151
|
Cao T, Heng BC, Ye CP, Liu H, Toh WS, Robson P, Li P, Hong YH, Stanton LW. Osteogenic differentiation within intact human embryoid bodies result in a marked increase in osteocalcin secretion after 12 days of in vitro culture, and formation of morphologically distinct nodule-like structures. Tissue Cell 2005; 37:325-34. [PMID: 15979113 DOI: 10.1016/j.tice.2005.03.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2004] [Revised: 03/11/2005] [Accepted: 03/14/2005] [Indexed: 11/19/2022]
Abstract
Osteogenic lineages derived from human embryonic stem cells hold much promise for clinical application in bone regeneration, in addition to providing a useful research model in developmental biology, and for pharmacological and cytotoxicity screening of bone-related biomaterials and drugs in vitro. Previously, osteogenic differentiation of human embryonic stem cells was achieved through dissociation of embryoid bodies by trypsinization, prior to culture with osteogenesis-promoting medium. This study therefore attempted a new approach: that is to achieve osteogenesis within intact human embryoid bodies. After 22 days of culture in osteogenesis-promoting medium comprising a cocktail of ascorbic acid, beta-glycerophosphate and dexamethasone, the attached embryoid bodies exhibited much cellular outgrowth and migration, and formed morphologically distinct nodule-like structures. These were somewhat similar to osteogenic nodules formed by mesenchymal stem cells, as reported by previous studies. Immunohistochemical staining and RT-PCR analysis confirmed the presence of osteogenic cells within these nodule-like structures. Additionally, the quantitative assay of osteocalcin secretion demonstrated a rapid sharp increase in osteocalcin expression on day 12 of in vitro culture, which could suggest the appearance of differentiated osteoblasts from day 12 onwards. Future work will attempt to investigate whether other cytokines, growth factors and chemical compounds could further enhance osteogenesis within intact human embryoid bodies.
Collapse
Affiliation(s)
- Tong Cao
- Stem Cell Laboratory, Faculty of Dentistry, National University of Singapore, 5 Lower Kent Ridge Road, 119074 Singapore, Singapore.
| | | | | | | | | | | | | | | | | |
Collapse
|
152
|
Baek KH, Lee WY, Oh KW, Tae HJ, Lee JM, Lee EJ, Han JH, Kang MI, Cha BY, Lee KW, Son HY, Kang SK. The effect of simvastatin on the proliferation and differentiation of human bone marrow stromal cells. J Korean Med Sci 2005; 20:438-44. [PMID: 15953866 PMCID: PMC2782200 DOI: 10.3346/jkms.2005.20.3.438] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Statins have been postulated to affect the bone metabolism. Recent experimental and epidemiologic studies have suggested that statins may also have bone protective effects. This study assessed the effects of simvastatin on the proliferation and differentiation of human bone marrow stromal cells (BMSCs) in an ex vivo culture. The bone marrow was obtained from healthy donors. Mononuclear cells were isolated and cultured to osteoblastic lineage. In the primary culture, 10(-6) M simvastatin diminished the mean size of the colony forming units-fibroblastic (CFU-Fs) and enhanced matrix calcification. At near confluence, the cells were sub-cultured. Thereafter, the alkaline phosphatase (ALP) activities of each group were measured by the time course of the secondary culture. Simvastatin increased the ALP activity in a dose dependent manner, and this stimulatory effect was more evident during the early period of culture. A 3-[4, 5-dimethylthiazol-2-yl]-2, 5-diphenyltetrazolium bromide (MTT) assay was performed during the secondary culture in order to estimate the effect of simvastatin on the proliferation of human BMSCs. When compared to the control group, simvastatin significantly decreased the proliferation of cells of each culture well. 10(-6) M of simvastatin also significantly enhanced the osteocalcin mRNA expression level. This study shows that simvastatin has a stimulatory effect on bone formation through osteoblastic differentiation, and has an inhibitory effect on the proliferative potential of human BMSCs.
Collapse
Affiliation(s)
- Ki Hyun Baek
- Department of Internal Medicine, The Catholic University of Korea, College of Medicine, Seoul
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
153
|
Yazawa H, Zimmermann B, Asami Y, Bernimoulin JP. Simvastatin Promotes Cell Metabolism, Proliferation, and Osteoblastic Differentiation in Human Periodontal Ligament Cells. J Periodontol 2005; 76:295-302. [PMID: 15974856 DOI: 10.1902/jop.2005.76.2.295] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Simvastatin is one of the cholesterol lowering drugs. Recent studies demonstrated that it has a bone stimulatory effect. Periodontal ligament (PDL) cells are believed to play an important role in periodontal regeneration; that is, they may differentiate into specific cells which make cementum, bone, and attachment apparatus. It would be of interest whether simvastatin has a positive effect on PDL cells. Therefore, effects of simvastatin on cell proliferation and osteoblastic differentiation in PDL cells were analyzed. METHODS Human PDL cells were cultured in monolayer with simvastatin for 24 and 72 hours and cell metabolism and proliferation were determined. To analyze osteoblastic differentiation, human PDL cells were cultured in organoid culture for 7, 14, and 21 days and alkaline phosphatase (ALP) activity, osteopontin (OPN), bone morphogenetic protein (BMP) -2, osteocalcin (OCN), and calcium contents were measured. They were co-treated by simvastatin and mevalonate. RESULTS Simvastatin enhanced cell proliferation and metabolism dose-dependently after 24 hours. Simvastatin also stimulated ALP activity of human PDL cells dose-dependently, and maximum effect was obtained at the concentration of 10(8) M. In time dependent analysis, 10(8) M simvastatin stimulated ALP activity and osteopontin content after 7 days and calcium contents after 21 days. BMP-2 and OCN contents were not detected. Moreover this statin-enhanced ALP activity was abolished by mevalonate. CONCLUSION These results suggest that at low concentration, simvastatin exhibits positive effect on proliferation and osteoblastic differentiation of human PDL cells, and these effects may be caused by the inhibition of the mevalonate pathway.
Collapse
Affiliation(s)
- Hiroshi Yazawa
- Department of Periodontology, Faculty of Dentistry, Campus Virchow Klinikum, Charité, University Medical School, Berlin, Germany.
| | | | | | | |
Collapse
|
154
|
Whang K, Grageda E, Khan A, McDonald J, Lawton M, Satsangi N. A novel osteotropic biomaterial OG-PLG:In vitro efficacy. J Biomed Mater Res A 2005; 74:247-53. [PMID: 15962265 DOI: 10.1002/jbm.a.30370] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Previously, a novel osteotropic biomaterial, OG-PLG [simvastatin grafted to poly(lactide-co-glycolide), PLG], was synthesized and shown to have degradation-controlled release kinetics. The objective here was to determine the effect of grafting statins to PLG on bone regeneration in vitro. Rat bone marrow cells were stimulated in vitro with simvastatin dissolved in media, saponified simvastatin dissolved in media, simvastatin released through diffusion from emulsion freeze-dried scaffolds, and OG-PLG. Unstimulated cultures and cultures stimulated with dexamethasone were used as negative and positive controls, respectively. In vitro bone formation was assessed using the alkaline phosphatase (ALP) and von Kossa assays at different times up to 16 days. ALP analysis revealed that saponified simvastatin at 10(-7)M and OG-PLG significantly increased ALP expression at various time points. von Kossa assay showed that simvastatin, saponified simvastatin, and OG-PLG significantly enhanced mineralization, with the effect from OG-PLG being the most significant. In short, OG-PLG significantly enhanced in vitro bone cell mineralization beyond the effect of simvastatin or saponified simvastatin dissolved in media and simvastatin released via diffusion from scaffolds.
Collapse
Affiliation(s)
- Kyumin Whang
- Department of Restorative Dentistry, Division of Biomaterials, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MSC 7890, San Antonio, TX 78229-3900, USA
| | | | | | | | | | | |
Collapse
|
155
|
Viereck V, Gründker C, Blaschke S, Frosch KH, Schoppet M, Emons G, Hofbauer LC. Atorvastatin stimulates the production of osteoprotegerin by human osteoblasts. J Cell Biochem 2005; 96:1244-53. [PMID: 16152630 DOI: 10.1002/jcb.20598] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Recently, HMG-CoA reductase inhibitors (statins), potent inhibitors of cholesterol biosynthesis, have been linked to protective effects on bone metabolism. Because of their widespread use, prevention of bone loss and fractures would be a desirable side effect. However, the mechanisms how statins may affect bone metabolism are poorly defined. Here, we evaluated the effect of atorvastatin on osteoblastic production of receptor activator of nuclear factor-kappaB ligand (RANKL) and osteoprotegerin (OPG), cytokines that are essential for osteoclast cell biology. While RANKL enhances osteoclast formation and activation, thereby, promoting bone loss, OPG acts as a soluble decoy receptor and antagonizes the effects of RANKL. In primary human osteoblasts (hOB), atorvastatin increased OPG mRNA levels and protein secretion by hOB by up to three fold in a dose-dependent manner with a maximum effect at 10(-6) M (P < 0.001). Time course experiments indicated a time-dependent stimulatory effect of atorvastatin on OPG mRNA levels after 24 h and on OPG protein secretion after 48-72 h (P < 0.001). Treatment of hOB with substrates of cholesterol biosynthesis that are downstream of the HMG-CoA reductase reaction (mevalonate, geranylgeranyl pyrophosphate) reversed atorvastatin-induced enhancement of OPG production. Of note, atorvastatin abrogated the inhibitory effect of glucocorticoids on OPG production. Treatment of hOB with atorvastatin enhanced the expression of osteoblastic differentiation markers, alkaline phosphatase and osteocalcin. In summary, our data suggest that atorvastatin enhances osteoblastic differentiation and production of OPG. This may contribute to the bone-sparing effects of statins.
Collapse
MESH Headings
- Adult
- Atorvastatin
- Cell Differentiation/drug effects
- Cells, Cultured
- Dose-Response Relationship, Drug
- Female
- Glucocorticoids/metabolism
- Glucocorticoids/pharmacology
- Glycoproteins/biosynthesis
- Glycoproteins/drug effects
- Glycoproteins/metabolism
- Heptanoic Acids/metabolism
- Heptanoic Acids/pharmacology
- Humans
- Male
- Mevalonic Acid/metabolism
- Models, Biological
- Osteoblasts/drug effects
- Osteoblasts/metabolism
- Osteoprotegerin
- Pyrroles/metabolism
- Pyrroles/pharmacology
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Receptors, Cytoplasmic and Nuclear/biosynthesis
- Receptors, Cytoplasmic and Nuclear/drug effects
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Tumor Necrosis Factor/biosynthesis
- Receptors, Tumor Necrosis Factor/drug effects
- Receptors, Tumor Necrosis Factor/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Time Factors
Collapse
Affiliation(s)
- Volker Viereck
- Department of Obstetrics and Gynecology, Georg-August-University, Goettingen, Germany.
| | | | | | | | | | | | | |
Collapse
|
156
|
Whang K, McDonald J, Khan A, Satsangi N. A novel osteotropic biomaterial OG-PLG: Synthesis andin vitro release. J Biomed Mater Res A 2005; 74:237-46. [PMID: 15981201 DOI: 10.1002/jbm.a.30309] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Statins (e.g., simvastatin) have shown to induce expression of the bone morphogenic protein-2 gene in bone cells, but they are not used clinically because of a lack of a suitable delivery device. The overall objective is to develop optimized statin delivery devices for bone regeneration. The specific objective was to determine the effect of grafting statins to biodegradable poly[lactide-co-glycolide] (PLG) on release kinetics. Simvastatin was grafted to PLG (OG-PLG) and characterized using contact-angle measurements, attenuated total reflectance-Fourier transform infrared, and ultraviolet-visible spectroscopy to determine success of the synthesis. An ultraviolet-visible assay for measuring release of statins and degraded OG-PLG in media was also developed. In vitro release studies using films and scaffolds made with PLG, PLG blended with simvastatin (PLG + Sim), and OG-PLG (simvastatin grafted to PLG) blended into PLG at different concentrations showed that release rate of OG-PLG from films was significantly greater than that of PLG + Sim. However, release rate from scaffolds showed PLG + Sim to be significantly higher than that of OG-PLG. The diffusion-controlled release kinetics of simvastatin from PLG + Sim seems to be more heavily affected by device morphology, whereas the degradation-controlled release kinetics seem to be less affected. In short, release kinetics can be modulated by grafting statins to PLG.
Collapse
Affiliation(s)
- Kyumin Whang
- Department of Restorative Dentistry, Division of Biomaterials, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MSC 7890, San Antonio, TX 78229-3900, USA.
| | | | | | | |
Collapse
|
157
|
Heng BC, Cao T, Stanton LW, Robson P, Olsen B. Strategies for directing the differentiation of stem cells into the osteogenic lineage in vitro. J Bone Miner Res 2004; 19:1379-94. [PMID: 15312238 DOI: 10.1359/jbmr.040714] [Citation(s) in RCA: 119] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2004] [Revised: 03/02/2004] [Accepted: 05/18/2004] [Indexed: 01/29/2023]
Abstract
A major area in regenerative medicine is the application of stem cells in bone reconstruction and bone tissue engineering. This will require well-defined and efficient protocols for directing the differentiation of stem cells into the osteogenic lineage, followed by their selective purification and proliferation in vitro. The development of such protocols would reduce the likelihood of spontaneous differentiation of stem cells into divergent lineages on transplantation, as well as reduce the risk of teratoma formation in the case of embryonic stem cells. Additionally, such protocols could provide useful in vitro models for studying osteogenesis and bone development, and facilitate the genetic manipulation of stem cells for therapeutic applications. The development of pharmokinetic and cytotoxicity/genotoxicity screening tests for bone-related biomaterials and drugs could also use protocols developed for the osteogenic differentiation of stem cells. This review critically examines the various strategies that could be used to direct the differentiation of stem cells into the osteogenic lineage in vitro.
Collapse
Affiliation(s)
- Boon Chin Heng
- Faculty of Dentistry, National University of Singapore, 119074 Singapore
| | | | | | | | | |
Collapse
|
158
|
Schoofs MWCJ, Sturkenboom MCJM, van der Klift M, Hofman A, Pols HAP, Stricker BHC. HMG-CoA reductase inhibitors and the risk of vertebral fracture. J Bone Miner Res 2004; 19:1525-30. [PMID: 15312254 DOI: 10.1359/jbmr.040607] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2004] [Revised: 04/19/2004] [Accepted: 05/03/2004] [Indexed: 11/18/2022]
Abstract
UNLABELLED Statins inhibit an enzyme in the mevalonate pathway and therefore may affect bone. In this first study on both symptomatic and nonsymptomatic vertebral fractures in the elderly (N = 3469), we show that long-term statin use is significantly associated with a 50% lower vertebral fracture risk. Randomized trials on statins and fractures, carried out in populations at risk for fractures, are needed. INTRODUCTION Statins are cholesterol-lowering agents that could potentially affect bone. Previous studies on statin use and fracture risk reported contradictory results and did not include both symptomatic and nonsymptomatic vertebral fractures. MATERIALS AND METHODS To examine the association between statin use, vertebral fractures, and lumbar spine BMD, we performed a prospective population-based cohort study in men and women (N = 3469) > or =55 years of age. These individuals had both baseline and follow-up spinal X-rays available. Statin use was obtained from detailed computerized pharmacy data, and the total number of days of exposure before second X-ray was calculated. A multivariate logistic regression model was fitted to calculate odds ratios and CIs. RESULTS During a mean follow-up of 6.5 years, 176 incident vertebral fractures occurred. There were 508 statin users and 16 exposed cases. The adjusted relative risk for incident vertebral fracture in users of statins (compared with nonusers) was 0.58 (95% CI, 0.34-0.99). The relative risk decreased on higher cumulative use to 0.52 (95% CI, 0.28-0.97) for use for more than 365 days during the study period. Use of (the hydrophilic statin) pravastatin and use of nonstatin cholesterol-lowering drugs was not significantly associated with vertebral fracture risk. Statin use was not significantly associated with lumbar spine BMD. CONCLUSION Statin use is associated with a lower risk of vertebral fracture. Randomized clinical trials in a population at risk for fracture are needed to examine this association.
Collapse
Affiliation(s)
- Mariette W C J Schoofs
- Department of Epidemiology and Biostatistics, Erasmus Medical Center, Rotterdam 3000 DR, The Netherlands
| | | | | | | | | | | |
Collapse
|
159
|
Ayukawa Y, Okamura A, Koyano K. Simvastatin promotes osteogenesis around titanium implants. A histological and histometrical study in rats. Clin Oral Implants Res 2004; 15:346-50. [PMID: 15142098 DOI: 10.1046/j.1600-0501.2003.01015.x] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVES Hydroxymethylglutaryl-coenzyme A reductase inhibitors, the so-called statins, have been widely used for hyperlipidemic patients, and it was recently reported that it promoted bone formation. In the present study, we examined the effect of simvastatin on the promotion of osteogenesis around titanium implants. MATERIALS AND METHODS Ten 30-week-old rats received pure titanium implants in both tibiae, and were then divided into experimental and control groups. The experimental group was administered simvastatin daily. Thirty days later, all animals were killed and then specimens were prepared. The bone contact ratio (BCR) to the implant and bone density (BD) around the implant, as well as histological findings, were obtained. RESULTS In the control group, newly formed bone could be seen around the implants. It was seen to be in direct contact with the implant surface, but otherwise unmineralized connective tissue was occasionally interposed. In the medullary canal, a scanty amount of bone trabeculae was observed. In the experimental group, in contrast, thicker bone trabeculae were abundantly seen in the medullary canal and showed a mesh-like structure. In the histometrical observations, both BCR and BD of the experimental group were significantly greater than those of the control group. CONCLUSION The administration of simvastatin increases the value of both BCR and BD. This drug may have the potential to improve the nature of osseointegration.
Collapse
Affiliation(s)
- Yasunori Ayukawa
- Section of Oral Reconstructive Biotechnology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Japan.
| | | | | |
Collapse
|
160
|
Abstract
OBJECTIVE To review the skeletal effects of hydroxy-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors (statins) and evaluate the use and potential benefit of statins in the management of osteoporosis. METHODS Background information on the topic of the effect of statins on bone and fracture risk is presented, and the pertinent published literature is reviewed. RESULTS Osteoporosis is the most common bone disease, affecting millions of people worldwide and leading to considerable morbidity, especially when it is not adequately managed. Although statins have primarily been known for their lipid-lowering effects, recent data have documented a potential association between statin use and improvement in fracture risk profile. Some statins have been shown to decrease bone-specific alkaline phosphatase or serum osteocalcin concentrations, but other studies have failed to demonstrate the beneficial effects of these agents. Therefore, additional studies should be undertaken to clarify the mechanism of action. CONCLUSION Observational studies suggest an association between HMG-CoA reductase inhibitors and reduction in fracture risk. Large randomized controlled clinical trials must be performed to confirm this association.
Collapse
Affiliation(s)
- Subhashini Yaturu
- Section of Endocrinology, Overton Brooks VA Medical Center/LSU Health Sciences Center, Shreveport, Louisiana 71101-4295, USA
| |
Collapse
|
161
|
Oxlund H, Andreassen TT. Simvastatin treatment partially prevents ovariectomy-induced bone loss while increasing cortical bone formation. Bone 2004; 34:609-18. [PMID: 15050891 DOI: 10.1016/j.bone.2003.12.014] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2003] [Revised: 10/08/2003] [Accepted: 12/03/2003] [Indexed: 11/22/2022]
Abstract
Statins are commonly prescribed drugs that inhibit hepatic cholesterol synthesis and thereby reduce serum cholesterol concentrations. Some of the statins are thought to possess bone anabolic properties. Effects of statin on tibia, femur, and vertebral cortical and cancellous bone were studied in ovariectomized (OVX) rats. Sixty Wistar female rats, 4 months old, were allocated into four groups: baseline control, sham + placebo group, OVX + placebo, OVX + simvastatin. Simvastatin, 20 mg/kg, or placebo was given twice daily by a gastric tube for 3 months. The rats were labeled with tetracycline at day 11 and calcein at day 4 before sacrifice. Concerning cortical bone, the tibial diaphysis bending strength was increased by 8% and the periosteal bone formation rate (BFR) at the mid-diaphysis increased by twofold in the OVX + simvastatin group compared with the OVX + placebo group, in harmony with increased serum osteocalcin concentrations. Simvastatin did not affect the endocortical bone formation. Concerning cancellous bone, the cancellous bone volumes in the proximal tibia and vertebral body were reduced in both OVX groups, but the reduction was less in the OVX + simvastatin group compared with the OVX + placebo group. This reduction in cancellous bone loss is in agreement with the 36% decreased activity of serum tartrate-resistant-acid-phosphatase 5b (TRAP-5b), indicating decreased osteoclast activity in the OVX + simvastatin group compared with the OVX + placebo group. In conclusion, simvastatin induces a moderate increase in cortical bone formation at the periosteal bone surface. The new cortical bone exhibits a normal lamellar structure, and simvastatin seems to respect the regional pattern of bone formation, bone resorption, and drift; for example, no periosteal bone formation is observed in the vertebral canal. Furthermore, simvastatin reduces the loss of cancellous bone induced by ovariectomy.
Collapse
Affiliation(s)
- Hans Oxlund
- Department of Connective Tissue Biology, Institute of Anatomy, University of Aarhus, DK-8000 Aarhus C, Denmark.
| | | |
Collapse
|
162
|
Schlienger RG, Meier CR. HMG-CoA reductase inhibitors in osteoporosis: do they reduce the risk of fracture? Drugs Aging 2004; 20:321-36. [PMID: 12696993 DOI: 10.2165/00002512-200320050-00002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Osteoporosis affects a large number of people in industrialised countries. It has clinical and public-health impacts, most importantly due to subsequent fractures. Osteoporotic fractures are one of the most common causes of disability and are associated with enormous healthcare expenditure. The majority of existing treatment options for osteoporosis only inhibit bone resorption and prevent excessive bone loss but are not capable of stimulating bone formation. However, several recent in vitro and in vivo studies in animals demonstrated that HMG-CoA reductase inhibitors stimulate the production of bone morphogenetic protein (BMP-2), which is a potent regulating protein in osteoblast differentiation and activity. This suggests that HMG-CoA reductase inhibitors may have an anabolic effect on bones, making them a potentially interesting treatment option for osteoporosis. Additionally, several studies in humans showed that some HMG-CoA reductase inhibitors may have a beneficial effect on bone turnover and may lead to an increase in bone mineral density. Consequently, several observational studies tried to evaluate whether use of HMG-CoA reductase inhibitors is associated with a decreased risk of fractures. Even though not all results of these epidemiological studies, using different designs in different study populations, were entirely consistent, they provided substantial evidence that HMG-CoA reductase inhibitor use may decrease the bone fracture risk by approximately 50%. On the other hand, reanalysis of two randomised controlled trials of HMG-CoA reductase inhibitor therapy, designed to assess cardiovascular outcomes, could not show that patients treated with HMG-CoA reductase inhibitors had a lower fracture risk in comparison with placebo-treated patients. Therefore, to conclusively assess the potential of HMG-CoA reductase inhibitors in the prevention and treatment of osteoporosis, randomised controlled trials need to be performed to address this conflicting issue. Until the results of such trials are available, practitioners should prescribe the drugs that have been proven to reduce the risk of osteoporotic fractures.
Collapse
Affiliation(s)
- Raymond G Schlienger
- Basel Pharmacoepidemiology Unit, Division of Clinical Pharmacology, University Hospital, Basel, Switzerland.
| | | |
Collapse
|
163
|
Hatano H, Maruo A, Bolander ME, Sarkar G. Statin stimulates bone morphogenetic protein-2, aggrecan, and type 2 collagen gene expression and proteoglycan synthesis in rat chondrocytes. J Orthop Sci 2004; 8:842-8. [PMID: 14648275 DOI: 10.1007/s00776-003-0724-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2003] [Accepted: 08/07/2003] [Indexed: 11/26/2022]
Abstract
Statins increase bone morphogenetic protein-2 (BMP-2) mRNA expression and subsequently increase new bone formation in vitro. However, the action of statins on the BMP-2 mRNA regulation of cartilage matrix synthesis by chondrocytes is unknown. We evaluated regulation of BMP-2, aggrecan, and type II collagen (COL2) mRNA and (35)S-labeled proteoglycan (PG) synthesis by mevastatin using cultured chondrocytes obtained from articular cartilage of fetal rats. Expression of BMP-2, aggrecan, and COL2 mRNAs were increased in the presence of 2 microM mevastatin on day 2. However, longer (10 day) culture in the presence of the drug decreased the expression of these mRNAs. PG synthesis was increased 3 days after treating the cells with mevastatin, which was also decreased with longer (10 day) mevastatin treatment. These results suggest that mevastatin increases mRNA expression of BMP-2, aggrecan, and COL2 as well as PG synthesis by fetal rat chondrocytes early in the treatment period. We suggest that statins have implications for fracture and cartilage repair.
Collapse
Affiliation(s)
- Hiroshi Hatano
- Department of Orthopedic Research, Mayo Clinic and Foundation, MSB 3-69, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
164
|
Hwang R, Lee EJ, Kim MH, Li SZ, Jin YJ, Rhee Y, Kim YM, Lim SK. Calcyclin, a Ca2+ ion-binding protein, contributes to the anabolic effects of simvastatin on bone. J Biol Chem 2004; 279:21239-47. [PMID: 14973129 DOI: 10.1074/jbc.m312771200] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In vitro treatment with a pharmacological dose of simvastatin, a potent pro-drug of a 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitor, stimulates bone formation. In our study, simvastatin stimulated differentiation of osteoblasts remarkably in a dose-dependent manner, with minimal effect on proliferation. To identify the mediators of the anabolic effects of simvastatin on osteoblasts, we tried to identify and characterize simvastatin-induced proteins by using proteomic analysis. Calcyclin was significantly up-regulated by more than 10 times, and annexin I was also up-regulated by simvastatin. However, annexin III, vimentin, and tropomyosin were down-regulated. Up-regulated calcyclin mRNA by simvastatin was validated by reverse transcription in mouse calvarial cells. In confocal microscope analysis, green fluorescence protein-calcyclin fusion protein was ubiquitously observed in the of MC3T3-E1 cells transfected with green fluorescence protein-calcyclin cDNA containing plasmid and was quickly concentrated in the nucleus 20 min after simvastatin treatment. Overexpression of calcyclin cDNA stimulated both the proliferation and expression of alkaline phosphatase mRNA significantly, without exposure to simvastatin in MC3T3-E1 cells. However, both the rate of proliferation of the osteoblasts and the expression of alkaline phosphatase mRNA were suppressed significantly 1 day after treatment with the calcyclin-specific small interference RNA, and furthermore, simvastatin did not overcome this suppression in the small interference RNA-pretreated MC3T3-E1 cells. In conclusion, calcyclin is one of the candidate proteins that plays a role in osteoblastogenesis in response to simvastatin, although the precise functions of calcyclin in osteoblast remain to be verified.
Collapse
Affiliation(s)
- Ranjoo Hwang
- Departments of Internal Medicine and Anatomy, College of Medicine, Yonsei University, Seoul 120-752, Korea
| | | | | | | | | | | | | | | |
Collapse
|
165
|
Maeda T, Matsunuma A, Kurahashi I, Yanagawa T, Yoshida H, Horiuchi N. Induction of osteoblast differentiation indices by statins in MC3T3-E1 cells. J Cell Biochem 2004; 92:458-71. [PMID: 15156558 DOI: 10.1002/jcb.20074] [Citation(s) in RCA: 188] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Statins inhibit 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase, which catalyzes conversion of HMG-CoA to mevalonate, a rate-limiting step in cholesterol synthesis. The present study was undertaken to understand the events of osteoblast differentiation induced by statins. Simvastatin at 10(-7) M markedly increased mRNA expression for bone morphogenetic protein-2 (BMP-2), vascular endothelial growth factor (VEGF), alkaline phosphatase, type I collagen, bone sialoprotein, and osteocalcin (OCN) in nontransformed osteoblastic cells (MC3T3-E1), while suppressing gene expression for collagenase-1, and collagenase-3. Extracellular accumulation of proteins such as VEGF, OCN, collagenase-digestive proteins, and noncollagenous proteins was increased in the cells treated with 10(-7) M simvastatin, or 10(-8) M cerivastatin. In the culture of MC3T3-E1 cells, statins stimulated mineralization; pretreating MC3T3-E1 cells with mevalonate, or geranylgeranyl pyrophosphate (a mevalonate metabolite) abolished statin-induced mineralization. Statins stimulate osteoblast differentiation in vitro, and may hold promise drugs for the treatment of osteoporosis in the future.
Collapse
Affiliation(s)
- Toyonobu Maeda
- Department of Biochemistry, School of Dentistry, Ohu University, Koriyama 963-8611, Japan
| | | | | | | | | | | |
Collapse
|
166
|
Whitney C, Warburton DER, Frohlich J, Chan SY, McKay H, Khan K. Are Cardiovascular Disease and Osteoporosis Directly Linked? Sports Med 2004; 34:779-807. [PMID: 15462612 DOI: 10.2165/00007256-200434120-00001] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
For years, osteoporosis and cardiovascular disease were thought to be two independent consequences of aging; however, mounting evidence supports an association between these diseases. Recently, a widespread class of cholesterol-lowering drugs known as statins have demonstrated (in rodents and cell cultures) the ability to induce bone formation. This finding is significant since current therapies are limited to the prevention or slowing down of bone loss rather than (enhancing/improving) bone formation. In humans, the ability of statins to generate new bone has not been consistent; however, several investigations have demonstrated a dramatic decrease in fracture risk. Although it has been proposed that statins induce new bone via increased bone morphogenetic protein-2, other conditions affected by statins such as dyslipidaemia, vascular calcification, endothelial dysfunction and impaired nitric oxide expression, may also contribute to the cardiovascular and bone health paradigm. Furthermore, the role of physical activity and its influence on cardiovascular and bone health, especially in postmenopausal women, may contribute to the discrepancy of findings in human data. In summary, it remains to be determined if statins contribute to bone health via improvements in vascular health or by pleiotropic properties unique to their pharmacology. This review provides information on our current understanding of the bone and cardiovascular association, as well as on novel areas of research to further our current understanding of these conditions.
Collapse
Affiliation(s)
- Crystal Whitney
- Healthy Heart Program, St Paul's Hospital, Vancouver, British Columbia V6T 1Z3, Canada
| | | | | | | | | | | |
Collapse
|
167
|
Baghdasarian SB, Jneid H, Hoogwerf BJ. Association of dyslipidemia and effects of statins on nonmacrovascular diseases. Clin Ther 2004; 26:337-51. [PMID: 15110128 DOI: 10.1016/s0149-2918(04)90031-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2004] [Indexed: 02/02/2023]
Abstract
BACKGROUND Statins have mechanisms of action that expand their effects beyond cholesterol lowering and atherosclerotic medical conditions. OBJECTIVE This review summarizes clinical evidence for the association of dyslipidemia and the effects of statin use on aortic stenosis, Alzheimer's dementia (AD), osteoporosis, prevention of diabetes mellitus (DM), diabetic retinopathy, age-related macular degeneration, and diabetic/nondiabetic nephropathy. METHODS An English-language literature search was conducted using MEDLINE (1966-June 2003). Bibliographies of retrieved articles were reviewed. Search terms included statin, HMG-CoA reductase inhibitors, aortic stenosis, Alzheimer's dementia, osteoporosis, prevention of diabetis, diabetic retinopathy, age-related macular degeneration, diabetic nephropathy, and nondiabetic nephropathy. RESULTS Three retrospective cohort trials have shown an association between statin use and the progression of aortic stenosis; one of these trials observed a 45% decrease in aortic valve area in 1 year. In AD, one cross-sectional analysis found 60% to 73% lower AD rates in lovastatin or pravastatin recipients ( P<0.001 ). Of the multiple observational studies on the effect of statins on fracture risk, some have shown a decreased risk, with an odds ratio as low as 0.50 (95% CI, 0.33-0.76); others have demonstrated no association. A post hoc analysis of the West of Scotland Coronary Prevention Study found a 30% reduction in the development of DM ( P=0.042 ), but this was not duplicated in the Anglo-Scandinavian Cardiac Outcomes Trial-Lipid Lowering Arm. A small clinical trial of 6 patients (11 eyes) demonstrated improved retinal hard exudates with pravastatin treatment in patients with diabetic retinopathy. In a cross-sectional analysis, age-related macular degeneration was found to be less common among statin users than nonusers (4% [ 1/27 ] vs 22% [ 76/352 ]; P=0.02. Multiple small clinical trials of 19 to 56 patients with diabetic and nondiabetic nephropathy at various stages generated inconsistent results for an association between statin use and decreased albumin excretion rate and decreased rate of decline in glomerular filtration. CONCLUSION Data of variable quantity and quality support the use of statins as adjuncts in the treatment of nonmacrovascular diseases.
Collapse
Affiliation(s)
- Sarkis B Baghdasarian
- Cardiovascular Medicine Department, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA.
| | | | | |
Collapse
|
168
|
Kizu A, Shioi A, Jono S, Koyama H, Okuno Y, Nishizawa Y. Statins inhibit in vitro calcification of human vascular smooth muscle cells induced by inflammatory mediators. J Cell Biochem 2004; 93:1011-9. [PMID: 15389884 DOI: 10.1002/jcb.20207] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Although lipid-lowering therapy with 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) decreases the progression of coronary artery and aortic valve calcification, the mechanism of action of these drugs to inhibit the calcification process remains unclear. In this study, we investigated the effect of statins such as cerivastatin and atorvastatin on vascular calcification by utilizing an in vitro model of inflammatory vascular calcification. Cerivastatin and atorvastatin dose-dependently inhibited in vitro calcification of human vascular smooth muscle cells (HVSMCs) induced by the following inflammatory mediators (IM): interferon-gamma, 1alpha,25-dihydroxyvitamin D3, tumor necrosis factor-alpha, and oncostatin M. These statins also depressed expression of alkaline phosphatase (ALP) in HVSMCs induced by these factors. Mevalonate and geranylgeranylpyrophosphate reversed the inhibitory effect of cerivastatin on ALP expression in HVSMCs, while farnesylpyrophosphate showed no effect on the ALP activities inhibited by this drug, suggesting that inhibition of Rho and its downstream target, Rho kinase may mediate the inhibitory effect of cerivastatin. Cerivastatin prevented RhoA activation in HVSMCs induced by the IM. A specific inhibitor of Rho kinase (Y-27632) inhibited in vitro calcification and induction of ALP in HVSMCs. These findings provide a possible mechanism of statins to prevent the progression of calcification in inflammatory vascular diseases such as atherosclerosis and cardiac valvular calcification.
Collapse
MESH Headings
- Alkaline Phosphatase/metabolism
- Atorvastatin
- Calcinosis
- Dose-Response Relationship, Drug
- Heptanoic Acids/pharmacology
- Humans
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology
- Inflammation/metabolism
- Mevalonic Acid/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/immunology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/immunology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Polyisoprenyl Phosphates/metabolism
- Pyridines/pharmacology
- Pyrroles/pharmacology
- Sesquiterpenes
Collapse
Affiliation(s)
- Akane Kizu
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | | | | | | | | | | |
Collapse
|
169
|
Chaudhry GR, Yao D, Smith A, Hussain A. Osteogenic Cells Derived From Embryonic Stem Cells Produced Bone Nodules in Three-Dimensional Scaffolds. J Biomed Biotechnol 2004; 2004:203-210. [PMID: 15467160 PMCID: PMC555770 DOI: 10.1155/s111072430431003x] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2003] [Revised: 02/27/2004] [Accepted: 03/18/2004] [Indexed: 11/17/2022] Open
Abstract
An approach for 3D bone tissue generation from embryonic stem (ES) cells was investigated. The ES cells were induced to differentiate into osteogenic precursors, capable of proliferating and subsequently differentiating into bone-forming cells. The differentiated cells and the seeded scaffolds were characterized using von Kossa and Alizarin Red staining, electron microscopy, and RT-PCR analysis. The results demonstrated that ES-derived bone-forming cells attached to and colonized the biocompatible and biodegradable scaffolds. Furthermore, these cells produced bone nodules when grown for 3-4 weeks in mineralization medium containing ascorbic acid and beta-glycerophosphate both in tissue culture plates and in scaffolds. The differentiated cells also expressed osteospecific markers when grown both in the culture plates and in 3D scaffolds. Osteogenic cells expressed alkaline phosphatase, osteocalcin, and osteopontin, but not an ES cell-specific marker, oct-4. These findings suggest that ES cell can be used for in vitro tissue engineering and cultivation of graftable skeletal structures.
Collapse
Affiliation(s)
- G. R. Chaudhry
- Department of Biological Sciences, Oakland University, Rochester,
MI 48309, USA
| | - D. Yao
- Department of Mechanical Engineering, Oakland University,
Rochester, MI 48309, USA
| | - A. Smith
- Department of Biological Sciences, Oakland University, Rochester,
MI 48309, USA
| | - A. Hussain
- Department of Biological Sciences, Oakland University, Rochester,
MI 48309, USA
| |
Collapse
|
170
|
Thunyakitpisal PD, Chaisuparat R. Simvastatin, an HMG-CoA Reductase Inhibitor, Reduced the Expression of Matrix Metalloproteinase-9 (Gelatinase B) in Osteoblastic Cells and HT1080 Fibrosarcoma Cells. J Pharmacol Sci 2004; 94:403-9. [PMID: 15107580 DOI: 10.1254/jphs.94.403] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
MMP-9 or Gelatinase B, a member of the matrix metalloproteinase family (MMPs), plays important roles in physiological events such as tissue remodeling and in pathological processes that lead to destructive bone diseases, including osteoarthritis and periodontitis. In addition to its effect on the increase of total bone mass, statin (an HMG-CoA reductase inhibitor) suppresses the expression of MMPs. In this study, we proposed that simvastatin reduces MMP-9 expression in osteoblasts and HT1080 fibrosarcoma cell line. Gelatin zymography, Western blot analysis and reverse transcriptase-PCR were used to investigate the effects of simvastatin on MMP-9 in primary calvaria cells, U2-OS osteosarcoma cells, and HT1080 fibrosarcoma cells. The results from gelatin zymography and Western blot analysis revealed that simvastatin suppressed MMP-9 activity in these cells in concentration- and time-dependent manners. The effective concentrations of simvastatin were 100 - 500 nM, 5 - 15 microM, and 2.5 - 10 microM in primary calvaria, U2-OS, and HT1080 cells, respectively. Collectively, these results suggest that simvastatin is a potent drug for inhibition of MMP-9 expression in osteoblastic cells and HT1080 fibrosarcoma cells.
Collapse
|
171
|
Calabro P, Yeh ETH. Multitasking of the 3-hydroxy-3-methylglutaryl coenzyme a reductase inhibitor: Beyond cardiovascular diseases. Curr Atheroscler Rep 2004; 6:36-41. [PMID: 14662106 DOI: 10.1007/s11883-004-0114-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Statins can profoundly affect cellular metabolism by inhibiting 3-hydroxy-3-methylglutaryl coenzyme A reductase, which is the rate-limiting enzyme responsible for cholesterol synthesis. Many physicians prescribe statins to lower plasma cholesterol levels, which has beneficial effects in both the primary and secondary prevention of coronary artery disease. However, in vitro, in vivo, animal, and clinical studies have all shown that statins may also have important pleiotropic properties. In fact, a number of clinical studies have suggested that statins are involved in modulating diseases such as cancer, osteoporosis, and dementia (including Alzheimer's disease). However, because these studies have been only preliminary and observational in nature, large randomized, placebo-controlled studies are needed to confirm the modulatory role of statins in these important diseases.
Collapse
Affiliation(s)
- Paolo Calabro
- Department of Cardiology, The University of Texas-MD Anderson Cancer Center, 1515 Holcombe Boulevard, Box 449, Houston, TX 77030-4095, USA
| | | |
Collapse
|
172
|
King DS, Wilburn AJ, Wofford MR, Harrell TK, Lindley BJ, Jones DW. Cognitive Impairment Associated with Atorvastatin and Simvastatin. Pharmacotherapy 2003; 23:1663-7. [PMID: 14695047 DOI: 10.1592/phco.23.15.1663.31953] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Clinical guidelines for cholesterol testing and management have been updated recently. With the evolving recognition of benefits and intensified recommendations for cholesterol management, many more patients will require cholesterol-lowering drugs. All the statins share similar adverse-effect profiles, with a low overall frequency of undesirable effects. Emerging data associate statins with a decreased risk of Alzheimer's disease; however, we report two women who experienced significant cognitive impairment temporally related to statin therapy. One woman took atorvastatin, and the other first took atorvastatin, then was rechallenged with simvastatin. Clinicians should be aware of cognitive impairment and dementia as potential adverse effects associated with statin therapy.
Collapse
Affiliation(s)
- Deborah S King
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA.
| | | | | | | | | | | |
Collapse
|
173
|
Yamada S, Ohara N, Hayashi Y. Mineralization of matrix vesicles isolated from a human osteosarcoma cell line in culture with water-soluble chitosan-containing medium. J Biomed Mater Res A 2003; 66:500-6. [PMID: 12918032 DOI: 10.1002/jbm.a.10009] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Chitosan is a natural bioactive material. Although it has been reported that chitosan promotes osteogenesis in bone lesions, little is known about how chitosan modulates this process. The present study was designed to investigate the effect of water-soluble chitosan relative to initiation of biologic mineralization, especially in the matrix-vesicles-(MVs) mediated process in vitro. A human osteoblastic cell line (NOS-1) was used. After 3 days of incubation, the number of cells and alkaline phosphatase (ALP) activity increased significantly in the chitosan group. RT-PCR analysis revealed that chitosan induced an increase in the expression of bone morphogenetic protein-2 mRNA after 7 days of incubation. MVs were isolated from NOS-1 cells using a collagenase digestion and ultracentrifugation method. ALP activity of MVs isolated from chitosan-supplemented cells was significantly higher than that of the control group. Furthermore, isolated MVs were incubated in medium supplemented with Na-beta-glycerophosphate without fetal bovine serum. Needle-like crystals were observed in association with MVs after 24 h of incubation. These needle-like crystals were densely accumulated in the chitosan group. The present findings suggest that water-soluble chitosan would promote osteoblast proliferation and differentiation and may be useful for the acceleration of initial biologic mineralization.
Collapse
Affiliation(s)
- Shizuka Yamada
- Division of Cardiology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | | | | |
Collapse
|
174
|
Song C, Guo Z, Ma Q, Chen Z, Liu Z, Jia H, Dang G. Simvastatin induces osteoblastic differentiation and inhibits adipocytic differentiation in mouse bone marrow stromal cells. Biochem Biophys Res Commun 2003; 308:458-62. [PMID: 12914771 DOI: 10.1016/s0006-291x(03)01408-6] [Citation(s) in RCA: 148] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
To clarify the mechanism of the stimulatory effect of statins on bone formation, we investigated the effect of simvastatin, a widely used statin, on osteoblastic and adipocytic differentiation in primary cultured mouse bone marrow stromal cells (BMSCs). Simvastatin treatment enhanced the expression level of mRNA for osteocalcin and protein for osteocalcin and osteopontin, and increased alkaline phosphatase activity significantly (p<0.05). After BMSCs were exposed to an adipocyte differentiation agonist, Oil Red O staining, fluorescence activated cell sorting, and decreased expression level of lipoprotein lipase mRNA showed that treatment with simvastatin significantly inhibits adipocytic differentiation compared to controls that did not receive simvastatin (p<0.05). Lastly, we found that simvastatin induces high expression of BMP(2) in BMSCs. These observations suggested that simvastatin acts on BMSCs to enhance osteoblastic differentiation and inhibits adipocytic differentiation; this effect is at least partially mediated by inducing BMP(2) expression in BMSCs.
Collapse
Affiliation(s)
- Chunli Song
- Department of Orthopedics, Peking University Third Hospital, Beijing, PR China
| | | | | | | | | | | | | |
Collapse
|
175
|
Wong RWK, Rabie ABM. Statin collagen grafts used to repair defects in the parietal bone of rabbits. Br J Oral Maxillofac Surg 2003; 41:244-8. [PMID: 12946667 DOI: 10.1016/s0266-4356(03)00081-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We compared the amount of new bone produced by statin collagen grafts with that produced by collagen grafts. Fifteen bone defects were created in the parietal bone of nine New Zealand White rabbits. In the experimental group, five defects were grafted with simvastatin dissolved in water for injection mixed with absorbable collagen sponge. In the control groups, five defects were grafted with water for injection mixed with absorbable collagen sponge alone (active control) and five were left empty (passive control). Animals were killed on day 14 and the defects were prepared for histological assessment. Serial sections were cut across the whole defect. Quantitative analysis of new bone formation was made on 100 sections using image analysis. A total of 308% more new bone was present in defects grafted with statin collagen grafts than those grafted with collagen grafts alone (P<0.0001). No bone was formed in the passive control group. In conclusion, statin collagen grafts were osteoinductive and can be used as a material for bone grafts.
Collapse
Affiliation(s)
- R W K Wong
- Orthodontics, University of Hong Kong, Government of Hong Kong SAR, Hong Kong
| | | |
Collapse
|
176
|
Bauer DC. HMG CoA reductase inhibitors and the skeleton: a comprehensive review. Osteoporos Int 2003; 14:273-82. [PMID: 12736772 DOI: 10.1007/s00198-002-1323-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2002] [Accepted: 09/10/2002] [Indexed: 02/06/2023]
Abstract
Recent studies suggest that the mevalonate pathway plays an important role in skeletal metabolism. HMG CoA reductase inhibitors ("statins"), which inhibit a key enzyme in the mevalonate pathway, are widely used for the treatment of hyperlipidemia. In vitro and animal studies demonstrate that statins stimulate the production of BMP-2, a potent regulator of osteoblast differentiation and activity, suggesting that statins may have an anabolic effect on bone. Statin use in most, but not all observational studies is associated with a reduced risk of fracture, particularly hip fracture, even after adjustment for the confounding effects of age, weight and other medication use. This beneficial effect has not been observed in clinical trials designed to assess cardiovascular endpoints. The effects of statins on bone mass and bone turnover are controversial, but increased bone mass and reduced bone turnover have been observed in controlled studies. Further studies of the skeletal effects of statins are needed, particularly their effects on surrogate markers such as bone mass, bone turnover, and microarchitecture, to determine the optimal formulation, dosing and route of administration. Clinical trials with fracture endpoints are needed before statins can be recommended as therapeutic agents for osteoporosis.
Collapse
Affiliation(s)
- D C Bauer
- Department of Medicine, Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California 94105, USA.
| |
Collapse
|
177
|
Waldman A, Kritharides L. The pleiotropic effects of HMG-CoA reductase inhibitors: their role in osteoporosis and dementia. Drugs 2003; 63:139-52. [PMID: 12515562 DOI: 10.2165/00003495-200363020-00002] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
HMG-CoA reductase is the rate-limiting enzyme for cholesterol synthesis and its inhibition exerts profound effects on cellular metabolism. Inhibitors of this enzyme are used in clinical practice to lower plasma cholesterol levels and are commonly collectively referred to as 'statins'. A number of in vitro, in vivo animal, and clinical studies suggest that properties of statins other than cholesterol lowering may be of biological importance. These diverse properties are often referred to as 'pleiotropic' and suggest that statins may affect a number of diseases of ageing. In this article we review the biological plausibility and clinical evidence of a role for statins in modulating two diseases of ageing: osteoporosis and dementia (including Alzheimer's disease). In both diseases, there is a sound cellular and laboratory basis for a plausible therapeutic effect of statins. In the case of osteoporosis, there are conflicting data regarding clinical benefit, with both negative and positive results reported. In particular, secondary analyses of randomised, controlled studies have shown no reduction of fracture risk by statins. In the case of dementia there are fewer clinical studies but there is clear anticipated benefit in macrovascular dementias attributable to statin-mediated reduction of the risk of stroke. Overall, there are a lack of prospective, placebo-controlled, randomised data testing statins and modulation of the risk of osteoporosis-related fracture or of clinical dementia, where these are primary outcomes. Until such data are available, the use of statins appears promising but cannot be recommended as a primary therapeutic modality for either condition.
Collapse
Affiliation(s)
- Alla Waldman
- Department of Cardiology, Concord Hospital, University of Sydney, NSW, Australia
| | | |
Collapse
|
178
|
Montagnani A, Gonnelli S, Cepollaro C, Pacini S, Campagna MS, Franci MB, Lucani B, Gennari C. Effect of simvastatin treatment on bone mineral density and bone turnover in hypercholesterolemic postmenopausal women: a 1-year longitudinal study. Bone 2003; 32:427-33. [PMID: 12689687 DOI: 10.1016/s8756-3282(03)00034-6] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Although several studies have reported a lower risk of osteoporotic fracture in hypercholesterolemic patients treated with statins, so far longitudinal studies on the effects of statins on bone are lacking. The aim of the present study was to evaluate bone mineral density (BMD) and bone turnover changes induced by 1-year simvastatin treatment on postmenopausal women. Thirty consecutive postmenopausal hypercholesterolemic women (61.2 +/- 4.9 years) were treated for 12 months with 40 mg/day simvastatin and 30 normocholesterolemic age-matched postmenopausal women provided control data. In all subjects, at baseline and at 3-month intervals, serum lipids, calcium, phosphate, total and bone alkaline phosphatase (Bone-ALP), and carboxy-terminal fragment of type I collagen (CTx) were measured in a fasting blood sample. At baseline and after 6 and 12 months BMD was measured at lumbar spine (BMD-LS) and at femur (BMD-Ftot) and at femoral neck (BMD-Fn) by DXA. In the simvastatin-treated group Bone-ALP showed a significant increase (P < 0.05) with respect to baseline from the sixth month, whereas serum CTx showed a weak and nonsignificant increase over the study period. In treated women BMD-LS, BMD-Fn, and BMD-Ftot increased respectively by 1.1, 0.9, and 0.4% at Month 6; and by 2.8, 1.0, and 0.8% at Month 12. In controls BMD-LS, BMD-Fn, and BMD-Ftot at the end of the study period decreased by 1.6, 1.4, and 1.2%, respectively. The difference between controls and simvastatin-treated patients was significant (P < 0.05) for both BMD-LS and BMD-Fn only at Month 12. In conclusion our results, although obtained from a small sample of postmenopausal hypercholesterolemic women, suggest a probable positive effect of simvastatin on bone formation and BMD.
Collapse
Affiliation(s)
- A Montagnani
- Department of Internal Medicine, University of Siena, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
179
|
Abstract
Anabolic agents represent an important new advance in the therapy of osteoporosis. Their potential might be substantially greater than the anti-resorptives. Because the anti-resorptives and anabolic agents work by completely distinct mechanisms of action, it is possible that the combination of agents could be significantly more potent than either agent alone. Recent evidence suggests that a plateau in BMD might occur after prolonged exposure to PTH. Anti-resorptive therapy during or after anabolic therapy might prevent this skeletal adaptation. Protocols to consider anabolic agents as intermittent recycling therapy would be of interest. Of all the anabolics, PTH is the most promising. However, there are unanswered questions about PTH. More studies are needed to document an anabolic effect on cortical bone. More large-scale studies are needed to further determine the reduction in nonvertebral fractures with PTH, especially at the hip. In the future, PTH is likely to be modified for easier and more targeted delivery. Oral or transdermal delivery systems may become available. Recently, Gowen et al have described an oral calcilytic molecule that antagonizes the parathyroid cell calcium receptor, thus stimulating the endogenous release of PTH. This approach could represent a novel endogenous delivery system for intermittent PTH administration. Rising expectations that anabolic therapies for osteoporosis will soon play a major role in treating this disease are likely to fuel further studies and the development of even more novel approaches to therapy.
Collapse
Affiliation(s)
- Mishaela R Rubin
- Department of Medicine, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | | |
Collapse
|
180
|
Maeda T, Kawane T, Horiuchi N. Statins augment vascular endothelial growth factor expression in osteoblastic cells via inhibition of protein prenylation. Endocrinology 2003; 144:681-92. [PMID: 12538631 DOI: 10.1210/en.2002-220682] [Citation(s) in RCA: 167] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Statins such as simvastatin are 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors that inhibit cholesterol synthesis. We presently investigated statin effects on vascular endothelial growth factor (VEGF) expression in osteoblastic cells. Hydrophobic statins including simvastatin, atorvastatin, and cerivastatin-but not a hydrophilic statin, pravastatin-markedly increased VEGF mRNA abundance in nontransformed osteoblastic cells (MC3T3-E1). Simvastatin (10(-6) M) time-dependently augmented VEGF mRNA expression in MC3T3-E1 cells, mouse stromal cells (ST2), and rat osteosarcoma cells (UMR-106). According to heterogeneous nuclear RNA and Northern analyses, 10(-6) M simvastatin stimulated gene expression for VEGF in MC3T3-E1 cells without altering mRNA stability. Transcriptional activation of a VEGF promoter-luciferase construct (-1128 to +827), significantly increased by simvastatin administration. As demonstrated by gel mobility shift assay, simvastatin markedly enhanced the binding of hypoxia-responsive element-protein complexes. These results indicate that the stimulation of the VEGF gene by simvastatin in MC3T3-E1 cells is transcriptional in nature. VEGF secretion into medium was increased in MC3T3-E1 by 10(-6) M simvastatin. Pretreating MC3T3-E1 cells with mevalonate or geranylgeranyl pyrophosphate, a mevalonate metabolite, abolished simvastatin-induced VEGF mRNA expression; manumycin A, a protein prenylation inhibitor, mimicked statin effects on VEGF expression. The effect of simvastatin was blocked by pretreatment with wortmannin and LY294002, specific phosphatidylinositide-3 kinase inhibitors. Simvastatin enhanced mineralized nodule formation in culture, whereas coincubation with mevalonate, geranylgeranyl pyrophosphate, LY294002, or VEGF receptor 2 inhibitor (SU1498) abrogated statin-induced mineralization. Thus, statins stimulate VEGF expression in osteoblasts via reduced protein prenylation and the phosphatidylinositide-3 kinase pathway, promoting osteoblastic differentiation.
Collapse
Affiliation(s)
- Toyonobu Maeda
- Department of Biochemistry, Ohu University School of Dentistry, Koriyama 963-8611, Japan
| | | | | |
Collapse
|
181
|
Whitfield JF, Morley P, Willick GE. Bone growth stimulators. New tools for treating bone loss and mending fractures. VITAMINS AND HORMONES 2003; 65:1-80. [PMID: 12481542 DOI: 10.1016/s0083-6729(02)65059-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In the new millennium, humans will be traveling to Mars and eventually beyond with skeletons that respond to microgravity by self-destructing. Meanwhile in Earth's aging populations growing numbers of men and many more women are suffering from crippling bone loss. During the first decade after menopause all women suffer an accelerating loss of bone, which in some of them is severe enough to result in "spontaneous" crushing of vertebrae and fracturing of hips by ordinary body movements. This is osteoporosis, which all too often requires prolonged and expensive care, the physical and mental stress of which may even kill the patient. Osteoporosis in postmenopausal women is caused by the loss of estrogen. The slower development of osteoporosis in aging men is also due at least in part to a loss of the estrogen made in ever smaller amounts in bone cells from the declining level of circulating testosterone and is needed for bone maintenance as it is in women. The loss of estrogen increases the generation, longevity, and activity of bone-resorbing osteoclasts. The destructive osteoclast surge can be blocked by estrogens and selective estrogen receptor modulators (SERMs) as well as antiosteoclast agents such as bisphosphonates and calcitonin. But these agents stimulate only a limited amount of bone growth as the unaffected osteoblasts fill in the holes that were dug by the now suppressed osteoclasts. They do not stimulate osteoblasts to make bone--they are antiresorptives not bone anabolic agents. (However, certain estrogen analogs and bisphosphates may stimulate bone growth to some extent by lengthening osteoblast working lives.) To grow new bone and restore bone strength lost in space and on Earth we must know what controls bone growth and destruction. Here we discuss the newest bone controllers and how they might operate. These include leptin from adipocytes and osteoblasts and the statins that are widely used to reduce blood cholesterol and cardiovascular damage. But the main focus of this article is necessarily the currently most promising of the anabolic agents, the potent parathyroid hormone (PTH) and certain of its 31- to 38-aminoacid fragments, which are either in or about to be in clinical trial or in the case of Lilly's Forteo [hPTH-(1-34)] tentatively approved by the Food and Drug Administration for treating osteoporosis and mending fractures.
Collapse
Affiliation(s)
- James F Whitfield
- Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario, Canada K1A 0R6
| | | | | |
Collapse
|
182
|
Staal A, Frith JC, French MH, Swartz J, Güngör T, Harrity TW, Tamasi J, Rogers MJ, Feyen JHM. The ability of statins to inhibit bone resorption is directly related to their inhibitory effect on HMG-CoA reductase activity. J Bone Miner Res 2003; 18:88-96. [PMID: 12510809 DOI: 10.1359/jbmr.2003.18.1.88] [Citation(s) in RCA: 128] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Statins, which are inhibitors of 3-hydroxy-3-glutaryl-coenzyme A (HMG-CoA) reductase, decrease the hepatic biosynthesis of cholesterol by blocking the mevalonate pathway. Nitrogen-containing bisphosphonate drugs also inhibit the mevalonate pathway, preventing the production of the isoprenoids, which consequently results in the inhibition of osteoclast formation and osteoclast function. Therefore, we hypothesized that statins could affect bone metabolism in vivo through effects on osteoclastic bone resorption. In vitro, cerivastatin inhibited the parathyroid hormone (PTH)-stimulated bone resorption. Using a panel of 40 statin analogs, which showed variable effects on HMG-CoA reductase activity, we found that the ability of compounds to inhibit bone resorption is directly related to HMG-CoA reductase activity. However, in the thyro-parathyrodectomy (TPTX) model for bone resorption in the rat in vivo, cerivastatin did not prevent experimentally induced increases in bone resorption. The lack of effect of cerivastatin in this model is not related to a limited penetration of the target tissue (bone marrow), because a significant effect on HMG-CoA reductase activity was demonstrated in the total rat bone marrow cell extracts of rats posttreatment in vivo. Furthermore, cerivastatin inhibited protein prenylation in osteoclasts isolated from the rabbit bone marrow of rabbits after treatment in vivo. In contrast to other studies, none of the statins tested showed anabolic effects in parietal bone explant cultures. Taken together, we conclude that statins inhibit bone resorption in vitro, which correlates directly with the potency of the compounds for inhibition of HMG-CoA reductase activity. However, cerivastatin does not affect bone resorption in the rat TPTX model in vivo.
Collapse
Affiliation(s)
- Ada Staal
- Osteoporosis Research, Metabolic and Cardiovascular Drug Discovery, Pharmaceutical Research Institute, Bristol-Myers Squibb Company, Pennington, New Jersey 08534, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
183
|
Abstract
Recently, several articles have been published dealing with the anabolic effects on bone by statins. Mundy and associates discovered that several statins were able to activate the promotor of bone morphogenetic protein (BMP) 2. Additionally, oral simvastatin and lovastatin increased the cancellous bone volume in rats, presumably an effect of the increase of BMP-2. Other studies have followed, with conflicting results; some have found a positive bone metabolic effect of statins and others have not. Studies published so far have focused on osteoporosis. In this study, femur fractures were produced in 81 mature male BALB/c mice and stabilized with marrow-nailing. Forty-one mice were given a diet prepared with simvastatin, so that each mouse received an approximate dose of 120 mg/kg of body weight per day. The remaining mice received the same diet with the exception of the simvastatin. Bilateral femurs were harvested at 8, 14, and 21 days postoperatively (po), the marrow-nail was extracted, and diameters were measured. Biomechanical tests were performed on 42 mice, by way of three-point bending. Histological specimens were prepared using standard techniques. For statistical analysis, ANOVA with Scheffés post hoc test was used. At 8 days, the fracture callus was too soft for meaningful biomechanical testing. At 14 days, the callus of the simvastatin-treated mice had a 53% larger transverse area than controls (p = 0.001), the force required to break the bone was 63% greater (p = 0.001), and the energy uptake was increased by 150% (p = 0.0008). Stiffness and modulus of elasticity were not significantly affected. At 21 days, the fractures were histologically healed and the mechanical differences had disappeared. The contralateral unbroken bone showed a slight increase in transverse area because of the simvastatin treatment, but there was no significant effect on the force required to break the bone or on energy uptake. These results point to a new possibility in the treatment of fractures.
Collapse
Affiliation(s)
- Bjorn Skoglund
- Department of Neuroscience and Locomotion, Faculty of Health Sciences, Linkoping, Sweden
| | | | | |
Collapse
|
184
|
Higuchi C, Myoui A, Hashimoto N, Kuriyama K, Yoshioka K, Yoshikawa H, Itoh K. Continuous inhibition of MAPK signaling promotes the early osteoblastic differentiation and mineralization of the extracellular matrix. J Bone Miner Res 2002; 17:1785-94. [PMID: 12369782 DOI: 10.1359/jbmr.2002.17.10.1785] [Citation(s) in RCA: 147] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We screened the small molecule compounds that stimulate osteogenesis by themselves or promote bone morphogenetic protein (BMP)-induced bone formation. We found that a specific inhibitor for MAPK/extracellular signal-regulated kinase kinase (MEK)-1, promoted the early osteoblastic differentiation and mineralization of extracellular matrix (ECM) in C2Cl2 pluripotent mesenchymal cells treated with recombinant human BMP-2 (rhBMP-2) and MC3T3-E1 preosteoblastic cells. ALP activity was synergistically increased by the treatment with a specific MEK-1 inhibitor PD98059 and rhBMP-2 in both cell lines. Twenty-five micromolar PD98059 promoted mineralization of ECM in rhBMP-2-treated C2Cl2 cells and MC3T3-E1 cells. In contrast, PD98059 reduced osteocalcin (OCN) secretion and its transcriptional level in rhBMP-2-treated C2Cl2 cells but increased its secretion and mRNA level in MC3T3-E1 cells. Stable expression of a dominant-negative MEK-1 mutant in C2Cl2 cells represented high ALP activity and low osteocalcin production in the presence of rhBMP-2, while a constitutively active mutant of MEK-1 attenuated both of them. Together, our results indicated that BMP-2-induced mineralization of ECM of pluripotent mesenchymal stem cells and preosteoblastic cells could be controlled by a fine tuning of the MAPK signaling pathway. Further, MEK-1 inhibitors would be useful for the promotion of bone formation, for instance, the treatments for delayed fracture healing or advance of localized osteoporotic change after fracture healing.
Collapse
Affiliation(s)
- Chikahisa Higuchi
- Department of Biology, Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | | | | | | | | | | | | |
Collapse
|
185
|
Thylin MR, McConnell JC, Schmid MJ, Reckling RR, Ojha J, Bhattacharyya I, Marx DB, Reinhardt RA. Effects of simvastatin gels on murine calvarial bone. J Periodontol 2002; 73:1141-8. [PMID: 12416771 DOI: 10.1902/jop.2002.73.10.1141] [Citation(s) in RCA: 113] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND The cholesterol-lowering drug simvastatin has been shown to stimulate murine calvarial bone growth after multiple injections. The purpose of this study was to test if similar bone stimulation could be induced by 2 single-dose drug delivery systems appropriate to periodontal therapy. METHODS ICR Swiss mice were treated with the following protocols: 1) injection of methylcellulose gel alone, subcutaneously over the calvarium (INJ-GEL; n = 8); 2) injection of gel with simvastatin (INJ-SIM; 2.2 mg, n = 16); 3) polylactide membrane (PLA) containing gel alone implanted over calvarium (MEM-GEL; n = 10); 4) implanted PLA membrane containing gel and simvastatin (MEM-SIM; n = 10); and 5) untreated mice (n = 12). Animals were sacrificed after 22 or 44 days, calvaria decalcified and stained with hematoxylin and eosin, and images digitized and measured for bone thickness and area. Data were compared using analysis of variance. RESULTS INJ-SIM stimulated a 53% (P = 0.02) increase at the thickest point of calvarial bone, while MEM-SIM caused a highly significant (P < or = 0.0005) increase in bone thickness (159% to 172%) and bone area (144% to 180%) compared to gel controls. Simvastatin gels caused soft tissue inflammation, which appeared to be related to bone increases. If INJ-SIM animals showing leakage of gel and/or no inflammation were excluded from analysis, INJ-SIM resulted in more bone (58% to 83%) than gel controls. An insignificant amount of SIM-stimulated bone was lost over the long term (44 days). CONCLUSIONS A single, high dose of simvastatin gel can stimulate murine cranial bone apposition, particularly when delivered under an occlusive membrane. Both approaches should be investigated further for possible development for periodontal therapy.
Collapse
Affiliation(s)
- Michael R Thylin
- Department of Surgical Specialties, University of Nebraska Medical Center, College of Dentistry, Lincoln 68583-0757, USA
| | | | | | | | | | | | | | | |
Collapse
|
186
|
Abstract
Pruritus, fatigue and metabolic bone disease represent three major extrahepatic manifestations of chronic cholestatic liver disease that considerably affect the patient's quality of life. The present article reviews pathogenetic aspects of and current therapeutic approaches to extrahepatic manifestations of cholestatic liver disease. Pathogenesis of pruritus of cholestasis remains poorly understood. The involvement of putative peripherally acting pruritogens, such as bile acids or endogenous opioids, is being discussed. More recently, central mechanisms, including an increased central opioidergic tone and pertubations in the serotonergic system have been proposed. Treatment of the underlying disease is beneficial also for the control of cholestasis-associated pruritus. Current therapeutic recommendations include ursodeoxycholic acid, cholestyramine, rifampicin and opioid antagonists. Liver transplantation may be indicated when severe pruritus is refractory to medical treatment. Fatigue is being recognized as the most frequent and one of the most disabling complaints in chronic cholestasis. Fatigue is presumably of central origin and its association with other neuropsychiatric disorders (e.g. depression, obsessive-compulsive disorders) is consistent with defective central neurotransmission. No specific therapies are currently available and a healthy lifestyle, regular sleep and avoidance of unnecessary stress and other precipiting factors are recommended. Antidepressant therapy may be warranted in selected patients. Osteopenia and osteoporosis are common in chronic cholestatic liver disease, whereas osteomalacia is rare. The pathophysiology of cholestasis-associated metabolic bone disease is regarded as multifactorial. Therapeutic recommendations include regular exercise, calcium and vitamin D supplementation in late stage disease, hormone replacement therapy in postmenopausal women and bisphosphonates.
Collapse
Affiliation(s)
- Helena Glasova
- Department of Medicine II, Klinikum of the University of Munich-Grosshadern, Munich, Germany
| | | |
Collapse
|
187
|
McLaughlin F, Mackintosh J, Hayes BP, McLaren A, Uings IJ, Salmon P, Humphreys J, Meldrum E, Farrow SN. Glucocorticoid-induced osteopenia in the mouse as assessed by histomorphometry, microcomputed tomography, and biochemical markers. Bone 2002; 30:924-30. [PMID: 12052464 DOI: 10.1016/s8756-3282(02)00737-8] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Glucocorticoids are potent anti-inflammatory molecules used in the treatment of asthma, rheumatoid arthritis, inflammatory bowel disease, and other inflammatory and dermatological diseases, as well as in posttransplantation immunotherapy. Although glucocorticoids have been prescribed for many years, their potential side effects, when administered orally, can prevent their long-term use. The most serious side effect observed in the clinic is glucocorticoid-induced osteoporosis (GIOP). To develop a small animal model to characterize glucocorticoid-induced bone loss, we carried out a series of experiments using BALB/c mice given daily intraperitoneal doses of the synthetic glucocorticoid, dexamethasone. Following dexamethasone treatment, the mice became osteopenic, with highly significant decreases in bone formation rate and mineral apposition rate, as assessed by standard histomorphometry. Moreover, 3 week treatment with dexamethasone resulted in a decrease in trabecular thickness and trabecular number with an increase in surface-to-volume ratio of trabeculae in the distal femur, as measured using microcomputed tomography (micro-CT). The serum bone formation marker, osteocalcin, was dose-dependently decreased in all mice treated with dexamethasone and showed a parallel extent of regulation to the bone formation rate changes. In addition, serum levels of leptin, recently identified as playing a role in the regulation of bone mass, increased following dexamethasone treatment. BALB/c mice therefore represent a useful model system in which the detrimental effects of glucocorticoids on bone can be studied.
Collapse
Affiliation(s)
- F McLaughlin
- Department of Asthma Cell Biology, GlaxoSmithKline, Stevenage, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
188
|
Hsia J, Morse M, Levin V. Effect of simvastatin on bone markers in osteopenic women: a placebo-controlled, dose-ranging trial [ISRCTN85429598]. BMC Musculoskelet Disord 2002; 3:7. [PMID: 11879528 PMCID: PMC79000 DOI: 10.1186/1471-2474-3-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2001] [Accepted: 02/15/2002] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hydroxymethylglutaryl coenzyme A reductase inhibitors increase new bone formation in vitro and in rodents. Results of epidemiologic analyses evaluating the association between use of these cholesterol-lowering drugs, bone mineral density and fracture have been mixed. METHODS Women (n = 24) with osteopenia, assessed by broad band ultrasound attenuation, were randomized to simvastatin 20 mg, 40 mg or identical-appearing placebo for 12 weeks. Fasting lipid profiles and biochemical markers of bone formation (bone-specific alkaline phosphatase) and resorption (N-telopeptides and C-terminal propeptide of type 1 collagen) were measured at baseline, 6 and 12 weeks. RESULTS Plasma low density lipoprotein-cholesterol concentration fell 7%, 39% (p < 0.01 vs baseline) and 47% (p < 0.01 vs baseline) after 12 weeks of treatment with placebo, simvastatin 20 mg and 40 mg, respectively. At baseline, bone marker concentrations were similar in the three treatment groups. At 6 and 12 weeks, bone marker concentrations were not different from baseline, and no significant differences in bone marker concentrations were observed between treatment groups at either 6 or 12 weeks. CONCLUSION Among osteopenic women, treatment with simvastatin for 12 weeks did not affect markers of bone formation or resorption.
Collapse
Affiliation(s)
- Judith Hsia
- Department of Medicine, George Washington University, Washington, DC, USA
| | - Megan Morse
- Department of Medicine, George Washington University, Washington, DC, USA
| | - Virginia Levin
- Department of Medicine, George Washington University, Washington, DC, USA
| |
Collapse
|
189
|
|
190
|
Reinholz GG, Getz B, Sanders ES, Karpeisky MY, Padyukova NS, Mikhailov SN, Ingle JN, Spelsberg TC. Distinct mechanisms of bisphosphonate action between osteoblasts and breast cancer cells: identity of a potent new bisphosphonate analogue. Breast Cancer Res Treat 2002; 71:257-68. [PMID: 12002344 DOI: 10.1023/a:1014418017382] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
While the effects of bisphosphonates on bone-resorbing osteoclasts have been well documented, the effects of bisphosphonates on other cell types are not as well studied. Recently, we reported that bisphosphonates have direct effects on bone-forming human fetal osteoblast cells (hFOB). In this report, the role of the mevalonate pathway in the actions of bisphosphonates on hFOB, and MDA-MB-231 human breast cancer cells was examined. These studies included a novel bisphosphonate analog, the anhydride formed between arabinocytidine 5' phosphate and etidronate (Ara-CBP). Ara-CBP was the most potent inhibitor of hFOB and MDA-MB-231 cell proliferation, and stimulator of hFOB cell mineralization compared to etidronate, the anhydride formed between AMP and etidronate (ABP), pamidronate, and zoledronate. Inhibition of hFOB cell proliferation by Ara-CBP and zoledronate was partially reversed by mevalonate pathway intermediates, and stimulation of hFOB cell mineralization was completely reversed by mevalonate pathway intermediates. These results suggest that zoledronate and Ara-CBP act, at least in part, via inhibition of the mevalonate pathway in hFOB cells. In contrast, none of the mevalonate pathway intermediates reversed the inhibition of MDA-MB-231 cell proliferation by the bisphosphonates, or the effects of pamidronate on hFOB cells. As a positive control, the effects of mevastatin on hFOB and MDA-MB-231 cells were completely reversed by mevalonate. In summary, these data suggest that zoledronate and Ara-CBP induce human osteoblast differentiation via inhibition of the mevalonate pathway. In contrast, the inhibition of MDA-MB-231 cell proliferation by the bisphosphonates appears to be through mechanisms other than inhibition of the mevalonate pathway.
Collapse
Affiliation(s)
- Gregory G Reinholz
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | |
Collapse
|
191
|
Garrett IR, Mundy GR. The role of statins as potential targets for bone formation. ARTHRITIS RESEARCH 2002; 4:237-40. [PMID: 12106493 PMCID: PMC128929 DOI: 10.1186/ar413] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2001] [Accepted: 01/10/2002] [Indexed: 02/25/2023]
Abstract
Inhibitors of the 3-hydroxy-3-methylglutaryl coenzyme A reductase enzyme have recently been shown to stimulate bone formation in rodents both in vitro and in vivo. In bone cells, these inhibitors increase the gene expression of bone morphogenetic protein-2, which is an autocrine-paracrine factor for osteoblast differentiation. The findings that statins increase bone formation and bone mass in rodents suggest a potential new action for these compounds, which may be beneficial in patients with established osteoporosis where marked bone loss has occurred. Recent clinical data suggest that they may reduce the risk of fracture in patients taking these drugs.
Collapse
|
192
|
Edwards CJ, Spector TD. Statins as modulators of bone formation. ARTHRITIS RESEARCH 2002; 4:151-3. [PMID: 12010563 PMCID: PMC128924 DOI: 10.1186/ar399] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/04/2001] [Revised: 11/20/2001] [Accepted: 11/22/2001] [Indexed: 01/03/2023]
Abstract
The use of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) to reduce serum cholesterol is well described. However, the recent finding that statins have direct effects on bone was unexpected. A number of epidemiological studies have recently been published that explore the effects of statins on bone mineral density and risk of fracture in humans. Statins may act by directly stimulating the expression of bone morphogenetic protein-2 and increasing osteoblast differentiation or, like nitrogen-containing bisphosphonates, may have effects on the mevalonate pathway that leads to inhibition of osteoclast activity and osteoblast apoptosis. In addition, the demonstration that statins can inhibit inflammation and encourage angiogenesis offers other possibilities for action.
Collapse
|
193
|
Watkins BA, Li Y, Seifert MF. Nutraceutical fatty acids as biochemical and molecular modulators of skeletal biology. J Am Coll Nutr 2001; 20:410S-416S; discussion 417S-420S. [PMID: 11603651 DOI: 10.1080/07315724.2001.10719177] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Several systemic hormones and localized growth factors coordinate events of bone formation and resorption to support bone growth in the young and maintain bone mineral content in the adult. Some of the more important factors produced in the bone microenvironment that impact skeletal biology include prostaglandins, cytokines, and insulin-like growth factors. Dietary fat sources that exert potent biological effects on the skeletal tissues belong to the omega-6 and omega-3 families of essential fatty acids. Specific long-chain polyunsaturated fatty acids (PUFA) belonging to these families are substrates for prostanoids that influence the differentiation and activity of cells in bone and cartilage tissues. These PUFA appear to alter prostanoid formation, cell-to-cell signaling processes, and impact transcription factors in vivo. Hence, these biologically active PUFA can be called nutraceutical fatty acids. This review highlights the role of nutraceutical fatty acids on bone metabolism and joint disease. The recent discovery of transcription factors controlling osteoblast function, and soluble proteins directing osteoclastogenesis and osteoblastogenesis offer new research opportunities for studying nutraceutical fatty acids in skeletal biology.
Collapse
Affiliation(s)
- B A Watkins
- Department of Food Science, Purdue University, West Lafayette, Indiana 47907, USA.
| | | | | |
Collapse
|
194
|
Maritz FJ, Conradie MM, Hulley PA, Gopal R, Hough S. Effect of statins on bone mineral density and bone histomorphometry in rodents. Arterioscler Thromb Vasc Biol 2001; 21:1636-41. [PMID: 11597938 DOI: 10.1161/hq1001.097781] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Statins have been postulated to affect bone metabolism. We investigated the effects of different doses of simvastatin (1, 5, 10, and 20 mg. kg(-1). d(-1)), atorvastatin (2.5 mg. kg(-1). d(-1)), and pravastatin (10 mg. kg(-1). d(-1)) administered orally for 12 weeks to intact female Sprague-Dawley rats and the effect of 20 mg. kg(-1). d(-1) simvastatin in sham-operated and ovariectomized rats on femoral bone mineral density (BMD) and quantitative bone histomorphometry (QBH) and compared them with controls. BMD was decreased by 1 mg. kg(-1). d(-1) simvastatin (P=0.042), atorvastatin (P=0.0002), and pravastatin (P=0.002). The effect on QBH parameters differed with different doses of simvastatin (ANOVA, P=0.00012). QBH parameters of both bone formation and resorption were equivalently and markedly increased by 20 mg. kg(-1). d(-1) simvastatin in 2 separate groups of intact rats and were reflected by a relatively unchanged BMD. At lower doses, 1 mg. kg(-1). d(-1) simvastatin decreased bone formation while increasing bone resorption, as reflected by a marked decrease in BMD. Ovariectomized animals receiving 20 mg. kg(-1). d(-1) simvastatin showed no change in BMD relative to the untreated, ovariectomized controls; their increase in bone formation was smaller than in sham-operated rats receiving simvastatin, and there was no change in bone resorption. Dose-response curves of simvastatin for bone formation and resorption differed. These studies indicate that (1) statins decrease BMD in rodents, (2) high-dose simvastatin increases bone formation and resorption, (3) low-dose simvastatin decreases bone formation and increases bone resorption, (4) the effects of simvastatin on QBH differ at different dosages, (5) the effects of simvastatin seen in intact rats are not observed in ovariectomized rats, and (6) simvastatin is unable to prevent bone loss caused by ovariectomy.
Collapse
Affiliation(s)
- F J Maritz
- Endocrinology and Metabolism Unit, Department of Internal Medicine, University of Stellenbosch and Tygerberg Hospital, Tygerberg, South Africa.
| | | | | | | | | |
Collapse
|
195
|
Ohnaka K, Shimoda S, Nawata H, Shimokawa H, Kaibuchi K, Iwamoto Y, Takayanagi R. Pitavastatin enhanced BMP-2 and osteocalcin expression by inhibition of Rho-associated kinase in human osteoblasts. Biochem Biophys Res Commun 2001; 287:337-42. [PMID: 11554731 DOI: 10.1006/bbrc.2001.5597] [Citation(s) in RCA: 152] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To clarify the mechanism of the stimulatory effect of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors (statins) on bone formation, we investigated the effect of pitavastatin, a newly developed statin, on expression of bone morphogenetic protein-2 (BMP-2) and osteocalcin in primary cultured human osteoblasts. Pitavastatin increased the expression level of mRNA for BMP-2, and much more effectively for osteocalcin. This stimulatory effect was abolished by the addition of geranylgeranyl pyrophosphate, an essential molecule for prenylation of small GTP-binding proteins such as Rho GTPase, but not by inhibitors of nitric oxide synthase and various protein kinases. Pitavastatin suppressed the Rho-associated kinase (Rho-kinase) activity. Hydroxyfasudil, a specific inhibitor of Rho-kinase, increased BMP-2 and osteocalcin expression. These mRNA levels were strongly suppressed by dexamethasone, but restored by co-treatment with hydroxyfasudil. These observations suggest that the Rho-kinase negatively regulates bone formation and the inhibition of Rho and Rho-kinase pathway is the major mechanism of the statin effect on bone. Moreover, a Rho-kinase inhibitor may be a new therapeutic reagent for the treatment of osteoporosis such as glucocorticoid-induced osteoporosis.
Collapse
Affiliation(s)
- K Ohnaka
- Department of Geriatric Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | | | | | | | | | | | | |
Collapse
|
196
|
Phillips BW, Belmonte N, Vernochet C, Ailhaud G, Dani C. Compactin enhances osteogenesis in murine embryonic stem cells. Biochem Biophys Res Commun 2001; 284:478-84. [PMID: 11394905 DOI: 10.1006/bbrc.2001.4987] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Embryonic stem (ES) cells have the capacity to differentiate into various cell types in vitro. In this study, we show that retinoic acid is important for the commitment of ES cells into osteoblasts. Culturing retinoic acid treated ES cells in the presence of the osteogenic supplements ascorbic acid and beta-glycerophosphate resulted in the expression of several osteoblast marker genes, osteocalcin, alkaline phosphatase, and osteopontin. However, there was only a slight amount of mineralized matrix secretion. Addition of bone morphogenic protein-2 or compactin, a drug of the statin family of HMG-CoA reductase inhibitors, resulted in a greatly enhanced formation of bone nodules. Compactin did not modify the expression of osteogenic markers, but at the late stage of differentiation promoted an increase in BMP-2 expression. These results establish ES-cell derived osteogenesis as an effective model system to study the molecular mechanisms by which the statin compactin promotes osteoblastic differentiation and bone nodule formation.
Collapse
Affiliation(s)
- B W Phillips
- Centre de Biochimie (UMR 6543 CNRS), Faculté de Sciences, Université de Nice-Sophia Antipolis, Parc Valrose, Nice, 06108, France
| | | | | | | | | |
Collapse
|
197
|
Mostaza JM, De la Piedra C, Curiel MD, Peña R, Lahoz C. Pravastatin therapy increases procollagen I N-terminal propeptide (PINP), a marker of bone formation in post-menopausal women. Clin Chim Acta 2001; 308:133-7. [PMID: 11412825 DOI: 10.1016/s0009-8981(01)00476-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The aim of our study was to evaluate whether pravastatin treatment affected biochemical markers of bone turnover. METHODS Thirty-six hypercholesterolemic post-menopausal women, not on hormonal replacement therapy, were selected from a population study evaluating factors affecting cholesterol response to pravastatin. After a 6-week period on a 30% fat diet, participants received treatment with 20 mg/day of pravastatin during a 16-week follow-up period. Pre- and post-treatment samples were analyzed for procollagen I aminoterminal peptide (PINP) and bone alkaline phosphatase (bAP) as markers of bone formation, carboxyterminal telopeptide of collagen I (CTX) as a marker of bone resorption, and procollagen III aminoterminal propeptide (PIIINP) as a marker of fibrogenesis. RESULTS Total cholesterol decreased from 7.26+/-0.83 to 6.1+/-0.77 mmol/l with pravastatin treatment. PINP levels significantly increased (from 33.6+/-13 to 37.4+/-16, p=0.03) without changes in bAP or CTX. Individual changes in PINP correlated with individual reduction in cholesterol levels (r=0.337, p=0.04). There was no significant change in PIIINP concentration. CONCLUSIONS Pravastatin treatment increased PINP levels, a marker of bone formation, in hypercholesterolemic, post-menopausal women, without affecting bone resorption. PIIINP concentration, a marker of liver fibrogenesis, was not affected by the treatment.
Collapse
Affiliation(s)
- J M Mostaza
- Atherosclerosis Unit, Hospital Carlos III, Sinesio Delgado 10, 28029 Madrid, Spain.
| | | | | | | | | |
Collapse
|
198
|
Watanabe S, Fukumoto S, Takeuchi Y, Fujita H, Nakano T, Fujita T. Effects of 1-year treatment with fluvastatin or pravastatin on bone. Am J Med 2001; 110:584-7. [PMID: 11343673 DOI: 10.1016/s0002-9343(01)00679-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- S Watanabe
- Department of Medicine and Laboratory Medicine, University of Tokyo Branch Hospital, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
199
|
Abstract
Statins are widely used lipid-lowering drugs that reduce cholesterol synthesis by inhibiting 3-hydroxy-3-glutaryl-coenzyme A (HMG-CoA) reductase activity. They also strongly stimulate bone formation in rodents. If the drugs' potent bone-building activity results directly from inhibition of HMG-CoA reductase, there should be less bone fracturing in humans who have taken statins to lower their serum cholesterol and prevent heart attacks, but the data gleaned from several databases are contradictory. According to some reports the lipid-lowering doses of oral statins increased bone mineral density and more than halved the risk of fracturing various bones, while according to others, including the very large Women's Health Initiative Observational Study (WHI-OS), the drugs did not significantly affect the fracturing risk. Such contradictory data could be due in part to one of the commonly used statins, pravastatin which only targets hepatocytes, or due to bone growth being stimulated by something other than inhibition of HMG-CoA reductase. Therefore, different doses of statins may be needed to build bone or optimally lower serum cholesterol. To be able to answer the question posed by the title of this editorial, it will be necessary to carry out a controlled trial using designer statins that are less liver-oriented and thus better for assessing the optimal doses needed, specifically for osteogenicity rather than for their cholesterol-lowering ability.
Collapse
|
200
|
Abstract
Bone remodelling processes are regulated by systemic hormones and a multitude of local and systemic factors, including prostaglandins, cytokines, and growth factors. Dietary fatty acids and their derivatives (eicosanoids) have been a recent focus of investigation on bone and cartilage metabolism. Specific fatty acids are recognized modulators of eicosanoid biosynthesis, signal transduction, and gene expression. The actions of polyunsaturated fatty acids have not been extensively examined in the skeletal system. Promising research on fatty acids and bone remodelling should evaluate the potential effects on pathways for osteoclastogenesis and osteoblastogenesis.
Collapse
Affiliation(s)
- B A Watkins
- Department of Food Science, Lipid Chemistry and Molecular Biology Laboratory, Purdue University, West Lafayette, Indiana 47907, USA.
| | | | | |
Collapse
|