Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Wound Repair Regen ; 24(4): 630-43, 2016 07.
Article in English | MEDLINE | ID: mdl-27102877

ABSTRACT

Current chronic wound treatments often fail to promote healing of diabetic foot ulcers (DFU), leading to amputation and increased patient morbidity. A critical mediator of proper wound healing is the production, assembly, and remodeling of the extracellular matrix (ECM) by fibroblasts. However, little is known about how these processes are altered in fibroblasts within the DFU microenvironment. Thus, we investigated the capacity of multiple, primary DFU-derived fibroblast strains to express, produce, and assemble ECM proteins compared to diabetic patient-derived fibroblasts and healthy donor-derived fibroblasts. Gene expression microarray analysis showed differential expression of ECM and ECM-regulatory genes by DFU-derived fibroblasts which translated to functional differences in a 3D in vitro ECM tissue model. DFU-derived fibroblasts produced thin, fibronectin-rich matrices, and responded abnormally when challenged with transforming growth factor-beta, a key regulator of matrix production during healing. These results provide novel evidence that DFU-derived fibroblasts contribute to the defective matrices of DFUs and chronic wound pathogenesis.


Subject(s)
Diabetic Foot/pathology , Diabetic Foot/physiopathology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibroblasts/metabolism , Fibronectins/metabolism , Wound Healing , Collagen Type I/metabolism , Diabetic Foot/metabolism , Extracellular Matrix/chemistry , Extracellular Matrix/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Expression Profiling , Humans , Immunohistochemistry , Microarray Analysis , Neovascularization, Physiologic , Transforming Growth Factor beta/pharmacology
2.
J Cell Sci ; 125(Pt 9): 2276-87, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22344267

ABSTRACT

Platelet-derived growth factor receptor-beta (PDGFRß) is required for the development of mesenchymal cell types, and plays a diverse role in the function of fibroblasts in tissue homeostasis and regeneration. In this study, we characterized the expression of PDGFRß in fibroblasts derived from human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), and showed that this expression is important for cellular functions such as migration, extracellular matrix production and assembly in 3D self-assembled tissues. To determine potential regulatory regions predictive of expression of PDGFRß following differentiation from ESCs and iPSCs, we analyzed the DNA methylation status of a region of the PDGFRB promoter that contains multiple CpG sites, before and after differentiation. We demonstrated that this promoter region is extensively demethylated following differentiation, and represents a developmentally regulated, differentially methylated region linked to PDGFRß expression. Understanding the epigenetic regulation of genes such as PDGFRB, and identifying sites of active DNA demethylation, is essential for future applications of iPSC-derived fibroblasts for regenerative medicine.


Subject(s)
Cell Differentiation/genetics , DNA Methylation/genetics , Fibroblasts/metabolism , Receptor, Platelet-Derived Growth Factor beta/genetics , Cell Movement/genetics , CpG Islands/genetics , DNA/analysis , DNA/chemistry , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Extracellular Matrix/metabolism , Fibroblasts/cytology , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic
3.
In Vitro Cell Dev Biol Anim ; 48(2): 112-22, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22259014

ABSTRACT

Reprogramming of somatic cells to induced pluripotent stem cells (iPSC) provides an important cell source to derive patient-specific cells for potential therapeutic applications. However, it is not yet clear whether reprogramming through pluripotency allows the production of differentiated cells with improved functional properties that may be beneficial in regenerative therapies. To address this, we compared the production and assembly of extracellular matrix (ECM) by iPSC-derived fibroblasts to that of the parental, dermal fibroblasts (BJ), from which these iPSC were initially reprogrammed, and to fibroblasts differentiated from human embryonic stem cells (hESC). iPSC- and hESC-derived fibroblasts demonstrated stable expression of surface markers characteristic of stromal fibroblasts during prolonged culture and showed an elevated growth potential when compared to the parental BJ fibroblasts. We found that in the presence of L: -ascorbic acid-2-phosphate, iPSC- and hESC-derived fibroblasts increased their expression of collagen genes, secretion of soluble collagen, and extracellular deposition of type I collagen to a significantly greater degree than that seen in the parental BJ fibroblasts. Under culture conditions that enabled the self-assembly of a 3D stromal tissue, iPSC- and hESC-derived fibroblasts generated a well organized, ECM that was enriched in type III collagen. By characterizing the functional properties of iPSC-derived fibroblasts compared to their parental fibroblasts, we demonstrate that these cells represent a promising, alternative source of fibroblasts to advance future regenerative therapies.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/metabolism , Extracellular Matrix Proteins/metabolism , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/metabolism , Ascorbic Acid/analogs & derivatives , Ascorbic Acid/pharmacology , Biomarkers/metabolism , Cell Culture Techniques , Cells, Cultured , Collagen/biosynthesis , Embryonic Stem Cells/cytology , Extracellular Matrix/metabolism , Fibroblasts/cytology , Humans , Induced Pluripotent Stem Cells/cytology
4.
PLoS One ; 5(5): e10528, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20502640

ABSTRACT

The alpha6beta4 integrin plays a significant role in tumor growth, angiogenesis and metastasis through modulation of growth factor signaling, and is a potentially important therapeutic target. However, alpha6beta4-mediated cell-matrix adhesion is critical in normal keratinocyte attachment, signaling and anchorage to the basement membrane through its interaction with laminin-5, raising potential risks for targeted therapy. Bioengineered Human Skin Equivalent (HSE), which have been shown to mimic their normal and wounded counterparts, have been used here to investigate the consequences of targeting beta4 to establish toxic effects on normal tissue homeostasis and epithelial wound repair. We tested two antibodies directed to different beta4 epitopes, one adhesion-blocking (ASC-8) and one non-adhesion blocking (ASC-3), and determined that these antibodies were appropriately localized to the basal surface of keratinocytes at the basement membrane interface where beta4 is expressed. While normal tissue architecture was not altered, ASC-8 induced a sub-basal split at the basement membrane in non-wounded tissue. In addition, wound closure was significantly inhibited by ASC-8, but not by ASC-3, as the epithelial tongue only covered 40 percent of the wound area at 120 hours post-wounding. These results demonstrate beta4 adhesion-blocking antibodies may have adverse effects on normal tissue, whereas antibodies directed to other epitopes may provide safer alternatives for therapy. Taken together, we conclude that these three-dimensional tissue models provide a biologically relevant platform to identify toxic effects induced by candidate therapeutics, which will allow generation of findings that are more predictive of in vivo responses early in the drug development process.


Subject(s)
Antibodies, Blocking/pharmacology , Epithelium/metabolism , Integrins/immunology , Keratinocytes/drug effects , Keratinocytes/metabolism , Models, Biological , Wound Healing/drug effects , Antibody Specificity/drug effects , Biological Assay , Cell Adhesion Molecules/immunology , Cell Movement/drug effects , Epithelium/drug effects , Fluorescent Antibody Technique , Humans , Integrin beta4/immunology , Male , Protein Binding/drug effects , Skin, Artificial , Staining and Labeling , Kalinin
SELECTION OF CITATIONS
SEARCH DETAIL
...