our study showed that the organ must be decellularized in full thickness and perfusion pressure must be controlled to minimize radial expansion. These factors were found to be critical in preserving the folded mucosa in the decellularized tissues.


Assuntos
Materiais Biocompatíveis/isolamento & purificação , Mucosa Esofágica/ultraestrutura , Perfusão/métodos , Animais , Desenho de Equipamento , Mucosa Esofágica/citologia , Mucosa Esofágica/efeitos dos fármacos , Microscopia Eletrônica , Perfusão/instrumentação , Dodecilsulfato de Sódio/farmacologia , Tensoativos/farmacologia , Suínos , Alicerces Teciduais
4.
Biofabrication ; 9(2): 025011, 2017 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-28393762

RESUMO

Cardiotoxicity is one of the major reasons for clinical drug attrition. In vitro tissue models that can provide efficient and accurate drug toxicity screening are highly desired for preclinical drug development and personalized therapy. Here, we report the fabrication and characterization of a human cardiac tissue model for high throughput drug toxicity studies. Cardiac tissues were fabricated via cellular self-assembly of human transgene-free induced pluripotent stem cells-derived cardiomyocytes in pre-fabricated polydimethylsiloxane molds. The formed tissue constructs expressed cardiomyocyte-specific proteins, exhibited robust production of extracellular matrix components such as laminin, collagen and fibronectin, aligned sarcomeric organization, and stable spontaneous contractions for up to 2 months. Functional characterization revealed that the cardiac cells cultured in 3D tissues exhibited higher contraction speed and rate, and displayed a significantly different drug response compared to cells cultured in age-matched 2D monolayer. A panel of clinically relevant compounds including antibiotic, antidiabetic and anticancer drugs were tested in this study. Compared to conventional viability assays, our functional contractility-based assays were more sensitive in predicting drug-induced cardiotoxic effects, demonstrating good concordance with clinical observations. Thus, our 3D cardiac tissue model shows great potential to be used for early safety evaluation in drug development and drug efficiency testing for personalized therapy.


Assuntos
Modelos Biológicos , Engenharia Tecidual , Alicerces Teciduais/química , Antibacterianos/toxicidade , Antineoplásicos/toxicidade , Técnicas de Cultura de Células , Diferenciação Celular , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Colágeno/química , Dimetilpolisiloxanos/química , Combinação de Medicamentos , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibronectinas/química , Humanos , Hipoglicemiantes/toxicidade , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Cariótipo , Laminina/química , Microscopia de Fluorescência , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Proteoglicanas/química
5.
Acta Biomater ; 46: 266-277, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27667015

RESUMO

The use of human induced pluripotent stem cells (hiPSCs) for clinical tissue engineering applications requires expansion and differentiation of the cells using defined, xeno-free substrates. The screening and selection of suitable synthetic substrates however, is tedious, as their performance relies on the inherent material properties. In the present work, we demonstrate an alternative concept for xeno-free expansion and differentiation of hiPSCs using synthetic substrates, which hinges on the structure-function relationship between electrospun polystyrene scaffolds (ESPS) and pluripotent stem cell growth. ESPS of differential porosity was obtained by fusing the fibers at different temperatures. The more porous, loosely fused scaffolds were found to efficiently trap the cells, leading to a large number of three-dimensional (3D) aggregates which were shown to be pluripotent colonies. Immunostaining, PCR analyses, in vitro differentiation and in vivo teratoma formation studies demonstrated that these hiPSC aggregates could be cultured for up to 10 consecutive passages (P10) with maintenance of pluripotency. Flow cytometry showed that more than 80% of the cell population stained positive for the pluripotent marker OCT4 at P1, P5 and P10. P10 cells could be differentiated to neuronal-like cells and cultured within the ESPS for up to 18months. Our results suggest the usefulness of a generic class of synthetic substrates, exemplified by ESPS, for 'trapped aggregate culture' of hiPSCs. STATEMENT OF SIGNIFICANCE: To realize the potential of human induced pluripotent stem cells (hiPSCs) in clinical medicine, robust, xeno-free substrates for expansion and differentiation of iPSCs are required. In the existing literature, synthetic materials have been reported that meet the requirement for non-xenogeneic substrates. However, the self-renewal and differentiation characteristics of hiPSCs are affected differently by the biocompatibility and physico-chemical properties of individual substrates. Although some rules based on chemical structure and substrate rigidity have been developed, most of these efforts are still empirical, and most synthetic substrates must still be rigorously screened for suitability. In this paper, we demonstrate an alternative concept for xeno-free expansion and differentiation of hiPSCs using synthetic substrates, which hinges on the structure-function relationship between electrospun polystyrene scaffolds (ESPS) and pluripotent stem cell growth. ESPS of differential porosity was obtained by fusing the fibers at different temperatures. The more porous, loosely fused scaffold was found to efficiently trap the cells, leading to a large number of three-dimensional (3D) aggregates. In the form of these trapped aggregates, we showed that hiPSCs could be cultured for up to 10 consecutive passages (P10) with maintenance of pluripotency, following which they could be differentiated to a chosen lineage. We believe that this novel, generic class of synthetic substrates that employs 'trapped aggregate culture' for expansion and differentiation of hiPSCs is an important conceptual advance, and would be of high interest to the readership of Acta Biomaterialia.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/citologia , Poliestirenos/farmacologia , Alicerces Teciduais/química , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/ultraestrutura , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Porosidade
6.
Tissue Eng Part C Methods ; 22(9): 884-94, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27484909

RESUMO

Pluripotent human embryonic stem cells (hESCs) are a potential renewable cell source for regenerative medicine and drug testing. To obtain adequate cell numbers for these applications, there is a need to develop scalable cell culture platforms to propagate hESCs. In this study, we encapsulated hESCs in calcium alginate microfibers as single cells, for expansion and differentiation under chemically defined conditions. hESCs were suspended in 1% (w/v) alginate solution at high cell density (>10(7) cells/mL) and extruded at 5 m/min into a low calcium concentration bath (10 mM) for gelation. Mild citrate buffer (2.5 mM), which did not affect hESCs viability, was used to release the cells from the calcium alginate hydrogel. Encapsulation as single cells was critical, as this allowed the hESCs to grow in the form of relatively small and uniform aggregates. This alginate microfiber system allowed for expansion of an hESC line, HUES7, for up to five passages while maintaining pluripotency. Immunohistochemistry, polymerase chain reaction, and other analyses showed that passage 5 (P5) HUES7 cells expressed proteins and genes characteristic of pluripotent stem cells, possessed normal karyotype, and were able to form representative tissues of the three embryonic germ layers in vitro and in vivo. Encapsulated HUES7 cells at P5 could also be induced to directly differentiate into liver-like cells. Collectively, our experiments show that the alginate microfiber system can be used as a three-dimensional cell culture platform for long-term expansion and differentiation of hESCs under defined conditions.


Assuntos
Alginatos/química , Diferenciação Celular , Células Imobilizadas , Células-Tronco Embrionárias Humanas/citologia , Células Cultivadas , Ácido Glucurônico/química , Ácidos Hexurônicos/química , Humanos
7.
Adv Healthc Mater ; 5(16): 2080-91, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27294565

RESUMO

Recent advances in developmental biology and stem cell technology have led to the engineering of functional organs in a dish. However, the limited size of these organoids and absence of a large circulatory system poses limits to its clinical translation. To overcome these issues, decellularized whole kidney scaffolds with native microstructure and extracellular matrix (ECM) are employed for kidney bioengineering, using human-induced pluripotent-stem-cell-derived renal progenitor cells and endothelial cells. To demonstrate ECM-guided cellular assembly, the present work is focused on generating the functional unit of the kidney, the glomerulus. In the repopulated organ, the presence of endothelial cells broadly upregulates the expression level of genes related to renal development. When the cellularized native scaffolds are implanted in SCID mice, glomeruli assembly can be achieved by co-culture of the renal progenitors and endothelial cells. These individual glomerular units are shown to be functional in the context of the whole organ using a simulated bio-reactor set-up with urea and creatinine excretion and albumin reabsorption. Our results indicate that the repopulation of decellularized native kidney using clinically relevant, expandable patient-specific renal progenitors and endothelial cells may be a viable approach for the generation of a functional whole kidney.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Rim , Engenharia Tecidual , Alicerces Teciduais/química , Animais , Humanos , Rim/química , Rim/metabolismo , Camundongos
8.
Biomed Mater ; 11(2): 025013, 2016 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-27041648

RESUMO

Repair of critical-size articular cartilage defects typically involves delivery of cells in biodegradable, 3D matrices. Differences in the developmental status of mesenchymal stem cells (MSCs) and terminally differentiated mature chondrocytes might be a critical factor in engineering appropriate 3D matrices for articular cartilage tissue engineering. This study examined the relationship between material-driven early cell morphological adaptations and chondrogenic outcomes, by studying the influence of aligned collagen type I (Col I) presentation on chondrocytes and MSC in interfacial polyelectrolyte complexation (IPC)-based hydrogels. In the absence of Col I, both chondrocytes and MSCs adopted rounded cell morphology and formed clusters, with chondrocyte clusters favoring the maintenance of hyaline phenotype, while MSC clusters differentiated to fibro-superficial zone-like chondrocytes. Encapsulated chondrocytes in IPC-Col I hydrogel adopted a fibroblastic morphology forming fibro-superficial zone-like phenotype, which could be reversed by inhibiting actin polymerization using cytochalasin D (CytD). In contrast, adoption of fibroblastic morphology by encapsulated MSCs in IPC-Col I facilitated superior chondrogenesis, generating a mature, hyaline neocartilage tissue. CytD treatment abrogated the elongation of MSCs and brought about a single cell-like state, resulting in insignificant chondrogenic differentiation, underscoring the essential requirement of providing matrix environments that are amenable to cell-cell interactions for robust MSC chondrogenic differentiation. Our study demonstrates that MSCs and culture-expanded chondrocytes favour differential microenvironmental niches and emphasizes the importance of designing biomaterials that meet cell type-specific requirements, in adopting chondrocyte or MSC-based approaches for regenerating hyaline, articular cartilage.


Assuntos
Condrócitos/citologia , Condrogênese , Células-Tronco Mesenquimais/citologia , Adaptação Fisiológica , Materiais Biocompatíveis/química , Cartilagem Articular/citologia , Diferenciação Celular , Forma Celular , Células Cultivadas , Microambiente Celular , Condrócitos/fisiologia , Condrogênese/genética , Condrogênese/fisiologia , Matriz Extracelular/metabolismo , Expressão Gênica , Humanos , Hidrogéis , Teste de Materiais , Células-Tronco Mesenquimais/fisiologia , Fenótipo , Polieletrólitos/química , Engenharia Tecidual/métodos
9.
Biomaterials ; 78: 62-73, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26684838

RESUMO

A hierarchy of cellular stemness exists in certain cancers, and any successful strategy to treat such cancers would have to eliminate the self-renewing tumor-initiating cells at the apex of the hierarchy. The cellular microenvironment, in particular the extracellular matrix (ECM), is believed to have a role in regulating stemness. In this work, U251 glioblastoma cells are cultured on electrospun polystyrene (ESPS) scaffolds coated with an array of 7 laminin isoforms to provide a 3D model for stem cell-related genes and proteins expression studies. We observed collaboration between 3D context and laminins in promoting glioma stemness. Depending on the laminin isoform presented, U251 cells cultured on ESPS scaffolds (3D) exhibited increased expression of stemness markers compared to those cultured on tissue culture polystyrene (2D). Our results indicate the influence of 3D (versus 2D) context on integrin expression, specifically, the upregulation of the laminin-binding integrins alpha 6 and beta 4. By a colony forming assay, we showed enhanced clonogenicity of cells grown on ESPS scaffolds in collaboration with laminins 411, 421, 511 and 521. Evaluation of patient glioma databases demonstrated significant enrichment of integrin and ECM pathway networks in tumors of worse prognosis, consistent with our observations. The present results demonstrate how 3D versus 2D context profoundly affects ECM signaling, leading to stemness.


Assuntos
Neoplasias Encefálicas/patologia , Matriz Extracelular/patologia , Glioma/patologia , Modelos Biológicos , Células-Tronco Neoplásicas/patologia , Transportadores de Cassetes de Ligação de ATP/metabolismo , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Humanos , Microscopia Eletrônica de Varredura
10.
Lancet Oncol ; 16(2): e84-92, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25638684

RESUMO

Removal of malignant tissue in patients with oesophageal cancer and replacement with autologous grafts from the stomach and colon can lead to problems. The need to reduce stenosis and anastomotic leakage after oesophagectomy is a high priority. Developments in tissue-engineering methods and cell-sheet technology have improved scaffold materials for oesophageal repair. Despite the many successful animal studies, few tissue-engineering approaches have progressed to clinical trials. In this Review, we discuss the status of oesophagus reconstruction after surgery. In particular, we highlight two clinical trials that used decellularised constructs and epithelial cell sheets to replace excised tissues after endoscopic submucosal dissection or mucosal resection procedures. Results from the trials showed that both decellularised grafts and epithelial-cell sheets prevented stenosis. By contrast, animal studies have shown that the use of tissue-engineered constructs after oesophagectomy remains a challenge.


Assuntos
Neoplasias Esofágicas/cirurgia , Procedimentos de Cirurgia Plástica , Medicina Regenerativa , Engenharia Tecidual , Humanos
11.
Biomaterials ; 35(23): 6006-14, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24780169

RESUMO

Liver tissue engineering requires a suitable cell source, methodologies to assemble the cells within their niche microenvironments in a spatially defined manner, and vascularization of the construct in vivo for maintenance of hepatocyte viability and function. Recently, we have developed methods of encapsulating cells within separate domains in multi-component hydrogel fibers and methods of assembling fibers to form 3D-patterned tissue constructs. In the present work, we have combined these approaches to encapsulate hepatocytes and endothelial cells within their specific niches, and to assemble them into endothelialized liver tissue constructs. The hepatocytes and endothelial cells were obtained in parallel by differentiating human recombinant protein-induced human pluripotent stem cells, resulting in a construct which contained genetically identical endothelial and parenchymal elements. We were able to demonstrate that the presence of endothelial cells in the scaffold significantly improved hepatocyte function in vitro and facilitated vascularization of the scaffold when implanted in a mouse partial hepatectomy model. The in vivo studies further asserted that integration of the scaffold with host vasculature had occurred, as demonstrated by the presence of human albumin in the mouse serum.


Assuntos
Células Endoteliais/citologia , Hepatócitos/citologia , Hidrogéis/química , Fígado Artificial , Fígado/crescimento & desenvolvimento , Células-Tronco Pluripotentes/citologia , Engenharia Tecidual/instrumentação , Diferenciação Celular/fisiologia , Células Cultivadas , Técnicas de Cocultura/instrumentação , Células Endoteliais/fisiologia , Desenho de Equipamento , Hepatócitos/fisiologia , Humanos , Fígado/citologia , Nanofibras/química , Nanofibras/ultraestrutura , Neovascularização Fisiológica/fisiologia , Células-Tronco Pluripotentes/fisiologia , Alicerces Teciduais
12.
Biomaterials ; 35(9): 2816-26, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24411336

RESUMO

A defined xeno-free system for patient-specific iPSC derivation and differentiation is required for translation to clinical applications. However, standard somatic cell reprogramming protocols rely on using MEFs and xenogeneic medium, imposing a significant obstacle to clinical translation. Here, we describe a well-defined culture system based on xeno-free media and LN521 substrate which supported i) efficient reprogramming of normal or diseased skin fibroblasts from human of different ages into hiPSCs with a 15-30 fold increase in efficiency over conventional viral vector-based method; ii) long-term self-renewal of hiPSCs; and iii) direct hiPSC lineage-specific differentiation. Using an excisable polycistronic vector and optimized culture conditions, we achieved up to 0.15%-0.3% reprogramming efficiencies. Subsequently, transgene-free hiPSCs were obtained by Cre-mediated excision of the reprogramming factors. The derived iPSCs maintained long-term self-renewal, normal karyotype and pluripotency, as demonstrated by the expression of stem cell markers and ability to form derivatives of three germ layers both in vitro and in vivo. Importantly, we demonstrated that Parkinson's patient transgene-free iPSCs derived using the same system could be directed towards differentiation into dopaminergic neurons under xeno-free culture conditions. Our approach provides a safe and robust platform for the generation of patient-specific iPSCs and derivatives for clinical and translational applications.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular , Células Alimentadoras/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Transgenes , Adulto , Animais , Proliferação de Células , Reprogramação Celular , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/metabolismo , Fibroblastos/citologia , Vetores Genéticos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Lentivirus/metabolismo , Camundongos , Camundongos SCID , Doença de Parkinson/patologia
13.
Biomaterials ; 35(9): 2607-16, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24388815

RESUMO

Cell-cell and cell-matrix interactions are important events in directing stem cell chondrogenesis, which can be promoted in matrix microenvironments presenting appropriate ligands. In this study, interfacial polyelectrolyte complexation (IPC) based hydrogels were employed, wherein the unique formation of submicron size fibers facilitated spatial orientation of ligands within such hydrogels. The influence of aligned, collagen type I (Col I) presentation in IPC hydrogel on chondrogenic differentiation of human mesenchymal stem cells (MSC) was investigated. Early morphological dynamics, onset of N-cadherin/ß-catenin mediated chondrogenic induction and differentiation were compared between MSCs encapsulated in IPC-Col I and IPC-control (without Col I) hydrogels, and a conventional Col I hydrogel. MSCs in IPC-Col I hydrogel aligned and packed uniformly, resulting in enhanced cell-cell interactions and cellular condensation, facilitating superior chondrogenesis and the generation of mature hyaline neocartilage, with notable downregulation of fibrocartilaginous marker. Inhibition study using function blocking ß1-integrin antibodies reversed the aforementioned outcomes, indicating the importance of coupling integrin mediated cell-matrix interactions and N-cadherin/ß-catenin mediated downstream signaling events. This study demonstrated the significance of oriented ligand presentation for MSC chondrogenesis, and the importance of facilitating an orderly sequence of differentiation events, initiated by proximal interactions between MSCs and the extracellular matrix for robust neocartilage formation.


Assuntos
Comunicação Celular/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Eletrólitos/farmacologia , Matriz Extracelular/metabolismo , Hidrogéis/farmacologia , Células-Tronco Mesenquimais/citologia , Engenharia Tecidual/métodos , Animais , Linhagem da Célula/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Colágeno Tipo I/farmacologia , Matriz Extracelular/efeitos dos fármacos , Humanos , Integrina beta1/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Ratos , Alicerces Teciduais/química
14.
Nat Commun ; 4: 2353, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23955534

RESUMO

The in vivo efficacy of engineered tissue constructs depends largely on their integration with the host vasculature. Prevascularisation has been noted to facilitate integration of the constructs via anastomosis of preformed microvascular networks. Here we report a technique to fabricate aligned, spatially defined prevascularised tissue constructs with endothelial vessels by assembling individually tailored cell-laden polyelectrolyte hydrogel fibres. Stable, aligned endothelial vessels form in vitro within these constructs in 24 h, and these vessels anastomose with the host circulation in a mouse subcutaneous model. We create vascularised adipose and hepatic tissues by co-patterning the respective cell types with the preformed endothelial vessels. Our study indicates that the formation of aligned endothelial vessels in a hydrogel is an efficient prevascularisation approach in the engineering of tissue constructs.


Assuntos
Tecido Adiposo/irrigação sanguínea , Endotélio Vascular/metabolismo , Hidrogel de Polietilenoglicol-Dimetacrilato/metabolismo , Fígado/irrigação sanguínea , Engenharia Tecidual/métodos , Animais , Sobrevivência Celular , Células Endoteliais , Células Hep G2 , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Camundongos , Camundongos SCID , Neovascularização Fisiológica
15.
Biomaterials ; 34(29): 7064-72, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23796577

RESUMO

The hair follicle is a regenerating organ that produces a new hair shaft during each growth cycle. Development and cycling of the hair follicle is governed by interactions between the epithelial and mesenchymal components. Therefore, development of an engineered 3D hair follicle would be useful for studying these interactions to identify strategies for treatment of hair loss. We have developed a technique suitable for assembly of different cell types in close proximity in fibrous hydrogel scaffolds with resolutions of ∼50 µm. By assembly of dermal papilla (DP) and keratinocytes, structures similar to the native hair bulb arrangement are formed. Gene expression of these constructs showed up-regulation of molecules involved in epithelial-mesenchymal interactions of the hair follicle. Implantation of the follicular structures in SCID mice led to the formation of hair follicle-like structures, thus demonstrating their hair inductive ability. The transparency of the fiber matrix and the small dimensions of the follicular structures allowed the direct quantitation of DP cell proliferation by confocal microscopy, clearly illustrating the promoting or inhibitory effects of hair growth regulating agents. Collectively, our results suggested a promising application of these 3D engineered follicular structures for in vitro screening and testing of drugs for hair growth therapy.


Assuntos
Derme/citologia , Folículo Piloso/citologia , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Linhagem Celular , Células Epiteliais/citologia , Folículo Piloso/transplante , Humanos , Queratinócitos/citologia , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos SCID
16.
Nanomedicine (Lond) ; 8(4): 555-66, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23560407

RESUMO

BACKGROUND: Conventional electrospun scaffolds have very small pores, thus limiting cellular infiltration, tissue ingrowth and vascularization in tissue engineering applications. The cryogenic electrospinning process overcame the small pore size constraints found in conventional electrospun scaffolds. AIM: The aim of this paper is to propose a mechanism for cryogenic electrospinning and how scaffold pore size can be controlled. MATERIALS & METHODS: We studied the roles of ice crystals in controlling the pore size of cryogenic electrospun scaffolds (CES). Based on this understanding, we have successfully fabricated a bilayered scaffold with distinctly different pore sizes. RESULTS: Our study showed that CES pore size was dependent on the structure of the frost layer formed and hence the factors affecting ice deposition. The bilayered scaffold was able to support the coculture of human dermal fibroblasts and keratinocytes. CONCLUSION: The larger pores of CES add versatility to the use of electrospun scaffolds in tissue engineering applications.


Assuntos
Engenharia Tecidual/métodos , Alicerces Teciduais/química , Fenômenos Biomecânicos
17.
Adv Healthc Mater ; 1(1): 101-5, 2012 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-23184693

RESUMO

In multi-interfacial polyelectrolyte complexation (MIPC), fusion of nascent fibers from multiple interfaces brings the interfaces to a point from which a composite fiber is drawn. MIPC applied to two, three, and four polyelectrolyte complex interfaces leads to various patterned multicomponent fibers. Cells encapsulated in these fibers exhibit migration, aggregation and spreading in relation to the initial cell or matrix pattern.


Assuntos
Materiais Biocompatíveis/química , Técnicas de Cultura de Células/métodos , Eletrólitos/química , Engenharia Tecidual/métodos , Células 3T3 , Animais , Sobrevivência Celular , Teste de Materiais , Camundongos
18.
Biomaterials ; 33(36): 9179-87, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22998816

RESUMO

Developing an efficient culture system for controlled human pluripotent stem cell (hPSC) differentiation into selected lineages is a major challenge in realizing stem cell-based clinical applications. Here, we report the use of chitin-alginate 3D microfibrous scaffolds, previously developed for hPSC propagation, to support efficient neuronal differentiation and maturation under chemically defined culture conditions. When treated with neural induction medium containing Noggin/retinoic acid, the encapsulated cells expressed much higher levels of neural progenitor markers SOX1 and PAX6 than those in other treatment conditions. Immunocytochemisty analysis confirmed that the majority of the differentiated cells were nestin-positive cells. Subsequently transferring the scaffolds to neuronal differentiation medium efficiently directed these encapsulated neural progenitors into mature neurons, as detected by RT-PCR and positive immunostaining for neuron markers ßIII tubulin and MAP2. Furthermore, flow cytometry confirmed that >90% ßIII tubulin-positive neurons was achieved for three independent iPSC and hESC lines, a differentiation efficiency much higher than previously reported. Implantation of these terminally differentiated neurons into SCID mice yielded successful neural grafts comprising MAP2 positive neurons, without tumorigenesis, suggesting a potential safe cell source for regenerative medicine. These results bring us one step closer toward realizing large-scale production of stem cell derivatives for clinical and translational applications.


Assuntos
Células-Tronco Pluripotentes/citologia , Alicerces Teciduais/química , Diferenciação Celular/fisiologia , Linhagem Celular , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Citometria de Fluxo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Imuno-Histoquímica , Regeneração Nervosa/fisiologia , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo
19.
Biomaterials ; 33(8): 2419-30, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22196900

RESUMO

Realizing the potential of human pluripotent stem cell (hPSC)-based therapy requires the development of defined scalable culture systems with efficient expansion, differentiation and isolation protocols. We report an engineered 3D microfiber system that efficiently supports long-term hPSCs self-renewal under chemically defined conditions. The unique feature of this system lies in the application of a 3D ECM-like environment in which cells are embedded, that affords: (i) uniform high cell loading density in individual cell-laden constructs (∼10(7) cells/ml); (ii) quick recovery of encapsulated cells (<10min at 37°C) with excellent preservation of cell viability and 3D multicellular structure; (iii) direct cryopreservation of the encapsulated cells in situ in the microfibers with >17-fold higher cell viability compared to those cultured on Matrigel surface; (iv) long-term hPSC propagation under chemically defined conditions. Four hPSC lines propagated in the microfibrous scaffold for 10 consecutive passages were capable of maintaining an undifferentiated phenotype as demonstrated by the expression of stem cell markers and stable karyotype in vitro and the ability to form derivatives of the three germ layers both in vitro and in vivo. Our 3D microfibrous system has the potential for large-scale cultivation of transplantable hESCs and derivatives for clinical applications.


Assuntos
Alginatos/farmacologia , Quitina/farmacologia , Células-Tronco Pluripotentes/citologia , Alicerces Teciduais/química , Animais , Agregação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Imobilizadas/citologia , Células Imobilizadas/efeitos dos fármacos , Células Imobilizadas/metabolismo , Colágeno/farmacologia , Criopreservação , Combinação de Medicamentos , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/ultraestrutura , Ácido Glucurônico/farmacologia , Ácidos Hexurônicos/farmacologia , Humanos , Laminina/farmacologia , Camundongos , Camundongos SCID , Fenótipo , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Proteoglicanas/farmacologia , Teratoma/patologia , Fatores de Tempo
20.
Biomaterials ; 33(2): 353-64, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22018387

RESUMO

We developed a library of industrial materials, which can be applied to any adherent cell type for determining cell-material interactions. Bulk and surface chemistry as well as other material properties were characterized. The library covered broad ranges of various material properties. We applied the library to primary human endothelial and epithelial cells, which play important roles in tissue engineering and biomedical applications. The results revealed that substrate stiffness was the major determinant of cell performance. The ability to grow and differentiate on stiff or more compliant materials was cell type-dependent, but cell performance was consistently best on stiff and smooth materials. These results give new insights into the nature of substrate-dependent performance of primary human cells and are potentially useful for the development of improved biomaterials. The materials of the library can be easily accessed by the scientific community to determine cell-material interactions of any adherent cell type of interest.


Assuntos
Materiais Biocompatíveis/química , Adesão Celular , Células Endoteliais da Veia Umbilical Humana/citologia , Pesquisa Biomédica , Diferenciação Celular , Proliferação de Células , Elasticidade , Humanos , Interações Hidrofóbicas e Hidrofílicas , Microscopia Eletrônica de Varredura , Polietileno , Porosidade , Reação em Cadeia da Polimerase em Tempo Real , Espectrometria de Massa de Íon Secundário , Espectroscopia de Infravermelho com Transformada de Fourier , Técnicas de Cultura de Tecidos , Engenharia Tecidual/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA