551
|
Behl Y, Siqueira M, Ortiz J, Li J, Desta T, Faibish D, Graves DT. Activation of the acquired immune response reduces coupled bone formation in response to a periodontal pathogen. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:8711-8. [PMID: 19050291 PMCID: PMC2638096 DOI: 10.4049/jimmunol.181.12.8711] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Osteoimmunolgy involves the interaction of the immune system with skeletal elements. This interaction can lead to the formation of osseous lesions. To investigate how the acquired immune response could contribute to osteolytic lesions, we injected the periodontal pathogen Porphyromonas gingivalis adjacent to calvarial bone with or without prior immunization against the bacterium. Activation of the acquired immune response increased osteoclastogenesis and decreased coupled bone formation. The latter was accompanied by an increase in nuclear translocation of the transcription factor FOXO1 in vivo, increased apoptosis of bone-lining cells measured by the TUNEL assay and number of activated caspase-3 positive cells and a decrease in bone lining cell density. Further studies were conducted with MC3T3 osteoblastic cells. Apoptosis and increased FOXO1 DNA binding activity were induced when a combination of cytokines was tested, IL-beta, TNF-alpha, and IFN-gamma. Knockdown of FOXO1 by small interfering RNA significantly reduced cytokine stimulated apoptosis, cleaved caspase-3/7 activity and decreased mRNA levels of the proapoptotic genes, TNF-alpha, FADD, and caspase-3, -8, and -9. These results indicate that activation of the acquired immunity by a periodontal pathogen reduces the coupling of bone formation and resorption. This may occur by enhancing bone lining cell apoptosis through a mechanism that involves increased FOXO1 activation. These studies give insight into inflammatory bone diseases such as periodontal disease and arthritis were the formation of lytic lesions occurs in conjunction with deficient bone formation and activation of an acquired immune response.
Collapse
Affiliation(s)
- Yugal Behl
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston, MA 02118, USA
| | | | | | | | | | | | | |
Collapse
|
552
|
Mansell JP. Bone phenotypes in response to gonadotropin misexpression: the role for gonadotropins in postmenopausal osteoporosis. Int J Gen Med 2008; 1:51-7. [PMID: 20428406 PMCID: PMC2840536 DOI: 10.2147/ijgm.s3879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Scant attention has been paid to the potential role of gonadotropins in bone tissue homeostasis. The focus on estrogen and estrogen replacement therapy for osteoporosis as far back as the 1940's may account for the paucity of gonadotropin studies in bone biology. It is conceivable that prevailing dogma may have subconsciously steered us away from addressing whether gonadotropins have a place in skeletal physiology. However an examination of bone tissue catabolism in ovariectomized (OVX) and luteinizing hormone-releasing hormone (LHRH) agonist (Zoladex((R)))-treated rats generated some interesting and conflicting data; Zoladex-treated rats, unlike the OVX group, failed to exhibit increased bone collagen catabolism despite clear evidence for estrogen deficiency. The findings, although controversial, supported the possibility that elevated gonadotropins in the OVX model were in some way accountable for increased bone catabolism. In response to these initial findings further studies were performed to determine if altered LH status may in some way impact on the skeleton To this end an investigation of bone mass and histomorphometry were conducted in LH receptor nullizygous mice and human chorionic gonadotropin (hCG) overexpressing mice. There were clear phenotypic differences; the LH receptor knockout mice displayed reduced bone mass whereas the hCG overexpressing animals had stark increases in bone mass. Much more recently the team of the Mount Sinai Bone Program have made a significant discovery that bone-resorbing osteoclasts express receptors for follicle-stimulating hormone (FSH) and that mice nullizygous for FSH receptor are resistant to bone loss despite severe estrogen deficiency. Details of these fascinating models will be presented together with additional findings that give credence for exploring gonadotropin action on the skeleton as we enter the twilight of this Decade of the Bone and Joint.
Collapse
Affiliation(s)
- Jason P Mansell
- Department of Oral and Dental Sciences, Division of Oral Medicine, University of Bristol Dental School, Lower Maudlin Street, Bristol, BS1 2LY, UK
| |
Collapse
|
553
|
Herrmann M, Umanskaya N, Wildemann B, Colaianni G, Widmann T, Zallone A, Herrmann W. Stimulation of osteoblast activity by homocysteine. J Cell Mol Med 2008; 12:1205-10. [PMID: 18782184 PMCID: PMC3865664 DOI: 10.1111/j.1582-4934.2008.00104.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Homocysteine (HCY) has recently been linked to fragility fractures. Moreover, HCY activates osteoclasts. Little is known about the effect of HCY on activity of human osteoblasts (OBs). We hypothesized that HCY decreases the activity of OBs. Osteoblasts obtained from tra-becular human bone specimens of eight donors were cultured with conditioned medium. Culture medium was adjusted to 0, 100, 500, 1000 and 2000 μM HCY. After 14 days alkaline phosphatase (AP) activity, pro-collagen type I N-terminal peptide (PINP) and osteocalcin (OC) secretion in the supernatant were measured. After 20 days the formation of mineralized matrix was analyzed. HCY-stimulated AP activity gradually (100 μM HCY: 118%, P= 0.006; 500 μM HCY: 125%, P < 0.001). At 1000 and 2000 μM HCY the increase of AP activity was reversible (1000 μM HCY: 106%, P= 0.317; 2000 μM HCY: 102%, P < 0.737). The PINP secretion was also stimulated by HCY reaching a maximum of 260 ± 154 μg/l at 500 μmol/l versus 205 ± 94 μ,g/l in controls. After 20 days of culture the formation of bone matrix was increased at 100 and 500 μM HCY. OC secretion was not significantly changed. The results of the present study consistently demonstrate a moderate stimulation of primary human OB activity by increasing concentrations of HCY. However, the magnitude of this effect seems to be less pronounced than recent observations on primary human osteoclasts, suggesting a dysbalance between OBs and osteoclasts in favour of osteoclasts
Collapse
Affiliation(s)
- Markus Herrmann
- Department of Clinical Chemistry and Laboratory Medicine, University Hospital of Saarland, Homburg/Saar, Germany.
| | | | | | | | | | | | | |
Collapse
|
554
|
|
555
|
Ogita M, Rached MT, Dworakowski E, Bilezikian JP, Kousteni S. Differentiation and proliferation of periosteal osteoblast progenitors are differentially regulated by estrogens and intermittent parathyroid hormone administration. Endocrinology 2008; 149:5713-23. [PMID: 18617606 PMCID: PMC2584601 DOI: 10.1210/en.2008-0369] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The periosteum is now widely recognized as a homeostatic and therapeutic target for actions of sex steroids and intermittent PTH administration. The mechanisms by which estrogens suppress but PTH promotes periosteal expansion are not known. In this report, we show that intermittent PTH(1-34) promotes differentiation of periosteal osteoblast precursors as evidenced by the stimulation of the expression or activity of alkaline phosphatase as well as of targets of the bone morphogenetic protein 2 (BMP-2) and Wnt pathways. In contrast, 17beta-estradiol (E2) had no effect by itself. However, it attenuated PTH- or BMP-2-induced differentiation of primary periosteal osteoblast progenitors. Administration of intermittent PTH to ovariectomized mice induced rapid phosphorylation of the BMP-2 target Smad1/5/8 in the periosteum. A replacement dose of E2 had no effect by itself but suppressed PTH-induced phosphorylation of Smad1/5/8. In contrast to its effects to stimulate periosteal osteoblast differentiation, PTH promoted and subsequently suppressed proliferation of periosteal osteoblast progenitors in vitro and in vivo. E2 promoted proliferation and attenuated the antiproliferative effect of PTH. Both hormones protected periosteal osteoblasts from apoptosis induced by various proapoptotic agents. These observations suggest that the different effects of PTH and estrogens on the periosteum result from opposing actions on the recruitment of early periosteal osteoblast progenitors. Intermittent PTH promotes osteoblast differentiation from periosteum-derived mesenchymal progenitors through ERK-, BMP-, and Wnt-dependent signaling pathways. Estrogens promote proliferation of early osteoblast progenitors but inhibit their differentiation by osteogenic agents such as PTH or BMP-2.
Collapse
Affiliation(s)
- Mami Ogita
- Department of Medicine, Columbia University, New York, New York 10032, USA
| | | | | | | | | |
Collapse
|
556
|
Palmer LC, Newcomb CJ, Kaltz SR, Spoerke ED, Stupp SI. Biomimetic systems for hydroxyapatite mineralization inspired by bone and enamel. Chem Rev 2008; 108:4754-83. [PMID: 19006400 PMCID: PMC2593885 DOI: 10.1021/cr8004422] [Citation(s) in RCA: 670] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Liam C Palmer
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, USA
| | | | | | | | | |
Collapse
|
557
|
Plotkin LI, Lezcano V, Thostenson J, Weinstein RS, Manolagas SC, Bellido T. Connexin 43 is required for the anti-apoptotic effect of bisphosphonates on osteocytes and osteoblasts in vivo. J Bone Miner Res 2008; 23:1712-21. [PMID: 18597631 PMCID: PMC2648607 DOI: 10.1359/jbmr.080617] [Citation(s) in RCA: 162] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Revised: 06/04/2008] [Accepted: 06/27/2008] [Indexed: 02/01/2023]
Abstract
Connexin (Cx)43 is required for inhibition of osteocyte and osteoblast apoptosis by bisphosphonates in vitro. Herein, we evaluated its requirement for the in vivo actions of bisphosphonates using mice in which Cx43 was deleted specifically from osteocytes and osteoblasts (Cx43(DeltaOb-Ot/-) mice). Effective removal of Cx43 was confirmed by the presence of the deleted form of the gene and by reduced mRNA and protein expression in osteoblastic cells and bones obtained from Cx43(DeltaOb-Ot/-) mice. The amino-bisphosphonate alendronate (2.3 micromol/kg/d) was injected daily into 5-mo-old female mice (n = 6-11) for 31 days, starting 3 days before implantation of pellets releasing the glucocorticoid prednisolone (2.1 mg/kg/d). Cx43(DeltaOb-Ot/-) mice and their littermates (Cx43(fl/-), Cx43(DeltaOb-Ot/+), and Cx43(fl/+)) gained bone with similar kinetics and exhibited identical bone mass from 2 to 4.5 mo of age, indicating that Cx43 deletion from osteocytes and mature osteoblasts does not impair bone acquisition. In addition, prednisolone induced a similar increase in osteocyte and osteoblast apoptosis in Cx43(DeltaOb-Ot/-) or in control Cx43(fl/-) littermates. However, whereas alendronate prevented prednisolone-induced apoptosis in control Cx43(fl/-) mice, it was ineffective in Cx43(DeltaOb-Ot/-) mice. In contrast, alendronate inhibited glucocorticoid-induced bone loss in both type of animals, suggesting that inhibition of resorption is the predominant effect of alendronate against the early phase of glucocorticoid-induced bone loss. Taken together with earlier in vitro evidence, these findings show that Cx43 is required for the anti-apoptotic effect of bisphosphonates on osteocytes and osteoblasts.
Collapse
Affiliation(s)
- Lilian I Plotkin
- Division of Endocrinology and Metabolism, the Center for Osteoporosis and Metabolic Bone Diseases, the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA.
| | | | | | | | | | | |
Collapse
|
558
|
Mizobuchi H, García-Castellano JM, Philip S, Healey JH, Gorlick R. Hypoxia markers in human osteosarcoma: an exploratory study. Clin Orthop Relat Res 2008; 466:2052-9. [PMID: 18528739 PMCID: PMC2493019 DOI: 10.1007/s11999-008-0328-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2007] [Accepted: 05/16/2008] [Indexed: 01/31/2023]
Abstract
Neoplastic cells growing under hypoxic conditions exhibit a more aggressive phenotype by activating a cascade of molecular events partly mediated by hypoxia-inducible transcription factor (HIF-1alpha) and vascular endothelial growth factor (VEGF). The roles of these markers have been studied previously in several cancer lines. We ascertained the frequency of HIF-1alpha expression, VEGF expression, the degree of neovascularization, and cell proliferation in osteosarcoma samples. Samples from osteosarcoma patients were assessed for HIF-1alpha and VEGF protein expression using immunohistochemistry, neovascularization using antibodies for Factor VIII, and cell proliferation using the Ki-67 labeling index. Associations between these parameters and clinical features were examined. HIF-1alpha staining was positive in 35% of patients and metastases were present in 61% of these HIF-1alpha-positive patients. VEGF protein expression was detected in 69% of patients, 92% of whom were female. We observed an insignificant trend for a higher frequency of VEGF expression in the high-grade as compared to low-grade osteosarcoma. We observed no association between vascular density and proliferation index and any clinical parameters. We found an association between HIF-1alpha expression and metastatic disease and between VEGF expression and female gender.
Collapse
Affiliation(s)
- Hiroo Mizobuchi
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY USA
| | | | - Shaji Philip
- Division of Hematology/Oncology, Department of Pediatrics, The Children’s Hospital at Montefiore, 3415 Bainbridge Avenue, Rosenthal 3rd Floor, Bronx, NY 10467 USA
| | - John H. Healey
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY USA
| | - Richard Gorlick
- Division of Hematology/Oncology, Department of Pediatrics, The Children’s Hospital at Montefiore, 3415 Bainbridge Avenue, Rosenthal 3rd Floor, Bronx, NY 10467 USA
| |
Collapse
|
559
|
Abstract
Age-dependent bone loss has been well documented in both human and animal models. Although the underlying causal mechanisms are probably multifactorial, it has been hypothesized that alterations in progenitor cell number or function are important. Little is known regarding the properties of bone marrow stromal cells (BMSCs) or bone progenitor cells during the aging process, so the question of whether aging alters BMSC/progenitor osteogenic differentiation remains unanswered. In this study, we examined age-dependent changes in bone marrow progenitor cell number and differentiation potential between mature (3 and 6 mo old), middle-aged (12 and 18 mo old), and aged (24 mo old) C57BL/6 mice. BMSCs or progenitors were isolated from five age groups of C57BL/6 mice using negative immunodepletion and positive immunoselection approaches. The osteogenic differentiation potential of multipotent BMSCs was determined using standard osteogenic differentiation procedures. Our results show that both BMSC/progenitor number and differentiation potential increase between the ages of 3 and 18 mo and decrease rapidly thereafter with advancing age. These results are consistent with the changes of the mRNA levels of osteoblast lineage-associated genes. Our data suggest that the decline in BMSC number and osteogenic differentiation capacity are important factors contributing to age-related bone loss.
Collapse
|
560
|
Yingling VR, Taylor G. Delayed pubertal development by hypothalamic suppression causes an increase in periosteal modeling but a reduction in bone strength in growing female rats. Bone 2008; 42:1137-43. [PMID: 18406225 PMCID: PMC2494865 DOI: 10.1016/j.bone.2008.02.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Revised: 01/28/2008] [Accepted: 02/05/2008] [Indexed: 11/23/2022]
Abstract
The timing of the pubertal growth is a critical event in skeletal development. A delay in the onset of puberty has been correlated with increased stress fracture incidence in young women and as a result, suboptimal skeletal development may affect long-term bone strength. Gonadotropin releasing hormone antagonist (GnRH-a) injections were used to delay the onset of puberty in growing female rats. 23-day-old female rats were injected with a GnRH-antagonist at 2 dosage levels (n=15/group). The Low Dose group (1.25 mg/kg/dose) received daily injections for 27 days (sacrifice 49 days). The High Dose group received (5.0 mg/kg/dose) only 5 days per week over a 26 day period (sacrifice 48 days). Calcein injections measured bone formation activity on the periosteal and endocortical surfaces. Standard histomorphometric and biomechanical analyses were performed on the femora and ash content was measured on the tibiae of all animals. Serum estradiol and insulin-like growth factor (IGF)-1 levels were assayed. Significant delays in pubertal development occurred in the two GnRH-a groups as evidenced by delayed vaginal openings, decreased uterine and ovarian weights and suppressed estradiol levels compared to control. Femoral lengths were significantly shorter in the experimental groups and serum IGF-1 levels were higher than control. Bone strength and stiffness were significantly lower in the GnRH-a groups. Cortical bone area was decreased and total area was not different between groups. There was a significant decrease in % Ct.Ar/T.Ar. The decreased bone strength may have resulted from a decrease in the amount and distribution of bone, however, stress and Young's modulus were also decreased. There was a different response between endocortical formation indices and periosteal formation indices to the GnRH-a protocol. Endocortical bone formation rates decreased and there was an increase in periosteal labeled surface. A dose response between bone strength and GnRH-a dosage was found. The data suggest that hypothalamic suppression during pubertal development resulted in decreased bone strength which may result in fracture development.
Collapse
Affiliation(s)
- Vanessa R Yingling
- Department of Kinesiology, College of Health Professions, Temple University, 1800 North Broad Street, Philadelphia, PA 19122, USA.
| | | |
Collapse
|
561
|
Karasik D, Kiel DP. Genetics of the musculoskeletal system: a pleiotropic approach. J Bone Miner Res 2008; 23:788-802. [PMID: 18269309 PMCID: PMC3280426 DOI: 10.1359/jbmr.080218] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2007] [Revised: 01/28/2008] [Accepted: 02/07/2008] [Indexed: 12/25/2022]
Abstract
The risk of osteoporotic fracture can be viewed as a function of loading conditions and the ability of the bone to withstand the load. Skeletal loads are dominated by muscle action. Recently, it has become clear that bone and muscle share genetic determinants. Involution of the musculoskeletal system manifests as bone loss (osteoporosis) and muscle wasting (sarcopenia). Therefore, the consideration of pleiotropy is an important aspect in the study of the genetics of osteoporosis and sarcopenia. This Perspective will provide the evidence for a shared genetic influence on bone and muscle. We will start with an overview of accumulating evidence that physical exercise produces effects on the adult skeleton, seeking to unravel some of the contradictory findings published thus far. We will provide indications that there are pleiotropic relationships between bone structure/mass and muscle mass/function. Finally, we will offer some insights and practical recommendations as to the value of studying shared genetic factors and will explore possible directions for future research. We consider several related questions that together comprise the general paradigm of bone responses to mechanical loading and the relationship between muscle strength and bone parameters, including the genetic factors that modulate these responses. We believe that further progress in understanding the common genetic etiology of osteoporosis and sarcopenia will provide valuable insight into important biological underpinnings for both conditions and may translate into new approaches to reduce the burdens of both conditions through improved diagnosis, prevention, and early targeted treatment.
Collapse
Affiliation(s)
- David Karasik
- Institute for Aging Research, Hebrew SeniorLife, Harvard Medical School, Boston, Massachusetts 02131, USA.
| | | |
Collapse
|
562
|
In vivo genome-wide expression study on human circulating B cells suggests a novel ESR1 and MAPK3 network for postmenopausal osteoporosis. J Bone Miner Res 2008; 23:644-54. [PMID: 18433299 PMCID: PMC2674539 DOI: 10.1359/jbmr.080105] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Osteoporosis is characterized by low BMD. Studies have shown that B cells may participate in osteoclastogenesis through expression of osteoclast-related factors, such as RANKL, transforming growth factor beta (TGFB), and osteoprotegerin (OPG). However, the in vivo significance of B cells in human bone metabolism and osteoporosis is still largely unknown, particularly at the systematic gene expression level. MATERIALS AND METHODS In this study, Affymetrix HG-U133A GeneChip arrays were used to identify genes differentially expressed in B cells between 10 low and 10 high BMD postmenopausal women. Significance of differential expression was tested by t-test and adjusted for multiple testing with the Benjamini and Hochberg (BH) procedure (adjusted p </= 0.05). RESULTS Twenty-nine genes were downregulated in the low versus high BMD group. These genes were further analyzed using Ingenuity Pathways Analysis (Ingenuity Systems). A network involving estrogen receptor 1 (ESR1) and mitogen activated protein kinase 3 (MAPK3) was identified. Real-time RT-PCR confirmed differential expression of eight genes, including ESR1, MAPK3, methyl CpG binding protein 2 (MECP2), proline-serine-threonine phosphatase interacting protein 1 (PSTPIP1), Scr-like-adaptor (SLA), serine/threonine kinase 11 (STK11), WNK lysine-deficient protein kinase 1 (WNK1), and zinc finger protein 446 (ZNF446). CONCLUSIONS This is the first in vivo genome-wide expression study on human B cells in relation to osteoporosis. Our results highlight the significance of B cells in the etiology of osteoporosis and suggest a novel mechanism for postmenopausal osteoporosis (i.e., that downregulation of ESR1 and MAPK3 in B cells regulates secretion of factors, leading to increased osteoclastogenesis or decreased osteoblastogenesis).
Collapse
|
563
|
Abstract
INTRODUCTION B-cell leukemia/lymphoma 2 (Bcl2) is a proto-oncogene best known for its ability to suppress cell death. However, the role of Bcl2 in the skeletal system is unknown. Bcl2 has been hypothesized to play an important anti-apoptotic role in osteoblasts during anabolic actions of PTH. Although rational, this has not been validated in vivo; hence, the impact of Bcl2 in bone remains unknown. MATERIALS AND METHODS The bone phenotype of Bcl2 homozygous mutant (Bcl2(-/-)) mice was analyzed with histomorphometry and muCT. Calvarial osteoblasts were isolated and evaluated for their cellular activity. Osteoclastogenesis was induced from bone marrow cells using RANKL and macrophage-colony stimulating factor (M-CSF), and their differentiation was analyzed. PTH(1-34) (50 microg/kg) or vehicle was administered daily to Bcl2(+/+) and Bcl2(-/-) mice (4 days old) for 9 days to clarify the influence of Bcl2 ablation on PTH anabolic actions. Western blotting and real-time PCR were performed to detect Bcl2 expression in calvarial osteoblasts in response to PTH ex vivo. RESULTS There were reduced numbers of osteoclasts in Bcl2(-/-) mice, with a resultant increase in bone mass. Bcl2(-/-) bone marrow-derived osteoclasts ex vivo were significantly larger in size and short-lived compared with wildtype, suggesting a pro-apoptotic nature of Bcl2(-/-) osteoclasts. In contrast, osteoblasts were entirely normal in their proliferation, differentiation, and mineralization. Intermittent administration of PTH increased bone mass similarly in Bcl2(+/+) and Bcl2(-/-) mice. Finally, Western blotting and real-time PCR showed that Bcl2 levels were not induced in response to PTH in calvarial osteoblasts. CONCLUSIONS Bcl2 is critical in osteoclasts but not osteoblasts. Osteoclast suppression is at least in part responsible for increased bone mass of Bcl2(-/-) mice, and Bcl2 is dispensable in PTH anabolic actions during bone growth.
Collapse
|
564
|
Abstract
The majority of bone cell biology focuses on activity on the surface of the bone with little attention paid to the activity that occurs below the surface. However, with recent new discoveries, osteocytes, cells embedded within the mineralized matrix of bone, are becoming the target of intensive investigation. In this article, the distinctions between osteoblasts and their descendants, osteocytes, are reviewed. Osteoblasts are defined as cells that make bone matrix and osteocytes are thought to translate mechanical loading into biochemical signals that affect bone (re)modeling. Osteoblasts and osteocytes should have similarities as would be expected of cells of the same lineage, yet these cells also have distinct differences, particularly in their responses to mechanical loading and utilization of the various biochemical pathways to accomplish their respective functions. For example, the Wnt/beta-catenin signaling pathway is now recognized as an important regulator of bone mass and bone cell functions. This pathway is important in osteoblasts for differentiation, proliferation and the synthesis bone matrix, whereas osteocytes appear to use the Wnt/beta-catenin pathway to transmit signals of mechanical loading to cells on the bone surface. New emerging evidence suggests that the Wnt/beta-catenin pathway in osteocytes may be triggered by crosstalk with the prostaglandin pathway in response to loading which then leads to a decrease in expression of negative regulators of the pathway such as Sost and Dkk1. The study of osteocyte biology is becoming an intense area of research interest and this review will examine some of the recent findings that are reshaping our understanding of bone/bone cell biology.
Collapse
Affiliation(s)
- Lynda F Bonewald
- University of Missouri, Kansas City School of Dentistry, Department of Oral Biology, 650 East 25th Street, Kansas City, MO 64108, USA.
| | | |
Collapse
|
565
|
Yavorskyy A, Hernandez-Santana A, McCarthy G, McMahon G. Detection of calcium phosphate crystals in the joint fluid of patients with osteoarthritis - analytical approaches and challenges. Analyst 2008; 133:302-18. [PMID: 18299743 PMCID: PMC2625400 DOI: 10.1039/b716791a] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Clinically, osteoarthritis (OA) is characterised by joint pain, stiffness after immobility, limitation of movement and, in many cases, the presence of basic calcium phosphate (BCP) crystals in the joint fluid. The detection of BCP crystals in the synovial fluid of patients with OA is fraught with challenges due to the submicroscopic size of BCP, the complex nature of the matrix in which they are found and the fact that other crystals can co-exist with them in cases of mixed pathology. Routine analysis of joint crystals still relies almost exclusively on the use of optical microscopy, which has limited applicability for BCP crystal identification due to limited resolution and the inherent subjectivity of the technique. The purpose of this Critical Review is to present an overview of some of the main analytical tools employed in the detection of BCP to date and the potential of emerging technologies such as atomic force microscopy (AFM) and Raman microspectroscopy for this purpose.
Collapse
Affiliation(s)
- Alexander Yavorskyy
- Bioanalytical Chemistry & Diagnostics Group, National Centre for Sensor Research, School of Chemical Sciences, Dublin City University, Dublin 9, Ireland. ; Tel: +353 1 7005914
| | - Aaron Hernandez-Santana
- Bioanalytical Chemistry & Diagnostics Group, National Centre for Sensor Research, School of Chemical Sciences, Dublin City University, Dublin 9, Ireland. ; Tel: +353 1 7005914
| | - Geraldine McCarthy
- Division of Rheumatology, Mater Misericordiae University Hospital, Eccles St, Dublin 7, Ireland
| | - Gillian McMahon
- Bioanalytical Chemistry & Diagnostics Group, National Centre for Sensor Research, School of Chemical Sciences, Dublin City University, Dublin 9, Ireland. ; Tel: +353 1 7005914
| |
Collapse
|
566
|
Nerenz RD, Martowicz ML, Pike JW. An enhancer 20 kilobases upstream of the human receptor activator of nuclear factor-kappaB ligand gene mediates dominant activation by 1,25-dihydroxyvitamin D3. Mol Endocrinol 2008; 22:1044-56. [PMID: 18202151 DOI: 10.1210/me.2007-0380] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Receptor activator of nuclear factor-kappaB ligand (RANKL) is a TNF-like factor that is both produced by osteoblasts, mesenchymal cells, and activated T cells and required for osteoclast maturation and survival. The gene is up-regulated by the two primary calcemic hormones, 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] and PTH. Previous studies have indicated that five enhancer regions located significantly upstream of the mouse Rankl transcriptional start site mediate up-regulation by 1,25(OH)2D3 and PTH. The most distal of these, termed mRLD5, is highly conserved in the human gene at -96 kb where it was also shown to be functionally active. Four additional mouse Rankl upstream enhancers are also highly conserved in the human gene at -20, -25, -75, and -87 kb. In the present studies, we characterized the activity of these regions, explored their capacity to mediate the actions of 1,25(OH)2D3, and identified the vitamin D response elements contained within the two most proximal segments. Interestingly, whereas the most distal of the five enhancers is the dominant mediator of 1,25(OH)2D3 activity in the mouse Rankl gene, that role in the human gene is manifested by the most proximal element at -20 kb. Importantly, activity at this region in response to 1,25(OH)2D3 was associated with a significant increase in histone acetylation as well as the enhanced recruitment of RNA polymerase II. Both likely reflect the primary role of this enhancer in human RANKL gene expression. Our studies confirm the complex nature of RANKL regulation and indicate that although the five enhancers are evolutionarily conserved across several species, their relative contributions to RANKL expression in response to 1,25(OH)2D3 may be different.
Collapse
Affiliation(s)
- Robert D Nerenz
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | | | | |
Collapse
|
567
|
Seeman E. Bone quality: the material and structural basis of bone strength. J Bone Miner Metab 2008; 26:1-8. [PMID: 18095057 DOI: 10.1007/s00774-007-0793-5] [Citation(s) in RCA: 199] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Accepted: 08/13/2007] [Indexed: 01/23/2023]
Abstract
The material composition and structural design of bone determine its strength. Structure determines loads that can be tolerated but loads also determine structure. Bone modifies its material composition and structure to accommodate loads by adaptive modeling and remodeling. Adaptation is successful during growth but not aging because accumulating insults, including a reduction in the volume of bone formed in the basic multicellular unit (BMU), increased resorption in the BMU, increased remodeling rate in midlife in women and in some men because of sex hormone deficiency, and in both sexes in old age as a consequence of secondary hyperparathyroidism and reduced periosteal bone formation, all of which compromises the material composition of bone and its structure. An understanding of the mechanisms of adaptation and failed adaptation provides rational approaches to interventions that can prevent or restore bone fragility.
Collapse
Affiliation(s)
- Ego Seeman
- Department of Endocrinology and Medicine, Austin Hospital, Austin Health, Heidelberg 3084, Melbourne, Australia.
| |
Collapse
|
568
|
Naliato ECDO, Violante AHD, Caldas D, Farias MLF, Bussade I, Lamounier Filho A, Loureiro CR, Fontes R, Schrank Y, Loures T, Colao A. Bone density in women with prolactinoma treated with dopamine agonists. Pituitary 2008; 11:21-8. [PMID: 17661178 DOI: 10.1007/s11102-007-0064-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVES (1) to evaluate bone density in women with prolactinoma treated with dopamine agonists and healthy controls, using dual energy x-ray absorptiometry (DXA), (2) to classify the results according to the current International Society for Clinical Densitometry (ISCD) criteria, and (3) to correlate bone density with lean and fat masses, biochemical data and clinical aspects of prolactinomas. MATERIALS AND METHODS A cross-sectional study was performed in two University referral centers. Forty-five premenopausal women with prolactinoma were submitted to DXA and blood analysis (prolactin, estradiol, testosterone, SHBG, calcium, phosphorus, PTH, C-telopeptides of type 1 collagen, and osteocalcin) by the time of their clinical evaluation. They were compared with 25 control women of similar age and body mass index distribution. RESULTS Women with prolactinoma had lower lumbar spine Z-score than controls. Femoral neck, trochanter, and total proximal femur Z-scores were similar in patients and controls. Twenty-two percent of the patients had Z-scores below the expected age range vs. 4% in the control group. Lumbar spine, femoral neck, and total proximal femur Z-scores were mainly correlated with the amenorrhea duration. The trochanter Z-score was associated with the gynoid lean/fat mass ratio. CONCLUSIONS Based on the current ISCD criteria, bone density evaluation in women with prolactinoma reveals bone loss, especially of trabecular type. Bone density in these patients was particularly associated with the duration of amenorrhea, which reinforces the importance of the adequate disease control in women with prolactinoma in order to avoid complications of this disease.
Collapse
|
569
|
Abstract
Cancer therapy can result in significant bone loss and increased risk of fragility fracture. Chemotherapy, aromatase inhibitors, and gonadotropin-releasing hormone analogues contribute to increases in the rate of bone remodelling and reduce bone mineral density. Patients with prostate cancer on androgen deprivation therapy experience an increase in the risk of fracture. New research has demonstrated the key role played by bisphosphonates in preventing declines in bone density and increases in bone remodelling. Novel antiresorptive agents targeting receptor activator of nuclear factor κB ligand have great potential in skeletal protection and prevention of bone loss related to cancer therapy. Early assessment of skeletal health, followed by initiation of calcium, vitamin D, and an exercise program are valuable in the prevention and treatment of osteoporosis. In addition, individuals at increased risk for fracture should be offered antiresorptive therapy. Early data have demonstrated that bisphosphonates are able to prevent the bone loss and increased bone remodelling associated with cancer therapy, including aromatase inhibition and androgen deprivation therapy. The present paper reviews the new research and advances in the management of bone loss associated with both cancer therapy and estrogen deficiency in the postmenopausal female.
Collapse
Affiliation(s)
| | - A.A. Khan
- Correspondence to: Aliya A. Khan, Department of Medicine, McMaster University, 331–209 Sheddon Avenue, Oakville, Ontario L6J1X8 E-mail:
| |
Collapse
|
570
|
Haine V, Fischer-Smith T, Rappaport J. Macrophage colony-stimulating factor in the pathogenesis of HIV infection: potential target for therapeutic intervention. J Neuroimmune Pharmacol 2007; 1:32-40. [PMID: 18040789 DOI: 10.1007/s11481-005-9003-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Macrophage colony stimulating factor (M-CSF) appears to play a major role in promoting and maintaining reservoirs of human immunodeficiency virus type 1 (HIV-1) in infected individuals. HIV-1 infection induces production of M-CSF by macrophages, which in turn promotes further infection of macrophages via increases in CD4 and CCR5 receptors, as well as increases in virus gene expression. M-CSF promotes the ontogeny and survival of macrophages, contributing to both the number and longevity of these infected cells. M-CSF dysregulation promotes the differentiation of monocytes toward macrophages and osteoclasts and at the same time may inhibit differentiation toward dendritic cells, resulting in immune impairment. The potential role of M-CSF in HIV-associated end organ diseases including HIV-associated dementia, HIV-associated nephropathy, and osteoporosis is discussed. This review emphasizes the need for developing M-CSF antagonists for treatment of HIV-1-infected patients.
Collapse
Affiliation(s)
- Valerie Haine
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, 1900 N. 12th Street, Philadelphia, PA 19122, USA
| | | | | |
Collapse
|
571
|
Abstract
A functional endocannabinoid system is present in several mammalian organs and tissues. Recently, endocannabinoids and their receptors have been reported in the skeleton. Osteoblasts, the bone forming cells, and osteoclasts, the bone resorbing cells, produce the endocannabinoids anandamide and 2-arachidonoylglycerol and express CB2 cannabinoid receptors. Although CB2 has been implicated in pathological processes in the central nervous system and peripheral tissues, the skeleton appears as the main system physiologically regulated by CB2. CB2-deficient mice show a markedly accelerated age-related bone loss and the CNR2 gene (encoding CB2) in women is associated with low bone mineral density. The activation of CB2 attenuates ovariectomy-induced bone loss in mice by restraining bone resorption and enhancing bone formation. Hence synthetic CB2 ligands, which are stable and orally available, provide a basis for developing novel anti-osteoporotic therapies. Activation of CB1 in sympathetic nerve terminals in bone inhibits norepinephrine release, thus balancing the tonic sympathetic restrain of bone formation. Low levels of CB1 were also reported in osteoclasts. CB1-null mice display a skeletal phenotype that is dependent on the mouse strain, gender and specific mutation of the CB1 encoding gene, CNR1.
Collapse
|
572
|
Horsch K, de Wet H, Schuurmans MM, Allie-Reid F, Cato ACB, Cunningham J, Burrin JM, Hough FS, Hulley PA. Mitogen-activated protein kinase phosphatase 1/dual specificity phosphatase 1 mediates glucocorticoid inhibition of osteoblast proliferation. Mol Endocrinol 2007; 21:2929-40. [PMID: 17761948 PMCID: PMC2838148 DOI: 10.1210/me.2007-0153] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Steroid-induced osteoporosis is a common side effect of long-term treatment with glucocorticoid (GC) drugs. GCs have multiple systemic effects that may influence bone metabolism but also directly affect osteoblasts by decreasing proliferation. This may be beneficial at low concentrations, enhancing differentiation. However, high-dose treatment produces a severe deficit in the proliferative osteoblastic compartment. We provide causal evidence that this effect of GC is mediated by induction of the dual-specificity MAPK phosphatase, MKP-1/DUSP1. Excessive MKP-1 production is both necessary and sufficient to account for the impaired osteoblastic response to mitogens. Overexpression of MKP-1 after either GC treatment or transfection ablates the mitogenic response in osteoblasts. Knockdown of MKP-1 using either immunodepletion of MKP-1 before in vitro dephosphorylation assay or short interference RNA transfection prevents inactivation of ERK by GCs. Neither c-jun N-terminal kinase nor p38 MAPK is activated by the mitogenic cocktail in 20% fetal calf serum, but their activation by a DNA-damaging agent (UV irradiation) was inhibited by either GC treatment or overexpression of MKP-1, indicating regulation of all three MAPKs by MKP-1 in osteoblasts. However, an inhibitor of the MAPK/ERK kinase-ERK pathway inhibited osteoblast proliferation whereas inhibitors of c-jun N-terminal kinase or p38 MAPK had no effect, suggesting that ERK is the MAPK that controls osteoblast proliferation. Regulation of ERK by MKP-1 provides a novel mechanism for control of osteoblast proliferation by GCs.
Collapse
Affiliation(s)
- Kay Horsch
- Division of Endocrinology and Metabolism, Department of Medicine, University of Stellenbosch, Stellenbosch 7505, South Africa
| | | | | | | | | | | | | | | | | |
Collapse
|
573
|
Conradie MM, de Wet H, Kotze DDR, Burrin JM, Hough FS, Hulley PA. Vanadate prevents glucocorticoid-induced apoptosis of osteoblasts in vitro and osteocytes in vivo. J Endocrinol 2007; 195:229-40. [PMID: 17951534 PMCID: PMC2173947 DOI: 10.1677/joe-07-0217] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Skeletal mass is maintained by a balance between formation and resorption, cell proliferation and apoptosis. In vitro, glucocorticoids (GCs) decrease extracellular signal-regulated kinases (ERK) activation by mitogens, thus inhibiting osteoblast proliferation. Both ERK activity and proliferation are restored by co-treatment with the protein tyrosine phosphatase inhibitor, vanadate. Since ERK signalling may also be anti-apoptotic, we explored the effects of vanadate on GC-induced apoptosis in vitro and in vivo. Apoptosis in MBA-15.4 pre-osteoblasts increased from 6 h and remained up to eightfold higher through 6 days of 10(- 6) M dexamethasone (Dex) treatment. Co-incubation with 10(- 7) M vanadate markedly reduced apoptosis at all time points. Vanadate also prevented GC-induced poly-ADP-ribose polymerase cleavage. We assessed the transcriptional profiles of seven anti-apoptotic proteins (Bcl-2, Bcl-X(L), inhibitors of apoptosis protein-1 (IAP-1), IAP-2, X-linked IAP (XIAP), Fas-associated death-domain-like IL-1beta-converting enzyme-inhibitory protein (FLIP(Long)) and FLIP(Short)) in osteoblasts subjected to various stimuli using real-time quantitative PCR. Although these anti-apoptotic genes responded to different mitogenic conditions, Dex failed to repress their expression, and in fact significantly up-regulated Bcl-X(L), IAP-2 and XIAP. Dex may therefore induce apoptosis by up-regulating pro-apoptotic gene expression. We have previously demonstrated that rats treated with GC develop low formation osteoporosis (bone histomorphometry and DEXA) and skeletal fragility (breaking strength) that were largely prevented by co-treatment with vanadate. We report here that vertebrae from rats treated with 3.5 mg/kg per day methylprednisolone for 9 weeks showed increased incidence of terminal deoxynucleotidyl transferase-mediated biotin-dUTP nick end-labelling-positive apoptotic osteocytes, which was reduced by vanadate co-treatment. We conclude that vanadate prevents GC-induced apoptosis of pre-osteoblasts in vitro and osteocytes in vivo, and this may contribute to its bone-sparing effects in vivo.
Collapse
Affiliation(s)
- M M Conradie
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Health Sciences, University of StellenboschTygerbergSouth Africa
| | - H de Wet
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Health Sciences, University of StellenboschTygerbergSouth Africa
| | - D D R Kotze
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Health Sciences, University of StellenboschTygerbergSouth Africa
| | - J M Burrin
- Department of Endocrinology, St Bart's Hospital, University of LondonLondonUK
| | - F S Hough
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Health Sciences, University of StellenboschTygerbergSouth Africa
| | - P A Hulley
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Health Sciences, University of StellenboschTygerbergSouth Africa
- Institute of Musculoskeletal Sciences, Botnar Research Centre, Nuffield Orthopaedic Centre, University of OxfordHeadington, Oxford OX3 7LDUK
- (Correspondence should be addressed to P A Hulley; )
| |
Collapse
|
574
|
Yates LB, Karasik D, Beck TJ, Cupples LA, Kiel DP. Hip structural geometry in old and old-old age: similarities and differences between men and women. Bone 2007; 41:722-32. [PMID: 17662680 PMCID: PMC2198902 DOI: 10.1016/j.bone.2007.06.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2006] [Revised: 05/25/2007] [Accepted: 06/04/2007] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Changes in hip structure and geometry during aging contribute to decreased bone strength. Little is known, however, about these characteristics at advanced age, when fragility fractures are common. We examined hip structural geometry in men and women of old age (72-84 years) and old-old age (85-96 years) to determine (1) gender differences; (2) whether or not these differences are consistent with the increased occurrence of hip fracture in elderly women, compared to men; and (3) whether or not gender-specific changes are consistent with the increased occurrence of fragility fractures after age 80 in both men and women. METHODS We used Hip Structure Analysis (HSA) software to analyze bone densitometry scans from 916 community-dwelling men and women aged 72-96 years. We examined gender differences in hip geometry by age group (72-74, 75-79, 80-84, and >or=85 years) and between gender-specific age groups using multivariable linear regression. RESULTS At the femoral narrow neck, there was no gender difference at age 72-74 in bone mineral density (BMD), cortical thickness (CT), and buckling ratio (BR). In contrast, at age 85 or older women had 13% less BMD and CT than men and 8% higher BR. At the intertrochanteric region, women >or=85 years had 25-31% less BMD, cross-sectional bone area (CSA), and CT than men of comparable age, and 38% higher BR. These gender differences were approximately 10-20% greater than those between men and women in their 70s. In gender-specific comparisons, women showed increasing change in structural geometry with increasing age. At both narrow neck and trochanteric regions, women >or=85 years had nearly 35% higher BR, 15% less BMD and CT, and 10% less CSA than women aged 72-74 years. At the narrow neck, they also had 6% greater outer diameter than the youngest women and 8% lower section modulus (Z), an index of bending strength. In contrast, men showed significant age differences only at the narrow neck region, and only at 85 years or older, including 22% higher BR, 10% less BMD and CT, and 5% greater outer diameter, compared to men in their early 70s. Unlike women, men showed no age-associated decline in section modulus. CONCLUSIONS Gender differences in hip geometry consistent with increased fragility and fracture risk in elderly women, compared to men, continue into old-old age. Both men and women 85 or older show the most unfavorable features, suggesting a structural basis for the increased occurrence of hip fracture in both sexes at advanced age.
Collapse
Affiliation(s)
- Laurel B Yates
- Division of Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02120, USA.
| | | | | | | | | |
Collapse
|
575
|
Almeida M, Han L, Martin-Millan M, Plotkin LI, Stewart SA, Roberson PK, Kousteni S, O'Brien CA, Bellido T, Parfitt AM, Weinstein RS, Jilka RL, Manolagas SC. Skeletal involution by age-associated oxidative stress and its acceleration by loss of sex steroids. J Biol Chem 2007; 282:27285-27297. [PMID: 17623659 PMCID: PMC3119455 DOI: 10.1074/jbc.m702810200] [Citation(s) in RCA: 537] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Both aging and loss of sex steroids have adverse effects on skeletal homeostasis, but whether and how they may influence each others negative impact on bone remains unknown. We report herein that both female and male C57BL/6 mice progressively lost strength (as determined by load-to-failure measurements) and bone mineral density in the spine and femur between the ages of 4 and 31 months. These changes were temporally associated with decreased rate of remodeling as evidenced by decreased osteoblast and osteoclast numbers and decreased bone formation rate; as well as increased osteoblast and osteocyte apoptosis, increased reactive oxygen species levels, and decreased glutathione reductase activity and a corresponding increase in the phosphorylation of p53 and p66(shc), two key components of a signaling cascade that are activated by reactive oxygen species and influences apoptosis and lifespan. Exactly the same changes in oxidative stress were acutely reproduced by gonadectomy in 5-month-old females or males and reversed by estrogens or androgens in vivo as well as in vitro. We conclude that the oxidative stress that underlies physiologic organismal aging in mice may be a pivotal pathogenetic mechanism of the age-related bone loss and strength. Loss of estrogens or androgens accelerates the effects of aging on bone by decreasing defense against oxidative stress.
Collapse
Affiliation(s)
- Maria Almeida
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, and the
| | - Li Han
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, and the
| | - Marta Martin-Millan
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, and the
| | - Lilian I Plotkin
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, and the
| | - Scott A Stewart
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, and the
| | - Paula K Roberson
- Department of Biostatistics, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Health Care System, Little Rock, Arkansas 72205
| | - Stavroula Kousteni
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, and the
| | - Charles A O'Brien
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, and the
| | - Teresita Bellido
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, and the
| | - A Michael Parfitt
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, and the
| | - Robert S Weinstein
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, and the
| | - Robert L Jilka
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, and the
| | - Stavros C Manolagas
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, and the.
| |
Collapse
|
576
|
Papapoulos SE, Schimmer RC. Changes in bone remodelling and antifracture efficacy of intermittent bisphosphonate therapy: implications from clinical studies with ibandronate. Ann Rheum Dis 2007; 66:853-8. [PMID: 17277001 PMCID: PMC1955119 DOI: 10.1136/ard.2006.064931] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2007] [Indexed: 11/04/2022]
Abstract
Bisphosphonates reduce the rate of bone resorption and bone remodelling. Given daily, they decrease the risk of fractures in postmenopausal osteoporosis. When bisphosphonates were given at extended drug-free intervals this antifracture efficacy was generally not seen. This may be due to the different pattern of bone remodelling changes. Data from randomised clinical studies of ibandronate, given orally or intravenously, at different doses and for variable time intervals to women with osteoporosis were examined to explore the relationship between intermittent bisphosphonate therapy, changes in bone resorption and fracture risk. The magnitude of the reduction of the rate of bone resorption at the end of the drug-free interval rather than its fluctuation pattern after bisphosphonate administration determines antifracture efficacy, provided that these fluctuations occur within the premenopausal range. Prolongation of the drug-free interval beyond 2 weeks should be compensated by a dose higher than the cumulative daily dose.
Collapse
Affiliation(s)
- S E Papapoulos
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Centre, Albinusdreef 2, 2333 ZA Leiden, The Netherlands.
| | | |
Collapse
|
577
|
|
578
|
Jilka RL. Molecular and cellular mechanisms of the anabolic effect of intermittent PTH. Bone 2007; 40:1434-46. [PMID: 17517365 PMCID: PMC1995599 DOI: 10.1016/j.bone.2007.03.017] [Citation(s) in RCA: 501] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2006] [Revised: 03/14/2007] [Accepted: 03/20/2007] [Indexed: 12/24/2022]
Abstract
Intermittent administration of parathyroid hormone (PTH) stimulates bone formation by increasing osteoblast number, but the molecular and cellular mechanisms underlying this effect are not completely understood. In vitro and in vivo studies have shown that PTH directly activates survival signaling in osteoblasts; and that delay of osteoblast apoptosis is a major contributor to the increased osteoblast number, at least in mice. This effect requires Runx2-dependent expression of anti-apoptotic genes like Bcl-2. PTH also causes exit of replicating progenitors from the cell cycle by decreasing expression of cyclin D and increasing expression of several cyclin-dependent kinase inhibitors. Exit from the cell cycle may set the stage for pro-differentiating and pro-survival effects of locally produced growth factors and cytokines, the level and/or activity of which are known to be influenced by PTH. Observations from genetically modified mice suggest that the anabolic effect of intermittent PTH requires insulin-like growth factor-I (IGF-I), fibroblast growth factor-2 (FGF-2), and perhaps Wnts. Attenuation of the negative effects of PPAR gamma may also lead to increased osteoblast number. Daily injections of PTH may add to the pro-differentiating and pro-survival effects of locally produced PTH related protein (PTHrP). As a result, osteoblast number increases beyond that needed to replace the bone removed by osteoclasts during bone remodeling. The pleiotropic effects of intermittent PTH, each of which alone may increase osteoblast number, may explain why this therapy reverses bone loss in most osteoporotic individuals regardless of the underlying pathophysiology.
Collapse
Affiliation(s)
- Robert L Jilka
- Division of Endocrinology and Metabolism, Slot 587 Center for Osteoporosis and Metabolic Bone Diseases, Central Arkansas Veterans Healthcare System, University of Arkansas for Medical Sciences, 4301 W. Markham, Little Rock, AR 72205, USA.
| |
Collapse
|
579
|
Hong X, Hsu YH, Terwedow H, Arguelles LM, Tang G, Liu X, Zhang S, Xu X, Xu X. CYP19A1 polymorphisms are associated with bone mineral density in Chinese men. Hum Genet 2007; 121:491-500. [PMID: 17216495 DOI: 10.1007/s00439-006-0303-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2006] [Accepted: 11/18/2006] [Indexed: 11/27/2022]
Abstract
Aromatase-dependent biosynthesis of estrogen plays an important role in maintenance of the male skeleton, and Cytochrome p450 aromatase is the key enzyme to catalyze the conversion of androgen precursors to estrogens. We investigated the association of polymorphisms in the CYP19A1 gene and bone mineral density in a Chinese cohort. 2392 extreme low femoral neck BMD cases or extreme high femoral neck BMD controls were selected from a population-based cohort and genotyped for eight SNPs in the CYP19A1 gene. Significant associations for rs17703883, rs12594287 and rs16964201 SNPs with BMD were found in men only. Men with TC/CC genotypes in the rs17703883 SNP had a 1.5 times higher risk of having extreme low femoral neck BMD (P = 0.003, empirical P value = 0.05), and decreased BMDs at total body (P = 0.004, empirical P value = 0.07) and total hip (P = 0.003, empirical P value = 0.05). Men carrying AA/AG genotypes in the rs12594287 SNP had a 30% reduced risk of having extreme low femoral neck BMD (P = 0.007, empirical P value = 0.12), and increased BMDs at total body (P = 0.0009, empirical P value = 0.018) and total hip (P = 0.001, empirical P value = 0.02). Men carrying TT/TC genotypes in the rs16964201 SNP had a 40% reduced risk of having extreme low femoral neck BMD (P = 0.005, empirical P value = 0.087), and increased BMDs at total body (P = 0.0001, empirical P value = 0.002) and total hip (P = 0.0006, empirical P value = 0.012). Haplotype analysis showed that the G-C-T-A-T haplotype was significantly related to higher BMD. Our finding suggests that genetic variations in the CYP19A1 gene are significantly associated with BMD at different skeletal sites in adult men, but not in women.
Collapse
Affiliation(s)
- Xiumei Hong
- School of Life Science, University of Science and Technology of China, Huangshan Road, Hefei City, Anhui Province, China
| | | | | | | | | | | | | | | | | |
Collapse
|
580
|
Sato T, Watanabe K, Masuhara M, Hada N, Hakeda Y. Production of IL-7 is increased in ovariectomized mice, but not RANKL mRNA expression by osteoblasts/stromal cells in bone, and IL-7 enhances generation of osteoclast precursors in vitro. J Bone Miner Metab 2007; 25:19-27. [PMID: 17187190 DOI: 10.1007/s00774-006-0723-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2006] [Accepted: 06/30/2006] [Indexed: 12/15/2022]
Abstract
Osteoclastogenic cytokines produced by T and B lineage cells and interleukin (IL)-7-induced expansion of the pool size of osteoclast precursors have been suggested to play an important role in acceleration of osteoclastogenesis induced by estrogen deficiency. However, the contribution of increased RANKL produced by osteoblasts/stromal cells to increase osteoclastogenesis in a mouse model of estrogen-deficient osteoporosis and in vitro effects of IL-7 on osteoclast precursor generation remain controversial. Thus, we investigated the effect of ovariectomy (OVX) of mice on production of RANKL, osteoprotegerin (OPG), and IL-7 in bone and the effect of IL-7 on osteoclast precursor generation in vitro. OVX did not significantly stimulate mRNA expressions of RANKL and OPG in whole femurs. Because the epiphysis, but not the femoral shaft (diaphysis) or bone marrow, is the main site of osteoclastogenesis, it is important to specifically analyze mRNA expression by osteoblasts/stromal cells at these parts of the femur. Therefore, we isolated RNA from bone marrow cell-free epiphysis, diaphysis, and flushed-out bone marrow and examined mRNA expression. The results showed no significant changes of RANKL and OPG mRNA expression in any part of the femur. In addition, OVX did not significantly affect RANKL and OPG mRNA expression by the adherent stromal cells isolated from flushed-out bone marrow cells but did stimulate RANKL mRNA expression by B220(+) cells in the nonadherent cell fraction. On the other hand, OVX increased IL-7 mRNA expression in the femur as well as IL-7 concentrations in bone fluid. In cultures of unfractionated bone cells isolated by vigorous agitation of minced whole long bones to release the cells tightly attached to the bone surfaces, but not in cocultures of clonal osteoblasts/stromal cells and flushed-out bone marrow cells, IL-7 stimulated generations of osteoclasts as well as osteoclast precursors. These data suggest that increased RANKL production by osteoblasts/stromal cells is unlikely to play a central role in acceleration of osteoclastogenesis in estrogen deficiency of mice and that IL-7 stimulates osteoclast precursor generation, presumably through an action of IL-7 on the cells attached to bone rather than on cells contained in the bone marrow cell population.
Collapse
Affiliation(s)
- Takuya Sato
- Division of Oral Anatomy, Department of Human Development and Fostering, Meikai University School of Dentistry, 1-1 Keyaki-dai, Sakado, 350-0283, Japan.
| | | | | | | | | |
Collapse
|
581
|
Migliaccio S, Brama M, Spera G. The differential effects of bisphosphonates, SERMS (selective estrogen receptor modulators), and parathyroid hormone on bone remodeling in osteoporosis. Clin Interv Aging 2007; 2:55-64. [PMID: 18044075 PMCID: PMC2684086 DOI: 10.2147/ciia.2007.2.1.55] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Osteoporosis is a skeletal metabolic disease characterized by a compromised bone fragility, leading to an increased risk of developing spontaneous and traumatic fractures. Osteoporosis is considered a multifactorial disease and fractures are the results of several different risk factors both extra- and intraskeletal. Thus bone fragility can be the end point of several different causes: a) failure to reach an optimal peak bone mass during growth; b) excessive bone resorption resulting in decreased bone mass and microarchitectural deterioration; c) inadequate formation upon an increased resorption during the process of bone remodeling. The pharmacological therapeutical options, available to date, are directed on prevention of fractures. The aim of this paper is to describe the activities and the mechanisms of action, as known at present, of the most used therapies for osteoporosis and their clinical implications. Improvement of knowledge in this field will allow us to further improve therapeutical choices and pharmacological interventions.
Collapse
Affiliation(s)
- Silvia Migliaccio
- Cattedra di Medicina Interna, Dipartimento di Fisiopatologia Medica, Università degli Studi di Roma La Sapienza, Italy.
| | | | | |
Collapse
|
582
|
Windahl SH, Galien R, Chiusaroli R, Clément-Lacroix P, Morvan F, Lepescheux L, Nique F, Horne WC, Resche-Rigon M, Baron R. Bone protection by estrens occurs through non-tissue-selective activation of the androgen receptor. J Clin Invest 2006; 116:2500-9. [PMID: 16955145 PMCID: PMC1555662 DOI: 10.1172/jci28809] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2006] [Accepted: 06/20/2006] [Indexed: 11/17/2022] Open
Abstract
The use of estrogens and androgens to prevent bone loss is limited by their unwanted side effects, especially in reproductive organs and breast. Selective estrogen receptor modulators (SERMs) partially avoid such unwanted effects, but their efficacy on bone is only moderate compared with that of estradiol or androgens. Estrens have been suggested to not only prevent bone loss but also exert anabolic effects on bone while avoiding unwanted effects on reproductive organs. In this study, we compared the effects of a SERM (PSK3471) and 2 estrens (estren-alpha and estren-beta) on bone and reproductive organs to determine whether estrens are safe and act via the estrogen receptors and/or the androgen receptor (AR). Estrens and PSK3471 prevented gonadectomy-induced bone loss in male and female mice, but none showed true anabolic effects. Unlike SERMs, the estrens induced reproductive organ hypertrophy in both male and female mice and enhanced MCF-7 cell proliferation in vitro. Estrens directly activated transcription in several cell lines, albeit at much higher concentrations than estradiol or the SERM, and acted for the most part through the AR. We conclude that the estrens act mostly through the AR and, in mice, do not fulfill the preclinical efficacy or safety criteria required for the treatment or prevention of osteoporosis.
Collapse
Affiliation(s)
- Sara H. Windahl
- Department of Orthopaedics, Yale University School of Medicine, New Haven, Connecticut, USA.
ProStrakan Pharmaceuticals, Romainville, France.
Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - René Galien
- Department of Orthopaedics, Yale University School of Medicine, New Haven, Connecticut, USA.
ProStrakan Pharmaceuticals, Romainville, France.
Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Riccardo Chiusaroli
- Department of Orthopaedics, Yale University School of Medicine, New Haven, Connecticut, USA.
ProStrakan Pharmaceuticals, Romainville, France.
Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Philippe Clément-Lacroix
- Department of Orthopaedics, Yale University School of Medicine, New Haven, Connecticut, USA.
ProStrakan Pharmaceuticals, Romainville, France.
Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Frederic Morvan
- Department of Orthopaedics, Yale University School of Medicine, New Haven, Connecticut, USA.
ProStrakan Pharmaceuticals, Romainville, France.
Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Liên Lepescheux
- Department of Orthopaedics, Yale University School of Medicine, New Haven, Connecticut, USA.
ProStrakan Pharmaceuticals, Romainville, France.
Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - François Nique
- Department of Orthopaedics, Yale University School of Medicine, New Haven, Connecticut, USA.
ProStrakan Pharmaceuticals, Romainville, France.
Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - William C. Horne
- Department of Orthopaedics, Yale University School of Medicine, New Haven, Connecticut, USA.
ProStrakan Pharmaceuticals, Romainville, France.
Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Michèle Resche-Rigon
- Department of Orthopaedics, Yale University School of Medicine, New Haven, Connecticut, USA.
ProStrakan Pharmaceuticals, Romainville, France.
Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Roland Baron
- Department of Orthopaedics, Yale University School of Medicine, New Haven, Connecticut, USA.
ProStrakan Pharmaceuticals, Romainville, France.
Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
583
|
Kim S, Yamazaki M, Zella LA, Shevde NK, Pike JW. Activation of receptor activator of NF-kappaB ligand gene expression by 1,25-dihydroxyvitamin D3 is mediated through multiple long-range enhancers. Mol Cell Biol 2006; 26:6469-86. [PMID: 16914732 PMCID: PMC1592822 DOI: 10.1128/mcb.00353-06] [Citation(s) in RCA: 157] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
RANKL is a tumor necrosis factor (TNF)-like factor secreted by mesenchymal cells, osteoblast derivatives, and T cells that is essential for osteoclastogenesis. In osteoblasts, RANKL expression is regulated by two major calcemic hormones, 1,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3)] and parathyroid hormone (PTH), as well as by several inflammatory/osteoclastogenic cytokines; the molecular mechanisms for this regulation are unclear. To identify such mechanisms, we screened a DNA microarray which tiled across the entire mouse RankL gene locus at a 50-bp resolution using chromatin immunoprecipitation (ChIP)-derived DNA precipitated with antibodies to the vitamin D receptor (VDR) and the retinoid X receptor (RXR). Five sites of dimer interaction were observed on the RankL gene centered at 16, 22, 60, 69, and 76 kb upstream of the TSS. These regions contained binding sites for not only VDR and RXR, but also the glucocorticoid receptor (GR). The most distant of these regions, termed the distal control region (RL-DCR), conferred both VDR-dependent 1,25(OH)(2)D(3) and GR-dependent glucocorticoid (GC) responses. We mapped these activities to an unusual but functionally active vitamin D response element and to several potential GC response elements located over a more extensive region within the RL-DCR. An evolutionarily conserved region within the human RANKL gene contained a similar vitamin D response element and exhibited an equivalent behavior. Importantly, hormonal activation of the RankL gene was also associated with chromatin modification and RNA polymerase II recruitment. Our studies demonstrate that regulation of RankL gene expression by 1,25(OH)(2)D(3) is complex and mediated by at least five distal regions, one of which contains a specific element capable of mediating direct transcriptional activation.
Collapse
Affiliation(s)
- Sungtae Kim
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Dr., Madison, WI 53706, USA
| | | | | | | | | |
Collapse
|
584
|
Abstract
Estrogen plays a fundamental role in skeletal growth and bone homeostasis in both men and women. Although remarkable progress has been made in our understanding of how estrogen deficiency causes bone loss, the mechanisms involved have proven to be complex and multifaceted. Although estrogen is established to have direct effects on bone cells, recent animal studies have identified additional unexpected regulatory effects of estrogen centered at the level of the adaptive immune response. Furthermore, a potential role for reactive oxygen species has now been identified in both humans and animals. One major challenge is the integration of a multitude of redundant pathways and cytokines, each apparently capable of playing a relevant role, into a comprehensive model of postmenopausal osteoporosis. This Review presents our current understanding of the process of estrogen deficiency-mediated bone destruction and explores some recent findings and hypotheses to explain estrogen action in bone. Due to the inherent difficulties associated with human investigation, many of the lessons learned have been in animal models. Consequently, many of these principles await further validation in humans.
Collapse
Affiliation(s)
- M. Neale Weitzmann
- Division of Endocrinology, Metabolism, and Lipids and
Molecular Pathogenesis Program, Emory University, Atlanta, Georgia, USA
| | - Roberto Pacifici
- Division of Endocrinology, Metabolism, and Lipids and
Molecular Pathogenesis Program, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
585
|
Phadke PA, Mercer RR, Harms JF, Jia Y, Frost AR, Jewell JL, Bussard KM, Nelson S, Moore C, Kappes JC, Gay CV, Mastro AM, Welch DR. Kinetics of metastatic breast cancer cell trafficking in bone. Clin Cancer Res 2006; 12:1431-40. [PMID: 16533765 PMCID: PMC1523260 DOI: 10.1158/1078-0432.ccr-05-1806] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
PURPOSE In vivo studies have focused on the latter stages of the bone metastatic process (osteolysis), whereas little is known about earlier events, e.g., arrival, localization, and initial colonization. Defining these initial steps may potentially identify the critical points susceptible to therapeutic intervention. EXPERIMENTAL DESIGN MDA-MB-435 human breast cancer cells engineered with green fluorescent protein were injected into the cardiac left ventricle of athymic mice. Femurs were analyzed by fluorescence microscopy, immunohistochemistry, real-time PCR, flow cytometry, and histomorphometry at times ranging from 1 hour to 6 weeks. RESULTS Single cells were found in distal metaphyses at 1 hour postinjection and remained as single cells up to 72 hours. Diaphyseal arrest occurred rarely and few cells remained there after 24 hours. At 1 week, numerous foci (2-10 cells) were observed, mostly adjacent to osteoblast-like cells. By 2 weeks, fewer but larger foci (> or =50 cells) were seen. Most bones had a single large mass at 4 weeks (originating from a colony or coalescing foci) which extended into the diaphysis by 4 to 6 weeks. Little change (<20%) in osteoblast or osteoclast numbers was observed at 2 weeks, but at 4 to 6 weeks, osteoblasts were dramatically reduced (8% of control), whereas osteoclasts were reduced modestly (to approximately 60% of control). CONCLUSIONS Early arrest in metaphysis and minimal retention in diaphysis highlight the importance of the local milieu in determining metastatic potential. These results extend the Seed and Soil hypothesis by demonstrating both intertissue and intratissue differences governing metastatic location. Ours is the first in vivo evidence that tumor cells influence not only osteoclasts, as widely believed, but also eliminate functional osteoblasts, thereby restructuring the bone microenvironment to favor osteolysis. The data may also explain why patients receiving bisphosphonates fail to heal bone despite inhibiting resorption, implying that concurrent strategies that restore osteoblast function are needed to effectively treat osteolytic bone metastases.
Collapse
Affiliation(s)
| | - Robyn R. Mercer
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA
| | | | | | - Andra R. Frost
- Departments of Pathology
- Comprehensive Cancer Center
- NFCR-Center for Metastasis Research, University of Alabama at Birmingham, Birmingham, AL and
| | - Jennifer L. Jewell
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA
| | - Karen M. Bussard
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA
| | - Shakira Nelson
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA
| | | | | | - Carol V. Gay
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA
| | - Andrea M. Mastro
- NFCR-Center for Metastasis Research, University of Alabama at Birmingham, Birmingham, AL and
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA
| | - Danny R. Welch
- Departments of Pathology
- Comprehensive Cancer Center
- Center for Metabolic Bone Disease
- NFCR-Center for Metastasis Research, University of Alabama at Birmingham, Birmingham, AL and
- Requests for reprints: Danny R. Welch, Ph.D. Department of Pathology, University of Alabama at Birmingham; 1670 University Blvd.; Volker Hall G-019A; Birmingham, AL 35294-0019; Phone: +1-205-934-2961 Fax: +1-205-975-1126; ; or, Andrea M. Mastro, Ph.D., Department of Biochemistry and Molecular Biology, 231 South Frear, Pennsylvania State University, University Park, PA 16802; Phone: +1-814-863-0152; Fax: +1-814-863-7024;
| |
Collapse
|
586
|
Abstract
The quest for effective treatment for osteoporosis merits great attention because of the widespread prevalence of this disease, which is not only associated with fragility fractures, but also with significant morbidity and mortality. The efficacy of the antiresorptive drugs in this disease is achieved by reducing bone turnover, increasing bone density and improving other aspects of bone quality. This article concentrates on another approach to the treatment of osteoporosis, namely the use of anabolic therapy, which has even greater prospects for improving bone quality. Parathyroid hormone (PTH) is currently available only as the recombinant amino-terminal fragment, PTH(1-34), known as teriparatide. The full-length molecule, human PTH(1-84), is currently being investigated, as are other PTH molecules. Teriparatide improves bone quality through actions on bone turnover, bone density, bone size and bone microarchitecture. In postmenopausal women with osteoporosis, teriparatide reduces the incidence of vertebral and nonvertebral fractures. In individuals who have previously been treated with an antiresorptive agent, the subsequent actions of teriparatide on bone density are transiently delayed if bone turnover has been markedly suppressed. Combination therapy with teriparatide or PTH(1-84) and an antiresorptive agent does not appear, at this time, to offer advantages over the use of PTH or an antiresorptive agent alone. However, in order to maintain the densitometric gains in bone density obtained with PTH, it is important to follow its use with that of an antiresorptive agent.
Collapse
Affiliation(s)
- Mishaela R Rubin
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | | |
Collapse
|
587
|
Richard S, Torabi N, Franco GV, Tremblay GA, Chen T, Vogel G, Morel M, Cléroux P, Forget-Richard A, Komarova S, Tremblay ML, Li W, Li A, Gao YJ, Henderson JE. Ablation of the Sam68 RNA binding protein protects mice from age-related bone loss. PLoS Genet 2005; 1:e74. [PMID: 16362077 PMCID: PMC1315279 DOI: 10.1371/journal.pgen.0010074] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2005] [Accepted: 11/11/2005] [Indexed: 02/02/2023] Open
Abstract
The Src substrate associated in mitosis of 68 kDa (Sam68) is a KH-type RNA binding protein that has been shown to regulate several aspects of RNA metabolism; however, its physiologic role has remained elusive. Herein we report the generation of Sam68-null mice by homologous recombination. Aged Sam68−/− mice preserved their bone mass, in sharp contrast with 12-month-old wild-type littermates in which bone mass was decreased up to approximately 75%. In fact, the bone volume of the 12-month-old Sam68−/− mice was virtually indistinguishable from that of 4-month-old wild-type or Sam68−/− mice. Sam68−/− bone marrow stromal cells had a differentiation advantage for the osteogenic pathway. Moreover, the knockdown of Sam68 using short hairpin RNA in the embryonic mesenchymal multipotential progenitor C3H10T1/2 cells resulted in more pronounced expression of the mature osteoblast marker osteocalcin when differentiation was induced with bone morphogenetic protein-2. Cultures of mouse embryo fibroblasts generated from Sam68+/+ and Sam68−/− littermates were induced to differentiate into adipocytes with culture medium containing pioglitazone and the Sam68−/− mouse embryo fibroblasts shown to have impaired adipocyte differentiation. Furthermore, in vivo it was shown that sections of bone from 12-month-old Sam68−/− mice had few marrow adipocytes compared with their age-matched wild-type littermate controls, which exhibited fatty bone marrow. Our findings identify endogenous Sam68 as a positive regulator of adipocyte differentiation and a negative regulator of osteoblast differentiation, which is consistent with Sam68 being a modulator of bone marrow mesenchymal cell differentiation, and hence bone metabolism, in aged mice. Osteoporosis is a debilitating bone disease that is characterized by reduced bone mass and microarchitectural damage, which result in increased bone fragility and susceptibility to fracture. Peak bone mass, which is achieved by the age of 30 in humans, has been identified as a major determinant of resistance or susceptibility to osteoporosis. The authors generated mice deficient for the Sam68 RNA binding protein, a protein of unknown physiologic function. The mice develop normally and are protected against bone loss during aging. Age-related bone loss has long been associated with an increase in marrow adipocytes, which are derived from the same mesenchymal lineage as osteoblasts in bone marrow. The authors showed that Sam68 regulates the differentiation of this mesenchymal lineage, such that in its absence, osteoblasts continued to be generated in aging bone, leading to preservation of bone mass. This study identifies a physiologic role for Sam68 as a modulator of the bone marrow stem cell niche and hence of bone metabolism. The data identify Sam68 as a potential therapeutic target for the prevention and treatment of age-related bone loss.
Collapse
Affiliation(s)
- Stéphane Richard
- Terry Fox Molecular Oncology Group and the Bloomfield Center for Research on Aging, Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, Montréal, Québec, Canada.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
588
|
Young N, Mikhalkevich N, Yan Y, Chen D, Zheng WP. Differential regulation of osteoblast activity by Th cell subsets mediated by parathyroid hormone and IFN-gamma. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2005; 175:8287-95. [PMID: 16339569 PMCID: PMC2647996 DOI: 10.4049/jimmunol.175.12.8287] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Bone loss is a typical pathological feature of chronic inflammatory bone diseases including rheumatoid arthritis, in which CD4 effector T cells play critical roles. We found that activated mouse Th2 and not Th1 cells produced the parathyroid hormone (PTH). Unlike in the parathyroid cells, PTH expression in Th2 cells was not regulated by the fluctuation of calcium level, but rather it required the full activation of the T cells. Although PTH was expressed in immature Th2 cells, and its receptor was transiently expressed during Th1 and Th2 cell differentiation, PTH did not significantly affect the outcome of the differentiation. In primary osteoblasts cultured in Th2 cell condition medium, the alkaline phosphatase (ALP) activity was maintained at a basal level. However, antagonizing PTH in the condition medium resulted in a significant reduction of the ALP activity. These results demonstrated an important role of the Th2 cell-derived PTH in maintaining the bone-forming activity of the osteoblasts under inflammatory conditions. In osteoblasts cultured in the Th1 cell condition medium, the ALP activity was significantly suppressed. Neutralizing IFN-gamma alleviated the suppression. Conversely, treatment of osteoblasts with IFN-gamma suppressed the ALP activity. Unlike ALP, expression of the major bone matrix proteins by the osteoblasts was only minimally affected by either Th1 or Th2 cytokine environment. In addition, the Th2 cytokine environment also regulated to expression of receptor activator of NF-kappaB ligand and osteoprotegerin through both PTH-dependent and -independent mechanisms. Our study therefore identified new regulatory events in bone remodeling under inflammatory conditions.
Collapse
Affiliation(s)
- Nathan Young
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Natallia Mikhalkevich
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Ying Yan
- Department of Orthopedics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Di Chen
- Department of Orthopedics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Wei-ping Zheng
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| |
Collapse
|
589
|
Ginaldi L, Di Benedetto MC, De Martinis M. Osteoporosis, inflammation and ageing. IMMUNITY & AGEING 2005; 2:14. [PMID: 16271143 PMCID: PMC1308846 DOI: 10.1186/1742-4933-2-14] [Citation(s) in RCA: 298] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/31/2005] [Accepted: 11/04/2005] [Indexed: 02/06/2023]
Abstract
Osteoporosis is a condition characterized by low bone mass and increased bone fragility, putting patients at risk of fractures, which are major causes of morbidity substantially in older people. Osteoporosis is currently attributed to various endocrine, metabolic and mechanical factors. However, emerging clinical and molecular evidence suggests that inflammation also exerts significant influence on bone turnover, inducing osteoporosis. Numerous proinflammatory cytokines have been implicated in the regulation of osteoblasts and osteoclasts, and a shift towards an activated immune profile has been hypothesized as important risk factor. Chronic inflammation and the immune system remodelling characteristic of ageing, as well as of other pathological conditions commonly associated with osteoporosis, may be determinant pathogenetic factors. The present article will review the current perspectives on the interaction between bone and immune system in the elderly, providing an interpretation of osteoporosis in the light of inflamm-ageing.
Collapse
Affiliation(s)
- Lia Ginaldi
- Department of Internal Medicine, University of L'Aquila, Italy
| | | | | |
Collapse
|
590
|
Rude RK, Gruber HE, Wei LY, Frausto A. Immunolocalization of RANKL is increased and OPG decreased during dietary magnesium deficiency in the rat. Nutr Metab (Lond) 2005; 2:24. [PMID: 16162295 PMCID: PMC1266035 DOI: 10.1186/1743-7075-2-24] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2005] [Accepted: 09/14/2005] [Indexed: 11/10/2022] Open
Abstract
Background Epidemiological studies have linked low dietary magnesium (Mg) to low bone mineral density and osteoporosis. Mg deficiency in animal models has demonstrated a reduction in bone mass and increase in skeletal fragility. One major mechanism appears to be an increase in osteoclast number and bone resorption. The final pathway of osteoclastogenesis involves three constituents of a cytokine system: receptor activator of nuclear factor kB ligand (RANKL); its receptor, receptor activator of nuclear factor kB (RANK); and its soluble decoy receptor, osteoprotegerin (OPG). The relative presence of RANKL and OPG dictates osteoclastogenesis. The objective of this study was to assess the presence of RANKL and OPG in rats on a low Mg diet. Methods RANKL and OPG were assessed by immunocytochemistry staining in the tibia for up to 6 months in control rats on regular Mg intake (0.5 g/kg) and experimental rats on reduction of dietary Mg (.04%, 25% and 50% of this Nutrient Requirement). Results At all dietary Mg intakes, alteration in the presence of immunocytochemical staining of RANKL and OPG was observed. In general, OPG was decreased and RANKL increased, reflecting an alteration in the RANKL/OPG ratio toward increased osteoclastogenesis. Conclusion We have, for the first time demonstrated that a reduction in dietary Mg in the rat alters the presence of RANKL and OPG and may explain the increase in osteoclast number and decrease in bone mass in this animal model. As some of these dietary intake reductions in terms of the RDA are present in a large segment of or population, Mg deficiency may be another risk factor for osteoporosis.
Collapse
Affiliation(s)
- Robert K Rude
- University of Southern California and the Orthopaedic Hospital, 1975 Zonal Ave., GNH 6602, Los Angeles, CA 90089-9317, USA
| | - Helen E Gruber
- Department of Orthopaedic Surgery, Carolinas Medical Center, P.O. Box 32861, Charlotte, NC 28203, USA
| | - Livia Y Wei
- University of Southern California and the Orthopaedic Hospital, 1975 Zonal Ave., GNH 6602, Los Angeles, CA 90089-9317, USA
| | - Angelica Frausto
- University of Southern California and the Orthopaedic Hospital, 1975 Zonal Ave., GNH 6602, Los Angeles, CA 90089-9317, USA
| |
Collapse
|
591
|
Bajayo A, Goshen I, Feldman S, Csernus V, Iverfeldt K, Shohami E, Yirmiya R, Bab I. Central IL-1 receptor signaling regulates bone growth and mass. Proc Natl Acad Sci U S A 2005; 102:12956-61. [PMID: 16126903 PMCID: PMC1200265 DOI: 10.1073/pnas.0502562102] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The proinflammatory cytokine IL-1, acting via the hypothalamic IL-1 receptor type 1 (IL-1RI), activates pathways known to suppress bone formation such as the hypothalamo pituitary-adrenocortical axis and the sympathetic nervous system. In addition, peripheral IL-1 has been implicated as a mediator of the bone loss induced by sex hormone depletion and TNF. Here, we report an unexpected low bone mass (LBM) phenotype, including impairment of bone growth, in IL-1RI-deficient mice (IL-1rKO mice). Targeted overexpression of human IL-1 receptor antagonist to the central nervous system using the murine glial fibrillary acidic protein promoter (IL-1raTG mice) resulted in a similar phenotype, implying that central IL-1RI silencing is the causative process in the LBM induction. Analysis of bone remodeling indicates that the process leading to the LBM in both IL-1rKO and IL-1raTG is characterized mainly by doubling the osteoclast number. Either genetic modification does not decrease testosterone or increase corticosterone serum levels, suggesting that systems other than the gonads and hypothalamo pituitary-adrenocortical axis mediate the central IL-1RI effect on bone. We further demonstrate that WT mice express mouse IL-1ra in bone but not in the hypothalamus. Because low levels of IL-1 are present in both tissues, it is suggested that skeletal IL-1 activity is normally suppressed, whereas central IL-1 produces a constant physiologic stimulation of IL-1RI signaling. Although the pathway connecting the central IL-1RI signaling to bone remodeling remains unknown, the outburst of osteoclastogenesis in its absence suggests that normally it controls bone growth and mass by tonically restraining bone resorption.
Collapse
Affiliation(s)
- Alon Bajayo
- Bone Laboratory, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | | | | | | | | | | | | | | |
Collapse
|
592
|
Cremers SCLM, Pillai G, Papapoulos SE. Pharmacokinetics/pharmacodynamics of bisphosphonates: use for optimisation of intermittent therapy for osteoporosis. Clin Pharmacokinet 2005; 44:551-70. [PMID: 15932344 DOI: 10.2165/00003088-200544060-00001] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Bisphosphonates suppress osteoclast-mediated bone resorption and are widely used in the management of osteoporosis. Daily oral administration of alendronic acid and risedronic acid have been shown to reduce the risk of vertebral and non-vertebral fractures. Once-weekly regimens with these bisphosphonates are pharmacologically equivalent to daily regimens. Regimens with treatment-free intervals longer than 1 week present an attractive therapeutic option as they may offer additional patient convenience and long-term adherence to treatment. However, until recently, such regimens, usually referred to as intermittent or cyclical, have not shown any convincing antifracture efficacy in clinical trials, probably because of the empirical manner in which the design of these regimens has been approached. Investigation of pharmacokinetics/pharmacodynamics of bisphosphonates may help in the design of effective intermittent dosage regimens. Bisphosphonates are poorly absorbed from the gastrointestinal tract and about 50% of the absorbed drug is taken up selectively by the skeleton, while the rest is excreted unaltered in urine. Bisphosphonates exert their action at the bone surface, where they are taken up by the osteoclasts during bone resorption. Therefore, when describing the pharmacokinetics of bisphosphonates in relation to the pharmacodynamics, the amount of bisphosphonate at the skeleton should be accounted for. Few of the reported clinical pharmacokinetic studies addressed this issue. This is partly due to the absence of study design elements to account for skeletal binding of the drugs. Pharmacokinetic studies have also been hampered by technical difficulties in determining the concentration of bisphosphonates in serum and urine. Moreover, most clinical pharmacokinetic (but also pharmacokinetic/pharmacodynamic) studies have primarily used noncompartmental analysis, leaving out the distinct advantages of modelling and simulation techniques. Clinically, the primary action of bisphosphonates can be assessed by the measurement of biochemical markers of bone resorption. Recent studies indicate that the pattern of these markers during bisphosphonate treatment may be predictive of antifracture efficacy; however, only limited data are available for the development of pharmacokinetic/pharmacodynamic models that are able to predict the response of these markers to different treatment regimens with bisphosphonates. Recently, pharmacokinetic/pharmacodynamic models for response to bisphosphonates have been described and, at present, some of them are being used in the design of bisphosphonate regimens with long drug-free intervals.
Collapse
Affiliation(s)
- Serge C L M Cremers
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, The Netherlands.
| | | | | |
Collapse
|
593
|
Karasik D, Demissie S, Cupples LA, Kiel DP. Disentangling the genetic determinants of human aging: biological age as an alternative to the use of survival measures. J Gerontol A Biol Sci Med Sci 2005; 60:574-87. [PMID: 15972604 PMCID: PMC1361266 DOI: 10.1093/gerona/60.5.574] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The choice of a phenotype is critical for the study of a complex genetically regulated process, such as aging. To date, most of the twin and family studies have focused on broad survival measures, primarily age at death or exceptional longevity. However, on the basis of recent studies of twins and families, biological age has also been shown to have a strong genetic component, with heritability estimates ranging from 27% to 57%. The aim of this review is twofold: first, to summarize growing consensus on reliable methods of biological age assessment, and second, to demonstrate validity of this phenotype for research in the genetics of aging in humans.
Collapse
Affiliation(s)
- David Karasik
- Hebrew Rehabilitation Center for Aged, Research and Training Institute, 1200 Centre Street, Boston, MA 02131, USA.
| | | | | | | |
Collapse
|
594
|
Abstract
Patterns of cell death have been divided into apoptosis, which is actively executed by specific proteases, the caspases, and accidental necrosis. However, there is now accumulating evidence indicating that cell death can occur in a programmed fashion but in complete absence and independent of caspase activation. Alternative models of programmed cell death (PCD) have therefore been proposed, including autophagy, paraptosis, mitotic catastrophe, and the descriptive model of apoptosis-like and necrosis-like PCD. Caspase-independent cell death pathways are important safeguard mechanisms to protect the organism against unwanted and potential harmful cells when caspase-mediated routes fail but can also be triggered in response to cytotoxic agents or other death stimuli. As in apoptosis, the mitochondrion can play a key role but also other organelles such as lysosomes and the endoplasmic reticulum have an important function in the release and activation of death factors such as cathepsins, calpains, and other proteases. Here we review the various models of PCD and their death pathways at molecular and organelle level and discuss the relevance of the growing knowledge of caspase-independent cell death pathways for cancer.
Collapse
Affiliation(s)
- Linda E Bröker
- Department of Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | | | | |
Collapse
|
595
|
Mentaverri R, Wattel A, Lemaire-Hurtel AS, Kamel S, Blesius A, Brazier M. [Partnership between academic research and industry to study a new anti-osteoporotic drug]. Med Sci (Paris) 2005; 21:663-8. [PMID: 15985213 DOI: 10.1051/medsci/2005216-7663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The activity of the osteoclast, the cell responsible for bone resorption, is subjected to different regulation factors. Amongst these, those issued from the matrix, particularly released minerals such as calcium, are determinants. We have shown that variations in calcium concentration in the medium regulates resorption activity and duration of the osteoclast lifespan. The development of a new therapeutic agent, strontium ranelate, has shown very interesting clinical effects reliant on the stimulation of bone formation activity by osteoblasts and modulation of bone resorption activity. From our knowledge regarding osteoclast physiology, in particular calcium signaling pathways, and the control of different osteoclast cellular models, a consequent collaboration was formed between our laboratory and Servier in order to elaborate on the effects of strontium ranelate on the osteoclast. In several years, this collaboration has been further enriched by other collaborators in order to better understand this mechanism. It has also been shown that strontium likely interacts with the calcium-sensing receptor and that the pathways of intracellular signaling pathways activated by calcium and strontium ranelate via this receptor are different. In fact, within the scope of this collaboration with Servier, exchanges with other academic laboratories were initiated and collaboration on numerous techniques became possible. Then, it has been possible to confirm the presence of the calcium-sensing receptor on the osteoclasts and to demonstrate its role in the molecular events associated with strontium ranelate's effects on the osteoclast.
Collapse
Affiliation(s)
- Romuald Mentaverri
- Unité de Recherche des mécanismes de la résorption osseuse, Faculté de Pharmacie, Amiens, France
| | | | | | | | | | | |
Collapse
|
596
|
Wu X, Ahn EY, McKenna MA, Yeo H, McDonald JM. Fas binding to calmodulin regulates apoptosis in osteoclasts. J Biol Chem 2005; 280:29964-70. [PMID: 15965236 PMCID: PMC1351015 DOI: 10.1074/jbc.m500710200] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Promotion of osteoclast apoptosis is one therapeutic approach to osteoporosis. Calmodulin, the major intracellular Ca(2+) receptor, modulates both osteoclastogenesis and bone resorption. The calmodulin antagonist, trifluoperazine, rescues bone loss in ovariectomized mice (Zhang, L., Feng, X., and McDonald, J. M. (2003) Endocrinology 144, 4536-4543). We show here that a 3-h treatment of mouse osteoclasts with either of the calmodulin antagonists, tamoxifen or trifluoperazine, induces osteoclast apoptosis dose-dependently. Tamoxifen, 10 microm, and trifluoperazine, 10 microm, induce 7.3 +/- 1.8-fold and 5.3 +/- 0.9-fold increases in osteoclast apoptosis, respectively. In Jurkat cells, calmodulin binds to Fas, the death receptor, and this binding is regulated during Fas-mediated apoptosis (Ahn, E. Y., Lim, S. T., Cook, W. J., and McDonald, J. M. (2004) J. Biol. Chem. 279, 5661-5666). In osteoclasts, calmodulin also binds Fas. When osteoclasts are treated with 10 microm trifluoperazine, the binding between Fas and calmodulin is dramatically decreased at 15 min and gradually recovers by 60 min. A point mutation of the Fas death domain in the Lpr(-cg) mouse renders Fas inactive. Using glutathione S-transferase fusion proteins, the human Fas cytoplasmic domain is shown to bind calmodulin, whereas a point mutation (V254N) comparable with the Lpr(-cg) mutation in mice has markedly reduced calmodulin binding. Osteoclasts derived from Lpr(-cg) mice have diminished calmodulin/Fas binding and are more sensitive to calmodulin antagonist-induced apoptosis than those from wild-type mice. Both tamoxifen- and trifluoperazine-induced apoptosis are increased 1.6 +/- 0.2-fold in Lpr(-cg)-derived osteoclasts compared with osteoclasts derived from wild-type mice. In summary, calmodulin antagonists induce apoptosis in osteoclasts by a mechanism involving interference with calmodulin binding to Fas. The effects of calmodulin/Fas binding on calmodulin antagonist-induced apoptosis may open a new avenue for therapy for osteoporosis.
Collapse
Affiliation(s)
- Xiaojun Wu
- From the Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama 35294 and the
| | - Eun-Young Ahn
- From the Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama 35294 and the
| | - Margaret A. McKenna
- From the Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama 35294 and the
| | - Hyeonju Yeo
- From the Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama 35294 and the
| | - Jay M. McDonald
- From the Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama 35294 and the
- Veterans Affairs Medical Center, Birmingham, Alabama 35233
- ¶ To whom correspondence should be addressed: University of Alabama at Birmingham, 509 LHRB, 1530 3rd Ave. S., Birmingham, AL 35294-0007. Tel.: 205-934-6666; Fax: 205-975-9927; E-mail:
| |
Collapse
|
597
|
Abstract
During growth, estrogen deficiency in females may produce increased bone size as a result of removal of inhibition of periosteal apposition, while failed endosteal apposition produces thin cortices and trabeculae in the smaller bone. In males, androgen deficiency produces reduced periosteal and endosteal apposition, reduced bone size, and cortical and trabecular thickness. At completion of longitudinal growth, advancing age is associated with emergence of a negative bone balance in each basic multicellular unit (BMU) because of reduced bone formation. Bone loss occurs, but slowly because the remodeling rate is slow. In midlife, in females, estrogen deficiency increases remodeling rate, increases the volume of bone resorbed, and decreases the volume of bone formed in each of the numerous BMUs remodeling bone on its endosteal (endocortical, trabecular, intracortical) surfaces so bone loss accelerates. In males, remodeling rate remains slow and is driven largely by reduced bone formation in the BMU. Hypogonadism in 20% to 30% of elderly men contributes to bone loss. In both sexes, calcium malabsorption and secondary hyperparathyroidism may partly be sex-hormone dependent and contributes to cortical bone loss. Concurrent periosteal apposition partly offsets endosteal bone loss, but less so in women than in men. More women than men fracture because their smaller skeleton incurs greater architectural damage and adapts less by periosteal apposition. Sex hormone deficiency during growth and aging is pivotal in the pathogenesis of bone fragility.
Collapse
Affiliation(s)
- Ego Seeman
- Department of Endocrinology, Austin Hospital, Heidelberg 3084, Melbourne, Australia.
| |
Collapse
|
598
|
Conway SP, Oldroyd B, Morton A, Truscott JG, Peckham DG. Effect of oral bisphosphonates on bone mineral density and body composition in adult patients with cystic fibrosis: a pilot study. Thorax 2004; 59:699-703. [PMID: 15282392 PMCID: PMC1747108 DOI: 10.1136/thx.2002.002568] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND Approximately two thirds of adult patients with cystic fibrosis have reduced bone mineral density and up to one quarter have osteoporosis at one or more sites. Any bone mineral deficits are likely to be exacerbated in patients following lung transplantation by their immunosuppressive regimen. Vertebral collapse and rib fractures will impair the ability to cough and the efficacy of physiotherapy treatments. METHODS Patients attending the Leeds Regional Adult Cystic Fibrosis Unit with either osteopenia or osteoporosis on dual energy x ray absorptiometry (DXA) scanning were offered treatment with oral bisphosphonates after exclusion of abnormal vitamin D, calcium, or phosphate levels, abnormal thyroid function, or hypogonadism. Those declining treatment or patients with a normal initial DXA scan formed the control group. A second DXA scan was performed after a mean of 2.4 years in the treatment group and 2.9 years in the non-treatment group. Patients in the active group were asked to complete a short questionnaire detailing their adherence to treatment. RESULTS The medians of the differences in annual changes in bone parameters between treatment and control groups showed significant differences in bone mineralisation in favour of the treatment group at the lumbar spine (L2-L4), the femoral neck, and for total body measurements. There were no significant differences in weight, height, or body composition in either patient group. Most treated patients stated that they adhered to treatment most of the time. CONCLUSION Treatment with oral bisphosphonates may improve bone mineralisation in adult patients with cystic fibrosis. The results of this pilot study need to be further explored in a randomised controlled trial.
Collapse
Affiliation(s)
- S P Conway
- Regional Cystic Fibrosis Unit, Seacroft Hospital, Leeds LS14 6UH, UK.
| | | | | | | | | |
Collapse
|
599
|
Wang MWH, Wei S, Faccio R, Takeshita S, Tebas P, Powderly WG, Teitelbaum SL, Ross FP. The HIV protease inhibitor ritonavir blocks osteoclastogenesis and function by impairing RANKL-induced signaling. J Clin Invest 2004; 114:206-13. [PMID: 15254587 PMCID: PMC449740 DOI: 10.1172/jci15797] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2002] [Accepted: 05/28/2004] [Indexed: 12/27/2022] Open
Abstract
Highly active antiretroviral therapy (HAART), which includes HIV protease inhibitors (PIs), has been associated with bone demineralization. To determine if this complication reflects accelerated resorptive activity, we studied the impact of two common HIV PIs, ritonavir and indinavir, on osteoclast formation and function. Surprisingly, we find that ritonavir, but not indinavir, inhibits osteoclast differentiation in a reversible manner and also abrogates bone resorption by disrupting the osteoclast cytoskeleton, without affecting cell number. Ritonavir given in vivo completely blunts parathyroid hormone-induced osteoclastogenesis in mice, which confirms that the drug is bone sparing. In keeping with its antiresorptive properties, ritonavir impairs receptor activator of nuclear factor kappaB ligand-induced (RANKL-induced) activation of NF-kappaB and Akt signaling pathways, both critical to osteoclast formation and function. In particular, ritonavir is found to inhibit RANKL-induced Akt signaling by disrupting the recruitment of TNF receptor-associated factor 6/c-Src complex to lipid rafts. Thus, ritonavir may represent a bone-sparing PI capable of preventing development of osteopenia in patients currently on HAART.
Collapse
Affiliation(s)
- Michael W-H Wang
- Department of Pathology and Immunology, Washington University School of Medicine, Campus Box 8118, 660 South Euclid Avenue, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
600
|
Karasik D, Hannan MT, Cupples LA, Felson DT, Kiel DP. Genetic contribution to biological aging: the Framingham Study. J Gerontol A Biol Sci Med Sci 2004; 59:218-26. [PMID: 15031305 PMCID: PMC1201381 DOI: 10.1093/gerona/59.3.b218] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
This study assessed the contribution of genetic and nongenetic factors to biological aging in American Caucasians. The studied sample included 1402 members of 288 pedigrees from the Framingham Heart Study. The original cohort participants received hand radiography in 1967-1969 (mean age, 58.7 years) and their offspring in 1992-1993 (mean age, 51.6 years). An osseographic score was applied to hand radiographs. Standardized residuals between Osseographic Scoring System-predicted age and actual age were used as a measure of biological age (BA). In variance component genetic analysis, sex, cohort, height, body mass index, and, in women, menopausal status and estrogen use, jointly explained approximately 6% of the total variance of BA. Genetic factors explained an additional 57%. Linkage analysis of covariate-adjusted BA suggested the presence of quantitative trait loci on chromosomes 3p, 7q, 11p, 16q, and 21q. In conclusion, the variation in BA measured by radiography was strongly governed by genetic factors in a sample of American adults.
Collapse
Affiliation(s)
- David Karasik
- Hebrew Rehabilitation Center for Aged, and Harvard Medical School, Boston, MA 02131, USA.
| | | | | | | | | |
Collapse
|