1
|
Forte G, Chiarotto I, Giannicchi I, Loreto MA, Martinelli A, Micci R, Pepi F, Rossi S, Salvitti C, Stringaro A, Tortora L, Vecchio Ciprioti S, Feroci M. Characterization of naproxen–polymer conjugates for drug-delivery. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2015; 27:69-85. [DOI: 10.1080/09205063.2015.1108637] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
2
|
The effect of nonsteroidal anti-inflammatory drugs on tissue healing. Knee Surg Sports Traumatol Arthrosc 2013; 21:540-9. [PMID: 22744434 DOI: 10.1007/s00167-012-2095-2] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 06/04/2012] [Indexed: 01/29/2023]
Abstract
PURPOSE Non-selective (NSAIDs) and selective (COX-2) nonsteroidal anti-inflammatory drugs are commonly used for their analgesic and anti-inflammatory effects. Their role after orthopaedic surgery has been infrequently described and remains controversial because of unclear effects on soft tissue and bone healing. This study critically reviews the available literature to describe the effects of NSAIDs and COX-2 inhibitors on soft tissue and bone healing. METHODS A Medline search was performed using NSAIDs or COX-2 inhibitors and tissue healing. The combined search yielded 637 articles. Following exclusion, 44 articles were deemed relevant with 9 articles on soft tissue healing and 35 articles on bone healing. The available evidence is based primarily on animal data (39 studies), with considerable variation in study methods. RESULTS In regard to soft tissue healing, there is insufficient evidence of a detrimental effect when using either NSAIDs or COX-2 inhibitors at standard doses for ≤2 weeks. For soft tissue to bone healing, a limited number of studies demonstrate impairment in healing. However, with respect to bone healing, indomethacin appears to have a clear detrimental effect, with less substantial evidence for other NSAIDs. CONCLUSIONS Short-term, low-dose use of NSAIDs and COX-2 inhibitors does not appear to have a detrimental effect following soft tissue injury, but is inhibitory in cases involving bony healing. However, additional well-controlled human studies are necessary to draw more definitive conclusions regarding their role. Clinically, the prudent use of anti-inflammatory medications following sports medicine injuries and surgeries appears to be a reasonable option in clinical practice unless bone healing is required. LEVEL OF EVIDENCE III.
Collapse
|
3
|
Pountos I, Georgouli T, Calori GM, Giannoudis PV. Do nonsteroidal anti-inflammatory drugs affect bone healing? A critical analysis. ScientificWorldJournal 2012; 2012:606404. [PMID: 22272177 PMCID: PMC3259713 DOI: 10.1100/2012/606404] [Citation(s) in RCA: 173] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2011] [Accepted: 10/18/2011] [Indexed: 12/21/2022] Open
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) play an essential part in our approach to control pain in the posttraumatic setting. Over the last decades, several studies suggested that NSAIDs interfere with bone healing while others contradict these findings. Although their analgesic potency is well proven, clinicians remain puzzled over the potential safety issues. We have systematically reviewed the available literature, analyzing and presenting the available in vitro animal and clinical studies on this field. Our comprehensive review reveals the great diversity of the presented data in all groups of studies. Animal and in vitro studies present so conflicting data that even studies with identical parameters have opposing results. Basic science research defining the exact mechanism with which NSAIDs could interfere with bone cells and also the conduction of well-randomized prospective clinical trials are warranted. In the absence of robust clinical or scientific evidence, clinicians should treat NSAIDs as a risk factor for bone healing impairment, and their administration should be avoided in high-risk patients.
Collapse
Affiliation(s)
- Ippokratis Pountos
- Academic Department of Trauma & Orthopaedics, School of Medicine, University of Leeds, Leeds LS1 3EX, UK
| | | | | | | |
Collapse
|
4
|
HO MEILING, CHANG JEKEN, TSAI HSIUTING, CHO MINGHSUANG, WANG GWOJAW. NONSTEROIDAL ANTI-INFLAMMATORY DRUGS ARREST CELL CYCLE IN G0/G1PHASE AND INDUCE CELL DEATH IN OSTEOBLAST-ENRICHED CULTURES. ACTA ACUST UNITED AC 2011. [DOI: 10.1142/s0218957701000623] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Nonsteroidal anti-inflammatory drugs have been widely prescribed for orthopaedic patients to relieve pain and chronic inflammation. However, it has been demonstrated that NSAIDs suppress bone repair and remodeling in vivo. We have reported that ketorolac inhibits bone repair in vivo and its critical effective timing is at the early stage of endochondral ossification. Our previous results showed that ketorolac and indomethacin inhibit osteoblast proliferation in vitro, suggesting that this effect may be one of the mechanisms contributing to the suppressive effect of NSAIDs on bone remodeling. Cell proliferation and death of osteoblasts should be well regulated through some relative apoptotic and mitotic factors during normal bone remodeling process. Accordingly, we proposed that the induction of osteoblastic cell death of NSAIDs might be one of the mechanisms involving their suppressive effect on bone remodeling in vivo. In this study, we investigated whether NSAIDs arrest osteoblastic cell cycle and/or induce cell death. Whether the mechanism was mediated through prostaglandin (PG) pathway. We tested the effects of ketorolac, indomethacin, diclofenac, piroxicam on cell cycle kinetics, cytotoxicity, and cell death pattern in osteoblast-enriched cultures derived from fetal rat calvaria. Our results showed that ketorolac and indomethacin arrested cell cycle at G0/G1phase. All the 4 NSAIDs had cytotoxic effects and these effects were concentration dependent. The sequence of the cytotoxic effects of these four NSAIDs at 10-4M were indomethacin > diclofenac > ketorolac > piroxicam. Both PGE1and PGE2(10-10-10-8M) also significantly elevated the LDH leakage of osteoblasts, while PGF2αhad no significant effect. These results revealed that the cytotoxic effects of NSAIDs on osteoblasts might not be through inhibiting prostaglandin synthesis. They may be through PG-independent pathways. The results from flow cytometry followed by AnnexinV-FITC and propidium iodide double staining showed that 24 hours treatment of all the 4 NSAIDs (10-6and 10-4M) significantly induced both apoptosis (p<0.01) and necrosis (p<0.01, or p<0.05) in osteoblast cultures. These effects of NSAIDs on cell cycle arrest and cell death induction in osteoblasts may be one of the important mechanisms contributing to their suppressive effect on bone repair and bone remodeling in vivo.
Collapse
Affiliation(s)
- MEI-LING HO
- Departments of Physiology, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, R.O.C
| | - JE-KEN CHANG
- Departments of Orthopaedics, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, R.O.C
| | - HSIU-TING TSAI
- Departments of Physiology, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, R.O.C
| | - MING-HSUANG CHO
- Departments of Physiology, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, R.O.C
| | - GWO-JAW WANG
- Departments of Orthopaedics, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, R.O.C
| |
Collapse
|
5
|
Wixted JJ, Fanning PJ, Gaur T, O'Connell SL, Silva J, Mason-Savas A, Ayers DC, Stein GS, Lian JB. Enhanced fracture repair by leukotriene antagonism is characterized by increased chondrocyte proliferation and early bone formation: a novel role of the cysteinyl LT-1 receptor. J Cell Physiol 2009; 221:31-9. [PMID: 19544365 DOI: 10.1002/jcp.21809] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Inflammatory mediators and drugs which affect inflammation can influence the healing of injured tissues. Leukotrienes are potent inflammatory mediators, and similar to prostaglandins, are metabolites of arachidonic acid which can have positive or negative effects on bone and cartilage tissues. Here we tested the hypothesis that blocking the negative regulation of leukotrienes, would lead to enhanced endochondral bone formation during fracture repair. A closed femoral fracture was created in mice. Animals were divided into three groups for treatment with either montelukast sodium, a cysteinyl leukotriene type 1 receptor antagonist (trade name Singulair), zileuton, a 5-lipoxygenase enzyme inhibitor (trade name Zyflo), or carrier alone. The fractures were analyzed using radiographs, quantitative gene expression, histology and histomorphometry, and immunohistochemistry. Both the montelukast sodium group and the zileuton group exhibited enhanced fracture repair when compared with controls. Both treatment groups exhibited increased callous size and earlier bone formation when compared to controls as early as day 7. Gene expression analysis of treatment groups showed increased markers of chondrocyte proliferation and differentiation, and increased early bone formation markers when compared with controls. Treatment with montelukast sodium directly targeted the cysteinyl leukotriene type 1 receptor, leading to increased chondrocyte proliferation at early time points. These novel findings suggests a potential mechanism by which the cysteinyl leukotriene type 1 receptor acts as a negative regulator of chondrocyte proliferation, with important and previously unrecognized implications for both fracture repair, and in a broader context, systemic chondrocyte growth and differentiation.
Collapse
Affiliation(s)
- John J Wixted
- Department of Orthopedic Surgery and Physical Rehabilitation, University of Massachusetts Medical School, Worcester, Massachusetts, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Dimmen S, Nordsletten L, Engebretsen L, Steen H, Madsen JE. The effect of parecoxib and indometacin on tendon-to-bone healing in a bone tunnel. ACTA ACUST UNITED AC 2009; 91:259-63. [DOI: 10.1302/0301-620x.91b2.21471] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Conventional non-steroidal anti-inflammatory drugs (NSAIDs) and newer specific cyclo-oxygenase-2 (cox-2) inhibitors are commonly used in musculoskeletal trauma and orthopaedic surgery to reduce the inflammatory response and pain. These drugs have been reported to impair bone metabolism. In reconstruction of the anterior cruciate ligament the hamstring tendons are mainly used as the graft of choice, and a prerequisite for good results is healing of the tendons in the bone tunnel. Many of these patients are routinely given NSAIDs or cox-2 inhibitors, although no studies have elucidated the effects of these drugs on tendon healing in the bone tunnel. In our study 60 female Wistar rats were randomly allocated into three groups of 20. One received parecoxib, one indometacin and one acted as a control. In all the rats the tendo-Achillis was released proximally from the calf muscles. It was then pulled through a drill hole in the distal tibia and sutured anteriorly. The rats were given parecoxib, indometacin or saline intraperitoneally twice daily for seven days. After 14 days the tendon/bone-tunnel interface was subjected to mechanical testing. Significantly lower maximum pull-out strength (p < 0.001), energy absorption (p < 0.001) and stiffness (p = 0.035) were found in rats given parecoxib and indometacin compared with the control group, most pronounced with parecoxib.
Collapse
Affiliation(s)
- S. Dimmen
- Orthopaedic Centre, Ullevaal University Hospital and Faculty of Medicine, University of Oslo, N-0407 Oslo, Norway
| | - L. Nordsletten
- Orthopaedic Centre, Ullevaal University Hospital and Faculty of Medicine, University of Oslo, N-0407 Oslo, Norway
| | - L. Engebretsen
- Orthopaedic Centre, Ullevaal University Hospital and Faculty of Medicine, University of Oslo, N-0407 Oslo, Norway
| | - H. Steen
- Institute for Surgical Research, Rikshospitalet-Radiumhospitalet Medical Centre and Faculty of Medicine, University of Oslo, N-0027, Oslo, Norway
| | - J. E. Madsen
- Orthopaedic Centre, Ullevaal University Hospital and Faculty of Medicine, University of Oslo, N-0407 Oslo, Norway
| |
Collapse
|
7
|
Dimmen S, Nordsletten L, Engebretsen L, Steen H, Madsen JE. Negative effect of parecoxib on bone mineral during fracture healing in rats. Acta Orthop 2008; 79:438-44. [PMID: 18626809 DOI: 10.1080/17453670710015373] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND AND PURPOSE Non-steroidal anti-inflammatory drugs (NSAIDs) are conventional cyclooxygen-ase (cox) inhibitors commonly used in musculoskeletal trauma to reduce the inflammatory response and pain, but they also seem to affect bone metabolism. Parecoxib is a cox inhibitor that selectively inhibits cox-2. Through their selective mechanism of action, these newer drugs are supposed to reduce the gastrointestinal side effects of conventional cox inhibitors. The effects on bone metabolism and healing have, however, not been fully elucidated. Thus, there are reasons for concern regarding the potential negative effects of these drugs on bone metabolism and bone repair. We investigated the effects of short-term administration of parecoxib on bone mineral formation and bone healing in rats. ANIMALS AND METHODS 26 female Wistar rats were given parecoxib intraperitoneally for 7 days after a closed tibial fracture that was stabilized with an intra-medullary nail, and 26 animals were given saline. At 2, 3, and 6 weeks after surgery bone mineral density (BMD) at the fracture site was measured using dualenergy X-ray absorptiometry (DEXA). 6 weeks after the fracture, 14 rats from the parecoxib group and 16 rats from the placebo group were killed for mechanical testing, and the rest of the animals were killed for tissue analysis. The healing fractures and the intact contralateral tibias were mechanically tested by three-point cantilever bending. RESULTS The BMD at the fracture site was calculated as the average of the results after 2,3, and 6 weeks. Mean BMD was lower in the parecoxib group, 0.23 (SD 0.06) g/ cm2, than in the control group, 0.27 (SD 0.05) g/cm2 (p = 0.01). There were no statistically significant differences in mechanical properties of the healing fractures after 6 weeks. However, the study may have lacked sufficient statistical power to determine whether a negative effect on healing had occurred. INTERPRETATION No mechanical differences were detected between the control and treatment groups after 6 weeks, but they may have been present earlier in the fracture healing process. Our findings do, however, indicate that parecoxib given postoperatively for a week has a negative effect on mineralization during the early phase of fracture healing.
Collapse
Affiliation(s)
- Sigbøjrn Dimmen
- Institute of Surgical Research and Biomechanics Laboratory, Orthopedics Department, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Norway.
| | | | | | | | | |
Collapse
|
8
|
Chang JK, Li CJ, Wu SC, Yeh CH, Chen CH, Fu YC, Wang GJ, Ho ML. Effects of anti-inflammatory drugs on proliferation, cytotoxicity and osteogenesis in bone marrow mesenchymal stem cells. Biochem Pharmacol 2007; 74:1371-82. [PMID: 17714695 DOI: 10.1016/j.bcp.2007.06.047] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2007] [Revised: 06/29/2007] [Accepted: 06/29/2007] [Indexed: 11/22/2022]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) were found to suppress proliferation and induce cell death in cultured osteoblasts, and steroids were found to decrease the osteogenesis potential of mesenchymal stem cells. In this study, we further tested the effects of anti-inflammatory drugs (AIDs) on the functions of bone marrow mesenchymal stem cells (BMSCs). The BMSCs from mice (D1-cells) and humans (hBMSCs) were treated with dexamethasone (10(-7) to 10(-6) M), cyclooxygenase-2 (COX-2) selective NSAIDs (10(-6) to 10(-5) M) and non-selective NSAIDs (10(-5) to 10(-4) M). Drug effects on proliferation, cell cycle kinetics, cytotoxicity and mRNA and protein expressions of cell cycle regulators were tested. The osteogenesis potential of D1-cells were evaluated by testing mRNA expressions of type Ialpha collagen and osteocalcin 2-8 days after treatments, and testing mineralization 1-3 weeks after treatments. The results showed that all the tested drugs suppressed proliferation and arrested cell cycle of D1-cells, but no significant cytotoxic effects was found. Prostaglandin E1, E2 and F2alpha couldn't rescue the effects of AIDs on proliferation. The p27kip1 expression was up-regulated by indomethacin, celecoxib and dexamethasone in both D1-cells and hBMSCs. Higher concentrations of indomethacin and dexamethasone also up-regulated p21Cip1/Waf1 expression in hBMSCs, and so did celecoxib on D1-cells. Expressions of cyclin E1 and E2 were down-regulated by these AIDs in D-cells, while only cyclin E2 was down-regulated by dexamethasone in hBMSCs. All the tested NSAIDs revealed no obvious detrimental effects on osteogenic differentiation of D1-cells. These results suggest that the proliferation suppression of AIDs on BMSCs may act via affecting expressions of cell cycle regulators, but not prostaglandin-related mechanisms.
Collapse
Affiliation(s)
- Je-Ken Chang
- Department of Orthopaedics, Faculty of Medicine, Kaohsiung Medical University, and Kaohsiung Medical University Hospital, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Busti AJ, Hooper JS, Amaya CJ, Kazi S. Effects of perioperative antiinflammatory and immunomodulating therapy on surgical wound healing. Pharmacotherapy 2006; 25:1566-91. [PMID: 16232020 DOI: 10.1592/phco.2005.25.11.1566] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Patients with various rheumatologic and inflammatory disease states commonly require drugs known to decrease the inflammatory or autoimmune response for adequate control of their condition. Such drugs include nonsteroidal antiinflammatory drugs (NSAIDs), cyclooxygenase (COX)-2 inhibitors, corticosteroids, disease-modifying antirheumatic drugs (DMARDs), and biologic response modifiers. These drugs affect inflammation and local immune responses, which are necessary for proper wound healing in the perioperative setting, thereby potentially resulting in undesirable postoperative complications. Such complications include wound dehiscence, infection, and impaired collagen synthesis. The end result is delayed healing of soft tissue and bone wounds. The current literature provides insight into the effect of some of these drugs on wound healing. For certain drugs, such as methotrexate, trials have been conducted in humans and direct us on what to do during the perioperative period. Whereas with other drugs, we must rely on either small-animal studies or extrapolation of data from human studies that did not specifically look at wound healing. Unfortunately, no clear consensus exists on the need and optimum time for withholding therapy before surgery. Likewise, clinicians are often uncertain of the appropriate time to resume therapy after the procedure. For those drugs with limited or no data in this setting, the use of pharmacokinetic properties and biologic effects of each drug should be considered individually. In some cases, discontinuation of therapy may be required up to 4 weeks before surgery because of the long half-lives of the drugs. In doing so, patients may experience an exacerbation or worsening of disease. Clinicians must carefully evaluate individual patient risk factors, disease severity, and the pharmacokinetics of available therapies when weighing the risks and benefits of discontinuing therapy in the perioperative setting.
Collapse
Affiliation(s)
- Anthony J Busti
- Texas Tech University Health Sciences Center School of Pharmacy, Dallas-Ft. Worth Regional Campus, Dallas, Texas 75216, USA.
| | | | | | | |
Collapse
|
10
|
Persson PE, Sisask G, Nilsson O. Indomethacin inhibits bone formation in inductive allografts but not in autografts: studies in rat. Acta Orthop 2005; 76:465-9. [PMID: 16195059 DOI: 10.1080/17453670510041420] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Non-steroidal anti-inflammatory drugs (NSAIDs) are known to have inhibitory effects on new bone formation. We wanted to analyze some effects of the NSAID indomethacin on different inductive stimuli for bone formation. METHODS We experimentally induced heterotopic new bone with demineralized allogeneic bone matrix (DABM) and with bone autografts in rats, in order to study the effects of indomethacin on new bone formation at 3 and 6 weeks. RESULTS Indomethacin inhibited net bone formation in DABMs by 30% at 6 weeks. At 6 weeks, the mineral accretion rate was unaffected, indicating that it is at the early phase of the inductive process that mineral accretion is sensitive to inhibition by indomethacin, but not at the later stages. In traumatized skeletal bone, the (45)Ca-specific activity was higher than in non-traumatized bone, while indomethacin significantly reduced the (45)Ca uptake at 3 weeks, but not at 6 weeks. In the autografts, a net mineral loss occurred, but neither mineral content nor (45)Ca incorporation was affected by Indomethacin treatment. INTERPRETATION Indomethacin inhibited the early phase of new bone formation in heterotopic DABMs and the early bone healing process in traumatized skeletal bone, but did not affect resorption or bone formation in heterotopic autografts. blacksquare, square, filled.
Collapse
|
11
|
Jessop HL, Suswillo RFL, Rawlinson SCF, Zaman G, Lee K, Das-Gupta V, Pitsillides AA, Lanyon LE. Osteoblast-like cells from estrogen receptor alpha knockout mice have deficient responses to mechanical strain. J Bone Miner Res 2004; 19:938-46. [PMID: 15190886 DOI: 10.1359/jbmr.2004.19.6.938] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
INTRODUCTION In vivo, bones' osteogenic response to mechanical loading involves proliferation of surface osteoblasts. This response is replicated in vitro and involves ERK-mediated activation of the estrogen receptor (ER) alpha and upregulation of estrogen response element activity. This proliferative response can be blocked by selective estrogen receptor modulators and increased by transfection of additional ERalpha. MATERIALS AND METHODS We have now investigated the mechanisms of ER involvement in osteoblast-like cells' early responses to strain by comparing the responses of primary cultures of these cells derived from homozygous ERalpha knockout (ERKO) mice (ERalpha-/-) with those from their wildtype (ERalpha+/+) and heterozygous (ERalpha+/-) littermates and from ER/beta knockout (BERKO) mice (ERbeta+/+, ERbeta+/-, and ERbeta-/-). RESULTS Whereas ERalpha+/+, ERalpha+/-, ERbeta+/+, and ERbeta-/- cells proliferate in response to a single 10-minute period of cyclic strain, ERalpha-/- cells do not. Transfection of fully functional, but not mutant, ERalpha rescues the proliferative response to strain in these cells. The strain-related response of ERalpha-/- cells is also deficient in that they show no increased activity of an AP-I driven reporter vector and no strain-related increases in NO production. Their strain-related increase in prostacyclin production is retained. They proliferate in response to fibroblast growth factor-2 but not insulin-like growth factor (IGF)-I or IGF-II, showing the importance of ERalpha in the IGF axis and the ability of ERalpha-/- cells to proliferate normally in response to a mitogenic stimulus that does not require functional ERalpha. CONCLUSIONS These data indicate ERalpha's obligatory involvement in a number of early responses to mechanical strain in osteoblast-like cells, including those that result in proliferation. They support the hypothesis that reduction in ERalpha expression or activity after estrogen withdrawal results in a less osteogenic response to loading. This could be important in the etiology of postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Helen L Jessop
- Department of Veterinary Basic Sciences, The Royal Veterinary College, London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Chang JK, Chuang LY, Ho ML, Wang GJ. Effects of nonsteroidal anti-inflammatory drugs on transforming growth factor-beta expression and bioactivity in rat osteoblast-enriched culture. Kaohsiung J Med Sci 2003; 19:278-88. [PMID: 12873036 DOI: 10.1016/s1607-551x(09)70474-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) have been reported to suppress bone remodeling in normal and repaired bones. Our previous results indicated that ketorolac and indomethacin suppressed proliferation, stimulated early differentiation, and induced apoptosis in cultured osteoblasts. Transforming growth factor-beta (TGF-beta) has been reported to enhance proliferation, suppress differentiation, and prevent apoptosis in osteoblasts. We proposed that one pathway of NSAID effects on osteoblast function might be through inhibition of the expression and/or bioactivity of TGF-beta in osteoblasts. We tested the effects of ketorolac and indomethacin on the expression of TGF-beta1 mRNA and protein and the bioactivity of TGF-beta in osteoblast-enriched cultures derived from fetal calvaria. The effects of prostaglandin E1 (PGE1) and PGE2 on TGF-beta expression and bioactivity were also examined in order to understand more about the role of prostaglandins in osteoblast function. Simultaneously, we estimated whether these NSAID effects on osteoblasts were prostaglandin-related. The results showed that 24-hour treatments with both PGEs and theoretic therapeutic concentrations of ketorolac and indomethacin had no significant effects on the levels of either transcription or translation of TGF-beta or the post-translational function of TGF-beta in osteoblasts. These results suggest that NSAIDs do not affect osteoblast function through changes in TGF-beta action in osteoblasts.
Collapse
Affiliation(s)
- Je-Ken Chang
- Department of Orthopedics, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | | | | |
Collapse
|
13
|
Abstract
BACKGROUND Nonsteroidal anti-inflammatory drugs have been implicated in the development of delayed unions and nonunion after fractures in animal models. Previous investigations have identified two important factors as determinants of delayed fracture healing: early drug administration and a dose-dependent effect. OBJECTIVE The purpose of this investigation was to study the effect of tenoxicam, a nonsteroidal anti-inflammatory drug, on the fracture healing process in rat tibiae. METHODS Fifty-eight Wistar rats were randomly divided in four groups (I, II, III, and IV). Group I (control group, n=12) was given 0.1ml saline solution per day intramuscularly. Groups II (n=12), III (n=12), and IV (n=12) were administered 10mg per kg per day of tenoxicam intramuscularly. Administration of substances was begun on a week before to 48h after the fracturing procedure and continued during the entire experiment. Callus formation was studied histologically and histomorphologically, using light microscopy. In addition, a histologic grading based on the morphologic stage of fracture healing was carried out at 4 weeks, according to the criteria proposed by Allen et al. RESULTS There was a significant difference in treatment effect between Group I (saline solution) and Groups II, III, and IV (tenoxicam) (P=0.07). Histologically and histomorphologically, there were qualitative and quantitative delay in callus formation at all tenoxicam groups. This was more pronounced the earlier the nonsteroidal anti-inflammatory drug was started, although no significant difference could be detected between Groups II, III, and IV (P>(alpha=10%)). Four weeks after fracture, Group I (n=3) showed complete osseous union, Groups II (n=3) and III (n=3), complete cartilaginous union, and Group IV (n=3), incomplete osseous union, according to Allen et al. By using this rating scale, the difference between control and drug-treated groups was statistically significant (P<0.1). CONCLUSION Under studied conditions, this investigation shows that administration of tenoxicam intramuscularly delays fracture healing process in rat tibiae. These results suggest the hypothesis that early drug administration may delay bone healing after experimental fractures in animals, although it could not be detected statistically significant.
Collapse
Affiliation(s)
- Vincenzo Giordano
- Departamento de Ortopedia e Traumatologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | | | | | | | | |
Collapse
|
14
|
Jessop HL, Rawlinson SCF, Pitsillides AA, Lanyon LE. Mechanical strain and fluid movement both activate extracellular regulated kinase (ERK) in osteoblast-like cells but via different signaling pathways. Bone 2002; 31:186-94. [PMID: 12110433 DOI: 10.1016/s8756-3282(02)00797-4] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Extracellular regulated kinases (ERKs)-1 and -2 are members of the MAPK family of protein kinases involved in the proliferation, differentiation, and apoptosis of bone cells. We have shown previously that ROS 17/2.8 cells show increased activation of ERK-1 or -2, which is sustained for 24 h, when the strips onto which they are seeded are subjected to a 10 min period of cyclic four point bending that produces physiological levels of mechanical strain along with associated fluid movement of the medium. Movement of the strips through the medium without bending causes fluid movement without strain. This also increases ERK-1/2 activation, but in a biphasic manner over the same time period. Our present study investigates the role of components of signaling pathways in the activation of ERK-1/2 in ROS 17/2.8 cells in response to these stimuli. Using a range of inhibitors we show specific differences by which ERK-1 and ERK-2 are activated in response to fluid movement alone, compared with those induced in response to strain plus its associated fluid movement. ERK-1 activation induced by fluid movement was markedly reduced by nifedipine, and therefore appears to involve L-type calcium channels, but was unaffected by either L-NAME or indomethacin. This suggests independence from prostacyclin (PGI(2)) and nitric oxide (NO) production. In contrast, ERK-1 activation induced by application of strain (and its associated fluid disturbance) was abrogated by TMB-8 hydrochloride, L-NAME, and indomethacin. This suggests that strain-induced ERK-1 activation is dependent upon calcium mobilization from intracellular stores and production of NO and PGI(2). ERK-2 activation appears to be mediated by a separate mechanism in these cells. Its activation by fluid movement alone involved both PGI(2) and NO production, but its activation by strain was not affected by any of the inhibitors used. The G protein inhibitor, pertussis toxin, did not cause a reduction in the activation of ERK-1 or -2 in response to either stimulus. These results are consistent with earlier observations of ERK activation in bone cells in response to both strain (with fluid movement) and fluid movement alone, and further demonstrate that these phenomena stimulate distinct signaling pathways.
Collapse
Affiliation(s)
- H L Jessop
- Department of Veterinary Basic Sciences, The Royal Veterinary College, London, UK
| | | | | | | |
Collapse
|
15
|
Ho ML, Chang JK, Chuang LY, Hsu HK, Wang GJ. Effects of nonsteroidal anti-inflammatory drugs and prostaglandins on osteoblastic functions. Biochem Pharmacol 1999; 58:983-90. [PMID: 10509750 DOI: 10.1016/s0006-2952(99)00186-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
It has been reported that nonsteroidal anti-inflammatory drugs (NSAIDs) suppress bone repair and bone remodeling but only mildly inhibit bone mineralization at the earlier stage of the repair process. We proposed that the proliferation and/or the earlier stage of differentiation of osteoblasts may be affected by NSAIDs. This study was designed to investigate whether NSAIDs affect the proliferation and/or differentiation of osteoblasts and whether these effects are prostaglandin (PG) mediated. The effects of PGE1 and PGE2, indomethacin, and ketorolac on thymidine incorporation, cell count, intracellular alkaline phosphatase (ALP) activity, and Type I collagen content in osteoblast-enriched cultures derived from fetal calvaria were evaluated. The results showed that both PGs and NSAIDs inhibited DNA synthesis and cell mitosis in a time- and concentration-dependent manner. However, intracellular ALP activity and Type I collagen content were stimulated at an earlier stage of differentiation in osteoblasts. These results suggested that (i) the inhibitory effect of ketorolac on osteoblastic proliferation contributes to its suppressive effects on bone repair and remodeling in vivo; (ii) PGEs and NSAIDs may be involved in matrix maturation and biologic bone mineralization in the earlier stage of osteoblast differentiation; and (iii) the effects of ketorolac and indomethacin on cell proliferation and differentiation may not be through the inhibition of the synthesis of PGE1 or PGE2.
Collapse
Affiliation(s)
- M L Ho
- Department of Physiology, Kaohsiung Medical College, Taiwan, ROC.
| | | | | | | | | |
Collapse
|
16
|
Sibonga JD, Bell NH, Turner RT. Evidence that ibuprofen antagonizes selective actions of estrogen and tamoxifen on rat bone. J Bone Miner Res 1998; 13:863-70. [PMID: 9610751 DOI: 10.1359/jbmr.1998.13.5.863] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Studies were performed to determine if the nonsteroidal anti-inflammatory drug ibuprofen alters bone and mineral metabolism in female rats. In experiment 1, four groups of growing rats underwent either sham operation or ovariectomy (OVX). One week later, controlled-release pellets with ibuprofen or placebo were implanted subcutaneously at the back of the neck. Following 3 weeks of treatment, rats were sacrificed and blood and bone samples were removed for serum assays and histomorphometric analysis. Body growth rate and the static cortical bone measurements made at the tibial diaphysis did not change in response to OVX. OVX, however, did increase radial bone growth, lowered serum 17beta-estradiol, reduced uterine weight, and decreased the cancellous bone area of the tibial metaphysis in the rats. Ibuprofen did not alter serum 17beta-estradiol or uterine weight but reduced radial bone growth as well as cancellous bone area of the tibial metaphysis in both sham-operated and OVX animals. In experiments 2 and 3, we tested the influence of ibuprofen on the effects of the tissue-selective estrogen agonist tamoxifen and of exogenous 17beta-estradiol in the OVX rat. Ibuprofen completely blocked the effects of tamoxifen and partially blocked the effects of 17beta-estradiol to prevent cancellous osteopenia. In contrast, ibuprofen did not influence the effects of tamoxifen and 17beta-estradiol to reduce radial bone growth. Besides the skeletal effects, ibuprofen suppressed estrogen-induced uterine growth. Our data suggest that ibuprofen blocks selective estrogen receptor-mediated activities in the rat.
Collapse
Affiliation(s)
- J D Sibonga
- Department of Orthopedics, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | |
Collapse
|
17
|
Giunta D, Keller J, Nielsen FF, Melsen B. Influence of indomethacin on bone turnover related to orthodontic tooth movement in miniature pigs. Am J Orthod Dentofacial Orthop 1995; 108:361-6. [PMID: 7572847 DOI: 10.1016/s0889-5406(95)70033-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The purpose was to evaluate the influence of a prostaglandin inhibitor, indomethacin, on the tissue reaction related to orthodontic tooth movement. Sixteen miniature pigs were chosen for the study, eight of which received indomethacin perorally every day of the 39-day observation period. Sentalloy expansion springs (GAC, Central Islip, N.Y.) delivering 100 cN were inserted on a segmented arch between the central lower incisors. Intravital labeling with tetracycline was used for the evaluation of the rate of bone formation. After the pigs were killed, the bone turnover was evaluated on undecalcified methacrylate embedded sections and on microradiographs. The histomorphometric analysis of bone turnover revealed that the relative extent of resorption surfaces was decreased significantly in the indomethacin treated animals. Formation surfaces were also decreased although not significantly. The bone turnover, but not the mineralization rate, was influenced. The results corroborate the recommendation that prostaglandin inhibitors should be avoided during orthodontic treatments.
Collapse
Affiliation(s)
- D Giunta
- Orthodontic Department, Dentistry School, University of Naples, Italy
| | | | | | | |
Collapse
|
18
|
Cook SD, Barrack RL, Dalton JE, Thomas KA, Brown TD. Effects of indomethacin on biologic fixation of porous-coated titanium implants. J Arthroplasty 1995; 10:351-8. [PMID: 7673914 DOI: 10.1016/s0883-5403(05)80184-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The effect of perioperative administration of indomethacin on attachment strength and bone growth into porous-coated titanium implants was evaluated in the canine transcortical plug model. Various drug administration protocols simulating clinical use of indomethacin were studied. These included chronic treatment (starting 2 weeks prior to surgery), treatment immediately after surgery, and treatment 3, 6, 9, and 18 weeks following surgery. Indomethacin therapy was continued until sacrifice at 3, 6, 12, 18, or 24 postoperative weeks. Push-out testing was performed to determine the maximum bone-implant interface shear strength, and quantitative histologic analysis was used to determine percentage of bone ingrowth. When indomethacin was administered chronically or immediately after surgery, a statistically significant decrease in bone-implant interface attachment strength was seen at 3 postoperative weeks but not at later periods. No adverse effect was observed in any group after the 3-week period. Quantitative histologic analysis demonstrated no significant differences in percentage bone ingrowth among any of the treatment protocols at 3 or 6 weeks after surgery. No significant difference was observed between any of the groups at 18 or 24 weeks. The results of this study suggest that perioperative administration of indomethacin does not significantly affect attachment strength or bone ingrowth into porous-coated implants except at early periods, in which cases a transient decrease in attachment strength occurs.
Collapse
Affiliation(s)
- S D Cook
- Department of Orthopaedic Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | | | | | | | | |
Collapse
|
19
|
Sennerby L, Kälebo P, Thomsen P, Albrektsson T. Influence of indomethacin on the regeneration of cortical bone within titanium implants in rabbits. Biomaterials 1993; 14:156-8. [PMID: 8435461 DOI: 10.1016/0142-9612(93)90231-p] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The influence of indomethacin on cortical bone regeneration was studied in bone harvest chambers made of commercially pure titanium and inserted in rabbit tibia. Newly formed bone was harvested in situ every 3 wk for 33 wk. Indomethacin (1 mg/kg body weight) was given daily as subcutaneous injections for two periods, followed by two control periods with no drug administration and the same schedule was followed for indomethacin at a dose of 4 mg/kg body weight. These indomethacin dosages did not statistically influence the cortical bone regeneration.
Collapse
Affiliation(s)
- L Sennerby
- Department of Handicap Research, University of Gothenburg, Sweden
| | | | | | | |
Collapse
|
20
|
Keller J, Schumacher B, Lind M. Effect of local prostaglandin E2 on periosteum and muscle in rabbits. ACTA ORTHOPAEDICA SCANDINAVICA 1992; 63:623-7. [PMID: 1471509 DOI: 10.1080/17453679209169722] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We assessed the target tissue for the stimulatory effect of prostaglandin E2 (PGE2) on bone formation previously observed during fracture healing. PGE2 was infused into tibial periosteal tissue in the right leg of 7 rabbits and into the anterior tibial muscle in the right leg of 7 other rabbits for 6 weeks. Solvent solution was infused into the left leg. PGE2 infusion at the periosteum caused the formation of primitive woven bone with large amounts of connective tissue; solvent infusion caused small amounts of normal periosteal bone formation. In the neighboring cortical bone, remodeling was increased after PGE2 infusion compared to solvent infusion. In the muscle, PGE2 infusion caused the formation of connective tissue with small amounts of woven bone. Thus, the major effects of PGE2 infusion at the site of the periosteum was the formation of primitive woven bone and in muscles the formation of connective tissue.
Collapse
Affiliation(s)
- J Keller
- Institute of Pathology, Aarhus Amtssygehus, Denmark
| | | | | |
Collapse
|
21
|
Hansen ES, He SZ, Søballe K, Kjølseth D, Henriksen TB, Hjortdal VE, Bünger C. [99mTc]diphosphonate uptake and hemodynamics in experimental arthritis: effect of naproxen in the canine carrageenan injection model. J Orthop Res 1992; 10:647-56. [PMID: 1500978 DOI: 10.1002/jor.1100100507] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The impact of naproxen treatment on juxta-articular hemodynamics and bone metabolism in experimental juvenile arthritis was studied in the articular carrageenan injection model. Unilateral gonarthritis was induced for 12 weeks in eight dogs receiving naproxen (dosage, 2 mg/kg) and eight controls. Regional blood flow was assessed by the microsphere method, plasma volume by the distribution space of [125I]fibrinogen, and bone metabolism by the 2-h uptake of [99mTc]diphosphonate ([99mTc]DPD). Synovial effusion was less prominent with naproxen treatment as judged by joint fluid volume and pressure. Naproxen reduced the arthritic capsular hyperemia, almost normalized a severe blood flow increase in patella and both juxta-articular epiphyses, ameliorated an expansion of plasma volume in the patella and the distal femoral epiphysis, and normalized an increased [99mTc]DPD uptake in subchondral femoral bone and the tibial cortex. Significantly increased arteriovenous shunting in the arthritic extremity was unaffected by naproxen. The study suggests that long-term cyclooxygenase inhibition offers protection against hemodynamic and metabolic changes in juxta-articular bone secondary to synovial inflammation.
Collapse
Affiliation(s)
- E S Hansen
- Department of Orthopedics, University of Aarhus, Denmark
| | | | | | | | | | | | | |
Collapse
|
22
|
Reis HJ, Küsswetter W, Schellinger T. The suppression of heterotopic ossification after total hip arthroplasty. INTERNATIONAL ORTHOPAEDICS 1992; 16:140-5. [PMID: 1428312 DOI: 10.1007/bf00180205] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In a double-blind prospective randomised study we examined the effects of Diclofenac on heterotopic ossification after hip arthroplasty. Either the drug, or a placebo, was given by mouth to 158 patients in doses of 3 x 50 mg for 6 weeks. Diclofenac resulted in highly significant improvement (p less than 0.0001 versus controls) without severe side-effects. Heterotopic ossification decreased from 55% in the placebo to 15%. Significant degrees of heterotopic ossification did not occur with the drug. Movement of the hip was considerably increased after operation. We recommend Diclofenac prophylaxis against heterotopic ossification after hip operations in view of the serious clinical effects of this complication and its quoted incidence of 15% to 80%.
Collapse
Affiliation(s)
- H J Reis
- Department of Orthopaedics, University of Tübingen, Federal Republic of Germany
| | | | | |
Collapse
|
23
|
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) affect bone metabolism in vitro and in vivo. They delay but do not alter the outcome of healing processes in bone. In some bone loss models, they block bone resorption and slow the rate of loss. We studied the effect of naproxen, a potent NSAID, on cancellous bone of the proximal tibial metaphysis of 6-month-old adult female ovariectomized rats. Animals were ovariectomized, divided into groups, and fed standard diets differing only in naproxen content for 42 days. The rats of the groups ate 2.0, 5.5, 12.7, and 32 mg naproxen per kg body weight per day, respectively. Serum levels of naproxen were determined. Bone volume, mineralizing surface, osteoblast activity, osteoclast surface, and bone resorption rate were determined by bone histomorphometric techniques. The rats' dose-related serum naproxen levels ranged from 4 to 28 micrograms/ml. Naproxen inhibited up to 70% of the bone loss occurring after ovariectomy at a serum level of 4 micrograms/ml. We deduced that naproxen blocked bone resorption in ovariectomized rats by slowing osteoclast activity at all doses. In contrast, naproxen slowed bone formation only at serum levels greater than 20 micrograms/ml in ovariectomized rats. These findings may have clinical relevance in helping to prevent postmenopausal bone loss in women.
Collapse
Affiliation(s)
- N Lane
- Syntex Labs, Palo Alto, CA 94305
| | | | | |
Collapse
|
24
|
Keller J, Kjaersgaard-Andersen P, Bayer-Kristensen I, Melsen F. Indomethacin and bone trauma. Effects on remodeling of rabbit bone. ACTA ORTHOPAEDICA SCANDINAVICA 1990; 61:66-9. [PMID: 2336956 DOI: 10.3109/17453679008993070] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The influence of indomethacin on remodeling activity in normal trabecular and cortical bone and its influence on cortical bone close to a midtibial drill hole, 2 mm in diameter were histomorphometrically evaluated. Eight rabbits were treated with indomethacin (12.5 mg/kg/day), and another 8 rabbits served as controls. After 3 days, the mean plasma indomethacin level was 542 ng/mL, resulting in an almost complete inhibition of prostaglandin synthesis as reflected by the serum levels. In the control rabbits the remodeling activity after 6 weeks was increased 1 mm away from the drill hole but not at 3 and 8 mm. In conclusion, indomethacin had no effect on the activated remodeling process in cortical bone neighboring a small drill hole or on remodeling in nontraumatized cortical and cancellous bone. This suggests that the inhibitory effect of indomethacin on the remodeling process following local trauma to bone depends on the extent of the trauma.
Collapse
Affiliation(s)
- J Keller
- University Department of Orthopedics, Arhus Amtssygehus, Denmark
| | | | | | | |
Collapse
|