1
|
Pérez-Chacón G, Santamaría PG, Redondo-Pedraza J, González-Suárez E. RANK/RANKL Signaling Pathway in Breast Development and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:309-345. [PMID: 39821032 DOI: 10.1007/978-3-031-70875-6_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
RANK pathway has attracted increasing interest as a promising target in breast cancer, given the availability of denosumab, an anti-RANKL drug. RANK signaling mediates progesterone-driven regulation of mammary gland development and favors breast cancer initiation by controlling mammary cell proliferation and stem cell fate. RANK activation promotes luminal mammary epithelial cell senescence, acting as an initial barrier to tumorigenesis but ultimately facilitating tumor progression and metastasis. Comprehensive analyses have demonstrated that RANK protein expression is an independent biomarker of poor prognosis in postmenopausal and estrogen receptor-negative breast cancer patients. RANK pathway also has multiple roles in immunity and inflammation, regulating innate and adaptive responses. In the tumor microenvironment, RANK and RANKL are expressed by different immune cell populations and contribute to the regulation of tumor immune surveillance, mainly driving immunosuppressive effects.Herein, we discuss the preventive and therapeutic potential of targeting RANK signaling in breast cancer given its tumor cell intrinsic and extrinsic effects. RANKL inhibition has been shown to induce mammary tumor cell differentiation and an antitumor immune response. Moreover, loss of RANK signaling increases sensitivity of breast cancer cells to chemotherapy, targeted therapies such as HER2 and CDK4/6 inhibitors, and immunotherapy. Finally, we describe clinical trials of denosumab for breast cancer prevention, such as those ongoing in women with high risk of developing breast cancer, large phase III clinical trials where the impact of adjuvant denosumab on disease-free survival has been assessed, and window trials to evaluate the immunomodulatory effects of denosumab in breast cancer and other solid tumors.
Collapse
Affiliation(s)
- Gema Pérez-Chacón
- Molecular Oncology, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | | | - Eva González-Suárez
- Molecular Oncology, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
2
|
Freeman C, A S MD, A S P. Unraveling the Intricacies of OPG/RANKL/RANK Biology and Its Implications in Neurological Disorders-A Comprehensive Literature Review. Mol Neurobiol 2024; 61:10656-10670. [PMID: 38777981 DOI: 10.1007/s12035-024-04227-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
The OPG/RANKL/RANK framework, along with its specific receptors, plays a crucial role in bone remodeling and the functioning of the central nervous system (CNS) and associated disorders. Recent research and investigations provide evidence that the components of osteoprotegerin (OPG), receptor activator of NF-kB ligand (RANKL), and receptor activator of NF-kB (RANK) are expressed in the CNS. The CNS structure encompasses cells involved in neuroinflammation, including local macrophages, inflammatory cells, and microglia that cross the blood-brain barrier. The OPG/RANKL/RANK trio modulates the neuroinflammatory response based on the molecular context. The levels of OPG/RANKL/RANK components can serve as biomarkers in the blood and cerebrospinal fluid. They act as neuroprotectants following brain injuries and also participate in the regulation of body weight, internal body temperature, brain ischemia, autoimmune encephalopathy, and energy metabolism. Although the OPG/RANKL/RANK system is primarily known for its role in bone remodeling, further exploring deeper into its multifunctional nature can uncover new functions and novel drug targets for diseases not previously associated with OPG/RANKL/RANK signaling.
Collapse
Affiliation(s)
- Chrisanne Freeman
- Department of Biotechnology, Bishop Heber College, Tamil Nadu, Tiruchirappalli, 620017, India.
| | - Merlyn Diana A S
- Department of Biotechnology, Bishop Heber College, Tamil Nadu, Tiruchirappalli, 620017, India
- Department of Zoology and Research Centre, Lady Doak College, Tamil Nadu, Madurai, 625002, India
| | - Priscilla A S
- Department of Zoology and Research Centre, Lady Doak College, Tamil Nadu, Madurai, 625002, India
| |
Collapse
|
3
|
Alraouji NN, Colak D, Al-Mohanna FH, Alaiya AA, Aboussekhra A. Endogenous osteoprotegerin (OPG) represses ERα and promotes stemness and chemoresistance in breast cancer cells. Cell Death Discov 2024; 10:377. [PMID: 39181873 PMCID: PMC11344809 DOI: 10.1038/s41420-024-02151-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/11/2024] [Accepted: 08/14/2024] [Indexed: 08/27/2024] Open
Abstract
Breast cancer (BC) is the most prevalent cancer and the leading cause of death among women worldwide. The osteoprotegerin (OPG) cytokine, a decoy receptor for RANKL and a key player in bone homeostasis, has pro-and anti-carcinogenic effects in various types of cancer, including breast neoplasms. In the present study, we have shown that ectopic expression of OPG in breast epithelial/cancer cells promotes the pro-metastatic processes epithelial-to-mesenchymal transition (EMT), stemness, angiogenesis as well as the activation of breast stromal fibroblasts. Furthermore, proteomics analysis, which allows the identification and quantification of a plethora of known and unknown proteins, has shown a strong and significant correlation between OPG upregulation and the expression of proteins with functions in EMT and stemness. On the other hand, OPG knockdown in triple-negative breast cancer (TNBC) cells inhibited the formation of cancer stem cells. Importantly, while OPG upregulation significantly enhanced the resistance of luminal BC cells to cisplatin and docetaxel, OPG downregulation sensitized TNBC cells to these chemotherapeutic drugs. We have also shown that OPG negatively controls estrogen receptor α (ERα), and OPG upregulation correlated well with the expression of genes related to ER-negative claudin low cells. Collectively, these results show that OPG promotes stemness and the consequent chemoresistance of breast cancer cells.
Collapse
Affiliation(s)
- Noura N Alraouji
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Dilek Colak
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Falah H Al-Mohanna
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Ayodele A Alaiya
- Department of Cell Therapy & Immunobiology, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Abdelilah Aboussekhra
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia.
| |
Collapse
|
4
|
Zhang L, Zeng F, Jiang M, Han M, Huang B. Roles of osteoprotegerin in endocrine and metabolic disorders through receptor activator of nuclear factor kappa-B ligand/receptor activator of nuclear factor kappa-B signaling. Front Cell Dev Biol 2022; 10:1005681. [PMID: 36407115 PMCID: PMC9671468 DOI: 10.3389/fcell.2022.1005681] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/17/2022] [Indexed: 11/21/2023] Open
Abstract
Endocrine and metabolic diseases show increasing incidence and high treatment costs worldwide. Due to the complexity of their etiology and mechanism, therapeutic strategies are still lacking. Osteoprotegerin (OPG), a member of the tumor necrosis factor receptor superfamily, appears to be a potential candidate for the treatment of these diseases. Studies based on clinical analysis and rodent animal models reveal the roles of OPG in various endocrine and metabolic processes or disorders, such as bone remodeling, vascular calcification, and β-cell proliferation, through the receptor activator of nuclear factor kappa-B ligand (RANKL) and the receptor activator of NF-κB (RANK). Thus, in this review, we mainly focus on relevant diseases, including osteoporosis, cardiovascular disease (CVD), diabetes, and gestational diabetes mellitus (GDM), to summarize the effects of the RANKL/RANK/OPG system in endocrine and metabolic tissues and diseases, thereby providing a comprehensive insight into OPG as a potential drug for endocrine and metabolic diseases.
Collapse
Affiliation(s)
- Luodan Zhang
- Department of Nephrology, Anhui Provincial Children’s Hospital, Children’s Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Fa Zeng
- Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, Guangdong, China
| | - Minmin Jiang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, MOE Key Laboratory of Population Health Across Life Cycle, NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei, Anhui, China
| | - Maozhen Han
- College of Life Science, Anhui Medical University, Hefei, Anhui, China
| | - Binbin Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, MOE Key Laboratory of Population Health Across Life Cycle, NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei, Anhui, China
| |
Collapse
|
5
|
Al-Nasrallah HK, Al-Ansari MM, Aboussekhra A. Osteoprotegerin (OPG) Upregulation Activates Breast Stromal Fibroblasts and Enhances Their Pro-Carcinogenic Effects through the STAT3/IL-6 Signaling. Cells 2022; 11:3369. [PMID: 36359766 PMCID: PMC9655455 DOI: 10.3390/cells11213369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 07/30/2023] Open
Abstract
Breast carcinomas are composed of cancer cells surrounded by various types of non-cancer cells such as fibroblasts. While active cancer-associated fibroblasts (CAFs) support tumor initiation and progression, quiescent breast stromal fibroblasts (BSFs) inhibit these effects through various cytokines such as osteoprotegerin (OPG). We showed here that OPG is upregulated in CAFs as compared to their adjacent normal tumor counterpart fibroblasts. Interestingly, breast cancer cells can upregulate OPG in BSFs in an IL-6-dependent manner through the IL-6/STAT3 pathway. When upregulated by ectopic expression, OPG activated BSFs through the NF-κB/STAT3/AUF1 signaling pathway and promoted their paracrine pro-carcinogenic effects in an IL-6-dependent manner. In addition, this increase in the OPG level enhanced the potential of BSFs to promote the growth of humanized orthotopic tumors in mice. However, specific OPG knock-down suppressed active CAFs and their paracrine pro-carcinogenic effects. Similar effects were observed when CAF cells were exposed to the pure recombinant OPG (rOPG) protein. Together, these findings show the importance of OPG in the activation of stromal fibroblasts and the possible use of rOPG or inhibitors of the endogenous protein to target CAFs as precision cancer therapeutics.
Collapse
Affiliation(s)
- Huda K. Al-Nasrallah
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - Mysoon M. Al-Ansari
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
- Department of Microbiology, Faculty of Science and Medical Studies, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdelilah Aboussekhra
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| |
Collapse
|
6
|
Abstract
Since the receptor activator of nuclear factor-kappa B ligand (RANKL), its cognate receptor activator of nuclear factor-kappa B (RANK), and the decoy receptor osteoprotegerin (OPG) were discovered, a number of studies have uncovered the crucial role of the RANKL-RANK-OPG pathway in controlling the key aspect of bone homeostasis, the immune system, inflammation, cancer, and other systems under pathophysiological condition. These findings have expanded the understanding of the multifunctional biology of the RANKL-RANK-OPG pathway and led to the development of therapeutic potential targeting this pathway. The successful development and application of anti-RANKL antibody in treating diseases causing bone loss validates the utility of therapeutic approaches based on the modulation of this pathway. Moreover, recent studies have demonstrated the involvement of the RANKL-RANK pathway in osteoblast differentiation and bone formation, shedding light on the RANKL-RANK dual signaling in coupling bone resorption and bone formation. In this review, we will summarize the current understanding of the RANKL-RANK-OPG system in the context of the bone and the immune system as well as the impact of this pathway in disease conditions, including cancer development and metastasis.
Collapse
Affiliation(s)
- Noriko Takegahara
- Departments of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Hyunsoo Kim
- Departments of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Yongwon Choi
- Departments of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
7
|
Ming J, Cronin SJF, Penninger JM. Targeting the RANKL/RANK/OPG Axis for Cancer Therapy. Front Oncol 2020; 10:1283. [PMID: 32850393 PMCID: PMC7426519 DOI: 10.3389/fonc.2020.01283] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022] Open
Abstract
RANKL and RANK are expressed in different cell types and tissues throughout the body. They were originally described for their essential roles in bone remodeling and the immune system but have subsequently been shown to provide essential signals from regulating mammary gland homeostasis during pregnancy to modulating tumorigenesis. The success of RANKL/RANK research serves as a paragon for translational research from the laboratory to the bedside. The case in point has been the development of Denosumab, a RANKL-blocking monoclonal antibody which has already helped millions of patients suffering from post-menopausal osteoporosis and skeletal related events in cancer. Here we will provide an overview of the pathway from its origins to its clinical relevance in disease, with a special focus on emerging evidence demonstrating the therapeutic value of targeting the RANKL/RANK/OPG axis not only in breast cancer but also as an addition to the cancer immunotherapy arsenal.
Collapse
Affiliation(s)
- Jie Ming
- Department of Breast and Thyroid Surgery, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Shane J F Cronin
- Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna Biocenter, Vienna, Austria
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna Biocenter, Vienna, Austria.,Department of Medical Genetics, Life Science Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
8
|
Goswami S, Sharma-Walia N. Osteoprotegerin rich tumor microenvironment: implications in breast cancer. Oncotarget 2018; 7:42777-42791. [PMID: 27072583 PMCID: PMC5173171 DOI: 10.18632/oncotarget.8658] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 03/31/2016] [Indexed: 12/18/2022] Open
Abstract
Osteoprotegerin (OPG) is a soluble decoy receptor for tumor necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL). It belongs to the tumor necrosis factor receptor superfamily (TNFRSF). OPG was initially discovered to contribute to homeostasis of bone turnover due to its capability of binding to receptor activator of nuclear factor-kappaB (NF-kB). However, apart from bone turnover, OPG plays important and diverse role(s) in many biological functions. Besides having anti-osteoclastic activity, OPG is thought to exert a protective anti-apoptotic action in OPG-expressing tumors by overcoming the physiologic mechanism of tumor surveillance exerted by TRAIL. Along with inhibiting TRAIL induced apoptosis, it can induce proliferation by binding to various cell surface receptors and thus turning on the canonical cell survival and proliferative pathways. OPG also induces angiogenesis, one of the hallmarks of cancer, thus facilitating tumor growth. Recently, the understanding of OPG and its different roles has been augmented substantially. This review is aimed at providing a very informative overview as to how OPG affects cancer progression especially breast cancer.
Collapse
Affiliation(s)
- Sudeshna Goswami
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Neelam Sharma-Walia
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| |
Collapse
|
9
|
Bernardi S, Toffoli B, Bossi F, Candido R, Stenner E, Carretta R, Barbone F, Fabris B. Circulating osteoprotegerin is associated with chronic kidney disease in hypertensive patients. BMC Nephrol 2017; 18:219. [PMID: 28683789 PMCID: PMC5500921 DOI: 10.1186/s12882-017-0625-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 06/19/2017] [Indexed: 12/26/2022] Open
Abstract
Background Osteoprotegerin (OPG) is a glycoprotein that plays an important regulatory role in the skeletal, vascular, and immune system. It has been shown that OPG predicts chronic kidney disease (CKD) in diabetic patients. We hypothesized that OPG could be a risk marker of CKD development also in non-diabetic hypertensive patients. Methods A case-control study was carried out to measure circulating OPG levels in 42 hypertensive patients with CKD and in 141 hypertensive patients without CKD. A potential relationship between OPG and the presence of CKD was investigated and a receiver-operating characteristic (ROC) curve was designed thereafter to identify a cut-off value of OPG that best explained the presence of CKD. Secondly, to evaluate whether OPG increase could affect the kidney, 18 C57BL/6J mice were randomized to be treated with saline or recombinant OPG every 3 weeks for 12 weeks. Results Circulating OPG levels were significantly higher in hypertensive patients with CKD, and there was a significant inverse association between OPG and renal function, that was independent from other variables. ROC analysis showed that OPG levels had a high statistically predictive value on CKD in hypertensive patients, which was greater than that of hypertension. The OPG best cut-off value associated with CKD was 1109.19 ng/L. In the experimental study, OPG delivery significantly increased the gene expression of pro-inflammatory and pro-fibrotic mediators, as well as the glomerular nitrosylation of proteins. Conclusions This study shows that OPG is associated with CKD in hypertensive patients, where it might have a higher predictive value than that of hypertension for CKD development. Secondly, we found that OPG delivery significantly increased the expression of molecular pathways involved in kidney damage. Further longitudinal studies are needed not only to evaluate whether OPG predicts CKD development but also to clarify whether OPG should be considered a risk factor for CKD. Electronic supplementary material The online version of this article (doi:10.1186/s12882-017-0625-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stella Bernardi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, Trieste, 34100, Italy. .,Azienda Sanitaria Universitaria Integrata di Trieste (ASUITS), Strada di Fiume, Trieste, 34100, Italy.
| | - Barbara Toffoli
- IRCCS Burlo Garofolo, Via dell'Istria, Trieste, 34100, Italy
| | - Fleur Bossi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, Trieste, 34100, Italy
| | - Riccardo Candido
- Azienda Sanitaria Universitaria Integrata di Trieste (ASUITS), Strada di Fiume, Trieste, 34100, Italy
| | - Elisabetta Stenner
- Azienda Sanitaria Universitaria Integrata di Trieste (ASUITS), Strada di Fiume, Trieste, 34100, Italy
| | - Renzo Carretta
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, Trieste, 34100, Italy.,Azienda Sanitaria Universitaria Integrata di Trieste (ASUITS), Strada di Fiume, Trieste, 34100, Italy
| | - Fabio Barbone
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, Trieste, 34100, Italy.,IRCCS Burlo Garofolo, Via dell'Istria, Trieste, 34100, Italy
| | - Bruno Fabris
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, Trieste, 34100, Italy.,Azienda Sanitaria Universitaria Integrata di Trieste (ASUITS), Strada di Fiume, Trieste, 34100, Italy
| |
Collapse
|
10
|
Prystupa A, Dąbrowska A, Sak JJ, Tarach J, Toruń-Jurkowska A, Lachowska-Kotowska P, Dzida G. Concentrations of fetuin-A, osteoprotegerin and α-Klotho in patients with alcoholic liver cirrhosis. Exp Ther Med 2016; 12:3464-3470. [PMID: 27882180 PMCID: PMC5103841 DOI: 10.3892/etm.2016.3754] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Accepted: 07/26/2016] [Indexed: 12/15/2022] Open
Abstract
The aim of the present study was to evaluate the concentrations of fetuin-A, osteoprotegerin (OPG) and α-Klotho protein in patients with alcoholic cirrhosis at different stages of the disease, and to demonstrate that fetuin-A, osteoprotegin and α-Klotho may be used as markers of the severity of cirrhosis. A total of 54 patients with alcoholic liver cirrhosis treated in various hospitals in the Lublin region of Poland were randomly enrolled. The control group consisted of 18 healthy individuals without liver disease, who did not drink alcohol. Serum levels of fetuin-A, OPG and α-Klotho were measured by ELISA kits. Levels of fetuin-A were significantly reduced in patients with alcoholic liver cirrhosis compared with the control group. OPG levels were higher in patients with alcoholic liver cirrhosis than in the controls, whereas the levels of α-Klotho were comparable in the cirrhosis and control groups. No statistically significant differences in the concentrations of fetuin-A, OPG and α-Klotho protein were demonstrated according to type of liver cirrhosis. The findings of the present study revealed a significant negative correlation between the level of α-Klotho protein and C-reactive protein in the patients with alcoholic liver cirrhosis. Concentrations of fetuin-A were lower, whereas those of OPG were higher, in the alcoholic liver cirrhosis group compared with the control group. Fetuin-A, OPG and α-Klotho may not be good indicators of liver cirrhosis severity. In conclusion, fetuin-A and OPG may be used in the diagnosis of liver cirrhosis.
Collapse
Affiliation(s)
- Andrzej Prystupa
- Department of Internal Medicine, Medical University of Lublin, 20-059 Lublin, Poland
| | - Anna Dąbrowska
- Department of Endocrinology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Jarosław Jerzy Sak
- Department of Nephrology, Medical University of Lublin, 20-059 Lublin, Poland; Department of Ethics and Human Philosophy, Medical University of Lublin, 20-059 Lublin, Poland
| | - Jerzy Tarach
- Department of Endocrinology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Anna Toruń-Jurkowska
- Department of Mathematics and Medical Biostatistics, Medical University of Lublin, 20-059 Lublin, Poland
| | | | - Grzegorz Dzida
- Department of Internal Medicine, Medical University of Lublin, 20-059 Lublin, Poland
| |
Collapse
|
11
|
Bernardi S, Bossi F, Toffoli B, Fabris B. Roles and Clinical Applications of OPG and TRAIL as Biomarkers in Cardiovascular Disease. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1752854. [PMID: 27200369 PMCID: PMC4856888 DOI: 10.1155/2016/1752854] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/28/2016] [Accepted: 04/05/2016] [Indexed: 12/19/2022]
Abstract
Cardiovascular diseases (CVD) remain the major cause of death and premature disability in Western societies. Assessing the risk of CVD is an important aspect in clinical decision-making. Among the growing number of molecules that are studied for their potential utility as CVD biomarkers, a lot of attention has been focused on osteoprotegerin (OPG) and its ligands, which are receptor activator of nuclear factor κB ligand (RANKL) and TNF-related apoptosis-inducing ligand. Based on the existing literature and on our experience in this field, here we review what the possible roles of OPG and TRAIL in CVD are and their potential utility as CVD biomarkers.
Collapse
Affiliation(s)
- Stella Bernardi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, 34149 Trieste, Italy
| | - Fleur Bossi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, 34149 Trieste, Italy
| | - Barbara Toffoli
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, 34149 Trieste, Italy
| | - Bruno Fabris
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, 34149 Trieste, Italy
| |
Collapse
|
12
|
Ayina Ayina CN, Sobngwi E, Essouma M, Noubiap JJN, Boudou P, Etoundi Ngoa LS, Gautier JF. Osteoprotegerin in relation to insulin resistance and blood lipids in sub-Saharan African women with and without abdominal obesity. Diabetol Metab Syndr 2015; 7:47. [PMID: 26034511 PMCID: PMC4450452 DOI: 10.1186/s13098-015-0042-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 05/13/2015] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Osteoprotegerin (OPG), a soluble member of the tumor necrosis factor receptor superfamily that inhibits bone resorption, has been suggested as a potential marker of cardiovascular risk. This study aimed to assess the relationship between insulin resistance, lipid profile and OPG levels in obese and non-obese sub-Saharan African women. METHODS Sixty obese (44) and non-obese (16) volunteer women aged 18 to 40 years were recruited in this cross-sectional study. Their clinical (age, height, weight, waist circumference, systolic and diastolic blood pressures) and biochemical parameters (fasting blood glucose, total cholesterol, high density lipoprotein-cholesterol (HDL-C)) were measured using standard methods. Insulin levels were measured using an electrochemiluminescence immunoassay, while OPG levels were measured using the ELISA technique. Low density lipoprotein-cholesterol (LDL-C), body mass index (BMI) and Homeostasis Model Assessment of Insulin Resistance (HOMA-IR) were calculated using standard methods. Abdominal obesity was defined as a waist circumference ≥ 80 cm. RESULTS OPG levels were higher in obese than in normal subjects, though the difference was not significant (p = 0.9). BMI, waist circumference, percent body fat and systolic blood pressure were significantly higher in obese than in non-obese subjects (p < 0.05). In these subjects, only age significantly correlated with OPG levels (r = 0.831, p = 0.003), while none of the anthropometric nor metabolic parameter did, even after adjustment for age. In obese subjects, OPG levels fairly correlated with HDL-C (r = 0.298, p = 0.058), and significantly correlated with HOMA-IR (r = -0.438, p = 0.018). After adjustment for age, OPG levels remained negatively correlated to HOMA-IR (r = -0.516, p = 0.020) and LDL-C (r = -0.535, p = 0.015) and positively correlated to HDL-C (r = 0.615, p = 0.004). In multiple linear regression analysis, age was a main determinant of OPG levels in non-obese (β = 0.647, p = 0.006) and obese (β = 0.356, p = 0.044) women. HDL-C was also associated to OPG levels in obese women (β = 0.535, p = 0.009). CONCLUSION The positive correlation of OPG with HDL-C and HOMA-IR, and its negative correlation with LDL-C suggest that it may be a marker of insulin sensitivity/resistance and atherogenic risk in obese African women.
Collapse
Affiliation(s)
| | - Eugene Sobngwi
- />Department of Internal Medicine and Specialties, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
- />Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
- />National Obesity Center, Yaoundé Central Hospital, Yaoundé, Cameroon
| | - Mickael Essouma
- />Department of Internal Medicine and Specialties, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
| | - Jean Jacques N. Noubiap
- />Department of Medicine, Groote Schuur Hospital and University of Cape Town, Cape Town, South Africa
- />Medical Diagnostic Center, Yaoundé, Cameroon
| | - Philippe Boudou
- />Department of Hormonal Biology, Saint-Louis Hospital, Public Assistance - Paris Hospitals, University Paris-Diderot Paris-7, Paris, France
- />Department of Diabetes and Endocrinology, Saint-Louis Hospital, Public Assistance - Paris Hospitals, University Paris-Diderot Paris-7, Paris, France
| | - Laurent Serge Etoundi Ngoa
- />Department of Animal Science, Higher Teacher’s Training College, University of Yaoundé I, Yaoundé, Cameroon
| | - Jean François Gautier
- />Department of Diabetes and Endocrinology, Saint-Louis Hospital, Public Assistance - Paris Hospitals, University Paris-Diderot Paris-7, Paris, France
- />INSERM UMRS 1138, Cordeliers Research Centre, University Pierre et Marie Curie-Paris 6, Paris, France
| |
Collapse
|
13
|
Walsh MC, Choi Y. Biology of the RANKL-RANK-OPG System in Immunity, Bone, and Beyond. Front Immunol 2014; 5:511. [PMID: 25368616 PMCID: PMC4202272 DOI: 10.3389/fimmu.2014.00511] [Citation(s) in RCA: 445] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 10/02/2014] [Indexed: 12/27/2022] Open
Abstract
Discovery and characterization of the cytokine receptor-cytokine-decoy receptor triad formed by receptor activator of nuclear factor kappa-B ligand (RANKL)–receptor activator of NF-κB (RANK)–osteoprotegerin (OPG) have led not only to immense advances in understanding the biology of bone homeostasis, but have also crystalized appreciation of the critical regulatory relationship that exists between bone and immunity, resulting in the emergence of the burgeoning field of osteoimmunology. RANKL–RANK–OPG are members of the tumor necrosis factor (TNF) and TNF receptor superfamilies, and share signaling characteristics common to many members of each. Developmentally regulated and cell-type specific expression patterns of each of these factors have revealed key regulatory functions for RANKL–RANK–OPG in bone homeostasis, organogenesis, immune tolerance, and cancer. Successful efforts at designing and developing therapeutic agents targeting RANKL–RANK–OPG have been undertaken for osteoporosis, and additional efforts are underway for other conditions. In this review, we will summarize the basic biology of the RANKL–RANK–OPG system, relate its cell-type specific functions to system-wide mechanisms of development and homeostasis, and highlight emerging areas of interest for this cytokine group.
Collapse
Affiliation(s)
- Matthew C Walsh
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA , USA
| | - Yongwon Choi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA , USA
| |
Collapse
|
14
|
Galeone A, Paparella D, Colucci S, Grano M, Brunetti G. The role of TNF-α and TNF superfamily members in the pathogenesis of calcific aortic valvular disease. ScientificWorldJournal 2013; 2013:875363. [PMID: 24307884 PMCID: PMC3836568 DOI: 10.1155/2013/875363] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 10/02/2013] [Indexed: 01/08/2023] Open
Abstract
Calcific aortic valve disease (CAVD) represents a slowly progressive pathologic process associated with major morbidity and mortality. The process is characterized by multiple steps: inflammation, fibrosis, and calcification. Numerous studies focalized on its physiopathology highlighting different "actors" for the multiple "acts." This paper focuses on the role of the tumor necrosis factor superfamily (TNFSF) members in the pathogenesis of CAVD. In particular, we discuss the clinical and experimental studies providing evidence of the involvement of tumor necrosis factor-alpha (TNF-α), receptor activator of nuclear factor-kappa B (NF-κB) ligand (RANKL), its membrane receptor RANK and its decoy receptor osteoprotegerin (OPG), and TNF-related apoptosis-inducing ligand (TRAIL) in valvular calcification.
Collapse
Affiliation(s)
- Antonella Galeone
- Division of Cardiac Surgery, Department of Emergencies and Organ Transplantation (DETO), University of Bari “Aldo Moro”, Italy
| | - Domenico Paparella
- Division of Cardiac Surgery, Department of Emergencies and Organ Transplantation (DETO), University of Bari “Aldo Moro”, Italy
| | - Silvia Colucci
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, Section of Human Anatomy and Histology, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Maria Grano
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, Section of Human Anatomy and Histology, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Giacomina Brunetti
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, Section of Human Anatomy and Histology, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy
| |
Collapse
|
15
|
Oñate B, Vilahur G, Camino-López S, Díez-Caballero A, Ballesta-López C, Ybarra J, Moscatiello F, Herrero J, Badimon L. Stem cells isolated from adipose tissue of obese patients show changes in their transcriptomic profile that indicate loss in stemcellness and increased commitment to an adipocyte-like phenotype. BMC Genomics 2013; 14:625. [PMID: 24040759 PMCID: PMC3848661 DOI: 10.1186/1471-2164-14-625] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 09/11/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The adipose tissue is an endocrine regulator and a risk factor for atherosclerosis and cardiovascular disease when by excessive accumulation induces obesity. Although the adipose tissue is also a reservoir for stem cells (ASC) their function and "stemcellness" has been questioned. Our aim was to investigate the mechanisms by which obesity affects subcutaneous white adipose tissue (WAT) stem cells. RESULTS Transcriptomics, in silico analysis, real-time polymerase chain reaction (PCR) and western blots were performed on isolated stem cells from subcutaneous abdominal WAT of morbidly obese patients (ASCmo) and of non-obese individuals (ASCn). ASCmo and ASCn gene expression clustered separately from each other. ASCmo showed downregulation of "stemness" genes and upregulation of adipogenic and inflammatory genes with respect to ASCn. Moreover, the application of bioinformatics and Ingenuity Pathway Analysis (IPA) showed that the transcription factor Smad3 was tentatively affected in obese ASCmo. Validation of this target confirmed a significantly reduced Smad3 nuclear translocation in the isolated ASCmo. CONCLUSIONS The transcriptomic profile of the stem cells reservoir in obese subcutaneous WAT is highly modified with significant changes in genes regulating stemcellness, lineage commitment and inflammation. In addition to body mass index, cardiovascular risk factor clustering further affect the ASC transcriptomic profile inducing loss of multipotency and, hence, capacity for tissue repair. In summary, the stem cells in the subcutaneous WAT niche of obese patients are already committed to adipocyte differentiation and show an upregulated inflammatory gene expression associated to their loss of stemcellness.
Collapse
Affiliation(s)
- Blanca Oñate
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Xie GQ, Lei DD, He HB, Gong JJ, Chen C, Chen P, Zhang H, Luo XH, Liao EY, Wu XP. Relationship between serum TGF-β1, OPG levels and osteoporotic risk in native Chinese women. Clin Chim Acta 2013; 423:116-21. [DOI: 10.1016/j.cca.2013.04.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 03/26/2013] [Accepted: 04/17/2013] [Indexed: 12/17/2022]
|
17
|
Abstract
Osteoprotegerin (OPG) is a 401 amino acid N-glycosylated protein, which is highly expressed in a large number of tissues. OPG mainly binds to two ligands, i.e. RANKL (receptor activator of nuclear factor κB ligand) and TRAIL (tumor necrosis factor- related apoptosis-inducing ligand). Upon binding to the former ligand, OPG inhibits the activation of osteoclasts and promotes apoptosis of osteoclasts, whereas the binding of OPG with TRAIL prevents apoptosis of tumor cells. There is now emerging evidence that OPG participates in the pathogenesis of atherosclerosis and cardiovascular diseases by amplifying the adverse effects of inflammation and several traditional risk factors such as hyperlipidemia, endothelial dysfunction, diabetes mellitus, and hypertension. Some epidemiological studies also showed a positive association between OPG levels and cardiovascular morbidity and mortality. The aim of this article is to provide an overview of the main biochemical, physiological, and pathological aspects of OPG biology in cardiovascular disease.
Collapse
Affiliation(s)
- Martina Montagnana
- Chemistry and Clinical Microscopy Section, Department of Life and Reproduction Sciences, University Hospital of Verona, Italy.
| | | | | | | |
Collapse
|
18
|
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
|
19
|
Stanley AC, de Labastida Rivera F, Haque A, Sheel M, Zhou Y, Amante FH, Bunn PT, Randall LM, Pfeffer K, Scheu S, Hickey MJ, Saunders BM, Ware C, Hill GR, Tamada K, Kaye PM, Engwerda CR. Critical roles for LIGHT and its receptors in generating T cell-mediated immunity during Leishmania donovani infection. PLoS Pathog 2011; 7:e1002279. [PMID: 21998581 PMCID: PMC3188526 DOI: 10.1371/journal.ppat.1002279] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 08/08/2011] [Indexed: 11/18/2022] Open
Abstract
LIGHT (TNFSF14) is a member of the TNF superfamily involved in inflammation and defence against infection. LIGHT signals via two cell-bound receptors; herpes virus entry mediator (HVEM) and lymphotoxin-beta receptor (LTβR). We found that LIGHT is critical for control of hepatic parasite growth in mice with visceral leishmaniasis (VL) caused by infection with the protozoan parasite Leishmania donovani. LIGHT-HVEM signalling is essential for early dendritic cell IL-12/IL-23p40 production, and the generation of IFNγ- and TNF-producing T cells that control hepatic infection. However, we also discovered that LIGHT-LTβR interactions suppress anti-parasitic immunity in the liver in the first 7 days of infection by mechanisms that restrict both CD4+ T cell function and TNF-dependent microbicidal mechanisms. Thus, we have identified distinct roles for LIGHT in infection, and show that manipulation of interactions between LIGHT and its receptors may be used for therapeutic advantage. Visceral leishmaniasis (VL) is a potentially fatal human disease caused by the intracellular protozoan parasites Leishmania donovani and L. infantum (chagasi). Parasites infect macrophages throughout the viscera, though the spleen and liver are the major sites of disease. VL is responsible for significant morbidity and mortality in the developing world, particularly in India, Sudan, Nepal, Bangladesh and Brazil. Because of the intrusive techniques required to analyse tissue in VL patients, our current understanding of the host immune response during VL largely derives from studies performed in genetically susceptible mice. We have discovered that mice which are unable to produce a cytokine called LIGHT have poor control of L. donovani infection in the liver, compared with wild-type control animals. In addition, we demonstrated that LIGHT has distinct roles during VL, depending on which of its two major cell-bound receptors it engages. Finally, we identified an antibody that stimulates the lymphotoxin β receptor (one of the LIGHT receptors), that can stimulate anti-parasitic activity during an established infection, thereby identifying this receptor as a therapeutic target during disease.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Cell Proliferation/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Female
- Immunity, Cellular
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Interleukin-12/biosynthesis
- Interleukin-23/biosynthesis
- Leishmania donovani/immunology
- Leishmania donovani/pathogenicity
- Leishmaniasis, Visceral/immunology
- Leishmaniasis, Visceral/parasitology
- Leishmaniasis, Visceral/pathology
- Liver/parasitology
- Liver/pathology
- Lymphotoxin beta Receptor/immunology
- Lymphotoxin beta Receptor/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, Tumor Necrosis Factor, Member 14/immunology
- Receptors, Tumor Necrosis Factor, Member 14/metabolism
- Signal Transduction
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
- Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism
Collapse
Affiliation(s)
- Amanda C. Stanley
- Queensland Institute of Medical Research and the Australian Centre for Vaccine Development, Herston, Queensland, Australia
- Institute for Molecular Biology, University of Queensland, St Lucia, Queensland, Australia
| | - Fabian de Labastida Rivera
- Queensland Institute of Medical Research and the Australian Centre for Vaccine Development, Herston, Queensland, Australia
| | - Ashraful Haque
- Queensland Institute of Medical Research and the Australian Centre for Vaccine Development, Herston, Queensland, Australia
| | - Meru Sheel
- Queensland Institute of Medical Research and the Australian Centre for Vaccine Development, Herston, Queensland, Australia
| | - Yonghong Zhou
- Queensland Institute of Medical Research and the Australian Centre for Vaccine Development, Herston, Queensland, Australia
| | - Fiona H. Amante
- Queensland Institute of Medical Research and the Australian Centre for Vaccine Development, Herston, Queensland, Australia
| | - Patrick T. Bunn
- Queensland Institute of Medical Research and the Australian Centre for Vaccine Development, Herston, Queensland, Australia
| | - Louise M. Randall
- Queensland Institute of Medical Research and the Australian Centre for Vaccine Development, Herston, Queensland, Australia
- Department of Pathobiology, School of Veterinary Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Klaus Pfeffer
- Institute of Medical Microbiology and Hospital Hygiene, University of Duesseldorf, Duesseldorf, Germany
| | - Stefanie Scheu
- Institute of Medical Microbiology and Hospital Hygiene, University of Duesseldorf, Duesseldorf, Germany
| | - Michael J. Hickey
- Centre for Inflammatory Diseases, Monash University, Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | | | - Carl Ware
- Infectious and Inflammatory Diseases Centre, Sanford|Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Geoff R. Hill
- Queensland Institute of Medical Research and the Australian Centre for Vaccine Development, Herston, Queensland, Australia
| | - Koji Tamada
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland, Baltimore, Maryland, Unites States of America
| | - Paul M. Kaye
- Hull York Medical School, Department of Biology, York University, York, United Kingdom
| | - Christian R. Engwerda
- Queensland Institute of Medical Research and the Australian Centre for Vaccine Development, Herston, Queensland, Australia
- * E-mail:
| |
Collapse
|
20
|
Malliga DE, Wagner D, Fahrleitner-Pammer A. The role of osteoprotegerin (OPG) receptor activator for nuclear factor kappaB ligand (RANKL) in cardiovascular pathology - a review. Wien Med Wochenschr 2011; 161:565-70. [PMID: 21870142 DOI: 10.1007/s10354-011-0022-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 06/29/2011] [Indexed: 10/17/2022]
Abstract
Initially described as key regulators in metabolic bone disease osteoprotegerin (OPG), receptor activator of nuclear factor kappa B (RANK) and RANK ligand (RANKL) have also been discriminated as regulators in immunologic function. Cardiovascular diseases (CVD) develop over many years in life and are often triggered by inflammatory processes within the vessel wall that lead to vascular remodeling. Recently some study groups have described OPG as a prognostic parameter for mortality and morbidity in cardiovascular patients.
Collapse
Affiliation(s)
- Daniela-Eugenia Malliga
- Division of Cardiac Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | | | | |
Collapse
|
21
|
Schramek D, Penninger JM. The many roles of RANKL-RANK signaling in bone, breast and cancer. ACTA ACUST UNITED AC 2011. [DOI: 10.1138/20110512] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
22
|
Receptor Activator for Nuclear Factor kappa B Ligand (RANKL) as an osteoimmune key regulator in bone physiology and pathology. Acta Histochem 2011; 113:73-81. [PMID: 19926120 DOI: 10.1016/j.acthis.2009.10.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Revised: 10/11/2009] [Accepted: 10/12/2009] [Indexed: 12/18/2022]
Abstract
The strength and integrity of the human skeleton depends on a delicate equilibrium between bone resorption and bone formation. Bone resorption is an elementary cellular activity in the modelling of the skeleton during growth and development. Later in life a most important physiological process in the skeleton is bone remodelling, which is locally initiated by resorption. During remodelling bone resorption is coupled to new bone formation that ensures renewal of bone with only minor local and temporary bone loss. Cells responsible for bone resorption and subsequent bone formation are the osteoclasts and osteoblasts, respectively. The osteoclast is derived from the pluripotent hematopoietic stem cell, which gives rise to a myeloid stem cell that can further differentiate into megakaryocytes, granulocytes, monocytes/macrophages and osteoclasts. The respective bone resorbing and forming actions of osteoclasts and osteoblasts are finely coupled, so that bone mass remains remarkably stable in a healthy adult. Imbalance between osteoclast and osteoblast activities can arise from a wide variety of hormonal changes or perturbations of inflammatory and growth factors resulting in postmenopausal osteoporosis, Paget's disease, lytic bone metastases, or rheumatoid arthritis, leading to increased bone resorption and crippling bone damage. In view of the critical role of osteoclasts in diverse pathology, there has been immense effort aimed at understanding the biology of this unique cell. The present review is focused on the current knowledge of the mechanisms that regulate the functional links between bone turnover and the immune system helping us to understand the main factors that lead to bone loss observed in osteoporosis, cancer and in rheumatoid arthritis. The aim of this review paper is to consider the key molecular interactions involved in the formation of osteoclast cells in normal and pathological conditions.
Collapse
|
23
|
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
|
24
|
Hofer EL, Labovsky V, La Russa V, Vallone VF, Honegger AE, Belloc CG, Wen HC, Bordenave RH, Bullorsky EO, Feldman L, Chasseing NA. Mesenchymal stromal cells, colony-forming unit fibroblasts, from bone marrow of untreated advanced breast and lung cancer patients suppress fibroblast colony formation from healthy marrow. Stem Cells Dev 2010; 19:359-70. [PMID: 19388812 DOI: 10.1089/scd.2008.0375] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
We have shown that bone marrow (BM) from untreated advanced lung and breast cancer patients (LCP and BCP) have a reduced number of colony-forming unit fibroblasts (CFU-Fs) or mesenchymal stem cells (MSCs). Factors that regulate the proliferation and differentiation of CFU-F are produced by the patients' BM microenvironment. We have now examined whether conditioned media (CM) from patients' CFU-F-derived stromal cells also inhibits the colony-forming efficiency (CFE) of CFU-F in primary cultures from healthy volunteers (HV)-BM. Thus the number and proliferation potential of HV-CFU-F were also found to be decreased and similar to colony numbers and colony size of patients' CFU-F. Stromal cells from both of these types of colonies appeared relatively larger and lacked the characteristic spindle morphology typically seen in healthy stromal cells. We developed an arbitrary mesenchymal stromal cell maturational index by taking three measures consisting of stromal cell surface area, longitudinal and horizontal axis. All stromal indices derived from HV-CFU-F grown in patients' CM were similar to those from stromal elements derived from patients' CFU-F. These indices were markedly higher than stromal indices typical of HV-CFU-F cultured in healthy CM or standard medium [alpha-medium plus 20% heat-inactivated fetal bovine serum (FBS)]. Patients' CM had increased concentrations of the CFU-F inhibitor, GM-CSF, and low levels of bFGF and Dkk-1, strong promoters of self-renewal of MSCs, compared to the levels quantified in CM from HV-CFU-F. Moreover, the majority of patients' MSCs were unresponsive in standard medium and healthy CM to give CFU-F, indicating that the majority of mesenchymal stromal cells from patients' CFU-F are locked in maturational arrest. These results show that alterations of GM-CSF, bFGF, and Dkk-1 are associated with deficient cloning and maturation arrest of CFU-F. Defective autocrine and paracrine mechanisms may be involved in the BM microenvironments of LCP and BCP.
Collapse
Affiliation(s)
- Erica Leonor Hofer
- Agencia Nacional de Promoción Científica, Tecnológica y de Innovación Productiva, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Randall LM, Engwerda CR. TNF family members and malaria: old observations, new insights and future directions. Exp Parasitol 2010; 126:326-31. [PMID: 20433831 DOI: 10.1016/j.exppara.2010.04.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Revised: 03/26/2010] [Accepted: 04/19/2010] [Indexed: 11/27/2022]
Abstract
Tumor necrosis factor (TNF) has long been recognized to promote malaria parasite killing, but also to contribute to the development of severe malaria disease. The precise molecular mechanisms that influence these different outcomes in malaria patients are not well understood, but the virulence and drug-resistance phenotype of malaria parasites and the genetic background and age of patients are likely to be important determinants. In the past few years, important roles for other TNF family members in host immune responses to malaria parasites and the induction of disease pathology have been discovered. In this review, we will summarize these more recent findings and highlight major gaps in our current knowledge. We will also discuss future research strategies that may allow us to better understand the sometimes subtle and intricate effects of TNF family molecules during malaria infection.
Collapse
Affiliation(s)
- Louise M Randall
- Immunology and Infection Laboratory, Queensland Institute of Medical Research and The Australian Centre for Vaccine Development, 300 Herston Road, Herston, Brisbane, Queensland, Qld 4006, Australia
| | | |
Collapse
|
26
|
Tyrovola JB, Perrea D, Halazonetis DJ, Dontas I, Vlachos IS, Makou M. Relation of soluble RANKL and osteoprotegerin levels in blood and gingival crevicular fluid to the degree of root resorption after orthodontic tooth movement. J Oral Sci 2010; 52:299-311. [DOI: 10.2334/josnusd.52.299] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
|
27
|
Tyrovola JB, Spyropoulos MN, Makou M, Perrea D. Root resorption and the OPG/RANKL/RANK system: a mini review. J Oral Sci 2009; 50:367-76. [PMID: 19106463 DOI: 10.2334/josnusd.50.367] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Odontoclastic root resorption is a significant clinical issue in relation to orthodontic tooth movement, and resorption of the roots of primary teeth is an intriguing biological phenomenon. The functional coordination of the OPG/RANKL/RANK system seems to contribute not only to alveolar remodeling, but also to resorption during orthodontic tooth movement and physiological root resorption. Serum OPG and s-RANKL are related to regulation of bone homeostasis by the OPG/RANKL/RANK system, and determination of their concentrations might be useful for predicting the rate of bone remodeling during orthodontic tooth movement, the net effect between bone remodeling and root resorption, and the degree of root resorption. It is therefore rational to speculate that a study of the levels of OPG and s-RANKL in blood and GCF, in relation to the degree of root resorption during orthodontic tooth movement, using healthy experimental animals and a carefully planned and organized experimental design, may be able to answer this intriguing question.
Collapse
Affiliation(s)
- Joanna B Tyrovola
- Department of Orthodontics, School of Dentistry, University of Athens, Athens, Greece.
| | | | | | | |
Collapse
|
28
|
Mohamed JA, DuPont HL, Jiang ZD, Flores J, Carlin LG, Belkind-Gerson J, Martinez-Sandoval FG, Guo D, White AC, Okhuysen PC. A single-nucleotide polymorphism in the gene encoding osteoprotegerin, an anti-inflammatory protein produced in response to infection with diarrheagenic Escherichia coli, is associated with an increased risk of nonsecretory bacterial diarrhea in North American travelers to Mexico. J Infect Dis 2009; 199:477-85. [PMID: 19128145 DOI: 10.1086/596319] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Osteoprotegerin (OPG), an immunoregulatory member of the TNF receptor superfamily, is expressed in inflamed intestinal mucosa. We investigated whether OPG is produced by intestinal epithelial cells and tested the hypothesis that single-nucleotide polymorphisms (SNPs) in the gene encoding OPG (TNFRSF11B) are associated with traveler's diarrhea (TD) among North American travelers to Mexico. METHODS OPG concentration was measured in the supernatants of T84 cells infected with various diarrheagenic Escherichia coli pathotypes. Genotyping was performed for 4 SNPs in the OPG gene for 968 North American travelers with or without TD. Stool samples from travelers with TD were evaluated for the presence of enteric pathogens. RESULTS T84 cells produced higher OPG levels in response to infection with various diarrheagenic E. coli pathotypes than with E. coli controls (P<.05). A SNP in the exon 1 region of the OPG gene (OPG+1181G>C) was associated with TD in white travelers who stayed in Mexico for >1 week during the summer (P=.009) and for TD due to nonsecretory pathogens (P=.001). CONCLUSIONS Our study suggests that OPG is secreted by intestinal epithelial cells in response to enteropathogens and that a polymorphism in the OPG gene is associated with an increased susceptibility to TD.
Collapse
Affiliation(s)
- Jamal A Mohamed
- Division of Infectious Diseases, The University of Texas Medical School, Center for Infectious Diseases, Baylor College of Medicine, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Paul S, Lee JC, Yeh LCC. A comparative study on BMP-induced osteoclastogenesis and osteoblastogenesis in primary cultures of adult rat bone marrow cells. Growth Factors 2009; 27:121-31. [PMID: 19180354 DOI: 10.1080/08977190802707324] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Bone remodeling is a process consisting of bone formation and resorption. The present study compared the relative osteoclastic and osteoblastic potency of bone morphogenetic proteins (BMP)-2, -4, -5, -6, and -7 in primary murine bone marrow cultures. All five BMPs stimulated, to varying degree, formation of tartrate-resistant acid phosphatase (TRAP)-positive multinucleated cells in a time- and protein concentration-dependent manner. The TRAP staining intensity correlated positively with the number of nuclei per TRAP-positive cell and the mRNA levels of colony-stimulating factor (CSF-1), receptor activator of nuclear factor kappaB ligand (RANKL), TRAP, and cathepsin K. Under osteogenic conditions, all five BMPs stimulated AP activity and mineralized bone nodule formation in a protein concentration-dependent manner in the same primary murine bone marrow cell culture system. These findings should be useful in designing treatment strategies for bone regeneration.
Collapse
Affiliation(s)
- Sangita Paul
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, USA
| | | | | |
Collapse
|
30
|
Leibbrandt A, Penninger JM. RANK/RANKL: regulators of immune responses and bone physiology. Ann N Y Acad Sci 2009; 1143:123-50. [PMID: 19076348 DOI: 10.1196/annals.1443.016] [Citation(s) in RCA: 292] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bone-related diseases, such as osteoporosis and rheumatoid arthritis, affect hundreds of millions of people worldwide and pose a tremendous burden to health care. By deepening our understanding of the molecular mechanisms of bone metabolism and bone turnover, it became possible over the past years to devise new and promising strategies for treating such diseases. In particular, three tumor necrosis factor (TNF) family molecules, the receptor activator of NF-kappaB (RANK), its ligand RANKL, and the decoy receptor of RANKL, osteoprotegerin (OPG), have attracted the attention of scientists and pharmaceutical companies alike. Genetic experiments revolving around these molecules established their pivotal role as central regulators of osteoclast development and osteoclast function. RANK-RANKL signaling not only activates a variety of downstream signaling pathways required for osteoclast development, but crosstalk with other signaling pathways also fine-tunes bone homeostasis both in normal physiology and disease. In addition, RANKL and RANK have essential roles in lymph node formation, establishment of the thymic microenvironment, and development of a lactating mammary gland during pregnancy. Consequently, novel drugs specifically targeting RANK, RANKL, and their signaling pathways in osteoclasts are expected to revolutionize the treatment of various ailments associated with bone loss, such as arthritis, periodontal disease, cancer metastases, and osteoporosis.
Collapse
Affiliation(s)
- Andreas Leibbrandt
- IMBA, Institute for Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | | |
Collapse
|
31
|
RANK(L) as a Key Target for Controlling Bone Loss. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 647:130-45. [DOI: 10.1007/978-0-387-89520-8_9] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
32
|
RANKL/RANK as key factors for osteoclast development and bone loss in arthropathies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 649:100-13. [PMID: 19731623 DOI: 10.1007/978-1-4419-0298-6_7] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Osteoporosis or rheumatoid arthritis are bone diseases affecting hundreds of millions of people worldwide and thus pose a tremendous burden to health care. Ground-breaking discoveries made in basic science over the last decade shed light on the molecular mechanisms of bone metabolism and bone turnover. Thereby, it became possible over the past years to devise new and promising strategies for treating such diseases. In particular, three molecules, the receptor activator of NF-kappaB (RANK), its ligand RANKL and the decoy receptor of RANKL, osteoprotegerin (OPG), have been a major focus of scientists and pharmaceutical companies alike, since experiments using mice in which these genes have been inactivated unanimously established their pivotal role as central regulators ofosteoclast function. RANK(L) signaling not only activates a variety of downstream signaling pathways required for osteoclast development, but crosstalk with other signaling pathways also fine-tunes bone homeostasis both in normal physiology and disease. Consequently, novel drugs specifically targeting RANK-RANKL and their signaling pathways in osteoclasts are expected to revolutionize the treatment ofvarious bone diseases, such as cancer metastases, osteoporosis, or arthropathies.
Collapse
|
33
|
Yasunori K, Masaaki T, Tetsuyuki N, Hayato K, Akira N. Reduction of urinary levels of pyridinoline and deoxypyridinoline and serum levels of soluble receptor activator of NF-kappaB ligand by etanercept in patients with rheumatoid arthritis. Clin Rheumatol 2008; 27:1093-101. [PMID: 18338203 DOI: 10.1007/s10067-008-0870-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Revised: 01/16/2008] [Accepted: 02/12/2008] [Indexed: 12/22/2022]
Abstract
The effects of soluble TNF-alpha receptor, etanercept, on bone metabolism were investigated in patients with rheumatoid arthritis (RA). Thirty RA patients were administered etanercept once or twice a week for more than 6 months. We evaluated clinical and laboratory parameters and measured urinary excretion levels of pyridinoline (PYD), deoxypyridinoline (DPD), cross-linked N-telopeptides of type I collagen (NTX), and serum levels of bone alkaline phosphatase (BAP), osteoprotegerin (OPG), and soluble receptor activator of NFkappaB ligand (sRANKL) at the baseline and at 3 and 6 months after initial treatment with etanercept. Etanercept treatment resulted in an improvement of symptoms due to RA and in a reduction of urinary excretion levels of PYD and DPD as well as serum sRANKL levels, with a significant difference at 6 months, and an increase of serum BAP levels at 3 and 6 months after the initial treatment with etanercept. Urinary NTX and serum OPG levels did not show a significant change at 3 and 6 months after the initial treatment, but serum OPG levels did show a reverse correlation with serum CRP levels, suggesting that the regulation of inflammation in RA may result in an induction of OPG production. Etanercept may have the ability to reduce the levels of bone resorption markers and to increase the levels of a bone formation marker while reducing sRANKL formation in RA patients.
Collapse
Affiliation(s)
- Kageyama Yasunori
- Department of Orthopaedic Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | | | | | | | | |
Collapse
|
34
|
Han X, Kawai T, Taubman MA. Interference with immune-cell-mediated bone resorption in periodontal disease. Periodontol 2000 2007; 45:76-94. [PMID: 17850450 DOI: 10.1111/j.1600-0757.2007.00215.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Xiaozhe Han
- Department of Immunology, The Forsyth Institute, Harvard Medical School, Harvard School of Dental Medicine, Boston, MA, USA
| | | | | |
Collapse
|
35
|
Kavurma MM, Bennett MR. Expression, regulation and function of trail in atherosclerosis. Biochem Pharmacol 2007; 75:1441-50. [PMID: 18061141 DOI: 10.1016/j.bcp.2007.10.020] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2007] [Revised: 10/03/2007] [Accepted: 10/11/2007] [Indexed: 01/22/2023]
Abstract
Atherosclerosis is a condition where vascular smooth muscle cells (VSMCs), inflammatory cells, lipids, cholesterol and cellular waste accumulate in the inner lining of an artery, producing a fibro-fatty plaque and resulting in the thickening of the arterial wall. The tumor necrosis factor (TNF) family of cytokines plays a major role in the progression of atherosclerosis. Recently, TNF-related apoptosis-inducing ligand (TRAIL), a member of the TNF superfamily, has been implicated in the development of atherosclerosis since it has been detected in normal and diseased atherosclerotic tissue. Not only is TRAIL involved in apoptosis and immune regulation, recent studies have provided a new function of TRAIL on vascular cells, such that TRAIL can promote endothelial cell (EC) and VSMCs migration and proliferation. In addition, TRAIL is implicated in regulating vascular tone. This review discusses our current understanding of TRAIL expression, regulation and function, and summarises the recent data implicating a role for TRAIL in atherosclerosis.
Collapse
Affiliation(s)
- Mary M Kavurma
- Centre for Vascular Research, The University of New South Wales, Kensington, Sydney, NSW 2052, Australia.
| | | |
Collapse
|
36
|
Vitovski S, Phillips JS, Sayers J, Croucher PI. Investigating the interaction between osteoprotegerin and receptor activator of NF-kappaB or tumor necrosis factor-related apoptosis-inducing ligand: evidence for a pivotal role for osteoprotegerin in regulating two distinct pathways. J Biol Chem 2007; 282:31601-9. [PMID: 17702740 DOI: 10.1074/jbc.m706078200] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Osteoprotegerin (OPG) binds the ligand for receptor activator of nuclear factor kappaB (RANKL) to prevent association with its receptor RANK and inhibit osteoclast-mediated bone resorption. OPG has been reported, recently, to inhibit tumor necrosis factor-related apoptosis-induced ligand (TRAIL)-induced tumor cell apoptosis. This raises the possibility that OPG may play a unique role in regulating these two signaling pathways. However, there are little data on the interactions between OPG, RANKL, and TRAIL, and the relative affinity of OPG for these two ligands is unknown. In the present study we examined the ability of OPG to bind native human TRAIL and RANKL under physiological conditions. Native TRAIL was expressed in Escherichia coli, purified to homogeneity, and shown to induce human myeloma cell apoptosis. OPG inhibited native TRAIL from binding the TRAILR1 at 37 degrees C in vitro. Similarly, OPG prevented RANKL from binding to RANK. TRAIL also prevented OPG-mediated inhibition of RANKL from binding RANK. The affinity of OPG for native TRAIL and RANKL at 37 degrees C was determined by plasmon surface resonance analysis. OPG had a binding affinity for TRAIL of 45 nM, whereas the affinity of OPG for RANKL was 23 nM. These data suggest that OPG can bind both RANKL and TRAIL and that the affinity of OPG for these two ligands is of a similar order of magnitude. Furthermore, OPG prevented TRAIL-mediated reductions in cell viability, whereas TRAIL inhibited OPG-mediated inhibition of osteoclastogenesis in vitro. This highlights the pivotal role of OPG in regulating the biology of both RANKL and TRAIL.
Collapse
Affiliation(s)
- Srdjan Vitovski
- Section of Musculoskeletal Science, University of Sheffield School of Medical and Biomedical Sciences, Beech Hill Road, Sheffield, Yorkshire S10 2RX, United Kingdom
| | | | | | | |
Collapse
|
37
|
Lau YS, Adamopoulos IE, Sabokbar A, Giele H, Gibbons CLMH, Athanasou NA. Cellular and humoral mechanisms of osteoclast formation in Ewing's sarcoma. Br J Cancer 2007; 96:1716-22. [PMID: 17533390 PMCID: PMC2359921 DOI: 10.1038/sj.bjc.6603774] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Cellular mechanisms that account for tumour osteolysis associated with Ewing's sarcoma are uncertain. Osteoclasts are marrow-derived multinucleated cells (MNCs) that effect tumour osteolysis. Osteoclasts are known to form from macrophages by both receptor activator for nuclear factor-κB (RANK) ligand (RANKL)-dependent and -independent mechanisms. In this study, our aim has been to determine whether tumour-associated macrophages (TAMs) isolated from Ewing's sarcoma are capable of differentiating into osteoclasts and to characterise the cellular and humoral mechanisms whereby this occurs. Tumour-associated macrophages were isolated from two Ewing's sarcomas and cultured on both coverslips and dentine slices for up to 21 days with soluble RANKL and macrophage colony stimulating factor (M-CSF). Osteoclast formation from TAMs (CD14+) was evidenced by the formation of tartrate-resistant acid phosphatase (TRAP) and vitronectin receptor (VNR)-positive MNCs, which were capable of carrying out lacunar resorption. This osteoclast formation was inhibited by the addition of bisphosphonates. Both Ewing's sarcoma-derived fibroblasts and some bone stromal cells expressed RANKL and supported osteoclast formation by a contact-dependent mechanism. We also found that osteoclast differentiation occurred when Ewing's TAMs were cultured with tumour necrosis factor (TNF)-α in the presence of M-CSF and that TC71 Ewing's sarcoma cells stimulated osteoclast formation through the release of a soluble factor, the action of which was abolished by an antibody to TNF-α. These results indicate that TAMs in Ewing's sarcoma are capable of osteoclast differentiation by both RANKL-dependent and TNF-α-dependent mechanisms and that Ewing's sarcoma cells produce osteoclastogenic factor(s). Our findings suggest that anti-resorptive and anti-osteoclastogenic therapies may be useful in inhibiting the osteolysis of Ewing's sarcoma.
Collapse
Affiliation(s)
- Y S Lau
- Department of Pathology, Nuffield Department of Orthopaedic Surgery, Nuffield Orthopaedic Centre, University of Oxford, Oxford OX3 7LD, UK
| | - I E Adamopoulos
- Department of Pathology, Nuffield Department of Orthopaedic Surgery, Nuffield Orthopaedic Centre, University of Oxford, Oxford OX3 7LD, UK
| | - A Sabokbar
- Department of Pathology, Nuffield Department of Orthopaedic Surgery, Nuffield Orthopaedic Centre, University of Oxford, Oxford OX3 7LD, UK
| | - H Giele
- Nuffield Department of Surgery, Nuffield Orthopaedic Centre, University of Oxford, Oxford OX3 7LD, UK
| | - C L M H Gibbons
- Nuffield Department of Surgery, Nuffield Orthopaedic Centre, University of Oxford, Oxford OX3 7LD, UK
| | - N A Athanasou
- Department of Pathology, Nuffield Department of Orthopaedic Surgery, Nuffield Orthopaedic Centre, University of Oxford, Oxford OX3 7LD, UK
- E-mail:
| |
Collapse
|
38
|
Lin JM, Callon KE, Lin CQ, Bava U, Zheng MH, Reid IR, Cornish J. Alteration of bone cell function by RANKL and OPG in different in vitro models. Eur J Clin Invest 2007; 37:407-15. [PMID: 17461987 DOI: 10.1111/j.1365-2362.2007.01800.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Receptor activator of nuclear factor-kappaB ligand (RANKL) and osteoprotegerin (OPG) are well-documented potent regulators of osteoclast development. However, their effects in mature bone cells and in organ cultures have not been well studied. It is uncertain whether their activities in different experimental models are comparable. MATERIALS AND METHODS RANKL and OPG were evaluated for their activities in mouse calvarial organ cultures, mouse bone marrow cultures, isolated rat mature osteoclast assays and rat primary osteoblast cultures. Results In murine calvarial organ culture, both muRANKL (> or = 10 ng mL(-1)) and rRANKL (> or = 100 ng mL(-1)) significantly stimulated (45)Ca release, while OPG (> or = 50 ng mL(-1)) was an inhibitor of bone resorption. Meanwhile, [(3)H]-thymidine incorporation in this assay was also modulated (indicating proliferation increases in the osteoblast lineage of cells) although these peptides had no direct effect on [(3)H]-thymidine incorporation in isolated osteoblast assays. In mouse bone marrow cultures, muRANKL (> or = 1 ng mL(-1)) and rRANKL (> or = 5 ng mL(-1)) significantly stimulated osteoclastogenesis. The number of nuclei per osteoclast was also significantly increased. OPG strongly inhibited this index, with over 90% suppression at 1 ng mL(-1). Both muRANKL (10 ng mL(-1)) and rRANKL (100 ng mL(-1)) stimulated, while OPG (10 ng mL(-1)) inhibited osteoclast activity in isolated mature osteoclast assays. CONCLUSION The current study demonstrated that bone resorption modulated by RANKL and OPG, in murine calvarial organ culture, leads to changes in osteoblast proliferation, suggesting a feedback mechanism from osteoclasts to osteoblasts. In addition, it was found that RANKL and OPG have more potent effects on osteoclastogenesis than on the activity of mature osteoclasts.
Collapse
Affiliation(s)
- J M Lin
- University of Auckland, Auckland, New Zealand
| | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Osteoimmunology is an interdisciplinary research field combining the exciting fields of osteology and immunology. An observation that contributed enormously to the emergence of osteoimmunology was the accelerated bone loss caused by inflammatory diseases such as rheumatoid arthritis. Receptor activator of nuclear factor kappaB ligand (RANKL), which is the main regulator of osteoclastogenesis, was found to be the primary culprit responsible for the enhanced activation of osteoclasts: activated T cells directly and indirectly increased the expression of RANKL, and thereby promoted osteoclastic activity. Excessive bone loss is not only present in inflammatory diseases but also in autoimmune diseases and cancer. Furthermore, there is accumulating evidence that the very prevalent skeletal disorder osteoporosis is associated with alterations in the immune system. Meanwhile, numerous connections have been discovered in osteoimmunology beyond merely the actions of RANKL. These include the importance of osteoblasts in the maintenance of the hematopoietic stem cell niche and in lymphocyte development as well as the functions of immune cells participating in osteoblast and osteoclast development. Furthermore, research is being done investigating cytokines, chemokines, transcription factors and co-stimulatory molecules which are shared by both systems. Research in osteoimmunology promises the discovery of new strategies and the development of innovative therapeutics to cure or alleviate bone loss in inflammatory and autoimmune diseases as well as in osteoporosis. This review gives an introduction to bone remodeling and the cells governing that process and summarizes the most recent discoveries in the interdisciplinary field of osteoimmunology. Furthermore, an alternative large animal model will be discussed and the pathophysiological alterations of the immune system in osteoporosis will be highlighted.
Collapse
Affiliation(s)
- Martina Rauner
- Ludwig Boltzmann Institute of Aging Research, Vienna, Austria
| | | | | |
Collapse
|
40
|
Fábrega E, Orive A, García-Unzueta M, Amado JA, Casafont F, Pons-Romero F. Osteoprotegerin and receptor activator of nuclear factor-kappaB ligand system in the early post-operative period of liver transplantation. Clin Transplant 2006; 20:383-8. [PMID: 16824158 DOI: 10.1111/j.1399-0012.2006.00497.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND The precise mechanism that leads to accelerated bone resorption in the early post-transplant period remains unclear. Recent data suggest that osteoprotegerin (OPG) and its ligand receptor activator of nuclear factor-kappaB ligand (RANKL) constitute a novel cytokine system that can influence the function of both bone and immune cells. The aim of our study was to assess OPG and RANKL concentrations in the early post-operative period of liver transplantation. METHODS Serum OPG and RANKL levels were measured in 30 patients who underwent liver transplantation at 1, 7 and 14 d post-operatively. These values were compared with 22 age- and sex-matched healthy controls. Plasma sodium, creatinine, aspartate-aminotransferase, alanine-amino transferase, gamma-glutamyl transferase, alkaline phosphatase, bilirubin, albumin, prothrombin time, tacrolimus and cyclosporine levels were measured in each patient. RESULTS We found a significant increase in OPG levels in the early post-operative period compared with the control group: day 1 (10.42 pmol/L, range 3.80-17.50 vs. 3.91 pmol/L, range 1.20-6.60; p = 0.0001), day 7 (6.90 pmol/L, range 3.00-15.30 vs. 3.91 pmol/L, range 1.20-6.60; p = 0.0001) and day 14 (5.76 pmol/L, range 2.60-10.70 vs. 3.91 pmol/L, range 1.20-6.60; p = 0.001). Similarly, serum RANKL levels were significantly higher than in the control group in this period, day 1 (0.123 pmol/L, range 0.010-0.420 vs. 0.054 pmol/L, range 0.010-0.300; p = 0.02), day 7 (0.236 pmol/L, range 0.010-0.720 vs. 0.054 pmol/L, range 0.010-0.300; p = 0.0004) and day 14 (0.137 pmol/L, range 0.010-0.520 vs. 0.054 pmol/L, range 0.010-0.300; p = 0.007). No correlation was found between OPG levels and RANKL, ischemic times, liver function tests, albumin, sodium or creatinine concentrations and tacrolimus or cyclosporine levels. CONCLUSIONS A significant amount of OPG and RANKL is released in the early post-transplant period of liver transplantation. This might be explained by an activation of the immune system caused by the allograft. Therefore, the RANKL/OPG system may be involved in the pathophysiological evolution of transplantation osteoporosis.
Collapse
Affiliation(s)
- Emilio Fábrega
- Gastroenterology and Hepatology Unit, University Hospital Marqués de Valdecilla, Faculty of Medicine, UC Santander, Spain
| | | | | | | | | | | |
Collapse
|
41
|
Grzegorzewska AE, Młot M. Serum markers of bone turnover in dialyzed patients separated according to age. Int Urol Nephrol 2006; 38:311-6. [PMID: 16868703 DOI: 10.1007/s11255-006-0079-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2005] [Indexed: 10/24/2022]
Abstract
BACKGROUND Bone metabolism changes with aging in healthy population. Our aim was to determine serum markers of bone turnover in dialysis patients separated according to age. METHODS Peritoneal dialysis (PD) or hemodialysis (HD) patients were divided into two groups. Group I (n = 30) consisted of patients older than 65 years. Patients at the age less or equal 65 years were included in group II (n = 37). In all patients we determined serum concentration of intact parathyroid hormone (iPTH), cyclase activating parathyroid hormone (CAP), osteoprotegrin (OPG) and osteoprotegrin ligand (OPGL). Cyclase inactive parathyroid hormone (CIP) was calculated. Healthy volunteers (n = 13) at the age of 42.1 years (range 23.5-70.9 years) served as controls. RESULTS When results of dialysis patients were adjusted to gender, dialysis modality and duration, group I revealed significantly lower iPTH (113.0, 10.3-606.3 pg/ml), CAP (70.0, 6.5-442.6 pg/ml) and CIP (53.3, 3.3-214.4 pg/ml) than group II (310.6, 13.7-1266.9 pg/ml for iPTH; 205.0, 9.3-887.9 pg/ml for CAP; 76.0, 2.4-399.0 pg/ml for CIP), but this group showed significantly higher serum OPG (7.39, 1.52-15.84 pg/ml) than group II (5.45, 0.95-16.47 pg/ml) and controls (2.17, 1.05-3.95 pg/ml). Only patients of group II showed significantly higher iPTH, CAP and CIP than controls (34.9, 18.9-76.9 pg/ml; 24.3, 11.2-42.6 pg/ml, 12.0, 1.0-34.2 pg/ml, respectively for iPTH, CAP and CIP). CONCLUSION Our results suggest that age over 65 years is a risk factor for low bone turnover in dialyzed patients. An increase in serum OPG probably reflects a paracrine mechanism of bone cells to compensate for age dependent bone loss.
Collapse
Affiliation(s)
- Alicja E Grzegorzewska
- Department of Nephrology, Transplantology and Internal Diseases, Karol Marcinkowski University of Medical Sciences, Poznań, Poland.
| | | |
Collapse
|
42
|
Kohanawa M. A Regulatory Effect of the Balance between TNF-α and IL-6 in the Granulomatous and Inflammatory Response toRhodococcus aurantiacusInfection in Mice. THE JOURNAL OF IMMUNOLOGY 2006; 177:642-50. [PMID: 16785562 DOI: 10.4049/jimmunol.177.1.642] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
After i.v. inoculation with Rhodococcus aurantiacus, wild-type (WT) mice develop nonnecrotic, epithelioid granulomas. Because a high level of TNF-alpha is observed during the initial phase postinfection, we examined the extent to which TNF-alpha contributes to granulomatous inflammation using TNF-alpha gene-deficient (TNF-alpha(-/-)) mice. Despite a lack of R. aurantiacus proliferation, TNF-alpha(-/-) mice displayed high mortality rates within 5 days postinfection, as well as a high level of IL-6 in their spleens. Histological examination showed an absence of granuloma formation in TNF-alpha(-/-) mice. Pretreatment of TNF-alpha(-/-) mice with rTNF-alpha failed to restore this granuloma formation but accelerated bacterial removal and cellular recruitment. This rTNF-alpha administration also attenuated IL-6 production, resulting in increased survival rates of TNF-alpha(-/-) mice. Heat-killed R. aurantiacus induced in vitro enhanced mRNA expression and production of IL-6 in macrophages and DCs from TNF-alpha(-/-) mice when compared with WT controls, and treatment of TNF-alpha(-/-) mouse cells with rTNF-alpha decreased the IL-6 secretion. Moreover, anti-TNF-alpha or anti-IL-6 treatment increased IL-6 or TNF-alpha production by WT mouse cells, respectively. These data suggest that the production of TNF-alpha and IL-6 can be negatively regulated by each other. Administration of rIFN-gamma to TNF-alpha(-/-) mice caused immature granulomas in livers, and treatment with both rTNF-alpha and rIFN-gamma led to the formation of mature granulomas. Overall, TNF-alpha appears crucial for bacterial clearance, cellular recruitment, and granuloma formation. The balance between TNF-alpha and IL-6 during the early phase of infection controls the development of the inflammatory response to R. aurantiacus infection.
Collapse
|
43
|
Gannagé-Yared MH, Fares F, Semaan M, Khalife S, Jambart S. Circulating osteoprotegerin is correlated with lipid profile, insulin sensitivity, adiponectin and sex steroids in an ageing male population. Clin Endocrinol (Oxf) 2006; 64:652-8. [PMID: 16712667 DOI: 10.1111/j.1365-2265.2006.02522.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE The relationship between osteoprotegerin (OPG) and lipid profile, insulin sensitivity, adipocytokines and sex steroids has been poorly studied and subject to controversy. The purpose of this study was to look at the correlates of OPG in an elderly male population. DESIGN One hundred and fifty-one nondiabetic, elderly Lebanese men (age range 50-83) were recruited in this cross-sectional study based on voluntary enrolment. MEASUREMENTS In all the subjects, serum OPG levels were measured and related to clinical parameters (age, waist, body mass index (BMI), systolic and diastolic blood pressure), as well as to metabolic and hormonal parameters. The following fasting laboratory measurements were performed: plasma glucose and insulin levels, total cholesterol, triglycerides and HDL cholesterol, adiponectin, leptin, as well as sex steroids (testosterone, SHBG, free androgen index, ooestradiol, DHEAS), GH and IGF-1. QUICKI index was calculated as a measure of insulin sensitivity. RESULTS OPG levels were significantly correlated with age (r = 0.28, P < 0.0001) but not with BMI, waist, systolic or diastolic blood pressure. There was a trend towards higher OPG levels in subjects without, compared to subjects with the metabolic syndrome (3.58 +/- 1.28 vs. 3.26 +/- 1.04 pmol/l, P = 0.09). OPG was negatively correlated with fasting glucose and triglyceride levels (r = -0.18, P = 0.031 and r = -0.19, P = 0.02, respectively) and positively correlated with the QUICKI index (r = 0.17, P = 0.033), HDL cholesterol (r = 0.21, P = 0.009) and adiponectin levels (r = 0.27, P = 0.001). No significant correlations were reported with total or LDL cholesterol levels and with leptin levels. After adjustment for age, OPG is still correlated with triglycerides (r = -0.19, P = 0.02), glucose (r = -0.21, P = 0.011) and adiponectin (r = 0.19, P = 0.02). Finally, OPG was positively associated with SHBG (r = 0.31, P < 0.001) and negatively associated with free androgen index (r =-0.346, P < 0.001); both correlations persisted after adjustment for age (r = 0.21, P = 0.009 and r = -0.23, P = 0.005, respectively). No significant correlation was found between OPG and oestradiol levels while a weak negative correlation was demonstrated with DHEAS (r = -0.18, P = 0.025). Also, no significant correlation was found between OPG and GH or IGF-1 values. In a multiple regression analysis with a stepwise model, the main determinants of OPG were free androgen index and adiponectin (P < 0.0001 and P = 0.015, respectively). CONCLUSION Our results show that circulating OPG levels are favourably associated with some components of the metabolic syndrome. Also, for the first time, an association between OPG and adiponectin is described. Finally, the negative correlation we found between OPG and free androgen index may suggest a potential role of OPG in the increase in cardiovascular disease related to ageing and sex steroid deficiency.
Collapse
|
44
|
Abstract
The role of the immune system in the development of senile osteoporosis, which arises primarily through the effects of estrogen deficiency and secondary hyperparathyroidism, is slowly being unraveled. This review focuses on our current understanding of how the components of this complex-interlinked system are regulated and how these fit with previous models of senile and postmenopausal osteoporosis. There is certainly substantial evidence that bone remodeling is a tightly regulated, finely balanced process influenced by subtle changes in proinflammatory and inhibitory cytokines as well as hormones and cellular components that act primarily but not exclusively through the receptor activator of nuclear factor-kappaB (RANK)/RANK ligand/osteoprotegerin system. In addition, an acute or chronic imbalance in the system due to infection or inflammation could contribute to systemic (or local) bone loss and increase the risk of fracture. Although significant progress has been made, there remains much to be done in unraveling this complex interaction between the immune system and bone.
Collapse
Affiliation(s)
- Jackie A Clowes
- Mayo Clinic College of Medicine, Endocrine Research Unit, St Mary's Hospital, Rochester, MN 55905, USA.
| | | | | |
Collapse
|
45
|
Nardelli B, Zaritskaya L, McAuliffe W, Ni Y, Lincoln C, Cho YH, Birse CE, Halpern W, Ullrich S, Moore PA. Osteostat/tumor necrosis factor superfamily 18 inhibits osteoclastogenesis and is selectively expressed by vascular endothelial cells. Endocrinology 2006; 147:70-8. [PMID: 16179414 DOI: 10.1210/en.2005-0518] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Vascular endothelial cells (EC) participate in the process of bone formation through the production of factors regulating osteoclast differentiation and function. In this study, we report the selective expression in primary human microvascular EC of Osteostat/TNF superfamily 18, a ligand of the TNF superfamily. Osteostat protein is detectable in human microvascular EC and is highly up-regulated by IFN-alpha and IFN-beta. Moreover, an anti-Osteostat antibody strongly binds to the vascular endothelium in human tissues, demonstrating that the protein is present in the EC layers surrounding blood vessels. Functional in vitro assays were used to define Osteostat involvement in osteoclastogenesis. Both recombinant and membrane-bound Osteostat inhibit differentiation of osteoclasts from monocytic precursor cells. Osteostat suppresses the early stage of osteoclastogenesis via inhibition of macrophage colony-stimulating factor-induced receptor activator of NF-kappaB (RANK) expression in the osteoclast precursor cells. This effect appears to be specific for the differentiation pathway of the osteoclast lineage, because Osteostat does not inhibit lipopolysaccharide-induced RANK expression in monocytes and dendritic cells, or activation-induced RANK expression in T cells. These findings demonstrate that Osteostat is a novel regulator of osteoclast generation and substantiate the major role played by the endothelium in bone physiology.
Collapse
|
46
|
Wan C, He Q, Li G. Osteoclastogenesis in the nonadherent cell population of human bone marrow is inhibited by rhBMP-2 alone or together with rhVEGF. J Orthop Res 2006; 24:29-36. [PMID: 16419966 DOI: 10.1002/jor.20010] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
During bone development and repair, angiogenesis, osteogenesis, and bone remodeling are closely associated processes that share some common mediators. In the present study nonadherent human bone marrow mononuclear cells under the induction of sRANKL and M-CSF, differentiated into osteoclasts with TRAP-positive staining, VNR expression, and Ca-P resorptive activity. The effects of various combinations of rhBMP-2 (0, 3, 30, and 300 ng/mL) and rhVEGF (0 and 25 ng/mL) on osteoclastogenesis potentials were examined in this experimental system. The percentages of TRAP-positive multiple nucleated cells represent osteoclast differentiation potential, and the percentages of resorptive areas in the Ca-P coated plates resemble osteoclast resorption capability. The presence of rhBMP-2 at 30 and 300 ng/mL showed inhibitory effects on osteoclast differentiation and their resorptive capability in the human osteoclast culture system. rhVEGF (25 ng/mL) enhanced the resorptive function of osteoclast whenever it was used alone or combined with 3 ng/mL rhBMP-2. However, rhVEGF-induced resorptive function was inhibited by 30 ng/mL and 300 ng/mL rhBMP-2 in a dose-dependent manner. Statistical analysis demonstrated that an interactive effect exists between rhBMP-2 and rhVEGF on human osteoclastogenesis. These findings suggested that an interactive regulation may exist between BMPs and VEGF signaling pathways during osteoclastogenesis; exact mechanisms are yet to be elucidated.
Collapse
Affiliation(s)
- Chao Wan
- Department of Trauma and Orthopaedic Surgery, Centre for Cancer Research and Cell Biology, School of Medicine, Queen's University Belfast, Musgrave Park Hospital, Belfast, BT9 7JB, UK
| | | | | |
Collapse
|
47
|
Shibata H, Abe M, Hiura K, Wilde J, Moriyama K, Sano T, Kitazoe KI, Hashimoto T, Ozaki S, Wakatsuki S, Kido S, Inoue D, Matsumoto T. Malignant B-Lymphoid Cells with Bone Lesions Express Receptor Activator of Nuclear Factor-κB Ligand and Vascular Endothelial Growth Factor to Enhance Osteoclastogenesis. Clin Cancer Res 2005; 11:6109-15. [PMID: 16144909 DOI: 10.1158/1078-0432.ccr-05-0181] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Receptor activator of nuclear factor-kappaB ligand (RANKL) is a key mediator of osteoclastogenesis. Because certain types of tumor cells aberrantly express RANKL, and because bone destruction also develops in B-cell lymphomas of bone origin, we investigated RANKL expression and the mechanisms of osteoclastogenesis in B-lymphoid neoplasms. EXPERIMENTAL DESIGN AND RESULTS Immunohistochemistry of bone specimens resected from patients with primary B-cell lymphoma of bone with bone destruction revealed that lymphoma cells express RANKL as well as vascular endothelial cell growth factor (VEGF). The tumor cells isolated from the bone specimens enhanced osteoclastogenesis in vitro. In contrast, B-cell lymphoma infiltrating to the bone marrow without bone destruction did not express RANKL. Both RANKL and VEGF were expressed by a portion of B-lymphoid cell lines, including Daudi and IM-9. These RANKL-expressing tumor cells enhanced osteoclastogenesis from RAW264.7 cells and human monocyte-derived preosteoclasts in the absence of stromal cells/osteoblasts in a RANKL-dependent manner. Furthermore, conditioned media from Daudi cells enhanced transmigration of preosteoclasts that was inhibited by anti-VEGF antibody, suggesting that tumor cell-derived VEGF mediates recruitment of osteoclast precursors. Moreover, cocultures of B-lymphoid cell lines with osteoclasts enhanced the growth of B-lymphoid cells. CONCLUSIONS Some malignant B cells aberrantly express functional RANKL as well as VEGF to enhance osteoclastogenesis. The coexpression of RANKL and VEGF may also contribute to the close cellular interactions with osteoclastic cells, thereby forming a vicious cycle between osteoclastic bone destruction and tumor expansion in bone.
Collapse
Affiliation(s)
- Hironobu Shibata
- Department of Medicine and Bioregulatory Sciences, University of Tokushima Graduate School of Health Biosciences, Tokushima, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Arko B, Prezelj J, Kocijancic A, Komel R, Marc J. Association of the osteoprotegerin gene polymorphisms with bone mineral density in postmenopausal women. Maturitas 2005; 51:270-9. [PMID: 15978970 DOI: 10.1016/j.maturitas.2004.08.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2004] [Revised: 08/11/2004] [Accepted: 08/12/2004] [Indexed: 11/17/2022]
Abstract
OBJECTIVES Osteoprotegerin (OPG) is a recently discovered member of the tumour necrosis factor receptor superfamily. It plays a crucial role in the control of bone resorption and its gene could therefore be a good candidate gene for osteoporosis. The aim of our work was to find polymorphisms in the OPG gene and to investigate their possible contribution to the genetic susceptibility to osteoporosis by testing for their association with bone mineral density (BMD). METHODS The whole OPG gene coding region was screened for the presence of polymorphisms in a group of 60 osteoporotic women by single-strand conformation polymorphism analysis (SSCP) approach. Association of the discovered polymorphisms with bone mineral density was investigated in 136 Slovenian postmenopausal women. RESULTS We detected eight OPG gene polymorphisms that were confirmed by direct DNA sequencing, deletion 4752_4753delCT and nucleotide substitutions 1181G>C, 1217C>T, 1284G>A, 4501C>T, 6893A>G, 6950A>C and 8738T>A. Nucleotide substitutions 1284G>A and 8738T>A have not been previously described. Polymorphisms 4752_4753delCT, 6893A>G and 6950A>C were in complete linkage and the same was true for 1217C>T and 4501C>T. The association with BMD was found only for polymorphism 1181G>C. Subjects with genotype 1181GG had significantly lower lumbar spine BMD than subjects displaying 1181GC. CONCLUSIONS By our approach we detected eight polymorphisms in the OPG gene. According to our analysis polymorphism 1181G>C is associated with BMD and could therefore be considered as an element of genetic susceptibility to osteoporosis.
Collapse
Affiliation(s)
- Barbara Arko
- Department of Clinical Biochemistry, Faculty of Pharmacy, Askerceva 7, SI-1000 Ljubljana, Slovenia
| | | | | | | | | |
Collapse
|
49
|
Fábrega E, Orive A, García-Suarez C, García-Unzueta M, Antonio Amado J, Pons-Romero F. Osteoprotegerin and RANKL in alcoholic liver cirrhosis. Liver Int 2005; 25:305-10. [PMID: 15780054 DOI: 10.1111/j.1478-3231.2005.01073.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND/AIMS The mechanisms leading to osteoporosis in alcoholic liver disease remain poorly understood. Recently identified soluble circulating osteoprotegerin (OPG), is the osteoclastogenesis inhibitory factor. It acts as a decoy receptor for osteoclast activating factor, receptor activator of nuclear factor-kappaB ligand (RANKL), and impairs osteoclast function. The aim of our study was to investigate the OPG/RANKL system in alcoholic cirrhotic patients and their correlation with biochemical marker of bone turnover. PATIENTS AND METHODS Serum OPG, RANKL, osteocalcin (OC), C-terminal cross-linking telopeptide of type I collagen (CTX-I), bone alkaline phosphatase activity (bALP), and urinary hydroxyproline were measured in 30 patients with alcoholic cirrhosis, and in 20 age- and sex-matched healthy controls. RESULTS OPG levels were significantly increased in patients with alcoholic cirrhosis compared with healthy subjects (5.9 pmol/l, range 2.7-9.0 vs 4.1 pmol/l, range 1.2-6.6; P < 0.001). RANKL levels were significantly higher in patients with cirrhosis (0.48 pmol/l, range 0.01-1.34) than in healthy subjects (0.11 pmol/l, range 0.01-0.90). There was a positive correlation between serum OPG and RANKL (r = 0.37; P < 0.001), bALP (r = 0.66; P < 0.001) and urinary hydroxyproline (r = 0.51; P < 0.05) but not with OC and CTX-I. CONCLUSIONS OPG might partly represent a compensating mechanism to the negative balance of bone remodelling in patients with alcoholic cirrhosis.
Collapse
Affiliation(s)
- Emilio Fábrega
- Gastroenterology and Hepatology Unit, Faculty of Medicine, University Hospital Marqués de Valdecilla, U.C. Santander, Avenida Valdecilla s/n, 39008 Santander, Cantabria, Spain.
| | | | | | | | | | | |
Collapse
|
50
|
Oh ES, Rhee EJ, Oh KW, Lee WY, Baek KH, Yoon KH, Kang MI, Yun EJ, Park CY, Choi MG, Yoo HJ, Park SW. Circulating osteoprotegerin levels are associated with age, waist-to-hip ratio, serum total cholesterol, and low-density lipoprotein cholesterol levels in healthy Korean women. Metabolism 2005; 54:49-54. [PMID: 15562379 DOI: 10.1016/j.metabol.2004.08.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Osteoprotegerin (OPG) is a recently identified cytokine that acts as a decoy receptor for the receptor activator of nuclear factor I masculine B ligand. Osteoprotegerin has been shown to be an important inhibitor of osteoclastogenesis and arterial calcification in animal models. Recently, OPG has been proposed as a link molecule between osteoporosis and arterial calcification, but the relationship between circulating OPG levels and cardiovascular disease in human populations is unclear. Thus, the aim of this study was to investigate the relationship between circulating OPG levels and cardiovascular risk factors in healthy Korean women. The subjects were 286 women aged 37 to 73 (mean +/- SD, 51.5 +/- 6.9 years). We examined blood pressure, body mass index, and waist-to-hip ratio. Serum concentrations of OPG were determined by enzyme-linked immunosorbent assay. Fasting plasma glucose levels, serum lipid profiles, insulin levels, and serum follicle-stimulating hormone (FSH) levels were determined by standard methods and homeostasis model assessment of insulin resistance was calculated. We observed a significant association between serum OPG levels and age, waist-to-hip ratio, total cholesterol, low-density lipoprotein cholesterol, and FSH levels (P < .05). Among the metabolic components, the older, obese, and hypercholsterolemic subjects had higher serum OPG levels (P < .05). However, no significant relationship was found between serum OPG levels and blood pressure and fasting plasma glucose levels. We found that mean serum OPG levels were about 11% greater in postmenopausal women (mean +/- SD, 1358.5 +/- 380.0 pg/mL) than in premenopausal women (mean +/- SD, 1228.8 +/- 407.7 pg/mL, P < .001). In multiple regression analysis with OPG as the dependent variable, serum FSH and low-density lipoprotein cholesterol levels were the significant predictor for serum OPG level (R(2) = 0.051, P < .05). In conclusion, our results show that circulating OPG levels are partly associated with cardiovascular risk factors and menopausal status in healthy Korean women. Out findings suggest that OPG may be an important paracrine factor of cardiovascular disease in the female population.
Collapse
Affiliation(s)
- Eun Sook Oh
- Department of Internal Medicine, Miz Medi Hospital, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|