Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
J Tissue Eng Regen Med ; 6(1): 21-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21312336

ABSTRACT

Tissue engineering sustains the need of a three-dimensional (3D) scaffold to promote the regeneration of tissues in volume. Usually, scaffolds are seeded with an adequate cell population, allowing their growth and maturation upon implantation in vivo. Previous studies obtained by our group evidenced significant growth patterns and osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs) when seeded and cultured on melt-based porous chitosan fibre mesh scaffolds (cell constructs). Therefore, it is crucial to test the in vivo performance of these in vitro 3D cell constructs. In this study, chitosan-based scaffolds were seeded and cultured in vitro with hBMSCs for 3 weeks under osteogenic stimulation conditions and analysed for cell adhesion, proliferation and differentiation. Implantation of 2 weeks precultured cell constructs in osteogenic culture conditions was performed into critical cranial size defects in nude mice. The objective of this study was to verify the scaffold integration and new bone formation. At 8 weeks of implantation, scaffolds were harvested and prepared for micro-computed tomography (µCT) analysis. Retrieved implants showed good integration with the surrounding tissue and significant bone formation, more evident for the scaffolds cultured and implanted with human cells. The results of this work demonstrated that chitosan-based scaffolds, besides supporting in vitro proliferation and osteogenic differentiation of hBMSCs, induced bone formation in vivo. Thus, their osteogenic potential in orthotopic location in immunodeficient mice was validated, evidencing good prospects for their use in bone tissue-engineering therapies.


Subject(s)
Bone Marrow Cells/cytology , Butylene Glycols/chemistry , Chitosan/chemistry , Polymers/chemistry , Stromal Cells/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Alkaline Phosphatase/metabolism , Animals , Bone and Bones/cytology , Bone and Bones/metabolism , Cell Survival , Cells, Cultured , Humans , Mice , Mice, Nude , Phenotype , Porosity , X-Ray Microtomography/methods
2.
In Vitro Cell Dev Biol Anim ; 47(4): 294-301, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21359818

ABSTRACT

Bone repair is a major concern in reconstructive surgery. Transplants containing osteogenically committed mesenchymal stem cells (MSCs) provide an alternative source to the currently used autologous bone transplants which have limited supply and require additional surgery to the patient. A major drawback, however is the lack of a critical mass of cells needed for successful transplantation. The purpose of the present study was to test the effects of FGF2 and FGF9 on expansion and differentiation of MSCs in order to establish an optimal culture protocol resulting in sufficient committed osteogenic cells required for successful in vivo transplantation. Bone marrow-derived MSCs cultured in αMEM medium supplemented with osteogenic supplements for up to three passages (control medium), were additionally treated with FGF2 and FGF9 in various combinations. Cultures were evaluated for viability, calcium deposition and in vivo osteogenic capacity by testing subcutaneous transplants in nude mice. FGF2 had a positive effect on the proliferative capacity of cultured MSCs compared to FGF9 and control medium treated cultures. Cultures treated with FGF2 followed by FGF9 showed an increased amount of extracted Alizarin red indicating greater osteogenic differentiation. Moreover, the osteogenic capacity of cultured cells transplanted in immunodeficient mice revealed that cells that were subjected to treatment with FGF2 in the first two passages and subsequently to FGF9 in the last passage only, were more successful in forming new bone. It is concluded that the protocol using FGF2 prior to FGF9 is beneficial to cell expansion and commitment, resulting in higher in vivo bone formation for successful bone tissue engineering.


Subject(s)
Bone Marrow Cells/physiology , Bone Regeneration/physiology , Cell Differentiation/physiology , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 9/pharmacology , Mesenchymal Stem Cells/physiology , Animals , Anthraquinones , Cell Culture Techniques/methods , Mice , Mice, Nude , Organic Chemicals
3.
J Endocrinol Invest ; 34(2): 106-10, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20543554

ABSTRACT

BACKGROUND: We demonstrated previously that phytoestrogens and vitamin D analogs like estradiol-17ß (E2) modulate bone morphology in rat female model. AIM: We now analyze the effects of phytoestrogens, E2, selective E2 re ceptor modulators, and the less-calcemic analogs of vitamin D: JKF1624F2-2 (JKF) or QW1624F2-2 (QW) on fat content in bone marrow (BM) from long bones in ovariectomized female rats (OVX). MATERIALS AND METHODS: OVX rats were injected with treatments known to affect bone formation, 5 days per week for 2.5 month for analysis of fat content in BM. RESULTS: In OVX young adults there is a decreased bone formation and a 10-fold increase in fat cells content in BM. Treatment with E2, raloxifene (Ral) or DT56a resulted in almost completely abolishment of fat cells content. Daidzein (D) decreased fat cells content by 80%, genistein (G) or biochainin A (BA) did not change fat cells content and carboxy BA (cBA) had a small but significant effect. JKF or QW did not affect fat cells content, whereas combined treatment of JKF or QW with E2 resulted in complete abolishment of fat cells content. These changes in fat cells content are inversely correlated with changes in bone formation. CONCLUSIONS: Our results demonstrate that adipogenesis induced by OVX is a reversible process which can be corrected by hormonal treatments. The awareness of a relationship between fat and bone at the marrow level might provide a better understanding of the pathophysiology of bone loss as well as a novel approach to diagnosis and treatment of postmenopausal osteoporosis.


Subject(s)
Adipocytes/drug effects , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Calcitriol/analogs & derivatives , Estrogens/pharmacology , Adipocytes/cytology , Animals , Calcitriol/pharmacology , Estradiol/pharmacology , Estrogen Receptor Modulators/pharmacology , Female , Genistein/pharmacology , Isoflavones/pharmacology , Ovariectomy , Phytoestrogens/pharmacology , Raloxifene Hydrochloride/pharmacology , Rats , Rats, Wistar
4.
Int J Oral Maxillofac Surg ; 39(8): 793-801, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20417057

ABSTRACT

Maxillary sinus membrane lifting is a common procedure aimed at increasing the volume of the maxillary sinus osseous floor prior to inserting dental implants. Clinical observations of bone formation in sinus lifting procedures without grafting bone substitutes were observed, but the biological nature of bone regeneration in sinus lifting procedures is unclear. This study tested whether this osteogenic activity relies on inherent osteogenic capacity residing in the sinus membrane by simulating the in vivo clinical condition of sinus lifting in an animal model. Maxillary sinus membrane cells were cultured in alpha-MEM medium containing osteogenic supplements (ascorbic acid, dexamethasone). Cultured cells revealed alkaline phosphatase activity and mRNA expression of osteogenic markers (alkaline phosphatase, bone sialoprotein, osteocalcin and osteonectin) verifying the osteogenic potential of the cells. Fresh tissue samples demonstrated positive alkaline phosphatase enzyme activity situated along the membrane-bone interface periosteum-like layer. To simulate the in vivo clinical conditions, the membranes were folded to form a pocket-like structure and were transplanted subcutaneously in immunodeficient mice for 8 weeks. New bone formation was observed in the transplants indicating the innate osteogenic potential within the maxillary Schneiderian sinus membrane and its possible contribution to bone regeneration in sinus lifting procedures.


Subject(s)
Adult Stem Cells/cytology , Bone Regeneration/physiology , Nasal Mucosa/cytology , Osteoblasts/cytology , Osteogenesis/physiology , Adolescent , Adult , Adult Stem Cells/metabolism , Adult Stem Cells/transplantation , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Biological Assay , Calcification, Physiologic/physiology , Humans , Integrin-Binding Sialoprotein/genetics , Integrin-Binding Sialoprotein/metabolism , Maxillary Sinus , Mice , Mice, Nude , Osteoblasts/metabolism , Osteoblasts/transplantation , Osteocalcin/genetics , Osteocalcin/metabolism , Osteonectin/genetics , Osteonectin/metabolism , RNA, Messenger/analysis , Transplantation, Heterologous , Young Adult
5.
J Tissue Eng Regen Med ; 4(1): 30-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19842114

ABSTRACT

Biocompatible and osteoconductive cell-scaffold constructs comprise the first and most important step towards successful in vivo bone repair. This study reports on a new cell-scaffold construct composed of gelatin-based hydrogel and ceramic (CaCO(3)/beta-TCP) particles loaded with human MSCs producing a tissue-like construct applied as a transplant for in vivo bone formation. Bone marrow-derived human MSCs were cultured in osteogenic induction medium. 5 x 10(5) (P(2)) cells were loaded on a mixture of hydrogel microspheres and ceramic particles, cultured in a rotating dynamic culture for up to 3 weeks. Both hydrogel microspheres and ceramic particles coalesced together to form a tissue-like construct, shown by histology to contain elongated spindle-like cells forming the new tissue between the individual particles. Cell proliferation and cell viability were confirmed by Alamar blue assay and by staining with CFDA, respectively. FACS analysis conducted before loading the cells, and after formation of the construct, revealed that the profile of cell surface markers remained unchanged throughout the dynamic culture. The osteogenic potential of the cells composing the tissue-like construct was further validated by subcutaneous transplants in athymic nude mice. After 8 weeks a substantial amount of new bone formation was observed in the cell-construct transplants, whereas no bone formation was observed in transplants containing no cells. This new cell construct provides a system for in vivo bone transplants. It can be tailored for a specific size and shape as needed for various transplant sites and for all aspects of regenerative medicine and biomaterial science.


Subject(s)
Bone Marrow Cells/cytology , Mesenchymal Stem Cells/cytology , Osteogenesis/physiology , Tissue Engineering/methods , Tissue Scaffolds , Animals , Bone Marrow Cells/physiology , Bone Regeneration/physiology , Ceramics , Flow Cytometry , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Mice , Mice, Nude , Microscopy, Electron, Scanning , Microspheres
6.
Calcif Tissue Int ; 84(2): 138-45, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19067018

ABSTRACT

Recent studies successfully demonstrated induction of new bone formation in the maxillary sinus by mucosal membrane lifting without the use of any graft material. The aim of this work was to test the osteogenic potential of human maxillary sinus Schneiderian membrane (hMSSM) using both in vitro and in vivo assays. Samples of hMSSM were used for establishment of cell cultures and for histological studies. Flow cytometry analysis was performed on P(0), P(1), and P(2) cultures using established mesenchymal progenitor cell markers (CD 105, CD 146, CD 71, CD 73, CD 166), and the ability of hMSSM cells to undergo osteogenic differentiation in culture was analyzed using relevant in vitro assays. Results showed that hMSSM cells could be induced to express alkaline phosphatase, bone morphogenic protein-2, osteopontin, osteonectin, and osteocalcin and to mineralize their extracellular matrix. Inherent osteogenic potential of hMSSM-derived cells was further proven by in vivo experiments, which demonstrated the formation of histology-proven bone at ectopic sites following transplantation of hMSSM-derived cells in conjunction with an osteoconductive scaffold. This study provides the biological background for understanding the observed clinical phenomena in sinus lifting. Our results show that a genuine osteogenic potential is associated with the hMSSM and can contribute to development of successful sinus augmentation techniques.


Subject(s)
Nasal Mucosa/cytology , Osteoblasts/cytology , Stem Cells/cytology , Animals , Calcification, Physiologic , Cell Differentiation , Cells, Cultured , Humans , Mice , Mice, Nude , Nasal Mucosa/chemistry , Nasal Mucosa/metabolism , Osteoblasts/metabolism , Osteoblasts/transplantation , Osteogenesis , Stem Cells/metabolism
7.
Histochem Cell Biol ; 129(5): 589-97, 2008 May.
Article in English | MEDLINE | ID: mdl-18274772

ABSTRACT

An intimate interplay exists between the bone and the immune system, which has been recently termed osteoimmunology. The activity of immune cells affects the intrinsic balance of bone mineralization and resorption carried out by the opposing actions of osteoblasts and osteoclasts. The aim of this study was to determine the possible interaction between inflammatory-induced conditions and matrix metalloproteinases-2,-9 (MMP-2,-9) synthesis and secretion by bone marrow-derived osteoprogenitor cells during advanced stages of osteogenesis. Rat bone marrow-derived mesenchymal stem cells (MSCs) were cultured in the presence of osteogenic supplements in order to direct the cells towards the osteogenic differentiation lineage. At the late stages of osteogenesis, assessed by histochemistry, immunohistochemistry and RT-PCR, cultures were exposed to pro-inflammatory cytokines, tumor necrosis factor-alpha (TNF-alpha) and interleukin-1 alpha (IL-1 alpha). Biochemical, histochemical and molecular biology techniques were used to discern the influence of pro-inflammatory cytokines on MMP-2,-9 synthesis and secretion. Results indicated that MMP-9 synthesis and secretion were significantly induced after exposure to the cytokines (TNF-alpha, IL-1 alpha) treatment, while MMP-2 levels remained unchanged. These results indicate that in response to inflammatory processes, osteoblasts, in addition to osteoclasts, can also be involved and contribute to the process of active bone resorption by secretion and activation of MMPs.


Subject(s)
Bone Marrow Cells/drug effects , Cytokines/pharmacology , Matrix Metalloproteinase 9/biosynthesis , Mesenchymal Stem Cells/drug effects , Animals , Blotting, Western , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Immunohistochemistry , Inflammation Mediators/pharmacology , Interleukin-1alpha/pharmacology , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteogenesis/drug effects , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/pharmacology
8.
J Mater Sci Mater Med ; 19(3): 1249-55, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17701297

ABSTRACT

UNLABELLED: Extra cellular matrix (ECM) is a natural cell environment, possesses complicated nano- and macro- architecture. Mimicking this three-dimensional (3-D) web is a challenge in the modern tissue engineering. This study examined the application of a novel 3-D construct, produced by multilayered organization of electrospun nanofiber membranes, for human bone marrow-derived mesenchymal stem cells (hMSCs) support. The hMSCs were seeded on an electrospun scaffold composed of poly epsilon-caproloactone (PCL) and collagen (COL) (1:1), and cultured in a dynamic flow bioreactor prior to in vivo implantation. Cell viability after seeding was analyzed by AlamarBlue Assay. At the various stages of experiment, cell morphology was examined by histology, scanning electron microscopy (SEM) and confocal microscopy. RESULTS: A porous 3-D network of randomly oriented nanofibers appeared to support cell attachment in a way similar to traditionally used tissue culture polysterene plate. The following 6 week culture process of the tested construct in the dynamic flow system led to massive cell proliferation with even distribution inside the scaffold. Subcutaneous implantation of the cultured construct into nude mice demonstrated good integration with the surrounding tissues and neovascularization. CONCLUSION: The combination of electrospinning technology with multilayer technique resulted in the novel 3-D nanofiber multilayered construct, able to contain efficient cell mass necessary for a successful in vivo grafting. The success of this approach with undifferentiated cells implies the possibility of its application as a platform for development of constructs with cells directed into various tissue types.


Subject(s)
Biomimetic Materials , Extracellular Matrix , Models, Anatomic , Tissue Scaffolds , Animals , Cell Movement , Cell Survival , Cells, Cultured , Extracellular Matrix/chemistry , Humans , Male , Materials Testing , Mice , Mice, Nude , Mice, Transgenic , Nanostructures , Porosity , Prostheses and Implants , Tissue Scaffolds/chemistry
9.
J Cell Biochem ; 103(6): 1826-32, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18059016

ABSTRACT

Ovariectomy of immature female rats, results in significant decrease of trabecular bone volume and in cortical bone thickness. Previously, we found that estradiol-17beta (E(2)) restored bone structure of ovariectomized (Ovx) female rats to values obtained in intact sham-operated female rats. E(2) also selectively stimulated creatine kinase (CK) specific activity a hormonal-genomic activity marker. In the present study, we compared the effects of E(2) and the phytoestrogens: daidzein (D), biochainin A (BA), genistein (G), carboxy-derivative of BA (cBA), and the SERM raloxifene (Ral) in Ovx, on both histological changes of bones and CK, when administered in multiple daily injections for 2.5 months. Bone from Ovx rats, showed significant disrupted architecture of the growth plate, with fewer proliferative cells and less chondroblasts. The metaphysis underneath the growth plate, contained less trabeculae but a significant increased number of adipocytes in the bone marrow. D like E(2) and Ral but not G, BA, or cBA, restored the morphology of the tibiae, similar to that of control sham-operated animals; the bony trabeculeae observed in the primary spongiosa was thicker, with almost no adipocytes in bone marrow. Ovariectomy resulted also in reduced CK, which in both epiphysis and diaphysis was stimulated by all estrogenic compounds tested. In summary, only D stimulated skeletal tissues growth and differentiation as effectively as E(2) or Ral, suggesting that under our experimental conditions, D is more effective in reversing menopausal changes than any of the other isolated phytoestrogens which cannot be considered as one entity.


Subject(s)
Bone and Bones/drug effects , Creatine Kinase/metabolism , Isoflavones/pharmacology , Phytoestrogens/pharmacology , Adipocytes/pathology , Animals , Bone Marrow/pathology , Bone and Bones/enzymology , Bone and Bones/pathology , Estradiol/administration & dosage , Estradiol/pharmacology , Female , Genistein/administration & dosage , Genistein/pharmacology , Growth Plate/drug effects , Growth Plate/pathology , Isoflavones/administration & dosage , Ovariectomy , Phytoestrogens/administration & dosage , Raloxifene Hydrochloride/administration & dosage , Raloxifene Hydrochloride/pharmacology , Rats , Rats, Wistar , Tibia/drug effects , Tibia/enzymology , Tibia/pathology , Trabecular Meshwork/drug effects , Trabecular Meshwork/enzymology , Trabecular Meshwork/pathology
10.
Pediatr Hematol Oncol ; 24(2): 111-5, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17454776

ABSTRACT

This pilot study aimed at determining serum VEGF levels (S-VEGF) at diagnosis and at restaging in children with Hodgkin lymphoma, and investigating whether this parameter provides prognostic information for remission after 2 courses of chemotherapy. PET-CT fusion was performed to assess response to treatment. Changes in S-VEGF levels were found to correlate with response to treatment for most of the children. This provides a rationale for exploring clinical interest in S-VEGF measurements in a larger group of children with Hodgkin lymphoma, and using the test for clinical trials of anti-angiogenic therapies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/blood , Hodgkin Disease/blood , Vascular Endothelial Growth Factor A/blood , Adolescent , Child , Child, Preschool , Diagnosis, Differential , Enzyme-Linked Immunosorbent Assay , Female , Hodgkin Disease/diagnosis , Hodgkin Disease/drug therapy , Humans , Male , Neoplasm Staging/methods , Pilot Projects , Positron-Emission Tomography , Remission Induction , Tomography, X-Ray Computed
11.
J Cell Biochem ; 100(6): 1406-14, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17163482

ABSTRACT

We demonstrated previously that daily injection for 3 days of the less calcemic vitamin D analogs: JK 1624 F(2)-2 (JKF) and QW 1624F(2)-2 (QW) followed by estradiol-17beta (E(2)) in female rats upregulated creatine kinase-specific activity (CK) in skeletal tissues. In this study, we evaluated both histomorphological and biochemical changes due to a regime of 4 days treatment with JKF or QW, followed by injection of E(2) on day 5, repeated for 2.5 months. Ovariectomized female rats (Ovx) were injected 2 weeks after surgery, with JKF or QW at 0.2 ng/g BW followed by injections of E(2) (1 microg/rat) on day 5 of each week for 2.5 months. Rats were sacrificed 24 h after the last injection and bones were analyzed. JKF alone decreased growth plate width, increased % total bone volume (%TBV), with no change in cortical thickness. In contrast, QW restored growth plate width and %TBV with no change in cortical thickness. Combined with E(2), JKF restored %TBV and growth plate width but with no change in cortical thickness, while QW restored significantly all parameters including cortical thickness. Moreover, there was also an increase in the responsiveness of CK to E(2) in epiphyseal cartilage and diaphyseal bone but not in uterus. Thus, vitamin D less calcemic analogs increased responsiveness to E(2) morphologically as well as biochemically. We, therefore, conclude that combined treatment of less calcemic analogs vitamin D and E(2) might be superior for treatment of bone damage caused by ovariectomy in female rats and might be applied for post-menopausal osteoporosis.


Subject(s)
Estradiol/pharmacology , Tibia/drug effects , Vitamin D/pharmacology , Animals , Creatine Kinase/metabolism , Diaphyses/drug effects , Enzyme Activation/drug effects , Female , Growth Plate/anatomy & histology , Growth Plate/drug effects , Growth Plate/metabolism , Molecular Structure , Ovariectomy , Rats , Rats, Wistar , Tibia/anatomy & histology , Tibia/metabolism , Time Factors , Vitamin D/analogs & derivatives , Vitamin D/chemistry , Vitamins/pharmacology
12.
Pediatr Hematol Oncol ; 22(6): 513-24, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16169818

ABSTRACT

The aim of this pilot study was to determine VEGF serum levels (S-VEGF) at diagnosis and at restaging in children diagnosed with cancer, and to investigate whether this parameter provides prognostic information for remission after induction therapy and response to treatment. S-VEGF levels of 35 consecutive pediatric patients with various types of cancer were assayed at diagnosis and at restaging. Levels of VEGF were determined using a commercially available ELISA anti-human VEGF immunoassay kit. Thirty-one children went into complete remission or had a very good partial response to first-line therapy; 4 patients developed tumor progression. At diagnosis average S-VEGF level was 495 pg/mL (range, 0.89--2220 pg/mL) and at restaging it decreased to 118.36 pg/mL (range, 7.44--487 pg/mL). (p=.0039). The 4 patients with tumor progression had increased S-VEGF levels at restaging. The comparison between the levels of S-VEGF at diagnosis and at restaging showed a significant difference for the patients who responded to treatment with decreased S-VEGF and the patients who developed tumor progression with increased S-VEGF (p=.0019). One child with metastatic Ewing sarcoma developed progressive disease after several weeks, with significantly progressively higher S-VEGF levels. One child with Hodgkin disease, who had a higher level at first restaging and developed progressive disease, responded to reinduction therapy and had a significantly lower level at the second restaging. The child with metastatic hepatoblastoma responded to first-line chemotherapy with concomitant decrease in S-VEGF and alpha-fetoprotein levels, but developed local recurrence with elevation in both parameters. Changes in S-VEGF levels correlated with response to treatment for most of the children diagnosed with cancer. This provides a rationale for exploring clinical interest in S-VEGF measurements of a larger group of children with malignancies, and using the test for clinical trials of antiangiogenic therapies.


Subject(s)
Biomarkers, Tumor/blood , Neoplasms/blood , Vascular Endothelial Growth Factor A/blood , Adolescent , Adult , Child , Child, Preschool , Enzyme-Linked Immunosorbent Assay , Female , Humans , Infant , Male , Neoplasm Staging/methods , Neoplasms/diagnostic imaging , Neoplasms/therapy , Radiography , Remission Induction
13.
Microsc Res Tech ; 66(2-3): 132-8, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15880496

ABSTRACT

Bone marrow contains progenitor cells that are able to differentiate into several mesenchymal lineages, including bone. These cells may also provide a potential therapy for bone repair. The purpose of this study was to select the osteoprogenitor cell subpopulation from bone marrow-derived mesenchymal stem cells (MSCs) and to test the ability of a hydrogel scaffold to support growth and osteogenic differentiation. MSCs isolated from rat femur bone marrow were cultured in DMEM medium supplemented with antibiotics, FCS, and L-glutamine. Osteogenic supplements (dexamethasone, sodium beta-glycerophosphate, and ascorbic acid) were added for one, two or three weeks. A selective subpopulation of osteoprogenitor cells was identified by immunohistochemistry, general morphology, scanning electron microscopy (SEM), and energy dispersive spectroscopy (EDS). Committed osteogenic cells were transferred to a 3-D hydrogel scaffold and cultured for an additional week. In standard culture, the osteoprogenitor cells formed cell clusters identified by Alizarin red S staining and by positive osteocalcin immunostaining. The number of osteoprogenitor cells, matrix synthesis, and mineralization increased gradually up to three weeks in culture. Mineral deposition in the matrix analyzed by EDS revealed the presence of calcium and phosphate ions at a Ca/P molar ratio of 1.73 in both the osteogenic cultures and the scaffold osteoprogenitor culture. Histological preparations revealed cell clusters within the hydrogel scaffold and SEM analysis revealed cell clusters attached to the scaffold surface. It is concluded that the hydrogel scaffold can support growth and differentiation of osteogenic cultures including mineralization and can potentially serve as a bone graft substitute containing committed osteoprogenitor cells.


Subject(s)
Bone Marrow Cells/cytology , Calcification, Physiologic , Hydrogel, Polyethylene Glycol Dimethacrylate , Mesenchymal Stem Cells/cytology , Animals , Biomarkers/metabolism , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Extracellular Matrix/metabolism , Immunohistochemistry , Mesenchymal Stem Cells/metabolism , Microscopy, Electron, Scanning , Osteocalcin/metabolism , Rats , Rats, Sprague-Dawley , Spectrum Analysis , Tissue Engineering
14.
Mech Ageing Dev ; 126(2): 281-7, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15621208

ABSTRACT

The loss of bone mass observed in aging enhances the risk of fractures. The process of bone repair in aging is slow and limited due to reduced activity of the osteoblasts. Bone marrow stem cells (MSCs) residing in the bone marrow are the progenitors for osteoblasts. The ability to enhance healing of bone defect in aging by MSCs can contribute in the prevention of the complications resulting from long-term immobilization that are especially fatal in old age. Our aim was to test the ability of MSCs inserted into a biological scaffold to enhance bone defect repair. Osteoprogenitor cells were selected from rat bone marrow stem cells cultured in DMEM medium supplemented with FCS, antibiotics, ascorbic acid, beta-glycerophosphate, and dexamethasone. The selected osteogenic subpopulation was identified by osteocalcin immunohistochemistry as well as Alizarin red S and von Kossa staining which are specific for bone matrix and mineral deposition. Committed osteoprogenitor cells cultured on the hydrogel scaffold were transplanted into the area of a rat tibia segmental bone defect and examined after 6 weeks. Radiology images revealed that 6 weeks post-implantaion, calcified material was present in the site of the defect, indicating new bone formation. It is concluded that committed osteogenic MSCs contained in a biocompatible scaffold can provide a promising surgical tool for enhancement of bone defect healing that will minimize the complications of bone repair in aging and disease.


Subject(s)
Aging , Bone Marrow Cells/cytology , Bone and Bones/pathology , Fracture Healing , Stem Cells/cytology , Animals , Anti-Bacterial Agents/pharmacology , Ascorbic Acid/pharmacology , Blood Proteins/pharmacology , Bone Marrow Cells/metabolism , Bone and Bones/metabolism , Bone and Bones/ultrastructure , Cells, Cultured , Dexamethasone/pharmacology , Glycerophosphates/pharmacology , Growth Substances , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Immunohistochemistry , Mesenchymal Stem Cells/metabolism , Mesoderm/cytology , Microscopy, Electron, Scanning , Models, Biological , Osteoblasts/metabolism , Osteocalcin/metabolism , Osteogenesis , Rats , Rats, Sprague-Dawley , Time Factors , Wound Healing
15.
Cell Tissue Bank ; 5(4): 223-30, 2004.
Article in English | MEDLINE | ID: mdl-15591825

ABSTRACT

Bone repair is one of the major challenges facing reconstructive surgery. Bone regeneration is needed for the repair of large defects and fractures. The ability of TGF-beta1 and IGF-1 incorporated into hydrogel scaffold to induce bone regeneration was evaluated in a rat tibia segmental defect model. External fixation was performed prior to the induction of the segmental bone defect in order to stabilize the defect site. Hydrogel scaffold containing either TGF-beta, IGF-1, TGF-beta + IGF-1, hydrogel containing saline or saline, were inserted in the defect. Calcified material was observed in the defects treated with TGF-beta 2 weeks following the start of treatment. Bone defects treated with TGF-beta, IGF-1 or TGF-beta + IGF-1 revealed significant bone formation after 4 and 6 weeks when compared to the control specimens. X-ray images showed that solid bone was present at the defect site after 6 weeks of treatment with TGF-beta or TGF-beta + IGF-1. A less pronounced bone induction was observed in the control specimens and bones treated with IGF-1. Percent closure ratio of bone defects after 6 weeks were 40, 80, 89, and 97% for saline, hydrogel, IGF-1, TGF-beta and IGF-1 + TGF-beta groups, respectively. It is concluded that hydrogel scaffold can serve as a good osteoconductive matrix for growth factors, and that it provides a site for bone regeneration and enhances bone defect healing and could be used as alternative graft material.


Subject(s)
Bone Regeneration/physiology , Bone and Bones/metabolism , Insulin-Like Growth Factor I/metabolism , Transforming Growth Factor beta/metabolism , Animals , Bone and Bones/diagnostic imaging , Bone and Bones/injuries , Hydrogels , Radiography , Rats , Tibia/diagnostic imaging , Tibia/injuries , Tibia/metabolism , Time Factors
16.
Osteoarthritis Cartilage ; 12(4): 284-95, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15023380

ABSTRACT

OBJECTIVES: To characterize the molecular events underlying cartilage injury in the early phase of mono-iodoacetate-induced osteoarthritis (OA) in rats. METHODS: Experimental osteoarthritis was induced by intra-articular injection of 0.03mg mono-iodoacetate (MIA) in Wistar rats. Animals were killed 2, 5, 10, 15 and 20 days post-injection. Synovial tissue and standardized biopsies from different areas of knee cartilage were examined. Proteoglycan synthesis ((35)S incorporation) and gelatinase activities (zymography), semi-quantitative RT-PCR and immunohistochemistry for IL1beta, iNOS, COX2 and PPARgamma, were performed on these samples. RESULTS: Changes in proteoglycan synthesis and gelatinase activities were time and site-dependent. Proteoglycan synthesis inhibition was maximal by day 2 while the highest gelatinase activities were observed at day 5. Central part of patella and posterior plateaus and condyles, i.e. the weight-bearing cartilage areas, were the most affected. IL1beta and iNOS transcripts were induced early in cartilage at time of maximal proteoglycan synthesis inhibition, especially in weight-bearing areas. COX-2 was slightly up-regulated whereas PPARgamma gene expression remained unchanged. Gene expression profile in synovium paralleled that of cartilage, except for PPARgamma which was up-regulated at day 15 and 20. Immunostaining for IL1beta and iNOS showed that proteins were located in diseased cartilage areas at early stage of the experimental OA (day 2). At later time-points (day 20), IL1beta and iNOS were expressed in perilesional areas whereas immunostaining became below control level for COX-2 and PPARgamma. CONCLUSIONS: Time-dependent degradation of cartilage after injection of low dose of MIA (0.03mg) into rat knee joint can be related to early loss of proteoglycan anabolism, increased gelatinase activities and expression of IL1beta and downstream inflammatory genes. Increased susceptibility to MIA in weight-bearing areas of cartilage further indicate that MIA-induced experimental OA is a relevant model to study not only metabolical but also biomechanical aspects of human OA.


Subject(s)
Cartilage, Articular/metabolism , Gene Expression/genetics , Osteoarthritis/genetics , Animals , Cyclooxygenase 2 , Enzyme Inhibitors , Gelatinases/metabolism , Immunohistochemistry/methods , Interleukin-1/analysis , Iodoacetic Acid , Isoenzymes/analysis , Male , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase Type II , Osteoarthritis/metabolism , Prostaglandin-Endoperoxide Synthases/analysis , Proteoglycans/biosynthesis , RNA, Messenger/analysis , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/analysis , Reverse Transcriptase Polymerase Chain Reaction/methods , Time Factors , Transcription Factors/analysis
17.
Exp Gerontol ; 37(4): 553-65, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11830358

ABSTRACT

Bone defects are often created in order to repair bone pathologies. In the aging population, the healing of such defects is very limited. Bone healing in aging depends on the availability of various hormone and growth factors. The ability of growth factors to enhance bone formation in femoral defects in old rats was tested. Bone defects were induced in femurs of old rats. A single dose of transforming growth factor-beta (TGF-beta), IGF-1, TGF-beta+IGF-1 or saline was inserted in the defect and bones were tested after 2 and 4 weeks. Radiology revealed that mineralization appeared in the 2 weeks group in defects treated with TGF-beta and in defects treated with TGF-beta, TGF-beta+IGF-1 in the 4 weeks groups. Computerized tomography (CT) coronal and axial images revealed that 4 weeks after treatment with TGF-beta+IGF-1, a complete bone bridge was observed. Morphology revealed that these defects were filled with trabecular bone. A less pronounced bone healing was observed after TGF-beta or IGF-1, while control specimens revealed partial healing of the bone defect. Biomechanical tests indicated that treatment with TGF-beta, IGF-1 or TGF-beta+IGF-1 resulted in a significant increase of bone bending rigidity compared to control in the 4 weeks group and that TGF-beta+IGF-1 was the most inductive in this respect. The ability to induce bone healing in aging by TGF-beta+IGF-1 is of a great clinical importance for restoration of bone strength and biomechanical properties of bone defects in aging.


Subject(s)
Aging/physiology , Bone Remodeling/drug effects , Fracture Healing/drug effects , Insulin-Like Growth Factor I/pharmacology , Transforming Growth Factor beta/pharmacology , Animals , Biomechanical Phenomena , Female , Rats , Rats, Wistar , Stress, Mechanical
18.
Arch Gerontol Geriatr ; 35(1): 59-69, 2002.
Article in English | MEDLINE | ID: mdl-14764345

ABSTRACT

Matrix metalloproteinases are important in the physiological and pathological degradation of extracellular matrix including that of bone and cartilage. The process of bone defect healing is associated with formation of cartilage callus and cancelous bone. With maturation and aging, the response of skeletal tissues to injury is limited. The ability of growth factors to enhance bone defect healing in aged rats was studied. Partial bone defects were induced in femurs of aged rats. A single dose of IGF-1, TGF-beta+IGF-1 or saline was inserted in the defect and bones were examined after 2 and 4 weeks. Morphology revealed that after 2 weeks of treatment with TGF-beta the defects were filled with mesenchyme-like tissue and delicate bone trabeculae. Positive staining for metalloproteinase-2 (MMP-2) was shown at the sites of new bone formation. In defects treated with IGF-1 or TGF-beta+IGF-1 nodules of cartilage and fine bone trabeculae along with positive staining for both MMP-2 and MMP-3 were demonstrated in the healing defects. After 4 weeks radiology revealed mineralization in defects treated with TGF-beta and less pronounced mineralization after treatment with IGF-1, or with TGF-beta+IGF-1, whereas only partial healing of the defects was observed in control specimens. MMP-2 and MMP-3 were detected at sites of new bone formation after treatment with TGF-beta, IGF-1, and TGF-beta+IGF-1. It is concluded that TGF-beta and IGF-1 induced bone defect healing in aged rats. TGF-beta induced bone formation while IGF-1 induced cartilage and than bone formation via endochondral ossification. The localization of MMP-2 and MMP-3 in the healing defects reflected the synthesis of bone or cartilage matrices in the defect, reflecting the involvement of MMPs in the process of bone formation and endochondral ossification. The ability to induce bone defect healing in aging is of great clinical importance and understanding the involvement of MMPs in this process can contribute to future treatment with growth factors to enhance bone defect healing in 20-month-old female rats.

19.
J Clin Invest ; 108(3): 407-14, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11489934

ABSTRACT

The study of human cardiac tissue development is hampered by the lack of a suitable in vitro model. We describe the phenotypic properties of cardiomyocytes derived from human embryonic stem (ES) cells. Human ES cells were cultivated in suspension and plated to form aggregates termed embryoid bodies (EBs). Spontaneously contracting areas appeared in 8.1% of the EBs. Cells from the spontaneously contracting areas within EBs were stained positively with anti-cardiac myosin heavy chain, anti--alpha-actinin, anti-desmin, anti--cardiac troponin I (anti-cTnI), and anti-ANP antibodies. Electron microscopy revealed varying degrees of myofibrillar organization, consistent with early-stage cardiomyocytes. RT-PCR studies demonstrated the expression of several cardiac-specific genes and transcription factors. Extracellular electrograms were characterized by a sharp component lasting 30 +/- 25 milliseconds, followed by a slow component of 347 +/- 120 milliseconds. Intracellular Ca(2+) transients displayed a sharp rise lasting 130 +/- 27 milliseconds and a relaxation component lasting 200--300 milliseconds. Positive and negative chronotropic effects were induced by application of isoproterenol and carbamylcholine, respectively. In conclusion, the human ES cell--derived cardiomyocytes displayed structural and functional properties of early-stage cardiomyocytes. Establishment of this unique differentiation system may have significant impact on the study of early human cardiac differentiation, functional genomics, pharmacological testing, cell therapy, and tissue engineering.


Subject(s)
Embryo, Mammalian/cytology , Myocardium/cytology , Stem Cells/cytology , Actinin/genetics , Actinin/metabolism , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Base Sequence , Calcium/metabolism , Cell Aggregation , Cell Differentiation , Cells, Cultured , DNA Primers/genetics , Desmin/genetics , Desmin/metabolism , Gene Expression , Humans , Microscopy, Electron , Myocardial Contraction , Myocardium/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Stem Cells/metabolism , Troponin I/genetics , Troponin I/metabolism
20.
Arch Gerontol Geriatr ; 31(2): 159-176, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11090910

ABSTRACT

Osteoarthritis is a degenerative joint disease characterized by destruction of the articular cartilage in aging and senescence. The aim of this study was to study the possible treatment of this disease by intraarticular injection of growth factors to osteoarthritic joints of aged animals. 20-month-old female ICR mice were injected with insulin-like growth factor (IGF-1), transforming growth factor-beta (TGF-beta) or TGF-beta+IGF-1 on days 1, 4, and 7. On day 9 the joints were dissected and cultured in the presence of 35S-sulfate and 3H-thymidine. Combined treatment of TGF-beta and IGF-1 resulted in elevated 3H-thymidine incorporation and DNA and protein contents, reduction of 35S-sulfate incorporation and alkaline phosphatase activity, with no significant change in the activity of acid phosphatase. Following injections of TGF-beta, contents of DNA and protein, and incorporations of 3H-thymidine were induced, and 35S-sulfate and alkaline phosphatase activity were reduced. Treatment with IGF-1 resulted in reduced incorporation of 3H-thymidine with no significant changes in the activity of acid phosphatase. Atypically hypertrophic chondrocytes were observed along the articular surface and the endogenous production of TGF-beta and of IGF-1, as revealed by immunohistochemistry, was reduced. It is concluded that although 3H-thymidine incorporation and alkaline phosphatase activity appeared to be induced by TGF-beta and IGF-1, the overall responsiveness of cartilage from aged mice to these growth factors appeared to be inhibitory. Moreover, their effects appeared to be limited to specific cell populations in the cartilage itself.

SELECTION OF CITATIONS
SEARCH DETAIL
...