51
|
Andreev D, Liu M, Weidner D, Kachler K, Faas M, Grüneboom A, Schlötzer-Schrehardt U, Muñoz LE, Steffen U, Grötsch B, Killy B, Krönke G, Luebke AM, Niemeier A, Wehrhan F, Lang R, Schett G, Bozec A. Osteocyte necrosis triggers osteoclast-mediated bone loss through macrophage-inducible C-type lectin. J Clin Invest 2021; 130:4811-4830. [PMID: 32773408 DOI: 10.1172/jci134214] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 06/03/2020] [Indexed: 12/17/2022] Open
Abstract
Although the control of bone-resorbing osteoclasts through osteocyte-derived RANKL is well defined, little is known about the regulation of osteoclasts by osteocyte death. Indeed, several skeletal diseases, such as bone fracture, osteonecrosis, and inflammation are characterized by excessive osteocyte death. Herein we show that osteoclasts sense damage-associated molecular patterns (DAMPs) released by necrotic osteocytes via macrophage-inducible C-type lectin (Mincle), which induced their differentiation and triggered bone loss. Osteoclasts showed robust Mincle expression upon exposure to necrotic osteocytes in vitro and in vivo. RNA sequencing and metabolic analyses demonstrated that Mincle activation triggers osteoclastogenesis via ITAM-based calcium signaling pathways, skewing osteoclast metabolism toward oxidative phosphorylation. Deletion of Mincle in vivo effectively blocked the activation of osteoclasts after induction of osteocyte death, improved fracture repair, and attenuated inflammation-mediated bone loss. Furthermore, in patients with osteonecrosis, Mincle was highly expressed at skeletal sites of osteocyte death and correlated with strong osteoclastic activity. Taken together, these data point to what we believe is a novel DAMP-mediated process that allows osteoclast activation and bone loss in the context of osteocyte death.
Collapse
Affiliation(s)
- Darja Andreev
- Department of Internal Medicine 3 - Rheumatology and Immunology.,Deutsches Zentrum für Immuntherapie (DZI), and
| | - Mengdan Liu
- Department of Internal Medicine 3 - Rheumatology and Immunology.,Deutsches Zentrum für Immuntherapie (DZI), and
| | - Daniela Weidner
- Department of Internal Medicine 3 - Rheumatology and Immunology.,Deutsches Zentrum für Immuntherapie (DZI), and
| | - Katerina Kachler
- Department of Internal Medicine 3 - Rheumatology and Immunology.,Deutsches Zentrum für Immuntherapie (DZI), and
| | - Maria Faas
- Department of Internal Medicine 3 - Rheumatology and Immunology.,Deutsches Zentrum für Immuntherapie (DZI), and
| | - Anika Grüneboom
- Department of Internal Medicine 3 - Rheumatology and Immunology.,Deutsches Zentrum für Immuntherapie (DZI), and
| | | | - Luis E Muñoz
- Department of Internal Medicine 3 - Rheumatology and Immunology.,Deutsches Zentrum für Immuntherapie (DZI), and
| | - Ulrike Steffen
- Department of Internal Medicine 3 - Rheumatology and Immunology.,Deutsches Zentrum für Immuntherapie (DZI), and
| | - Bettina Grötsch
- Department of Internal Medicine 3 - Rheumatology and Immunology.,Deutsches Zentrum für Immuntherapie (DZI), and
| | - Barbara Killy
- Institute of Clinical Microbiology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3 - Rheumatology and Immunology.,Deutsches Zentrum für Immuntherapie (DZI), and
| | | | - Andreas Niemeier
- Department of Orthopaedics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Falk Wehrhan
- Department of Oral and Maxillofacial Surgery, FAU and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Roland Lang
- Institute of Clinical Microbiology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology.,Deutsches Zentrum für Immuntherapie (DZI), and
| | - Aline Bozec
- Department of Internal Medicine 3 - Rheumatology and Immunology.,Deutsches Zentrum für Immuntherapie (DZI), and
| |
Collapse
|
52
|
MiR-206 regulates the progression of osteoporosis via targeting HDAC4. Eur J Med Res 2021; 26:8. [PMID: 33461610 PMCID: PMC7812640 DOI: 10.1186/s40001-021-00480-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 01/06/2021] [Indexed: 11/16/2022] Open
Abstract
Background More and more studies have confirmed that miRNAs play an important role in maintaining bone remodeling and bone metabolism. This study investigated the expression level of miR-206 in the serum of osteoporosis (OP) patients and explored the effect and mechanism of miR-206 on the occurrence and development of osteoporosis. Methods 120 postmenopausal women were recruited, including 63 cases with OP and 57 women without OP. The levels of miR-206 were determined by qRT-PCR technology. Spearman correlation coefficient was used to evaluate the correlation of miR-206 with bone mineral density (BMD). An ROC curve was used to evaluate the diagnostic value of miR-206 in osteoporosis. The effects of miR-206 on cell proliferation and cell apoptosis of hFOBs were measured by CCK-8 assay and flow cytometry, respectively. Luciferase reporter gene assay was used to confirm the interaction of miR-206 and the 3′UTR of HDAC4. Results Serum miR-206 had low expression level in osteoporosis patient group compared with control group. The expression level of serum miR-206 had diagnostic value for osteoporosis, and the serum miR-206 levels were positively correlated with BMD. The down-regulated miR-206 could inhibit cell proliferation and promote cell apoptosis. Luciferase analysis indicated that HDAC4 was the target gene of miR-206. Conclusions MiR-206 could be used as a new potential diagnostic biomarker for osteoporosis, and in in vitro cell experiments, miR-206 may regulate osteoblast cell proliferation and apoptosis by targeting HDAC4.
Collapse
|
53
|
Florencio-Silva R, Sasso GRDS, Sasso-Cerri E, Simões MDJ, Cerri PS. Immunoexpression pattern of autophagy mediators in alveolar bone osteoclasts following estrogen withdrawal in female rats. J Mol Histol 2021; 52:321-333. [PMID: 33409945 DOI: 10.1007/s10735-020-09953-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 12/28/2020] [Indexed: 12/30/2022]
Abstract
It is known that estrogen deficiency increases osteoclast formation and activity. Autophagy, a cell survival pathway, has been shown to be crucial for osteoclast function. However, little is known about the effects of estrogen depletion on osteoclast autophagy. Here, we evaluated the effects of estrogen deficiency in the immunoexpression of autophagy mediators in alveolar bone osteoclasts of ovariectomized rats. Twelve adult female rats were ovariectomized (OVX-group) or SHAM-operated (SHAM-group). After three weeks, the rats were euthanized and maxillary fragments containing alveolar bone of the first molars were processed for light microscopy or transmission electron microscopy (TEM). Paraffin-sections were subjected to the TRAP method (osteoclast marker) or to the immunohistochemical detections of beclin-1, LC3α, and p62 (autophagy mediators); araldite-sections were processed for TEM. The number of TRAP-positive osteoclasts and the number of immunolabeled-multinucleated cells (MNCs) along the alveolar bone surface of the first molar were computed. The number of TRAP-positive osteoclasts and the number of beclin-1-, LC3α- and p62-immunolabelled osteoclasts were significantly higher in OVX-group than the SHAM-group. MNCs were frequently located juxtaposed to Howship lacunae along the alveolar bone surface, indicating that these cells are osteoclasts. TEM revealed osteoclasts exhibiting autophagosomes. Our data indicate that autophagy plays an important role during estrogen deficiency-induced osteoclastogenesis. Thus, our results contribute to a better understanding on the role of autophagy on osteoclasts under estrogenic deficiency, and reinforce the idea that modulation of autophagy may be a useful tool to inhibit excessive oral bone resorption in post-menopausal women.
Collapse
Affiliation(s)
- Rinaldo Florencio-Silva
- Disciplina de Histologia e Biologia Estrutural, Departamento de Morfologia e Genética, Escola Paulista de Medicina - EPM, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brasil.
| | - Gisela Rodrigues da Silva Sasso
- Departamento de Ginecologia, Escola Paulista de Medicina - EPM, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brasil
| | - Estela Sasso-Cerri
- Araraquara - Laboratory of Histology and Embryology, School of Dentistry, São Paulo State University (UNESP), Araraquara, SP, Brasil
| | - Manuel de Jesus Simões
- Disciplina de Histologia e Biologia Estrutural, Departamento de Morfologia e Genética, Escola Paulista de Medicina - EPM, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brasil
| | - Paulo Sérgio Cerri
- Araraquara - Laboratory of Histology and Embryology, School of Dentistry, São Paulo State University (UNESP), Araraquara, SP, Brasil
| |
Collapse
|
54
|
Alteration of the oral microbiota may be a responsible factor, along with estrogen deficiency, by the development of larger periapical lesions. Clin Oral Investig 2020; 25:3651-3662. [PMID: 33188615 DOI: 10.1007/s00784-020-03688-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/06/2020] [Indexed: 01/09/2023]
Abstract
OBJECTIVES To answer the questions: (1) Does reducing estrogen levels influence the microbial composition of the oral cavity? (2) Does the presence of periapical lesion (PL) cause changes in the oral microbiota? (3) Since estrogen deficiency alters the oral microbiota, can this be one of the factors that contribute to the increase of the PL? MATERIALS AND METHODS Thirty-six rats were divided into four groups: sham (control), ovariectomy (OVX), control with PL (Sham + PL), and OVX + PL. After 9 weeks of OVX, the lower first molars were submitted to PL induction. After 21 days, the microbiological collection of the oral cavity was performed, and the animals were euthanized. The contents were evaluated by the checkerboard DNA-DNA hybridization method, to verify the prevalence of 40 bacterial species (divided into 7 microbial complexes). The blocks containing the lower first molars were submitted to histotechnical processing and staining with hematoxylin and eosin (HE), for the measurement of the periapical lesion area. The results were submitted to ANOVA and Kruskal-Wallis tests and Tukey and Dunn post-tests, with a significance level of 5%. RESULTS In conditions of estrogen deficiency, there was alteration of the oral microbiota. The OVX groups had a higher amount of bacteria compared to the SHAM group in most of the microbial complexes (p < 0.001). The animals in the control group (with or without lesion) did not present a statistically significant difference (p > 0.001) in any of the microbial complexes. The PLs in OVX animals were significantly higher compared to SHAM animals (p < 0.001). CONCLUSIONS Hypoestrogenicity conditions interfere in the oral microbiota by increasing the amount of bacteria in the saliva and influencing the progression of periapical lesions. CLINICAL RELEVANCE This inedited study shows that deficiency of estrogen leads to alteration of the oral microbiota.
Collapse
|
55
|
Osteocyte apoptosis: the roles and key molecular mechanisms in resorption-related bone diseases. Cell Death Dis 2020; 11:846. [PMID: 33046704 PMCID: PMC7552426 DOI: 10.1038/s41419-020-03059-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 01/18/2023]
Abstract
Vital osteocytes have been well known to function as an important orchestrator in the preservation of robustness and fidelity of the bone remodeling process. Nevertheless, some key pathological factors, such as sex steroid deficiency and excess glucocorticoids, and so on, are implicated in inducing a bulk of apoptotic osteocytes, subsequently resulting in resorption-related bone loss. As much, osteocyte apoptosis, under homeostatic conditions, is in an optimal state of balance tightly controlled by pro- and anti-apoptotic mechanism pathways. Importantly, there exist many essential signaling proteins in the process of osteocyte apoptosis, which has a crucial role in maintaining a homeostatic environment. While increasing in vitro and in vivo studies have established, in part, key signaling pathways and cross-talk mechanism on osteocyte apoptosis, intrinsic and complex mechanism underlying osteocyte apoptosis occurs in various states of pathologies remains ill-defined. In this review, we discuss not only essential pro- and anti-apoptotic signaling pathways and key biomarkers involved in these key mechanisms under different pathological agents, but also the pivotal role of apoptotic osteocytes in osteoclastogenesis-triggered bone loss, hopefully shedding new light on the attractive and proper actions of pharmacotherapeutics of targeting apoptosis and ensuing resorption-related bone diseases such as osteoporosis and fragility fractures.
Collapse
|
56
|
Aggarwal A, Sharma N, Khera A, Sandhir R, Rishi V. Quercetin alleviates cognitive decline in ovariectomized mice by potentially modulating histone acetylation homeostasis. J Nutr Biochem 2020; 84:108439. [DOI: 10.1016/j.jnutbio.2020.108439] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/08/2020] [Accepted: 05/27/2020] [Indexed: 12/20/2022]
|
57
|
Schwarze UY, Strauss FJ, Gruber R. Caspase inhibitor attenuates the shape changes in the alveolar ridge following tooth extraction: A pilot study in rats. J Periodontal Res 2020; 56:101-107. [PMID: 32935871 PMCID: PMC7891322 DOI: 10.1111/jre.12798] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/04/2020] [Accepted: 08/09/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVE The aim of the study was to determine whether the inhibition of apoptosis via pan-caspase inhibitors can attenuate the changes in the alveolar ridge upon tooth extraction. BACKGROUND Cells undergoing apoptosis might play a central role in the onset of alveolar bone resorption and the ensuing bone atrophy following tooth extraction. Caspases are proteases that regulate apoptotic cell death. It is, therefore, reasonable to hypothesize that blocking apoptosis with pan-caspase inhibitors attenuates the changes in the alveolar ridge following tooth extraction. METHODS In 16 inbred rats, the mandibular first (M1) and second (M2) molars of one side were extracted. Following random allocation, the rats received either a cell-permeable pan-caspase inhibitor or diluent. After a healing period of 10 days, changes in shape and height of the alveolar ridge were examined using geometric morphometrics and linear measurements based on micro-computed tomography. RESULTS Geometric morphometric analysis revealed that the pan-caspase inhibitor prevented major shape changes of the alveolar ridge following M1 tooth extraction (P < .05). Furthermore, linear measurements confirmed that the pan-caspase inhibitor significantly prevented the atrophy of the alveolar ridge height following M1 tooth extraction compared to the diluent controls (-0.53 mm vs -0.24 mm; P = .012). M2 tooth extraction caused no shape changes of the alveolar ridge, and thus, the pan-caspase inhibitor group did not differ from the control group (-0.14 mm vs -0.05 mm; P = .931). CONCLUSIONS These findings suggest that the inhibition of apoptosis may attenuate shape changes of the alveolar ridge following M1 tooth extraction in rodents.
Collapse
Affiliation(s)
- Uwe Yacine Schwarze
- Department of Oral Biology, Medical University of Vienna, Vienna, Austria.,Division of Oral Surgery and Orthodontics, Department of Dental Medicine and Oral Health, Medical University of Graz, Graz, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Franz-Josef Strauss
- Clinic of Reconstructive Dentistry, Center of Dental Medicine, University of Zurich, Zurich, Switzerland.,Department of Conservative Dentistry, Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Reinhard Gruber
- Department of Oral Biology, Medical University of Vienna, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland
| |
Collapse
|
58
|
Dell'Aquila E, Armento G, Iuliani M, Simonetti S, D'Onofrio L, Zeppola T, Madaudo C, Russano M, Citarella F, Ribelli G, Pantano F, Vincenzi B, Tonini G, Santini D. Denosumab for cancer-related bone loss. Expert Opin Biol Ther 2020; 20:1261-1274. [PMID: 32835531 DOI: 10.1080/14712598.2020.1814731] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Prolonged use of anti-cancer treatments in breast and prostate tumors alters physiological bone turnover leading to adverse skeletal related events, such as osteoporosis, loss of bone mass, and increased risk of fractures. These complications known as cancer treatment-induced bone loss (CTIBL) should be managed with bone targeting agents such as the bisphosphonates and denosumab. The latter is a monoclonal antibody against the receptor activator of nuclear factor-kB ligand (RANKL) that suppresses osteoclasts function and survival increasing bone mass. AREAS COVERED This review will focus on the mechanisms associated with bone loss induced by cancer treatments and the most recent evidence about the use of denosumab as preventive and therapeutic strategy to protect bone health. Moreover, we will discuss several key aspects regarding the clinical practical use of denosumab to optimize the management of CTLIB in breast and prostate cancer. EXPERT OPINION Denosumab treatment strongly prevents cancer therapies-related skeletal issues in breast and prostate cancer with a good safety profile. Adjuvant six-monthly denosumab delays the time to first fracture onset in early stage breast cancer patients with normal or altered bone mineral density (BMD). Similarly, denosumab treatment is able to prevent fractures and BMD loss in nonmetastatic prostate cancer patients.
Collapse
Affiliation(s)
| | - Grazia Armento
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Michele Iuliani
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Sonia Simonetti
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Loretta D'Onofrio
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Tea Zeppola
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Cristina Madaudo
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Marco Russano
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Fabrizio Citarella
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Giulia Ribelli
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Francesco Pantano
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Bruno Vincenzi
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Giuseppe Tonini
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Daniele Santini
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| |
Collapse
|
59
|
Wang XY, Gong LJ, Huang JM, Jiang C, Yan ZQ. Pinocembrin alleviates glucocorticoid-induced apoptosis by activating autophagy via suppressing the PI3K/Akt/mTOR pathway in osteocytes. Eur J Pharmacol 2020; 880:173212. [PMID: 32470335 DOI: 10.1016/j.ejphar.2020.173212] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/14/2020] [Accepted: 05/19/2020] [Indexed: 12/20/2022]
Abstract
Glucocorticoids are widely used in clinical practice, but are associated with potentially severe side effects like glucocorticoid-induced osteoporosis (GIOP) and glucocorticoid-associated osteonecrosis of the femoral head (GA-ONFH). Glucocorticoid-induced osteocyte apoptosis plays critical roles in the pathological processes of both GIOP and GA-ONFH. Pinocembrin is a natural flavonoid that may exert protective effects on osteocytes. The present study investigated the effects of pinocembrin on glucocorticoid-induced apoptosis of murine long bone osteocyte Y4 (MLO-Y4) cells and sought to elucidate the underlying molecular mechanism. We found that pinocembrin attenuated glucocorticoid-induced cell viability injury and apoptosis of MLO-Y4 cells. Moreover, pinocembrin increased Beclin-1 and LC3B-II level, but decreased p62 expression, suggesting that pinocembrin activates autophagy in glucocorticoid-treated MLO-Y4 cells. The protective effects of pinocembrin on glucocorticoid-induced apoptosis of MLO-Y4 cells were mimicked by a known stimulator of autophagy but prevented by a known inhibitor of autophagy. Pinocembrin also suppressed the PI3K/Akt/mTOR signaling pathway, which regulates cell autophagy, in glucocorticoid-treated MLO-Y4 cells. In conclusion, the results indicate that pinocembrin alleviates glucocorticoid-induced osteocyte apoptosis by activating autophagy via suppressing the PI3K/Akt/mTOR pathway. Pinocembrin may represent a potential natural agent for preventing and treating GIOP and GA-ONFH.
Collapse
Affiliation(s)
- Xin-Yuan Wang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Lin-Jing Gong
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Jun-Ming Huang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Chang Jiang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Zuo-Qin Yan
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
60
|
Oliveira MC, Pieters BCH, Guimarães PB, Duffles LF, Heredia JE, Silveira ALM, Oliveira ACC, Teixeira MM, Ferreira AVM, Silva TA, van de Loo FAJ, Macari S. Bovine Milk Extracellular Vesicles Are Osteoprotective by Increasing Osteocyte Numbers and Targeting RANKL/OPG System in Experimental Models of Bone Loss. Front Bioeng Biotechnol 2020; 8:891. [PMID: 32850743 PMCID: PMC7411003 DOI: 10.3389/fbioe.2020.00891] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 07/10/2020] [Indexed: 12/24/2022] Open
Abstract
Studying effects of milk components on bone may have a clinical impact as milk is highly associated with bone maintenance, and clinical studies provided controversial associations with dairy consumption. We aimed to evaluate the impact of milk extracellular vesicles (mEVs) on the dynamics of bone loss in mice. MEVs are nanoparticles containing proteins, mRNA and microRNA, and were supplemented into the drinking water of mice, either receiving diet-induced obesity or ovariectomy (OVX). Mice receiving mEVs were protected from the bone loss caused by diet-induced obesity. In a more severe model of bone loss, OVX, higher osteoclast numbers in the femur were found, which were lowered by mEV treatment. Additionally, the osteoclastogenic potential of bone marrow-derived precursor cells was lowered in mEV-treated mice. The reduced stiffness in the femur of OVX mice was consequently reversed by mEV treatment, accompanied by improvement in the bone microarchitecture. In general, the RANKL/OPG ratio increased systemically and locally in both models and was rescued by mEV treatment. The number of osteocytes, as primary regulators of the RANKL/OPG system, raised in the femur of the OVX mEVs-treated group compared to OVX non-treated mice. Also, the osteocyte cell line treated with mEVs demonstrated a lowered RANKL/OPG ratio. Thus, mEVs showed systemic and local osteoprotective properties in two mouse models of bone loss reflected in reduced osteoclast presence. Data reveal mEV potential in bone modulation, acting via osteocyte enhancement and RANKL/OPG regulation. We suggest that mEVs could be a therapeutic candidate for the treatment of bone loss.
Collapse
Affiliation(s)
- Marina C Oliveira
- Laboratory of Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Experimental Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands.,Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Bartijn C H Pieters
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Polianna B Guimarães
- Laboratory of Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Letícia F Duffles
- Department of Oral Surgery and Pathology, Faculty of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Joyce E Heredia
- Laboratory of Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana L M Silveira
- Laboratory of Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Amanda C C Oliveira
- Laboratory of Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro M Teixeira
- Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Adaliene V M Ferreira
- Laboratory of Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Tarcilia A Silva
- Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Department of Oral Surgery and Pathology, Faculty of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fons A J van de Loo
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Soraia Macari
- Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Department of Restorative Dentistry, Faculty of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
61
|
Naqvi SM, Panadero Pérez JA, Kumar V, Verbruggen ASK, McNamara LM. A Novel 3D Osteoblast and Osteocyte Model Revealing Changes in Mineralization and Pro-osteoclastogenic Paracrine Signaling During Estrogen Deficiency. Front Bioeng Biotechnol 2020; 8:601. [PMID: 32656194 PMCID: PMC7326002 DOI: 10.3389/fbioe.2020.00601] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/18/2020] [Indexed: 11/13/2022] Open
Abstract
Recent in vitro studies have revealed that the mechanobiological responses of osteoblasts and osteocytes are fundamentally impaired during estrogen deficiency. However, these two-dimensional (2D) cell culture studies do not account for in vivo biophysical cues. Thus, the objectives of this study are to (1) develop a three-dimensional (3D) osteoblast and osteocyte model integrated into a bioreactor and (2) apply this model to investigate whether estrogen deficiency leads to changes in osteoblast to osteocyte transition, mechanosensation, mineralization, and paracrine signaling associated with bone resorption by osteoclasts. MC3T3-E1s were expanded in media supplemented with estrogen (17β-estradiol). These cells were encapsulated in gelatin-mtgase before culture in (1) continued estrogen (E) or (2) no further estrogen supplementation. Constructs were placed in gas permeable and water impermeable cell culture bags and maintained at 5% CO2 and 37°C. These bags were either mechanically stimulated in a custom hydrostatic pressure (HP) bioreactor or maintained under static conditions (control). We report that osteocyte differentiation, characterized by the presence of dendrites and staining for osteocyte marker dentin matrix acidic phosphoprotein 1 (DMP1), was significantly greater under estrogen withdrawal (EW) compared to under continuous estrogen treatment (day 21). Mineralization [bone sialoprotein (BSP), osteopontin (OPN), alkaline phosphatase (ALP), calcium] and gene expression associated with paracrine signaling for osteoclastogenesis [receptor activator of nuclear factor kappa-β ligand (RANKL)/osteoprotegerin OPG ratio] were significantly increased in estrogen deficient and mechanically stimulated cells. Interestingly, BSP and DMP-1 were also increased at day 1 and day 21, respectively, which play a role in regulation of biomineralization. Furthermore, the increase in pro-osteoclastogenic signaling may be explained by altered mechanoresponsiveness of osteoblasts or osteocytes during EW. These findings highlight the impact of estrogen deficiency on bone cell function and provide a novel in vitro model to investigate the mechanisms underpinning changes in bone cells after estrogen deficiency.
Collapse
Affiliation(s)
| | | | | | | | - Laoise M. McNamara
- Mechanobiology and Medical Device Research Group, Department of Biomedical Engineering, College of Engineering and Informatics, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
62
|
McCutcheon S, Majeska RJ, Spray DC, Schaffler MB, Vazquez M. Apoptotic Osteocytes Induce RANKL Production in Bystanders via Purinergic Signaling and Activation of Pannexin Channels. J Bone Miner Res 2020; 35:966-977. [PMID: 31910292 PMCID: PMC8009310 DOI: 10.1002/jbmr.3954] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 11/25/2019] [Accepted: 12/14/2019] [Indexed: 12/15/2022]
Abstract
Localized apoptosis of osteocytes, the tissue-resident cells within bone, occurs with fatigue microdamage and activates bone resorption. Osteoclasts appear to target and remove dying osteocytes, resorbing damaged bone matrix as well. Osteocyte apoptosis similarly activates bone resorption with estrogen loss and in disuse. Apoptotic osteocytes trigger viable neighbor (ie, bystander) osteocytes to produce RANKL, the cytokine required for osteoclast activation. Signals from apoptotic osteocytes that trigger this bystander RANKL expression remain obscure. Studying signaling among osteocytes has been hampered by lack of in vitro systems that model the limited communication among osteocytes in vivo (ie, via gap junctions on cell processes and/or paracrine signals through thin pericellular fluid spaces around osteocytes). Here, we used a novel multiscale fluidic device (the Macro-micro-nano, or Mμn) that reproduces these key anatomical features. Osteocytes in discrete compartments of the device communicate only via these limited pathways, which allows assessment of their roles in triggering osteocytes RANKL expression. Apoptosis of MLOY-4 osteocytes in the Mμn device caused increased osteocyte RANKL expression in the neighboring compartment, consistent with in vivo findings. This RANKL upregulation in bystander osteocytes was prevented by blocking Pannexin 1 channels as well as its ATP receptor. ATP alone caused comparable RANKL upregulation in bystander osteocytes. Finally, blocking Connexin 43 gap junctions did not abolish osteocyte RANKL upregulation, but did alter the distribution of RANKL expressing bystander osteocytes. These findings point to extracellular ATP, released from apoptotic osteocytes via Panx1 channels, as a major signal for triggering bystander osteocyte RANKL expression and activating bone remodeling. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Sean McCutcheon
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Robert J Majeska
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - David C Spray
- Dominick Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mitchell B Schaffler
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Maribel Vazquez
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
63
|
Wang T, Yu X, He C. Pro-inflammatory Cytokines: Cellular and Molecular Drug Targets for Glucocorticoid-induced-osteoporosis via Osteocyte. Curr Drug Targets 2020; 20:1-15. [PMID: 29618305 DOI: 10.2174/1389450119666180405094046] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 03/11/2018] [Accepted: 03/21/2018] [Indexed: 02/08/2023]
Abstract
Glucocorticoids are widely used to treat varieties of allergic and autoimmune diseases, however, long-term application results in glucocorticoid-induced osteoporosis (GIOP). Inflammatory cytokines: tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) play important regulatory roles in bone metabolism, but their roles in GIOP remain largely unknown. Osteocytes can modulate the formation and function of both osteoblasts and osteoclasts, directly via gap junctions, or indirectly by transferring molecule signaling. Apoptotic osteocytes release RANKL, HMGB1 and pro-inflammatory cytokines to stimulate osteoclastogenesis. Moreover, osteocytes can secrete FGF23 to regulate bone metabolism. Exposure to high levels of GCs can drive osteocyte apoptosis and influence gap junctions, leading to bone loss. GCs treatment is regarded to produce more FGF23 to inhibit bone mineralization. GCs also disrupt the vascular to decrease osteocyte feasibility and mineral appositional rate, resulting in a decline in bone strength. Apoptotic bodies from osteocytes induced by GCs treatment can enhance production of TNF-α and IL-6. On the other hand, TNF-α and IL-6 show synergistic effects by altering osteocytes signaling towards osteoclasts and osteoblasts. In addition, TNF-α can induce osteocyte apoptosis and attribute to a worsened bone quality in GCs. IL-6 and osteocytes may interact with each other. Therefore, we hypothesize that GCs regulate osteocyteogenesis through TNF-α and IL-6, which are highly expressed around osteocyte undergoing apoptosis. In the present review, we summarized the roles of osteocytes in regulating osteoblasts and osteoclasts. Furthermore, the mechanism of GCs altered relationship between osteocytes and osteoblasts/osteoclasts. In addition, we discussed the roles of TNF-α and IL-6 in GIOP by modulating osteocytes. Lastly, we discussed the possibility of using pro-inflammatory signaling pathway as therapeutic targets to develop drugs for GIOP.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.,Laboratory of Endocrinology and Metabolism, Department of Endocrinology, National Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, 610041, China
| | - Xijie Yu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology, National Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, 610041, China
| | - Chengqi He
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
64
|
Li J, Chen X, Lu L, Yu X. The relationship between bone marrow adipose tissue and bone metabolism in postmenopausal osteoporosis. Cytokine Growth Factor Rev 2020; 52:88-98. [PMID: 32081538 DOI: 10.1016/j.cytogfr.2020.02.003] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 02/06/2020] [Accepted: 02/06/2020] [Indexed: 02/06/2023]
Abstract
Postmenopausal osteoporosis (PMOP) is a prevalent skeletal disorder associated with menopause-related estrogen withdrawal. PMOP is characterized by low bone mass, deterioration of the skeletal microarchitecture, and subsequent increased susceptibility to fragility fractures, thus contributing to disability and mortality. Accumulating evidence indicates that abnormal expansion of marrow adipose tissue (MAT) plays a crucial role in the onset and progression of PMOP, in part because both bone marrow adipocytes and osteoblasts share a common ancestor lineage. The cohabitation of MAT adipocytes, mesenchymal stromal cells, hematopoietic cells, osteoblasts and osteoclasts in the bone marrow creates a microenvironment that permits adipocytes to act directly on other cell types in the marrow. Furthermore, MAT, which is recognized as an endocrine organ, regulates bone remodeling through the secretion of adipokines and cytokines. Although an enhanced MAT volume is linked to low bone mass and fractures in PMOP, the detailed interactions between MAT and bone metabolism remain largely unknown. In this review, we examine the possible mechanisms of MAT expansion under estrogen withdrawal and further summarize emerging findings regarding the pathological roles of MAT in bone remodeling. We also discuss the current therapies targeting MAT in osteoporosis. A comprehensive understanding of the relationship between MAT expansion and bone metabolism in estrogen deficiency conditions will provide new insights into potential therapeutic targets for PMOP.
Collapse
Affiliation(s)
- Jiao Li
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiang Chen
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lingyun Lu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China; Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xijie Yu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
65
|
Guder C, Gravius S, Burger C, Wirtz DC, Schildberg FA. Osteoimmunology: A Current Update of the Interplay Between Bone and the Immune System. Front Immunol 2020; 11:58. [PMID: 32082321 PMCID: PMC7004969 DOI: 10.3389/fimmu.2020.00058] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 01/09/2020] [Indexed: 12/11/2022] Open
Abstract
Immunology, already a discipline in its own right, has become a major part of many different medical fields. However, its relationship to orthopedics and trauma surgery has unfortunately, and perhaps unjustly, been developing rather slowly. Discoveries in recent years have emphasized the immense breadth of communication and connection between both systems and, importantly, the highly promising therapeutic opportunities. Recent discoveries of factors originally assigned to the immune system have now also been shown to have a significant impact on bone health and disease, which has greatly changed how we approach treatment of bone pathologies. In case of bone fracture, immune cells, especially macrophages, are present throughout the whole healing process, assure defense against pathogens and discharge a complex variety of effectors to regulate bone modeling. In rheumatoid arthritis and osteoporosis, the immune system contributes to the formation of the pathological and chronic conditions. Fascinatingly, prosthesis failure is not at all solely a mechanical problem of improper strain but works in conjunction with an active contribution of the immune system as a reaction to irritant debris from material wear. Unraveling conjoined mechanisms of the immune and osseous systems heralds therapeutic possibilities for ailments of both. Contemplation of the bone as merely an unchanging support pillar is outdated and obsolete. Instead it is mandatory that this highly diverse network be incorporated in our understanding of the immune system and hematopoiesis.
Collapse
Affiliation(s)
- Christian Guder
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Sascha Gravius
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany.,Department of Orthopedics and Trauma Surgery, University Medical Center Mannheim of University Heidelberg, Mannheim, Germany
| | - Christof Burger
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Dieter C Wirtz
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Frank A Schildberg
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
66
|
Creecy A, Damrath JG, Wallace JM. Control of Bone Matrix Properties by Osteocytes. Front Endocrinol (Lausanne) 2020; 11:578477. [PMID: 33537002 PMCID: PMC7848033 DOI: 10.3389/fendo.2020.578477] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/01/2020] [Indexed: 12/13/2022] Open
Abstract
Osteocytes make up 90-95% of the cellular content of bone and form a rich dendritic network with a vastly greater surface area than either osteoblasts or osteoclasts. Osteocytes are well positioned to play a role in bone homeostasis by interacting directly with the matrix; however, the ability for these cells to modify bone matrix remains incompletely understood. With techniques for examining the nano- and microstructure of bone matrix components including hydroxyapatite and type I collagen becoming more widespread, there is great potential to uncover novel roles for the osteocyte in maintaining bone quality. In this review, we begin with an overview of osteocyte biology and the lacunar-canalicular system. Next, we describe recent findings from in vitro models of osteocytes, focusing on the transitions in cellular phenotype as they mature. Finally, we describe historical and current research on matrix alteration by osteocytes in vivo, focusing on the exciting potential for osteocytes to directly form, degrade, and modify the mineral and collagen in their surrounding matrix.
Collapse
Affiliation(s)
- Amy Creecy
- Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, United States
| | - John G. Damrath
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States
| | - Joseph M. Wallace
- Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, United States
- *Correspondence: Joseph M. Wallace,
| |
Collapse
|
67
|
Chai S, Wan L, Wang JL, Huang JC, Huang HX. Gushukang inhibits osteocyte apoptosis and enhances BMP-2/Smads signaling pathway in ovariectomized rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 64:153063. [PMID: 31419728 DOI: 10.1016/j.phymed.2019.153063] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/22/2019] [Accepted: 08/02/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Traditional herbal formula Gushukang (GSK) has been clinically applied to treat primary osteoporosis, which can stimulate osteoblastogenesis and improve calcium homeostasis. However, it remains unknown the mechanism that GSK against ovariectomized (OVX) induced damage. PURPOSE The aim of this study was to investigate the effect of GSK on BMP-2/Smsds signaling pathway and osteocyte apoptosis which has been reported to play a central role in bone remodeling. STUDY DESIGN OVX in rat was established and GSK was administered. RESULTS BMP-2/Smsds signaling pathway was inhibited and the number of apoptotic osteocytes was increased in OVX rats. Treatment with GSK significantly enhanced BMP-2/Smsds signaling pathway by up-regulating the expression of BMP-2, p-Smad1 and p-Smad5, Osterix and Runx2, and inhibited osteocyte apoptosis by up-regulating Bcl-xl and down-regulating Bak, which were consistent with histological changes revealed by ALP, Trap and TUNEL staining. GSK treatment improved bone mass and micro-structure of trabecular bone at distal femur in OVX rats shown by BMD, micro-CT measurement and HE staining. CONCLUSION These data suggest that GSK exhibited protective effects on promoting bone formation and precluding osteocyte apoptosis. The underlying mechanism may be attributed to its regulation on BMP-2/Smads signaling pathway and Bcl2 family.
Collapse
Affiliation(s)
- Shuang Chai
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lei Wan
- The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ji-Li Wang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jia-Chun Huang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hong-Xing Huang
- The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Laboratory Affiliated to National Key Discipline of Orthopaedic and Traumatology of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
68
|
Roberts BC, Giorgi M, Oliviero S, Wang N, Boudiffa M, Dall'Ara E. The longitudinal effects of ovariectomy on the morphometric, densitometric and mechanical properties in the murine tibia: A comparison between two mouse strains. Bone 2019; 127:260-270. [PMID: 31254730 DOI: 10.1016/j.bone.2019.06.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 06/24/2019] [Accepted: 06/24/2019] [Indexed: 12/19/2022]
Abstract
Oestrogen deficiency-related bone loss in the ovariectomized (OVX) mouse is a common model for osteoporosis. However, a comprehensive in vivo assessment of intervention-related changes in multiple bone properties, and in multiple mouse strains, is required in order to identify an appropriate model for future evaluation of novel anti-osteoporotic therapies. The aim of this study was to evaluate the effect of OVX on the morphometric and densitometric properties measured in the microCT images and the mechanical properties estimated with finite element models of the tibia in two mouse strains, C57BL/6 and BALB/c. 14-weeks-old female C57BL/6 and BALB/c mice were divided into two groups per strain: (1) ovariectomized, (2) non-operated control. The right tibia was scanned at baseline (14 weeks) and then every two weeks thereafter, until 24-weeks-old, using in vivo microCT. Changes in trabecular and cortical bone morphometry, spatiotemporal changes in densitometric properties and in mechanical properties (from micro-finite element (μFE) analysis) were computed. Differences between OVX and non-operated controls were evaluated by ANCOVA, adjusted for 14-weeks baseline. In morphometry, trabecular bone mass was significantly reduced in both C57BL/6 and BALB/c from four weeks following surgery. Though the OVX-effect was transient in BALB/c as bone mass reached skeletal homeostasis. OVX inhibited the age-related thickening of cortical bone only in C57BL/6. In both strains, increments in bone mineral content were significantly lower with OVX only in the proximal tibia, with intervention-related differences increasing with time. OVX had no effect on μFE estimates of stiffness nor failure load in either strain. The results of this study show strain-, time- and region-(trabecular or cortical) dependent changes in morphometric and densitometric properties. These findings highlight the importance of choosing an appropriate mouse model and time points for research of treatments against accelerated bone resorption.
Collapse
Affiliation(s)
- Bryant C Roberts
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK; Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, UK
| | - Mario Giorgi
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK; Certara QSP, Certara UK Ltd., Simcyp Division, Sheffield, UK
| | - Sara Oliviero
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK; Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, UK
| | - Ning Wang
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK; MRC Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA), University of Sheffield, Sheffield, UK
| | - Maya Boudiffa
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK; MRC Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA), University of Sheffield, Sheffield, UK
| | - Enrico Dall'Ara
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK; Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, UK; MRC Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA), University of Sheffield, Sheffield, UK.
| |
Collapse
|
69
|
S-nitrosoglutathione prevents cognitive impairment through epigenetic reprogramming in ovariectomised mice. Biochem Pharmacol 2019; 168:352-365. [DOI: 10.1016/j.bcp.2019.07.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/23/2019] [Indexed: 12/22/2022]
|
70
|
Silva RAB, Sousa‐Pereira AP, Lucisano MP, Romualdo PC, Paula‐Silva FWG, Consolaro A, Silva LAB, Nelson‐Filho P. Alendronate inhibits osteocyte apoptosis and inflammation via
IL
‐6, inhibiting bone resorption in periapical lesions of ovariectomized rats. Int Endod J 2019; 53:84-96. [DOI: 10.1111/iej.13206] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 08/16/2019] [Indexed: 01/29/2023]
Affiliation(s)
- R. A. B. Silva
- Department of Pediatric Dentistry School of Dentistry of Ribeirão Preto University of São Paulo Ribeirão Preto Brazil
| | | | - M. P. Lucisano
- Department of Pediatric Dentistry School of Dentistry of Ribeirão Preto University of São Paulo Ribeirão Preto Brazil
| | - P. C. Romualdo
- Department of Pediatric Dentistry School of Dentistry of Ribeirão Preto University of São Paulo Ribeirão Preto Brazil
| | - F. W. G. Paula‐Silva
- Department of Pediatric Dentistry School of Dentistry of Ribeirão Preto University of São Paulo Ribeirão Preto Brazil
| | - A. Consolaro
- Department of Stomatology Bauru School of Dentistry University of São Paulo Bauru Brazil
| | - L. A. B. Silva
- Department of Pediatric Dentistry School of Dentistry of Ribeirão Preto University of São Paulo Ribeirão Preto Brazil
| | - P. Nelson‐Filho
- Department of Pediatric Dentistry School of Dentistry of Ribeirão Preto University of São Paulo Ribeirão Preto Brazil
| |
Collapse
|
71
|
McKenzie J, Smith C, Karuppaiah K, Langberg J, Silva MJ, Ornitz DM. Osteocyte Death and Bone Overgrowth in Mice Lacking Fibroblast Growth Factor Receptors 1 and 2 in Mature Osteoblasts and Osteocytes. J Bone Miner Res 2019; 34:1660-1675. [PMID: 31206783 PMCID: PMC6744314 DOI: 10.1002/jbmr.3742] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 03/27/2019] [Accepted: 04/05/2019] [Indexed: 01/11/2023]
Abstract
Fibroblast growth factor (FGF) signaling pathways have well-established roles in skeletal development, with essential functions in both chondrogenesis and osteogenesis. In mice, previous conditional knockout studies suggested distinct roles for FGF receptor 1 (FGFR1) signaling at different stages of osteogenesis and a role for FGFR2 in osteoblast maturation. However, the potential for redundancy among FGFRs and the mechanisms and consequences of stage-specific osteoblast lineage regulation were not addressed. Here, we conditionally inactivate Fgfr1 and Fgfr2 in mature osteoblasts with an Osteocalcin (OC)-Cre or Dentin matrix protein 1 (Dmp1)-CreER driver. We find that young mice lacking both receptors or only FGFR1 are phenotypically normal. However, between 6 and 12 weeks of age, OC-Cre Fgfr1/Fgfr2 double- and Fgfr1 single-conditional knockout mice develop a high bone mass phenotype with increased periosteal apposition, increased and disorganized endocortical bone with increased porosity, and biomechanical properties that reflect increased bone mass but impaired material properties. Histopathological and gene expression analyses show that this phenotype is preceded by a striking loss of osteocytes and accompanied by activation of the Wnt/β-catenin signaling pathway. These data identify a role for FGFR1 signaling in mature osteoblasts/osteocytes that is directly or indirectly required for osteocyte survival and regulation of bone mass during postnatal bone growth. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jennifer McKenzie
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA.,Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Craig Smith
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kannan Karuppaiah
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Joshua Langberg
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Matthew J Silva
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA.,Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - David M Ornitz
- Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
72
|
Connexin 43 hemichannels protect bone loss during estrogen deficiency. Bone Res 2019; 7:11. [PMID: 31016065 PMCID: PMC6476886 DOI: 10.1038/s41413-019-0050-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 02/28/2019] [Accepted: 03/18/2019] [Indexed: 02/02/2023] Open
Abstract
Estrogen deficiency in postmenopausal women is a major cause of bone loss, resulting in osteopenia, osteoporosis, and a high risk for bone fracture. Connexin 43 (Cx43) hemichannels (HCs) in osteocytes play an important role in osteocyte viability, bone formation, and remodeling. We showed here that estrogen deficiency reduced Cx43 expression and HC function. To determine if functional HCs protect osteocytes and bone loss during estrogen deficiency, we adopted an ovariectomy model in wild-type (WT) and two transgenic Cx43 mice: R76W (dominant-negative mutant inhibiting only gap junction channels) and Cx43 Δ130–136 (dominant-negative mutant compromising both gap junction channels and HCs). The bone mineral density (BMD), bone structure, and histomorphometric changes of cortical and trabecular bones after ovariectomy were investigated. Our results showed that the Δ130–136 transgenic cohort had greatly decreased vertebral trabecular bone mass compared to WT and R76W mice, associated with a significant increase in the number of apoptotic osteocyte and empty lacunae. Moreover, osteoclast surfaces in trabecular and cortical bones were increased after ovariectomy in the R76W and WT mice, respectively, but not in ∆130–136 mice. These data demonstrate that impairment of Cx43 HCs in osteocytes accelerates vertebral trabecular bone loss and increase in osteocyte apoptosis, and further suggest that Cx43 HCs in osteocytes protect trabecular bone against catabolic effects due to estrogen deficiency. Channels that form between cells and their extracellular environment help protect bone tissue from the damage wrought by low estrogen levels, a major cause of bone loss in post-menopausal women. Jean Jiang from the UT Health San Antonio and colleagues showed that depleting the estrogen hormone in mouse bone cells reduced levels of connexin 43 and impaired the protein’s ability to forms pores known as ‘hemichannels’. The researchers surgically removed the ovaries of various mouse strains to induce estrogen deficiencies. They found that transgenic mice without working hemichannels had reduced bone mass compared to normal mice or mice that could make hemichannels but lacked the ability for those channels to come together to form complete passageways. The findings highlight the importance of connexin 43 hemichannels in protecting bone tissue against osteoporosis.
Collapse
|
73
|
Xing L, Zhang X, Feng H, Liu S, Li D, Hasegawa T, Guo J, Li M. Silencing FOXO1 attenuates dexamethasone-induced apoptosis in osteoblastic MC3T3-E1 cells. Biochem Biophys Res Commun 2019; 513:1019-1026. [PMID: 31010677 DOI: 10.1016/j.bbrc.2019.04.112] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 04/15/2019] [Indexed: 12/12/2022]
Abstract
Dexamethasone (DEX), a widely used glucocorticoid with strong anti-inflammatory and immunosuppressive activities, has been reported to induce apoptosis in osteoblasts, but the underlying mechanisms are still not comprehensively investigated. FOXO1 plays an important role in the regulation of cell proliferation and apoptosis. Our study aims to explore the role of FOXO1 in DEX-induced apoptosis of osteoblastic MC3T3-E1 cells through bioinformatics and experiments. We first employed bioinformatics to identify DEX-related genes and revealed their functions by GO enrichment analysis including FOXO1 associated biological processes. Expression level of FOXO1 was validated by GEO data. Then, experiments were performed to verify the hypothesis. CCK8 was used to detect cell viability and apoptosis was detected by flow cytometry. SiRNA was used to silence FOXO1 and western-blot was employed to detect protein expression. Results demonstrated DEX-related genes involved in cell proliferation, apoptosis and angiogenesis and FOXO1 was a regulator of apoptosis. DEX could up-regulate FOXO1 expression, inhibit cell viability, promote apoptosis. SiRNA-FOXO1 could attenuate DEX-induced apoptosis in MC3T3-E1. These findings suggested DEX could affect some vital biological processes of MC3T3-E1 and FOXO1 played an essential role in DEX-induced apoptosis in MC3T3-E1.
Collapse
Affiliation(s)
- Lu Xing
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Bone Metabolism, School of Stomatology Shandong University, Jinan, 250012, China
| | - Xiaoqi Zhang
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Bone Metabolism, School of Stomatology Shandong University, Jinan, 250012, China
| | - Hao Feng
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Bone Metabolism, School of Stomatology Shandong University, Jinan, 250012, China
| | - Shanshan Liu
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Bone Metabolism, School of Stomatology Shandong University, Jinan, 250012, China
| | - Dongfang Li
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Bone Metabolism, School of Stomatology Shandong University, Jinan, 250012, China
| | - Tomoka Hasegawa
- Department of Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586, Japan
| | - Jie Guo
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Bone Metabolism, School of Stomatology Shandong University, Jinan, 250012, China
| | - Minqi Li
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Bone Metabolism, School of Stomatology Shandong University, Jinan, 250012, China.
| |
Collapse
|
74
|
El Hage C, Hallit S, Akel M, Dagher E. Osteoporosis awareness and health beliefs among Lebanese women aged 40 years and above. Osteoporos Int 2019; 30:771-786. [PMID: 30840113 DOI: 10.1007/s00198-019-04901-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 02/12/2019] [Indexed: 10/27/2022]
Abstract
UNLABELLED Our study investigated the characteristics of Lebanese women population groups with the poorest knowledge of osteoporosis definition, risk factors, and preventive measures in order to identify categories that should be targeted in future campaigns and educational programs. Higher knowledge scores were reached in women who already heard about the disease and had a formal education. As a result, osteoporosis awareness campaigns and educational programs are mostly needed in populations with lower educational levels. INTRODUCTION Our study investigated the characteristics of Lebanese women population groups with the poorest knowledge of osteoporosis definition, risk factors, and preventive measures. METHODS A cross-sectional study, conducted between March and June 2018, enrolled 560 community dwelling women aged 40 years and above. A questionnaire was used to collect data. A proportionate random sample from all Lebanese Mohafazat was used. Data collection was performed through personal interviews. The median was used as a cutoff point for both the Knowledge and Health Belief scales. RESULTS The study results showed that 47.3% of participants had a poor knowledge score. Women who received no education, compared to a higher education and have not previously heard about the disease, had lower knowledge scores. In addition, women not taking calcium and vitamin D supplements and not exercising or exercising less than 20 min per day compared to those having these characteristics had lower osteoporosis knowledge levels. A lower knowledge score was associated with less recognized benefits of adequate calcium intake and regular physical activity, more perceived barriers towards their practice, and a less important health motivation. A lower level of education correlated to the same results. CONCLUSION Lower knowledge scores were reached in women who have never heard of osteoporosis and had a lower level of education. As a result, osteoporosis awareness campaigns and educational programs need to target population categories with lower educational levels.
Collapse
Affiliation(s)
- C El Hage
- Faculty of Medicine and Medical Sciences, Holy Spirit University of Kaslik (USEK), Jounieh, Lebanon.
- Notre Dame des Secours Hospital University Hospital Center, Byblos City, Lebanon.
- , Building Carole El-Boustany, Holy Virgin Street, 1st Floor, Damour, Lebanon.
| | - S Hallit
- Faculty of Medicine and Medical Sciences, Holy Spirit University of Kaslik (USEK), Jounieh, Lebanon.
- INSPECT-LB: Institut National de Sante Publique, Epidemiologie Clinique et Toxicologie, Beirut, Lebanon.
| | - M Akel
- INSPECT-LB: Institut National de Sante Publique, Epidemiologie Clinique et Toxicologie, Beirut, Lebanon
- School of Pharmacy, Lebanese International University, Beirut, Lebanon
| | - E Dagher
- Faculty of Medicine and Medical Sciences, Holy Spirit University of Kaslik (USEK), Jounieh, Lebanon
- Notre Dame des Secours Hospital University Hospital Center, Byblos City, Lebanon
| |
Collapse
|
75
|
Estrogen deficiency impairs integrin α vβ 3-mediated mechanosensation by osteocytes and alters osteoclastogenic paracrine signalling. Sci Rep 2019; 9:4654. [PMID: 30874595 PMCID: PMC6420496 DOI: 10.1038/s41598-019-41095-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/26/2019] [Indexed: 02/06/2023] Open
Abstract
The integrin αvβ3 has been shown to play an important role in osteocyte mechanotransduction. It has been reported that there are fewer β3 integrin-containing cells in osteoporotic bone cells. Osteocytes cultured in vitro under estrogen deficient conditions demonstrate altered mechanotransduction. However, it is unknown whether the altered mechanotransduction in estrogen deficient osteocytes is directly associated with defective αvβ3 expression or signalling. The objective of this study is to investigate the role of estrogen deficiency for regulating MLO-Y4 cell morphology, αvβ3 expression, focal adhesion formation and mechanotransduction by osteocytes. Here, we report that estrogen withdrawal leads to a smaller focal adhesion area and reduced αvβ3 localisation at focal adhesion sites, resulting in an increased Rankl/Opg ratio and defective Cox-2 responses to oscillatory fluid flow. Interestingly, αvβ3 antagonism had a similar effect on focal adhesion assembly, Rankl/Opg ratio, and Cox-2 responses to oscillatory fluid flow. Taken together, our results provide the first evidence for a relationship between estrogen withdrawal and defective αvβ3-mediated signalling. Specifically, this study implicates estrogen withdrawal as a putative mechanism responsible for altered αvβ3 expression and resultant changes in downstream signalling in osteocytes during post-menopausal osteoporosis, which might provide an important, but previously unidentified, contribution to the bone loss cascade.
Collapse
|
76
|
Osteocytes respond to particles of clinically-relevant conventional and cross-linked polyethylene and metal alloys by up-regulation of resorptive and inflammatory pathways. Acta Biomater 2019; 87:296-306. [PMID: 30690207 DOI: 10.1016/j.actbio.2019.01.047] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 01/18/2019] [Accepted: 01/24/2019] [Indexed: 02/06/2023]
Abstract
Periprosthetic osteolysis is a major cause of implant failure in total hip replacements. Aseptic loosening caused by osteolytic lesions is associated with the production of bioactive wear particles from the articulations of implants. Wear particles infiltrate the surrounding tissue of implants, promoting inflammation as well as bone resorption. Osteocytes have been shown to both regulate physiological osteoclastogenesis and directly remodel their perilacunar bone matrix by the process of osteocytic osteolysis. We hypothesise that osteocytes respond to wear debris of orthopaedic implant materials by adopting a pro-catabolic phenotype and thus contribute to periprosthetic osteolysis through the known pathways of bone loss. Osteocyte responses to particles derived from clinically relevant materials, ultra-high molecular weight polyethylene (UHMWPE), highly cross-linked polyethylene (XLPE) and metal alloys, Ti6Al4V and CoCrMo, were examined in vitro in human primary osteocyte-like cultures. Osteocyte-like cells exposed to both polyethylene and metal wear particle types showed upregulated expression of catabolic markers associated with osteocytic osteolysis, MMP13, carbonic anhydrase 2 (CA2) and cathepsin K (CTSK). In addition, pro-osteoclastogenesis markers RANKL and M-CSF were induced, as well as the expression of pro-inflammatory cytokines, IL-6 and TNFα, albeit with different kinetics. These findings suggest a previously unrecognised action of wear particles of multiple orthopaedic materials on osteocytes, and suggest a multifaceted role for osteocytes in periprosthetic osteolysis. STATEMENT OF SIGNIFICANCE: This study addresses periprosthetic osteolysis, a major clinical problem leading to aseptic loosening of orthopaedic implants. It is well accepted that wear particles of polyethylene and of other implant materials stimulate the activity of bone resorbing osteoclasts. Our recent work provided evidence that commercial particles of ultra-high molecular weight polyethylene (UHMWPE) stimulated osteocytes to adopt a bone catabolic state. In this study we demonstrate for the first time that particles derived from materials in clinical use, conventional UHMWPE, highly cross-linked polyethylene (XLPE), and Ti6Al4V and CoCrMo metal alloys, all stimulate human osteocyte activities of osteocyte-regulated osteoclastogenesis, osteocytic osteolysis, proinflammatory responses, osteocyte apoptosis, albeit to varying extents. This study provides further mechanistic insight into orthopaedic wear particle mediated bone disease in terms of the osteocyte, the most abundant and key controlling cell type in bone.
Collapse
|
77
|
Paschalis EP, Gamsjaeger S, Condon K, Klaushofer K, Burr D. Estrogen depletion alters mineralization regulation mechanisms in an ovariectomized monkey animal model. Bone 2019; 120:279-284. [PMID: 30414509 DOI: 10.1016/j.bone.2018.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 11/05/2018] [Accepted: 11/06/2018] [Indexed: 12/14/2022]
Abstract
Ovariectomized animal models have been extensively used in osteoporosis research due to the resulting loss of bone mass. The purpose of the present study was to test the hypothesis that estrogen depletion alters mineralization regulation mechanisms in an ovariectomized monkey animal model. To achieve this we used Raman microspectroscopy to analyze humeri from monkeys that were either SHAM-operated or ovariectomized (N = 10 for each group). Measurements were made as a function of tissue age and cortical surface (periosteal, osteonal, endosteal) based on the presence of calcein fluorescent double labels. In the present work we focused on osteoid seams (defined as a surface with evident calcein labels, 1 μm distance away from the mineralizing front, and for which the Raman spectra showed the presence of organic matrix but not mineral), as well as the youngest mineralized tissue between the second fluorescent label and the mineralizing front, 1 μm inwards from the front with the phosphate mineral peak evident in the Raman spectra (TA1). The spectroscopically determined parameters of interest were the relative glycosaminoglycan (GAG) and pyridinoline (Pyd) contents in the osteoid, and the mineral content in TA1. At all three cortical surfaces, significant correlations were evident in the SHAM-operated animals between osteoid GAG (negative) and Pyd content, and mineral content, unlike the OVX animals. These results suggest that in addition to the well-established effects on turnover rates and bone mass, estrogen depletion alters the regulation of mineralization by GAGs and Pyd.
Collapse
Affiliation(s)
- E P Paschalis
- Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of WGKK and AUVA Trauma Centre Meidling, 1st Medical Department, Hanusch Hospital, Heinrich Collin Str. 30, A-1140 Vienna, Austria.
| | - S Gamsjaeger
- Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of WGKK and AUVA Trauma Centre Meidling, 1st Medical Department, Hanusch Hospital, Heinrich Collin Str. 30, A-1140 Vienna, Austria
| | - K Condon
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Biomedical Engineering, Indiana University-Purdue University, Indianapolis (IUPUI), Indianapolis, IN 46202, USA
| | - K Klaushofer
- Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of WGKK and AUVA Trauma Centre Meidling, 1st Medical Department, Hanusch Hospital, Heinrich Collin Str. 30, A-1140 Vienna, Austria
| | - D Burr
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Biomedical Engineering, Indiana University-Purdue University, Indianapolis (IUPUI), Indianapolis, IN 46202, USA
| |
Collapse
|
78
|
He F, Bai J, Wang J, Zhai J, Tong L, Zhu G. Irradiation‐induced osteocyte damage promotes HMGB1‐mediated osteoclastogenesis in vitro. J Cell Physiol 2019; 234:17314-17325. [DOI: 10.1002/jcp.28351] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 01/06/2023]
Affiliation(s)
- Feilong He
- Department of Radiation Health Institute of Radiation Medicine, Fudan University Shanghai China
| | - Jiangtao Bai
- Department of Radiation Health Institute of Radiation Medicine, Fudan University Shanghai China
| | - Jianping Wang
- Department of Radiation Health Institute of Radiation Medicine, Fudan University Shanghai China
| | - Jianglong Zhai
- Department of Radiation Health Institute of Radiation Medicine, Fudan University Shanghai China
| | - Ling Tong
- Department of Radiation Health Institute of Radiation Medicine, Fudan University Shanghai China
| | - Guoying Zhu
- Department of Radiation Health Institute of Radiation Medicine, Fudan University Shanghai China
| |
Collapse
|
79
|
Tang Q, Su YW, Fan CM, Chung R, Hassanshahi M, Peymanfar Y, Xian CJ. Release of CXCL12 From Apoptotic Skeletal Cells Contributes to Bone Growth Defects Following Dexamethasone Therapy in Rats. J Bone Miner Res 2019; 34:310-326. [PMID: 30395366 DOI: 10.1002/jbmr.3597] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 08/31/2018] [Accepted: 09/23/2018] [Indexed: 12/17/2022]
Abstract
Dexamethasone (Dex) is known to cause significant bone growth impairment in childhood. Although previous studies have suggested roles of osteocyte apoptosis in the enhanced osteoclastic recruitment and local bone loss, whether it is so in the growing bone following Dex treatment requires to be established. The current study addressed the potential roles of chemokine CXCL12 in chondroclast/osteoclast recruitment and bone defects following Dex treatment. Significant apoptosis was observed in cultured mature ATDC5 chondrocytes and IDG-SW3 osteocytes after 48 hours of 10-6 M Dex treatment, and CXCL12 was identified to exhibit the most prominent induction in Dex-treated cells. Conditioned medium from the treated chondrocytes/osteocytes enhanced migration of RAW264.7 osteoclast precursor cells, which was significantly inhibited by the presence of the anti-CXCL12 neutralizing antibody. To investigate the roles of the induced CXCL12 in bone defects caused by Dex treatment, young rats were orally gavaged daily with saline or Dex at 1 mg/kg/day for 2 weeks, and received an intraperitoneal injection of anti-CXCL12 antibody or control IgG (1 mg/kg, three times per week). Aside from oxidative stress induction systemically, Dex treatment caused reductions in growth plate thickness, primary spongiosa height, and metaphysis trabecular bone volume, which are associated with induced chondrocyte/osteocyte apoptosis and enhanced chondroclast/osteoclast recruitment and osteoclastogenic differentiation potential. CXCL12 was induced in apoptotic growth plate chondrocytes and metaphyseal bone osteocytes. Anti-CXCL12 antibody supplementation considerably attenuated Dex-induced chondroclast/osteoclast recruitment and loss of growth plate cartilage and trabecular bone. CXCL12 neutralization did not affect bone marrow osteogenic potential, adiposity, and microvasculature. Thus, CXCL12 was identified as a potential molecular linker between Dex-induced skeletal cell apoptosis and chondroclastic/osteoclastic recruitment, as well as growth plate cartilage/bone loss, revealing a therapeutic potential of CXCL12 functional blockade in preventing bone growth defects during/after Dex treatment. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Qian Tang
- School of Pharmacy and Medical Sciences, and University of South Australia Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Yu-Wen Su
- School of Pharmacy and Medical Sciences, and University of South Australia Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Chia-Ming Fan
- School of Pharmacy and Medical Sciences, and University of South Australia Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Rosa Chung
- School of Pharmacy and Medical Sciences, and University of South Australia Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Mohammadhossein Hassanshahi
- School of Pharmacy and Medical Sciences, and University of South Australia Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Yaser Peymanfar
- School of Pharmacy and Medical Sciences, and University of South Australia Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Cory J Xian
- School of Pharmacy and Medical Sciences, and University of South Australia Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia.,Ningbo No. 6 Hospital, Ningbo, 315040, China
| |
Collapse
|
80
|
Liu Z, Solesio ME, Schaffler MB, Frikha-Benayed D, Rosen CJ, Werner H, Kopchick JJ, Pavlov EV, Abramov AY, Yakar S. Mitochondrial Function Is Compromised in Cortical Bone Osteocytes of Long-Lived Growth Hormone Receptor Null Mice. J Bone Miner Res 2019; 34:106-122. [PMID: 30216544 PMCID: PMC7080402 DOI: 10.1002/jbmr.3573] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 08/03/2018] [Accepted: 08/08/2018] [Indexed: 12/12/2022]
Abstract
Despite increased longevity and resistance to multiple stressors, growth hormone receptor null (GHRKO) mice exhibit severe skeletal impairment. The role of GHR in maintaining osteocyte mitochondrial function is unknown. We found that GHR ablation was detrimental to osteocyte mitochondrial function. In vivo multiphoton microscopy revealed significant reductions of >10% in mitochondrial membrane potential (MMP) in GHRKO osteocytes and reduced mitochondrial volumetric density. Reductions in MMP were accompanied by reductions in glucose transporter-1 levels, steady state ATP, NADH redox index, oxygen consumption rate, and mitochondrial reserve capacity in GHRKO osteocytes. Glycolytic capacity did not differ between control and GHRKO males' osteocytes. However, osteocytes from aged female GHRKO mice exhibited reductions in glycolytic parameters, indicating impairments in glucose metabolism, which may be sex dependent. GHRKO osteocytes exhibited increased levels of cytoplasmic reactive oxygen species (ROS) (both basal and in response to high glucose), insulin-like growth factor-1 (IGF-1), and insulin. Mitochondrial ROS levels were increased and correlated with reduced glutathione in GHRKO osteocytes. Overall, the compromised osteocyte mitochondrial function and responses to metabolic insults strongly correlated with skeletal impairments, suggesting that despite increased life span of the GHRKO mice, skeletal health span is decreased. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Zhongbo Liu
- David B. Kriser Dental Center, Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, USA
| | - Maria E Solesio
- David B. Kriser Dental Center, Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, USA
| | - Mitchell B Schaffler
- Department of Biomedical Engineering, City College of New York, New York, NY, USA
| | - Dorra Frikha-Benayed
- Department of Biomedical Engineering, City College of New York, New York, NY, USA
| | | | - Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - John J Kopchick
- Edison Biotechnology Institute and Department of Biomedical Sciences, Ohio University, Athens, OH, USA
| | - Evgeny V Pavlov
- David B. Kriser Dental Center, Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, USA
| | - Andrey Y Abramov
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
| | - Shoshana Yakar
- David B. Kriser Dental Center, Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, USA
| |
Collapse
|
81
|
Hinton PV, Rackard SM, Kennedy OD. In Vivo Osteocyte Mechanotransduction: Recent Developments and Future Directions. Curr Osteoporos Rep 2018; 16:746-753. [PMID: 30406580 DOI: 10.1007/s11914-018-0485-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PURPOSE OF REVIEW Mechanical loading is an essential stimulus for skeletal tissues. Osteocytes are primarily responsible for sensing mechanical stimuli in bone and for orchestrating subsequent responses. This is critical for maintaining homeostasis, and responding to injury/disease. The osteocyte mechanotransduction pathway, and the downstream effects it mediates, is highly complex. In vivo models have proved invaluable in understanding this process. This review summarizes the commonly used models, as well as more recently developed ones, and describes how they are used to address emerging questions in the field. RECENT FINDINGS Minimally invasive animal models can be used to determine mechanisms of osteocyte mechanotransduction, at the cell and molecular level, while simultaneously reducing potentially confounding responses such as inflammation/wound-healing. The details of osteocyte mechanotransduction in bone are gradually becoming clearer. In vivo model systems are a key tool in pursing this question. Advances in this field are explored and discussed in this review.
Collapse
Affiliation(s)
- Paige V Hinton
- Department of Anatomy & Tissue Engineering Research Group, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland
| | - Susan M Rackard
- School of Veterinary Medicine, Veterinary Science Centre, University College Dublin, Dublin 4, Ireland
| | - Oran D Kennedy
- Department of Anatomy & Tissue Engineering Research Group, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland.
| |
Collapse
|
82
|
Fornetti J, Welm AL, Stewart SA. Understanding the Bone in Cancer Metastasis. J Bone Miner Res 2018; 33:2099-2113. [PMID: 30476357 DOI: 10.1002/jbmr.3618] [Citation(s) in RCA: 303] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/17/2018] [Accepted: 10/18/2018] [Indexed: 12/11/2022]
Abstract
The bone is the third most common site of metastasis for a wide range of solid tumors including lung, breast, prostate, colorectal, thyroid, gynecologic, and melanoma, with 70% of metastatic prostate and breast cancer patients harboring bone metastasis.1 Unfortunately, once cancer spreads to the bone, it is rarely cured and is associated with a wide range of morbidities including pain, increased risk of fracture, and hypercalcemia. This fact has driven experts in the fields of bone and cancer biology to study the bone, and has revealed that there is a great deal that each can teach the other. The complexity of the bone was first described in 1889 when Stephen Paget proposed that tumor cells have a proclivity for certain organs, where they "seed" into a friendly "soil" and eventually grow into metastatic lesions. Dr. Paget went on to argue that although many study the "seed" it would be paramount to understand the "soil." Since this original work, significant advances have been made not only in understanding the cell-autonomous mechanisms that drive metastasis, but also alterations which drive changes to the "soil" that allow a tumor cell to thrive. Indeed, it is now clear that the "soil" in different metastatic sites is unique, and thus the mechanisms that allow tumor cells to remain in a dormant or growing state are specific to the organ in question. In the bone, our knowledge of the components that contribute to this fertile "soil" continues to expand, but our understanding of how they impact tumor growth in the bone remains in its infancy. Indeed, we now appreciate that the endosteal niche likely contributes to tumor cell dormancy, and that osteoclasts, osteocytes, and adipocytes can impact tumor cell growth. Here, we discuss the bone microenvironment and how it impacts cancer cell seeding, dormancy, and growth. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jaime Fornetti
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Alana L Welm
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Sheila A Stewart
- Departments of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA.,Integrating Communication within the Cancer Environment (ICCE) Institute, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
83
|
Pacheco-Costa R, Davis HM, Atkinson EG, Dilley JE, Byiringiro I, Aref MW, Allen MR, Bellido T, Plotkin LI. Reversal of loss of bone mass in old mice treated with mefloquine. Bone 2018; 114:22-31. [PMID: 29879544 PMCID: PMC6056320 DOI: 10.1016/j.bone.2018.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/16/2018] [Accepted: 06/04/2018] [Indexed: 11/16/2022]
Abstract
Aging is accompanied by imbalanced bone remodeling, elevated osteocyte apoptosis, and decreased bone mass and mechanical properties; and improved pharmacologic approaches to counteract bone deterioration with aging are needed. We examined herein the effect of mefloquine, a drug used to treat malaria and systemic lupus erythematosus and shown to ameliorate bone loss in glucocorticoid-treated patients, on bone mass and mechanical properties in young and old mice. Young 3.5-month-old and old 21-month-old female C57BL/6 mice received daily injections of 5 mg/kg/day mefloquine for 14 days. Aging resulted in the expected changes in bone volume and mechanical properties. In old mice mefloquine administration reversed the lower vertebral cancellous bone volume and bone formation; and had modest effects on cortical bone volume, thickness, and moment of inertia. Mefloquine administration did not change the levels of the circulating bone formation markers P1NP or alkaline phosphatase, whereas levels of the resorption marker CTX showed trends towards increase with mefloquine treatment. In addition, and as expected, aging bones exhibited an accumulation of active caspase3-expressing osteocytes and higher expression of apoptosis-related genes compared to young mice, which were not altered by mefloquine administration at either age. In young animals, mefloquine induced higher periosteal bone formation, but lower endocortical bone formation. Further, osteoclast numbers were higher on the endocortical bone surface and circulating CTX levels were increased, in mefloquine- compared to vehicle-treated young mice. Consistent with this, addition of mefloquine to bone marrow cells isolated from young mice led to increased osteoclastic gene expression and a tendency towards increased osteoclast numbers in vitro. Taken together our findings identify the age and bone-site specific skeletal effects of mefloquine. Further, our results highlight a beneficial effect of mefloquine administration on vertebral cancellous bone mass in old animals, raising the possibility of using this pharmacologic inhibitor to preserve skeletal health with aging.
Collapse
Affiliation(s)
- Rafael Pacheco-Costa
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hannah M Davis
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Emily G Atkinson
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Julian E Dilley
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Innocent Byiringiro
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Mohammad W Aref
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Matthew R Allen
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, USA.
| | - Teresita Bellido
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, USA; Division of Endocrinology, Department of Internal Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Lilian I Plotkin
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, USA.
| |
Collapse
|
84
|
Tsourdi E, Jähn K, Rauner M, Busse B, Bonewald LF. Physiological and pathological osteocytic osteolysis. JOURNAL OF MUSCULOSKELETAL & NEURONAL INTERACTIONS 2018; 18:292-303. [PMID: 30179206 PMCID: PMC6146198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Osteocytes, the most abundant bone cell in the adult skeleton, can function as mechanosensors directing osteoblast and osteoclast function in order to maintain optimal load bearing bone in addition to functioning as endocrine cells regulating phosphate metabolism. A controversial function, previously overlooked or denied, has been osteocytes as regulators of calcium metabolism. Early histologists upon observing enlarged osteocyte lacunae in bone sections proposed that mature osteocytes could remove their perilacunar matrix, a term called "osteocytic osteolysis". New insights into this process have occurred during the last decade using novel technology thereby providing a means to identify molecular mechanisms responsible for osteocytic osteolysis. As release of calcium from a mineralized matrix requires a more acidic pH and specialized enzymes, it was proposed that osteocytes may utilize similar molecular mechanisms as osteoclasts to remove mineral. The idea that a cell descended from mesenchymal progenitors (the osteocyte) could function similarly to a cell descended from hematopoietic progenitors (the osteoclast) was challenged as being improbable. Here we review the molecular mechanisms behind this osteocyte function, the role of osteocytic osteolysis in health and disease, and the capacity of the osteocyte to reverse the osteolytic process by replacing the removed matrix, a revived osteoblast function.
Collapse
Affiliation(s)
- Elena Tsourdi
- Department of Medicine III, Technische Universität Dresden Medical Center, Dresden, Germany,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany,Corresponding author: Elena Tsourdi, M.D., Division of Endocrinology, Diabetes and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden Medical Center, Fetscherstrasse 74, 01307 Dresden, Germany E-mail:
| | - Katharina Jähn
- Department for Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martina Rauner
- Department of Medicine III, Technische Universität Dresden Medical Center, Dresden, Germany,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Björn Busse
- Department for Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lynda F. Bonewald
- Indiana Center for Musculoskeletal Health, VanNuys Medical Science Building, Indianapolis, USA
| |
Collapse
|
85
|
Lazzaro L, Tonkin BA, Poulton IJ, McGregor NE, Ferlin W, Sims NA. IL-6 trans-signalling mediates trabecular, but not cortical, bone loss after ovariectomy. Bone 2018; 112:120-127. [PMID: 29679733 DOI: 10.1016/j.bone.2018.04.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 04/04/2018] [Accepted: 04/17/2018] [Indexed: 12/21/2022]
Abstract
Bone loss associated with estrogen deficiency occurs due to a high level of bone remodelling, with a greater increase in the level of osteoclast-mediated bone resorption than osteoblast-mediated bone formation. Early studies showed that Interleukin-6 (IL-6) inhibition could prevent the increase in osteoclast numbers associated with ovariectomy. However, IL-6 signals through two possible pathways: classic IL-6 signalling (cis) utilizes a membrane-bound IL-6 receptor (IL-6R), while IL-6 trans-signalling occurs through a soluble IL-6R (sIL-6R). It is not known which of these pathways mediates the bone loss after ovariectomy. We therefore sought to determine whether specific pharmacological inhibition of IL-6 trans-signalling could prevent ovariectomy-induced bone loss in mice. We report that IL-6 trans-signalling inhibition prevented the increase in osteoclasts, and trabecular bone loss, associated with ovariectomy. IL-6 trans-signalling inhibition also reduced bone formation rate, but did not prevent the increase in osteoblast numbers. In contrast, cortical bone loss was not prevented by any IL-6 signalling inhibitor. This suggests that local production of sIL-6R mediates trabecular bone loss in estrogen deficiency, but the increased cortical bone resorption that leads to marrow expansion is independent of IL-6 signalling.
Collapse
Affiliation(s)
- Leah Lazzaro
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; The University of Melbourne, Department of Medicine at St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Brett A Tonkin
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Ingrid J Poulton
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Narelle E McGregor
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | | | - Natalie A Sims
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; The University of Melbourne, Department of Medicine at St. Vincent's Hospital, Fitzroy, Victoria, Australia.
| |
Collapse
|
86
|
Manolagas SC. The Quest for Osteoporosis Mechanisms and Rational Therapies: How Far We've Come, How Much Further We Need to Go. J Bone Miner Res 2018; 33:371-385. [PMID: 29405383 PMCID: PMC6816306 DOI: 10.1002/jbmr.3400] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/25/2018] [Accepted: 01/27/2018] [Indexed: 12/30/2022]
Abstract
During the last 40 years, understanding of bone biology and the pathogenesis of osteoporosis, the most common and impactful bone disease of old age, has improved dramatically thanks to basic and clinical research advances, genetic insights from humans and rodents, and newer imaging technologies. Culprits of osteoporosis are no longer a matter of speculation based on in vitro observations. Instead, they can be identified and dissected at the cellular and molecular level using genetic approaches; and their effect on distinct bone envelopes and anatomic regions can be functionally assessed in vivo. The landscape of pharmacotherapies for osteoporosis has also changed profoundly with the emergence of several potent antiresorptive drugs as well as anabolic agents, displacing estrogen replacement as the treatment of choice. In spite of these major positive developments, the optimal duration of the available therapies and their long-term safety remain matters of conjecture and some concern. Moreover, antiresorptive therapies are used indiscriminately for patients of all ages on the assumption that suppressing remodeling is always beneficial for bone, but rebound remodeling upon their discontinuation suggests otherwise. In this invited perspective, I highlight the latest state of knowledge of bone-intrinsic and extrinsic mechanisms responsible for the development of osteoporosis in both sexes; differences between the mechanisms responsible for the effects of aging and estrogen deficiency; and the role of old osteocytes in the development of cortical porosity. In addition, I highlight advances toward the goal of developing drugs for several degenerative diseases of old age at once, including osteoporosis, by targeting shared mechanisms of aging. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Stavros C Manolagas
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| |
Collapse
|
87
|
Silva LHFME, Silva JMDEME, Salama M, Pinheiro LGP, Lunardi FO, Silva PGBDA, Hirth CG, Lucena IFDE, Gomes GJDAC, Leite JAD. Criopreserved ovarian tissue transplantation and bone restoration metabolism in castrated rats. Rev Col Bras Cir 2018; 45:e1577. [PMID: 29451647 DOI: 10.1590/0100-6991e-20181577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 11/30/2017] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVES to evaluate estradiol levels and autotransplantation heated ovarian tissue effects, after vitrification, on rats bone metabolism previously oophorectomized bilaterally. METHODS experimental study with 27 rats aged 11 to 12 weeks and weighing 200g to 300g, submitted to bilateral oophorectomy and ovarian tissue cryopreservation for subsequent reimplantation. Animals were divided into two groups, A and B, with 8 and 19 rats, respectively. Autotransplantation occurred in two periods according to castration time: after one week, in group A, and after one month in group B. Serum estradiol measurements and ovary and tibia histological analysis were performed before and after oophorectomy period (early or late) and one month after reimplantation. RESULTS in groups A and B, tibia median cortical thickness was 0.463±0.14mm (mean±SD) at the baseline, 0.360±0.14mm after oophorectomy and 0.445±0.17mm one month after reimplantation p<0.005). Trabecular means were 0.050±0.08mm (mean±SD) at baseline, 0.022±0.08mm after oophorectomy and 0.049±0.032mm one month after replantation (p<0.005). There was no statistical difference in estradiol variation between the two study groups (p=0.819). CONCLUSION cryopreserved ovarian tissue transplantation restored bone parameters, and these results suggest that ovarian reimplantation in women may have the same beneficial effects on bone metabolism.
Collapse
|
88
|
Abstract
Considerable advances in oncology over recent decades have led to improved survival, while raising concerns about long-term consequences of anticancer treatments. In patients with breast or prostate malignancies, bone health is a major issue due to the high risk of bone metastases and the frequent prolonged use of hormone therapies that alter physiological bone turnover, leading to increased fracture risk. Thus, the onset of cancer treatment-induced bone loss (CTIBL) should be considered by clinicians and recent guidelines should be routinely applied to these patients. In particular, baseline and periodic follow-up evaluations of bone health parameters enable the identification of patients at high risk of osteoporosis and fractures, which can be prevented by the use of bone-targeting agents (BTAs), calcium and vitamin D supplementation and modifications of lifestyle. This review will focus upon the pathophysiology of breast and prostate cancer treatment-induced bone loss and the most recent evidence about effective preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Catherine Handforth
- Department of Oncology and Metabolism, Academic Unit of Clinical Oncology, Weston Park Hospital, University of Sheffield, Sheffield, UK
| | - Stella D’Oronzo
- Department of Oncology and Metabolism, Academic Unit of Clinical Oncology, Weston Park Hospital, University of Sheffield, Sheffield, UK
| | - Robert Coleman
- Department of Oncology and Metabolism, Academic Unit of Clinical Oncology, Weston Park Hospital, University of Sheffield, Sheffield, UK
| | - Janet Brown
- Department of Oncology and Metabolism, Academic Unit of Clinical Oncology, Weston Park Hospital, University of Sheffield, Sheffield, UK
| |
Collapse
|
89
|
Florencio-Silva R, Sasso GRS, Sasso-Cerri E, Simões MJ, Cerri PS. Effects of estrogen status in osteocyte autophagy and its relation to osteocyte viability in alveolar process of ovariectomized rats. Biomed Pharmacother 2017; 98:406-415. [PMID: 29276969 DOI: 10.1016/j.biopha.2017.12.089] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/05/2017] [Accepted: 12/18/2017] [Indexed: 12/13/2022] Open
Abstract
Estrogen maintains osteocyte viability, whereas its deficiency induces osteocyte apoptosis. As autophagy is important for osteocyte viability, we hypothesized whether the anti-apoptotic effect of estrogen is related to autophagy in osteocytes. Thirty adult female rats were sham-operated (SHAM) or ovariectomized (OVX). After three weeks, twelve rats of SHAM and OVX groups were killed before treatment (basal period), whereas the remaining rats received estrogen (OVXE) or vehicle (OVX) for 45 days. Fragments of maxilla containing alveolar process of the first molars were embedded in paraffin or Araldite. Paraffin-sections were stained with hematoxylin/eosin for histomorphometry, or subjected to the silver impregnation method for morphological analysis of osteocyte cytoplasmic processes. Autophagy was analyzed by immunohistochemical detections of beclin-1, MAP-LC3α and p62, whereas apoptosis was evaluated by immunohistochemical detections of cleaved caspase-3 and BAX, TUNEL (Terminal deoxynucleotidyl transferase dUTP nick end labeling) method and by ultrastructural analysis. Araldite-semithin sections were subjected to the Sudan-black method for detection of lipids. OVX-basal group showed high frequency of caspase-3-, TUNEL- and p62-positive osteocytes accompanied with low frequency of beclin-1- and MAP-LC3α-positive osteocytes. At 45 days, OVXE group exhibited higher number of osteocytes, higher frequency of beclin-1- and MAP-LC3α-positive osteocytes, and lower frequency of caspase-3, BAX-, TUNEL- and p62-positive osteocytes than OVX group. Significant reduction in bone area was observed in the OVX compared to OVXE and SHAM groups. The highest frequency of Sudan-Black-positive osteocytes and osteocytes with scarce cytoplasmic processes, or showing apoptotic features were mainly observed in OVX groups. Our results indicate that estrogen deficiency decreases autophagy and increases apoptosis, whereas estrogen replacement enhances osteocyte viability by inhibiting apoptosis and maintaining autophagy in alveolar process osteocytes. These results suggest that the anti-apoptotic effect of estrogen may be, at least in part, related to autophagy regulation in osteocytes.
Collapse
Affiliation(s)
- Rinaldo Florencio-Silva
- Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, São Paulo, SP, Brazil
| | - Gisela R S Sasso
- Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departmento de Ginecologia, São Paulo, SP, Brazil
| | - Estela Sasso-Cerri
- São Paulo State University (UNESP), School of Dentistry, Araraquara - Laboratory of Histology and Embryology, Araraquara, SP, Brazil
| | - Manuel J Simões
- Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, São Paulo, SP, Brazil
| | - Paulo S Cerri
- São Paulo State University (UNESP), School of Dentistry, Araraquara - Laboratory of Histology and Embryology, Araraquara, SP, Brazil.
| |
Collapse
|
90
|
Osteocyte calcium signals encode strain magnitude and loading frequency in vivo. Proc Natl Acad Sci U S A 2017; 114:11775-11780. [PMID: 29078317 DOI: 10.1073/pnas.1707863114] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Osteocytes are considered to be the major mechanosensory cells of bone, but how osteocytes in vivo process, perceive, and respond to mechanical loading remains poorly understood. Intracellular calcium (Ca2+) signaling resulting from mechanical stimulation has been widely studied in osteocytes in vitro and in bone explants, but has yet to be examined in vivo. This is achieved herein by using a three-point bending device which is capable of delivering well-defined mechanical loads to metatarsal bones of living mice while simultaneously monitoring the intracellular Ca2+ responses of individual osteocytes by using a genetically encoded fluorescent Ca2+ indicator. Osteocyte responses are imaged by using multiphoton fluorescence microscopy. We investigated the in vivo responses of osteocytes to strains ranging from 250 to 3,000 [Formula: see text] and frequencies from 0.5 to 2 Hz, which are characteristic of physiological conditions reported for bone. At all loading frequencies examined, the number of responding osteocytes increased strongly with applied strain magnitude. However, Ca2+ intensity within responding osteocytes did not change significantly with physiological loading magnitudes. Our studies offer a glimpse into how these critical bone cells respond to mechanical load in vivo, as well as provide a technique to determine how the cells encode magnitude and frequency of loading.
Collapse
|
91
|
Ye T, Cao P, Qi J, Zhou Q, Rao DS, Qiu S. Protective effect of low-dose risedronate against osteocyte apoptosis and bone loss in ovariectomized rats. PLoS One 2017; 12:e0186012. [PMID: 29045447 PMCID: PMC5646759 DOI: 10.1371/journal.pone.0186012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 09/22/2017] [Indexed: 01/23/2023] Open
Abstract
Osteocyte apoptosis is the first reaction to estrogen depletion, thereby stimulating osteoclastic bone resorption resulting in bone loss. We investigated the effects of two different risedronate (RIS) doses (high and low) on osteocyte apoptosis, osteoclast activity and bone loss in ovariectomized rats. Forty rats with ovariectomy (OVX) and sham ovariectomy (SHAM) were divided into 4 groups: 1) SHAM rats treated with saline (SHAM); 2) OVX rats treated with saline (OVX); 3) OVX rats treated with low-dose RIS (OVX-LR, 0.08 μg/kg/day); 4) OVX rats treated with high-dose RIS (OVX-HR, 0.8 μg/kg/day). All animals were sacrificed 90 days after surgery for the examinations of osteocyte apoptosis by caspase-3 staining, osteoclast activity by TRAP staining and bone volume by micro-CT scanning in lumbar vertebral cancellous bone. Both low and high dose RIS significantly reduced caspase-3 positive osteocytes, empty lacunae and TRAP positive osteoclasts in OVX rats. Although the difference in caspase-3 positive osteocytes was not significant between the OVX-LR and OVX-HR groups, numerically these cells were significantly more prevalent in OVX-HR (not OVX-LR) group than in SHAM group. TRAP positive osteoclasts were significantly higher in OVX-LR group than in SHAM or OVX-HR group. There was no significant difference in bone volume among the OVX-LR, OVX-HR and SHAM groups, but lower in OVX group alone. However, significant increase in trabecular thickness only occurred in OVX-LR group. We conclude that both low and high dose RIS significantly inhibit osteocyte apoptosis and osteoclast activity in OVX rats, but the low-dose RIS has weaker effect on osteoclast activity. However, low-dose RIS preserves cancellous bone mass and microarchitecture as well as high-dose RIS after estrogen depletion.
Collapse
Affiliation(s)
- Tingjun Ye
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Peng Cao
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- * E-mail: (SQ); (PC)
| | - Jin Qi
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qi Zhou
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - D. Sudhaker Rao
- Bone and Mineral Research Laboratory, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Shijing Qiu
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Bone and Mineral Research Laboratory, Henry Ford Hospital, Detroit, Michigan, United States of America
- * E-mail: (SQ); (PC)
| |
Collapse
|
92
|
Brunetti G, Faienza MF, Colaianni G, Grano M, Colucci S. Mechanisms of Altered Bone Remodeling in Multiple Myeloma. Clin Rev Bone Miner Metab 2017. [DOI: 10.1007/s12018-017-9236-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
93
|
Su YW, Chen KM, Hassanshahi M, Tang Q, Howe PR, Xian CJ. Childhood cancer chemotherapy-induced bone damage: pathobiology and protective effects of resveratrol and other nutraceuticals. Ann N Y Acad Sci 2017; 1403:109-117. [PMID: 28662275 DOI: 10.1111/nyas.13380] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/13/2017] [Accepted: 04/18/2017] [Indexed: 11/30/2022]
Abstract
Intensive cancer chemotherapy causes significant bone loss, for which the mechanisms remain unclear and effective treatments are lacking. This is a significant issue particularly for childhood cancers, as the most common ones have a >75% cure rate following chemotherapy; there is an increasing population of survivors who live with chronic bone defects. Studies suggest that these defects are the result of reduced bone from increased marrow fat formation and increased bone resorption following chemotherapy. These changes probably result from altered expression/activation of regulatory molecules or pathways regulating skeletal cell formation and activity. Treatment with methotrexate, an antimetabolite commonly used in childhood oncology, has been shown to increase levels of proinflammatory/pro-osteoclastogenic cytokines (e.g., enhanced NF-κB activation), leading to increased osteoclast formation and bone resorption, as well as to attenuate Wnt signaling, leading to both decreased bone and increased marrow fat formation. In recent years, understanding the mechanisms of action and potential health benefits of selected nutraceuticals, including resveratrol, genistein, icariin, and inflammatory fatty acids, has led to preclinical studies that, in some cases, indicate efficacy in reducing chemotherapy-induced bone defects. We summarize the supporting evidence.
Collapse
Affiliation(s)
- Yu-Wen Su
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Ke-Ming Chen
- Institute of Orthopaedics, Lanzhou General Hospital, Lanzhou Command of People's Liberation Army, Lanzhou, PR China
| | - Mohammadhossein Hassanshahi
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Qian Tang
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Peter R Howe
- Clinical Nutrition Research Centre, University of Newcastle, Callaghan, New South Wales, Australia
| | - Cory J Xian
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
94
|
Davis HM, Pacheco-Costa R, Atkinson EG, Brun LR, Gortazar AR, Harris J, Hiasa M, Bolarinwa SA, Yoneda T, Ivan M, Bruzzaniti A, Bellido T, Plotkin LI. Disruption of the Cx43/miR21 pathway leads to osteocyte apoptosis and increased osteoclastogenesis with aging. Aging Cell 2017; 16:551-563. [PMID: 28317237 PMCID: PMC5418188 DOI: 10.1111/acel.12586] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2017] [Indexed: 12/25/2022] Open
Abstract
Skeletal aging results in apoptosis of osteocytes, cells embedded in bone that control the generation/function of bone forming and resorbing cells. Aging also decreases connexin43 (Cx43) expression in bone; and osteocytic Cx43 deletion partially mimics the skeletal phenotype of old mice. Particularly, aging and Cx43 deletion increase osteocyte apoptosis, and osteoclast number and bone resorption on endocortical bone surfaces. We examined herein the molecular signaling events responsible for osteocyte apoptosis and osteoclast recruitment triggered by aging and Cx43 deficiency. Cx43‐silenced MLO‐Y4 osteocytic (Cx43def) cells undergo spontaneous cell death in culture through caspase‐3 activation and exhibit increased levels of apoptosis‐related genes, and only transfection of Cx43 constructs able to form gap junction channels reverses Cx43def cell death. Cx43def cells and bones from old mice exhibit reduced levels of the pro‐survival microRNA miR21 and, consistently, increased levels of the miR21 target phosphatase and tensin homolog (PTEN) and reduced phosphorylated Akt, whereas PTEN inhibition reduces Cx43def cell apoptosis. miR21 reduction is sufficient to induce apoptosis of Cx43‐expressing cells and miR21 deletion in miR21fl/fl bones increases apoptosis‐related gene expression, whereas a miR21 mimic prevents Cx43def cell apoptosis, demonstrating that miR21 lies downstream of Cx43. Cx43def cells release more osteoclastogenic cytokines [receptor activator of NFκB ligand (RANKL)/high‐mobility group box‐1 (HMGB1)], and caspase‐3 inhibition prevents RANKL/HMGB1 release and the increased osteoclastogenesis induced by conditioned media from Cx43def cells, which is blocked by antagonizing HMGB1‐RAGE interaction. These findings identify a novel Cx43/miR21/HMGB1/RANKL pathway involved in preventing osteocyte apoptosis that also controls osteoclast formation/recruitment and is impaired with aging.
Collapse
Affiliation(s)
- Hannah M. Davis
- Department of Anatomy & Cell Biology; Indiana University School of Medicine; Indianapolis IN USA
| | - Rafael Pacheco-Costa
- Department of Anatomy & Cell Biology; Indiana University School of Medicine; Indianapolis IN USA
| | - Emily G. Atkinson
- Department of Anatomy & Cell Biology; Indiana University School of Medicine; Indianapolis IN USA
| | - Lucas R. Brun
- Department of Anatomy & Cell Biology; Indiana University School of Medicine; Indianapolis IN USA
| | - Arancha R. Gortazar
- Instituto de Medicina Molecular Aplicada; Facultad de Medicina; Universidad San Pablo-CEU; Madrid Spain
| | - Julia Harris
- Department of Anatomy & Cell Biology; Indiana University School of Medicine; Indianapolis IN USA
| | - Masahiro Hiasa
- Division of Hematology/Oncology; Department of Internal Medicine; Indiana University School of Medicine; Indianapolis IN USA
| | - Surajudeen A. Bolarinwa
- Department of Anatomy & Cell Biology; Indiana University School of Medicine; Indianapolis IN USA
| | - Toshiyuki Yoneda
- Division of Hematology/Oncology; Department of Internal Medicine; Indiana University School of Medicine; Indianapolis IN USA
| | - Mircea Ivan
- Division of Hematology/Oncology; Department of Internal Medicine; Indiana University School of Medicine; Indianapolis IN USA
| | - Angela Bruzzaniti
- Department of Oral Biology; Indiana University School of Dentistry; Indianapolis IN USA
| | - Teresita Bellido
- Department of Anatomy & Cell Biology; Indiana University School of Medicine; Indianapolis IN USA
- Division of Endocrinology; Department of Internal Medicine; Indiana University School of Medicine; Indianapolis IN USA
- Roudebush Veterans Administration Medical Center; Indianapolis IN USA
| | - Lilian I. Plotkin
- Department of Anatomy & Cell Biology; Indiana University School of Medicine; Indianapolis IN USA
- Roudebush Veterans Administration Medical Center; Indianapolis IN USA
| |
Collapse
|
95
|
Almeida M, Laurent MR, Dubois V, Claessens F, O'Brien CA, Bouillon R, Vanderschueren D, Manolagas SC. Estrogens and Androgens in Skeletal Physiology and Pathophysiology. Physiol Rev 2017. [PMID: 27807202 DOI: 10.1152/physrev.00033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
Abstract
Estrogens and androgens influence the growth and maintenance of the mammalian skeleton and are responsible for its sexual dimorphism. Estrogen deficiency at menopause or loss of both estrogens and androgens in elderly men contribute to the development of osteoporosis, one of the most common and impactful metabolic diseases of old age. In the last 20 years, basic and clinical research advances, genetic insights from humans and rodents, and newer imaging technologies have changed considerably the landscape of our understanding of bone biology as well as the relationship between sex steroids and the physiology and pathophysiology of bone metabolism. Together with the appreciation of the side effects of estrogen-related therapies on breast cancer and cardiovascular diseases, these advances have also drastically altered the treatment of osteoporosis. In this article, we provide a comprehensive review of the molecular and cellular mechanisms of action of estrogens and androgens on bone, their influences on skeletal homeostasis during growth and adulthood, the pathogenetic mechanisms of the adverse effects of their deficiency on the female and male skeleton, as well as the role of natural and synthetic estrogenic or androgenic compounds in the pharmacotherapy of osteoporosis. We highlight latest advances on the crosstalk between hormonal and mechanical signals, the relevance of the antioxidant properties of estrogens and androgens, the difference of their cellular targets in different bone envelopes, the role of estrogen deficiency in male osteoporosis, and the contribution of estrogen or androgen deficiency to the monomorphic effects of aging on skeletal involution.
Collapse
Affiliation(s)
- Maria Almeida
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Michaël R Laurent
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Vanessa Dubois
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Frank Claessens
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Charles A O'Brien
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Roger Bouillon
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Dirk Vanderschueren
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Stavros C Manolagas
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| |
Collapse
|
96
|
Kalyanaraman H, Ramdani G, Joshua J, Schall N, Boss GR, Cory E, Sah RL, Casteel DE, Pilz RB. A Novel, Direct NO Donor Regulates Osteoblast and Osteoclast Functions and Increases Bone Mass in Ovariectomized Mice. J Bone Miner Res 2017; 32:46-59. [PMID: 27391172 PMCID: PMC5199609 DOI: 10.1002/jbmr.2909] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 07/01/2016] [Accepted: 07/05/2016] [Indexed: 01/06/2023]
Abstract
Most US Food and Drug Administration (FDA)-approved treatments for osteoporosis target osteoclastic bone resorption. Only PTH derivatives improve bone formation, but they have drawbacks, and novel bone-anabolic agents are needed. Nitrates, which generate NO, improved BMD in estrogen-deficient rats and may improve bone formation markers and BMD in postmenopausal women. However, nitrates are limited by induction of oxidative stress and development of tolerance, and may increase cardiovascular mortality after long-term use. Here we studied nitrosyl-cobinamide (NO-Cbi), a novel, direct NO-releasing agent, in a mouse model of estrogen deficiency-induced osteoporosis. In murine primary osteoblasts, NO-Cbi increased intracellular cGMP, Wnt/β-catenin signaling, proliferation, and osteoblastic gene expression, and protected cells from apoptosis. Correspondingly, in intact and ovariectomized (OVX) female C57Bl/6 mice, NO-Cbi increased serum cGMP concentrations, bone formation, and osteoblastic gene expression, and in OVX mice, it prevented osteocyte apoptosis. NO-Cbi reduced osteoclasts in intact mice and prevented the known increase in osteoclasts in OVX mice, partially through a reduction in the RANKL/osteoprotegerin gene expression ratio, which regulates osteoclast differentiation, and partially through direct inhibition of osteoclast differentiation, observed in vitro in the presence of excess RANKL. The positive NO effects in osteoblasts were mediated by cGMP/protein kinase G (PKG), but some of the osteoclast-inhibitory effects appeared to be cGMP-independent. NO-Cbi increased trabecular bone mass in both intact and OVX mice, consistent with its in vitro effects on osteoblasts and osteoclasts. NO-Cbi is a novel direct NO-releasing agent that, in contrast to nitrates, does not generate oxygen radicals, and combines anabolic and antiresorptive effects in bone, making it an excellent candidate for treating osteoporosis. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Hema Kalyanaraman
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093-0652
| | - Ghania Ramdani
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093-0652
| | - Jisha Joshua
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093-0652
| | - Nadine Schall
- Institute of Pharmacology and Toxicology, University of Bonn, 53105 Bonn, Germany
| | - Gerry R. Boss
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093-0652
| | - Esther Cory
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093-0652
| | - Robert L. Sah
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093-0652
| | - Darren E. Casteel
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093-0652
| | - Renate B. Pilz
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093-0652
| |
Collapse
|
97
|
Almeida M, Laurent MR, Dubois V, Claessens F, O'Brien CA, Bouillon R, Vanderschueren D, Manolagas SC. Estrogens and Androgens in Skeletal Physiology and Pathophysiology. Physiol Rev 2017; 97:135-187. [PMID: 27807202 PMCID: PMC5539371 DOI: 10.1152/physrev.00033.2015] [Citation(s) in RCA: 546] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Estrogens and androgens influence the growth and maintenance of the mammalian skeleton and are responsible for its sexual dimorphism. Estrogen deficiency at menopause or loss of both estrogens and androgens in elderly men contribute to the development of osteoporosis, one of the most common and impactful metabolic diseases of old age. In the last 20 years, basic and clinical research advances, genetic insights from humans and rodents, and newer imaging technologies have changed considerably the landscape of our understanding of bone biology as well as the relationship between sex steroids and the physiology and pathophysiology of bone metabolism. Together with the appreciation of the side effects of estrogen-related therapies on breast cancer and cardiovascular diseases, these advances have also drastically altered the treatment of osteoporosis. In this article, we provide a comprehensive review of the molecular and cellular mechanisms of action of estrogens and androgens on bone, their influences on skeletal homeostasis during growth and adulthood, the pathogenetic mechanisms of the adverse effects of their deficiency on the female and male skeleton, as well as the role of natural and synthetic estrogenic or androgenic compounds in the pharmacotherapy of osteoporosis. We highlight latest advances on the crosstalk between hormonal and mechanical signals, the relevance of the antioxidant properties of estrogens and androgens, the difference of their cellular targets in different bone envelopes, the role of estrogen deficiency in male osteoporosis, and the contribution of estrogen or androgen deficiency to the monomorphic effects of aging on skeletal involution.
Collapse
Affiliation(s)
- Maria Almeida
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Michaël R Laurent
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Vanessa Dubois
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Frank Claessens
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Charles A O'Brien
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Roger Bouillon
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Dirk Vanderschueren
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Stavros C Manolagas
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| |
Collapse
|
98
|
Cell Death in Chondrocytes, Osteoblasts, and Osteocytes. Int J Mol Sci 2016; 17:ijms17122045. [PMID: 27929439 PMCID: PMC5187845 DOI: 10.3390/ijms17122045] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 11/13/2016] [Accepted: 11/23/2016] [Indexed: 12/04/2022] Open
Abstract
Cell death in skeletal component cells, including chondrocytes, osteoblasts, and osteocytes, plays roles in skeletal development, maintenance, and repair as well as in the pathogenesis of osteoarthritis and osteoporosis. Chondrocyte proliferation, differentiation, and apoptosis are important steps for endochondral ossification. Although the inactivation of P53 and RB is involved in the pathogenesis of osteosarcomas, the deletion of p53 and inactivation of Rb are insufficient to enhance chondrocyte proliferation, indicating the presence of multiple inhibitory mechanisms against sarcomagenesis in chondrocytes. The inflammatory processes induced by mechanical injury and chondrocyte death through the release of danger-associated molecular patterns (DAMPs) are involved in the pathogenesis of posttraumatic osteoarthritis. The overexpression of BCLXL increases bone volume with a normal structure and maintains bone during aging by inhibiting osteoblast apoptosis. p53 inhibits osteoblast proliferation and enhances osteoblast apoptosis, thereby reducing bone formation, but also exerts positive effects on osteoblast differentiation through the Akt–FoxOs pathway. Apoptotic osteocytes release ATP, which induces the receptor activator of nuclear factor κ-B ligand (Rankl) expression and osteoclastogenesis, from pannexin 1 channels. Osteocyte death ultimately results in necrosis; DAMPs are released to the bone surface and promote the production of proinflammatory cytokines, which induce Rankl expression, and osteoclastogenesis is further enhanced.
Collapse
|
99
|
Zhang W, Liu C, Hai B, Du G, Wang H, Leng H, Xu Y, Song C. A Convenient In Vivo Model Using Small Interfering RNA Silencing to Rapidly Assess Skeletal Gene Function. PLoS One 2016; 11:e0167222. [PMID: 27893850 PMCID: PMC5125699 DOI: 10.1371/journal.pone.0167222] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 11/10/2016] [Indexed: 12/15/2022] Open
Abstract
It is difficult to study bone in vitro because it contains various cell types that engage in cross-talk. Bone biologically links various organs, and it has thus become increasingly evident that skeletal physiology must be studied in an integrative manner in an intact animal. We developed a model using local intraosseous small interfering RNA (siRNA) injection to rapidly assess the effects of a target gene on the local skeletal environment. In this model, 160-g male Sprague-Dawley rats were treated for 1-2 weeks. The left tibia received intraosseous injection of a parathyroid hormone 1 receptor (Pth1r) or insulin-like growth factor 1 receptor (Igf-1r) siRNA transfection complex loaded in poloxamer 407 hydrogel, and the right tibia received the same volume of control siRNA. All the tibias received an intraosseous injection of recombinant human parathyroid hormone (1-34) (rhPTH (1-34)) or insulin-like growth factor-1 (IGF-1). Calcein green and alizarin red were injected 6 and 2 days before euthanasia, respectively. IGF-1R and PTH1R expression levels were detected via RT-PCR assays and immunohistochemistry. Bone mineral density (BMD), microstructure, mineral apposition rates (MARs), and strength were determined by dual-energy X-ray absorptiometry, micro-CT, histology and biomechanical tests. The RT-PCR and immunohistochemistry results revealed that IGF-1R and PTH1R expression levels were dramatically diminished in the siRNA-treated left tibias compared to the right tibias (both p<0.05). Using poloxamer 407 hydrogel as a controlled-release system prolonged the silencing effect of a single dose of siRNA; the mRNA expression levels of IGF-1R were lower at two weeks than at one week (p<0.01). The BMD, bone microstructure parameters, MAR and bone strength were significantly decreased in the left tibias compared to the right tibias (all p<0.05). This simple and convenient local intraosseous siRNA injection model achieved gene silencing with very small quantities of siRNA over a short treatment period (≤7 days).
Collapse
Affiliation(s)
- Wen Zhang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
| | - Can Liu
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
| | - Bao Hai
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
| | - Guohong Du
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Hong Wang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
| | - Huijie Leng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
| | - Yingsheng Xu
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Chunli Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
- * E-mail:
| |
Collapse
|
100
|
Cui H, Zhu Y, Yang Q, Zhao W, Zhang S, Zhou A, Jiang D. Necrostatin-1 treatment inhibits osteocyte necroptosis and trabecular deterioration in ovariectomized rats. Sci Rep 2016; 6:33803. [PMID: 27703177 PMCID: PMC5050438 DOI: 10.1038/srep33803] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 09/01/2016] [Indexed: 12/31/2022] Open
Abstract
Estrogen (E2) deficiency has been associated with accelerated osteocyte apoptosis. Our previous study showed necroptosis accelerated the loss of osteocytes in E2 deficiency-induced osteoporosis in rats in addition to apoptosis, but the mechanism involved remains. Necroptosis is a caspase-independent form of programmed cell death. In the necroptosis pathway, receptor interaction proteins 1 and 3 (RIP1/3) play vital roles. Necrostatin-1 (Nec-1) has been confirmed to be a specific inhibitor of necroptosis. However, the effect of Nec-1 on postmenopausal osteoporosis remains ambiguous. The aim of this study was to investigate the effect of Nec-1 on osteocytes in ovariectomized (OVX) rats. We found that an increased number of necroptotic osteocytes was related to the production of tumor necrosis factor-alpha (TNF-α) in OVX rats. Treatment with Nec-1 significantly decreased RIP1 and RIP3 expression in OVX rats and inhibited osteocyte necroptosis induced by TNF-α in vitro. Both E2 and Nec-1 treatment markedly ameliorated trabecular bone deterioration. Nec-1 also significantly elevated the levels of bone formation markers and decreased bone resorption markers. These data suggest that the role of Nec-1 on alleviating bone loss might be associated with Nec-1 restraining TNF-α-induced osteocyte necroptosis in rats with E2 deficiency-induced osteoporosis. This process may represent a novel therapeutic strategy for the treatment of postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Hongwang Cui
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yongjun Zhu
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiming Yang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weikang Zhao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shiyang Zhang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ao Zhou
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dianming Jiang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|