Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Stem Cells Transl Med ; 8(9): 925-934, 2019 09.
Article in English | MEDLINE | ID: mdl-31020798

ABSTRACT

Ionizing radiation, commonly used in the treatment of solid tumors, has unintended but deleterious effects on overlying skin and is associated with chronic nonhealing wounds. Skin-derived mesenchymal stromal cells (SMSCs) are a pluripotent population of cells that are critically involved in skin homeostasis and wound healing. The aim of this study was to isolate and functionally characterize SMSCs from human skin that was previously irradiated as part of neoadjuvant or adjuvant cancer therapy. To this end, SMSCs were isolated from paired irradiated and nonirradiated human skin samples. Irradiated SMSCs expressed characteristic SMSC markers at lower levels, had disorganized cytoskeletal structure, and had disordered morphology. Functionally, these cells had diminished proliferative capacity and substantial defects in colony-forming capacity and differentiation in vitro. These changes were associated with significant differential expression of genes known to be involved in skin physiology and wound healing. Conditioned media obtained from irradiated SMSCs affected fibroblast but not endothelial cell proliferation and migration. These results suggest that in situ damage to SMSCs during neoadjuvant or adjuvant radiation may play a critical role in the pathogenesis of slow or nonhealing radiation wounds. Stem Cells Translational Medicine 2019;8:925&934.


Subject(s)
Cell Differentiation , Cell Proliferation , Mesenchymal Stem Cells/cytology , Paracrine Communication , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adipogenesis , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Formins/genetics , Formins/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Neoplasms/pathology , Neoplasms/radiotherapy , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Osteogenesis , Paracrine Communication/radiation effects , Radiation, Ionizing , Skin/cytology , Skin/pathology , Skin/radiation effects , Transcriptome/radiation effects
2.
Ann Plast Surg ; 80(5S Suppl 5): S317-S321, 2018 05.
Article in English | MEDLINE | ID: mdl-29553981

ABSTRACT

Impaired healing of the skin is a notable cause of patient morbidity and mortality. In diabetic individuals, dysregulated inflammation contributes to delayed wound healing. Specific immunomodulatory agents may have a role in the treatment of diabetic wounds. One of these molecules is interleukin-1 receptor antagonist (Anakinra; Amgen Corp.). Although interleukin-1 receptor antagonist (Anakinra; Amgen Corp.) is approved by the Food and Drug Administration (FDA) for the treatment of rheumatoid arthritis and neonatal-onset multisystem inflammatory disease, little is known about the local use this drug in cutaneous wound healing. Therefore, the aim of this study is to determine the effect of locally administered interleukin-1 receptor antagonist on delayed wound healing, specifically, in a diabetic mouse model. Two 6-mm full-thickness wounds were created on the dorsa of diabetic (db/db) mice and stented. One-hour postwounding, wound margins were subcutaneously injected with either (1) low-dose interleukin-1 receptor antagonist in a gelatin-transglutaminase gel vehicle or (2) the gel vehicle only. Wounds were imaged on days 0, 7, 14, and 21 postwounding, and wound area was determined. Wound biopsies were collected on day 21 and immunohistochemically stained for neutrophil and macrophage infiltration. Wounds treated with interleukin-1 receptor antagonist had significantly smaller wound area than nontreated wounds on day 7 and day 14 postwounding. Treated wounds also showed significantly less neutrophil and macrophage infiltration. These findings support the hypothesis that interleukin-1 receptor antagonist may have an important role in cutaneous wound healing, possibly by promoting successful resolution of acute inflammation and hence accelerating wound closure. Thereby, administration of IL-1Ra may be useful in the treatment of nonhealing wounds.


Subject(s)
Diabetes Complications/drug therapy , Diabetes Mellitus, Experimental/drug therapy , Interleukin 1 Receptor Antagonist Protein/administration & dosage , Wound Healing/drug effects , Administration, Cutaneous , Animals , Diabetes Complications/pathology , Diabetes Mellitus, Experimental/pathology , Disease Models, Animal , Humans , Leukocyte Count , Male , Mice , Mice, Inbred NOD , Neutrophils/drug effects , Neutrophils/pathology , Skin/drug effects , Skin/pathology
3.
Sci Transl Med ; 10(432)2018 03 14.
Article in English | MEDLINE | ID: mdl-29540618

ABSTRACT

Mesenchymal stem cells (MSCs) are capable of secreting exosomes, extracellular vesicles, and cytokines to regulate cell and tissue homeostasis. However, it is unknown whether MSCs use a specific exocytotic fusion mechanism to secrete exosomes and cytokines. We show that Fas binds with Fas-associated phosphatase-1 (Fap-1) and caveolin-1 (Cav-1) to activate a common soluble N-ethylmaleimide-sensitive factor (NSF) attachment protein receptor (SNARE)-mediated membrane fusion mechanism to release small extracellular vesicles (sEVs) in MSCs. Moreover, we reveal that MSCs produce and secrete interleukin-1 receptor antagonist (IL-1RA) associated with sEVs to maintain rapid wound healing in the gingiva via the Fas/Fap-1/Cav-1 cascade. Tumor necrosis factor-α (TNF-α) serves as an activator to up-regulate Fas and Fap-1 expression via the nuclear factor κB pathway to promote IL-1RA release. This study identifies a previously unknown Fas/Fap-1/Cav-1 axis that regulates SNARE-mediated sEV and IL-1RA secretion in stem cells, which contributes to accelerated wound healing.


Subject(s)
Caveolin 1/metabolism , Interleukin 1 Receptor Antagonist Protein/metabolism , Mesenchymal Stem Cells/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 13/metabolism , fas Receptor/metabolism , Animals , Caveolin 1/genetics , Female , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Knockout , Protein Tyrosine Phosphatase, Non-Receptor Type 13/genetics , Wound Healing/genetics , Wound Healing/physiology , fas Receptor/genetics
4.
Ann Biomed Eng ; 44(6): 1908-20, 2016 06.
Article in English | MEDLINE | ID: mdl-27009085

ABSTRACT

Repair and regeneration of muscle tissue following traumatic injuries or muscle diseases often presents a challenging clinical situation. If a significant amount of tissue is lost the native regenerative potential of skeletal muscle will not be able to grow to fill the defect site completely. Dental-derived mesenchymal stem cells (MSCs) in combination with appropriate scaffold material, present an advantageous alternative therapeutic option for muscle tissue engineering in comparison to current treatment modalities available. To date, there has been no report on application of gingival mesenchymal stem cells (GMSCs) in three-dimensional scaffolds for muscle tissue engineering. The objectives of the current study were to develop an injectable 3D RGD-coupled alginate scaffold with multiple growth factor delivery capacity for encapsulating GMSCs, and to evaluate the capacity of encapsulated GMSCs to differentiate into myogenic tissue in vitro and in vivo where encapsulated GMSCs were transplanted subcutaneously into immunocompromised mice. The results demonstrate that after 4 weeks of differentiation in vitro, GMSCs as well as the positive control human bone marrow mesenchymal stem cells (hBMMSCs) exhibited muscle cell-like morphology with high levels of mRNA expression for gene markers related to muscle regeneration (MyoD, Myf5, and MyoG) via qPCR measurement. Our quantitative PCR analyzes revealed that the stiffness of the RGD-coupled alginate regulates the myogenic differentiation of encapsulated GMSCs. Histological and immunohistochemical/fluorescence staining for protein markers specific for myogenic tissue confirmed muscle regeneration in subcutaneous transplantation in our in vivo animal model. GMSCs showed significantly greater capacity for myogenic regeneration in comparison to hBMMSCs (p < 0.05). Altogether, our findings confirmed that GMSCs encapsulated in RGD-modified alginate hydrogel with multiple growth factor delivery capacity is a promising candidate for muscle tissue engineering.


Subject(s)
Alginates/chemistry , Gingiva/metabolism , Hydrogels/chemistry , Intercellular Signaling Peptides and Proteins/pharmacology , Mesenchymal Stem Cells/metabolism , Muscle, Skeletal/metabolism , Tissue Engineering , Animals , Cells, Immobilized/cytology , Cells, Immobilized/metabolism , Female , Gingiva/cytology , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Mesenchymal Stem Cells/cytology , Mice , Mice, Nude , Muscle, Skeletal/cytology , Oligopeptides/pharmacology , Tissue Scaffolds/chemistry
5.
Adv Funct Mater ; 25(15): 2296-2307, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-26120294

ABSTRACT

The host immune system is known to influence mesenchymal stem cell (MSC)-mediated bone tissue regeneration. However, the therapeutic capacity of hydrogel biomaterial to modulate the interplay between MSCs and T-lymphocytes is unknown. Here it is shown that encapsulating hydrogel affects this interplay when used to encapsulate MSCs for implantation by hindering the penetration of pro-inflammatory cells and/or cytokines, leading to improved viability of the encapsulated MSCs. This combats the effects of the host pro-inflammatory T-lymphocyte-induced nuclear factor kappaB pathway, which can reduce MSC viability through the CASPASE-3 and CAS-PASE-8 associated proapoptotic cascade, resulting in the apoptosis of MSCs. To corroborate rescue of engrafted MSCs from the insult of the host immune system, the incorporation of the anti-inflammatory drug indomethacin into the encapsulating alginate hydrogel further regulates the local microenvironment and prevents pro-inflammatory cytokine-induced apoptosis. These findings suggest that the encapsulating hydrogel can regulate the MSC-host immune cell interplay and direct the fate of the implanted MSCs, leading to enhanced tissue regeneration.

6.
J Mater Sci Mater Med ; 26(3): 153, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25773231

ABSTRACT

Dental-derived mesenchymal stem cells (MSCs) provide an advantageous therapeutic option for tissue engineering due to their high accessibility and bioavailability. However, delivering MSCs to defect sites while maintaining a high MSC survival rate is still a critical challenge in MSC-mediated tissue regeneration. Here, we tested the osteogenic and adipogenic differentiation capacity of dental pulp stem cells (DPSCs) in a thermoreversible Pluronic F127 hydrogel scaffold encapsulation system in vitro. DPSCs were encapsulated in Pluronic (®) F-127 hydrogel and stem cell viability, proliferation and differentiation into adipogenic and osteogenic tissues were evaluated. The degradation profile and swelling kinetics of the hydrogel were also analyzed. Our results confirmed that Pluronic F-127 is a promising and non-toxic scaffold for encapsulation of DPSCs as well as control human bone marrow MSCs (hBMMSCs), yielding high stem cell viability and proliferation. Moreover, after 2 weeks of differentiation in vitro, DPSCs as well as hBMMSCs exhibited high levels of mRNA expression for osteogenic and adipogenic gene markers via PCR analysis. Our histochemical staining further confirmed the ability of Pluronic F-127 to direct the differentiation of these stem cells into osteogenic and adipogenic tissues. Furthermore, our results revealed that Pluronic F-127 has a dense tubular and reticular network morphology, which contributes to its high permeability and solubility, consistent with its high degradability in the tested conditions. Altogether, our findings demonstrate that Pluronic F-127 is a promising scaffold for encapsulation of DPSCs and can be considered for cell delivery purposes in tissue engineering.


Subject(s)
Hydrogels , Mesenchymal Stem Cells/cytology , Poloxamer/chemistry , Tissue Scaffolds , Tooth/cytology , Adolescent , Adult , Humans , Male , Young Adult
7.
J Exp Med ; 212(1): 73-91, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25534817

ABSTRACT

Fibrillin-1 (FBN1) deficiency-induced systemic sclerosis is attributed to elevation of interleukin-4 (IL4) and TGF-ß, but the mechanism underlying FBN1 deficiency-associated osteopenia is not fully understood. We show that bone marrow mesenchymal stem cells (BMMSCs) from FBN1-deficient (Fbn1(+/-)) mice exhibit decreased osteogenic differentiation and increased adipogenic differentiation. Mechanistically, this lineage alteration is regulated by IL4/IL4Rα-mediated activation of mTOR signaling to down-regulate RUNX2 and up-regulate PPARγ2, respectively, via P70 ribosomal S6 protein kinase (P70S6K). Additionally, we reveal that activation of TGF-ß/SMAD3/SP1 signaling results in enhancement of SP1 binding to the IL4Rα promoter to synergistically activate mTOR pathway in Fbn1(+/-) BMMSCs. Blockage of mTOR signaling by osteoblastic-specific knockout or rapamycin treatment rescues osteopenia phenotype in Fbn1(+/-) mice by improving osteogenic differentiation of BMMSCs. Collectively, this study identifies a previously unrecognized role of the FBN1/TGF-ß/IL4Rα/mTOR cascade in BMMSC lineage selection and provides experimental evidence that rapamycin treatment may provide an anabolic therapy for osteopenia in Fbn1(+/-) mice.


Subject(s)
Bone Diseases, Metabolic/metabolism , Microfilament Proteins/metabolism , Receptors, Cell Surface/metabolism , Scleroderma, Systemic/metabolism , TOR Serine-Threonine Kinases/metabolism , Adipogenesis/drug effects , Adipogenesis/genetics , Animals , Bone Diseases, Metabolic/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Female , Fibrillin-1 , Fibrillins , Flow Cytometry , Immunoblotting , Immunosuppressive Agents/pharmacology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microfilament Proteins/genetics , Microscopy, Fluorescence , Osteogenesis/drug effects , Osteogenesis/genetics , RNA Interference , Receptors, Cell Surface/genetics , Scleroderma, Systemic/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Transforming Growth Factor beta/metabolism
8.
EMBO Mol Med ; 6(3): 322-34, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24401839

ABSTRACT

Bone marrow mesenchymal stem cells (BMMSCs) are capable of differentiating into multiple cell types and regulating immune cell response. However, the mechanisms that govern the immunomodulatory properties of BMMSCs are still not fully elucidated. Here we show that telomerase-deficient BMMSCs lose their capacity to inhibit T cells and ameliorate the disease phenotype in systemic sclerosis mice. Restoration of telomerase activity by telomerase reverse transcriptase (TERT) transfection in TERT(-/-) BMMSCs rescues their immunomodulatory functions. Mechanistically, we reveal that TERT, combined with ß-catenin and BRG1, serves as a transcriptional complex, which binds the FAS ligand (FASL) promoter to upregulate FASL expression, leading to an elevated immunomodulatory function. To test the translational value of these findings in the context of potential clinical therapy, we used aspirin treatment to upregulate telomerase activity in BMMSCs, and found a significant improvement in the immunomodulatory capacity of BMMSCs. Taken together, these findings identify a previously unrecognized role of TERT in improving the immunomodulatory capacity of BMMSCs, suggesting that aspirin treatment is a practical approach to significantly reduce cell dosage in BMMSC-based immunotherapies.


Subject(s)
Fas Ligand Protein/metabolism , Gene Expression Regulation , Mesenchymal Stem Cells/metabolism , Telomerase/metabolism , Adipogenesis , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Bone Marrow Cells/cytology , Cell Differentiation , Cells, Cultured , DNA Helicases/metabolism , Disease Models, Animal , Fas Ligand Protein/genetics , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Scleroderma, Systemic/surgery , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Telomerase/deficiency , Telomerase/genetics , Transcription Factors/metabolism , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism
9.
Biomaterials ; 35(9): 2642-50, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24397989

ABSTRACT

Tendon injuries are often associated with significant dysfunction and disability due to tendinous tissue's very limited self-repair capacity and propensity for scar formation. Dental-derived mesenchymal stem cells (MSCs) in combination with appropriate scaffold material present an alternative therapeutic option for tendon repair/regeneration that may be advantageous compared to other current treatment modalities. The MSC delivery vehicle is the principal determinant for successful implementation of MSC-mediated regenerative therapies. In the current study, a co-delivery system based on TGF-ß3-loaded RGD-coupled alginate microspheres was developed for encapsulating periodontal ligament stem cells (PDLSCs) or gingival mesenchymal stem cells (GMSCs). The capacity of encapsulated dental MSCs to differentiate into tendon tissue was investigated in vitro and in vivo. Encapsulated dental-derived MSCs were transplanted subcutaneously into immunocompromised mice. Our results revealed that after 4 weeks of differentiation in vitro, PDLSCs and GMSCs as well as the positive control human bone marrow mesenchymal stem cells (hBMMSCs) exhibited high levels of mRNA expression for gene markers related to tendon regeneration (Scx, DCn, Tnmd, and Bgy) via qPCR measurement. In a corresponding in vivo animal model, ectopic neo-tendon regeneration was observed in subcutaneous transplanted MSC-alginate constructs, as confirmed by histological and immunohistochemical staining for protein markers specific for tendons. Interestingly, in our quantitative PCR and in vivo histomorphometric analyses, PDLSCs showed significantly greater capacity for tendon regeneration than GMSCs or hBMMSCs (P < 0.05). Altogether, these findings indicate that periodontal ligament and gingival tissues can be considered as suitable stem cell sources for tendon engineering. PDLSCs and GMSCs encapsulated in TGF-ß3-loaded RGD-modified alginate microspheres are promising candidates for tendon regeneration.


Subject(s)
Gingiva/cytology , Mesenchymal Stem Cells/cytology , Periodontal Ligament/cytology , Regeneration/physiology , Tendons/physiology , Adolescent , Adult , Animals , Biocompatible Materials/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Lineage/drug effects , Cells, Immobilized/cytology , Cells, Immobilized/drug effects , Gene Expression Regulation/drug effects , Humans , Immunohistochemistry , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Regeneration/drug effects , Tendons/drug effects , Transforming Growth Factor beta3/pharmacology , Young Adult
10.
Tissue Eng Part A ; 20(3-4): 611-21, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24070211

ABSTRACT

Mesenchymal stem cells (MSCs) provide an advantageous alternative therapeutic option for bone regeneration in comparison to current treatment modalities. However, delivering MSCs to the defect site while maintaining a high MSC survival rate is still a critical challenge in MSC-mediated bone regeneration. Here, we tested the bone regeneration capacity of periodontal ligament stem cells (PDLSCs) and gingival mesenchymal stem cells (GMSCs) encapsulated in a novel RGD- (arginine-glycine-aspartic acid tripeptide) coupled alginate microencapsulation system in vitro and in vivo. Five-millimeter-diameter critical-size calvarial defects were created in immunocompromised mice and PDLSCs and GMSCs encapsulated in RGD-modified alginate microspheres were transplanted into the defect sites. New bone formation was assessed using microcomputed tomography and histological analyses 8 weeks after transplantation. Results confirmed that our microencapsulation system significantly enhanced MSC viability and osteogenic differentiation in vitro compared with non-RGD-containing alginate hydrogel microspheres with larger diameters. Results confirmed that PDLSCs were able to repair the calvarial defects by promoting the formation of mineralized tissue, while GMSCs showed significantly lower osteogenic differentiation capability. Further, results revealed that RGD-coupled alginate scaffold facilitated the differentiation of oral MSCs toward an osteoblast lineage in vitro and in vivo, as assessed by expression of osteogenic markers Runx2, ALP, and osteocalcin. In conclusion, these results for the first time demonstrated that MSCs derived from orofacial tissue encapsulated in RGD-modified alginate scaffold show promise for craniofacial bone regeneration. This treatment modality has many potential dental and orthopedic applications.


Subject(s)
Alginates/pharmacology , Bone Regeneration/drug effects , Gingiva/cytology , Mesenchymal Stem Cells/cytology , Oligopeptides/pharmacology , Periodontal Ligament/cytology , Tissue Scaffolds/chemistry , Adolescent , Adult , Animals , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Survival/drug effects , Cells, Immobilized/cytology , Cells, Immobilized/drug effects , Cells, Immobilized/metabolism , Colony-Forming Units Assay , Glucuronic Acid/pharmacology , Hexuronic Acids/pharmacology , Humans , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Microfluidics , Models, Animal , Osteogenesis/drug effects , Skull/pathology , Transcription Factors/metabolism , Young Adult
11.
J Biomed Mater Res A ; 101(11): 3285-94, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23983201

ABSTRACT

Bone grafts are currently the major family of treatment options in modern reconstructive dentistry. As an alternative, stem cell-scaffold constructs seem to hold promise for bone tissue engineering. However, the feasibility of encapsulating dental-derived mesenchymal stem cells in scaffold biomaterials such as alginate hydrogel remains to be tested. The objectives of this study were, therefore, to: (1) develop an injectable scaffold based on oxidized alginate microbeads encapsulating periodontal ligament stem cells (PDLSCs) and gingival mesenchymal stem cells (GMSCs); and (2) investigate the cell viability and osteogenic differentiation of the stem cells in the microbeads both in vitro and in vivo. Microbeads with diameters of 1 ± 0.1 mm were fabricated with 2 × 10(6) stem cells/mL of alginate. Microbeads containing PDLSCs, GMSCs, and human bone marrow mesenchymal stem cells as a positive control were implanted subcutaneously and ectopic bone formation was analyzed by micro CT and histological analysis at 8-weeks postimplantation. The encapsulated stem cells remained viable after 4 weeks of culturing in osteo-differentiating induction medium. Scanning electron microscopy and X-ray diffraction results confirmed that apatitic mineral was deposited by the stem cells. In vivo, ectopic mineralization was observed inside and around the implanted microbeads containing the immobilized stem cells. These findings demonstrate for the first time that immobilization of PDLSCs and GMSCs in alginate microbeads provides a promising strategy for bone tissue engineering.


Subject(s)
Bone and Bones/physiology , Gingiva/cytology , Mesenchymal Stem Cells/cytology , Periodontal Ligament/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Adolescent , Adult , Alginates/pharmacology , Animals , Biodegradation, Environmental , Biomarkers/metabolism , Bone and Bones/diagnostic imaging , Bone and Bones/drug effects , Cell Count , Cells, Immobilized/cytology , Cells, Immobilized/drug effects , Gene Expression Regulation/drug effects , Glucuronic Acid/pharmacology , Hexuronic Acids/pharmacology , Humans , Injections , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Osteogenesis/drug effects , Osteogenesis/genetics , Radiography , Staining and Labeling , Young Adult
12.
Acta Biomater ; 9(12): 9343-50, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23891740

ABSTRACT

Dental-derived mesenchymal stem cells (MSCs) are promising candidates for cartilage regeneration, with a high capacity for chondrogenic differentiation. This property helps make dental MSCs an advantageous therapeutic option compared to current treatment modalities. The MSC delivery vehicle is the principal determinant for the success of MSC-mediated cartilage regeneration therapies. The objectives of this study were to: (1) develop a novel co-delivery system based on TGF-ß1 loaded RGD-coupled alginate microspheres encapsulating periodontal ligament stem cells (PDLSCs) or gingival mesenchymal stem cells (GMSCs); and (2) investigate dental MSC viability and chondrogenic differentiation in alginate microspheres. The results revealed the sustained release of TGF-ß1 from the alginate microspheres. After 4 weeks of chondrogenic differentiation in vitro, PDLSCs and GMSCs as well as human bone marrow mesenchymal stem cells (hBMMSCs) (as positive control) revealed chondrogenic gene expression markers (Col II and Sox-9) via qPCR, as well as matrix positively stained by Toluidine Blue and Safranin-O. In animal studies, ectopic cartilage tissue regeneration was observed inside and around the transplanted microspheres, confirmed by histochemical and immunofluorescent staining. Interestingly, PDLSCs showed more chondrogenesis than GMSCs and hBMMSCs (p<0.05). Taken together, these results suggest that RGD-modified alginate microencapsulating dental MSCs make a promising candidate for cartilage regeneration. Our results highlight the vital role played by the microenvironment, as well as value of presenting inductive signals for viability and differentiation of MSCs.


Subject(s)
Alginates/chemistry , Cartilage/physiology , Drug Delivery Systems , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Mesenchymal Stem Cells/cytology , Periodontium/cytology , Regeneration/physiology , Adolescent , Adult , Animals , Cartilage/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Cellular Microenvironment/drug effects , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrogenesis/drug effects , Drug Compounding , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Humans , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Microspheres , Regeneration/drug effects , Subcutaneous Tissue/drug effects , Transforming Growth Factor beta1/pharmacology , Young Adult
13.
Biomaterials ; 34(28): 6572-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23773817

ABSTRACT

Recently, it has been shown that tethered anti-BMP2 monoclonal antibodies (mAbs) can trap BMP ligands and thus provide BMP inductive signals for osteo-differentiation of progenitor cells. The objectives of this study were to: (1) develop a co-delivery system based on murine anti-BMP2 mAb-loaded alginate microspheres encapsulating human bone marrow mesenchymal stem cells (hBMMSCs); and (2) investigate osteogenic differentiation of encapsulated stem cells in alginate microspheres in vitro and in vivo. Alginate microspheres of 1 ± 0.1 mm diameter were fabricated with 2 × 10(6) hBMMSCs per mL of alginate. Critical-size calvarial defects (5 mm diameter) were created in immune-compromised mice and alginate microspheres preloaded with anti-BMP mAb encapsulating hBMMSCs were transplanted into defect sites. Alginate microspheres pre-loaded with isotype-matched non-specific antibody were used as the negative control. After 8 weeks, micro CT and histologic analyses were used to analyze bone formation. In vitro analysis demonstrated that anti-BMP2 mAbs tethered BMP2 ligands that can activate the BMP receptors on hBMMSCs. The co-delivery system described herein, significantly enhanced hBMMSC-mediated osteogenesis, as confirmed by the presence of BMP signal pathway-activated osteoblast determinants Runx2 and ALP. Our results highlight the importance of engineering the microenvironment for stem cells, and particularly the value of presenting inductive signals for osteo-differentiation of hBMMSCs by tethering BMP ligands using mAbs. This strategy of engineering the microenvironment with captured BMP signals is a promising modality for repair and regeneration of craniofacial, axial and appendicular bone defects.


Subject(s)
Alginates/chemistry , Bone Morphogenetic Protein 2/immunology , Mesenchymal Stem Cells/cytology , Microspheres , Tissue Engineering/methods , Adult , Animals , Antibodies, Monoclonal/immunology , Cell Differentiation , Cells, Cultured , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Humans , Immunohistochemistry , Mice , Young Adult
14.
J Mater Sci Mater Med ; 23(12): 3041-51, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22945383

ABSTRACT

The objectives of this study were to: (1) develop an injectable and biodegradable scaffold based on oxidized alginate microbeads encapsulating periodontal ligament (PDLSCs) and gingival mesenchymal stem cells (GMSCs); and (2) investigate the stem cell viability, and osteogenic differentiation of the stem cells in vitro. Stem cells were encapsulated using alginate hydrogel. The stem cell viability, proliferation and differentiation to adipogenic and osteogenic tissues were studied. To investigate the expression of both adipogenesis and ontogenesis related genes, the RNA was extracted and RT-PCR was performed. The degradation behavior of hydrogel based on oxidized sodium alginate with different degrees of oxidation was studied in PBS at 37 °C as a function of time by monitoring the changes in weight loss. The swelling kinetics of alginate hydrogel was also investigated. The results showed that alginate is a promising candidate as a non-toxic scaffold for PDLSCs and GMSCs. It also has the ability to direct the differentiation of these stem cells to osteogenic and adipogenic tissues as compared to the control group in vitro. The encapsulated stem cells remained viable in vitro and both osteo-differentiated and adipo-differentiated after 4 weeks of culturing in the induction media. It was found that the degradation profile and swelling kinetics of alginate hydrogel strongly depends on the degree of oxidation showing its tunable chemistry and degradation rate. These findings demonstrate for the first time that immobilization of PDLSCs and GMSCs in the alginate microspheres provides a promising strategy for bone tissue engineering.


Subject(s)
Alginates/chemistry , Hydrogels/chemistry , Stem Cells/cytology , Tissue Scaffolds/chemistry , Adipogenesis , Adolescent , Adult , Bone and Bones/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Cell Survival , Culture Media/pharmacology , Gingiva/pathology , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Humans , Kinetics , Male , Mesenchymal Stem Cells/cytology , Periodontal Ligament/pathology , Temperature , Tissue Engineering/methods
15.
Cell Stem Cell ; 10(5): 544-55, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22542159

ABSTRACT

Systemic infusion of bone marrow mesenchymal stem cells (BMMSCs) yields therapeutic benefit for a variety of autoimmune diseases, but the underlying mechanisms are poorly understood. Here we show that in mice systemic infusion of BMMSCs induced transient T cell apoptosis via the FAS ligand (FASL)-dependent FAS pathway and could ameliorate disease phenotypes in fibrillin-1 mutated systemic sclerosis (SS) and dextran-sulfate-sodium-induced experimental colitis. FASL(-/-) BMMSCs did not induce T cell apoptosis in recipients, and could not ameliorate SS and colitis. Mechanistic analysis revealed that FAS-regulated monocyte chemotactic protein 1 (MCP-1) secretion by BMMSCs recruited T cells for FASL-mediated apoptosis. The apoptotic T cells subsequently triggered macrophages to produce high levels of TGFß, which in turn led to the upregulation of CD4(+)CD25(+)Foxp3(+) regulatory T cells and, ultimately, immune tolerance. These data therefore demonstrate a previously unrecognized mechanism underlying BMMSC-based immunotherapy involving coupling via FAS/FASL to induce T cell apoptosis.


Subject(s)
Colitis/immunology , Fas Ligand Protein/metabolism , Immunotherapy , Mesenchymal Stem Cells/immunology , Scleroderma, Systemic/immunology , fas Receptor/metabolism , Animals , Apoptosis , Bone Marrow Cells/immunology , Chemokine CCL2/metabolism , Colitis/chemically induced , Colitis/therapy , Dextran Sulfate/administration & dosage , Fas Ligand Protein/genetics , Fibrillin-1 , Fibrillins , Humans , Immune Tolerance , Mesenchymal Stem Cell Transplantation , Mice , Mice, Knockout , Microfilament Proteins/genetics , Scleroderma, Systemic/genetics , Scleroderma, Systemic/therapy , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism
16.
Nat Med ; 17(12): 1594-601, 2011 Nov 20.
Article in English | MEDLINE | ID: mdl-22101767

ABSTRACT

Stem cell-based regenerative medicine is a promising approach in tissue reconstruction. Here we show that proinflammatory T cells inhibit the ability of exogenously added bone marrow mesenchymal stem cells (BMMSCs) to mediate bone repair. This inhibition is due to interferon γ (IFN-γ)-induced downregulation of the runt-related transcription factor 2 (Runx-2) pathway and enhancement of tumor necrosis factor α (TNF-α) signaling in the stem cells. We also found that, through inhibition of nuclear factor κB (NF-κB), TNF-α converts the signaling of the IFN-γ-activated, nonapoptotic form of TNF receptor superfamily member 6 (Fas) in BMMSCs to a caspase 3- and caspase 8-associated proapoptotic cascade, resulting in the apoptosis of these cells. Conversely, reduction of IFN-γ and TNF-α concentrations by systemic infusion of Foxp3(+) regulatory T cells, or by local administration of aspirin, markedly improved BMMSC-based bone regeneration and calvarial defect repair in C57BL/6 mice. These data collectively show a previously unrecognized role of recipient T cells in BMMSC-based tissue engineering.


Subject(s)
Bone Regeneration , Interferon-gamma/metabolism , Mesenchymal Stem Cells/cytology , T-Lymphocytes, Regulatory/cytology , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis , Aspirin/pharmacology , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Cell Survival , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/antagonists & inhibitors , Osteogenesis/drug effects , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , Tissue Engineering/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...