Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Mater Today Bio ; 22: 100791, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37731960

ABSTRACT

Osteoarthritis (OA) is a degenerative disease of the joints for which no curative treatment exists. Intra-articular injection of stem cells is explored as a regenerative approach, but rapid clearance of cells from the injection site limits the therapeutic outcome. Microencapsulation of mesenchymal stem cells (MSCs) can extend the retention time of MSCs, but the outcomes of the few studies currently performed are conflicting. We hypothesize that the composition of the micromaterial's shell plays a deciding factor in the treatment outcome of intra-articular MSC injection. To this end, we microencapsulate MSCs using droplet microfluidic generators in flow-focus mode using various polymers and polymer concentrations. We demonstrate that polymer composition and concentration potently alter the metabolic activity as well as the secretome of MSCs. Moreover, while microencapsulation consistently prolongs the retention time of MSC injected in rat joints, distinct biodistribution within the joint is demonstrated for the various microgel formulations. Furthermore, intra-articular injections of pristine and microencapsulated MSC in OA rat joints show a strong material-dependent effect on the reduction of cartilage degradation and matrix loss. Collectively, this study highlights that micromaterial composition and concentration are key deciding factors for the therapeutic outcome of intra-articular injections of microencapsulated stem cells to treat degenerative joint diseases.

2.
Polymers (Basel) ; 14(20)2022 Oct 12.
Article in English | MEDLINE | ID: mdl-36297870

ABSTRACT

Previously, 5% w/v hyaluronic acid-tyramine (HA-TA) and dextran-tyramine (Dex-TA) enzymatically cross-linked hybrid hydrogels were demonstrated to provide a mechanically stable environment, maintain cell viability, and promote cartilaginous-specific matrix deposition in vitro. In this study, 5% w/v hybrid hydrogels were combined with human mesenchymal stem cells (hMSCs), bovine chondrocytes (bCHs), or a combination of both in a 4:1 ratio and subcutaneously implanted in the backs of male and female nude rats to assess the performance of cell-laden hydrogels in tissue formation. Subcutaneous implantation of these biomaterials showed signs of integration of the gels within the host tissue. Histological analysis showed residual fibrotic capsules four weeks after implantation. However, enhanced tissue invasion and some giant cell infiltration were observed in the HA-TA/Dex-TA hydrogels laden with either hMSCs or bCHs but not with the co-culture. Moreover, hMSC-bCH co-cultures showed beneficial interaction with the hydrogels, for instance, in enhanced cell proliferation and matrix deposition. In addition, we provide evidence that host gender has an impact on the performance of bCHs encapsulated in HA-TA/Dex-TA hydrogels. This study revealed that hydrogels laden with different types of cells result in distinct host responses. It can be concluded that 5% w/v hydrogels with a higher concentration of Dex-TA (≥50%) laden with bCH-hMSC co-cultures are adequate for injectable applications and in situ cell delivery in cartilage regeneration approaches.

3.
Polymers (Basel) ; 13(9)2021 May 10.
Article in English | MEDLINE | ID: mdl-34068542

ABSTRACT

The ideal scaffold for cartilage regeneration is expected to provide adequate mechanical strength, controlled degradability, adhesion, and integration with the surrounding native tissue. As it does this, it mimics natural ECMs functions, which allow for nutrient diffusion and promote cell survival and differentiation. Injectable hydrogels based on tyramine (TA)-functionalized hyaluronic acid (HA) and dextran (Dex) are a promising approach for cartilage regeneration. The properties of the hydrogels used in this study were adjusted by varying polymer concentrations and ratios. To investigate the changes in properties and their effects on cellular behavior and cartilage matrix formation, different ratios of HA- and dextran-based hybrid hydrogels at both 5 and 10% w/v were prepared using a designed mold to control generation. The results indicated that the incorporation of chondrocytes in the hydrogels decreased their mechanical properties. However, rheological and compression analysis indicated that 5% w/v hydrogels laden with cells exhibit a significant increase in mechanical properties after 21 days when the constructs are cultured in a chondrogenic differentiation medium. Moreover, compared to the 10% w/v hydrogels, the 5% w/v hybrid hydrogels increased the deposition of the cartilage matrix, especially in constructs with a higher Dex-TA content. These results indicated that 5% w/v hybrid hydrogels with 25% HA-TA and 75% Dex-TA have a high potential as injectable scaffolds for cartilage tissue regeneration.

4.
J Mater Sci Mater Med ; 29(11): 174, 2018 Nov 09.
Article in English | MEDLINE | ID: mdl-30413974

ABSTRACT

Extrahepatic transplantation of islets of Langerhans could aid in better survival of islets after transplantation. When islets are transfused into the liver 60-70% of them are lost immediately after transplantation. An important factor for a successful extrahepatic transplantation is a well-vascularized tissue surrounding the implant. There are many strategies known for enhancing vessel formation such as adding cells with endothelial potential, the combination with angiogenic factors and / or applying surface topography at the exposed surface of the device. Previously we developed porous, micropatterned membranes which can be applied as a lid for an islet encapsulation device and we showed that the surface topography induces human umbilical vein endothelial cell (HUVEC) alignment and interconnection. This was achieved without the addition of hydrogels, often used in angiogenesis assays. In this work, we went one step further towards clinical implementation of the device by combining this micropatterned lid with Mesenchymal Stem Cells (MSCs) to facilitate prevascularization in vivo. As for HUVECs, the micropatterned membranes induced MSC alignment and organization in vitro, an important contributor to vessel formation, whereas in vivo (subcutaneous rat model) they contributed to improved implant prevascularization. In fact, the combination of MSCs seeded on the micropatterned membrane induced the highest vessel formation score in 80% of the sections.


Subject(s)
Drug Compounding , Islets of Langerhans/growth & development , Membranes, Artificial , Mesenchymal Stem Cells , Tissue Scaffolds , Human Umbilical Vein Endothelial Cells , Humans , Islets of Langerhans/blood supply , Neovascularization, Physiologic
5.
Tissue Eng Part B Rev ; 23(6): 515-528, 2017 12.
Article in English | MEDLINE | ID: mdl-28490258

ABSTRACT

Mesenchymal stem cells (MSCs) are considered to hold great therapeutic value for cell-based therapy and for tissue regeneration in particular. Recent evidence indicates that the main underlying mechanism for MSCs' beneficial effects in tissue regeneration is based on their capability to produce a large variety of bioactive trophic factors that stimulate neighboring parenchymal cells to start repairing damaged tissues. These new findings could potentially replace the classical paradigm of MSC differentiation and cell replacement. These bioactive factors have diverse actions like modulating the local immune system, enhancing angiogenesis, preventing cell apoptosis, and stimulating survival, proliferation, and differentiation of resident tissue specific cells. Therefore, MSCs are referred to as conductors of tissue repair and regeneration by secreting trophic mediators. In this review article, we have summarized the studies that focused on the trophic effects of MSC within the context of tissue regeneration. We will also highlight the various underlying mechanisms used by MSCs to act as trophic mediators. Besides the secretion of growth factors, we discuss two additional mechanisms that are likely to mediate MSC's beneficial effects in tissue regeneration, namely the production of extracellular vesicles and the formation of membrane nanotubes, which can both connect different cells and transfer a variety of trophic factors varying from proteins to mRNAs and miRNAs. Furthermore, we postulate that apoptosis of the MSCs is an integral part of the trophic effect during tissue repair.


Subject(s)
Cardiovascular Diseases/therapy , Cell- and Tissue-Based Therapy , Extracellular Vesicles/physiology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Regeneration , Animals , Humans , Tissue Engineering , Wound Healing
6.
J Tissue Eng Regen Med ; 10(8): 669-78, 2016 08.
Article in English | MEDLINE | ID: mdl-24170734

ABSTRACT

The aim of this study was to evaluate the effect of PLGA microsphere dimensions on bone formation after injection of calcium phosphate cement (CPC)/PLGA in a guinea pig tibial intramedullarly model. To this end, injectable CPC/PLGA formulations were prepared using PLGA microspheres with either a small (~25 µm) or large (~100 µm) diameter, which were incorporated at a 20:80 ratio (wt%) within apatite CPC. Both CPC/PLGA formulations were injected into a marrow-ablated tibial intramedullary cavity and, after an implantation period of 12 weeks, histology and histomorphometry were used to address bone formation. The results demonstrated bone ingrowth throughout the entire scaffold material for both CPC/PLGA formulations upon PLGA microsphere degradation. More importantly, bone formation within the CPC matrix was > two-fold higher for CPC-PLGA with 25 µm PLGA microspheres. Additionally, the pattern of bone and marrow formation showed distinct differences related to PLGA microsphere dimension. In general, this study demonstrates that PLGA microsphere dimensions of ~25 µm, leading to pores of ~25 µm within CPC, are sufficient for bone ingrowth and allow substantial bone formation. Further, the results demonstrate that PLGA microsphere dimensions provide a tool to control bone formation for injectable CPC/PLGA bone substitutes. Copyright © 2013 John Wiley & Sons, Ltd.


Subject(s)
Bone Cements , Calcium Phosphates , Lactic Acid , Microspheres , Osteogenesis/drug effects , Polyglycolic Acid , Tibia , Animals , Bone Cements/chemistry , Bone Cements/pharmacology , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Female , Guinea Pigs , Lactic Acid/chemistry , Lactic Acid/pharmacology , Polyglycolic Acid/chemistry , Polyglycolic Acid/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer , Tibia/injuries , Tibia/metabolism , Tibia/pathology
7.
Acta Biomater ; 26: 136-44, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26292264

ABSTRACT

A terpyridine end-functionalized 8-arm poly(ethylene glycol) was prepared using the reaction of a 4'-aminopentanoxy substituted terpyridine with a p-nitrophenyl chloroformate activated PEG-(OH)8. Supramolecular complexation of the polymer terpyridine moieties by Fe(2+) ions was investigated using NMR, UV-Vis and dynamic light scattering experiments. At low concentrations addition of Fe(2+) ions to an aqueous solution of the polymer conjugate afforded nanogels with a single size distribution around 250 nm. At concentrations above 3 wt%, and at a 1:2 metal to ligand molar ratio, hydrogels were formed with increasing mechanical properties at increasing polymer concentrations. Using bovine chondrocytes, the biocompatibility and potential cytotoxicity of the polymer conjugate, nanogels and hydrogels were studied. The polymer conjugate with free ligands was toxic to the cells likely due to depletion of essential metal ions. When the terpyridine groups were complexed with Fe(2+) ions, both nanogel suspensions and hydrogels showed no cytotoxicity in direct contact with chondrocytes. Indirect contact of gels with chondrocytes using transwells revealed the absence of toxic components by leaching. A Live-Dead assay on chondrocytes encapsulated in the hydrogels indicated that the hydrogels are cytocompatible, revealing the potential use of these materials for biomedical and pharmaceutical applications. STATEMENT OF SIGNIFICANCE: The binding between transition metal ions and ligands with multiple binding sites can be almost as strong as covalent bonds. This metal-ligand charge transfer (MLCT) complexation was used to crosslink water soluble polymers into hydrogels. This approach to novel materials may find applications in the biomedical and pharmaceutical fields. Transition metal ions are essential trace elements present in tissue but up to now no cytotoxicity data of free ligands are available. Data presented show that free ligands are toxic to cells likely by depletion of trace metal ions, whereas kinetically stable complexes are not cytotoxic even when embedded in hydrogels. These results provide fundamental issues to be considered in the design of hydrogels crosslinked through metal ligand complexation.


Subject(s)
Chondrocytes/drug effects , Cross-Linking Reagents/chemistry , Hydrogels/chemistry , Hydrogels/toxicity , Metal Nanoparticles/chemistry , Metal Nanoparticles/toxicity , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cattle , Cell Survival/drug effects , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/physiology , Compressive Strength , Dose-Response Relationship, Drug , Elastic Modulus , Ions , Kinetics , Metal Nanoparticles/ultrastructure , Static Electricity , Stress, Mechanical
8.
J Biomed Mater Res A ; 103(7): 2251-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25370308

ABSTRACT

Biomaterial scaffolds meant to function as supporting structures to osteogenic cells play a pivotal role in bone tissue engineering. Recently, we synthesized an aliphatic polyurethane (PU) scaffold via a foaming method using non-toxic components. Through this procedure a uniform interconnected porous structure was created. Furthermore, hydroxyapatite (HA) particles were introduced into this process to increase the bioactivity of the PU matrix. To evaluate the biological performances of these PU-based scaffolds, their influence on in vitro cellular behavior and in vivo bone forming capacity of the engineered cell-scaffold constructs was investigated in this study. A simulated body fluid test demonstrated that the incorporation of 40 wt % HA particles significantly promoted the biomineralization ability of the PU scaffolds. Enhanced in vitro proliferation and osteogenic differentiation of the seeded mesenchymal stem cells were also observed on the PU/HA composite. Next, the cell-scaffold constructs were implanted subcutaneously in a nude mice model. After 8 weeks, a considerable amount of vascularized bone tissue with initial marrow stroma development was generated in both PU and PU/HA40 scaffold. In conclusion, the PU/HA composite is a potential scaffold for bone regeneration applications.


Subject(s)
Bone and Bones/physiology , Durapatite/pharmacology , Polyurethanes/pharmacology , Tissue Engineering , Animals , Biocompatible Materials , Microscopy, Electron, Scanning , Rats
9.
Acta Biomater ; 13: 254-65, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25463490

ABSTRACT

Recapitulation of endochondral ossification leads to a new concept of bone tissue engineering via a cartilage intermediate as an osteoinductive template. In this study, we aimed to investigate the influence of in vitro chondrogenic priming time for the creation of cartilage template on the in vivo endochondral bone formation both qualitatively and quantitatively. To this end, rat bone-marrow-derived mesenchymal stromal cells (MSCs) were seeded onto two scaffolds with distinguished features: a fibrous poly(lactic-co-glycolic acid)/poly(ε-caprolactone) electrospun scaffold (PLGA/PCL) and a porous hydroxyapatite/tricalcium phosphate composite (HA/TCP). The constructs were then chondrogenically differentiated for 2, 3 and 4 weeks in vitro, followed by subcutaneous implantation in vivo for up to 8 weeks. A longer chondrogenic priming time resulted in a significantly increased amount and homogeneous deposition of the cartilage matrix on both the PLGA/PCL and HA/TCP scaffolds in vitro. In vivo, all implanted constructs gave rise to endochondral bone formation, whereas the bone volume was not affected by the length of priming time. An unpolarized woven bone-like structure, with significant amounts of cartilage remaining, was generated in fibrous PLGA/PCL scaffolds, while porous HA/TCP scaffolds supported progressive lamellar-like bone formation with mature bone marrow development. These data suggest that, by utilizing a chondrogenically differentiated MSC-scaffold construct as cartilage template, 2 weeks of in vitro priming time is sufficient to generate a substantial amount of vascularized endochondral bone in vivo. The structure of the bone depends on the chemical and structural cues provided by the scaffold design.


Subject(s)
Bone Marrow Cells/metabolism , Chondrogenesis , Lactic Acid/chemistry , Mesenchymal Stem Cells/metabolism , Osteogenesis , Polyesters/chemistry , Polyglycolic Acid/chemistry , Tissue Scaffolds/chemistry , Animals , Bone Marrow Cells/cytology , Cells, Cultured , Male , Mesenchymal Stem Cells/cytology , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Inbred F344
10.
J Tissue Eng Regen Med ; 9(7): 779-88, 2015 Jul.
Article in English | MEDLINE | ID: mdl-23364774

ABSTRACT

Human bone marrow-derived mesenchymal stem cells (BM-MSCs) and human adipose tissue-derived mesenchymal stem cells (AT-MSCs) are the most frequently used stem cells in tissue engineering. Due to major clinical demands, it is necessary to find an optimally safe and efficient way for large-scale expansion of these cells. Considering the nutritional source in the culture medium and method, this study aimed to analyze the effects of FBS- and PL-supplemented media on osteogenesis in stem cell mono- and co-cultures with human umbilical vein endothelial cells (HUVECs). Results showed that cell metabolic activity and proliferation increased in PL- compared to FBS-supplemented media in mono- and co-cultures for both BM-MSCs and AT-MSCs. In addition, calcium deposition was cell type dependent and decreased for BM-MSCs but increased for AT-MSCs in PL-supplemented medium in both mono- and co-cultures. Based on the effects of co-cultures, BM-MSCs/HUVECs enhanced osteogenesis compared to BM-MSCs monocultures in both FBS- and PL-supplemented media whereas AT-MSCs/HUVECs showed similar results compared to AT-MSCs monocultures.


Subject(s)
Adipose Tissue/cytology , Bone Marrow Cells/cytology , Coculture Techniques/methods , Culture Media , Human Umbilical Vein Endothelial Cells/cytology , Mesenchymal Stem Cells/cytology , Adipose Tissue/metabolism , Bone Marrow Cells/metabolism , Culture Media/chemistry , Culture Media/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mesenchymal Stem Cells/metabolism
11.
J Biomed Mater Res A ; 103(2): 439-50, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24737694

ABSTRACT

The aim of this study was to compare the effect of implantation site (i.e., subcutaneous, SQ vs. intramuscular, IM) on bone forming capacity of cell-based and growth factor-based scaffolds in athymic nude rats after an implantation period of 8 weeks. Cell-based scaffolds consisted of porous hydroxyapatite/tricalcium phosphate (HA/TCP) scaffolds seeded with either human adipose tissue-derived mesenchymal stem cells (AT-MSCs) only or both AT-MSCs and human umbilical vein endothelial cells (HUVECs), which were precultured in osteogenic medium for 7 days. Growth factor-based scaffolds consisted of porous HA/TCP scaffolds with 20 µg preadsorbed bone morphogenetic protein-2 (BMP-2). Histological and histomorphometrical analysis were used to assess bone formation. A differentiation experiment was performed in parallel to compare the in vitro osteogenic capacity of cell-based scaffolds. The results showed that cell-based scaffolds showed evident osteogenic differentiation in vitro, with only marginal differences between AT-MSCs only and AT-MSCs/HUVECs. In vivo, none of the cell-based scaffolds showed bone formation, irrespective of the site of implantation. In contrast, all growth factor-based scaffolds showed bone formation at both implantation sites without differences in the amount of formed bone. In conclusion, the results of this study demonstrated that the bone forming capacity of HA/TCP scaffolds with pre-adsorbed BMP-2 was equal at different ectopic implantation sites. Further, despite obvious in vitro osteogenic differentiation of AT-MSCs and AT-MSCS/HUVECs on HA/TCP scaffolds, no bone formation of these cell-based scaffolds was observed in vivo. This indicates further investigation on bone formation mechanisms of AT-MSCs is needed before AT-MSCs can be used as a cytotherapeutic treatment in clinics.


Subject(s)
Calcium Phosphates/chemistry , Cell Differentiation , Durapatite/chemistry , Human Umbilical Vein Endothelial Cells/metabolism , Implants, Experimental , Osteogenesis , Tissue Scaffolds/chemistry , Animals , Bone Morphogenetic Protein 2/chemistry , Heterografts , Human Umbilical Vein Endothelial Cells/transplantation , Humans , Male , Rats , Rats, Nude
12.
Biomed Res Int ; 2014: 145423, 2014.
Article in English | MEDLINE | ID: mdl-24864228

ABSTRACT

OBJECTIVES: STRO-1 positive periodontal ligament cells (PDLCs) and unsorted PDLCs have demonstrated potential for periodontal regeneration, but the comparison between unsorted cells and the expanded STRO-1 sorted cells has never been reported. Additionally, Wnt3a is involved in cell proliferation thus may benefit in vitro PDLC expansion. The aim was to evaluate the effect of STRO-1 cell sorting and Wnt3a treatment on cell behavior of human PDLCs (hPDLCs). MATERIALS AND METHODS: STRO-1 positive hPDLCs were sorted and the sorted cells were expanded and compared with their unsorted parental cells. Thereafter, hPDLCs were treated with or without Wnt3a and the cell proliferation, self-renewal, and osteogenic differentiation were evaluated. RESULTS: No differences were measured between the expanded STRO-1-sorted cells and unsorted parental cells in terms of proliferation, CFU, and mineralization capacity. Wnt3a enhanced the proliferation and self-renewal ability of hPDLCs significantly as displayed by higher DNA content values, a shorter cell population doubling time, and higher expression of the self-renewal gene Oct4. Moreover, Wnt3a promoted the expansion of hPDLCs for 5 passages without affecting cell proliferation, CFU, and osteogenic capacity. CONCLUSIONS: Expanded STRO-1-sorted hPDLCs showed no superiority compared to their unsorted parental cells. On the other hand, Wnt3a promotes the efficient hPDLC expansion and retains the self-renewal and osteogenic differentiation capacity.


Subject(s)
Antigens, Surface/metabolism , Flow Cytometry , Periodontal Ligament/cytology , Stem Cells/cytology , Stem Cells/metabolism , Wnt3A Protein/pharmacology , Adult , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Humans , Osteogenesis/drug effects , Real-Time Polymerase Chain Reaction , Stem Cells/drug effects , Young Adult
13.
Biomaterials ; 35(21): 5482-90, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24731712

ABSTRACT

The prevalence of osteoporosis will increase within the next decades due to the aging world population, which can affect the bone healing response to dental and orthopedic implants. Consequently, local drug targeting of peri-implant bone has been proposed as a strategy for the enhancement of bone-implant integration in osteoporotic conditions. In the present study, an established in-vivo femoral condyle implantation model in osteoporotic and healthy bone is used to analyze the osteogenic capacity of titanium implants coated with bisphosphonate (BP)-loaded calcium phosphate nanoparticles (nCaP) under compromised medical conditions. After 4 weeks of implantation, peri-implant bone volume (%BV; by µCT) and bone area (%BA; by histomorphometry) were significantly increased within a distance of 500 µm from implant surfaces functionalized with BP compared to control implants in osteoporotic and healthy conditions. Interestingly, the deposition of nCaP/BP coatings onto implant surfaces increased both peri-implant bone contact (%BIC) and volume (%BV) compared to the deposition of nCaP or BP coatings individually, in osteoporotic and healthy conditions. The results of real-time PCR revealed similar osteogenic gene expression levels to all implant surfaces at 4-weeks post-implantation. In conclusion, simultaneous targeting of bone formation (by nCaP) and bone resorption (by BP) using nCaP/BP surface coatings represents an effective strategy for synergistically improvement of bone-implant integration, especially in osteoporotic conditions.


Subject(s)
Bone and Bones/drug effects , Calcium Phosphates/pharmacology , Diphosphonates/pharmacology , Nanoparticles/chemistry , Prostheses and Implants , Animals , Bone Resorption/drug therapy , Bone and Bones/metabolism , Calcium Phosphates/chemistry , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Diphosphonates/chemistry , Disease Models, Animal , Drug Synergism , Male , Osteogenesis/drug effects , Osteoporosis/therapy , Rats , Rats, Wistar , Surface Properties , Titanium/chemistry
14.
Macromol Biosci ; 14(7): 1004-14, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24687628

ABSTRACT

The aim of this study is to optimize enzymatic control over gelation of chitosan-based hydrogels for the delivery of periodontal ligament cells (PDLCs). The results reveal that the gelation time, strength, and degradation rate of the chitosan hydrogels can be controlled precisely by variation of the urea and urease concentrations. PDLCs remain viable inside these hydrogels for up to 30 days. Cells released from the hydrogel upon degradation and collected after 3, 15, and 30 days are able to form colonies and osteogenically differentiate. In conclusion, the enzymatic control over the gelation of chitosan hydrogels offers options for the delivery of PDLCs.


Subject(s)
Chitosan/pharmacology , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Periodontal Ligament/cytology , Urease/metabolism , Adolescent , Alkaline Phosphatase/metabolism , Cell Survival/drug effects , Cells, Cultured , Colony-Forming Units Assay , Elastic Modulus/drug effects , Female , Humans , Hydrogen-Ion Concentration , Kinetics , Periodontal Ligament/drug effects , Rheology/drug effects , Time Factors , Water/chemistry
15.
Biofabrication ; 6(1): 015005, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24429700

ABSTRACT

The aim of this study was to comparatively evaluate the angiogenic capacity of cocultures using either human bone marrow- or human adipose tissue-derived mesenchymal stem cells (MSCs) (BM- or AT-MSCs) with human umbilical vein endothelial cells (HUVECs) both in vitro and in vivo at early time points (i.e. days 3 and 7). In vitro, cells were either monocultured (i.e. BM-MSCs, AT-MSCs or HUVECs) or cocultured (i.e. BM-MSCs/HUVECs and AT-MSCs/HUVECs) on Thermanox® (2-dimensional, 2D) or in collagen gels (3-dimensional, 3D). For the in vivo experiment, cells (cocultures) were embedded in collagen gels and implanted subcutaneously in nude mice. For both in vitro and in vivo experiments, samples were collected on days 3 and 7 and histologically processed for hematoxylin-eosin and platelet endothelial cell adhesion molecule (PECAM-1; CD31) staining. For in vivo samples, quantitative parameters for evaluating angiogenesis included CD31-positive staining percentage, total vessel-like structure (VLS) area percentage, VLS density, and average VLS area (i.e. the size of per VLS). In vitro results showed the formation of VLS in both cocultures, while none of the monocultures showed VLS formation, irrespective of 2D or 3D culture condition. Although VLS formation occurred after in vivo implantation, no significant difference in angiogenic capacity was observed between the two cocultures, either on day 3 or on day 7. Further, VLS density decreased and anastomosis of the new human vessels with the murine host vasculature occurred over time. In conclusion, this study demonstrated that AT-MSCs/HUVECs and BM-MSCs/HUVECs have equal angiogenic capacity both in vitro and in vivo, and that vessels from donor origin can anastomose with the host vasculature within seven days of implantation.


Subject(s)
Adipose Tissue/cytology , Blood Vessels/growth & development , Bone Marrow Cells/physiology , Human Umbilical Vein Endothelial Cells/physiology , Mesenchymal Stem Cells/physiology , Neovascularization, Physiologic , Tissue Engineering , Adipose Tissue/metabolism , Adult , Animals , Blood Vessels/metabolism , Bone Marrow Cells/cytology , Cells, Cultured , Coculture Techniques , Female , Human Umbilical Vein Endothelial Cells/cytology , Humans , Male , Mesenchymal Stem Cells/cytology , Mice , Mice, Nude , Middle Aged , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Tissue Engineering/methods
16.
J Biomed Mater Res A ; 102(4): 1026-36, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23640784

ABSTRACT

The aim of this study was to compare the osteogenic capacity between human adipose tissue-derived mesenchymal stem cells (AT-MSCs) and their cocultures with human umbilical vein endothelial cells (HUVECs) in vitro and their biological performance in vivo. First, the optimal cell ratio in cocultures for osteogenic differentiation was determined by seeding AT-MSCs and HUVECs in ratios varying from 100:0 to 0:100 on tissue culture plates. Afterward, AT-MSCs and AT-MSCs/HUVECs (50:50) were seeded on porous titanium fiber mesh scaffolds (Ti) for both in vitro and in vivo osteogenic evaluation. For in vitro evaluation, cell osteogenic differentiation was assessed by alkaline phosphatase (ALP) activity and calcium assay. For in vivo evaluation, the scaffolds were implanted bilaterally into rat cranial defects (5 mm diameter) and bone formation was assessed histologically and histomorphometrically after 8 weeks. The ratio of 50:50 was chosen in the cocultures because this coculture condition retained similar amount of calcium deposition while using the least amount of AT-MSCs. Moreover, AT-MSCs showed higher osteogenic differentiation in comparison to AT-MSCs/HUVECs on Ti in vitro. Furthermore, superior bone formation was observed in AT-MSCs compared to AT-MSCs/HUVECs in rat cranial defects. In conclusion, AT-MSCs showed significantly higher osteogenic potential compared to AT-MSCs/HUVECs both in vitro and in vivo.


Subject(s)
Adipose Tissue/cytology , Human Umbilical Vein Endothelial Cells/cytology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Skull/pathology , Adult , Animals , Cell Differentiation/drug effects , Cells, Cultured , Coculture Techniques , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/ultrastructure , Humans , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/ultrastructure , Middle Aged , Neovascularization, Physiologic/drug effects , Osteogenesis/drug effects , Prosthesis Implantation , Rats , Rats, Nude , Skull/diagnostic imaging , Skull/drug effects , Tissue Scaffolds/chemistry , Titanium/pharmacology , X-Ray Microtomography
17.
Tissue Eng Part B Rev ; 20(4): 340-54, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24168361

ABSTRACT

Stem cells, having the potential for self-renewal and multilineage differentiation, are the building blocks for tissue/organ regeneration. Stem cells can be isolated from various sources but are, in general, available in too small numbers to be used directly for clinical purpose without intermediate expansion procedures in vitro. Although this in vitro expansion of undifferentiated stem cells is necessary, stem cells typically diminish their ability to self-renew and proliferate during passaging. Consequently, maintaining the stemness of stem cells has been recognized as a major challenge in stem cell-based research. This review focuses on the latest developments in maintaining the self-renewal ability of stem cells during in vitro expansion by biomaterial strategies. Further, this review highlights what should be the focus for future studies using stem cells for regenerative applications.


Subject(s)
Biocompatible Materials/chemistry , Cell Culture Techniques/methods , Cell Proliferation , Stem Cells/cytology , Humans , Stem Cells/metabolism
18.
J Biomed Mater Res A ; 102(4): 935-46, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23650286

ABSTRACT

Bone regenerative medicine, based on the combined use of cells and scaffolds, represents a promising strategy in bone regeneration. Hydrogels have attracted huge interests for application as a scaffold for minimally invasive surgery. Collagen and oligo(poly(ethylene glycol)fumarate) (OPF) hydrogels are the representatives of two main categories of hydrogels, that is, natural- and synthetic-based hydrogels. With these the optimal cell-loading (i.e., cell distribution inside the hydrogels) method was assessed. The cell behavior of both bone marrow- and adipose tissue-derived mesenchymal stem cells (BM- and AT-MSCs) in three loading methods, which are dispersed (i.e., homogeneous cell encapsulation, D), sandwich (i.e., cells located in between two hydrogel layers, S), and spheroid (i.e., cell pellets encapsulation, Sp) loading in two hydrogel systems (i.e., collagen and OPF), was compared. The results suggested that the cell behavior was influenced by the hydrogel type, meaning cells cultured in collagen hydrogels had higher proliferation and osteogenic differentiation capacity than in OPF hydrogels. In addition, AT-MSCs exhibited higher proliferation and osteogenic properties compared to BM-MSCs. However, no difference was observed for mineralization among the three loading methods, which did not approve the hypothesis that S and Sp loading would increase osteogenic capacity compared to D loading. In conclusion, D and Sp loading represents two promising cell loading methods for injectable bone substitute materials that allow application of minimally invasive surgery for cell-based regenerative treatment.


Subject(s)
Cell Culture Techniques/methods , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Mesenchymal Stem Cells/cytology , Adipose Tissue/cytology , Adult , Aged , Animals , Bone Marrow Cells/cytology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Cells, Cultured , Female , Humans , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Polyesters/pharmacology , Polyethylene Glycols/pharmacology , Rats , Spheroids, Cellular/cytology , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Staining and Labeling
19.
Tissue Eng Part A ; 20(1-2): 139-46, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23865551

ABSTRACT

The use of bioreactors for the in vitro culture of constructs for bone tissue engineering has become prevalent as these systems may improve the growth and differentiation of a cultured cell population. Here we utilize a tubular perfusion system (TPS) bioreactor for the in vitro culture of human mesenchymal stem cells (hMSCs) and implant the cultured constructs into rat femoral condyle defects. Using nanofibrous electrospun poly(lactic-co-glycolic acid)/poly(ε-caprolactone) scaffolds, hMSCs were cultured for 10 days in vitro in the TPS bioreactor with cellular and acellular scaffolds cultured statically for 10 days as a control. After 3 and 6 weeks of in vivo culture, explants were removed and subjected to histomorphometric analysis. Results indicated more rapid bone regeneration in defects implanted with bioreactor cultured scaffolds with a new bone area of 1.23 ± 0.35 mm(2) at 21 days compared to 0.99 ± 0.43 mm(2) and 0.50 ± 0.29 mm(2) in defects implanted with statically cultured scaffolds and acellular scaffolds, respectively. At the 21 day timepoint, statistical differences (p<0.05) were only observed between defects implanted with cell containing scaffolds and the acellular control. After 42 days, however, defects implanted with TPS cultured scaffolds had the greatest new bone area with 1.72 ± 0.40 mm(2). Defects implanted with statically cultured and acellular scaffolds had a new bone area of 1.26 ± 0.43 mm(2) and 1.19 ± 0.33 mm(2), respectively. The increase in bone growth observed in defects implanted with TPS cultured scaffolds was statistically significant (p<0.05) when compared to both the static and acellular groups at this timepoint. This study demonstrates the efficacy of the TPS bioreactor to improve bone tissue regeneration and highlights the benefits of utilizing perfusion bioreactor systems to culture MSCs for bone tissue engineering.


Subject(s)
Bioreactors , Bone Regeneration , Nanofibers/chemistry , Perfusion , Tissue Engineering/instrumentation , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Cells, Cultured , Femur/pathology , Humans , Implants, Experimental , Lactic Acid/chemistry , Nanofibers/ultrastructure , Polyesters/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Nude
20.
Tissue Eng Part C Methods ; 20(3): 198-204, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23898861

ABSTRACT

The murine-derived MC3T3-E1 cell line provided by the American Type Culture Collection (ATCC) is a well-known osteogenic cell culture model system to test materials in vitro. However, the effect of passaging on its mineralization capacity has never been described and their culture supplements can be further optimized. Therefore, we evaluated the influence of the passage number and different osteogenic culture supplements, including ascorbic acid (AsAP) and dexamethasone (Dex) on the osteogenic capacity of MC3T3-E1 cells. This capacity was measured by the deposited calcium, the alkaline phosphatase activity, and the expression of osteogenic-related genes, including bone sialoprotein (BSP), osteocalcin (OC), and osteopontin (OPN). The results indicated that the mineralization capacity of MC3T3-E1 cells significantly decreased during passaging and got exhausted at passage 34, as assessed by measuring calcium deposition after 28 days of osteogenic induction. Moreover, the combination of AsAP and Dex triggered significantly more mineralization in MC3T3-E1 cells than the ATCC recommended addition of AsAP alone, as indicated by increased calcium deposition and higher expression of BSP and OPN. However, Dex alone could not trigger this effect, but only in combination with the AsAP, which indicates that Dex has no direct effect on mineralization. In conclusion, the passage number of MC3T3-E1 cells is of great importance and the use of cells above 30 passages should be avoided. In addition, the favored osteogenic supplements providing an improved osteogenic differentiation of MC3T3-E1 cells are the combination of AsAP and Dex.


Subject(s)
Calcification, Physiologic , Cell Culture Techniques/methods , Osteoblasts/cytology , Osteogenesis , Alkaline Phosphatase/metabolism , Animals , Calcium/metabolism , Cell Line , Cell Shape , Cell Survival , Gene Expression Regulation , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...