Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Ovarian Res ; 11(1): 22, 2018 Mar 06.
Article in English | MEDLINE | ID: mdl-29510732

ABSTRACT

BACKGROUND: The recurrence, metastasis and poor prognosis are important characteristics of ovarian carcinoma (OC), which are associated with exfoliation of cells from the primary tumor and colonization of the cells in pelvic cavity. On the other hand, the life quality of the patients undergoing surgical resection of OC was influenced by postoperative adhesions. Therefore, preventing postoperative implant tumor and adhesion may be effective methods to improve OC treatment. HyaRegen Gel, a cross-linked hyaluronan gel (CHAG), has been widely used as an anti-adhesive agent following pelvic operation in clinic. However, whether it can affect the implantation and growth of OC cells or not is still not clear. METHODS: Migration and invasion assays were applied to detect the effect of CHAG on migration and invasion of OC cells. Western blotting was performed to detect the phosphorylation/activation of EGFR and ERK, and the expression of PCNA and MMP7. Pull down assay was used to analyze the effect of CHAG on the activation of small G protein Rac1. Nude mice implantation tumor model was applied to observe the effect of CHAG on implantation tumor of OC cells. RESULTS: The results of in vitro experiments showed that CHAG suppressed both basic and EGF-induced migration and invasion of OC cells, blocked the activation of EGF-initiated EGFR activation, inhibited downstream signal transduction of EGFR, and decreased expression of proliferation and migration/invasion related proteins. Meanwhile, results of in vivo experiments showed that CHAG not only inhibited the formation of implantation tumor of OC cells but also delayed the of the growth of the tumors. CONCLUSIONS: CHAG inhibited migration, invasion and proliferation of OC cells in vitro, and suppressed development of implantation tumor of OC in vivo. This made it as both anti-tumor and anti-adhesion agents.


Subject(s)
Antineoplastic Agents/administration & dosage , Gels , Hyaluronic Acid/administration & dosage , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Female , Gels/chemistry , Humans , Hyaluronic Acid/chemistry , Mice , Ovarian Neoplasms , Signal Transduction/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , rac1 GTP-Binding Protein/metabolism
2.
Oncotarget ; 7(40): 65418-65428, 2016 Oct 04.
Article in English | MEDLINE | ID: mdl-27589842

ABSTRACT

Cross-linked hyaluronic acid gel (CHAG) has been used to prevent postoperative adhesion of abdominal tumorectomy. However, its effect on tumor cells is still unknown. This paper was designed to investigate the effect of CHAG on metastasis and growth of tumor cells. Migration and invasion assays, Western blotting, pull down assay, siRNA interference, and nude mice implantation tumor model were applied in this study. The results of in vitro experiments with gastric cancer cell line AGS and hepatic cancer cell line HepG2 showed that CHAG inhibited the migration and invasion activities, the MAPK and PI3K/Akt mediated signaling, the activation of small G proteins Rac1 and RhoA, and the expression of MMPs and PCNA initiated by EGF, through blocking the activation of EGFR. CHAG also had inhibitory effect on activation of other membrane receptors, including integrin and VEGFR. When the expression of hyaluronic acid receptors (CD44 or RHAMM) was interfered, the above inhibitory effects of CHAG still existed. In vivo experimental results showed that CHAG suppressed colonization, growth and metastasis of gastric cancer cell line SGC-7901 in peritoneal cavity of nude mice. In conclusion, CHAG had inhibitory effect on tumor cells, through covering cell surface and blocking the interaction between extracellular stimulative factors and their receptors.


Subject(s)
Hyaluronic Acid/pharmacology , Liver Neoplasms/surgery , Postoperative Complications/prevention & control , Stomach Neoplasms/surgery , Tissue Adhesions/prevention & control , Animals , Cell Growth Processes/drug effects , Cell Movement/drug effects , Gels/therapeutic use , Hep G2 Cells , Humans , Hyaluronic Acid/therapeutic use , Liver Neoplasms/therapy , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Neoplasm Recurrence, Local , Stomach Neoplasms/therapy , Tissue Adhesions/etiology , Xenograft Model Antitumor Assays
3.
Int J Mol Sci ; 17(4): 411, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-27092487

ABSTRACT

Cross-linked hyaluronic acid hydrogel (cHA gel) and dexamethasone (Dex) have been used to treat knee osteoarthritis (OA) in clinical practice owing to their chondroprotective and anti-inflammatory effects, respectively. The aim of the present study was to compare the treatment effects of the cHA gel pre-mixed with/without Dex in a surgery-induced osteoarthritis model in rats. Anterior cruciate ligament transection (ACLT) surgery was performed on the right knee of rats to induce OA. Male 2-month-old Sprague-Dawley rats were randomly divided into five groups (n = 10/per group): (1) ACLT + saline; (2) ACLT + cHA gel; (3) ACLT + cHA-Dex (0.2 mg/mL) gel; (4) ACLT + cHA-Dex (0.5 mg/mL) gel; (5) Sham + saline. Intra-joint injections were performed four weeks after ACLT in the right knee. All animals were euthanized at 12 weeks post-surgery. Cartilage damage and changes in the synovial membrane were assessed by micro X-ray, Indian ink articular surface staining, Safranin-O/Fast Green staining, immunohistochemistry, hematoxylin and eosin staining of the synovial membrane, and quantitative reverse transcription-polymerase chain reaction for changes in gene expression. Micro X-ray revealed that the knee joint treated with the cHA-Dex gel was wider than those treated with cHA gel alone or saline. The cHA-Dex gel group had less Indian ink staining (indicator of cartilage fibrillation) than the cHA gel or saline injection groups. Safranin-O/Fast Green staining indicated that increased proteoglycan staining and less cartilage damage were found in the cHA-Dex gel group compared with the cHA gel or saline injection groups. Quantification of histology findings from saline, cHA gel, cHA-Dex (0.2 mg/mL) gel, cHA-Dex (0.5 mg/mL) gel, and sham groups were 5.84 ± 0.29, 4.50 ± 0.87, 3.00 ± 1.00, 2.00 ± 0.48, and 0.30 ± 0.58 (p < 0.05), respectively. A strong staining of type II collagen was found in both the cHA-Dex gel groups compared with saline group or cHA alone group. Similar result was found for the mRNA level of aggrecan and opposite result for type X collagen. Hematoxylin and eosin staining in the synovial membrane showed less synovial lining cell layers and reduced inflammatory cell infiltration in cHA-Dex gel-treated animals compared with saline or cHA only groups. Altogether, cHA-Dex gel has better chondroprotective and anti-inflammatory effects in rat surgery-induced osteoarthritis than cHA alone.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Dexamethasone/administration & dosage , Hyaluronic Acid/administration & dosage , Hydrogels/chemistry , Osteoarthritis, Knee/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Collagen Type II/genetics , Cross-Linking Reagents , Dexamethasone/pharmacology , Disease Models, Animal , Drug Synergism , Gene Expression Regulation/drug effects , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/therapeutic use , Injections, Intra-Articular , Knee Joint/drug effects , Male , Osteoarthritis, Knee/genetics , Random Allocation , Rats , Rats, Sprague-Dawley
4.
Cartilage ; 5(1): 28-36, 2014 Jan.
Article in English | MEDLINE | ID: mdl-26069682

ABSTRACT

OBJECTIVE: Cartilage injury is one of the most common disorders of synovial joints. Fresh osteochondral allografts are becoming a standard treatment; however, they are supply constrained with a potential risk of disease transmission. There are no known virucidal processes available for osteochondral allografts and most methods presently available are detrimental to cartilage. Methylene blue light treatment has been shown to be successful in the literature for viral inactivation of fresh frozen plasma. The purpose of this study was to determine the capacity of methylene blue light treatment to inactivate a panel of clinically relevant viruses inoculated onto osteochondral allografts. DESIGN: Osteochondral grafts recovered from human cadaveric knees were inoculated with one of the following viruses: bovine viral diarrhea virus (BVDV), hepatitis A virus (HAV), human immunodeficiency virus type 1 (HIV-1), porcine parvovirus (PPV), and pseudorabies virus (PrV). The samples were processed through a methylene blue light treatment, which consisted of an initial soak in nonilluminated circulating methylene blue at ambient temperature, followed by light exposure with circulating methylene blue at cool temperatures. The final titer was compared with the recovery control for the viral log reduction. RESULTS: HIV-1, BVDV, and PrV were reduced to nondetectable levels while HAV and PPV were reduced by 3.1 and 5.6 logs, respectively. CONCLUSIONS: The methylene blue light treatment was effective in reducing (a) enveloped DNA and RNA viruses to nondetectable levels and (b) nonenveloped DNA and RNA viruses of inoculated human osteochondral grafts by 3.1 to 5.6 logs. This study demonstrates the first practical method for significantly reducing viral load in osteochondral implants.

5.
Am J Sports Med ; 41(11): 2658-67, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24043472

ABSTRACT

BACKGROUND: Articular cartilage undergoes substantial age-related changes in molecular composition, matrix structure, and mechanical properties. These age-related differences between juvenile and adult cartilage manifest themselves as markedly distinct potentials for tissue repair and regeneration. PURPOSE: To compare the biological properties and tissue regeneration capabilities of juvenile and adult bovine articular cartilage. STUDY DESIGN: Controlled laboratory study. METHODS: Articular cartilage harvested from juvenile (age, 4 months) and adult (age, 6-8 years) bovine femoral condyles was cultured for 4 weeks to monitor chondrocyte migration, glycosaminoglycan content conservation, and new tissue formation. The cartilage cell density and proliferative activity were also compared. Additionally, the effects of age-related changes on cartilage gene expression were analyzed using the Affymetrix GeneChip array. RESULTS: Compared with adult cartilage, juvenile bovine cartilage demonstrated a significantly greater cell density, higher cell proliferation rate, increased cell outgrowth, elevated glycosaminoglycan content, and enhanced matrix metallopeptidase 2 activity. During 4 weeks in culture, only juvenile cartilage was able to generate new cartilaginous tissues, which exhibited pronounced labeling for proteoglycan and type II collagen but not type I collagen. With over 19,000 genes analyzed, distinctive gene expression profiles were identified. The genes mostly involved in cartilage growth and expansion, such as COL2A1, COL9A1, MMP2, MMP14, and TGFB3, were upregulated in juvenile cartilage, whereas the genes primarily responsible for structural integrity, such as COMP, FN1, TIMP2, TIMP3, and BMP2, were upregulated in adult cartilage. CONCLUSION: As the first comprehensive comparison between juvenile and adult bovine articular cartilage at the tissue, cellular, and molecular levels, the results strongly suggest that juvenile cartilage possesses superior chondrogenic activity and enhanced regenerative potential over its adult counterpart. Additionally, the differential gene expression profiles of juvenile and adult cartilage suggest possible mechanisms underlying cartilage age-related changes in their regeneration capabilities, structural components, and biological properties. CLINICAL RELEVANCE: The results of this comparative study between juvenile and adult bovine articular cartilage suggest an enhanced regenerative potential of juvenile cartilage tissue in the restoration of damaged articular cartilage.


Subject(s)
Aging/physiology , Cartilage, Articular/physiology , Chondrocytes/physiology , Glycosaminoglycans/metabolism , Regeneration , Animals , Cattle , Cell Count , Cell Proliferation , Gene Expression Profiling , Matrix Metalloproteinase 2/metabolism , Oligonucleotide Array Sequence Analysis
6.
J Biomed Mater Res B Appl Biomater ; 97(2): 355-63, 2011 May.
Article in English | MEDLINE | ID: mdl-21442745

ABSTRACT

The purpose of the study was to design and develop unique drug delivery systems with controllable multiple burst releases of drugs for treating osteoarthritis. Chondroitin sulfate (CS) was encapsulated into four types of PLGA materials, that is, PLGA 50:50, PLGA 65:35, PLGA 75:25, and PLGA 85:15. The effects of microsphere size and various combinations of blend PLGA microspheres on CS release were investigated. The cytotoxicity of the CS-encapsulated microspheres was investigated according to the ISO 10993 guideline. Our study showed that the encapsulation efficiency of CS into PLGA 50:50 microspheres varied with the size of microspheres; however, the encapsulation efficiencies of CS into PLGA microspheres were independent of the types of PLGA materials. The size of PLGA microspheres was shown to affect the rate of CS release. With the increase of microsphere size from 75-150 µm to 300-355 µm, the initial CS release decreased. Further increase in microsphere size led to an increase in the initial CS release. In addition, combination of different types of PLGA microspheres was shown to be capable of achieving multiple burst CS releases. Moreover, the CS encapsulated PLGA microspheres were shown to be non-cytotoxic. This study proved the concept of multiple burst drug releases that were achieved by encapsulating CS into different types of PLGA microspheres and delivering CS from systems consisting of mixed types of PLGA microspheres, which may be applied to treat osteoarthritis by mimicking multiple intra-joint injection of therapeutic agents.


Subject(s)
Chondroitin Sulfates/chemistry , Chondroitin Sulfates/therapeutic use , Drug Delivery Systems , Lactic Acid/chemistry , Microspheres , Osteoarthritis/drug therapy , Polyglycolic Acid/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Chondroitin Sulfates/metabolism , Drug Compounding , Humans , Lactic Acid/metabolism , Materials Testing , Microscopy, Electron, Scanning , Particle Size , Polyglycolic Acid/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer
7.
Osteoarthritis Cartilage ; 13(4): 297-309, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15780643

ABSTRACT

OBJECTIVE: The natural repair of osteochondral defects can be enhanced with biocompatible, biodegradable materials that support the repair process. It is our hypothesis that hyaluronan-based scaffolds are superior to synthetic scaffolds because they provide biological cues. We tested this thesis by comparing two hyaluronan-based scaffolds [auto cross-linked polysaccharide polymer (ACP) and HYAFF-11] to polyester-based scaffolds [poly(DL-lactic-co-glycolic acid) (PLGA) and poly(L-lactic acid) (PLLA)] with similar pore size, porosity and degradation times. DESIGN: Fifty-four rabbits received bilateral osteochondral defects. One defect received a hyaluronan-based scaffold and the contralateral defect received the corresponding polyester-based scaffold. Rabbits were euthanized 4, 12 and 20 weeks after surgery and the condyles dissected and processed for histology. RESULTS: Only ACP-treated defects presented bone at the base of the defect at 4 weeks. At 12 weeks, only defects treated with rapidly dissolving implants (ACP and PLGA) presented bone reconstitution consistently, while bone was present in only one third of those treated with slowly dissolving scaffolds (HYAFF-11 and PLLA). After 20 weeks, the articular surface of PLGA-treated defects presented fibrillation more frequently than in ACP-treated defects. The surface of defects treated with slowly dissolving scaffolds presented more cracks and fissures. CONCLUSIONS: The degradation rate of the scaffolds is critical for the repair process. Slowly dissolving scaffolds sustain thicker cartilage at the surface but, it frequently presents cracks and discontinuities. These scaffolds also delay bone formation at the base of the defects. Hyaluronan-based scaffolds appear to allow faster cell infiltration leading to faster tissue formation. The degradation of ACP leads to rapid bone formation while the slow degradation of HYAFF-11 prolongs the presence of cartilage and delays endochondral bone formation.


Subject(s)
Bone Substitutes/therapeutic use , Cartilage, Articular/injuries , Hyaluronic Acid/therapeutic use , Tissue Engineering/methods , Animals , Biocompatible Materials , Cartilage, Articular/pathology , Chemical Phenomena , Chemistry, Physical , Chondrogenesis , Hyaluronic Acid/ultrastructure , Materials Testing/methods , Microscopy, Electron, Scanning , Polyesters/chemistry , Polyesters/therapeutic use , Rabbits , Wound Healing/physiology
8.
J Cell Physiol ; 203(1): 6-14, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15389636

ABSTRACT

Wnt genes encode a number of secreted glycoproteins which are closely associated with the cell surface and the extracellular matrix. Recently, members of Wnt family have been implicated in regulating chondrocyte differentiation, but their roles in the chondrogenic process are not fully understood. To contribute to an understanding of the roles of Wnts during chondrogenesis, we have analysed the spatiotemporal expression patterns of Wnt using in vitro models for chondrogenesis of human bone marrow-derived mesenchymal stem cells (hMSCs). In chondrogenic aggregate culture system, RT-PCR analysis revealed expression of Wnt5a and Wnt4 during late chondrogenesis (days 10 and 15). Immunohistochemical analysis showed widespread distribution of Wnt5a and Wnt4 throughout the aggregates at this late phase of culture (days 14 and 21). In addition, in this aggregate culture system, immunohistochemical staining of Wnt4 and Wnt5a showed similar spatiotemporal expression patterns to that of type II collagen or type X collagen. To confirm the results obtained by immunostaining, the specificity of the anti-Wnt4 or anti-Wnt5a antibody was assessed by Western blot analysis. Of Wnt4 and Wnt5a, only Wnt5a was immunodetectable by Western blot analysis. Western blot analysis showed that Wnt5a was expressed as two different molecular weight forms of 40 and 44 kDa. Treatment with PNGase F, which removes N-linked oligosaccharides, revealed that the mass difference between these two forms could be accounted for by the N-glycosylation status of the protein. When hMSCs were seeded on a porous gelatin sponge, immunolocalization studies showed that type II collagen and type X collagen were detected particularly at the periphery at day 7 of culture. In contrast, Wnt4 and Wnt5a showed even distribution throughout the hMSC/gelatin sponge constructs. Their different spatial expression patterns suggest that Wnt4 and Wnt5a proteins are not functionally linked to type II collagen and type X collagen synthesis in in vitro chondrogenic models of hMSCs.


Subject(s)
Chondrogenesis/physiology , Hematopoietic Stem Cells/physiology , Proto-Oncogene Proteins/genetics , Blotting, Western , Cell Aggregation , Cell Culture Techniques , Cell Differentiation/physiology , Cells, Cultured , Collagen Type II/genetics , Collagen Type II/metabolism , Collagen Type X/genetics , Collagen Type X/metabolism , Extracellular Matrix/metabolism , Frizzled Receptors , Gelatin , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Humans , Immunohistochemistry , Mesoderm/cytology , Proto-Oncogene Proteins/biosynthesis , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled , Receptors, Neurotransmitter/genetics , Reverse Transcriptase Polymerase Chain Reaction , Wnt Proteins , Wnt-5a Protein , Wnt4 Protein
9.
Clin Orthop Relat Res ; (427 Suppl): S62-6, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15480077

ABSTRACT

It has been reported that demineralized bone matrix (cortical or trabecular bone) contains intrinsic cytokines. In the present study, we tested allogeneic demineralized bone matrix for its capacity to resurface osteochondral defects in a rabbit model with the assumption that the intrinsic cytokines in demineralized bone matrix will facilitate the recruitment of progenitor cells from bone marrow into the defect. It was further assumed that these intrinsic bioactive factors would modulate these cells to differentiate into osteochondrogenic lineage and, thus, functionally repair the osteochondral defect. The biocompatibility of demineralized bone matrix was first tested by loading rabbit bone-marrow-derived mesenchymal stem cells into porous demineralized trabecular bone matrix that was then cultured for 3 days. The cell growth in demineralized trabecular bone matrix was examined by scanning electron microscopy. Loaded rabbit bone-marrow-derived mesenchymal stem cells attached to the trabeculae of demineralized trabecular bone matrix; some cells appeared to be round and others were spread and contacted other cells. Allogeneic rabbit demineralized cortical bone matrix or demineralized trabecular bone matrix was implanted into a full-thickness osteochondral defect in the load-bearing area of the medial femoral condyle of young adult rabbits. At 6 and 12 weeks after surgery, gross and histological examination showed that the defects were repaired up to 95% of their depth. The repair tissue using demineralized cortical bone matrix was composed of subchondral bone and a top layer of cartilage that was smooth and integrated with the adjacent cartilage in most of the specimens. Most of the repair tissue in the defect filled with demineralized trabecular bone matrix had a fibrillated surface without integration with the adjacent cartilage. These results indicate that demineralized cortical bone matrix may be potentially useful to repair osteochondral defects by managing the host's intrinsic reparative cells.


Subject(s)
Bone Matrix , Cartilage Diseases/surgery , Cartilage, Articular , Animals , Bone Demineralization Technique , Rabbits
10.
J Exp Med ; 200(2): 123-35, 2004 Jul 19.
Article in English | MEDLINE | ID: mdl-15263023

ABSTRACT

Here a new, intrinsically pluripotent, CD45-negative population from human cord blood, termed unrestricted somatic stem cells (USSCs) is described. This rare population grows adherently and can be expanded to 10(15) cells without losing pluripotency. In vitro USSCs showed homogeneous differentiation into osteoblasts, chondroblasts, adipocytes, and hematopoietic and neural cells including astrocytes and neurons that express neurofilament, sodium channel protein, and various neurotransmitter phenotypes. Stereotactic implantation of USSCs into intact adult rat brain revealed that human Tau-positive cells persisted for up to 3 mo and showed migratory activity and a typical neuron-like morphology. In vivo differentiation of USSCs along mesodermal and endodermal pathways was demonstrated in animal models. Bony reconstitution was observed after transplantation of USSC-loaded calcium phosphate cylinders in nude rat femurs. Chondrogenesis occurred after transplanting cell-loaded gelfoam sponges into nude mice. Transplantation of USSCs in a noninjury model, the preimmune fetal sheep, resulted in up to 5% human hematopoietic engraftment. More than 20% albumin-producing human parenchymal hepatic cells with absence of cell fusion and substantial numbers of human cardiomyocytes in both atria and ventricles of the sheep heart were detected many months after USSC transplantation. No tumor formation was observed in any of these animals.


Subject(s)
Cell Line , Fetal Blood/cytology , Placenta/blood supply , Stem Cells/cytology , Adipocytes/cytology , Albumins/metabolism , Animals , Blotting, Western , Bone and Bones/cytology , Cell Culture Techniques , Cell Differentiation , Cell Division , Cell Transplantation , Cord Blood Stem Cell Transplantation , Femur/metabolism , Flow Cytometry , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/cytology , Hippocampus/cytology , Humans , Immunophenotyping , Leukocyte Common Antigens/biosynthesis , Leukocytes, Mononuclear/metabolism , Myocardium/cytology , Myocytes, Cardiac/metabolism , Neurotransmitter Agents , Osteoblasts/metabolism , Phenotype , Polymerase Chain Reaction , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Sheep , Time Factors , Umbilical Veins
11.
Tissue Eng ; 8(5): 827-37, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12459061

ABSTRACT

Articular cartilage has limited capacity for repair. In the present study, tissue-engineered two-phase composite material was used for the repair of osteochondral defects in young adult rabbit knee. This composite material is composed of an injectable calcium phosphate (ICP) and a hyaluronan (HA) derivate of either ACP or HYAFF 11 sponge. The osteochondral defect, 3 mm in diameter and 3 mm deep, was created in the weight-bearing region of the medial femoral condyle. The bone portion of the defect was first filled with ICP to a level approximately 1 mm below the articular surface. HA sponge (3 mm in diameter and 1-1.2 mm thick), with or without loading of autologous bone marrow-derived progenitor cells (MPCs), was then inserted into the defect on top of the ICP as it hardened. Animals were allowed free cage activity postoperatively, and killed 4 or 12 weeks (for the HYAFF 11 sponge group) after the surgery. At 4 weeks, histological examination showed that the defect was filled up to 90-100% of its depth. Whitish repair tissue on the top appeared to be integrated with the surrounding articular cartilage. Four distinct zones of repair tissue were identified: a superficial layer, a chondroid tissue layer, an interface between HA sponge and ICP, and the ICP material. Evidence of extensive osteoclastic and osteoblastic activities was observed in the bone tissue surrounding the defect edge and in ICP material. By 12 weeks, the zonal features of the repair tissue became more distinct; chondrocytes were arranged in a columnar array, and a calcified layer of cartilage was formed beneath the chondroid tissue in some specimens. The healing tissue of the HA sponge material loaded with MPCs had higher cellular density and better integration with the surrounding cartilage than HA sponge material not loaded with MPCs. This study suggests that using a two-phase composite graft may hold potential for the repair of osteochondral defects by providing mechanical support that mimicks subchondral bone, while also providing a chondrogenic scaffold for the top cartilage repair.


Subject(s)
Biocompatible Materials , Bioprosthesis , Bone and Bones/injuries , Calcium Phosphates/metabolism , Cartilage/injuries , Hyaluronic Acid/metabolism , Animals , Bone and Bones/surgery , Cartilage/surgery , Fracture Fixation/methods , Lower Extremity , Rabbits
12.
Tissue Eng ; 8(2): 333-47, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12031121

ABSTRACT

The natural repair of osteochondral defects can be enhanced with biocompatible, biodegradable and bioactive materials that provide structural support and molecular cuing to stimulate repair. Since bone marrow contains osteochondral progenitor cells and bioactive agents, it is hypothesized that the combination of scaffold and bone marrow would be a superior composite material for osteochondral repair. This hypothesis will be tested by comparing the outcome of osteochondral defects filled with a fibronectin-coated hyaluronan-based sponge (ACP) with or without autologous bone marrow. Thirty-three 4-month-old rabbits received 3-mm diameter osteochondral defects that were then filled with ACP loaded or not with autologous bone marrow. Rabbits were sacrificed at 2, 3, 4, 12, and 24 weeks after surgery and the condyles processed for histologic and immunohistochemical evaluation. The defects were graded with a histologic scoring scale. Except for the 3-week specimens, the histologic appearance of the defects was similar in both groups. Four weeks after surgery, the defects were filled with bone with a top layer of cartilage well integrated with the adjacent cartilage. Twelve and 24 weeks after surgery, the defects again showed bone filling. The primary difference between the 4-week samples and the 12- and 24-week samples was that the layer of cartilage that appeared to be thinner than the adjacent cartilage. At each harvest time, the overall histologic scores of the specimens did not reveal statistical differences between the treatment groups. However, as revealed by the results of the 3-week sacrifices, bone marrow loading appeared to accelerate the first stages of the repair process. The fibronectin-coated hyaluronan-based scaffold appears to organize the natural response and facilitate the integration of the neo-cartilage with the adjacent tissue. The fundamental tissue engineering principles derived from this study should provide guidelines for the development of comparable clinical reconstructive therapies.


Subject(s)
Biocompatible Materials , Bone Marrow Transplantation , Cartilage, Articular/pathology , Hyaluronic Acid , Transplantation, Autologous , Animals , Bone Marrow Cells/pathology , Bone Marrow Transplantation/methods , Cartilage, Articular/injuries , Cell Differentiation , Fibronectins , Immunohistochemistry , Microspheres , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL
...