Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
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
2.
Bone ; 95: 162-174, 2017 02.
Article in English | MEDLINE | ID: mdl-27913271

ABSTRACT

Histone deacetylases (HDACs)2 play important roles in the epigenetic regulation of gene expression in cells and are emerging therapeutic targets for treating a wide range of diseases. HDAC inhibitors (HDACi)3 that act on multiple HDAC enzymes have been used clinically to treat a number of solid and hematological malignancies. HDACi are also currently being studied for their efficacy in non-malignant diseases, including pathologic bone loss, but this has necessitated a better understanding of the roles of individual HDAC enzymes, particularly the eleven zinc-containing isozymes. Selective isozyme-specific inhibitors currently being developed against class I HDACs (1, 2, 3 and 8) and class II HDACs (4, 5, 6, 7, 9 and 10) will be valuable tools for elucidating the roles played by individual HDACs in different physiological and pathological settings. Isozyme-specific HDACi promise to have greater efficacy and reduced side effects, as required for treating chronic disease over extended periods of time. This article reviews the current understanding of roles for individual HDAC isozymes and effects of HDACi on bone cells, (osteoblasts, osteoclasts and osteocytes), in relation to bone remodelling in conditions characterised by pathological bone loss, including periodontitis, rheumatoid arthritis and myeloma bone disease.


Subject(s)
Bone Remodeling/physiology , Histone Deacetylases/metabolism , Animals , Cell Differentiation/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism
3.
Oncogene ; 35(40): 5282-5294, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27041566

ABSTRACT

Osteosarcoma (OS) is the most common cancer of bone and the 5th leading cause of cancer-related death in young adults. Currently, 5-year survival rates have plateaued at ~70% for patients with localized disease. Those with disseminated disease have an ~20% 5-year survival. An improved understanding of the molecular genetics of OS may yield new approaches to improve outcomes for OS patients. To this end, we applied murine models that replicate human OS to identify and understand dysregulated microRNAs (miRNAs) in OS. miRNA expression patterns were profiled in murine primary osteoblasts, osteoblast cultures and primary OS cell cultures (from primary and paired metastatic locations) isolated from two genetically engineered murine models of OS. The differentially expressed miRNA were further assessed by a cross-species comparison with human osteoblasts and OS cultures. We identified miR-155-5p and miR-148a-3p as deregulated in OS. miR-155-5p suppression or miR-148a-3p overexpression potently reduced proliferation and induced apoptosis in OS cells, yet strikingly, did not impact normal osteoblasts. To define how these miRNAs regulated OS cell fate, we used an integrated computational approach to identify putative candidate targets and then correlated these with the cell biological impact. Although we could not resolve the mechanism through which miR-148a-3p impacts OS, we identified that miR-155-5p overexpression suppressed its target Ripk1 (receptor (TNFRSF)-interacting serine-threonine kinase 1) expression, and miR-155-5p inhibition elevated Ripk1 levels. Ripk1 is directly involved in apoptosis/necroptosis. In OS cells, small interfering RNA against Ripk1 prevented cell death induced by the sequestration of miR-155-5p. Collectively, we show that miR-148a-3p and miR-155-5p are species-conserved deregulated miRNA in OS. Modulation of these miRNAs was specifically toxic to tumor cells but not normal osteoblasts, raising the possibility that these may be tractable targets for miRNA-based therapies for OS.


Subject(s)
MicroRNAs/biosynthesis , Osteosarcoma/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Mice , MicroRNAs/genetics , Osteosarcoma/pathology
4.
Leukemia ; 29(10): 2075-85, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25921247

ABSTRACT

Hypoxia-inducible factor (HIF)-1α accumulation promotes hematopoietic stem cells' quiescence and is necessary to maintain their self-renewal. However, the role of HIF-2α in hematopoietic cells is less clear. We investigated the role of HIF-2α in leukemia and lymphoma cells. HIF-2α expression was high in subsets of human and mouse leukemia and lymphoma cells, whereas it was low in normal bone marrow leukocytes. To investigate the role of HIF-2α, we transduced human HIF-2α cDNA in mouse syngeneic models of myeloid preleukemia and a transgenic model of B lymphoma. Ectopic expression of HIF-2α accelerated leukemia cell proliferation in vitro. Mice transplanted with cells transduced with HIF-2α died significantly faster of leukemia or B lymphoma than control mice transplanted with empty vector-transduced cells. Conversely, HIF-2α knockdown in human myeloid leukemia HL60 cells decreased proliferation in vitro and significantly prolonged animal survival following transplantation. In human acute myeloid leukemia (AML), HIF-2α mRNA was significantly elevated in several subsets such as the t(15;17), inv(16), complex karyotype and favorable cytogenetic groups. However, patients with high HIF-2α expression had a trend to higher disease-free survival in univariate analysis. The different effects of HIF-2α overexpression in mouse models of leukemia and human AML illustrates the complexity of this mutliclonal disease.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Disease Models, Animal , Hematopoietic Stem Cells/pathology , Leukemia, Myeloid, Acute/pathology , Lymphoma/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Blotting, Western , Cell Hypoxia , Cells, Cultured , Cohort Studies , Disease Progression , Female , Follow-Up Studies , Hematopoietic Stem Cells/metabolism , Humans , Immunoenzyme Techniques , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Lymphoma/genetics , Lymphoma/mortality , Male , Mice , Mice, Transgenic , Middle Aged , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Young Adult
5.
Intern Med J ; 45(3): 335-43, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25735577

ABSTRACT

Options for treatment of elderly patients with multiple myeloma have expanded substantially following the development of immunomodulatory drugs (IMiD), proteasome inhibitors and with enhancement in safety of high-dose therapy and autologous stem cell transplant (HDT + ASCT). The recognition of biological heterogeneity among elderly patients has made delivery of therapy more challenging. An individualised approach to treatment selection is recommended in an era in which highly efficacious treatment options are available for transplant-ineligible patients. Here, we summarise recommendations for patients who are considered unsuitable for HDT + ASCT, including pretreatment considerations, and induction, maintenance and supportive care therapies.


Subject(s)
Advisory Committees/standards , Foundations/standards , Multiple Myeloma/epidemiology , Multiple Myeloma/therapy , Stem Cell Transplantation , Australia/epidemiology , Humans , Immunologic Factors/therapeutic use , Multiple Myeloma/diagnosis , Proteasome Inhibitors/therapeutic use , Transplantation, Autologous , Treatment Outcome
6.
Intern Med J ; 45(1): 94-105, 2015 01.
Article in English | MEDLINE | ID: mdl-25582938

ABSTRACT

The survival of patients with multiple myeloma (MM) has improved substantially since the introduction in the late 1980s of high-dose chemotherapy (HDT) supported by autologous stem cell transplantation (ASCT). Further improvements have been observed following the availability of immunomodulatory drugs (IMiD) such as thalidomide and lenalidomide, and the proteasome inhibitor, bortezomib. Here, we summarise the recommendations of the Medical Scientific Advisory Group to the Myeloma Foundation of Australia for patients considered suitable for HDT + ASCT as part of initial therapy. These recommendations incorporate the various phases of treatment: induction, HDT conditioning and maintenance therapy.


Subject(s)
Hematopoietic Stem Cell Transplantation/standards , Multiple Myeloma/drug therapy , Practice Guidelines as Topic , Societies, Scientific , Advisory Committees , Australia/epidemiology , Disease-Free Survival , Humans , Multiple Myeloma/epidemiology , Survival Rate/trends , Transplantation, Autologous , Treatment Outcome
7.
Intern Med J ; 45(4): 371-82, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25169210

ABSTRACT

Systemic AL amyloidosis is a plasma cell dyscrasia with a characteristic clinical phenotype caused by multi-organ deposition of an amyloidogenic monoclonal protein. This condition poses a unique management challenge due to the complexity of the clinical presentation and the narrow therapeutic window of available therapies. Improved appreciation of the need for risk stratification, standardised use of sensitive laboratory testing for monitoring disease response, vigilant supportive care and the availability of newer agents with more favourable toxicity profiles have contributed to the improvement in treatment-related mortality and overall survival seen over the past decade. Nonetheless, with respect to the optimal management approach, there is a paucity of high-level clinical evidence due to the rarity of the disease, and enrollment in clinical trials is still the preferred approach where available. This review will summarise the Clinical Practice Guidelines on the Management of Systemic Light Chain (AL) Amyloidosis recently prepared by the Medical Scientific Advisory Group of the Myeloma Foundation of Australia. It is hoped that these guidelines will assist clinicians in better understanding and optimising the management of this difficult disease.


Subject(s)
Advisory Committees/standards , Amyloidosis/therapy , Disease Management , Foundations/standards , Multiple Myeloma/therapy , Amyloidosis/diagnosis , Amyloidosis/epidemiology , Australia/epidemiology , Humans , Multiple Myeloma/diagnosis , Multiple Myeloma/epidemiology
8.
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
9.
Blood Cancer J ; 2: e91, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22983434

ABSTRACT

In the adult mammal, normal haematopoiesis occurs predominantly in the bone marrow, where primitive haematopoietic stem cells (HSC) and their progeny reside in specialised microenvironments. The bone marrow microenvironment contains specific anatomical areas (termed niches) that are highly specialised for the development of certain blood cell types, for example HSCs. The HSC niche provides important cell-cell interactions and signalling molecules that regulate HSC self-renewal and differentiation processes. These same signals and interactions are also important in the progression of haematological malignancies, such as multiple myeloma (MM). This review provides an overview of the bone marrow microenvironment and its involvement in normal, physiological HSC maintenance and plasma cell growth throughout MM disease progression.

10.
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
11.
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
12.
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
14.
Leukemia ; 24(4): 765-70, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20147974

ABSTRACT

Active influx of imatinib in chronic myeloid leukemia (CML) cells is mediated by the organic cation transporter 1 (OCT-1). Functional activity of OCT-1 (OCT-1 Activity) in mononuclear cells is an excellent predictor of molecular response over the first 24 months of imatinib therapy for chronic phase patients. CML progenitor cells are less sensitive to imatinib-induced apoptosis and are likely contributors to disease persistence. We investigated whether alterations in the expression and function of OCT-1 have a role in imatinib resistance in progenitors. We found the intracellular uptake and retention (IUR) of imatinib, OCT-1 Activity and OCT-1 mRNA expression are all significantly lower in CML CD34+ cells compared with mature CD34- cells (P<0.001). However, no differences in IUR or OCT-1 Activity were observed between these subsets in healthy donors. In contrast to OCT-1, ABCB1 and ABCG2 seemed to have no functional role in the transport of imatinib in CML CD34+ cells. Consistent with the observation that nilotinib uptake is not OCT-1 dependent, the IUR of nilotinib did not differ between CML CD34+ and CD34- cells. These results indicate that low imatinib accumulation in primitive CML cells, mediated through reduced OCT-1 Activity may be a critical determinant of long-term disease persistence.


Subject(s)
Antigens, CD34/metabolism , Bone Marrow Cells/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Organic Cation Transporter 1/metabolism , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adrenergic alpha-Antagonists/pharmacology , Benzamides , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Flow Cytometry , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Organic Cation Transporter 1/antagonists & inhibitors , Organic Cation Transporter 1/genetics , Prazosin/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
15.
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
16.
Placenta ; 31(3): 203-12, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20060164

ABSTRACT

The chorionic villi of human term placentae are a rich source of mesenchymal stem cells (PMSCs). The stem cell "niche" within the chorionic villi regulates how PMSCs participate in placental tissue generation, maintenance and repair, but the anatomic location of the niche has not been defined. A number of cell surface markers for phenotypic characterisation of mesenchymal stem cells (MSCs) were employed to identify the stem cell niche within the chorionic villi of first trimester and term human placenta. This included antibodies to pericyte cell surface markers STRO-1 and 3G5, which have been used to identify mesenchymal stem cells in other tissues, but have not been studied in placental tissues. PMSCs were isolated from term human placentae and shown to have stem cell properties by their ability to grow on untreated plastic culture ware, capacity for forming clones (i.e. clonogenicity) and their capability to differentiate into adipocytes, chondrocytes and osteocytes. Western analysis confirmed that STRO-1 and 3G5 are present in placental protein extracts and in PMSCs. Immunocytochemistry revealed PMSCs were positive for MSC cell surface markers (STRO-1, 3G5, CD105, CD106, CD146, CD49a, alpha-SMA) and negative for haematopoietic stem cell markers (CD117, CD34) and endothelial markers (CD34, vWF). Immunohistochemistry with antibodies to MSC cell surface markers on first trimester and term tissues revealed a vascular niche for PMSCs. Dual-label immunofluorescence analysis was used to compare STRO-1 antibody staining with that of endothelial cell marker vWF and found no significant overlap in staining. This indicated that some PMSCs have a pericyte-like phenotype. We propose that the vascular niche harbours a pool of PMSCs that can give rise to committed progenitors for tissue maintenance and repair, and that PMSCs contribute to vessel maturation and stabilization.


Subject(s)
Blood Vessels/cytology , Chorionic Villi/physiology , Mesenchymal Stem Cells/cytology , Stem Cell Niche/cytology , Adult , Antigens, Surface/physiology , Biomarkers/analysis , Cell Differentiation/physiology , Female , Humans , Immunohistochemistry , Infant, Newborn , Male , Pregnancy
18.
Intern Med J ; 39(5): 304-16, 2009 May.
Article in English | MEDLINE | ID: mdl-19220531

ABSTRACT

Osteonecrosis of the Jaw (ONJ) is a recently recognised and potentially highly morbid complication of bisphosphonate therapy in the setting of metastatic malignancy, including myeloma. Members of the Medical and Scientific Advisory Group of the Myeloma Foundation of Australia formulated guidelines for the management of bisphosphonates around the issue of ONJ, based on the best available evidence in June 2008. Prior to commencement of therapy, patients should have an oral health assessment and be educated about the risks of ONJ. Dental assessment should occur 6 monthly during therapy. If tooth extraction is required, sufficient time should be allowed for complete healing to occur prior to commencement of bisphosphonate. As the risk of ONJ increases with duration of bisphosphonate therapy, we recommend annual assessment of dose with modification to 3 monthly i.v. therapy or to oral therapy with clodronate for those with all but the highest risk of skeletal-related event. Established ONJ should be managed conservatively; a bisphosphonate "drug holiday" is usually indicated and invasive surgery should generally be avoided. These recommendations will assist with clinical decision making for myeloma patients who are at risk of bisphosphonate-associated ONJ.


Subject(s)
Diphosphonates/adverse effects , Health Planning Guidelines , Jaw Diseases/chemically induced , Multiple Myeloma/drug therapy , Osteonecrosis/chemically induced , Animals , Bone Diseases/chemically induced , Bone Diseases/prevention & control , Bone Diseases/therapy , Humans , Jaw Diseases/prevention & control , Jaw Diseases/therapy , Multiple Myeloma/pathology , Osteonecrosis/prevention & control , Osteonecrosis/therapy , Treatment Outcome
20.
J Cell Physiol ; 214(2): 413-21, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17654479

ABSTRACT

Mesenchymal stem-like cells identified in different tissues reside in a perivascular niche. In the present study, we investigated the putative niche of adipose-derived stromal/stem cells (ASCs) using markers, associated with mesenchymal and perivascular cells, including STRO-1, CD146, and 3G5. Immunofluorescence staining of human adipose tissue sections, revealed that STRO-1 and 3G5 co-localized with CD146 to the perivascular regions of blood vessels. FACS was used to determine the capacity of the CD146, 3G5, and STRO-1 specific monoclonal antibodies to isolate clonogenic ASCs from disassociated human adipose tissue. Clonogenic fibroblastic colonies (CFU-F) were found to be enriched in those cell fractions selected with either STRO-1, CD146, or 3G5. Flow cytometric analysis revealed that cultured ASCs exhibited similar phenotypic profiles in relation to their expression of cell surface markers associated with stromal cells (CD44, CD90, CD105, CD106, CD146, CD166, STRO-1, alkaline phosphatase), endothelial cells (CD31, CD105, CD106, CD146, CD166), haematopoietic cells (CD14, CD31, CD45), and perivascular cells (3G5, STRO-1, CD146). The immunoselected ASCs populations maintained their characteristic multipotential properties as shown by their capacity to form Alizarin Red positive mineralized deposits, Oil Red O positive lipid droplets, and Alcian Blue positive proteoglycan-rich matrix in vitro. Furthermore, ASCs cultures established from either STRO-1, 3G5, or CD146 selected cell populations, were all capable of forming ectopic bone when transplanted subcutaneously into NOD/SCID mice. The findings presented here, describe a multipotential stem cell population within adult human adipose tissue, which appear to be intimately associated with perivascular cells surrounding the blood vessels.


Subject(s)
Adipose Tissue/cytology , Phenotype , Pluripotent Stem Cells/cytology , Stromal Cells/cytology , Adipogenesis , Adult , Animals , Antibodies, Monoclonal/metabolism , Biomarkers/metabolism , CD146 Antigen/metabolism , Cell Differentiation , Cell Separation/methods , Cells, Cultured , Chondrogenesis , DNA, Complementary/biosynthesis , Female , Flow Cytometry , Fluorescein-5-isothiocyanate , Fluorescent Antibody Technique, Indirect , Fluorescent Dyes , Humans , Immunohistochemistry , In Vitro Techniques , Male , Mice , Mice, SCID , Osteogenesis , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/transplantation , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL