Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Stem Cell Res Ther ; 12(1): 26, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33413652

ABSTRACT

Bone marrow stromal cells (BMSC) show promise in cartilage repair, and sheep are the most common large animal pre-clinical model. OBJECTIVE: The objective of this study was to characterise ovine BMSC (oBMSC) in vitro, and to evaluate the capacity of chondrogenic micro-pellets manufactured from oBMSC or ovine articular chondrocytes (oACh) to repair osteochondral defects in sheep. DESIGN: oBMSC were characterised for surface marker expression using flow cytometry and evaluated for tri-lineage differentiation capacity. oBMSC micro-pellets were manufactured in a microwell platform, and chondrogenesis was compared at 2%, 5%, and 20% O2. The capacity of cartilage micro-pellets manufactured from oBMSC or oACh to repair osteochondral defects in adult sheep was evaluated in an 8-week pilot study. RESULTS: Expanded oBMSC were positive for CD44 and CD146 and negative for CD45. The common adipogenic induction ingredient, 3-Isobutyl-1-methylxanthine (IBMX), was toxic to oBMSC, but adipogenesis could be restored by excluding IBMX from the medium. BMSC chondrogenesis was optimal in a 2% O2 atmosphere. Micro-pellets formed from oBMSC or oACh appeared morphologically similar, but hypertrophic genes were elevated in oBMSC micro-pellets. While oACh micro-pellets formed cartilage-like repair tissue in sheep, oBMSC micro-pellets did not. CONCLUSION: The sensitivity of oBMSC, compared to human BMSC, to IBMX in standard adipogenic assays highlights species-associated differences. Micro-pellets manufactured from oACh were more effective than micro-pellets manufactured from oBMSC in the repair of osteochondral defects in sheep. While oBMSC can be driven to form cartilage-like tissue in vitro, the effective use of these cells in cartilage repair will depend on the successful mitigation of hypertrophy and tissue integration.


Subject(s)
Cartilage, Articular , Mesenchymal Stem Cells , Animals , Bone Marrow , Bone Marrow Cells , Cartilage , Cell Differentiation , Cells, Cultured , Chondrocytes , Chondrogenesis , Pilot Projects , Sheep
2.
Biomaterials ; 247: 119998, 2020 07.
Article in English | MEDLINE | ID: mdl-32251928

ABSTRACT

Many skeletal tissue regenerative strategies centre around the multifunctional properties of bone marrow derived stromal cells (BMSC) or mesenchymal stem/stromal cells (MSC)/bone marrow derived skeletal stem cells (SSC). Specific identification of these particular stem cells has been inconclusive. However, enriching these heterogeneous bone marrow cell populations with characterised skeletal progenitor markers has been a contributing factor in successful skeletal bone regeneration and repair strategies. In the current studies we have isolated, characterised and enriched ovine bone marrow mesenchymal stromal cells (oBMSCs) using a specific antibody, Stro-4, examined their multipotential differentiation capacity and, in translational studies combined Stro-4+ oBMSCs with a bovine extracellular matrix (bECM) hydrogel and a biocompatible melt electro-written medical-grade polycaprolactone scaffold, and tested their bone regenerative capacity in a small in vivo, highly vascularised, chick chorioallantoic membrane (CAM) model and a preclinical, critical-sized ovine segmental tibial defect model. Proliferation rates and CFU-F formation were similar between unselected and Stro-4+ oBMSCs. Col1A1, Col2A1, mSOX-9, PPARG gene expression were upregulated in respective osteogenic, chondrogenic and adipogenic culture conditions compared to basal conditions with no significant difference between Stro-4+ and unselected oBMSCs. In contrast, proteoglycan expression, alkaline phosphatase activity and adipogenesis were significantly upregulated in the Stro-4+ cells. Furthermore, with extended cultures, the oBMSCs had a predisposition to maintain a strong chondrogenic phenotype. In the CAM model Stro-4+ oBMSCs/bECM hydrogel was able to induce bone formation at a femur fracture site compared to bECM hydrogel and control blank defect alone. Translational studies in a critical-sized ovine tibial defect showed autograft samples contained significantly more bone, (4250.63 mm3, SD = 1485.57) than blank (1045.29 mm3, SD = 219.68) ECM-hydrogel (1152.58 mm3, SD = 191.95) and Stro-4+/ECM-hydrogel (1127.95 mm3, SD = 166.44) groups. Stro-4+ oBMSCs demonstrated a potential to aid bone repair in vitro and in a small in vivo bone defect model using select scaffolds. However, critically, translation to a large related preclinical model demonstrated the complexities of bringing small scale reported stem-cell material therapies to a clinically relevant model and thus facilitate progression to the clinic.


Subject(s)
Mesenchymal Stem Cells , Animals , Bone Marrow , Bone Marrow Cells , Cattle , Cell Differentiation , Cells, Cultured , Extracellular Matrix , Hydrogels , Osteogenesis , Polyesters , Sheep
3.
Stem Cells Int ; 2018: 2601945, 2018.
Article in English | MEDLINE | ID: mdl-29731776

ABSTRACT

Mesenchymal stromal cell-like populations have been derived from mouse-induced pluripotent stem cells (miPSC-MSC) with the capability for tissue regeneration. In this study, murine iPSC underwent differentiation towards an MSC-like immunophenotype. Stable miPSC-MSC cultures expressed the MSC-associated markers, CD73, CD105, and Sca-1, but lacked expression of the pluripotency marker, SSEA1, and hematopoietic markers, CD34 and CD45. Functionally, miPSC-MSC exhibited the potential for trilineage differentiation into osteoblasts, adipocytes, and chondrocytes and the capacity to suppress the proliferation of mitogen-activated splenocytes. The efficacy of miPSC-MSC was assessed in an acute inflammation model following systemic or local delivery into mice with subcutaneous implants containing heat-inactivated P. gingivalis. Histological analysis revealed less inflammatory cellular infiltrate within the sponges in mice treated with miPSC-MSC cells delivered locally rather than systemically. Assessment of proinflammatory cytokines in mouse spleens found that CXCL1 transcripts and protein were reduced in mice treated with miPSC-MSC. In a periodontitis model, mice subjected to oral inoculation with P. gingivalis revealed less bone tissue destruction and inflammation within the jaws when treated with miPSC-MSC compared to PBS alone. Our results demonstrated that miPSC-MSC derived from iPSC have the capacity to control acute and chronic inflammatory responses associated with the destruction of periodontal tissue. Therefore, miPSC-MSC present a promising novel source of stromal cells which could be used in the treatment of periodontal disease and other inflammatory systemic diseases such as rheumatoid arthritis.

5.
J Periodontal Res ; 51(1): 1-15, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25900048

ABSTRACT

Attainment of periodontal regeneration is a significant clinical goal in the management of advanced periodontal defects arising from periodontitis. Over the past 30 years numerous techniques and materials have been introduced and evaluated clinically and have included guided tissue regeneration, bone grafting materials, growth and other biological factors and gene therapy. With the exception of gene therapy, all have undergone evaluation in humans. All of the products have shown efficacy in promoting periodontal regeneration in animal models but the results in humans remain variable and equivocal concerning attaining complete biological regeneration of damaged periodontal structures. In the early 2000s, the concept of tissue engineering was proposed as a new paradigm for periodontal regeneration based on molecular and cell biology. At this time, tissue engineering was a new and emerging field. Now, 14 years later we revisit the concept of tissue engineering for the periodontium and assess how far we have come, where we are currently situated and what needs to be done in the future to make this concept a reality. In this review, we cover some of the precursor products, which led to our current position in periodontal tissue engineering. The basic concepts of tissue engineering with special emphasis on periodontal tissue engineering products is discussed including the use of mesenchymal stem cells in bioscaffolds and the emerging field of cell sheet technology. Finally, we look into the future to consider what CAD/CAM technology and nanotechnology will have to offer.


Subject(s)
Periodontium , Animals , Guided Tissue Regeneration, Periodontal , Humans , Periodontal Ligament , Regeneration , Tissue Engineering
6.
Adv Exp Med Biol ; 881: 207-36, 2015.
Article in English | MEDLINE | ID: mdl-26545752

ABSTRACT

The unique anatomy and composition of the periodontium make periodontal tissue healing and regeneration a complex process. Periodontal regeneration aims to recapitulate the crucial stages of wound healing associated with periodontal development in order to restore lost tissues to their original form and function and for regeneration to occur, healing events must progress in an ordered and programmed sequence both temporally and spatially, replicating key developmental events. A number of procedures have been employed to promote true and predictable regeneration of the periodontium. Principally, the approaches are based on the use of graft materials to compensate for the bone loss incurred as a result of periodontal disease, use of barrier membranes for guided tissue regeneration and use of bioactive molecules. More recently, the concept of tissue engineering has been integrated into research and applications of regenerative dentistry, including periodontics, to aim to manage damaged and lost oral tissues, through reconstruction and regeneration of the periodontium and alleviate the shortcomings of more conventional therapeutic options. The essential components for generating effective cellular based therapeutic strategies include a population of multi-potential progenitor cells, presence of signalling molecules/inductive morphogenic signals and a conductive extracellular matrix scaffold or appropriate delivery system. Mesenchymal stem cells are considered suitable candidates for cell-based tissue engineering strategies owing to their extensive expansion rate and potential to differentiate into cells of multiple organs and systems. Mesenchymal stem cells derived from multiple tissue sources have been investigated in pre-clinical animal studies and clinical settings for the treatment and regeneration of the periodontium.


Subject(s)
Dental Cementum/physiopathology , Periodontal Ligament/physiopathology , Regeneration , Tissue Engineering/methods , Wound Healing , Biocompatible Materials/metabolism , Dental Cementum/injuries , Guided Tissue Regeneration, Periodontal/methods , Humans , Mesenchymal Stem Cells/cytology , Periodontal Diseases/physiopathology , Periodontal Diseases/surgery , Periodontal Diseases/therapy , Periodontal Ligament/injuries , Periodontium/injuries , Periodontium/physiopathology , Regenerative Medicine/methods , Regenerative Medicine/trends
7.
J Dent Res ; 94(11): 1508-15, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26285811

ABSTRACT

Induced pluripotent stem cells (iPSCs) are the newest member of a growing list of stem cell populations that hold great potential for use in cell-based treatment approaches in the dental field. This review summarizes the dental tissues that have successfully been utilized to generate iPSC lines, as well as the potential uses of iPSCs for tissue regeneration in different dental applications. While iPSCs display great promise in a number of dental applications, there are safety concerns with these cells that need to be addressed before they can be used in clinical settings. This review outlines some of the apprehensions to the use of iPSCs clinically, and it details approaches that are being employed to ensure the safety and efficacy of these cells. One of the major approaches being investigated is the differentiation of iPSCs prior to use in patients. iPSCs have successfully been differentiated into a wide range of cells and tissue types. This review focuses on 2 differentiation approaches-the differentiation of iPSCs into mesenchymal stem cells and the differentiation of iPSCs into osteoprogenitor cells. Both these resulting populations of cells are particularly relevant to the dental field.


Subject(s)
Dentistry/methods , Induced Pluripotent Stem Cells/physiology , Cell Differentiation , Gingiva/cytology , Guided Tissue Regeneration/methods , Guided Tissue Regeneration, Periodontal/methods , Humans , Induced Pluripotent Stem Cells/transplantation , Multipotent Stem Cells/physiology , Multipotent Stem Cells/transplantation , Periodontium/cytology , Stem Cells/physiology , Tooth/cytology
8.
J Periodontal Res ; 50(2): 160-72, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24965968

ABSTRACT

BACKGROUND AND OBJECTIVE: Implantation of periodontal ligament stem cells is emerging as a potential periodontal regenerative procedure. This systematic review considers the evidence from animal models investigating the use of periodontal ligament stem cells for successful periodontal regeneration. MATERIAL AND METHODS: PubMed, Embase, MEDLINE and Google Scholar were searched to December 2013 for quantitative studies examining the outcome of implanting periodontal ligament stem cells into experimental periodontal defects in animals. Inclusion criteria were: implantation of periodontal ligament stem cells into surgically created periodontal defects for periodontal regeneration; animal models only; source of cells either human or animal; and published in English. We followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. RESULTS: From the literature search, 43 studies met the inclusion criteria. A wide variety of surgical defects were created in four species of animal (dog, rat, pig and sheep). Owing to wide variability in defect type, cell source and cell scaffold, no meta-analysis was possible. Outcome measures included new bone, new cementum and new connective tissue formation. In 70.5% of the results, statistically significant improvements of these measures was recorded. CONCLUSION: These results are notable in that they indicate that irrespective of the defect type and animal model used, periodontal ligament stem cell implantation can be expected to result in a beneficial outcome for periodontal regeneration. It is recommended that there is sufficient evidence from preclinical animal studies to warrant moving to human studies to examine the efficacy, safety, feasibility (autologous vs. allogeneic transplantation) and delivery of periodontal ligament stem cells for periodontal regeneration.


Subject(s)
Disease Models, Animal , Guided Tissue Regeneration, Periodontal/methods , Periodontal Ligament/cytology , Stem Cells/physiology , Animals , Cementogenesis/physiology , Humans , Osteogenesis/physiology , Periodontal Diseases/therapy , Regeneration/physiology
9.
J Dent Res ; 93(12): 1212-21, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25139362

ABSTRACT

For a successful clinical outcome, periodontal regeneration requires the coordinated response of multiple soft and hard tissues (periodontal ligament, gingiva, cementum, and bone) during the wound-healing process. Tissue-engineered constructs for regeneration of the periodontium must be of a complex 3-dimensional shape and adequate size and demonstrate biomechanical stability over time. A critical requirement is the ability to promote the formation of functional periodontal attachment between regenerated alveolar bone, and newly formed cementum on the root surface. This review outlines the current advances in multiphasic scaffold fabrication and how these scaffolds can be combined with cell- and growth factor-based approaches to form tissue-engineered constructs capable of recapitulating the complex temporal and spatial wound-healing events that will lead to predictable periodontal regeneration. This can be achieved through a variety of approaches, with promising strategies characterized by the use of scaffolds that can deliver and stabilize cells capable of cementogenesis onto the root surface, provide biomechanical cues that encourage perpendicular alignment of periodontal fibers to the root surface, and provide osteogenic cues and appropriate space to facilitate bone regeneration. Progress on the development of multiphasic constructs for periodontal tissue engineering is in the early stages of development, and these constructs need to be tested in large animal models and, ultimately, human clinical trials.


Subject(s)
Guided Tissue Regeneration, Periodontal/methods , Tissue Engineering/methods , Tissue Scaffolds/classification , Animals , Biocompatible Materials/therapeutic use , Biomechanical Phenomena , Biomimetic Materials/therapeutic use , Bone Regeneration/physiology , Guided Tissue Regeneration, Periodontal/instrumentation , Humans , Prosthesis Design , Tissue Engineering/instrumentation
10.
Aust Dent J ; 59 Suppl 1: 117-30, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24111843

ABSTRACT

The aim of this review is to discuss the clinical utility of stem cells in periodontal regeneration by reviewing relevant literature that assesses the periodontal-regenerative potential of stem cells. We consider and describe the main stem cell populations that have been utilized with regard to periodontal regeneration, including bone marrow-derived mesenchymal stem cells and the main dental-derived mesenchymal stem cell populations: periodontal ligament stem cells, dental pulp stem cells, stem cells from human exfoliated deciduous teeth, stem cells from apical papilla and dental follicle precursor cells. Research into the use of stem cells for tissue regeneration has the potential to significantly influence periodontal treatment strategies in the future.


Subject(s)
Periodontium/physiology , Regeneration/physiology , Stem Cells/physiology , Tissue Engineering , Bone Transplantation , Dental Cementum/cytology , Dental Pulp/cytology , Dental Sac/cytology , Gingiva/cytology , Guided Tissue Regeneration, Periodontal , Humans , Intercellular Signaling Peptides and Proteins/therapeutic use , Mesenchymal Stem Cells/cytology , Periodontal Ligament/cytology , Periodontal Ligament/physiology , Periodontium/embryology , Periodontium/injuries , Stem Cells/cytology , Tissue Scaffolds , Tooth, Deciduous/cytology , Wound Healing
11.
J Periodontal Res ; 49(3): 333-45, 2014 Jun.
Article in English | MEDLINE | ID: mdl-23841948

ABSTRACT

BACKGROUND AND OBJECTIVE: The complex microenvironment of the periodontal wound creates many challenges associated with multitissue regeneration of periodontal lesions. Recent characterization of mesenchymal stem cell-like populations residing in periodontal ligament tissues has shown that these cells exhibit features of postnatal stem cells. Despite these advances, a lack of consistency in design of preclinical studies and a limited study of allogeneic transplantation applications has restricted our understanding of their clinical utility in the treatment of periodontal disease. The aim of this study was to assess the regenerative potential of allogeneic periodontal ligament stem cells (PDLSCs) in a rat periodontal fenestration defect mode and to identify an optimal end time-point suitable for quantitative assessment of tissue regeneration. MATERIAL AND METHODS: Periodontal fenestration defects, created in Sprague Dawley rats, were treated with allogeneic PDLSCs seeded onto Gelfoam(®) (Absorbable gelatin sponge; Pharmacia Corporation, Kalamazoo, MI, USA) or with Gelfoam(®) alone, or remained untreated. Experimental rats were killed at 7, 14, 21 or 28 d after surgery and the tissues were processed for immunohistochemical and histomorphometric examination. RESULTS: Defects treated with PDLSCs showed significantly greater percentage bone fill and length of new bone bridge compared with the untreated group or the group treated with Gelfoam(®) alone on days 14 and 21. Similarly, a statistically significant difference was achieved within specimens retrieved on day 21 for analysis of regeneration of cementum/periodontal ligament (PDL)-like structures. CONCLUSION: The present investigation shows that allogeneic PDLSCs have a marked ability to repair periodontal defects by forming bone, PDL and cementum-like tissue in vivo. The results suggest that treatment periods of 14 and 21 d are optimal end time-points for quantitative assessment of periodontal regeneration within the rodent fenestration-defect model utilized in the present study.


Subject(s)
Allografts/transplantation , Alveolar Bone Loss/therapy , Periodontal Ligament/cytology , Regeneration/physiology , Stem Cell Transplantation/methods , Alveolar Process/pathology , Animals , Bone Regeneration/physiology , Cell Differentiation/physiology , Cell Separation/methods , Cementogenesis/physiology , Collagen/ultrastructure , Connective Tissue/pathology , Disease Models, Animal , Female , Flow Cytometry , Gelatin Sponge, Absorbable/chemistry , Guided Tissue Regeneration, Periodontal/methods , Osteogenesis/physiology , Periodontal Ligament/pathology , Rats , Rats, Sprague-Dawley , Tissue Engineering/methods , Tissue Scaffolds/chemistry
12.
Biomaterials ; 35(4): 1150-62, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24215733

ABSTRACT

Previous reports in the literature investigating chondrogenesis in mesenchymal progenitor cell (MPC) cultures have confirmed the chondro-inductive potential of pentosan polysulphate (PPS), a highly sulphated semi-synthetic polysaccharide, when added as a soluble component to culture media under standard aggregate-assay conditions or to poly(ethylene glycol)/hyaluronic acid (PEG/HA)-based hydrogels, even in the absence of inductive factors (e.g. TGFß). In this present study, we aimed to assess whether a 'bound' PPS would have greater activity and availability over a soluble PPS, as a media additive or when incorporated into PEG/HA-based hydrogels. We achieved this by covalently pre-binding the PPS to the HA component of the gel (forming a new molecule, HA-PPS). We firstly investigated the activity of HA-PPS compared to free PPS, when added as a soluble factor to culture media. Cell proliferation, as determined by CCK8 and EdU assay, was decreased in the presence of either bound or free PPS whilst chondrogenic differentiation, as determined by DMMB assay and histology, was enhanced. In all cases, the effect of the bound PPS (HA-PPS) was more potent than that of the unbound form. These results alone suggest wider applications for this new molecule, either as a culture supplement or as a coating for scaffolds targeted at chondrogenic differentiation or maturation. We then investigated the incorporation of HA-PPS into a PEG/HA-based hydrogel system, by simply substituting some of the HA for HA-PPS. Rheological testing confirmed that incorporation of either HA-PPS or PPS did not significantly affect gelation kinetics, final hydrogel modulus or degradation rate but had a small, but significant, effect on swelling. When encapsulated in the hydrogels, MPCs retained good viability and rapidly adopted a rounded morphology. Histological analysis of both GAG and collagen deposition after 21 days showed that the incorporation of the bound-PPS into the hydrogel resulted in increased matrix formation when compared to the addition of soluble PPS to the hydrogel, or the hydrogel alone. We believe that this new generation injectable, degradable hydrogel, incorporating now a covalently bound-PPS, when combined with MPCs, has the potential to assist cartilage regeneration in a multitude of therapeutic targets, including for intervertebral disc (IVD) degeneration.


Subject(s)
Biocompatible Materials/metabolism , Hydrogels/chemistry , Intervertebral Disc/physiology , Pentosan Sulfuric Polyester/metabolism , Polyethylene Glycols/chemistry , Regeneration , Biocompatible Materials/chemistry , Cell Differentiation , Cell Line , Cell Proliferation , Cell Survival , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , Hydrogels/metabolism , Mesenchymal Stem Cells/cytology , Pentosan Sulfuric Polyester/chemistry , Polyethylene Glycols/metabolism , Solubility , Tissue Engineering
13.
Lab Anim ; 47(4): 291-300, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23836849

ABSTRACT

In vivo assessment of ventricular function in rodents has largely been restricted to transthoracic echocardiography (TTE). However 1.5 T cardiac magnetic resonance (CMR) and transoesophageal echocardiography (TOE) have emerged as possible alternatives. Yet, to date, no study has systematically assessed these three imaging modalities in determining ejection fraction (EF) in rats. Twenty rats underwent imaging four weeks after surgically-induced myocardial infarction. CMR was performed on a 1.5 T scanner, TTE was conducted using a 9.2 MHz transducer and TOE was performed with a 10 MHz intracardiac echo catheter. Correlation between the three techniques for EF determination and analysis reproducibility was assessed. Moderate-strong correlation was observed between the three modalities; the greatest between CMR and TOE (intraclass correlation coefficient (ICC) = 0.89), followed by TOE and TTE (ICC = 0.70) and CMR and TTE (ICC = 0.63). Intra- and inter-observer variations were excellent with CMR (ICC = 0.99 and 0.98 respectively), very good with TTE (0.90 and 0.89) and TOE (0.87 and 0.84). Each modality is a viable option for evaluating ventricular function in rats, however the high image quality and excellent reproducibility of CMR offers distinct advantages even at 1.5 T with conventional coils and software.


Subject(s)
Echocardiography, Transesophageal/veterinary , Echocardiography/veterinary , Heart Ventricles/pathology , Magnetic Resonance Imaging/veterinary , Ventricular Function , Animals , Heart Ventricles/diagnostic imaging , Male , Rats , Rats, Sprague-Dawley , Reproducibility of Results
14.
J Dent Res ; 92(9): 833-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23884555

ABSTRACT

Mesenchymal stem cells (MSC) have been considered as a potential therapy for the treatment of periodontal defects arising from periodontitis. However, issues surrounding their accessibility and proliferation in culture significantly limit their ability to be used as a mainstream treatment approach. It is therefore important that alternative, easily accessible, and safe populations of stem cells be identified. Controlled induction of induced pluripotent stem cells (iPSC) into MSC-like cells is emerging as an attractive source for obtaining large populations of stem cells for regenerative medicine. We have successfully induced iPSC to differentiate into MSC-like cells. The MSC-like cells generated satisfied the International Society of Cellular Therapy's minimal criteria for defining multipotent MSC, since they had plastic adherent properties, expressed key MSC-associated markers, and had the capacity to undergo tri-lineage differentiation. Importantly, the resulting iPSC-MSC-like cells also had the capacity, when implanted into periodontal defects, to significantly increase the amount of regeneration and newly formed mineralized tissue present. Our results demonstrate, for the first time, that MSC derived from iPSC have the capacity to aid periodontal regeneration and are a promising source of readily accessible stem cells for use in the clinical treatment of periodontitis.


Subject(s)
Mesenchymal Stem Cells/physiology , Periodontal Diseases/therapy , Pluripotent Stem Cells/physiology , Alveolar Bone Loss/surgery , Animals , Antimetabolites , Bone Regeneration/physiology , Bromodeoxyuridine , Calcification, Physiologic/physiology , Cell Culture Techniques , Cell Differentiation/physiology , Cell Lineage/physiology , Disease Models, Animal , Flow Cytometry , Humans , Image Processing, Computer-Assisted/methods , Mesenchymal Stem Cell Transplantation/methods , Osteoblasts/pathology , Osteocytes/pathology , Osteogenesis/physiology , Periodontium/pathology , Periodontium/physiopathology , Rats , Rats, Nude , Regeneration/physiology
15.
Reprod Sci ; 19(12): 1302-14, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22886285

ABSTRACT

Mesenchymal stromal cells (MSCs) from gestational tissues represent promising cell populations with stem cell-like properties for use in regenerative medicine. Previously, we reported that MSCs in the chorionic villi of the human placenta reside in a vascular niche. However, the niche(s) in which MSCs reside in the fetal membranes, another rich source of MSCs, remains to be determined. The cell surface markers STRO-1 and 3G5 were previously employed to identify niches in a variety of tissues and here we use these markers to report the location of the MSC niche in the human decidua parietalis. The cultured decidua parietalis MSCs (DPMSCs) isolated from the choriodecidua component of the fetal membranes possessed stem cell-like properties such as adherence to plastic, colony forming ability, and multipotent differentiation potential. Fluorescence in situ hybridization analysis showed cultured DPMSCs were of maternal origin. Immunocytochemistry demonstrated that cultured DPMSCs stained positively with stem cell surface markers 3G5, CD105, CD106, STRO-1, CD146, CD49a, and α-SMA but were negative for hematopoietic markers (CD117, CD34) and vascular markers (CD34, von Willebrand factor [vWF]). Immunohistochemistry with antibodies to stem cell surface markers and the endothelial markers on term fetal membranes revealed a vascular niche for DPMSCs, which was confirmed by immunofluorescence analysis. Both STRO-1 and vWF fluorescence signals showed substantial overlap, while CD146 and vWF signals showed partial overlap. These observations were consistent with a vascular niche.


Subject(s)
Chorionic Villi/blood supply , Decidua/blood supply , Decidua/cytology , Mesenchymal Stem Cells/cytology , Stem Cell Niche/physiology , Antigens, Surface/analysis , Biomarkers/analysis , Cell Differentiation , Cells, Cultured , Female , Flow Cytometry , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Mesenchymal Stem Cells/chemistry , Pregnancy
16.
Leukemia ; 25(10): 1533-42, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21637285

ABSTRACT

Hypoxia is an imbalance between oxygen supply and demand, which deprives cells or tissues of sufficient oxygen. It is well-established that hypoxia triggers adaptive responses, which contribute to short- and long-term pathologies such as inflammation, cardiovascular disease and cancer. Induced by both microenvironmental hypoxia and genetic mutations, the elevated expression of the hypoxia-inducible transcription factor-1 (HIF-1) and HIF-2 is a key feature of many human cancers and has been shown to promote cellular processes, which facilitate tumor progression. In this review, we discuss the emerging role of hypoxia and the HIFs in the pathogenesis of multiple myeloma (MM), an incurable hematological malignancy of BM PCs, which reside within the hypoxic BM microenvironment. The need for current and future therapeutic interventions to target HIF-1 and HIF-2 in myeloma will also be discussed.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Multiple Myeloma/physiopathology , Adaptation, Physiological , Bone Marrow Cells/physiology , Humans , Multiple Myeloma/therapy
17.
J Periodontal Res ; 46(4): 438-47, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21443752

ABSTRACT

BACKGROUND AND OBJECTIVE: Human induced pluripotent stem (iPS) cells, which have similar properties to human embryonic stem (hES) cells, have been generated from neonatal and adult human dermal fibroblasts by reprogramming. iPS cells have high pluripotency and differentiation potential, and may be a potential autologous stem cell source for future regenerative therapy. MATERIAL AND METHODS: iPS cell lines from human gingival fibroblasts and, for the first time, from periodontal ligament fibroblasts, were generated by reprogramming using a retroviral transduction cocktail of OCT3/4, SOX2, KLF4 and c-MYC. iPS induction was investigated through expression of the embryonic stem cell markers SSEA4, OCT4, NANOG, GCTM-2, TG30 and TRA-1-60. Following in vitro differentiation, the expression of genes for differentiation markers for ectoderm (SOX1, PAX6), mesoderm [RUNX1, T(Brachyury)] and endoderm (GATA4, AFP) was assessed by real-time RT-PCR. The ability to form teratomas following implantation into mouse testes was assessed by histology. RESULTS: Human gingival fibroblast- and periodontal ligament fibroblast-derived iPS cells showed similar characteristics to hES cells. Both sets of iPS cells displayed colony morphology comparable to that of hES cells and expressed the hES cell-associated cell-surface antigens, SSEA3, SSEA4, GCTM-2, TG30 (CD9) and Tra-1-60, and the hES cell marker genes, OCT4, NANOG and GDF3. These iPS cells showed differentiation potential to form embryoid bodies in vitro and expressed genes for endoderm, ectoderm and mesoderm. Teratoma formation following implantation into mouse testes was observed. CONCLUSION: These results demonstrate that iPS cells can be successfully generated from adult human gingival and periodontal ligament fibroblasts.


Subject(s)
Fibroblasts/physiology , Gingiva/cytology , Induced Pluripotent Stem Cells/physiology , Periodontal Ligament/cytology , Animals , Antigens, Surface/analysis , Cell Differentiation/physiology , Cell Line , Core Binding Factor Alpha 2 Subunit/analysis , Eye Proteins/analysis , Fetal Proteins/analysis , GATA4 Transcription Factor/analysis , Genes, myc/genetics , Glycosphingolipids/analysis , Growth Differentiation Factor 3/analysis , Homeodomain Proteins/analysis , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, SCID , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , PAX6 Transcription Factor , Paired Box Transcription Factors/analysis , Proteoglycans/analysis , Repressor Proteins/analysis , SOXB1 Transcription Factors/analysis , SOXB1 Transcription Factors/genetics , Stage-Specific Embryonic Antigens/analysis , T-Box Domain Proteins/analysis , Teratoma/pathology , Testicular Neoplasms/pathology , Transduction, Genetic , alpha-Fetoproteins/analysis
18.
Vet Comp Orthop Traumatol ; 24(2): 113-21, 2011.
Article in English | MEDLINE | ID: mdl-21225086

ABSTRACT

INTRODUCTION: Synthetic void-fillers offer an alternative to autograft or allograft bone in the repair of segmental defects. However, the reparative process is delayed as only osteoconductive elements are present. The inclusion of pluripotential cells may resolve this limitation, and the use of allogeneic tissue provides the opportunity for an off-the-shelf remedy. The current study evaluated the utilisation of mesenchymal precursor cells (MPC) for the repair of an ovine critical-size tibial segmental defect. METHODS: Twenty-four, mature female sheep underwent surgery for the creation of a 3 cm tibial diaphyseal defect. In one group of 12 sheep the scaffold was used alone, and in the second group the scaffold was seeded with MPC. The defect was stabilised using a locking intramedullary nail and allowed to heal over a nine-month-period. Outcome assessments of healing included radiology of callus formation, computed tomography, assessment of new-bone volume, mechanical attributes, and histological evaluation of linear bone apposition rate and tissue response. RESULTS: The MPC-treated group displayed a significantly greater level of callus formation and rate of bone apposition in the defect. DISCUSSION: The incorporation of allogeneic MPC to a synthetic void filler stimulated early repair of critical-size diaphyseal segmental defects and holds potential as an off-the-shelf therapy for augmenting bone regeneration.


Subject(s)
Diaphyses/surgery , Orthopedic Procedures/methods , Sheep, Domestic/surgery , Surgery, Veterinary/methods , Tibial Fractures/surgery , Transplantation, Homologous/veterinary , Animals , Biocompatible Materials , Bony Callus/diagnostic imaging , Bony Callus/growth & development , Diaphyses/diagnostic imaging , Diaphyses/pathology , Female , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cell Transplantation/veterinary , Radiography , Tibial Fractures/diagnostic imaging , Tibial Fractures/pathology , Tissue Engineering/methods , Transplantation, Homologous/methods
19.
J Cell Biochem ; 110(6): 1288-98, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20564230

ABSTRACT

Bone marrow-derived mesenchymal stem cells (MSC), are multipotent cells that give rise to multiple lineages including osteoblasts, adipocytes, muscle, and fibroblasts. MSCs are useful for clinical applications such as cell therapy because they can be isolated from an individual and expanded for use in tissue repair, as well as other therapeutic applications, without immune rejection. However, one of the key problems in the use of MSCs for these applications is the efficiency of these cells to engraft and fully regenerate damaged tissues. Therefore, to optimize this process, a comprehensive understanding of the key regulators of MSCs self-renewal and maintenance are critical to the success of future cell therapy as well as other clinical applications. The basic helix loop helix transcription factor, Twist, plays a master regulatory role in all of these processes and, therefore, a thorough understanding of the mechanistic insights in the role of Twist in lineage specification/differentiation and tumorigenesis is vital to the success of future clinical applications for the therapeutic use of MSCs. In this article, we highlight the basic mechanisms and signaling pathways that are important to MSC fate, maintenance, and differentiation, as well as the critical role that Twist plays in these processes. In addition, we review the known literature suggesting a critical role for Twist in the generation of cancer stem cells, as this information may contribute to a broader understanding of stem cell biology and stem-cell-based therapeutics.


Subject(s)
Cell Differentiation/physiology , Mesenchymal Stem Cells/physiology , Multipotent Stem Cells/physiology , Twist-Related Protein 1/physiology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Cell Lineage , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Models, Biological , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Twist-Related Protein 1/metabolism
20.
J Cell Physiol ; 223(2): 530-40, 2010 May.
Article in English | MEDLINE | ID: mdl-20162565

ABSTRACT

The cardiovascular therapeutic potential of bone marrow mesenchymal stromal/stem cells (MSC) is largely mediated by paracrine effects. Traditional preparation of MSC has involved plastic adherence-isolation. In contrast, prospective immunoselection aims to improve cell isolation by enriching for mesenchymal precursor cells (MPC) at higher purity. This study compared the biological characteristics and cardiovascular trophic activity of plastic adherence-isolated MSC (PA-MSC) and MPC prepared from the same human donors by immunoselection for stromal precursor antigen-1 (STRO-1). Compared to PA-MSC, STRO-1-MPC displayed greater (1) clonogenicity, (2) proliferative capacity, (3) multilineage differentiation potential, and (4) mRNA expression of mesenchymal stem cell-related transcripts. In vitro assays demonstrated that conditioned medium from STRO-1-MPC had greater paracrine activity than PA-MSC, with respect to cardiac cell proliferation and migration and endothelial cell migration and tube formation. In keeping with this, STRO-1-MPC exhibited higher gene and protein expression of CXCL12 and HGF. Inhibition of these cytokines attenuated endothelial tube formation and cardiac cell proliferation, respectively. Paracrine responses were enhanced by using supernatant from STRO-1(Bright) MPC and diminished with STRO-1(Dim) conditioned medium. Together, these findings indicate that prospective isolation gives rise to mesenchymal progeny that maintain a higher proportion of immature precursor cells compared to traditional plastic adherence-isolation. Enrichment for STRO-1 is also accompanied by increased expression of cardiovascular-relevant cytokines and enhanced trophic activity. Immunoselection thus provides a strategy for improving the cardiovascular reparative potential of mesenchymal cells.


Subject(s)
Antigens, Surface/metabolism , Bone Marrow Cells/metabolism , Cardiovascular Diseases/therapy , Cell Differentiation/physiology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Paracrine Communication/physiology , Antigens, Surface/genetics , Antigens, Surface/immunology , Biomarkers/analysis , Biomarkers/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cell Adhesion/immunology , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Movement/drug effects , Cell Movement/physiology , Cell Proliferation/drug effects , Cells, Cultured , Colony-Forming Units Assay , Culture Media, Conditioned/pharmacology , Endothelial Cells/drug effects , Humans , Immunomagnetic Separation/methods , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Multipotent Stem Cells/cytology , Multipotent Stem Cells/immunology , Multipotent Stem Cells/metabolism , Myocytes, Cardiac/drug effects , Paracrine Communication/drug effects , Plastics/chemistry , RNA, Messenger/metabolism , Regeneration/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...