101
|
Receptor activator of nuclear factor-κB ligand (RANKL) and its relationship to coronary atherosclerosis in HIV patients. J Acquir Immune Defic Syndr 2013; 61:359-63. [PMID: 22842843 DOI: 10.1097/qai.0b013e31826a6c16] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
HIV-infected individuals have an increased prevalence of coronary artery disease. Receptor activator of nuclear factor-κB ligand (RANKL) and osteoprotegerin have been postulated as mediators of vascular calcification. 78 HIV-infected men and 32 healthy controls without history of coronary artery disease were prospectively recruited to undergo cardiac computed tomography and computed tomography angiography to assess coronary artery calcium and plaque burden. Soluble receptor activator of nuclear factor-κB ligand was lower in HIV-infected individuals than controls [2.52 (1.08-3.98) vs. 3.33 (2.44-4.64) pg/mL, P = 0.01, median (IQR) respectively]. Soluble receptor activator of nuclear factor-κB ligand was negatively associated with the number of coronary segments with plaque (Spearman ρ = -0.41, P < 0.001) and Agatston calcium score (ρ = -0.30, P < 0.01) in HIV-infected individuals even after adjusting for traditional cardiovascular risk factors.
Collapse
|
102
|
Carulli C, Innocenti M, Brandi ML. Bone vascularization in normal and disease conditions. Front Endocrinol (Lausanne) 2013; 4:106. [PMID: 23986744 PMCID: PMC3752619 DOI: 10.3389/fendo.2013.00106] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 08/06/2013] [Indexed: 01/14/2023] Open
Abstract
Bone vasculature is essential for many processes, such as skeletal development and growth, bone modeling and remodeling, and healing processes. Endothelium is an integral part of bone tissue, expressing a physiological paracrine function via growth factors and chemokines release, and interacting with several cellular lines. Alterations of the complex biochemical interactions between vasculature and bone cells may lead to various clinical manifestations. Two different types of pathologies result: a defect or an excess of bone vasculature or endothelium metabolism. Starting from the molecular basis of the interactions between endothelial and bone cells, the Authors present an overview of the recent acquisitions in the physiopathology of the most important clinical patterns, and the modern therapeutic strategies for their treatments.
Collapse
Affiliation(s)
- Christian Carulli
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Massimo Innocenti
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Maria Luisa Brandi
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
- *Correspondence: Maria Luisa Brandi, Department of Surgery and Translational Medicine, University of Florence, Viale Pieraccini, 650139 Florence, Italy e-mail:
| |
Collapse
|
103
|
Clarkin CE, Gerstenfeld LC. VEGF and bone cell signalling: an essential vessel for communication? Cell Biochem Funct 2012; 31:1-11. [PMID: 23129289 DOI: 10.1002/cbf.2911] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 08/31/2012] [Accepted: 09/10/2012] [Indexed: 01/17/2023]
Abstract
Vascular endothelial growth factor (VEGF) is an endothelial cell survival factor and is required for effective coupling of angiogenesis and osteogenesis. Although central to bone homeostasis, repair and the pathobiology that affect these processes, the precise mechanisms coupling endothelial cell function within bone formation and remodelling remain unclarified. This review will (i) focus on the potential directionality of VEGF signalling in adult bone by identifying the predominant source of VEGF within the bone microenvironment, (ii) will summarize current VEGF receptor expression studies by bone cells and (iii) will provide evidence for a role for VEGF signalling during postnatal repair and osteoporosis. A means of understanding the directionality of VEGF signalling in adult bone would allow us to most effectively target angiogenic pathways in diseases characterized by changes in bone remodelling rates and enhance bone repair when compromised.
Collapse
Affiliation(s)
- Claire E Clarkin
- Centre for Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK.
| | | |
Collapse
|
104
|
Elevated serum osteoprotegerin levels predict in-hospital major adverse cardiac events in patients with ST elevation myocardial infarction. J Cardiol 2012; 60:355-60. [PMID: 22727437 DOI: 10.1016/j.jjcc.2012.05.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 04/23/2012] [Accepted: 05/12/2012] [Indexed: 11/20/2022]
|
105
|
Davenport C, Kenny H, Ashley DT, O'Sullivan EP, Smith D, O'Gorman DJ. The effect of exercise on osteoprotegerin and TNF-related apoptosis-inducing ligand in obese patients. Eur J Clin Invest 2012; 42:1173-9. [PMID: 22803952 DOI: 10.1111/j.1365-2362.2012.02703.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Biomarkers of cardiovascular (CV) risk are tests that predict a patient's risk of future CV events. Recently, two proteins involved in vascular calcification; serum levels of osteoprotegerin (OPG) and tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) have emerged as potentially useful biomarkers. OPG levels are positively correlated with CV risk, whereas TRAIL levels show a negative correlation. Exercise training is known to reduce risk factors for CV disease by improving metabolism, vascular biology and blood flow. This study examined the effects of a 6-month exercise training programme on levels of OPG and TRAIL. Pulse wave velocity (PWV) and high-sensitivity C-reactive protein (hsCRP) were measured for comparative purposes. MATERIALS AND METHODS Overweight and obese patients undertook a 6-month exercise programme. Patients participated in 4 h of primarily aerobic exercise per week of which 2 h were supervised. At the beginning and end of the programme, anthropometric measurements, PWV and serum levels of OPG, TRAIL and hsCRP were measured. RESULTS A total of 21 patients (17 men) aged 55.2 ± 10 years completed the programme. Mean body mass index decreased from 34.1 ± 5.8 to 32.6 ± 5.4 kg/m(2) (P<0.05), while waist circumference decreased from 111.8 ± 12.4 to 109.6 ± 12.8 cm (P<0.05). PWV decreased from 9.2 to 8.5 m/s (P<0.02). OPG, TRAIL and hsCRP levels did not change significantly. CONCLUSIONS Exercise training reduced PWV but not OPG, TRAIL or hsCRP in this population. These data suggest that while an intervention of this nature improves vascular tone, it does not exert significant effects on serum biomarkers related to atherosclerotic inflammation and calcification.
Collapse
Affiliation(s)
- Colin Davenport
- Department of Diabetes and Endocrinology, Beaumont Hospital, Dublin, Ireland Centre for Preventive Medicine, School of Health and Human Performance, Dublin City University, Dublin, Ireland.
| | | | | | | | | | | |
Collapse
|
106
|
Thadani U. Osteoprotegerin and ST segment myocardial infarction: a novel risk marker but not for routine use at present. Cardiology 2012; 123:91-3. [PMID: 23018624 DOI: 10.1159/000342799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 08/21/2012] [Indexed: 11/19/2022]
|
107
|
Abstract
Bone never forms without vascular interactions. This simple statement of fact does not adequately reflect the physiological and pharmacological implications of the relationship. The vasculature is the conduit for nutrient exchange between bone and the rest of the body. The vasculature provides the sustentacular niche for development of osteoblast progenitors and is the conduit for egress of bone marrow cell products arising, in turn, from the osteoblast-dependent haematopoietic niche. Importantly, the second most calcified structure in humans after the skeleton is the vasculature. Once considered a passive process of dead and dying cells, vascular calcification has emerged as an actively regulated form of tissue biomineralization. Skeletal morphogens and osteochondrogenic transcription factors are expressed by cells within the vessel wall, which regulates the deposition of vascular calcium. Osteotropic hormones, including parathyroid hormone, regulate both vascular and skeletal mineralization. Cellular, endocrine and metabolic signals that flow bidirectionally between the vasculature and bone are necessary for both bone health and vascular health. Dysmetabolic states including diabetes mellitus, uraemia and hyperlipidaemia perturb the bone-vascular axis, giving rise to devastating vascular and skeletal disease. A detailed understanding of bone-vascular interactions is necessary to address the unmet clinical needs of an increasingly aged and dysmetabolic population.
Collapse
Affiliation(s)
- Bithika Thompson
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, Campus Box 8127, 660 South Euclid Avenue, St Louis, MO 63110, USA
| | | |
Collapse
|
108
|
Deuell KA, Callegari A, Giachelli CM, Rosenfeld ME, Scatena M. RANKL enhances macrophage paracrine pro-calcific activity in high phosphate-treated smooth muscle cells: dependence on IL-6 and TNF-α. J Vasc Res 2012; 49:510-21. [PMID: 22948607 DOI: 10.1159/000341216] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Accepted: 06/10/2012] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Vascular calcification is highly correlated with cardiovascular disease (CVD) morbidity and mortality, and it is associated with inflammation. Receptor activator of NF-ĸB ligand (RANKL) inhibition in vivo has been shown to reduce vascular calcification in a mouse model of atherosclerosis. Therefore, we tested the hypothesis that RANKL regulates smooth muscle cell (SMC) calcification by modulating macrophage production of pro-calcific cytokines. METHODS We used a bone marrow-derived macrophage (BMDM)/SMC co-culture system and examined the effects of RANKL on BMDM activation and SMC matrix calcification. RESULTS Treatment with RANKL alone did not stimulate SMC calcification induced by elevated phosphate. BMDMs differentiated with macrophage colony-stimulating factor and placed in co-culture with SMCs increased phosphate-induced SMC calcification. RANKL added to the BMDM/SMC co-cultures further enhanced SMC calcification. Treatment of BMDMs with RANKL resulted in increased expression of IL-6 and TNF-α. Thus, increased expression of these pro-calcific cytokines in macrophages may mediate RANKL-induced SMC calcification in a paracrine fashion. Addition of neutralizing IL-6 and TNF-α antibodies together with RANKL treatment significantly reduced the RANKL induction of SMC calcification. CONCLUSION RANKL activation of pro-inflammatory and pro-calcific pathways in macrophages may contribute to vascular calcification and inflammation.
Collapse
Affiliation(s)
- Kate Ann Deuell
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | |
Collapse
|
109
|
Abstract
Remodeling, a continuous physiological process maintains the strength of the bones, which maintains a delicate balance between bone formation and resorption process. This review gives an insight to the complex interaction and correlation between the bone remodeling and the corresponding changes in host immunological environment and also summarises the most recent developments occuring in the understanding of this complex field. T cells, both directly and indirectly increase the expression of receptor activator of nuclear factor kB ligand (RANKL); a vital step in the activation of osteoclasts, thus positively regulates the osteoclastogenesis. Though various cytokines, chemikines, transcription factors and co-stimulatory molecules are shared by both skeletal and immune systems, but researches are being conducted to establish and analyse their role and / or control on this complex but vital process. The understanding of this part of research may open new horizons in the management of inflammatory and autoimmune diseases, resulting into bone loss and that of osteoporosis also.
Collapse
Affiliation(s)
- Ajai Singh
- Department of Orthopaedics, Co Trauma Center I/C, C S M Medical University, Lucknow, India
| | | | | | | |
Collapse
|
110
|
Serum Levels of IL-6, IL-10, IL-12, IL-17 and IFN-γ and Their Association with Markers of Bone Metabolism in Vitamin D-Deficient Female Students. Inflammation 2012; 36:164-8. [DOI: 10.1007/s10753-012-9531-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
111
|
Riegel A, Maurer T, Prior B, Stegmaier S, Heppert V, Wagner C, Hänsch GM. Human polymorphonuclear neutrophils express RANK and are activated by its ligand, RANKL. Eur J Immunol 2012; 42:975-81. [PMID: 22531921 DOI: 10.1002/eji.201141786] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The receptor activator of NF-κB (RANK) is especially well studied in the context of bone remodeling, and RANK and its ligand, RANKL, are key molecules in the induction of bone resorbing osteoclasts. We now report that polymorphonuclear neutrophils (PMNs) contain preformed RANK, stored in secretory vesicles and in specific granules. Upon stimulation of PMNs in vitro, RANK was translocated to the cell membrane. In patients with persistent bacterial infections, RANK surface expression was enhanced compared with that of healthy individuals. The functional activity of RANK was assessed by determining migration of PMNs toward RANKL. A time- and dose-dependent migration was seen, leading to the conclusion that RANK on PMNs is functional. We presume that regulated RANK expression contributes to the fine tuning of PMN migration, for example, on and through inflamed endothelium that is known to express RANKL.
Collapse
|
112
|
Hosbond SE, Poulsen TS, Diederichsen ACP, Nybo M, Rasmussen LM, Mickley H. Osteoprotegerin as a marker of atherosclerosis: a systematic update. SCAND CARDIOVASC J 2012; 46:203-11. [PMID: 22506827 DOI: 10.3109/14017431.2012.685491] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Osteoprotegerin (OPG) may be involved in development of atherosclerosis. To evaluate plasma concentrations of OPG in individuals with stable coronary artery disease (CAD), acute coronary syndrome (ACS), peripheral artery disease (PAD), and cerebrovascular disease (CBVD) a systematic literature review was performed. DESIGN AND METHODS Studies investigating OPG concentrations in stable CAD, ACS, PAD, and CBVD were extracted from PubMed and the Cochrane Library, retrieving 280 articles. Nonrelevant articles were excluded and after thorough evaluation, and only 14 studies with clearly defined cohorts qualified for this review. RESULTS In 11 studies, OPG concentrations were elevated. Severity of atherosclerosis was significantly associated with higher OPG concentrations compared to healthy controls. No association between PAD and OPG concentrations was observed. CONCLUSION OPG concentrations are associated with the presence and severity of stable CAD, ACS, and CBVD. Larger studies are needed to reach conclusions concerning OPG concentrations in PAD. Studies addressing a putative role for OPG in suspected CAD and CBVD are warranted.
Collapse
|
113
|
Morena M, Jaussent I, Halkovich A, Dupuy AM, Bargnoux AS, Chenine L, Leray-Moragues H, Klouche K, Vernhet H, Canaud B, Cristol JP. Bone biomarkers help grading severity of coronary calcifications in non dialysis chronic kidney disease patients. PLoS One 2012; 7:e36175. [PMID: 22567137 PMCID: PMC3342257 DOI: 10.1371/journal.pone.0036175] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 03/27/2012] [Indexed: 02/06/2023] Open
Abstract
Background Osteoprotegerin (OPG) and fibroblast growth factor-23 (FGF23) are recognized as strong risk factors of vascular calcifications in non dialysis chronic kidney disease (ND-CKD) patients. The aim of this study was to investigate the relationships between FGF23, OPG, and coronary artery calcifications (CAC) in this population and to attempt identification of the most powerful biomarker of CAC: FGF23? OPG? Methodology/Principal Findings 195 ND-CKD patients (112 males/83 females, 70.8 [27.4–94.6] years) were enrolled in this cross-sectional study. All underwent chest multidetector computed tomography for CAC scoring. Vascular risk markers including FGF23 and OPG were measured. Logistic regression analyses were used to study the potential relationships between CAC and these markers. The fully adjusted-univariate analysis clearly showed high OPG (≥10.71 pmol/L) as the only variable significantly associated with moderate CAC ([100–400[) (OR = 2.73 [1.03;7.26]; p = 0.04). Such association failed to persist for CAC scoring higher than 400. Indeed, severe CAC was only associated with high phosphate fractional excretion (FEPO4) (≥38.71%) (OR = 5.47 [1.76;17.0]; p = 0.003) and high FGF23 (≥173.30 RU/mL) (OR = 5.40 [1.91;15.3]; p = 0.002). In addition, the risk to present severe CAC when FGF23 level was high was not significantly different when OPG was normal or high. Conversely, the risk to present moderate CAC when OPG level was high was not significantly different when FGF23 was normal or high. Conclusions Our results strongly suggest that OPG is associated to moderate CAC while FGF23 rather represents a biomarker of severe CAC in ND-CKD patients.
Collapse
Affiliation(s)
- Marion Morena
- Laboratoire de Biochimie, CHRU de Montpellier, Montpellier, France
- Institut de Recherche et de Formation en Dialyse, Montpellier, France
- UMR 204, Nutripass, Université Montpellier I, Montpellier, France
| | | | - Aurore Halkovich
- Laboratoire de Biochimie, CHRU de Montpellier, Montpellier, France
| | - Anne-Marie Dupuy
- Laboratoire de Biochimie, CHRU de Montpellier, Montpellier, France
| | - Anne-Sophie Bargnoux
- Laboratoire de Biochimie, CHRU de Montpellier, Montpellier, France
- UMR 204, Nutripass, Université Montpellier I, Montpellier, France
| | - Leila Chenine
- Service de Néphrologie-Hémodialyse et Soins Intensifs, CHRU de Montpellier, Montpellier, France
| | - Hélène Leray-Moragues
- Service de Néphrologie-Hémodialyse et Soins Intensifs, CHRU de Montpellier, Montpellier, France
| | - Kada Klouche
- UMR 204, Nutripass, Université Montpellier I, Montpellier, France
- Service de Réanimation Métabolique, CHRU de Montpellier, Montpellier, France
| | - Hélène Vernhet
- Service de Radiologie, CHRU de Montpellier, Montpellier, France
| | - Bernard Canaud
- Institut de Recherche et de Formation en Dialyse, Montpellier, France
- UMR 204, Nutripass, Université Montpellier I, Montpellier, France
- Service de Néphrologie-Hémodialyse et Soins Intensifs, CHRU de Montpellier, Montpellier, France
| | - Jean-Paul Cristol
- Laboratoire de Biochimie, CHRU de Montpellier, Montpellier, France
- UMR 204, Nutripass, Université Montpellier I, Montpellier, France
- * E-mail:
| |
Collapse
|
114
|
Glisic S, Jailwala P. Interaction between Treg apoptosis pathways, Treg function and HLA risk evolves during type 1 diabetes pathogenesis. PLoS One 2012; 7:e36040. [PMID: 22563437 PMCID: PMC3338571 DOI: 10.1371/journal.pone.0036040] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 03/29/2012] [Indexed: 11/23/2022] Open
Abstract
We have previously reported increased apoptosis of regulatory T cells (Tregs) in recent-onset Type 1 Diabetes subjects (RO T1D) in the honeymoon phase and in multiple autoantibody-positive (Ab+) subjects, some of which are developing T1D. We have also reported that increased Treg apoptosis was associated with High HLA risk and that it subsided with cessation of honeymoon period. In this report, we present results generated using genetics, genomics, functional cell-based assays and flow cytometry to assess cellular changes at the T-cell level during T1D pathogenesis. We measured ex vivo Treg apoptosis and Treg function, surface markers expression, expression of HLA class II genes, the influence of HLA risk on Treg apoptosis and function, and evaluated contribution of genes reported to be involved in the apoptosis process. This integrated comprehensive approach uncovered important information that can serve as a basis for future studies aimed to modulate Treg cell responsiveness to apoptotic signals in autoimmunity. For example, T1D will progress in those subjects where increased Treg apoptosis is accompanied with decreased Treg function. Furthermore, Tregs from High HLA risk healthy controls had increased Treg apoptosis levels and overexpressed FADD but not Fas/FasL. Tregs from RO T1D subjects in the honeymoon phase were primarily dying through withdrawal of growth hormones with contribution of oxidative stress, mitochondrial apoptotic pathways, and employment of TNF-receptor family members. Ab+ subjects, however, expressed high inflammation level, which probably contributed to Treg apoptosis, although other apoptotic pathways were also activated: withdrawal of growth hormones, oxidative stress, mitochondrial apoptosis and Fas/FasL apoptotic pathways. The value of these results lie in potentially different preventive treatment subjects would receive depending on disease progression stage when treated.
Collapse
Affiliation(s)
- Sanja Glisic
- Department of Pediatrics, Max McGee National Research Center for Juvenile Diabetes, Medical College of Wisconsin, Children's Hospital of Wisconsin, Milwaukee, Wisconsin, United States of America.
| | | |
Collapse
|
115
|
Kular J, Tickner J, Chim SM, Xu J. An overview of the regulation of bone remodelling at the cellular level. Clin Biochem 2012; 45:863-73. [PMID: 22465238 DOI: 10.1016/j.clinbiochem.2012.03.021] [Citation(s) in RCA: 378] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 03/07/2012] [Accepted: 03/13/2012] [Indexed: 01/11/2023]
Abstract
OBJECTIVES To review the current literature on the regulation of bone remodelling at the cellular level. DESIGN AND METHODS The cellular activities of the cells in the basic multicellular unit (BMU) were evaluated. RESULTS Bone remodelling requires an intimate cross-talk between osteoclasts and osteoblasts and is tightly coordinated by regulatory proteins that interact through complex autocrine/paracrine mechanisms. Osteocytes, bone lining cells, osteomacs, and vascular endothelial cells also regulate bone remodelling in the BMU via cell signalling networks of ligand-receptor complexes. In addition, through secreted and membrane-bound factors in the bone microenvironment, T and B lymphocytes mediate bone homeostasis in osteoimmunology. CONCLUSIONS Osteoporosis and other bone diseases occur because multicellular communication within the BMU is disrupted. Understanding the cellular and molecular basis of bone remodelling and the discovery of novel paracrine or coupling factors, such as RANKL, sclerostin, EGFL6 and semaphorin 4D, will lay the foundation for drug development against bone diseases.
Collapse
Affiliation(s)
- Jasreen Kular
- School of Pathology and Laboratory Medicine, The University of Western Australia, Western Australia, Australia
| | | | | | | |
Collapse
|
116
|
Identifying coronary artery disease in men with type 2 diabetes: osteoprotegerin, pulse wave velocity, and other biomarkers of cardiovascular risk. J Hypertens 2012; 29:2469-75. [PMID: 21970938 DOI: 10.1097/hjh.0b013e32834c1e95] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVES In patients with type 2 diabetes, high serum levels of osteoprotegerin (OPG) have been associated with a greater risk of cardiovascular events. However, it remains unclear how well OPG performs when compared with traditional biomarkers of cardiovascular risk such as high-sensitivity C-reactive protein (hsCRP). Furthermore, OPG levels are also high in the presence of diabetes-related microvascular disease, and it is unclear whether OPG can distinguish microvascular disease from large-vessel atherosclerosis. The first aim of this study was to compare OPG levels against other biomarkers of cardiovascular risk in the identification of patients with documented multivessel coronary artery disease (CAD). The second aim was to compare OPG levels in patients with microvascular complications (microalbuminuria) against those with established CAD. METHODS Three groups of male patients with type 2 diabetes were recruited: patients without microvascular complications or large-vessel atherosclerosis (n = 24), patients with microalbuminuria only (n = 23), and patients with microalbuminuria and documented multivessel CAD (n = 25). OPG, hsCRP, interleukin 6, urate, and pulse wave velocity were measured. RESULTS Serum OPG levels were significantly higher in patients with a combination of microalbuminuria and CAD than in those with microalbuminuria alone. There were no significant differences in any of the other biomarkers between the groups. CONCLUSION OPG was found to be superior to the other biomarkers studied in identifying patients with documented CAD. The presence of CAD was a greater determinant of serum OPG levels than microalbuminuria in our population. These findings support the use of OPG as a biomarker of cardiovascular risk.
Collapse
|
117
|
The Anti-Inflammatory, Phytoestrogenic, and Antioxidative Role of Labisia pumila in Prevention of Postmenopausal Osteoporosis. Adv Pharmacol Sci 2012; 2012:706905. [PMID: 22611381 PMCID: PMC3353141 DOI: 10.1155/2012/706905] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 01/08/2012] [Indexed: 11/17/2022] Open
Abstract
Osteoporosis is characterized by skeletal degeneration with low bone mass and destruction of microarchitecture of bone tissue which is attributed to various factors including inflammation. Women are more likely to develop osteoporosis than men due to reduction in estrogen during menopause which leads to decline in bone-formation and increase in bone-resorption activity. Estrogen is able to suppress production of proinflammatory cytokines such as IL-1, IL-6, IL-7, and TNF-α. This is why these cytokines are elevated in postmenopausal women. Studies have shown that estrogen reduction is able to stimulate focal inflammation in bone. Labisia pumila (LP) which is known to exert phytoestrogenic effect can be used as an alternative to ERT which can produce positive effects on bone without causing side effects. LP contains antioxidant as well as exerting anti-inflammatory effect which can act as free radical scavenger, thus inhibiting TNF-α production and COX-2 expression which leads to decline in RANKL expression, resulting in reduction in osteoclast activity which consequently reduces bone loss. Hence, it is the phytoestrogenic, anti-inflammatory, and antioxidative properties that make LP an effective agent against osteoporosis.
Collapse
|
118
|
Sakai N, Van Sweringen HL, Schuster R, Blanchard J, Burns JM, Tevar AD, Edwards MJ, Lentsch AB. Receptor activator of nuclear factor-κB ligand (RANKL) protects against hepatic ischemia/reperfusion injury in mice. Hepatology 2012; 55:888-97. [PMID: 22031462 PMCID: PMC3276725 DOI: 10.1002/hep.24756] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 10/04/2011] [Indexed: 12/16/2022]
Abstract
UNLABELLED The transcription factor nuclear factor kappaB (NF-κB) plays diverse roles in the acute injury response to hepatic ischemia/reperfusion (I/R). Activation of NF-κB in Kupffer cells promotes inflammation through cytokine expression, whereas activation in hepatocytes may be cell protective. The interaction of receptor activator of NF-κB (RANK) and its ligand (RANKL) promotes NF-κB activation; however, this ligand-receptor system has not been studied in acute liver injury. In the current study, we sought to determine if RANK and RANKL were important in the hepatic response to I/R. Mice were subjected to partial hepatic ischemia followed by reperfusion. In some experiments, mice received recombinant RANKL or neutralizing antibodies to RANKL 1 hour prior to surgery or at reperfusion to assess the role of RANK/RANKL signaling during I/R injury. RANK was constitutively expressed in the liver and was not altered by I/R. RANK was strongly expressed in hepatocytes and very weakly expressed in Kupffer cells. Serum RANKL concentrations increased after I/R and peaked 4 hours after reperfusion. Serum levels of osteoprotegerin (OPG), a decoy receptor for RANKL, steadily increased over the 8-hour period of reperfusion. Treatment with RANKL, before ischemia or at reperfusion, increased hepatocyte NF-κB activation and significantly reduced liver injury. These beneficial effects occurred without any effect on cytokine expression or liver inflammation. Treatment with anti-RANKL antibodies had no effect on liver I/R injury. CONCLUSION During the course of injury, endogenous OPG appears to suppress the effects of RANKL. However, exogenous administration of RANKL, given either prophylactically or postinjury, reduces liver injury in a manner associated with increased hepatocyte NF-κB activation. The data suggest that RANK/RANKL may be a viable therapeutic target in acute liver injury.
Collapse
|
119
|
Combination gene therapy targeting on interleukin-1β and RANKL for wear debris-induced aseptic loosening. Gene Ther 2012; 20:128-35. [PMID: 22318091 PMCID: PMC3349796 DOI: 10.1038/gt.2012.1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
This study investigated the efficacy of a combination gene therapy to repress interleukin-1 (IL-1) and receptor activator of nuclear factor NF-kappa B ligand (RANKL) for the treatment of particulate debris-induced aseptic loosening, and tried to explore the molecular mechanism of the exogenous gene modifications on osteoclastogenesis. RAW cells activated by titanium particles were transduced with DFG-IL-1Ra (retroviral vector encoding IL-1 receptor antagonist) and AAV-OPG (adeno-associated viral vectors-osteoprotegerin) individually or in combination for 4 weeks. Pro-inflammatory cytokines in culture media were determined by enzyme-linked immunosorbent assay, and gene expressions of RANK, IL-1β, c-Fos, TRAF6, JNK1 and CPK were examined using real-time PCR. An established knee-implant-failure mouse model was employed to evaluate the efficacy of the in vivo double-gene therapy. The surgical implantation of a titanium alloy pin into the proximal tibia was followed by monthly challenge with titanium debris. Peri-implant gene transfers of IL-1Ra and OPG (respectively or in combination) were given 3 weeks after surgery. The combination of OPG and IL-1Ra gene transfer exhibited strong synergetic effects in blockage of inflammation and osteoclastogenesis at 8 weeks after gene modification. The combination therapy reversed peri-implant bone resorption and restored implant stability when compared with either single gene transduction. Real-time PCR data indicated that the action of IL-1Ra gene therapy may be mediated via the JNK1 pathway, while the reduction of osteoclastogenesis by OPG gene modification may be regulated by c-Fos expression. In addition, both gene modifications resulted in significant diminishment of TRAF6 expression.
Collapse
|
120
|
MAPK usage in periodontal disease progression. JOURNAL OF SIGNAL TRANSDUCTION 2012; 2012:308943. [PMID: 22315682 PMCID: PMC3270463 DOI: 10.1155/2012/308943] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Accepted: 10/05/2011] [Indexed: 12/12/2022]
Abstract
In periodontal disease, host recognition of bacterial constituents, including lipopolysaccharide (LPS), induces p38 MAPK activation and subsequent inflammatory cytokine expression, favoring osteoclastogenesis and increased net bone resorption in the local periodontal environment. In this paper, we discuss evidence that the p38/MAPK-activated protein kinase-2 (MK2) signaling axis is needed for periodontal disease progression: an orally administered p38α inhibitor reduced the progression of experimental periodontal bone loss by reducing inflammation and cytokine expression. Subsequently, the significance of p38 signaling was confirmed with RNA interference to attenuate MK2-reduced cytokine expression and LPS-induced alveolar bone loss. MAPK phosphatase-1 (MKP-1), a negative regulator of MAPK activation, was also critical for periodontal disease progression. In MPK-1-deficient mice, p38-sustained activation increased osteoclast formation and bone loss, whereas MKP-1 overexpression dampened p38 signaling and subsequent cytokine expression. Finally, overexpression of the p38/MK2 target RNA-binding tristetraprolin (TTP) decreased mRNA stability of key inflammatory cytokines at the posttranscriptional level, thereby protecting against periodontal inflammation. Collectively, these studies highlight the importance of p38 MAPK signaling in immune cytokine production and periodontal disease progression.
Collapse
|
121
|
Hess E, Duheron V, Decossas M, Lézot F, Berdal A, Chea S, Golub R, Bosisio MR, Bridal SL, Choi Y, Yagita H, Mueller CG. RANKL induces organized lymph node growth by stromal cell proliferation. THE JOURNAL OF IMMUNOLOGY 2011; 188:1245-54. [PMID: 22210913 DOI: 10.4049/jimmunol.1101513] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
RANK and its ligand RANKL play important roles in the development and regulation of the immune system. We show that mice transgenic for Rank in hair follicles display massive postnatal growth of skin-draining lymph nodes. The proportions of hematopoietic and nonhematopoietic stromal cells and their organization are maintained, with the exception of an increase in B cell follicles. The hematopoietic cells are not activated and respond to immunization by foreign Ag and adjuvant. We demonstrate that soluble RANKL is overproduced from the transgenic hair follicles and that its neutralization normalizes lymph node size, inclusive area, and numbers of B cell follicles. Reticular fibroblastic and vascular stromal cells, important for secondary lymphoid organ formation and organization, express RANK and undergo hyperproliferation, which is abrogated by RANKL neutralization. In addition, they express higher levels of CXCL13 and CCL19 chemokines, as well as MAdCAM-1 and VCAM-1 cell-adhesion molecules. These findings highlight the importance of tissue-derived cues for secondary lymphoid organ homeostasis and identify RANKL as a key molecule for controlling the plasticity of the immune system.
Collapse
Affiliation(s)
- Estelle Hess
- Centre National de la Recherche Scientifique, Laboratoire d'Immunopathologie et Chimie Thérapeutique, Unité Propre de Recherche 9021, Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, 67084 Strasbourg, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
122
|
Findlay DM, Atkins GJ. Relationship between serum RANKL and RANKL in bone. Osteoporos Int 2011; 22:2597-602. [PMID: 21850548 DOI: 10.1007/s00198-011-1740-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 07/25/2011] [Indexed: 11/25/2022]
Abstract
It is now well accepted that the molecule receptor activator of NFκB ligand (RANKL) and osteoprotegerin play key roles in regulating physiological and pathological bone turnover. There are a large number of published reports of circulating RANKL levels in both health and pathology. However, interpretation of these data has been elusive, and the relationship between circulating RANKL and RANKL levels in bone is still not clear. This review explores this subject, documenting the possible origins of circulating RANKL and suggesting additional information that is required before serum RANKL levels can provide useful diagnostic or research information.
Collapse
Affiliation(s)
- D M Findlay
- Discipline of Orthopaedics and Trauma, University of Adelaide, Level 4 Bice Building, Royal Adelaide Hospital, Adelaide 5000, SA, Australia.
| | | |
Collapse
|
123
|
Xiong J, Onal M, Jilka RL, Weinstein RS, Manolagas SC, O'Brien CA. Matrix-embedded cells control osteoclast formation. Nat Med 2011; 17:1235-41. [PMID: 21909103 PMCID: PMC3192296 DOI: 10.1038/nm.2448] [Citation(s) in RCA: 991] [Impact Index Per Article: 70.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 07/20/2011] [Indexed: 02/08/2023]
Abstract
Osteoclasts resorb the mineralized matrices formed by chondrocytes or osteoblasts. The cytokine receptor activator of nuclear factor-κB ligand (RANKL) is essential for osteoclast formation and thought to be supplied by osteoblasts or their precursors, thereby linking bone formation to resorption. However, RANKL is expressed by a variety of cell types, and it is unclear which of them are essential sources for osteoclast formation. Here we have used a mouse strain in which RANKL can be conditionally deleted and a series of Cre-deleter strains to demonstrate that hypertrophic chondrocytes and osteocytes, both of which are embedded in matrix, are essential sources of the RANKL that controls mineralized cartilage resorption and bone remodeling, respectively. Moreover, osteocyte RANKL is responsible for the bone loss associated with unloading. Contrary to the current paradigm, RANKL produced by osteoblasts or their progenitors does not contribute to adult bone remodeling. These results suggest that the rate-limiting step of matrix resorption is controlled by cells embedded within the matrix itself.
Collapse
Affiliation(s)
- Jinhu Xiong
- Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences (UAMS), Little Rock, Arkansas, USA
| | | | | | | | | | | |
Collapse
|
124
|
Barrow AD, Raynal N, Andersen TL, Slatter DA, Bihan D, Pugh N, Cella M, Kim T, Rho J, Negishi-Koga T, Delaisse JM, Takayanagi H, Lorenzo J, Colonna M, Farndale RW, Choi Y, Trowsdale J. OSCAR is a collagen receptor that costimulates osteoclastogenesis in DAP12-deficient humans and mice. J Clin Invest 2011; 121:3505-16. [PMID: 21841309 PMCID: PMC3163954 DOI: 10.1172/jci45913] [Citation(s) in RCA: 153] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2010] [Accepted: 07/01/2011] [Indexed: 12/14/2022] Open
Abstract
Osteoclasts are terminally differentiated leukocytes that erode the mineralized bone matrix. Osteoclastogenesis requires costimulatory receptor signaling through adaptors containing immunoreceptor tyrosine-based activation motifs (ITAMs), such as Fc receptor common γ (FcRγ) and DNAX-activating protein of 12 kDa. Identification of these ITAM-containing receptors and their ligands remains a high research priority, since the stimuli for osteoclastogenesis are only partly defined. Osteoclast-associated receptor (OSCAR) was proposed to be a potent FcRγ-associated costimulatory receptor expressed by preosteoclasts in vitro, but OSCAR lacks a cognate ligand and its role in vivo has been unclear. Using samples from mice and patients deficient in various ITAM signaling pathways, we show here that OSCAR costimulates one of the major FcRγ-associated pathways required for osteoclastogenesis in vivo. Furthermore, we found that OSCAR binds to specific motifs within fibrillar collagens in the ECM that become revealed on nonquiescent bone surfaces in which osteoclasts undergo maturation and terminal differentiation in vivo. OSCAR promoted osteoclastogenesis in vivo, and OSCAR binding to its collagen motif led to signaling that increased numbers of osteoclasts in culture. Thus, our results suggest that ITAM-containing receptors can respond to exposed ligands in collagen, leading to the functional differentiation of leukocytes, which provides what we believe to be a new concept for ITAM regulation of cytokine receptors in different tissue microenvironments.
Collapse
Affiliation(s)
- Alexander David Barrow
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Nicolas Raynal
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Thomas Levin Andersen
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - David A. Slatter
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Dominique Bihan
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Nicholas Pugh
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Marina Cella
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Taesoo Kim
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Jaerang Rho
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Takako Negishi-Koga
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Jean-Marie Delaisse
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Hiroshi Takayanagi
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Joseph Lorenzo
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Marco Colonna
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Richard W. Farndale
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Yongwon Choi
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - John Trowsdale
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| |
Collapse
|
125
|
Edsfeldt A, Dias N, Elmståhl B, Müller MF, Berg K, Nitulescu M, Persson A, Ekberg O, Gonçalves I. Low carotid calcium score is associated with higher levels of glycosaminoglycans, tumor necrosis factor-alpha, and parathyroid hormone in human carotid plaques. Stroke 2011; 42:2966-9. [PMID: 21836100 DOI: 10.1161/strokeaha.111.620658] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Computed tomography (CT) is used to study coronary artery plaques, but little is known about its potential to characterize plaque composition. This study assesses the relation between carotid calcium score (CCS) by CT and plaque composition, namely extracellular matrix, inflammatory mediators, and calcium metabolites. METHODS Thirty patients with significant carotid stenosis underwent preoperative CT. CCS was quantified by Agaston calcium score. Plaque components were studied histologically and biochemically (collagen, elastin, and glycosaminoglycans). Fraktalkine, interferon-γ, interleukin-10, interleukin-12 p70, interleukin-1β, interleukin-6, monocyte chemoattractant protein-1, platelet-derived growth factor-AB/BB, RANTES and tumor necrosis factor-α, and parathyroid hormone were measured using Luminex technology. RESULTS Plaques with CCS ≥400 had more calcium (P=0.012), less glycosaminoglycan (P=0.002), tumor necrosis factor-α (P=0.013), and parathyroid hormone (P=0.028) than those with CCS <400. CCS correlated with plaque content of calcium (r=0.62; P<0.001) and inversely with glycosaminoglycan (r=-0.49; P=0.006) and tumor necrosis factor-α (r=-0.56; P=0.001). CONCLUSIONS Human carotid plaques with high CCS are richer in calcium and have lower amounts of glycosaminoglycan, parathyroid hormone, and tumor necrosis factor-α, which is one of the main proinflammatory cytokines involved in atherosclerosis. This suggests that CCS not only reflects the degree of calcification, but also other important biological components relevant for stability such as inflammation.
Collapse
Affiliation(s)
- Andreas Edsfeldt
- Experimental Cardiovascular Research Unit, Clinical Research Center, Lund University, and Department of Radiology, Entrance 72, House 91:12, Malmö University Hospital, SE-20502 Malmö, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
126
|
Raju R, Balakrishnan L, Nanjappa V, Bhattacharjee M, Getnet D, Muthusamy B, Kurian Thomas J, Sharma J, Rahiman BA, Harsha HC, Shankar S, Prasad TSK, Mohan SS, Bader GD, Wani MR, Pandey A. A comprehensive manually curated reaction map of RANKL/RANK-signaling pathway. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2011; 2011:bar021. [PMID: 21742767 PMCID: PMC3170171 DOI: 10.1093/database/bar021] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Receptor activator of nuclear factor-kappa B ligand (RANKL) is a member of tumor necrosis factor (TNF) superfamily that plays a key role in the regulation of differentiation, activation and survival of osteoclasts and also in tumor cell migration and bone metastasis. Osteoclast activation induced by RANKL regulates hematopoietic stem cell mobilization as part of homeostasis and host defense mechanisms thereby linking regulation of hematopoiesis with bone remodeling. Binding of RANKL to its receptor, Receptor activator of nuclear factor-kappa B (RANK) activates molecules such as NF-kappa B, mitogen activated protein kinase (MAPK), nuclear factor of activated T cells (NFAT) and phosphatidyl 3-kinase (PI3K). Although the molecular and cellular roles of these molecules have been reported previously, a systematic cataloging of the molecular events induced by RANKL/RANK interaction has not been attempted. Here, we present a comprehensive reaction map of the RANKL/RANK-signaling pathway based on an extensive manual curation of the published literature. We hope that the curated RANKL/RANK-signaling pathway model would enable new biomedical discoveries, which can provide novel insights into disease processes and development of novel therapeutic interventions. Database URL:http://www.netpath.org/pathways?path_id=NetPath_21
Collapse
Affiliation(s)
- Rajesh Raju
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Ashley DT, O'Sullivan EP, Davenport C, Devlin N, Crowley RK, McCaffrey N, Moyna NM, Smith D, O'Gorman DJ. Similar to adiponectin, serum levels of osteoprotegerin are associated with obesity in healthy subjects. Metabolism 2011; 60:994-1000. [PMID: 21087777 DOI: 10.1016/j.metabol.2010.10.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Revised: 09/09/2010] [Accepted: 10/04/2010] [Indexed: 12/11/2022]
Abstract
An increase in serum osteoprotegerin (OPG) is associated with type 2 diabetes mellitus, the severity of vascular calcification, and coronary artery disease. Obesity is a risk factor for diabetes and cardiovascular disease, but little is known about the relationship between OPG and obesity. The purpose of this study was to determine if changes in body mass index (BMI) and insulin sensitivity influence circulating OPG in healthy subjects. A total of 100 subjects (36 lean, 41 overweight, and 23 obese) with normal glucose tolerance, blood pressure, and electrocardiogram stress test result volunteered for this study. Insulin sensitivity was estimated using a 2-hour oral glucose tolerance test with oral glucose insulin sensitivity analysis. Osteoprotegerin, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL),soluble receptor activator of nuclear factor-κβ ligand (sRANKL), and adiponectin were analyzed using commercially available enzyme-linked immunosorbent assays. Osteoprotegerin (P < .01) and adiponectin (P < .001) were significantly decreased in the obese compared with lean subjects. There was no significant difference between BMI categories for TRAIL or sRANKL. Controlling for age and sex, there was a significant correlation between OPG and adiponectin (r = 0.391, P < .001), BMI (r = -0.331, P < .001), waist circumference (r = -0.268, P < .01), homeostasis model assessment of insulin resistance (r = -0.222, P < .05), and oral glucose insulin sensitivity (r = 0.221, P < .05). Both OPG and adiponectin were negatively correlated with body weight, BMI, waist circumference, and fasting plasma insulin while being positively correlated with insulin sensitivity (P < .05). Controlling for age, sex, and BMI, TRAIL was positively related to fat mass (r = 0.373, P < .001) and waist circumference (r = 0.257, P < .05). In contrast to patients with type 2 diabetes mellitus, circulating OPG is lower in obese, but otherwise healthy subjects and is positively correlated with indices of insulin sensitivity.
Collapse
Affiliation(s)
- David T Ashley
- Centre for Preventive Medicine, Dublin City University, Dublin 9, Ireland
| | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Chim SM, Qin A, Tickner J, Pavlos N, Davey T, Wang H, Guo Y, Zheng MH, Xu J. EGFL6 promotes endothelial cell migration and angiogenesis through the activation of extracellular signal-regulated kinase. J Biol Chem 2011; 286:22035-46. [PMID: 21531721 PMCID: PMC3121348 DOI: 10.1074/jbc.m110.187633] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Revised: 03/21/2011] [Indexed: 01/08/2023] Open
Abstract
Angiogenesis is required for bone development, growth, and repair. It is influenced by the local bone environment that involves cross-talks between endothelial cells and adjacent bone cells. However, data regarding factors that directly contribute to angiogenesis by bone cells remain poorly understood. Here, we report that EGFL6, a member of the epidermal growth factor (EGF) repeat superfamily proteins, induces angiogenesis by a paracrine mechanism in which EGFL6 is expressed in osteoblastic-like cells but promotes migration and angiogenesis of endothelial cells. Co-immunoprecipitation assays revealed that EGFL6 is secreted in culture medium as a homodimer protein. Using scratch wound healing and transwell assays, we found that conditioned medium containing EGFL6 potentiates SVEC (a simian virus 40-transformed mouse microvascular endothelial cell line) endothelial cell migration. In addition, EGFL6 promotes the endothelial cell tube-like structure formation in Matrigel assays and angiogenesis in a chick embryo chorioallantoic membrane. Furthermore, we show that EGFL6 recombinant protein induces phosphorylation of ERK in SVEC endothelial cells. Inhibition of ERK impaired EGFL6-induced ERK activation and endothelial cell migration. Together, these results demonstrate, for the first time, that osteoblastic-like cells express EGFL6 that is capable of promoting endothelial cell migration and angiogenesis via ERK activation. Thus, the EGLF6 mediates a paracrine mechanism of cross-talk between vascular endothelial cells and osteoblasts and might offer an important new target for the potential treatment of bone diseases, including osteonecrosis, osteoporosis, and fracture healing.
Collapse
Affiliation(s)
- Shek Man Chim
- From the School of Pathology and Laboratory Medicine and
| | - An Qin
- Centre for Orthopaedic Research, School of Surgery, The University of Western Australia, Western Australia 6009, Australia
- the Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011
| | | | - Nathan Pavlos
- Centre for Orthopaedic Research, School of Surgery, The University of Western Australia, Western Australia 6009, Australia
| | - Tamara Davey
- Centre for Orthopaedic Research, School of Surgery, The University of Western Australia, Western Australia 6009, Australia
| | - Hao Wang
- the International Joint Cancer Institute and 301 General Hospital Cancer Center, Second Military Medical University, Shanghai 200433
- the National Engineering Research Center for Antibody Medicine and Shanghai Key Laboratory of Cell Engineering and Antibody, Shanghai 201203, and
- the Schools of Medicine and Pharmacy, Center for Antibody Medicine of Ministry of Education, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Yajun Guo
- the International Joint Cancer Institute and 301 General Hospital Cancer Center, Second Military Medical University, Shanghai 200433
- the National Engineering Research Center for Antibody Medicine and Shanghai Key Laboratory of Cell Engineering and Antibody, Shanghai 201203, and
- the Schools of Medicine and Pharmacy, Center for Antibody Medicine of Ministry of Education, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Ming Hao Zheng
- Centre for Orthopaedic Research, School of Surgery, The University of Western Australia, Western Australia 6009, Australia
| | - Jiake Xu
- From the School of Pathology and Laboratory Medicine and
| |
Collapse
|
129
|
Demer L, Tintut Y. The roles of lipid oxidation products and receptor activator of nuclear factor-κB signaling in atherosclerotic calcification. Circ Res 2011; 108:1482-93. [PMID: 21659652 PMCID: PMC3128471 DOI: 10.1161/circresaha.110.234245] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Accepted: 05/05/2011] [Indexed: 12/20/2022]
Abstract
This review focuses on the roles of oxylipids and receptor activator of nuclear factor-κB ligand signaling in calcific cardiovascular disease. Both intimal and valvular calcifications are closely associated with atherosclerosis, leading investigators to study the role of atherogenic oxidatively modified lipids (oxylipids). Results have identified the molecular signaling through which oxylipids induce osteogenic differentiation and calcification in vascular cells. A surprising concomitant finding was that, in bona fide osteoblasts from skeletal bone, oxylipids have the opposite effect, ie, inhibiting osteoblastic maturation. This is the basis for the lipid hypothesis of osteoporosis. Oxylipids also induce resorptive osteoclastic cells within the bone environment, raising the question of whether resorptive osteoclasts can be harnessed in the vascular context for cell-based therapy to remove artery wall mineral deposits. The challenge is that vascular cells produce antiosteoclastogenic factors, including the soluble decoy receptor for receptor activator of nuclear factor-κB ligand, possibly accounting for the paucity of resorptive cells and the dominance of mineral in atherosclerotic plaque. These factors may have therapeutic use in osteoclastogenic removal of mineral deposits from arteries.
Collapse
Affiliation(s)
- Linda Demer
- Department of Medicine, University of California, Los Angeles, CA, USA.
| | | |
Collapse
|
130
|
Byon CH, Sun Y, Chen J, Yuan K, Mao X, Heath JM, Anderson PG, Tintut Y, Demer LL, Wang D, Chen Y. Runx2-upregulated receptor activator of nuclear factor κB ligand in calcifying smooth muscle cells promotes migration and osteoclastic differentiation of macrophages. Arterioscler Thromb Vasc Biol 2011; 31:1387-96. [PMID: 21454810 PMCID: PMC3098301 DOI: 10.1161/atvbaha.110.222547] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Accepted: 03/15/2011] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Clinical and experimental studies demonstrate the important roles of vascular smooth muscle cells (VSMC) in the pathogenesis of atherosclerosis. We have previously determined that the osteogenic transcription factor Runx2 is essential for VSMC calcification. The present study characterized Runx2-regulated signals and their potential roles in vascular calcification. METHODS AND RESULTS In vivo studies with atherogenic apolipoprotein E(-/-) mice demonstrated that increased oxidative stress was associated with upregulation of Runx2 and receptor activator of nuclear factor κB ligand (RANKL), which colocalized in the calcified atherosclerotic lesions and were juxtaposed to infiltrated macrophages and osteoclast-like cells that are positively stained for an osteoclast marker, tartrate-resistant acid phosphatase. Mechanistic studies using RNA interference, a luciferase reporter system, chromatin immunoprecipitation, and electrophoretic mobility shift assays indicated that Runx2 regulated the expression of RANKL via a direct binding to the 5'-flanking region of the RANKL. Functional characterization revealed that RANKL did not induce VSMC calcification, nor was RANKL required for oxidative stress-induced VSMC calcification. Using a coculture system, we demonstrated that VSMC-expressed RANKL induced migration as well as differentiation of bone marrow-derived macrophages into multinucleated, tartrate-resistant acid phosphatase-positive osteoclast-like cells. These effects were inhibited by the RANKL antagonist osteoprotegerin and with VSMC deficient in Runx2 or RANKL. CONCLUSION We demonstrate that Runx2 directly binds to the promoter and controls the expression of RANKL, which mediates the crosstalk between calcifying VSMC and migration and differentiation of macrophages into osteoclast-like cells in the atherosclerotic lesions. Our studies provide novel mechanistic insights into the regulation and function of VSMC-derived RANKL in the pathogenesis of atherosclerosis and vascular calcification.
Collapse
Affiliation(s)
- Chang Hyun Byon
- Department of Cell Biology University of Alabama at Birmingham
| | - Yong Sun
- Department of Pathology, University of Alabama at Birmingham
| | - Jianfeng Chen
- Department of Pathology, University of Alabama at Birmingham
| | - Kaiyu Yuan
- Department of Pathology, University of Alabama at Birmingham
| | - Xia Mao
- Department of Pathology, University of Alabama at Birmingham
| | - Jack M Heath
- Department of Pathology, University of Alabama at Birmingham
| | | | - Yin Tintut
- Department of Medicine, University of California at Los Angeles
| | - Linda L Demer
- Department of Medicine, University of California at Los Angeles
| | - Deli Wang
- Biostatistics Unit, University of Alabama at Birmingham
| | - Yabing Chen
- Department of Pathology, University of Alabama at Birmingham
- VA Medical Center, Birmingham, AL
| |
Collapse
|
131
|
Kariya Y, Honma M, Hanamura A, Aoki S, Ninomiya T, Nakamichi Y, Udagawa N, Suzuki H. Rab27a and Rab27b are involved in stimulation-dependent RANKL release from secretory lysosomes in osteoblastic cells. J Bone Miner Res 2011; 26:689-703. [PMID: 20939018 DOI: 10.1002/jbmr.268] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The quantity of the receptor activator of NF-κB ligand (RANKL) expressed at the cell surface of osteoblastic cells is an important factor regulating osteoclast activation. Previously, RANKL was found to be localized to secretory lysosomes in osteoblastic cells and to translocate to the cell surface in response to stimulation with RANK-Fc-conjugated beads. However, the in vivo significance of stimulation-dependent RANKL release has not been elucidated. In this study we show that small GTPases Rab27a and Rab27b are involved in the stimulation-dependent RANKL release pathway in osteoblastic cells. Suppression of either Rab27a or Rab27b resulted in a marked reduction in RANKL release after stimulation. Slp4-a, Slp5, and Munc13-4 acted as effector molecules that coordinated Rab27a/b activity in this pathway. Suppression of Rab27a/b or these effector molecules did not inhibit accumulation of RANKL in lysosomal vesicles around the stimulated sites but did inhibit the fusion of these vesicles to the plasma membrane. In osteoblastic cells, suppression of the effector molecules resulted in reduced osteoclastogenic ability. Furthermore, Jinx mice, which lack a functional Munc13-4 gene, exhibited a phenotype characterized by increased bone volume near the tibial metaphysis caused by low bone resorptive activity. In conclusion, stimulation-dependent RANKL release is mediated by Rab27a/b and their effector molecules, and this mechanism may be important for osteoclast activation in vivo.
Collapse
Affiliation(s)
- Yoshiaki Kariya
- Department of Pharmacy, University of Tokyo Hospital, Faculty of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
132
|
Islander U, Jochems C, Lagerquist MK, Forsblad-d'Elia H, Carlsten H. Estrogens in rheumatoid arthritis; the immune system and bone. Mol Cell Endocrinol 2011; 335:14-29. [PMID: 20685609 DOI: 10.1016/j.mce.2010.05.018] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Revised: 05/28/2010] [Accepted: 05/29/2010] [Indexed: 01/29/2023]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that is more common in women than in men. The peak incidence in females coincides with menopause when the ovarian production of sex hormones drops markedly. RA is characterized by skeletal manifestations where production of pro-inflammatory mediators, connected to the inflammation in the joint, leads to bone loss. Animal studies have revealed distinct beneficial effects of estrogens on arthritis, and a positive effect of hormone replacement therapy has been reported in women with postmenopausal RA. This review will focus on the influence of female sex hormones in the pathogenesis and progression of RA.
Collapse
Affiliation(s)
- Ulrika Islander
- Center for Bone and Arthritis Research (CBAR), Department of Rheumatology and Inflammation Research, The Sahlgrenska Academy, University of Gothenburg, Sweden.
| | | | | | | | | |
Collapse
|
133
|
Receptor activator of NF-kappaB (RANK) stimulates the proliferation of epithelial cells of the epidermo-pilosebaceous unit. Proc Natl Acad Sci U S A 2011; 108:5342-7. [PMID: 21402940 DOI: 10.1073/pnas.1013054108] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Receptor activator of NF-κB (RANK), known for controlling bone mass, has been recognized for its role in epithelial cell activation of the mammary gland. Because bone and the epidermo-pilosebaceous unit of the skin share a lifelong renewal activity where similar molecular players operate, and because mammary glands and hair follicles are both skin appendages, we have addressed the function of RANK in the hair follicle and the epidermis. Here, we show that mice deficient in RANK ligand (RANKL) are unable to initiate a new growth phase of the hair cycle and display arrested epidermal homeostasis. However, transgenic mice overexpressing RANK in the hair follicle or administration of recombinant RANKL both activate the hair cycle and epidermal growth. RANK is expressed by the hair follicle germ and bulge stem cells and the epidermal basal cells, cell types implicated in the renewal of the epidermo-pilosebaceous unit. RANK signaling is dispensable for the formation of the stem cell compartment and the inductive hair follicle mesenchyme, and the hair cycle can be rescued by Rankl knockout skin transplantation onto nude mice. RANKL is actively transcribed by the hair follicle at initiation of its growth phase, providing a mechanism for stem cell RANK engagement and hair-cycle entry. Thus, RANK-RANKL regulates hair renewal and epidermal homeostasis and provides a link between these two activities.
Collapse
|
134
|
Fernández M, Pino AM, Figueroa P, Rodríguez JP. The increased expression of receptor activator of nuclear-kappaB ligand (RANKL) of multiple myeloma bone marrow stromal cells is inhibited by the bisphosphonate ibandronate. J Cell Biochem 2011; 111:130-7. [PMID: 20506157 DOI: 10.1002/jcb.22676] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The receptor activator of nuclear factor-kappaB ligand (RANKL) and interleukin-1beta are osteoclast activating factors which are abnormally expressed in bone marrow stromal cells and plasma cells of multiple myeloma patients. In this work we analyzed RANKL expression in human bone marrow mesenchymal stromal cells and the effect of the bisphosphonate ibandronate on RANKL expression after IL-1beta activation of ERK pathway. Mesenchymal stromal cells were obtained from bone marrow iliac aspirates from multiple myeloma patients at stages II/III and non-osteoporotics control donors; these cells were maintained under long-term culture conditions. Cells were cultured in the presence or the absence of 5 ng/ml IL-1beta and/or 5 microM ibandronate, during selected periods. mRNA for RANKL and protein levels were assayed by RT-PCR and Western blot, respectively. Human bone marrow stromal cell line HS-5 was used for assessing IL 1beta- and ibandronate-ERK phosphorylation responses. Multiple myeloma mesenchymal stromal cells differentiate from control cells by increased basal RANKL expression. IL-1beta up regulated RANKL expression showed dependent on activated MEK/ERK pathway. Finally, the bisphosphonate ibandronate, that hindered activation of the MEK/ERK pathway significantly inhibited both basal and IL-1beta dependent RANKL expression by cells. Results indicate that RANKL expression involves the MEK/ERK pathway in multiple myeloma mesenchymal stromal cells, and that early obstruction of this path, such as that achieved with ibandronate, significantly deters RANKL protein expression.
Collapse
Affiliation(s)
- Mireya Fernández
- Laboratorio de Biología Celular y Molecular, INTA, Universidad de Chile, Santiago, Chile.
| | | | | | | |
Collapse
|
135
|
den Uyl D, Nurmohamed MT, van Tuyl LH, Raterman HG, Lems WF. (Sub)clinical cardiovascular disease is associated with increased bone loss and fracture risk; a systematic review of the association between cardiovascular disease and osteoporosis. Arthritis Res Ther 2011; 13:R5. [PMID: 21241491 PMCID: PMC3241350 DOI: 10.1186/ar3224] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Revised: 11/12/2010] [Accepted: 01/17/2011] [Indexed: 02/06/2023] Open
Abstract
Introduction Both cardiovascular disease and osteoporosis are important causes of morbidity and mortality in the elderly. The co-occurrence of cardiovascular disease and osteoporosis prompted us to review the evidence of an association between cardiovascular (CV) disease and osteoporosis and potential shared common pathophysiological mechanisms. Methods A systematic literature search (Medline, Pubmed and Embase) was conducted to identify all clinical studies that investigated the association between cardiovascular disease and osteoporosis. Relevant studies were screened for quality according to guidelines as proposed by the Dutch Cochrane Centre and evidence was summarized. Results Seventy studies were included in this review. Due to a large heterogeneity in study population, design and outcome measures a formal meta-analysis was not possible. Six of the highest ranked studies (mean n = 2,000) showed that individuals with prevalent subclinical CV disease had higher risk for increased bone loss and fractures during follow-up compared to persons without CV disease (range of reported risk: hazard ratio (HR) 1.5; odds ratio (OR) 2.3 to 3.0). The largest study (n = 31,936) reported a more than four times higher risk in women and more than six times higher risk in men. There is moderate evidence that individuals with low bone mass had higher CV mortality rates and incident CV events than subjects with normal bone mass (risk rates 1.2 to 1.4). Although the shared common pathophysiological mechanisms are not fully elucidated, the most important factors that might explain this association appear to be, besides age, estrogen deficiency and inflammation. Conclusions The current evidence indicates that individuals with prevalent subclinical CV disease are at increased risk for bone loss and subsequent fractures. Presently no firm conclusions can be drawn as to what extent low bone mineral density might be associated with increased cardiovascular risk.
Collapse
Affiliation(s)
- Debby den Uyl
- Department of Rheumatology, VU Medical Centre, De Boelelaan 1117, 1081 NV Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
136
|
Park JS, Cho MH, Nam JS, Yoo JS, Ahn CW, Cha BS, Kim KR, Lee HC. Effect of pioglitazone on serum concentrations of osteoprotegerin in patients with type 2 diabetes mellitus. Eur J Endocrinol 2011; 164:69-74. [PMID: 20961967 PMCID: PMC3000683 DOI: 10.1530/eje-10-0875] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Osteoprotegerin (OPG) acts as an important regulatory molecule in atherosclerosis. Recent studies report that thiazolidinediones could affect OPG expression. We investigated the relationship between OPG and inflammatory cytokines and the effects of pioglitazone (a PPARγ (PPARG) agonist) versus metformin on serum OPG levels in type 2 diabetic patients. DESIGN AND METHODS Sixty-seven type 2 diabetic patients were included in this study. They were assigned to pioglitazone (15 mg/day, n=34) or metformin (1000 mg/day, n=33) during 24 weeks. Various anthropometric and metabolic parameters, OPG, interleukin 6 (IL6), C-reactive protein (CRP), adiponectin, and homeostasis model assessment of insulin resistance (HOMA-IR), were measured at baseline and at 6 months of treatment. RESULTS Serum OPG levels correlated significantly with fasting plasma glucose (FPG), HbAlc, HOMA-IR, IL6, and CRP, and inversely correlated with adiponectin after adjusting for age (P<0.05). Multiple regression analysis showed that FPG, HbAlc, and adioponectin were independently correlated with OPG level. After 6 months of treatment, the reduction in FPG and HbAlc levels was similar between the two groups. Pioglitazone treatment significantly increased body mass index (P<0.05) and waist circumference (P<0.05) and decreased triglycerides (P<0.05) and HOMA-IR (P<0.01). The adiponectin concentration was increased (P<0.05), and OPG and CRP levels were decreased in the pioglitazone group (P<0.05), but were unchanged in the metformin group. The changes in serum OPG in the pioglitazone group showed significant correlation with changes in FPG, HbAlc, and adiponectin. CONCLUSIONS In type 2 diabetic patients, pioglitazone decreases OPG levels, and this decrease in OPG levels might be associated with the increase in adiponectin.
Collapse
Affiliation(s)
- Jong Suk Park
- Division of Endocrinology, Department of Internal MedicineKangnam Severance HospitalSeoul, 135-720Republic of Korea
- Severance Institute for Vascular and Metabolic ResearchYonsei University College of Medicine, 146-92, Dogok-Dong, Kangnam-KuSeoul, 135-720Republic of Korea
| | - Min Ho Cho
- Division of Endocrinology, Department of Internal MedicineKangnam Severance HospitalSeoul, 135-720Republic of Korea
| | - Ji Sun Nam
- Division of Endocrinology, Department of Internal MedicineKangnam Severance HospitalSeoul, 135-720Republic of Korea
| | - Jeong Seon Yoo
- Division of Endocrinology, Department of Internal MedicineKangnam Severance HospitalSeoul, 135-720Republic of Korea
| | - Chul Woo Ahn
- Division of Endocrinology, Department of Internal MedicineKangnam Severance HospitalSeoul, 135-720Republic of Korea
- Severance Institute for Vascular and Metabolic ResearchYonsei University College of Medicine, 146-92, Dogok-Dong, Kangnam-KuSeoul, 135-720Republic of Korea
- (Correspondence should be addressed to C W Ahn; )
| | - Bong Soo Cha
- Division of Endocrinology, Department of Internal MedicineKangnam Severance HospitalSeoul, 135-720Republic of Korea
| | - Kyung Rae Kim
- Division of Endocrinology, Department of Internal MedicineKangnam Severance HospitalSeoul, 135-720Republic of Korea
| | - Hyun Chul Lee
- Division of Endocrinology, Department of Internal MedicineKangnam Severance HospitalSeoul, 135-720Republic of Korea
| |
Collapse
|
137
|
Abstract
Blood vessels have a fundamental role both in skeletal homeostasis and in bone repair. Angiogenesis is also important for a successful bone engineering. Therefore, scaffolds should be tested for their ability to favour endothelial cell adhesion, proliferation and functions. The type of endothelial cell to use for in vitro assays should be carefully considered, because the properties of these cells may depend on their source. Morphological and functional relationships between endothelial cells and osteoblasts are evaluated with co-cultures, but this model should still be standardized, particularly for distinguishing the two cell types. Platelet-rich plasma and recombinant growth factors may be useful for stimulating angiogenesis.
Collapse
|
138
|
Alexopoulos A, Peroukides S, Bravou V, Varakis J, Pyrgakis V, Papadaki H. Implication of bone regulatory factors in human coronary artery calcification. Artery Res 2011. [DOI: 10.1016/j.artres.2011.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
139
|
Pepene CE, Ilie IR, Marian I, Duncea I. Circulating osteoprotegerin and soluble receptor activator of nuclear factor κB ligand in polycystic ovary syndrome: relationships to insulin resistance and endothelial dysfunction. Eur J Endocrinol 2011; 164:61-8. [PMID: 20974706 DOI: 10.1530/eje-10-0720] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE There is plenty of evidence that osteoprotegerin (OPG) is linked to subclinical vascular damage and predicts cardiovascular disease in high-risk populations. Our aim is to investigate the relationships of OPG/free soluble receptor activator of nuclear factor κB ligand (sRANKL) to insulin resistance, brachial artery flow-mediated vasodilation (FMD), and the carotid artery intima-media thickness (CIMT) in polycystic ovary syndrome (PCOS), a disorder characterized by hyperandrogenism, impaired glucose control, and endothelial injury. DESIGN A cross-sectional, observational study. METHODS Hormonal and metabolic profiles, FMD, CIMT, serum OPG, and ampli-sRANKL were assessed in 64 young PCOS patients and 20 controls of similar age. Body composition was measured by dual energy X-ray absorptiometry. RESULTS OPG was significantly lower in PCOS and related negatively to free testosterone and positively to estradiol (E(2)) levels. In multivariate analysis, OPG but not ampli-sRANKL correlated positively to fasting insulin, insulin sensitivity indices, and FMD. Neither OPG nor ampli-sRANKL was associated with CIMT. Significantly lower adjusted FMD values were demonstrated in women in the upper OPG quartile group (>2.65 pmol/l) compared with all other quartile groups together (P=0.012). In PCOS, multiple regression analysis retained E(2)/sex hormone-binding globulin ratio, fat mass, and homeostasis model assessment of insulin resistance as independent predictors of OPG. CONCLUSIONS In PCOS, circulating OPG is related to both endothelial dysfunction and insulin resistance, independent of obesity and androgen excess, suggesting OPG as a useful biomarker of these effects. Further studies are needed to evaluate OPG in relation to cardiovascular events and cardiovascular mortality in PCOS.
Collapse
Affiliation(s)
- Carmen Emanuela Pepene
- Departments of Endocrinology Internal Medicine-Cardiology, University of Medicine and Pharmacy Cluj-Napoca, 3-5 Louis Pasteur, 400349 Cluj-Napoca, Romania.
| | | | | | | |
Collapse
|
140
|
Pennisi P, Russo E, Gaudio A, Veca R, D’Amico F, Mangiafico R, Laspina M, Tringali G, Signorelli S, Fiore C. The association between carotid or femoral atherosclerosis and low bone mass in postmenopausal women referred for osteoporosis screening. Does osteoprotegerin play a role? Maturitas 2010; 67:358-62. [DOI: 10.1016/j.maturitas.2010.07.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Revised: 07/21/2010] [Accepted: 07/24/2010] [Indexed: 11/25/2022]
|
141
|
Vik A, Mathiesen EB, Brox J, Wilsgaard T, Njølstad I, Jørgensen L, Hansen JB. Relation between serum osteoprotegerin and carotid intima media thickness in a general population - the Tromsø Study. J Thromb Haemost 2010; 8:2133-9. [PMID: 20738762 DOI: 10.1111/j.1538-7836.2010.03990.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
BACKGROUND Previous studies have reported conflicting results on the relation between serum osteoprotegerin (OPG) concentration and carotid intima media thickness (CIMT). PATIENTS/METHODS The present study was conducted to investigate the relations between OPG, risk factors for cardiovascular diseases (CVD) and carotid intima media thickness (CIMT) in a large cross-sectional study including 6516 subjects aged 25-85years who participated in a population-based health survey. RESULTS CIMT increased significantly across tertiles of OPG after adjustment for traditional cardiovascular risk factors such as age, gender, smoking, total cholesterol, high-density lipoprotein (HDL) cholesterol, C-reactive protein (CRP), body mass index (BMI), systolic blood pressure, CVD and diabetes mellitus (P<0.0001). There was a significant interaction between age and OPG (P=0.026). The risk of being in the uppermost quartile of CIMT was reduced (OR 0.52, 95% CI 0.30-0.88) with each standard deviation (SD) higher level of OPG in subjects <45years (n=444), whereas subjects ≥55years of age (n=4884) had an increased risk of being in the uppermost quartile of CIMT (OR 1.19, 95% CI 1.10-1.29) after adjustment for traditional CVD risk factors. CONCLUSIONS Age has a differential impact on the association between OPG and CIMT in a general population. The present findings may suggest that increased serum OPG does not promote early atherosclerosis in younger subjects.
Collapse
Affiliation(s)
- A Vik
- Department of Medicine, Center for Atherothrombotic Research in Tromsø (CART), University of Tromsø, Tromsø,Norway.
| | | | | | | | | | | | | |
Collapse
|
142
|
Aoki S, Honma M, Kariya Y, Nakamichi Y, Ninomiya T, Takahashi N, Udagawa N, Suzuki H. Function of OPG as a traffic regulator for RANKL is crucial for controlled osteoclastogenesis. J Bone Miner Res 2010; 25:1907-21. [PMID: 20560139 DOI: 10.1002/jbmr.89] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The amount of the receptor activator of NF-κB ligand (RANKL) on the osteoblastic cell surface is considered to determine the magnitude of the signal input to osteoclast precursors and the degree of osteoclastogenesis. Previously, we have shown that RANKL is localized predominantly in lysosomal organelles, but little is found on the osteoblastic cell surface, and consequently, the regulated subcellular trafficking of RANKL in osteoblastic cells is important for controlled osteoclastogenesis. Here we have examined the involvement of osteoprotegerin (OPG), which is currently recognized as a decoy receptor for RANKL, in the regulation of RANKL behavior. It was suggested that OPG already makes a complex with RANKL in the Golgi apparatus and that the complex formation is necessary for RANKL sorting to the secretory lysosomes. It was also shown that each structural domain of OPG is indispensable for exerting OPG function as a traffic regulator. In particular, the latter domains of OPG, whose physiologic functions have been unclear, were indicated to sort RANKL molecules to lysosomes from the Golgi apparatus. In addition, the overexpression of RANK-OPG chimeric protein, which retained OPG function as a decoy receptor but lost the function as a traffic regulator, inhibited endogenous OPG function as a traffic regulator selectively in osteoblastic cells and resulted in the upregulation of osteoclastogenic ability despite the increased number of decoy receptor molecules. Conclusively, OPG function as a traffic regulator for RANKL is crucial for regulating osteoclastogenesis at least as well as that as a decoy receptor.
Collapse
Affiliation(s)
- Shigeki Aoki
- Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
143
|
Bitto A, De Caridi G, Polito F, Calò M, Irrera N, Altavilla D, Spinelli F, Squadrito F. Evidence for markers of hypoxia and apoptosis in explanted human carotid atherosclerotic plaques. J Vasc Surg 2010; 52:1015-21. [PMID: 20719466 DOI: 10.1016/j.jvs.2010.05.116] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Revised: 05/11/2010] [Accepted: 05/30/2010] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Apoptosis and inflammation are important features of atherosclerotic plaques. We investigated whether a common signal molecule can trigger these two apparently separate pathways. Hypoxia inducible factor (HIF-1α) is known to participate in atherosclerosis and to stimulate apoptosis signal-regulating kinase 1 (ASK-1), one of the mitogen-activated protein kinases, which is activated by various extracellular stimuli and involved in a variety of cellular function. METHODS We tested carotid artery specimens from 50 subjects who underwent angioplasty and five age-matched controls for either Western blot or histologic analysis. The hypoxic status was investigated by means of HIF-1α expression in carotid specimens. RESULTS HIF-1α was significantly upregulated in carotid specimens with respect to controls (P < .05), ASK-1 was detected in plaques of any composition from lipidic to calcific, and this expression increased with the stage of the plaque and with the expression of inflammatory (p-ERK, RANK-L, OPG) and apoptotic molecules (caspase 9, p-p-38, and p-JNK). CONCLUSION Our data suggest that hypoxia is the key regulating factor that triggers inflammation as well as apoptosis in the human atherosclerotic plaque.
Collapse
Affiliation(s)
- Alessandra Bitto
- Department of Clinical and Experimental Medicine and Vascular Surgery, Section of Pharmacology, School of Medicine, Messina, Italy
| | | | | | | | | | | | | | | |
Collapse
|
144
|
Laird E, Ward M, McSorley E, Strain JJ, Wallace J. Vitamin D and bone health: potential mechanisms. Nutrients 2010; 2:693-724. [PMID: 22254049 PMCID: PMC3257679 DOI: 10.3390/nu2070693] [Citation(s) in RCA: 169] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2010] [Revised: 06/22/2010] [Accepted: 06/29/2010] [Indexed: 12/31/2022] Open
Abstract
Osteoporosis is associated with increased morbidity, mortality and significant economic and health costs. Vitamin D is a secosteriod hormone essential for calcium absorption and bone mineralization which is positively associated with bone mineral density [BMD]. It is well-established that prolonged and severe vitamin D deficiency leads to rickets in children and osteomalacia in adults. Sub-optimal vitamin D status has been reported in many populations but it is a particular concern in older people; thus there is clearly a need for effective strategies to optimise bone health. A number of recent studies have suggested that the role of vitamin D in preventing fractures may be via its mediating effects on muscle function (a defect in muscle function is one of the classical signs of rickets) and inflammation. Studies have demonstrated that vitamin D supplementation can improve muscle strength which in turn contributes to a decrease in incidence of falls, one of the largest contributors to fracture incidence. Osteoporosis is often considered to be an inflammatory condition and pro-inflammatory cytokines have been associated with increased bone metabolism. The immunoregulatory mechanisms of vitamin D may thus modulate the effect of these cytokines on bone health and subsequent fracture risk. Vitamin D, therefore, may influence fracture risk via a number of different mechanisms.
Collapse
Affiliation(s)
- Eamon Laird
- School of Biomedical Sciences, University of Ulster, Coleraine, UK.
| | | | | | | | | |
Collapse
|
145
|
Jiang XY, Chen HH, Cao FF, Li L, Lin RY, Wen H, Jin L, Wang XF. A polymorphism near osteoprotegerin gene confer risk of obesity in Uyghurs. Endocrine 2010; 37:383-8. [PMID: 20960157 DOI: 10.1007/s12020-010-9318-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Accepted: 02/19/2010] [Indexed: 10/19/2022]
Abstract
To investigate the association of single nucleotide polymorphism (SNP) rs4355801 near osteoprotegerin (OPG) gene and rs3736228 in low-density lipoprotein receptor-related protein 5 (LRP5) gene with metabolic phenotypes [body mass index (BMI), waist-hip ratio, glucose, total cholesterol (CHO), and triglyceride], we carried out a population-based association study in Uyghur population living in Xinjiang Uyghur Autonomous Region of China. We observed a significant higher level of BMI in AG/AA carriers than in GG carriers (P = 0.022) for rs4355801. Subjects with the AG/GG genotype significantly increased the risk of BMI related obesity than subjects with the AA genotype, with an odds ratio of 1.31 (95% CI 1.09-1.56, P = 0.005). The association remained significant after controlling for covariates of age and gender. In addition, we observed a significant higher level of CHO in CT/TT carriers than in CC carriers (P = 0.021) for rs3736228. Our observations provide the first evidence that rs4355801 near OPG gene may confer susceptibility to obesity. In addition, SNP rs3736228 in LPR5 gene may affects the level of CHO in Uyghur population.
Collapse
Affiliation(s)
- Xiao-yan Jiang
- Department of Pharmacology, Tongji University School of Medicine, 1239 Si-Ping Road, 200092 Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
146
|
Shaker OG, El-Shehaby A, Nabih M. Possible Role of Osteoprotegerin and Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand as Markers of Plaque Instability in Coronary Artery Disease. Angiology 2010; 61:756-62. [DOI: 10.1177/0003319710370961] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Osteoprotegerin (OPG) produced by cardiovascular system raising the possibility that alterations of OPG serum levels may be associated with coronary artery disease (CAD). Our aim is to assess the possible role of serum OPG and soluble tumor necrosis factor-related apoptosis-inducing ligand (s-TRAIL) in the pathology of CAD and their uses as markers of plaque stability. A total of 80 male participants were categorized into 3 groups: 28 patients with acute myocardial infarction (AMI), 32 established stable CAD, and 20 healthy controls were enrolled in this study. Acute myocardial infarction and CAD groups exhibited significantly higher OPG levels and lower s-TRAIL levels compared to the stable CAD and control participants. These results are aggravated as the number of affected coronary vessels increase in AMI and stable CAD groups. Conclusion: There is an association between raised serum OPG and reduced s-TRAIL in patients with CAD. Elevation of circulating OPG levels may represent a crucial compensatory mechanism to limit further vascular damage.
Collapse
Affiliation(s)
- Olfat G. Shaker
- Medical Biochemistry, Faculty of Medicine, Cairo University, Egypt,
| | - Amal El-Shehaby
- Medical Biochemistry, Faculty of Medicine, Cairo University, Egypt
| | - Mona Nabih
- Internal medicine, Faculty of Medicine, Cairo University, Egypt
| |
Collapse
|
147
|
Singh DK, Winocour P, Farrington K. Review: Endothelial cell dysfunction, medial arterial calcification and osteoprotegerin in diabetes. ACTA ACUST UNITED AC 2010. [DOI: 10.1177/1474651409355453] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Macrovascular complications such as cardiovascular disease and peripheral vascular disease are the leading cause of increased mortality and morbidity, respectively, in patients with diabetes mellitus. The aetiopathogenesis of macrovasculopathy in diabetes is multifactorial and differs in types 1 and 2 diabetes. Endothelial cell dysfunction is an early feature of diabetic vasculopathy and is associated with poor glycaemic control. Chronic hyperglycaemia may promote an adverse vascular milieu leading to early endothelial cell apoptosis, in the long run. The presence of apoptotic cells in the vascular lumen may trigger a cascade of reactions between the promoters and inhibitors of arterial calcification. Medial arterial calcification, a characteristic feature of diabetes, is an important predictor of cardiovascular disease and occurs independently of atherosclerosis. Medial arterial calcification may occur in the presence of normal serum calcium and phosphate levels. Osteoprotegerin is an important modulator of mineral metabolism and manifests its effects in the bone and arteries. It is hypothesised that osteoprotegerin is a key inhibitor of arterial calcification which is released by endothelial cells as a protective measure for survival in adverse conditions. It is a potential risk marker for early identification and monitoring of disturbed mineral metabolism and vasculopathy in diabetes.
Collapse
Affiliation(s)
- Dhruv K Singh
- Department of Diabetes and Endocrinology, QE Hospital, Welwyn Garden City, UK, , Renal Unit, Lister Hospital, Stevenage, UK
| | - Peter Winocour
- Department of Diabetes and Endocrinology, QE Hospital, Welwyn Garden City, UK
| | | |
Collapse
|
148
|
Kim YS, Min KS, Lee HD, Oh HW, Kim EC. Effect of Cytosolic Phospholipase A2 on Proinflammatory Cytokine-induced Bone Resorptive Genes Including Receptor Activator of Nuclear Factor Kappa B Ligand in Human Dental Pulp Cells. J Endod 2010; 36:636-41. [DOI: 10.1016/j.joen.2009.12.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Revised: 12/08/2009] [Accepted: 12/12/2009] [Indexed: 10/19/2022]
|
149
|
Abstract
Osteoclasts are highly specialized cells capable of degrading mineralized tissue and form at different regions of bone to meet different physiological needs, such as mobilization of calcium, modeling of bone structure, and remodeling of bone matrix. Osteoclast production is elevated in a number of pathological conditions, many of which lead to loss of bone mass. Whether normal or pathological, osteoclastogenesis strictly depends upon support from accessory cells which supply cytokines required for osteoclast differentiation. Only one of these cytokines, receptor activator of NFkappaB ligand (RANKL), is absolutely essential for osteoclast formation throughout life and is thus expressed by all cell types that support osteoclast differentiation. The central role of RANKL in bone resorption is highlighted by the fact that it is the basis for a new therapy to inhibit bone loss. This review will discuss mechanisms that control RANKL gene expression in different osteoclast-support cells and how the study of such mechanisms may lead to a better understanding of the cellular interactions that drive normal and pathological bone resorption.
Collapse
Affiliation(s)
- Charles A O'Brien
- Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, and Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA.
| |
Collapse
|
150
|
Nybo M, Poulsen MK, Grauslund J, Henriksen JE, Rasmussen LM. Plasma osteoprotegerin concentrations in peripheral sensory neuropathy in Type 1 and Type 2 diabetic patients. Diabet Med 2010; 27:289-94. [PMID: 20536491 DOI: 10.1111/j.1464-5491.2010.02940.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
AIMS Osteoprotegerin (OPG) has been linked to different diabetes complications, including cardiovascular disease, and new findings have indicated a specific role in diabetic peripheral neuropathy, but the exact mechanism is unknown. To investigate a possible association between OPG and diabetic peripheral sensory neuropathy, we therefore analysed plasma OPG in Type 1 and Type 2 diabetic patients with and without peripheral neuropathy. SUBJECTS AND METHODS Two hundred Type 1 diabetes mellitus (T1DM) patients and 305 Type 2 diabetes mellitus (T2DM) patients participated in the study. Plasma OPG was measured with a sandwich immunoassay. Peripheral neuropathy was assessed by the Semmes-Weinstein monofilament test. RESULTS In T2DM, plasma OPG concentrations were significantly higher in the peripheral neuropathy group (P < 0.001). Furthermore, there was a significant relationship between the presence of neuropathy in T2DM and plasma OPG levels on logistic regression (P = 0.006). However, when investigated in a full multiple regression model including other long-term diabetes complications, the association became insignificant (P = 0.092). In T1DM, the difference in plasma OPG between groups did not reach significance (P = 0.066). However, plasma OPG significantly correlated to peripheral neuropathy in this group also (P = 0.022), although this correlation was not significant in a multiple linear regression model (P = 0.051). CONCLUSION Plasma OPG levels are related to peripheral neuropathy in both Type 1 and Type 2 diabetes, although with the strongest relationship in T2DM. Before understanding the significance of this, the pathological mechanism involved and, speculatively, a possible use of plasma OPG as a peripheral sensory neuropathy marker, a larger prospective study is needed.
Collapse
Affiliation(s)
- M Nybo
- Department of Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark.
| | | | | | | | | |
Collapse
|