Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Tissue Eng Part B Rev ; 26(2): 105-115, 2020 04.
Article in English | MEDLINE | ID: mdl-31774026

ABSTRACT

Damage to osteochondral (OC) tissues can lead to pain, loss of motility, and progress to osteoarthritis. Tissue engineering approaches offer the possibility of replacing damaged tissues and restoring joint function; however, replicating the spatial and functional heterogeneity of native OC tissue remains a pressing challenge. Chondrocytes in healthy cartilage exist in relatively low-oxygen conditions, while osteoblasts in the underlying bone experience higher oxygen pressures. Such oxygen gradients also exist in the limb bud, where they influence OC tissue development. The cellular response to these spatial variations in oxygen pressure, which is mediated by the hypoxia inducible factor (HIF) pathway, plays a central role in regulating osteo- and chondrogenesis by directing progenitor cell differentiation and promoting and maintaining appropriate extracellular matrix production. Understanding the role of the HIF pathway in OC tissue development may enable new approaches to engineer OC tissue. In this review, we discuss strategies to spatially and temporarily regulate the HIF pathway in progenitor cells to create functional OC tissue for regenerative therapies. Impact statement Strategies to engineer osteochondral (OC) tissue are limited by the complex and varying microenvironmental conditions in native bone and cartilage. Indeed, native cartilage experiences low-oxygen conditions, while the underlying bone is relatively normoxic. The cellular response to these low-oxygen conditions, which is mediated through the hypoxia inducible factor (HIF) pathway, is known to promote and maintain the chondrocyte phenotype. By using tissue engineering scaffolds to spatially and temporally harness the HIF pathway, it may be possible to improve OC tissue engineering strategies for the regeneration of damaged cartilage and its underlying subchondral bone.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Chondrocytes/cytology , Chondrogenesis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Osteoarthritis/therapy , Osteoblasts/cytology , Osteogenesis , Tissue Engineering/methods , Animals , Chondrocytes/metabolism , Humans , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoblasts/metabolism , Tissue Scaffolds
2.
Acta Biomater ; 89: 73-83, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30844569

ABSTRACT

Tissue engineering strategies often aim to direct tissue formation by mimicking conditions progenitor cells experience within native tissues. For example, to create cartilage in vitro, researchers often aim to replicate the biochemical and mechanical milieu cells experience during cartilage formation in the developing limb bud. This includes stimulating progenitors with TGF-ß1/3, culturing under hypoxic conditions, and regulating mechanosensory pathways using biomaterials that control substrate stiffness and/or cell shape. However, as progenitors differentiate down the chondrogenic lineage, the pathways that regulate their responses to mechanotransduction, hypoxia and TGF-ß may not act independently, but rather also impact one another, influencing overall cell response. Here, to better understand hypoxia's influence on mechanoregulatory-mediated chondrogenesis, we cultured human marrow stromal/mesenchymal stem cells (hMSC) on soft (0.167 kPa) or stiff (49.6 kPa) polyacrylamide hydrogels in chondrogenic medium containing TGF-ß3. We then compared cell morphology, phosphorylated myosin light chain 2 staining, and chondrogenic gene expression under normoxic and hypoxic conditions, in the presence and absence of pharmacological inhibition of cytoskeletal tension. We show that on soft compared to stiff substrates, hypoxia prompts hMSC to adopt more spread morphologies, assemble in compact mesenchymal condensation-like colonies, and upregulate NCAM expression, and that inhibition of cytoskeletal tension negates hypoxia-mediated upregulation of molecular markers of chondrogenesis, including COL2A1 and SOX9. Taken together, our findings support a role for hypoxia in regulating hMSC morphology, cytoskeletal tension and chondrogenesis, and that hypoxia's effects are modulated, at least in part, by mechanosensitive pathways. Our insights into how hypoxia impacts mechanoregulation of chondrogenesis in hMSC may improve strategies to develop tissue engineered cartilage. STATEMENT OF SIGNIFICANCE: Cartilage tissue engineering strategies often aim to drive progenitor cell differentiation by replicating the local environment of the native tissue, including by regulating oxygen concentration and mechanical stiffness. However, the pathways that regulate cellular responses to mechanotransduction and hypoxia may not act independently, but rather also impact one another. Here, we show that on soft, but not stiff surfaces, hypoxia impacts human MSC (hMSC) morphology and colony formation, and inhibition of cytoskeletal tension negates the hypoxia-mediated upregulation of molecular markers of chondrogenesis. These observations suggest that hypoxia's effects during hMSC chondrogenesis are modulated, at least in part, by mechanosensitive pathways, and may impact strategies to develop scaffolds for cartilage tissue engineering, as hypoxia's chondrogenic effects may be enhanced on soft materials.


Subject(s)
Acrylic Resins , Cell Differentiation , Chondrogenesis , Hydrogels , Mechanotransduction, Cellular , Mesenchymal Stem Cells/metabolism , Acrylic Resins/chemistry , Acrylic Resins/pharmacology , Cell Hypoxia , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Mesenchymal Stem Cells/cytology , Stress, Mechanical , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta3/metabolism
3.
Nat Commun ; 9(1): 5419, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30560926

ABSTRACT

The original version of this Article contained an error in the author affiliations. The affiliation of Marjan Enayati with 'Ludwig Boltzmann Cluster for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Austria' was inadvertently omitted. This has now been corrected in both the PDF and HTML versions of the Article.

4.
Nat Commun ; 9(1): 4851, 2018 11 14.
Article in English | MEDLINE | ID: mdl-30429483

ABSTRACT

In the original version of this Article the dataset identifier in the Data Availability statement was incorrect. The correct dataset identifier is PXD009500. This has been corrected in the HTML and PDF versions of this Article.

5.
Nat Commun ; 9(1): 4049, 2018 10 03.
Article in English | MEDLINE | ID: mdl-30282987

ABSTRACT

Modifiable hydrogels have revealed tremendous insight into how physical characteristics of cells' 3D environment drive stem cell lineage specification. However, in native tissues, cells do not passively receive signals from their niche. Instead they actively probe and modify their pericellular space to suit their needs, yet the dynamics of cells' reciprocal interactions with their pericellular environment when encapsulated within hydrogels remains relatively unexplored. Here, we show that human bone marrow stromal cells (hMSC) encapsulated within hyaluronic acid-based hydrogels modify their surroundings by synthesizing, secreting and arranging proteins pericellularly or by degrading the hydrogel. hMSC's interactions with this local environment have a role in regulating hMSC fate, with a secreted proteinaceous pericellular matrix associated with adipogenesis, and degradation with osteogenesis. Our observations suggest that hMSC participate in a bi-directional interplay between the properties of their 3D milieu and their own secreted pericellular matrix, and that this combination of interactions drives fate.


Subject(s)
Cell Communication , Cell Lineage , Cell-Matrix Junctions/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Amides/pharmacology , Cell Communication/drug effects , Cell Lineage/drug effects , Cell-Matrix Junctions/drug effects , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Paclitaxel/pharmacology , Pyridines/pharmacology , Stem Cells/drug effects
6.
Stem Cells ; 36(9): 1380-1392, 2018 09.
Article in English | MEDLINE | ID: mdl-29726060

ABSTRACT

The transcriptional profile induced by hypoxia plays important roles in the chondrogenic differentiation of marrow stromal/stem cells (MSC) and is mediated by the hypoxia inducible factor (HIF) complex. However, various compounds can also stabilize HIF's oxygen-responsive element, HIF-1α, at normoxia and mimic many hypoxia-induced cellular responses. Such compounds may prove efficacious in cartilage tissue engineering, where microenvironmental cues may mediate functional tissue formation. Here, we investigated three HIF-stabilizing compounds, which each have distinct mechanisms of action, to understand how they differentially influenced the chondrogenesis of human bone marrow-derived MSC (hBM-MSC) in vitro. hBM-MSCs were chondrogenically-induced in transforming growth factor-ß3-containing media in the presence of HIF-stabilizing compounds. HIF-1α stabilization was assessed by HIF-1α immunofluorescence staining, expression of HIF target and articular chondrocyte specific genes by quantitative polymerase chain reaction, and cartilage-like extracellular matrix production by immunofluorescence and histochemical staining. We demonstrate that all three compounds induced similar levels of HIF-1α nuclear localization. However, while the 2-oxoglutarate analog dimethyloxalylglycine (DMOG) promoted upregulation of a selection of HIF target genes, desferrioxamine (DFX) and cobalt chloride (CoCl2 ), compounds that chelate or compete with divalent iron (Fe2+ ), respectively, did not. Moreover, DMOG induced a more chondrogenic transcriptional profile, which was abolished by Acriflavine, an inhibitor of HIF-1α-HIF-ß binding, while the chondrogenic effects of DFX and CoCl2 were more limited. Together, these data suggest that HIF-1α function during hBM-MSC chondrogenesis may be regulated by mechanisms with a greater dependence on 2-oxoglutarate than Fe2+ availability. These results may have important implications for understanding cartilage disease and developing targeted therapies for cartilage repair. Stem Cells 2018;36:1380-1392.


Subject(s)
Bone Marrow Cells/drug effects , Cell Hypoxia/drug effects , Enzyme Inhibitors/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Mesenchymal Stem Cells/drug effects , Amino Acids, Dicarboxylic/pharmacology , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cell Hypoxia/physiology , Child , Chondrogenesis/drug effects , Cobalt/pharmacology , Deferoxamine/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism
7.
Regen Med ; 9(6): 723-32, 2014.
Article in English | MEDLINE | ID: mdl-25431909

ABSTRACT

AIM: Umbilical cord contains, within Wharton's jelly (WJ), multipotent mesenchymal stromal/stem cells (MSCs) of fetal origin that can be isolated and expanded in vitro with a minimal manipulation and very high efficiency. Our aim was to develop a highly reproducible protocol that has the unique potential to be scaled up and adapted to cGMP requirements for the use in cellular therapy. RESULTS: We found that derivation of WJ MSCs under defined conditions in low oxygen resulted in several folds higher populations of MSCA-1(+) cells (6.0-19.2%) when compared with WJ MSCs derived in the presence of serum (0.1-2.8%) or clinical-grade bone marrow (BM) MSCs cultured under atmospheric O2 (20%). We demonstrate that WJ MSCs derived following our protocol display antiproliferative activity similar to clinical-grade BM MSCs. We also show that these WJ MSCs can be differentiated into adipo-, chondro- and osteo-genic lineages. CONCLUSION: Easy accessibility, abundance and genetic 'naivety' make WJ MSCs logistically a more attractive source for clinical applications than BM MSCs.


Subject(s)
Cell Culture Techniques/methods , Mesenchymal Stem Cells/cytology , Oxygen/metabolism , Umbilical Cord/cytology , Wharton Jelly/cytology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Female , Flow Cytometry , Humans , Serum/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...