Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
Osteoporos Int ; 21(7): 1133-49, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19960185

ABSTRACT

UNLABELLED: Osteoporosis, a skeletal disorder characterized by a reduction in bone strength, increases fracture risk. Primary osteoporosis is usually a result of reduced bone mineral density as a consequence of natural aging. Secondary osteoporosis is usually a result of a disease, such as cystic fibrosis, or medical treatment, such as corticosteroids or cancer treatment. INTRODUCTION: Currently, ten million Americans are osteoporotic and an additional 34 million have the precursor condition, osteopenia. Osteoporosis leads to 1.5 million fractures and 500,000 hospitalizations annually. Osteoporosis-related fractures increase mortality and reduce quality of life. Calcitriol, the active form of vitamin D, regulates intestinal calcium absorption, among other actions. During the past four decades, many clinical trials investigating the effect of calcitriol on bone loss have been performed. METHODS: We conducted a systematic qualitative review of clinical trials that assessed calcitriol for the treatment of osteoporosis and bone loss. In these clinical trials, calcitriol was used as a monotherapy and in combination with other therapeutic bone agents. RESULTS AND CONCLUSION: Studies using calcitriol monotherapy, although not conclusive, found that calcitriol slowed the rate of bone loss in a variety of populations. Calcitriol in combination with other therapeutic bone agents was shown to have additional bone-preserving effects when compared to the use of therapeutic bone agents alone. A common side-effect of calcitriol therapy was hypercalcemia and hypercalciuria, but the degree of hypercalcemia was mild. Recent research found that intermittent dosing can reduce hypercalcemia rates. Calcitriol, alone or in combination with other agents, should be considered for the therapy of osteoporosis.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Calcitriol/therapeutic use , Osteoporosis/drug therapy , Osteoporotic Fractures/prevention & control , Biomarkers/metabolism , Bone Density/drug effects , Drug Administration Schedule , Drug Therapy, Combination , Female , Humans , Male , Osteoporosis/physiopathology , Osteoporotic Fractures/physiopathology , Treatment Outcome
2.
Oncogene ; 27(25): 3605-14, 2008 Jun 05.
Article in English | MEDLINE | ID: mdl-18223689

ABSTRACT

The Runx family of transcription factors regulate cell growth and differentiation, and control the expression of target genes involved in cell fate decisions. We examined the role of the bone-related member of this family, Runx2, in regulating apoptosis via modulation of the Bcl2 family of genes in the osteosarcoma cell line Saos2. Our data demonstrate that Runx2 directly binds to two Runx-specific regulatory elements on the human bax promoter thereby inducing Bax expression. Furthermore, bone morphogenetic protein-induced or vector-mediated expression of Runx2 resulted in upregulation of Bax expression, and subsequent increased sensitivity of Saos2 cells to apoptosis. Finally, the observed upregulation of Bax expression and increased apoptosis were Runx2 dependent as Runx2 loss of function abrogated these effects. Our study provides the first evidence for Bax as a direct target of Runx2, suggesting that Runx2 may act as a proapoptotic factor in osteosarcoma cells.


Subject(s)
Apoptosis , Bone Neoplasms/metabolism , Core Binding Factor Alpha 1 Subunit/physiology , Gene Expression Regulation, Neoplastic , Osteosarcoma/metabolism , bcl-2-Associated X Protein/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Gene Expression Regulation , Humans , Models, Biological , Mutagenesis, Site-Directed , Promoter Regions, Genetic , Transcriptional Activation
3.
J Clin Pathol ; 58(3): 328-30, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15735172

ABSTRACT

BACKGROUND: Dominant negative inhibition of nuclear factor kappa B (NF kappa B) signalling activity in a human osteosarcoma cell line (Saos2) results in malignant reversion and the induction of the osteoblast differentiating transcription factor, Runx2/Cbfa1. This observation suggests that there is an inverse relation between a transcription factor associated with malignant progression and chemoresistance (NF kappa B) and an osteoblast differentiating transcription factor (Runx2/Cbfa1). AIMS: To assess and correlate Runx2/Cbfa1 and NF kappa B (p65) immunoreactivity in human osteosarcoma. METHODS: Runx2/Cbfa1 and NFkappaB (p65) immunoreactivity was assessed on 11 paraffin wax embedded archival specimens of human primary osteosarcoma by standard immunohistochemical methods and scored on a scale of 0-3. A Pearson correlation analysis between Runx2/Cbfa1 and NF kappa B (p65) scores was established. RESULTS: Runx2/Cbfa1 was expressed constitutively in all pathology specimens of human osteosarcoma. Of note, a chondroblastic osteosarcoma showed the highest Runx2/Cbfa1 immunoreactivity. A Pearson correlation did not support an inverse correlation between Runx2/Cbfa1 and NF kappa B (p65) scores (r = 0.57) in human osteosarcoma. CONCLUSION: Runx2/Cbfa1 immunoreactivity does not inversely correlate with NF kappa B immunoreactivity, and thus cannot serve as an indirect measure of NF kappa B activity or an independent predictive or prognostic indicator.


Subject(s)
Biomarkers, Tumor/metabolism , Bone Neoplasms/metabolism , DNA-Binding Proteins/metabolism , NF-kappa B/metabolism , Osteosarcoma/metabolism , Transcription Factors/metabolism , Bone Neoplasms/pathology , Core Binding Factor Alpha 1 Subunit , Humans , Immunoenzyme Techniques , Neoplasm Proteins/metabolism , Osteosarcoma/pathology , Transcription Factor AP-2
6.
J Cell Physiol ; 188(3): 343-51, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11473361

ABSTRACT

PTHrP regulates the rate of chondrocyte differentiation during endochondral bone formation. The expression of PTHrP and its regulation by TGF-beta, BMP-2, and PTHrP was examined in upper sternal chondrocytes following 1, 3, and 5 days of continuous treatment. While TGF-beta stimulated the expression of PTHrP (5-fold), PTHrP caused a slight inhibition, and BMP-2 markedly inhibited PTHrP mRNA expression. The effect of these factors on PTHrP expression was not simply related to the maturational state of the cells, since BMP-2 increased, while both PTHrP and TGF-beta decreased the expression of type X collagen. TGF-beta isoforms 1, 2, and 3 all stimulated PTHrP expression. Signaling events involved in the induction of PTHrP by TGF-beta were further evaluated in a PTHrP-promoter CAT construct. The effect of TGF-beta, BMP-2, and PTHrP on the PTHrP-promoter paralleled their effects on mRNA expression, with TGF-beta significantly increasing CAT activity, BMP-2 decreasing CAT activity, and PTHrP having a minimal effect. Co-transfection of the TGF-beta signaling molecule, Smad 3, mimicked the effect of TGF-beta (induction of PTHrP promoter), while dominant negative Smad 3 inhibited the induction of the PTHrP promoter by TGF-beta. Furthermore, infection with a Smad 3-expressing retrovirus mimicked the effects of exogenously added TGF-beta, and induced PTHrP mRNA expression in the infected chondrocyte culture. In contrast, a dominant negative Smad 3 completely inhibited PTHrP promoter stimulation by TGF-beta, but only partially blocked the effect of TGF-beta on PTHrP mRNA synthesis. These findings demonstrate that PTHrP is expressed in chondrocytes undergoing endochondral ossification, and show regulation, at least in part, by TGF-beta through Smad mediated signaling events.


Subject(s)
Chondrocytes/metabolism , DNA-Binding Proteins/metabolism , Protein Biosynthesis , Signal Transduction/physiology , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation/physiology , Cells, Cultured , Chick Embryo , Chickens , Chondrocytes/cytology , Chondrocytes/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/pharmacology , Gene Expression/drug effects , Gene Expression/physiology , Parathyroid Hormone-Related Protein , Promoter Regions, Genetic/drug effects , Proteins/genetics , Proteins/pharmacology , RNA, Messenger/metabolism , Signal Transduction/drug effects , Smad3 Protein , Sternum , Trans-Activators/genetics , Trans-Activators/pharmacology , Transcriptional Activation/drug effects , Transfection , Transforming Growth Factor beta/pharmacology
7.
J Bone Joint Surg Am ; 83(7): 1057-61, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11451976

ABSTRACT

BACKGROUND: Pentoxifylline (Trental) is a methylxanthine-derivative drug that has been used for more than twenty years in the treatment of peripheral vascular disease. Pentoxifylline is also a potent inhibitor of tumor necrosis factor-alpha (TNF-alpha) secretion, both in vitro and in vivo, and has demonstrated efficacy in the treatment of certain animal and human inflammatory diseases. Pentoxifylline has a potential therapeutic role in the treatment of aseptic loosening of total joint replacement components because it inhibits TNF-alpha secretion by particle-stimulated human peripheral blood monocytes. The purpose of our study was to determine whether the particle-stimulated secretion of TNF-alpha by peripheral blood monocytes was inhibited in volunteers who had received pentoxifylline orally. METHODS: Human peripheral blood monocytes were harvested from eight healthy volunteers and were exposed to three different concentrations of titanium particles or to 500 ng/mL of lipopolysaccharide as a positive control. The same volunteers were then given pentoxifylline (400 mg, five times per day) for seven days. Their peripheral blood monocytes were again isolated and exposed to experimental conditions, and the TNF-alpha levels were measured. RESULTS: The peripheral blood monocytes from all eight volunteers showed a significant reduction in TNF-alpha release following oral treatment with pentoxifylline. This reduction was observed at exposures of 10(7) and 10(6) titanium particles/mL and in the lipopolysaccharide-treated group, but not at 10(5) particles/mL. CONCLUSIONS: To our knowledge, this is the first study to demonstrate the ability of an oral drug to decrease the release of TNF-alpha from human peripheral blood monocytes exposed ex vivo to particle debris. TNF-alpha is involved in the pathogenesis of osteolysis and subsequent loosening of total joint arthroplasty components. The ability to suppress the release of TNF-alpha in patients with a total joint replacement may help to control osteolysis and to reduce the development of aseptic loosening. This effect could increase implant longevity and decrease the need for revision arthroplasty.


Subject(s)
Monocytes/metabolism , Pentoxifylline/administration & dosage , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/drug effects , Administration, Oral , Adult , Analysis of Variance , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Equipment Failure Analysis , Female , Humans , Joint Prosthesis , Lipopolysaccharides/pharmacology , Male , Monocytes/drug effects , Probability , Reference Values , Titanium/pharmacology
8.
Radiat Res ; 155(6): 847-57, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11352768

ABSTRACT

Radiation therapy plays an important role as part of the multimodality treatment for a number of childhood malignancies. Dose-limiting complications of radiotherapy include skeletal abnormalities and disturbances in skeletal development within the irradiated field. The current study was undertaken to investigate the molecular mechanisms involved in radiation-induced arrest of bone growth. Our hypotheses were: (1) Expression of autocrine growth factors that regulate chondrocyte proliferation is inhibited by radiation in a specific pattern; (2) the disparity in radiosensitivity of growth plate chondrocytes and epiphyseal chondrocytes is due to differential modulation of autocrine growth factor expression by radiation. Given the important role these cells play in skeletal growth and development, we examined the comparative effects of radiation on expression of specific mitogenic growth factors in growth plate chondrocytes. The effect of radiation on the expression of autocrine/paracrine growth factors was examined in an established avian model of epiphyseal growth plate maturation. Exposure of growth plate chondrocytes to radiation resulted in a specific pattern of biochemical and morphological alterations that were dependent on dose and were progressive over time. While radiation did not affect the mRNA expression of some of the autocrine and paracrine factors important in endochondral ossification (such as FGF2 and TGFB isoforms), it did lead to a decrease in the mRNA expression of PTHrP, a critically important mitogen in growth plate chondrocytes, and a dose-dependent decrease in the PTH/PTHrP receptor mRNA. Interestingly, PTHrP mRNA levels were not affected in irradiated epiphyseal chondrocytes, the main source of PTHrP. Given evidence indicating a role for intracellular calcium levels in regulating PTHrP expression, basal calcium levels in irradiated growth plate chondrocytes and epiphyseal chondrocytes were examined 24 h after treatment. While cytosolic calcium levels were significantly higher in irradiated growth plate chondrocytes, they were not significantly affected in irradiated epiphyseal chondrocytes. The importance of calcium in mediating radiation damage to growth plate chondrocytes was further demonstrated by the finding that the addition of 4.0 mM EGTA (a calcium chelator) to the cell cultures before irradiation prevented the decrease in PTHrP mRNA levels. Since PTHrP up-regulates BCL2 levels and prevents growth plate chondrocyte maturation and apoptosis, BCL2 mRNA levels were examined in irradiated growth plate chondrocytes, and a dose-dependent decrease was found. An increase in apoptosis was further confirmed by a fivefold increase in caspase 3 levels in irradiated growth plate chondrocytes. The results of the current study suggest that radiation may interfere with proliferation of growth plate chondrocytes in part by causing an increase in cytosolic calcium levels which in turn leads to a decrease in PTHrP mRNA. Growth plate chondrocyte PTHrP receptor mRNA expression is also inhibited by radiation, further decreasing PTHrP signaling. Despite subtle differences between the chick and mammalian growth plates, further studies should provide an enhanced understanding of the mechanism(s) of radiation injury to the growth plate, as well as possibilities for new therapeutic strategies to protect the growing skeleton from the detrimental effects of radiotherapy.


Subject(s)
Fibroblast Growth Factor 2/physiology , Growth Plate/radiation effects , Transforming Growth Factor beta/physiology , Animals , Base Sequence , Chickens , DNA Primers , Growth Plate/metabolism , Growth Plate/physiology , In Vitro Techniques , Parathyroid Hormone-Related Protein , Proteins/genetics , RNA, Messenger/genetics , Radiotherapy/adverse effects , Receptor, Parathyroid Hormone, Type 1 , Receptors, Parathyroid Hormone/metabolism , Reverse Transcriptase Polymerase Chain Reaction
9.
J Bone Miner Res ; 16(4): 660-70, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11315993

ABSTRACT

Aseptic loosening is a major complication of prosthetic joint surgery and is manifested as chronic inflammation, pain, and osteolysis at the bone implant interface. The osteolysis is believed to be driven by a host inflammatory response to wear debris generated from the implant. In our current study, we use a selective inhibitor (celecoxib) of cyclo-oxygenase 2 (COX-2) and mice that lack either COX-1 (COX-1-/-) or COX-2 (COX-2-/-) to show that COX-2, but not COX-1, plays an important role in wear debris-induced osteolysis. Titanium (Ti) wear debris was implanted surgically onto the calvaria of the mice. An intense inflammatory reaction and extensive bone resorption, which closely resembles that observed in patients with aseptic loosening, developed within 10 days of implantation in wild-type and COX-1-/- mice. COX-2 and prostaglandin E2 (PGE2) production increased in the calvaria and inflammatory tissue overlying it after Ti implantation. Celecoxib (25 mg/kg per day) significantly reduced the inflammation, the local PGE2 production, and osteolysis. In comparison with wild-type and COX-1-/- mice, COX-2-/- mice implanted with Ti had a significantly reduced calvarial bone resorption response, independent of the inflammatory response, and significantly fewer osteoclasts were formed from cultures of their bone marrow cells. These results provide direct evidence that COX-2 is an important mediator of wear debris-induced osteolysis and suggests that COX-2 inhibitors are potential therapeutic agents for the prevention of wear debris-induced osteolysis.


Subject(s)
Isoenzymes/physiology , Osteolysis/enzymology , Prostaglandin-Endoperoxide Synthases/physiology , Prosthesis Failure , Animals , Bone Resorption/etiology , Celecoxib , Cells, Cultured/drug effects , Crosses, Genetic , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/biosynthesis , Female , Interleukin-1/biosynthesis , Interleukin-6/biosynthesis , Isoenzymes/antagonists & inhibitors , Isoenzymes/deficiency , Isoenzymes/genetics , Macrophage Activation , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Osteoclasts/pathology , Osteolysis/etiology , Osteolysis/pathology , Prostaglandin-Endoperoxide Synthases/deficiency , Prostaglandin-Endoperoxide Synthases/genetics , Prostheses and Implants , Pyrazoles , Skull , Sulfonamides/pharmacology , Titanium , Tumor Necrosis Factor-alpha/biosynthesis
10.
Clin Orthop Relat Res ; (385): 36-45, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11302324

ABSTRACT

Osteoarthritis affects more patients than almost any other musculoskeletal disorder. The number of patients suffering joint pain and stiffness as a result of this disease will increase rapidly in the next decade. Although operative treatments of patients with osteoarthritis will continue to improve and the number of operative procedures will increase slightly in the next decade, only a small fraction of the patients with osteoarthritis will require operative procedures. The most pressing healthcare need for the majority of patients with osteoarthritis is nonoperative care that helps relieve symptoms and improve function, and in some instances slows progression. In rare instances, the symptoms of osteoarthritis improve spontaneously, but most patients need nonoperative care for decades. Orthopaedists need to improve their ability to provide nonoperative care for patients with osteoarthritis. They should be skilled in the early diagnosis of osteoarthritis and in the use of current common nonoperative treatments including patient education, activity modification, shoe modifications, braces, oral analgesics, oral nonsteroidal antiinflammatory medications, oral dietary supplements, and intraarticular injections. Furthermore, orthopaedists should be prepared to incorporate new nonoperative treatments for patients with osteoarthritis into their practice.


Subject(s)
Osteoarthritis/therapy , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Chondroitin Sulfates/therapeutic use , Disease Progression , Exercise , Glucosamine/therapeutic use , Humans , Hyaluronic Acid/therapeutic use , Orthotic Devices , Osteoarthritis/etiology , Osteoarthritis, Knee/therapy
11.
Clin Orthop Relat Res ; (385): 57-67, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11302327

ABSTRACT

Osteoporosis is an increasingly prevalent disease among the aging population, and osteoporotic features account for substantial morbidity, mortality, and healthcare costs associated with this disease. Because the disease is silent until a fracture occurs, the orthopaedic surgeon often may be the physician in the best position to establish the diagnosis and consider the initiation of appropriate treatment. Historically, osteoporosis has been underdiagnosed and treated, but new methods allow accurate diagnosis using bone densitometry, and a range of effective treatment options that can reduce fracture risk. Diagnosis and treatment of osteoporosis fits readily into an efficient algorithmic approach in the office practice of orthopaedics. Orthopaedic surgeons can play a major role in improving the treatment of osteoporosis and decreasing morbidity from this disease. In addition, this can augment the office practice of orthopaedics with a large yet relatively underserved patient population. Finally, densitometry services can provide modest supplemental revenue sources for an orthopaedic practice.


Subject(s)
Orthopedics , Osteoporosis/therapy , Absorptiometry, Photon , Diphosphonates/pharmacology , Estrogen Replacement Therapy , Female , Humans , Middle Aged , Osteoporosis/drug therapy , Osteoporosis, Postmenopausal/prevention & control , Physician's Role
12.
J Biol Chem ; 276(15): 11639-47, 2001 Apr 13.
Article in English | MEDLINE | ID: mdl-11136722

ABSTRACT

During the process of differentiation, chondrocytes integrate a complex array of signals from local or systemic factors like parathyroid hormone-related peptide (PTHrP), Indian hedgehog, bone morphogenetic proteins and transforming growth factor beta. While PTHrP is known to be a critical regulator of chondrocyte proliferation and differentiation, the signaling pathways through which this factor acts remain to be elucidated. Here we show that both cAMP response element-binding protein (CREB) and AP-1 activation are critical to PTHrP signaling in chondrocytes. PTHrP treatment leads to rapid CREB phosphorylation and activation, while CREB DNA binding activity is constitutive. In contrast, PTHrP induces AP-1 DNA binding activity through induction of c-Fos protein expression. PTHrP activates CRE and TRE reporter constructs primarily through PKA-mediated signaling events. Both signaling pathways were found to be important mediators of PTHrP effects on chondrocyte phenotype. Alone, PTHrP suppresses maturation and stimulates proliferation of the chondrocyte cultures. However, in the presence of dominant negative inhibitors of CREB and c-Fos, these PTHrP effects were suppressed, and chondrocyte maturation was accelerated. Moreover, in combination, the effects of dominant negative c-Fos and CREB are synergistic, suggesting interaction between these signaling pathways during chondrocyte differentiation.


Subject(s)
Cell Differentiation/physiology , Chondrocytes/cytology , Cyclic AMP Response Element-Binding Protein/metabolism , Proteins/physiology , Signal Transduction/physiology , Transcription Factor AP-1/metabolism , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Parathyroid Hormone-Related Protein , Phosphorylation , Protein Kinase C/metabolism
13.
Cancer Res ; 60(23): 6557-62, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11118032

ABSTRACT

To investigate the role of the transcription factor nuclear factor kappaB (NFkappaB) in tumor metastasis, we generated a murine lung alveolar carcinoma cell line (Line 1) defective in NFkappaB-signaling by retroviral delivery of a dominant negative inhibitor of NFkappaB. The NFkappaB signal blockade resulted in the down-regulation of prometastatic matrix metalloproteinase 9, a urokinase-like plasminogen activator, and heparanase and reciprocal up-regulation of antimetastatic tissue inhibitors of matrix metalloproteinases 1 and 2 and plasminogen activator inhibitor 2. NFkappaB signal blockade did not affect tumor cell proliferation in vitro or in vivo but prevented intravasation of tumor cells in an in vivo chick chorioallantoic membrane model of metastasis as well as spontaneous metastasis in a murine model. These findings suggest that NFkappaB plays a central and specific role in the regulation of tumor metastasis and may be an important therapeutic target for development of antimetastatic cancer treatments.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , NF-kappa B/physiology , Neoplasm Metastasis/genetics , Adenocarcinoma, Bronchiolo-Alveolar/enzymology , Adenocarcinoma, Bronchiolo-Alveolar/genetics , Adenocarcinoma, Bronchiolo-Alveolar/secondary , Allantois/blood supply , Animals , Cell Division/physiology , Chick Embryo , Chorion/blood supply , Down-Regulation , Glucuronidase/biosynthesis , Glucuronidase/genetics , Humans , I-kappa B Proteins/genetics , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred BALB C , NF-kappa B/antagonists & inhibitors , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-2/biosynthesis , Tissue Inhibitor of Metalloproteinase-2/genetics , Transfection , Up-Regulation , Urokinase-Type Plasminogen Activator/biosynthesis , Urokinase-Type Plasminogen Activator/genetics
14.
Endocrinology ; 141(12): 4728-35, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11108288

ABSTRACT

Transforming growth factor-beta (TGF-beta) is a multifunctional regulator of a variety of cellular functions, including proliferation, differentiation, matrix synthesis, and apoptosis. In growth plate chondrocytes, TGF-beta slows the rate of maturation. Because the current paradigm of TGF-beta signaling involves Smad proteins as downstream regulators of target genes, we have characterized their role as mediators of TGF-beta effects on chondrocyte maturation. Both Smad2 and 3 translocated to the nucleus upon TGF-beta1 signaling, but not upon BMP-2 signaling. Cotransfection experiments using the TGF-beta responsive and Smad3 sensitive p3TP-Lux luciferase reporter demonstrated that wild-type Smad3 potentiated, whereas dominant negative Smad3 inhibited TGF-beta1 induced luciferase activity. To confirm the role of Smad2 and 3 as essential mediators of TGF-beta1 effects on chondrocyte maturation, we overexpressed both wild-type and dominant negative Smad2 and 3 in virally infected chondrocyte cultures. Overexpression of both wild-type Smad2 and 3 potentiated the inhibitory effect of TGF-beta on chondrocyte maturation, as determined by colx and alkaline phosphatase activity, whereas dominant negative Smad2 and 3 blocked these effects. Wild-type and dominant negative forms of Smad3 had more pronounced effects than Smad2. Our results define Smad2 and 3 as key mediators of the inhibitory effect of TGF-beta1 signaling on chondrocyte maturation.


Subject(s)
Chondrocytes/cytology , DNA-Binding Proteins/physiology , Trans-Activators/physiology , Transforming Growth Factor beta/pharmacology , Alkaline Phosphatase/metabolism , Animals , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Nucleus/metabolism , Chick Embryo , Chondrocytes/drug effects , Collagen/genetics , DNA-Binding Proteins/genetics , Gene Expression , Luciferases/genetics , RNA, Messenger/analysis , Signal Transduction , Smad2 Protein , Smad3 Protein , Trans-Activators/genetics , Transfection
15.
J Orthop Res ; 18(3): 472-80, 2000 May.
Article in English | MEDLINE | ID: mdl-10937636

ABSTRACT

Due to irreversible joint destruction caused by the various arthritides, more than 400,000 total joint arthroplasties are performed each year in the United States. As many as 20% of these require revision surgery because of aseptic loosening. The current paradigm to explain aseptic loosening is that wear debris generated from the prosthesis stimulates the release of proinflammatory cytokines (i.e., tumor necrosis factor-alpha and interleukins 1 and 6) following phagocytosis by resident macrophages. These cytokines, in turn, initiate an inflammatory response, with the development of an erosive pannus that stimulates bone resorption by osteoclasts. In support of this model, we have previously shown that human monocytes produce large quantities of tumor necrosis factor-alpha in response to titanium particles in vitro. In the current study, we characterized the role of tumor necrosis factor-alpha/nuclear transcription factor-kappaB signaling in the proinflammatory response to titanium particles in vitro and in vivo. Using the mouse macrophage cell line J774, we showed that these cells produce an amount of tumor necrosis factor-alpha in response to titanium particles similar to that produced by human peripheral blood monocytes. The production of tumor necrosis factor-alpha was preceded by a drop in cellular levels of inhibitory factor-kappaBalpha protein and translocation of p50/p65 nuclear transcription factor-KB to the nucleus 30 minutes after stimulation. Levels of tumor necrosis factor-alpha and inhibitory factor-kappaBalpha mRNA increased 30 minutes after stimulation, consistent with the activation of nuclear transcription factor-kappaB. Interleukin-6 mRNA was first seen 4 hours after the addition of the titanium particles, indicating that the production of this cytokine is secondary to the immediate nuclear transcription factor-kappaB response. To test the relevance of tumor necrosis factor-alpha/nuclear transcription factor-kappaB signaling in response to titanium particles in vivo, we adopted an animal model in which the particles were surgically implanted on the calvaria of mice. The animals displayed a dramatic histological response to the debris, with the formation of fibrous tissue and extensive bone resorption after only 1 week. With use of immunohistochemistry and tartrate-resistant acid phosphatase staining, tumor necrosis factor-alpha and osteoclasts were readily detected at the site of inflammation and bone resorption in the calvaria of the treated mice. By testing mice that genetically over-produce tumor necrosis factor-alpha (hTNFalpha-Tg), those defective in tumor necrosis factor-alpha signaling (TNF-RI-/-), and those that are nuclear transcription factor-kappaB1-deficient (NFkappaB1-/-), we evaluated the importance of tumor necrosis factor-alpha/nuclear transcription factor-kappaB signaling in the biological processes responsible for aseptic loosening. The hTNFalpha-Tg mice had a grossly exaggerated response, the TNF-RI(-/-) mice showed little evidence of inflammation or bone resorption, and the nuclear transcription factor-kappaB1(-/-) mice had an inflammatory response without bone resorption. On the basis of these results, we propose a model for periprosthetic osteolysis in which wear debris particles are phagocytosed by macrophages, resulting in the activation of nuclear transcription factor-kappaB and the production of tumor necrosis factor-alpha. Tumor necrosis factor-alpha directly induces fibroblast proliferation and tissue fibrosis and recruits or activates, or both, osteoclasts to resorb adjacent bone.


Subject(s)
Arthroplasty, Replacement/adverse effects , NF-kappa B/physiology , Osteolysis/etiology , Tumor Necrosis Factor-alpha/physiology , Animals , Cell Line , Female , Immunohistochemistry , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred CBA , Osteoclasts/physiology , Signal Transduction , Titanium/pharmacology
16.
Mol Cell Biol ; 20(17): 6496-507, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10938126

ABSTRACT

Multiple inositol polyphosphate phosphatase (Minpp1) metabolizes inositol 1,3,4,5,6-pentakisphosphate (InsP(5)) and inositol hexakisphosphate (InsP(6)) with high affinity in vitro. However, Minpp1 is compartmentalized in the endoplasmic reticulum (ER) lumen, where access of enzyme to these predominantly cytosolic substrates in vivo has not previously been demonstrated. To gain insight into the physiological activity of Minpp1, Minpp1-deficient mice were generated by homologous recombination. Tissue extracts from Minpp1-deficient mice lacked detectable Minpp1 mRNA expression and Minpp1 enzyme activity. Unexpectedly, Minpp1-deficient mice were viable, fertile, and without obvious defects. Although Minpp1 expression is upregulated during chondrocyte hypertrophy, normal chondrocyte differentiation and bone development were observed in Minpp1-deficient mice. Biochemical analyses demonstrate that InsP(5) and InsP(6) are in vivo substrates for ER-based Minpp1, as levels of these polyphosphates in Minpp1-deficient embryonic fibroblasts were 30 to 45% higher than in wild-type cells. This increase was reversed by reintroducing exogenous Minpp1 into the ER. Thus, ER-based Minpp1 plays a significant role in the maintenance of steady-state levels of InsP(5) and InsP(6). These polyphosphates could be reduced below their natural levels by aberrant expression in the cytosol of a truncated Minpp1 lacking its ER-targeting N terminus. This was accompanied by slowed cellular proliferation, indicating that maintenance of cellular InsP(5) and InsP(6) is essential to normal cell growth. Yet, depletion of cellular inositol polyphosphates during erythropoiesis emerges as an additional physiological activity of Minpp1; loss of this enzyme activity in erythrocytes from Minpp1-deficient mice was accompanied by upregulation of a novel, substitutive inositol polyphosphate phosphatase.


Subject(s)
Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/physiology , 3T3 Cells , Animals , Blotting, Northern , Cell Differentiation , Cell Division , Cells, Cultured , Chondrocytes/cytology , Chromatography, High Pressure Liquid , Cytosol/metabolism , Embryo, Mammalian/metabolism , Endoplasmic Reticulum/metabolism , Fibroblasts/metabolism , Fluorescent Antibody Technique , In Situ Hybridization , Inositol Phosphates/metabolism , Mice , Mice, Transgenic , Models, Genetic , Phenotype , Phosphoric Monoester Hydrolases/biosynthesis , Phytic Acid/metabolism , RNA, Messenger/metabolism , Recombination, Genetic , Time Factors , Up-Regulation
17.
Instr Course Lect ; 49: 495-502, 2000.
Article in English | MEDLINE | ID: mdl-10829203

ABSTRACT

A significant amount of evidence exists suggesting beneficial effects of intra-articular hyaluronate preparations in treating the symptoms of OA. The beneficial effects of these compounds tend to persist for several months after cessation of therapy, unlike anti-inflammatory drug effects. In addition, several clinical trials comparing these agents to anti-inflammatory medications indicate efficacy of symptom improvement equivalent to that of the anti-inflammatories. A few clinical studies have failed to demonstrate an effect, and clearly much work remains to be done in this area to fully determine rational treatment strategies with these agents. The symptom improvement is generally moderate, and additive effects of these agents with anti-inflammatories have not been demonstrated. Adverse local reactions have been reported in some clinical series of hyaluronate injections. Although the invasive nature of the procedure is a drawback, overall it appears to be well tolerated and is probably not harmful to articular cartilage, a major advantage over the widely practiced introduction of intra-articular steroids. Documentation of true chondroprotective effects or alteration of the natural history of cartilage degeneration by these agents is lacking in the clinical literature and awaits further study. Cost effectiveness of this therapy versus other treatments, including the costs of treatment of side effects, also requires further rigorous evaluation, and may influence acceptability of this form of treatment to various health care providers and organizations.


Subject(s)
Hyaluronic Acid/administration & dosage , Osteoarthritis/drug therapy , Animals , Humans , Hyaluronic Acid/adverse effects , Injections, Intra-Articular , Knee Joint , Treatment Outcome
18.
Gene ; 248(1-2): 233-43, 2000 May 02.
Article in English | MEDLINE | ID: mdl-10806368

ABSTRACT

The astacin-related metalloproteases Bone Morphogenetic Protein-1 (BMP1) and Tolloid possess multiple functions in the maturation of extracellular matrices containing fibrillar collagens. We are interested in developing an in-vitro model system to study the role of BMP1 and Tolloid in chondrocytes and osteoblasts. Cloning of the cDNAs for chick BMP1 and Tolloid reveals that the two gene products are more than 80% identical to their human and mouse homologs and are similarly derived from the same genetic locus. Anti-BMP1/Tolloid antibodies have been developed, and detect two proteins of 80 and 116kDa. Chick BMP1 and Tolloid message and proteins are found in a variety of embryonic and juvenile tissues, including chondrocytes and osteoblasts. Tolloid message and protein are generally less abundant than BMP1 message; this discrepancy is greatest in growth plate chondrocytes. Tolloid protein is more tightly bound than BMP1 to the extracellular matrix produced by cultured osteoblasts. The Chordin gene is also expressed in chondrocytes and osteoblasts, suggesting that BMP1 and Tolloid influence BMP signaling as well as matrix maturation during skeletogenesis.


Subject(s)
Bone Morphogenetic Proteins/genetics , Bone and Bones/metabolism , DNA, Complementary/genetics , Glycoproteins , Intercellular Signaling Peptides and Proteins , Metalloendopeptidases/genetics , Proteins/genetics , Alternative Splicing , Amino Acid Sequence , Animals , Bone Morphogenetic Protein 1 , Bone Morphogenetic Proteins/metabolism , Bone and Bones/embryology , Chick Embryo , Chondrocytes/metabolism , Cloning, Molecular , DNA, Complementary/chemistry , Gene Expression Regulation, Developmental , Immunoblotting , Mammals , Metalloendopeptidases/metabolism , Metalloproteases , Molecular Sequence Data , Osteoblasts/metabolism , Protein Isoforms/genetics , Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Tissue Distribution , Tolloid-Like Metalloproteinases
19.
Exp Cell Res ; 256(2): 555-62, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10772827

ABSTRACT

Although PTHrP has been identified as a key regulator of chondrocyte differentiation in the growth plate, the factors directly regulating PTHrP expression have not been identified. Furthermore, while cells from the epiphysis are considered the physiologic source of PTHrP, the relative expression of PTHrP in epiphyseal and growth plate chondrocytes has not been defined. PTHrP expression was examined in chondrocytes isolated from 3- to 5-week-old chick long bones. The expression of PTHrP mRNA was 10-fold higher in epiphyseal chondrocytes compared to cells from the growth plate. Growth plate chondrocytes were isolated into populations with distinct maturational characteristics by countercurrent centrifugal elutriation and analyzed for PTHrP expression. The expression was highest in the least mature cells and progressively declined with the onset of maturation. The regulation of PTHrP expression was further examined in epiphyseal chondrocytes. Both TGF-beta1 and cis-retinoic acid stimulation markedly increased PTHrP mRNA levels, while BMP-2 and PTHrP stimulation decreased the expression of this transcript. The effects of TGF-beta1 (8.9-fold stimulation) and TGF-beta3 (9.2-fold) were slightly greater than the effects of TGF-beta2 (4.9-fold). The effect of TGF-beta was dose-dependent and increases could be detected after 68 h of treatment. To analyze the paracrine effect of epiphyseal and growth plate chondrocytes on each other, these cells were placed in coculture and the mRNA from each of the populations was harvested separately after 24 h. Following coculture the PTHrP mRNA levels increased in the epiphyseal cells while the expression of type X collagen and Indian hedgehog transcripts decreased in growth plate chondrocytes. The results demonstrate potentially important paracrine interactions between these cell populations, possibly mediated by TGF-beta and PTHrP.


Subject(s)
Chondrocytes/metabolism , Growth Plate/metabolism , Proteins/metabolism , Transforming Growth Factor beta/physiology , Animals , Cells, Cultured , Chickens , Coculture Techniques , Growth Plate/cytology , Parathyroid Hormone-Related Protein , Transforming Growth Factor beta/pharmacology
20.
J Orthop Res ; 18(6): 849-55, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11192243

ABSTRACT

Individuals who suffer from severe joint destruction caused by the various arthritidies often undergo total joint arthroplasty. A major limitation of this treatment is the development of aseptic loosening of the prosthesis in as many as 20% of patients. The current paradigm to explain aseptic loosening proposes that wear debris generated from the prosthesis initiates a macrophage-mediated inflammatory response by resident macrophages, leading to osteoclast activation and bone resorption at the implant interface. No therapeutic interventions have been proved to prevent or inhibit aseptic loosening. The development of therapeutic strategies is limited due to the absence of a quantitative surrogate in which drugs can be screened rapidly in large numbers of animals. We have previously described a model in which titanium particles implanted on mouse calvaria induce an inflammatory response with osteolysis similar to that observed in clinical aseptic loosening. Here, we present new methods by which the osteolysis in this model can be quantified. We determined that 6-8-week-old mice in normal health have a sagittal suture area of 50 (+/-6) microm2, which contains approximately five osteoclasts. As a result of the titanium-induced inflammation and osteolysis, the sagittal suture area increases to 197 (+/-27) microm2, with approximately 30 osteoclasts, after 10 days of treatment. The sagittal suture area and the number of osteoclasts in the calvaria of sham-treated mice remained unchanged during the 10 days. We also determined the effects of pentoxifylline, a drug that blocks the responses of tumor necrosis factor-alpha to wear debris, and the osteoclast inhibitor alendronate. We found that both drugs effectively block wear debris-induced osteolysis but not osteoclastogenesis. In conclusion, we found the measurements made with this model to be reproducible and to permit quantitative analysis of agents that are to be screened for their potential to prevent aseptic loosening.


Subject(s)
Arthritis/surgery , Arthroplasty/adverse effects , Disease Models, Animal , Osteolysis/prevention & control , Postoperative Complications/prevention & control , Prostheses and Implants/adverse effects , Alendronate/pharmacology , Animals , Cell Division/drug effects , Cell Division/physiology , Female , Male , Mice , Mice, Inbred CBA , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteolysis/drug therapy , Osteolysis/etiology , Pentoxifylline/pharmacology , Postoperative Complications/etiology , Postoperative Complications/physiopathology , Skull/drug effects , Skull/pathology , Skull/physiopathology , Stress, Mechanical , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...