Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Tissue Eng Part A ; 26(11-12): 591-601, 2020 06.
Article in English | MEDLINE | ID: mdl-31739755

ABSTRACT

Secondary alveolar bone grafts (ABGs) are the standard treatment for the alveolar defect in patients with cleft lip and palate (CLP), but remain invasive and have several disadvantages such as delayed timing of alveolar repair, donor-site complications, graft resorption, and need for multiple surgeries. Earlier management of the alveolar defect (primary ABG) would be ideal, but is limited by the minimal bony donor sites available in the infant. In this study we used a critical-size alveolar bone defect model in the rat to investigate the use of Wharton's Jelly (WJ), the stem cell-rich connective tissue matrix of the umbilical cord, to generate bone within the alveolar cleft. Human WJ was isolated and implanted into a critical-size alveolar bone defect model representative of secondary cleft ABG surgery in 10-11-week-old male Sprague-Dawley rats. The defects were monitored with CT imaging of living animals to evaluate bone regrowth and healing over 24 weeks, followed by histomorphometric evaluation at 24 weeks, after the last CT scan. CT data confirmed that the defect size was critical and did not lead to the union of the bones in the control animals (n = 12) for the entire duration of the study. New bone growth was stimulated leading to partial-to-full closure of the defect in the animals treated with WJ (n = 12). Twenty four weeks postoperatively, the percent increase in new bone formation in the WJ-treated group (156.58% ± 20.67%) was markedly higher than that in the control group (50.36% ± 21.07%) (p < 0.05). Histomorphometric data also revealed significantly greater new bone formation in WJ-treated versus control animals, confirming CT findings. qPCR analysis of human Alu elements was unable to detect any appreciable long-term persistence of human cells in the new bone, indicating that WJ may enhance bone growth by mediating osteoinduction in the host tissue, rather than through osteogenic differentiation of WJ-embedded cells. Impact statement In this study, Wharton's Jelly enhanced bone growth in a preclinical alveolar defect model, indicating its potential use as a natural adjunct in the repair of the alveolar cleft defect in patients with cleft lip and palate (CLP). The clinical success of this approach would represent a paradigm shift in the treatment of patients with CLP by reducing or eliminating the need for subsequent secondary alveolar bone graft and reducing their number of lifetime surgeries.


Subject(s)
Cleft Palate/surgery , Wharton Jelly , Animals , Bone Regeneration/physiology , Cell Differentiation/physiology , Cell Survival/physiology , Cells, Cultured , Humans , Osteogenesis/physiology , Rats , Rats, Sprague-Dawley , X-Ray Microtomography
2.
Biotechniques ; 61(4): 206-209, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27712584

ABSTRACT

Immobilizing hydrated soft tissue specimens for atomic force microscopy (AFM) is a challenge. Here, we describe a simple and very cost-effective immobilization method, based on the use of transglutaminase in an aqueous environment, and successfully apply it to AFM characterization of human native Wharton's Jelly (nWJ), the gelatinous connective tissue matrix of the umbilical cord. A side-by-side comparison with a widely used polyphenolic protein-based tissue adhesive (Corning Cell-Tak), which is known to bind strongly to virtually all inorganic and organic surfaces in aqueous environments, shows that both adhesives successfully immobilize nWJ in its physological hydrated state. The cost of transglutaminase, however, is over 3000-fold lower than that of Cell-Tak, making it a very attractive method for immobilizing soft tissues for AFM characterization.


Subject(s)
Histocytological Preparation Techniques/methods , Microscopy, Atomic Force/methods , Wharton Jelly/diagnostic imaging , Wharton Jelly/physiology , Biocompatible Materials , Biomechanical Phenomena , Elastic Modulus , Humans , Tissue Adhesives
3.
Tissue Eng Part A ; 19(21-22): 2330-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23668629

ABSTRACT

Adipose-derived stem cells (ADSCs) possess potent angiogenic properties and represent a source for cell-based approaches to delivery of bioactive factors to drive vascularization of tissues. Hypoxic signaling appears to be largely responsible for triggering release of these angiogenic cytokines, including vascular endothelial growth factor (VEGF). Three-dimensional (3D) culture may promote activation of hypoxia-induced pathways, and has furthermore been shown to enhance cell survival by promoting cell-cell interactions while increasing angiogenic potential. However, the development of hypoxia within ADSC spheroids is difficult to characterize. In the present study, we investigated the impact of spheroid size on hypoxia-inducible transcription factor (HIF)-1 activity in spheroid cultures under atmospheric and physiological oxygen conditions using a fluorescent marker. Hypoxia could be induced and modulated by controlling the size of the spheroid; HIF-1 activity increased with spheroid size and with decreasing external oxygen concentration. Furthermore, VEGF secretion was impacted by the hypoxic status of the culture, increasing with elevated HIF-1 activity, up to the point at which viability was compromised. Together, these results suggest the ability to use 3D culture geometry as a means to control output of angiogenic factors from ADSCs, and imply that at a particular environmental oxygen concentration an optimal culture size for cytokine production exists. Consideration of culture geometry and microenvironmental conditions at the implantation site will be important for successful realization of ADSCs as a pro-angiogenic therapy.


Subject(s)
Adipocytes/cytology , Stem Cells/cytology , Stem Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cell Survival/physiology , Cells, Cultured , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
4.
Tissue Eng Part A ; 19(13-14): 1583-91, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23394201

ABSTRACT

Tissue engineering is a promising approach to repair critical-size defects in bone. Damage to vasculature at the defect site can create a lower O2 environment compared with healthy bone. Local O2 levels influence stem cell behavior, as O2 is not only a nutrient, but also a signaling molecule. The hypoxia-inducible factor-1 (HIF-1) is a transcription factor that regulates a wide range of O2-related genes and its contribution in bone repair/formation is an important area that can be exploited. In this study, we examined the effect of low O2 environments (1% and 2% O2) on the osteogenic differentiation of adipose-derived stem cells in both two-dimensional (2-D) and three-dimensional (3-D) culture systems. To determine the role of HIF-1 in the differentiation process, an inhibitor was used to block the HIF-1 activity. The samples were examined for osteogenesis markers as measured by quantification of the alkaline phosphatase (ALP) activity, mineral deposition, and expression of osteonectin (ON) and osteopontin (OPN). Results show a downregulation of the osteogenic markers (ALP activity, mineralization, ON, OPN) in both 1% and 2% O2 when compared to 20% O2 in both 2-D and 3-D culture. Vascular endothelial growth factor secretion over 28 days was significantly higher in low O2 environments and HIF-1 inhibition reduced this effect. The inhibition of the HIF-1 activity did not have a significant impact on the expression of the osteogenic markers, suggesting HIF-1-independent inhibition of osteogenic differentiation in hypoxic conditions.


Subject(s)
Adipose Tissue/cytology , Hypoxia-Inducible Factor 1/metabolism , Stem Cells/cytology , Cell Differentiation/physiology , Cells, Cultured , Humans , Osteogenesis/physiology , Osteonectin/metabolism , Osteopontin/metabolism , Tissue Engineering/methods
5.
Int J Artif Organs ; 36(3): 175-83, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23404645

ABSTRACT

PURPOSE: Hypoxia is a major cause for failure of encapsulated islet grafts. Three-dimensional (3D) re-aggregation and hypoxic preconditioning are used to help overcome this obstacle. However, it is still difficult to identify hypoxic cells in a 3D system. We evaluate the efficacy of a fluorescent system for detecting HIF-1 activity in live ß-cells. Identification of HIF-1 activity and correlation with insulin secretion and viability will allow for more informed implant construction and better prediction of post-transplantational function.
 METHODS: MIN6 cells were infected with the marker virus and rotationally cultured to form clusters. Clusters were encapsulated in PEG hydrogels and incubated in 20%, 2%, or 1% O2. Gels were imaged daily for hypoxia marker signaling and for morphological observation. Daily GSIS was quantified by insulin ELSIA and cell viability was assessed by LIVE/DEAD staining.
 RESULTS: Clusters cultured in 2% and 1% O2 displayed high levels of HIF activity compared to 20% O2 clusters. 20% O2 clusters maintained viability and achieved a smooth, islet-like morphology by Day 14. Clusters in 2% and 1% O2 failed to associate cohesively and showed reduced viability. As a whole, constructs cultured in 20% O2 exhibited 10-fold higher GSIS than constructs in 2% and 1% O2.
 CONCLUSIONS: Our marker is an effective approach for identifying cellular hypoxia in 3D cultures. ß-cell clusters in 2% and 1% O2 are similarly affected by reduced oxygen tension, with HIF-1 activity correlating to reduced GSIS and impaired cell/cluster morphology. Simultaneous aggregative culture and hypoxic conditioning may not be beneficial to ß-cell transplantation.


Subject(s)
Cell Hypoxia/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Animals , Cell Line, Tumor , Cell Shape , Insulin-Secreting Cells/cytology , Insulinoma/metabolism , Islets of Langerhans/cytology , Mice , Pancreatic Neoplasms/metabolism
6.
Tissue Eng Part C Methods ; 18(7): 557-65, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22250882

ABSTRACT

Oxygen is not only a nutrient but also an important signaling molecule whose concentration can influence the fate of stem cells. This study details the development of a marker of hypoxic signaling for use with encapsulated cells. Testing of the marker was performed with adipose-derived stem cells (ADSCs) in two-dimensional (2D) and 3D culture conditions in varied oxygen environments. The cells were genetically modified with our hypoxia marker, which produces a red fluorescent protein (DsRed-DR), under the control of a hypoxia-responsive element (HRE) trimer. For 3D culture, ADSCs were encapsulated in poly(ethylene glycol)-based hydrogels. The hypoxia marker (termed HRE DsRed-DR) is built on a recombinant adenovirus and ADSCs infected with the marker will display red fluorescence when hypoxic signaling is active. This marker was not designed to measure local oxygen concentration but rather to show how a cell perceives its local oxygen concentration. ADSCs cultured in both 2D and 3D were exposed to 20% or 1% oxygen environments for 96 h. In 2D at 20% O(2), the marker signal was not observed during the study period. In 1% O(2), the fluorescent signal was first observed at 24 h, with maximum prevalence observed at 96 h as 59%±3% cells expressed the marker. In 3D, the signal was observed in both 1% and 20% O(2). The onset of signal in 1% O(2) was observed at 4 h, reaching maximum prevalence at 96 h with 76%±4% cells expressing the marker. Interestingly, hypoxic signal was also observed in 20% O(2), with 13%±3% cells showing positive marker signal after 96 h. The transcription factor subunit hypoxia inducible factor-1α was tracked in these cells over the same time period by immunostaining and western blot analysis. Immunostaining results in 2D correlated well with our marker at 72 h and 96 h, but 3D results did not correlate well. The western blotting results in 2D and 3D correlated well with the fluorescent marker. The HRE DsRed-DR virus can be used to track the onset of this response for encapsulated, mesenchymal stem cells. Due to the importance of hypoxic signaling in determination of stem cell differentiation, this marker could be a useful tool for the tissue engineering community.


Subject(s)
Adipose Tissue/cytology , Biomarkers/metabolism , Cell Differentiation , Cell Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mesenchymal Stem Cells/cytology , Signal Transduction , Adipose Tissue/metabolism , Biomarkers/analysis , Cell Communication , Cell Culture Techniques , Cell Proliferation , Cells, Cultured , Humans , Hydrogels/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunoblotting , Luminescent Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Oxygen/metabolism , Polyethylene Glycols/metabolism , Response Elements/genetics , Red Fluorescent Protein
7.
J Biomed Mater Res B Appl Biomater ; 97(1): 148-55, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21290593

ABSTRACT

A common obstacle to the survival of encapsulated tissue is oxygen insufficiency. This appears particularly true of encapsulated pancreatic ß-cells. Our work investigates a fluorescent hypoxia detection system for early recognition of hypoxic stress in encapsulated pancreatic tissue. Murine insulinoma (MIN6) cells were engineered to produce a red fluorescent protein under the control of hypoxia-inducible-factor-1. Aggregates of these cells were encapsulated in poly(ethylene glycol) hydrogels at densities of 200,000, 600,000, and 1 million cells per capsule then incubated in either a 1% or 20% oxygen environment. Cell function was evaluated by daily measurement of glucose-stimulated insulin secretion. Encapsulated cells were also fluorescently imaged periodically over 72 h for expression of the marker signal. Results indicate that oxygen insufficiency severely impacts insulin release from MIN6 cells, and that large aggregates are especially vulnerable to oxygen limitations. Our marker was found to be successfully indicative of hypoxia and could be used as a predictor of subsequent insulin release. Further work will be required to fully characterize signal dynamics and to evaluate in vivo efficacy. The method presented here represents a unique and valuable approach to detecting hypoxic stress in living tissues which may prove useful to a variety of fields of biological research.


Subject(s)
Biological Assay/methods , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Luminescent Proteins/biosynthesis , Oxygen Consumption , Animals , Biomarkers/metabolism , Cell Hypoxia/physiology , Cell Line, Tumor , Cells, Immobilized/cytology , Cells, Immobilized/metabolism , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Luminescent Proteins/genetics , Mice , Red Fluorescent Protein
8.
J Vis Exp ; (58)2011 Dec 16.
Article in English | MEDLINE | ID: mdl-22215075

ABSTRACT

In Diabetes mellitus type 1, autoimmune destruction of the pancreatic ß-cells results in loss of insulin production and potentially lethal hyperglycemia. As an alternative treatment option to exogenous insulin injection, transplantation of functional pancreatic tissue has been explored. This approach offers the promise of a more natural, long-term restoration of normoglycemia. Protection of the donor tissue from the host's immune system is required to prevent rejection and encapsulation is a method used to help achieve this aim. Biologically-derived materials, such as alginate and agarose, have been the traditional choice for capsule construction but may induce inflammation or fibrotic overgrowth which can impede nutrient and oxygen transport. Alternatively, synthetic poly(ethylene glycol) (PEG)-based hydrogels are non-degrading, easily functionalized, available at high purity, have controllable pore size, and are extremely biocompatible. As an additional benefit, PEG hydrogels may be formed rapidly in a simple photo-crosslinking reaction that does not require application of non-physiological temperatures. Such a procedure is described here. In the crosslinking reaction, UV degradation of the photoinitiator, 1-[4-(2-Hydroxyethoxy)-phenyl]-2-hydroxy-2-methyl-1-propane-1-one (Irgacure 2959), produces free radicals which attack the vinyl carbon-carbon double bonds of dimethacrylated PEG (PEGDM) inducing crosslinking at the chain ends. Crosslinking can be achieved within 10 minutes. PEG hydrogels constructed in such a manner have been shown to favorably support cells, and the low photoinitiator concentration and brief exposure to UV irradiation is not detrimental to viability and function of the encapsulated tissue. While we methacrylate our PEG with the method described below, PEGDM can also be directly purchased from vendors such as Sigma. An inherent consequence of encapsulation is isolation of the cells from a vascular network. Supply of nutrients, notably oxygen, is therefore reduced and limited by diffusion. This reduced oxygen availability may especially impact ß-cells whose insulin secretory function is highly dependent on oxygen. Capsule composition and geometry will also impact diffusion rates and lengths for oxygen. Therefore, we also describe a technique for identifying hypoxic cells within our PEG capsules. Infection of the cells with a recombinant adenovirus allows for a fluorescent signal to be produced when intracellular hypoxia-inducible factor (HIF) pathways are activated. As HIFs are the primary regulators of the transcriptional response to hypoxia, they represent an ideal target marker for detection of hypoxic signaling. This approach allows for easy and rapid detection of hypoxic cells. Briefly, the adenovirus has the sequence for a red fluorescent protein (Ds Red DR from Clontech) under the control of a hypoxia-responsive element (HRE) trimer. Stabilization of HIF-1 by low oxygen conditions will drive transcription of the fluorescent protein (Figure 1). Additional details on the construction of this virus have been published previously. The virus is stored in 10% glycerol at -80° C as many 150 µL aliquots in 1.5 mL centrifuge tubes at a concentration of 3.4 x 10(10) pfu/mL. Previous studies in our lab have shown that MIN6 cells encapsulated as aggregates maintain their viability throughout 4 weeks of culture in 20% oxygen. MIN6 aggregates cultured at 2 or 1% oxygen showed both signs of necrotic cells (still about 85-90% viable) by staining with ethidium bromide as well as morphological changes relative to cells in 20% oxygen. The smooth spherical shape of the aggregates displayed at 20% was lost and aggregates appeared more like disorganized groups of cells. While the low oxygen stress does not cause a pronounced drop in viability, it is clearly impacting MIN6 aggregation and function as measured by glucose-stimulated insulin secretion. Western blot analysis of encapsulated cells in 20% and 1% oxygen also showed a significant increase in HIF-1α for cells cultured in the low oxygen conditions which correlates with the expression of the DsRed DR protein.


Subject(s)
Cell Aggregation/physiology , Cell Hypoxia/physiology , Insulin-Secreting Cells/cytology , Animals , Biocompatible Materials/chemistry , Cell Line , Hydrogels/chemistry , Insulin-Secreting Cells/chemistry , Insulin-Secreting Cells/transplantation , Mice , Polyethylene Glycols/chemistry , Propane/analogs & derivatives , Propane/chemistry , Signal Transduction
9.
Biomed Mater ; 5(6): 061001, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21060146

ABSTRACT

We have studied the impact of overexpression of an intracellular signaling protein, integrin-linked kinase (ILK), on the survival and function of encapsulated islet tissue used for the treatment of type 1 diabetes. The dimensions of the encapsulated tissue can impact the stresses placed on the tissue and ILK overexpression shows the ability to extend function of dissociated cells as well as intact islets. These results suggest that lost cell-extracellular matrix interactions in cell encapsulation systems can lead to decreased insulin secretion and ILK signaling is a target to overcome this phenomenon.


Subject(s)
Islets of Langerhans Transplantation/methods , Islets of Langerhans/cytology , Islets of Langerhans/physiology , Organ Culture Techniques/methods , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Line , Cell Proliferation , Cell Survival , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/genetics , Recombinant Proteins/metabolism , Transfection/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...