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.
Nat Commun ; 11(1): 114, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31913286

ABSTRACT

Stem cell therapies are limited by poor cell survival and engraftment. A hurdle to the use of materials for cell delivery is the lack of understanding of material properties that govern transplanted stem cell functionality. Here, we show that synthetic hydrogels presenting integrin-specific peptides enhance the survival, persistence, and osteo-reparative functions of human bone marrow-derived mesenchymal stem cells (hMSCs) transplanted in murine bone defects. Integrin-specific hydrogels regulate hMSC adhesion, paracrine signaling, and osteoblastic differentiation in vitro. Hydrogels presenting GFOGER, a peptide targeting α2ß1 integrin, prolong hMSC survival and engraftment in a segmental bone defect and result in improved bone repair compared to other peptides. Integrin-specific hydrogels have diverse pleiotropic effects on hMSC reparative activities, modulating in vitro cytokine secretion and in vivo gene expression for effectors associated with inflammation, vascularization, and bone formation. These results demonstrate that integrin-specific hydrogels improve tissue healing by directing hMSC survival, engraftment, and reparative activities.


Subject(s)
Bone Diseases/therapy , Integrin alpha2beta1/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Animals , Bone Diseases/metabolism , Bone Diseases/physiopathology , Bone Marrow/chemistry , Bone Marrow/metabolism , Bone Regeneration , Cell Adhesion , Cell Survival , Cell- and Tissue-Based Therapy , Humans , Hydrogels/chemistry , Integrin alpha2beta1/genetics , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred NOD , Peptides/metabolism
2.
J Cell Sci ; 132(20)2019 10 21.
Article in English | MEDLINE | ID: mdl-31558679

ABSTRACT

Synthetic hydrogels with controlled physicochemical matrix properties serve as powerful in vitro tools to dissect cell-extracellular matrix (ECM) interactions that regulate epithelial morphogenesis in 3D microenvironments. In addition, these fully defined matrices overcome the lot-to-lot variability of naturally derived materials and have provided insights into the formation of rudimentary epithelial organs. Therefore, we engineered a fully defined synthetic hydrogel with independent control over proteolytic degradation, mechanical properties, and adhesive ligand type and density to study the impact of ECM properties on epithelial tubulogenesis for inner medullary collecting duct (IMCD) cells. Protease sensitivity of the synthetic material for membrane-type matrix metalloproteinase-1 (MT1-MMP, also known as MMP14) was required for tubulogenesis. Additionally, a defined range of matrix elasticity and presentation of RGD adhesive peptide at a threshold level of 2 mM ligand density were required for epithelial tubulogenesis. Finally, we demonstrated that the engineered hydrogel supported organization of epithelial tubules with a lumen and secreted laminin. This synthetic hydrogel serves as a platform that supports epithelial tubular morphogenetic programs and can be tuned to identify ECM biophysical and biochemical properties required for epithelial tubulogenesis.


Subject(s)
Cellular Microenvironment , Epithelial Cells/metabolism , Extracellular Matrix/chemistry , Hydrogels/chemistry , Kidney Tubules, Collecting/metabolism , Kidney Tubules/metabolism , Animals , Cell Line, Transformed , Epithelial Cells/cytology , Kidney Tubules/cytology , Kidney Tubules, Collecting/cytology , Matrix Metalloproteinase 14/metabolism , Mice , Oligopeptides/chemistry
3.
Acta Biomater ; 67: 53-65, 2018 02.
Article in English | MEDLINE | ID: mdl-29246650

ABSTRACT

The use of human mesenchymal stromal cells (hMSC) for treating diseased tissues with poor vascularization has received significant attention, but low cell survival has hampered its translation to the clinic. Bioglasses and glass-ceramics have also been suggested as therapeutic agents for stimulating angiogenesis in soft tissues, but these effects need further evaluation in vivo. In this study, calcium-releasing particles and hMSC were combined within a hydrogel to examine their vasculogenic potential in vitro and in vivo. The particles provided sustained calcium release and showed proangiogenic stimulation in a chorioallantoic membrane (CAM) assay. The number of hMSC encapsulated in a degradable RGD-functionalized PEG hydrogel containing particles remained constant over time and IGF-1 release was increased. When implanted in the epidydimal fat pad of immunocompromised mice, this composite material improved cell survival and stimulated vessel formation and maturation. Thus, the combination of hMSC and calcium-releasing glass-ceramics represents a new strategy to achieve vessel stabilization, a key factor in the revascularization of ischemic tissues. STATEMENT OF SIGNIFICANCE: Increasing blood vessel formation in diseased tissues with poor vascularization is a current clinical challenge. Cell therapy using human mesenchymal stem cells has received considerable interest, but low cell survival has hampered its translation to the clinic. Bioglasses and glass-ceramics have been explored as therapeutic agents for stimulating angiogenesis in soft tissues, but these effects need further evaluation in vivo. By incorporating both human mesenchymal stem cells and glass-ceramic particles in an implantable hydrogel, this study provides insights into the vasculogenic potential in soft tissues of the combined strategies. Enhancement of vessel formation and maturation supports further investigation of this strategy.


Subject(s)
Blood Vessels/growth & development , Calcium/metabolism , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Mesenchymal Stem Cells/metabolism , Polyethylene Glycols/chemistry , Adipose Tissue/drug effects , Adipose Tissue/physiology , Angiogenesis Inducing Agents/pharmacology , Animals , Blood Vessels/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Immobilized/drug effects , Cells, Immobilized/metabolism , Chickens , Chorioallantoic Membrane/drug effects , Chorioallantoic Membrane/metabolism , Epididymis/drug effects , Epididymis/physiology , Humans , Implants, Experimental , Male , Maleimides/chemistry , Mesenchymal Stem Cells/drug effects , Mice , Models, Biological , Neovascularization, Physiologic/drug effects , Particle Size
5.
J Biomed Mater Res A ; 104(4): 889-900, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26662727

ABSTRACT

Vascularization of bone defects is considered a crucial component to the successful regeneration of large bone defects. Although vascular endothelial growth factor (VEGF) has been delivered to critical-size bone defect models to augment blood vessel infiltration into the defect area, its potential to increase bone repair remains ambiguous. In this study, we investigated whether integrin-specific biomaterials modulate the effects of VEGF on bone regeneration. We engineered protease-degradable, VEGF-loaded poly(ethylene glycol) (PEG) hydrogels functionalized with either a triple-helical, α2 ß1 integrin-specific peptide GGYGGGP(GPP)5 GFOGER(GPP)5 GPC (GFOGER) or an αv ß3 integrin-targeting peptide GRGDSPC (RGD). Covalent incorporation of VEGF into the PEG hydrogel allowed for protease degradation-dependent release of the protein while maintaining VEGF bioactivity. When applied to critical-size segmental defects in the murine radius, GFOGER-functionalized VEGF-free hydrogels exhibited significantly increased vascular volume and density and resulted in a larger number of thicker blood vessels compared to RGD-functionalized VEGF-free hydrogels. VEGF-loaded RGD hydrogels increased vascularization compared to VEGF-free RGD hydrogels, but the levels of vascularization for these VEGF-containing RGD hydrogels were similar to those of VEGF-free GFOGER hydrogels. VEGF transiently increased bone regeneration in RGD hydrogels but had no effect at later time points. In GFOGER hydrogels, VEGF did not show an effect on bone regeneration. However, VEGF-free GFOGER hydrogels resulted in increased bone regeneration compared to VEGF-free RGD hydrogels. These findings demonstrate the importance of integrin-specificity in engineering constructs for vascularization and associated bone regeneration.


Subject(s)
Biocompatible Materials/metabolism , Hydrogels/metabolism , Integrin alpha2beta1/metabolism , Integrin alphaVbeta3/metabolism , Peptides/metabolism , Polyethylene Glycols/metabolism , Vascular Endothelial Growth Factor A/administration & dosage , Animals , Biocompatible Materials/chemistry , Bone Regeneration/drug effects , Drug Delivery Systems/methods , Human Umbilical Vein Endothelial Cells , Humans , Hydrogels/chemistry , Male , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Peptides/chemistry , Polyethylene Glycols/chemistry , Radius/injuries , Radius/physiology , Rheology , Vascular Endothelial Growth Factor A/pharmacology
6.
J Mater Sci Mater Med ; 27(2): 38, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26704555

ABSTRACT

The development of elastomeric, bioresorbable and biocompatible segmented polyurethanes (SPUs) for use in tissue-engineering applications has attracted considerable interest because of the existing need of mechanically tunable scaffolds for regeneration of different tissues, but the incorporation of osteoinductive molecules into SPUs has been limited. In this study, SPUs were synthesized from poly (ε-caprolactone)diol, 4,4'-methylene bis(cyclohexyl isocyanate) using biologically active compounds such as ascorbic acid, L-glutamine, ß-glycerol phosphate, and dexamethasone as chain extenders. Fourier transform infrared spectroscopy (FTIR) revealed the formation of both urethanes and urea linkages while differential scanning calorimetry, dynamic mechanical analysis, X-ray diffraction and mechanical testing showed that these polyurethanes were semi-crystalline polymers exhibiting high deformations. Cytocompatibility studies showed that only SPUs containing ß-glycerol phosphate supported human mesenchymal stem cell adhesion, growth, and osteogenic differentiation, rendering them potentially suitable for bone tissue regeneration, whereas other SPUs failed to support either cell growth or osteogenic differentiation, or both. This study demonstrates that modification of SPUs with osteogenic compounds can lead to new cytocompatible polymers for regenerative medicine applications.


Subject(s)
Biocompatible Materials/chemistry , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Polyurethanes/chemistry , Tissue Scaffolds/chemistry , Bone and Bones/cytology , Bone and Bones/drug effects , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Humans , Materials Testing , Mechanical Phenomena , Mesenchymal Stem Cells/physiology , Osteogenesis/drug effects , Tissue Engineering/instrumentation , Tissue Engineering/methods
7.
ACS Biomater Sci Eng ; 2(4): 606-615, 2016 Apr 11.
Article in English | MEDLINE | ID: mdl-33465862

ABSTRACT

Plant viruses have been highlighted among material research due to their well-defined structures in nanoscale, monodispersity, stability, and chemical functionalities. Each of the thousands coat protein subunits on a viral nanoparticle can be homogeneously modified, chemically and genetically, with a functional ligand leading to a high-density and spatial distribution of ligands on each particle (multivalency). Previous reports from our group have evidenced that substrates coated with Tobacco mosaic virus (TMV) and its mutant promote early osteogenesis of mesenchymal stem cells (MSCs). We then fabricated a three-dimensional (3D) biopolymeric scaffold with rod-like TMV in the form of a sponge-like hydrogel for tissue engineering purposes. The hydrogel was functionalized with the cellular recognition peptide, arginine-glycine-aspartic acid (RGD), through an incorporation of an RGD mutant of TMV (TMV-RGD). The virus-functionalized hydrogel materials were shown to aid bone differentiation of MSCs in vitro. Herein, we performed an in vivo study based on the TMV and TMV-RGD hydrogels in Sprague-Dawley rats with cranial bone defects. This report substantiated the hypothesis that TMV-functionalized hydrogel scaffolds did not cause systemic toxicity when implanted in the defect site and that the TMV-based hydrogel platform can support cell localization and can be further optimized for bone regeneration and repair.

8.
Biomed Microdevices ; 16(5): 727-36, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24907052

ABSTRACT

While many advanced liver models support hepatic phenotypes necessary for drug and disease studies, these models are characterized by intricate features such as co-culture with one of more supporting cell types or advanced media perfusion systems. These systems have helped elucidate some of the critical biophysical features missing from standard well-plate based hepatocyte culture, but their advanced designs add to their complexity. Additionally, regardless of the culture system, primary hepatocyte culture systems suffer from reproducibility issues due to phenotypic variation and expensive, limited supplies of donor lots. Here we describe a microfluidic bilayer device that sustains primary human hepatocyte phenotypes, including albumin production, factor IX production, cytochrome P450 3A4 drug metabolism and bile canaliculi formation for at least 14 days in a simple monoculture format with static media. Using a variety of channel architectures, we describe how primary cell phenotype is promoted by spatial confinement within the microfluidic channel, without the need for perfusion or co-culture. By sourcing human hepatocytes expanded in the Fah, Rag2, and Il2rg-knockout (FRG™-KO) humanized mouse model, utilizing a few hundred hepatocytes within each channel, and maintaining hepatocyte function for weeks in vitro within a relatively simple model, we demonstrate a basic primary human hepatocyte culture system that addresses many of the major hurdles in human hepatocyte culture research.


Subject(s)
Cell Culture Techniques , Cell Proliferation , Hepatocytes/metabolism , Liver , Microfluidic Analytical Techniques , Animals , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Hep G2 Cells , Hepatocytes/cytology , Humans , Mice , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods
9.
Biomaterials ; 35(21): 5453-61, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24726536

ABSTRACT

Non-healing bone defects present tremendous socioeconomic costs. Although successful in some clinical settings, bone morphogenetic protein (BMP) therapies require supraphysiological dose delivery for bone repair, raising treatment costs and risks of complications. We engineered a protease-degradable poly(ethylene glycol) (PEG) synthetic hydrogel functionalized with a triple helical, α2ß1 integrin-specific peptide (GFOGER) as a BMP-2 delivery vehicle. GFOGER-functionalized hydrogels lacking BMP-2 directed human stem cell differentiation and produced significant enhancements in bone repair within a critical-sized bone defect compared to RGD hydrogels or empty defects. GFOGER functionalization was crucial to the BMP-2-dependent healing response. Importantly, these engineered hydrogels outperformed the current clinical carrier in repairing non-healing bone defects at low BMP-2 doses. GFOGER hydrogels provided sustained in vivo release of encapsulated BMP-2, increased osteoprogenitor localization in the defect site, enhanced bone formation and induced defect bridging and mechanically robust healing at low BMP-2 doses which stimulated almost no bone regeneration when delivered from collagen sponges. These findings demonstrate that GFOGER hydrogels promote bone regeneration in challenging defects with low delivered BMP-2 doses and represent an effective delivery vehicle for protein therapeutics with translational potential.


Subject(s)
Bone Morphogenetic Protein 2/genetics , Bone Regeneration/drug effects , Gene Transfer Techniques , Hydrogels/pharmacology , Integrin alpha2beta1/chemistry , Animals , Bone Morphogenetic Protein 2/metabolism , Bone Regeneration/physiology , Bone and Bones/cytology , Bone and Bones/drug effects , Cell Differentiation/drug effects , Collagen/chemistry , Collagen/pharmacology , Humans , Hydrogels/chemistry , Male , Mesenchymal Stem Cells , Mice , Osteogenesis/drug effects , Peptides/chemistry , Peptides/pharmacology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tissue Engineering , Wound Healing/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL