51
|
Kamakura T, Nadol Jr. JB. Evidence of Osteoclastic Activity in the Human Temporal Bone. Audiol Neurootol 2017; 22:218-225. [DOI: 10.1159/000481279] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 09/06/2017] [Indexed: 12/25/2022] Open
Abstract
Bone remodeling within the otic capsule has been reported to be inhibited especially at or near the cochlea, except under some pathological conditions such as otosclerosis, Paget's disease, or mastoiditis, when bone remodeling can occur. Microcavitations found in periosteal and endosteal layers of human temporal bone specimens without otosclerosis, Paget's disease, or inflammation as reported in the current study are consistent with osteoclastic bone resorption. Thirty-three temporal bones from 33 patients were prepared for light microscopy and classified into 4 groups: histologically proven dehiscence of the superior semicircular canal (SSCD) (n = 3, group 1), age 20 years or younger (n = 10, group 2), age 90 years or older and with otosclerosis (n = 10, group 3), and age 90 years or older without otosclerosis (n = 10, group 4). Microcavitation was seen at 7 anatomic locations in the temporal bone in all 4 groups, but not in the cochlea or vestibule. Microcavitation within the temporal bone is likely due to osteoclastic activity, and it is seen in both young and old patients, patients with and without otosclerosis, and in cases with SSCD.
Collapse
|
52
|
Lim R, Li L, Chew N, Yong EL. The prenylflavonoid Icaritin enhances osteoblast proliferation and function by signal transducer and activator of transcription factor 3 (STAT-3) regulation of C-X-C chemokine receptor type 4 (CXCR4) expression. Bone 2017; 105:122-133. [PMID: 28863947 DOI: 10.1016/j.bone.2017.08.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/23/2017] [Accepted: 08/28/2017] [Indexed: 12/12/2022]
Abstract
In this study, we examined the effects of a natural prenylflavonoid Icaritin (ICT), on human osteoblast proliferation and osteogenic function. We observed that ICT dose-dependently enhanced osteoblast proliferation by ~15% over a 7day period. This increase in cell proliferation was associated with corresponding increases in osteoblast functions as measured by ALP secretion, intracellular calcium ions influx and calcium deposition. These anabolic effects were associated with a 4-fold increase in CXCR4 mRNA and protein expression. Silencing of CXCR4 protein expression using small interfering RNA reversed ICT-induced increase in cell proliferation, ALP activity and calcium deposition. Interestingly, we observed that ICT dose-dependently increased STAT-3 phosphorylation; and this resulted in increased binding of phosphorylated STAT-3 to the promoter region of the CXCR4 gene, to increase CXCR4 protein expression. Furthermore, we found that inhibition of STAT-3 phosphorylation resulted in a decrease in CXCR4 protein expression; whilst increasing phosphorylation of STAT-3 using a constitutive active STAT-3 vector significantly increased CXCR4 levels. Moreover, the chemical inhibition of STAT-3 phosphorylation annulled our previously observed ICT-induced increases of osteoblast proliferation and function. Finally, in a rat model of estrogen-deficient osteoporosis, ICT restored both osteoblasts numbers and CXCR4 expression. Taken together, both cellular and animal models support the novel findings that ICT; through the phosphorylation of STAT-3, up-regulated CXCR4, to increase osteoblast proliferation and function.
Collapse
Affiliation(s)
- Rzl Lim
- Department of Obstetrics & Gynaecology, National University of Singapore, Singapore.
| | - L Li
- Department of Medicine, National University of, Singapore, Singapore
| | - N Chew
- Department of Medicine, National University of, Singapore, Singapore; Division of Infectious Diseases, National University Hospital Singapore, Singapore.
| | - E L Yong
- Department of Obstetrics & Gynaecology, National University of Singapore, Singapore.
| |
Collapse
|
53
|
Park S, Jang H, Kim BS, Hwang C, Jeong GS, Park Y. Directional migration of mesenchymal stem cells under an SDF-1α gradient on a microfluidic device. PLoS One 2017; 12:e0184595. [PMID: 28886159 PMCID: PMC5590985 DOI: 10.1371/journal.pone.0184595] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 08/25/2017] [Indexed: 12/13/2022] Open
Abstract
Homing of peripheral stem cells is regulated by one of the most representative homing factors, stromal cell-derived factor 1 alpha (SDF-1α), which specifically binds to the plasma membrane receptor CXCR4 of mesenchymal stem cells (MSCs) in order to initiate the signaling pathways that lead to directional migration and homing of stem cells. This complex homing process and directional migration of stem cells have been mimicked on a microfluidic device that is capable of generating a chemokine gradient within the collagen matrix and embedding endothelial cell (EC) monolayers to mimic blood vessels. On the microfluidic device, stem cells showed directional migration toward the higher concentration of SDF-1α, whereas treatment with the CXCR4 antagonist AMD3100 caused loss of directionality of stem cells. Furthermore, inhibition of stem cell's main migratory signaling pathways, Rho-ROCK and Rac pathways, caused blockage of actomyosin and lamellipodia formation, decreasing the migration distance but maintaining directionality. Stem cell homing regulated by SDF-1α caused directional migration of stem cells, while the migratory ability was affected by the activation of migration-related signaling pathways.
Collapse
Affiliation(s)
- Siwan Park
- Department of Biomedical Engineering, Biomedical Science of Brain Korea 21, College of Medicine, Korea University, Seoul, Korea
| | - Hwanseok Jang
- Department of Biomedical Engineering, Biomedical Science of Brain Korea 21, College of Medicine, Korea University, Seoul, Korea
| | - Byung Soo Kim
- Department of Biomedical Science, Graduate School of Medicine, Korea University, Seoul Korea
| | - Changmo Hwang
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Gi Seok Jeong
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
- * E-mail: (YP); (GSJ)
| | - Yongdoo Park
- Department of Biomedical Engineering, Biomedical Science of Brain Korea 21, College of Medicine, Korea University, Seoul, Korea
- * E-mail: (YP); (GSJ)
| |
Collapse
|
54
|
Stimulation of osteoclast migration and bone resorption by C-C chemokine ligands 19 and 21. Exp Mol Med 2017; 49:e358. [PMID: 28729639 PMCID: PMC5565950 DOI: 10.1038/emm.2017.100] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 01/22/2017] [Accepted: 01/31/2017] [Indexed: 01/26/2023] Open
Abstract
Osteoclasts are responsible for the bone erosion associated with rheumatoid arthritis (RA). The upregulation of the chemokines CCL19 and CCL21 and their receptor CCR7 has been linked to RA pathogenesis. The purpose of this study was to evaluate the effects of CCL19 and CCL21 on osteoclasts and to reveal their underlying mechanisms. The expression of CCL19, CCL21 and CCR7 was higher in RA patients than in osteoarthritis patients. In differentiating osteoclasts, tumor necrosis factor-α, interleukin-1β and lipopolysaccharide stimulated CCR7 expression. CCL19 and CCL21 promoted osteoclast migration and resorption activity. These effects were dependent on the presence of CCR7 and abolished by the inhibition of the Rho signaling pathway. CCL19 and CCL21 promoted bone resorption by osteoclasts in an in vivo mice calvarial model. These findings demonstrate for the first time that CCL19, CCL21 and CCR7 play important roles in bone destruction by increasing osteoclast migration and resorption activity. This study also suggests that the interaction of CCL19 and CCL21 with CCR7 is an effective strategic focus in developing therapeutics for alleviating inflammatory bone destruction.
Collapse
|
55
|
Kindstedt E, Holm CK, Sulniute R, Martinez-Carrasco I, Lundmark R, Lundberg P. CCL11, a novel mediator of inflammatory bone resorption. Sci Rep 2017; 7:5334. [PMID: 28706221 PMCID: PMC5509729 DOI: 10.1038/s41598-017-05654-w] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 06/01/2017] [Indexed: 01/21/2023] Open
Abstract
Normal bone homeostasis, which is regulated by bone-resorbing osteoclasts and bone-forming osteoblasts is perturbed by inflammation. In chronic inflammatory disease with disturbed bone remodelling, e.g. rheumatoid arthritis, patients show increased serum levels of the chemokine eotaxin-1 (CCL11). Herein, we demonstrate an inflammatory driven expression of CCL11 in bone tissue and a novel role of CCL11 in osteoclast migration and resorption. Using an inflammatory bone lesion model and primary cell cultures, we discovered that osteoblasts express CCL11 in vivo and in vitro and that expression increased during inflammatory conditions. Osteoclasts did not express CCL11, but the high affinity receptor CCR3 was significantly upregulated during osteoclast differentiation and found to colocalise with CCL11. Exogenous CCL11 was internalised in osteoclast and stimulated the migration of pre-osteoclast and concomitant increase in bone resorption. Our data pinpoints that the CCL11/CCR3 pathway could be a new target for treatment of inflammatory bone resorption.
Collapse
Affiliation(s)
- Elin Kindstedt
- Department of Odontology/Molecular Periodontology, Umeå University, SE-901 87, Umeå, Sweden
| | - Cecilia Koskinen Holm
- Department of Odontology/Molecular Periodontology, Umeå University, SE-901 87, Umeå, Sweden
| | - Rima Sulniute
- Department of Odontology/Molecular Periodontology, Umeå University, SE-901 87, Umeå, Sweden
| | - Irene Martinez-Carrasco
- Department of Medical Biochemistry and Biophysics, Laboratory for Molecular Infection Medicine Sweden, Umeå University, SE-901 87, Umeå, Sweden
| | - Richard Lundmark
- Department of Medical Biochemistry and Biophysics, Laboratory for Molecular Infection Medicine Sweden, Umeå University, SE-901 87, Umeå, Sweden.,Department of Integrative Medical Biology, Umeå University, SE-901 87, Umeå, Sweden
| | - Pernilla Lundberg
- Department of Odontology/Molecular Periodontology, Umeå University, SE-901 87, Umeå, Sweden.
| |
Collapse
|
56
|
Meshcheryakova A, Mechtcheriakova D, Pietschmann P. Sphingosine 1-phosphate signaling in bone remodeling: multifaceted roles and therapeutic potential. Expert Opin Ther Targets 2017; 21:725-737. [PMID: 28524744 PMCID: PMC5470107 DOI: 10.1080/14728222.2017.1332180] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Sphingolipids belong to a complex class of lipid molecules that are crucially involved in the regulation of important biological processes including proliferation, migration and apoptosis. Given the significant progress made in understanding the sphingolipid pathobiology of several diseases, sphingolipid-related checkpoints emerge as attractive targets. Recent data indicate the multifaceted contribution of the sphingolipid machinery to osteoclast – osteoblast crosstalk, representing one of the pivotal interactions underlying bone homeostasis. Imbalances in the interplay of osteoblasts and osteoclasts might lead to bone-related diseases such as osteoporosis, rheumatoid arthritis, and bone metastases. Areas covered: We summarize and analyze the progress made in bone research in the context of the current knowledge of sphingolipid-related mechanisms regulating bone remodeling. Particular emphasis was given to bioactive sphingosine 1-phosphate (S1P) and S1P receptors (S1PRs). Moreover, the mechanisms of how dysregulations of this machinery cause bone diseases, are covered. Expert opinion: In the context of bone diseases, pharmacological interference with sphingolipid machinery may lead to novel directions in therapeutic strategies. Implementation of knowledge derived from in vivo animal models and in vitro studies using pharmacological agents to manipulate the S1P/S1PRs axes suggests S1PR2 and S1PR3 as potential drug targets, particularly in conjunction with technology for local drug delivery.
Collapse
Affiliation(s)
- Anastasia Meshcheryakova
- a Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology , Medical University of Vienna , Vienna , Austria
| | - Diana Mechtcheriakova
- a Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology , Medical University of Vienna , Vienna , Austria
| | - Peter Pietschmann
- a Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology , Medical University of Vienna , Vienna , Austria
| |
Collapse
|
57
|
Xuan W, Feng X, Qian C, Peng L, Shi Y, Xu L, Wang F, Tan W. Osteoclast differentiation gene expression profiling reveals chemokine CCL4 mediates RANKL-induced osteoclast migration and invasion via PI3K pathway. Cell Biochem Funct 2017; 35:171-177. [PMID: 28370169 DOI: 10.1002/cbf.3260] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/26/2017] [Accepted: 02/27/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Wenhua Xuan
- Department of Rheumatology; The First Affiliated Hospital of Nanjing Medical University; Nanjing China
| | - Xiaoke Feng
- Department of Traditional Chinese Medicine; The First Affiliated Hospital of Nanjing Medical University; Nanjing China
| | - Chen Qian
- Department of Rheumatology; The First Affiliated Hospital of Nanjing Medical University; Nanjing China
| | - Liuying Peng
- Department of Rheumatology; The First Affiliated Hospital of Nanjing Medical University; Nanjing China
| | - Yumeng Shi
- Department of Rheumatology; The First Affiliated Hospital of Nanjing Medical University; Nanjing China
| | - Lingxiao Xu
- Department of Rheumatology; The First Affiliated Hospital of Nanjing Medical University; Nanjing China
| | - Fang Wang
- Department of Cardiology; The First Affiliated Hospital of Nanjing Medical University; Nanjing China
| | - Wenfeng Tan
- Department of Rheumatology; The First Affiliated Hospital of Nanjing Medical University; Nanjing China
| |
Collapse
|
58
|
Ucer S, Iyer S, Kim HN, Han L, Rutlen C, Allison K, Thostenson JD, de Cabo R, Jilka RL, O’Brien C, Almeida M, Manolagas SC. The Effects of Aging and Sex Steroid Deficiency on the Murine Skeleton Are Independent and Mechanistically Distinct. J Bone Miner Res 2017; 32:560-574. [PMID: 27714847 PMCID: PMC5340621 DOI: 10.1002/jbmr.3014] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 09/22/2016] [Accepted: 09/30/2016] [Indexed: 12/17/2022]
Abstract
Old age and sex steroid deficiency are the two most critical factors for the development of osteoporosis. It remains unknown, however, whether the molecular culprits of the two conditions are similar or distinct. We show herein that at 19.5 months of age-a time by which the age-dependent decline of cortical and cancellous bone mass and cortical porosity were fully manifested in C57BL/6J mice-these animals remained functionally estrogen sufficient. Transgenic mice with conditional expression of mitochondria-targeted catalase-a potent H2 O2 inactivating enzyme-in cells of the myeloid lineage (mitoCAT;LysM-Cre mice) were protected from the loss of cortical, but not cancellous, bone caused by gonadectomy in either sex. Consistent with these findings, in vitro studies with ERα-deficient Prx1+ cells and gonadectomized young adult mice showed that in both sexes decreased ERα signaling in Prx1+ cells leads to an increase in SDF1, a.k.a. CXCL12, an osteoclastogenic cytokine whose effects were abrogated in macrophages from mitoCAT;LysM-Cre mice. In contrast to sex steroid deficiency, the adverse effects of aging on either cortical or cancellous bone were unaffected in mitoCAT;LysM-Cre mice. On the other hand, attenuation of H2 O2 generation in cells of the mesenchymal lineage targeted by Prx1-Cre partially prevented the loss of cortical bone caused by old age. Our results suggest the effects of sex steroid deficiency and aging on the murine skeleton are independent and result from distinct mechanisms. In the former, the prevailing mechanism of the cortical bone loss in both sexes is increased osteoclastogenesis caused by estrogen deficiency; this is likely driven, at least in part, by mesenchymal/stromal cell-derived SDF1. Decreased osteoblastogenesis, owing in part to increased H2 O2, combined with increased osteoclastogenesis caused by aging mechanisms independent of estrogen deficiency, are the prevailing mechanisms of the loss of cortical bone with old age. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Serra Ucer
- 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
| | - Srividhya Iyer
- 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
| | - Ha-Neui Kim
- 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
| | - Li Han
- 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
| | - Christine Rutlen
- 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
| | - Kelly Allison
- 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
| | - Jeff D Thostenson
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Robert L Jilka
- 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
| | - Charles 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, AR, USA
| | - 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, AR, USA
| | - 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
|
59
|
Costa D, Principi E, Lazzarini E, Descalzi F, Cancedda R, Castagnola P, Tavella S. LCN2 overexpression in bone enhances the hematopoietic compartment via modulation of the bone marrow microenvironment. J Cell Physiol 2017; 232:3077-3087. [PMID: 28004388 DOI: 10.1002/jcp.25755] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/20/2016] [Accepted: 12/20/2016] [Indexed: 12/15/2022]
Abstract
Lipocalin-2 (LCN2) is a member of the lipocalin family whose expression is modulated in several conditions, including cell differentiation, innate immunity, stress, and cancer. Although it is known that it is expressed in bone, its function in this tissue remains poorly studied. To this end, we took advantage of transgenic mice lines that expressed LCN2 driven by a bone specific type I collagen (LCN2-Tg). In the bone marrow (BM) of LCN2-Tg mice we observed an increased number of phenotypically long-term hematopoietic stem cells (LT-HSC) that also displayed a higher proliferation rate compared to wild-type controls (Wt). Furthermore, hematopoietic progenitor cells, obtained from LCN2-Tg BM showed an increased clonogenic capacity compared to those obtained from LCN2-Tg spleen, a higher concentration of serum erythropoietin and a higher number of mature erythrocytes in the peripheral blood of old LCN2-Tg animals compared to aged-matched wt. The findings of a combined increase in the BM of the LCN2-Tg mice of SDF-1, SCF, and TIMP-1 levels along with the reduction of both MMP-9 activity and cathepsin K concentration may explain the observed effects on the HSC compartment. This study shows that LCN2 overexpression in bones modifies the BM microenvironment via modulation of the expression of key secreted factors and cytokines, which in turn regulate the HSC niche behavior enhancing both HSC homing in young mice and erythrocytes production in older mice.
Collapse
Affiliation(s)
- Delfina Costa
- Dipartimento di Medicina Sperimentale, Universita' di Genova, Genova, Italy
| | - Elisa Principi
- Dipartimento di Medicina Sperimentale, Universita' di Genova, Genova, Italy
| | - Edoardo Lazzarini
- Dipartimento di Medicina Sperimentale, Universita' di Genova, Genova, Italy
| | - Fiorella Descalzi
- Istituto di Bioimmagini e Fisiologia Molecolare, Consiglio Nazionale delle Ricerche, Genova, Italy
| | - Ranieri Cancedda
- Dipartimento di Medicina Sperimentale, Universita' di Genova, Genova, Italy
| | - Patrizio Castagnola
- IRCCS AOU San Martino-IST, Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Sara Tavella
- Dipartimento di Medicina Sperimentale, Universita' di Genova, Genova, Italy.,IRCCS AOU San Martino-IST, Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| |
Collapse
|
60
|
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease that primarily affects the joints. Self-reactive B and T lymphocytes cooperate to promote antibody responses against self proteins and are major drivers of disease. T lymphocytes also promote RA independently of B lymphocytes mainly through the production of key inflammatory cytokines, such as IL-17, that promote pathology. While the innate signals that initiate self-reactive adaptive immune responses are poorly understood, the disease is predominantly caused by inflammatory cellular infiltration and accumulation in articular tissues, and by bone erosions driven by bone-resorbing osteoclasts. Osteoclasts are giant multinucleated cells formed by the fusion of multiple myeloid cells that require short-range signals, such as the cytokines MCSF and RANKL, for undergoing differentiation. The recruitment and positioning of osteoclast precursors to sites of osteoclast differentiation by chemoattractants is an important point of control for osteoclastogenesis and bone resorption. Recently, the GPCR EBI2 and its oxysterol ligand 7a, 25 dihydroxycholesterol, were identified as important regulators of osteoclast precursor positioning in proximity to bone surfaces and of osteoclast differentiation under homeostasis. In chronic inflammatory diseases like RA, osteoclast differentiation is also driven by inflammatory cytokines such as TNFa and IL-1, and can occur independently of RANKL. Finally, there is growing evidence that the chemotactic signals guiding osteoclast precursors to inflamed articular sites contribute to disease and are of great interest. Furthering our understanding of the complex osteoimmune cell interactions should provide new avenues of therapeutic intervention for RA.
Collapse
|
61
|
Matsuura T, Ichinose S, Akiyama M, Kasahara Y, Tachikawa N, Nakahama KI. Involvement of CX3CL1 in the Migration of Osteoclast Precursors Across Osteoblast Layer Stimulated by Interleukin-1ß. J Cell Physiol 2017; 232:1739-1745. [PMID: 27579490 DOI: 10.1002/jcp.25577] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 08/24/2016] [Indexed: 12/25/2022]
Abstract
The trigger for bone remodeling is bone resorption by osteoclasts. Osteoclast differentiation only occurs on the old bone, which needs to be repaired under physiological conditions. However, uncontrolled bone resorption is often observed in pro-inflammatory bone diseases, such as rheumatoid arthritis. Mature osteoclasts are multinuclear cells that differentiate from monocyte/macrophage lineage cells by cell fusion. Although Osteoclast precursors should migrate across osteoblast layer to reach bone matrix before maturation, the underlying mechanisms have not yet been elucidated in detail. We herein found that osteoclast precursors utilize two routes to migrate across osteoblast layer by confocal- and electro-microscopic observations. The osteoclast supporting activity of osteoblasts inversely correlated with osteoblast density and was positively related to the number of osteoclast precursors under the osteoblast layer. Osteoclast differentiation was induced by IL-1ß, but not by PGE2 in high-density osteoblasts. Osteoblasts and osteoclast precursors expressed CX3CL1 and CX3CR1, respectively, and the expression of CX3CL1 increased in response to interleukin-1ß. An anti-CX3CL1-neutralizing antibody inhibited the migration of osteoclast precursors and osteoclast differentiation. These results strongly suggest the involvement of CX3CL1 in the migration of osteoclast precursors and osteoclastogenesis, and will contribute to the development of new therapies for bone diseases. J. Cell. Physiol. 232: 1739-1745, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tsuyoshi Matsuura
- Department of Cellular Physiological Chemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan.,Oral Implantology and Regenerative Dental Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Shizuko Ichinose
- Research Center for Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Masako Akiyama
- Department of Cellular Physiological Chemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Yuki Kasahara
- Department of Cellular Physiological Chemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Noriko Tachikawa
- Oral Implantology and Regenerative Dental Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Ken-Ichi Nakahama
- Department of Cellular Physiological Chemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
62
|
Hung CM, Hsu YC, Chen TY, Chang CC, Lee MJ. Cyclophosphamide promotes breast cancer cell migration through CXCR4 and matrix metalloproteinases. Cell Biol Int 2017; 41:345-352. [PMID: 28035725 DOI: 10.1002/cbin.10726] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 12/24/2016] [Indexed: 12/21/2022]
Abstract
Cyclophosphamide is indicated for the treatment of cancerous diseases such as breast cancer and cervical cancer. Recent studies have shown that cyclophosphamide may induce cancer metastasis, but the cause of this unexpected adverse effect is not fully understood. In this study, we investigate the effect of cyclophosphamide on cancer cell migration and its correlation to chemokine (C-X-C motif) receptor 4 (CXCR4), a biomarker for cancer metastasis. Two human cancer cell lines with significant difference in endogenous CXCR4 expression, the breast cancer cell line, MDA-MB-231, and the melanoma cell line, MDA-MB-435S, were treated with various concentrations of cyclophosphamide, followed by the assessment of CXCR4 expression and cell migration. We found that the migration ability of MDA-MB-231 cells was enhanced with increasing concentrations of cyclophosphamide, which induced the cell-surface expression of CXCR4, but had no effect on the overall amount of CXCR4. In MDA-MB-435S cells, in which CXCR4 was barely detectable, cyclophosphamide was unable to activate cell-surface CXCR4, and did not promote cell migration. Studies on the mRNA expression profile of matrix metalloproteinases (MMPs) in MDA-MB-231 cells further indicate that MMP9 and MMP13 may be involved in the action of cyclophosphamide. The protein expression of both MMP9 and MMP13 was increased in the presence of cyclophosphamide. Results from this study provide the molecular basis for the possible pathway of cyclophosphamide to induce cancer metastasis.
Collapse
Affiliation(s)
- Chao-Ming Hung
- Department of General Surgery, E-Da Hospital, I-Shou, University, Kaohsiung, Taiwan
| | - Yi-Chiang Hsu
- Graduate Institute of Medical Sciences, Chang Jung Christian University, Tainan, Taiwan.,Innovative Research Center of Medicine, Chang Jung Christian University, Tainan, Taiwan
| | - Tzu-Yu Chen
- Graduate Institute of Medical Sciences, Chang Jung Christian University, Tainan, Taiwan.,Department of Bioscience Technology, Chang Jung Christian University, Tainan, Taiwan
| | - Chi-Chang Chang
- Department of Obstetrics and Gynecology, E-Da Hospital, Kaohsiung, Taiwan
| | - Mon-Juan Lee
- Innovative Research Center of Medicine, Chang Jung Christian University, Tainan, Taiwan.,Department of Bioscience Technology, Chang Jung Christian University, Tainan, Taiwan
| |
Collapse
|
63
|
Takayama T, Dai J, Tachi K, Shohara R, Kasai H, Imamura K, Yamano S. The potential of stromal cell-derived factor-1 delivery using a collagen membrane for bone regeneration. J Biomater Appl 2017; 31:1049-1061. [PMID: 28056602 DOI: 10.1177/0885328216686727] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Stromal cell-derived factor-1 (SDF-1) is a cytokine that is important in stem and progenitor cell recruitment in tissue repair after injury. Regenerative procedures using collagen membranes (CMs) are presently well established in periodontal and implant dentistry. The objective of this study is to test the subsequent effects of the released SDF-1 from a CM on bone regeneration compared to platelet-derived growth factor (PDGF) in vitro and in vivo. For in vitro studies, cell proliferation, alkaline phosphatase activity, and osteoblastic differentiation marker genes were assessed after MC3T3-E1 mouse preosteoblasts were cultured with CMs containing factors. In vivo effects were investigated by placement of CMs containing SDF-1 or PDGF using a rat mandibular bone defect model. At 4 weeks after the surgery, the new bone formation was measured using micro-computed tomography (µCT) and histological analysis. The results of in vitro studies revealed that CM delivery of SDF-1 significantly induced cell proliferation, ALP activity, and gene expression of all osteogenic markers compared to the CM alone or control, similar to PDGF. Quantitative and qualitative µCT analysis for volume of new bone formation and the percentage of new bone area showed that SDF-1-treated groups significantly increased and accelerated bone regeneration compared to control and CM alone. The enhancement of bone formation in SDF-1-treated animals was dose-dependent and with levels similar to those measured with PDGF. These results suggest that a CM with SDF-1 may be a great candidate for growth factor delivery that could be a substitute for PDGF in clinical procedures where bone regeneration is necessary.
Collapse
Affiliation(s)
- Tadahiro Takayama
- 1 Department of Periodontology, Nihon University School of Dentistry, Tokyo, Japan.,2 Division of Advanced Dental Treatment, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| | - Jisen Dai
- 3 Mouse Genotyping Core, New York University Langone Medical Center, New York, NY, USA
| | - Keita Tachi
- 4 Department of Prosthodontics, New York University College of Dentistry, New York, NY, USA
| | - Ryutaro Shohara
- 4 Department of Prosthodontics, New York University College of Dentistry, New York, NY, USA
| | - Hironori Kasai
- 4 Department of Prosthodontics, New York University College of Dentistry, New York, NY, USA
| | - Kentaro Imamura
- 4 Department of Prosthodontics, New York University College of Dentistry, New York, NY, USA
| | - Seiichi Yamano
- 4 Department of Prosthodontics, New York University College of Dentistry, New York, NY, USA
| |
Collapse
|
64
|
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: 539] [Impact Index Per Article: 67.4] [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
|
65
|
Lerner UH. Inflammation-induced Bone Remodeling in Periodontal Disease and the Influence of Post-menopausal Osteoporosis. J Dent Res 2016; 85:596-607. [PMID: 16798858 DOI: 10.1177/154405910608500704] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
During physiological conditions, the skeleton is remodeled in so-called bone multi-cellular units. Such units have been estimated to exist at 1–2 x 106 sites in the adult skeleton. The number and activities of these units are regulated by a variety of hormones and cytokines. In post-menopausal osteoporosis, lack of estrogen leads to increased numbers of bone multi-cellular units and to uncoupling of bone formation and bone resorption, resulting in too little bone laid down by osteoblasts compared with the amount of bone resorbed by osteoclasts. Inflammatory processes in the vicinity of the skeleton, e.g., marginal and apical periodontitis, will affect the remodeling of the nearby bone tissue in such a way that, in most patients, the amount of bone resorbed exceeds that being formed, resulting in net bone loss (inflammation-induced osteolysis). In some patients, however, inflammation-induced bone formation exceeds resorption, and a sclerotic lesion will develop. The cellular and molecular pathogenetic mechanisms in inflammation-induced osteolysis and sclerosis are discussed in the present review. The cytokines believed to be involved in inflammation-induced remodeling are very similar to those suggested to play crucial roles in post-menopausal osteoporosis. In patients with periodontal disease and concomitant post-menopausal osteoporosis, the possibility exists that the lack of estrogen influences the activities of bone cells and immune cells in such a way that the progression of alveolar bone loss will be enhanced. In the present paper, the evidence for and against this hypothesis is presented.
Collapse
Affiliation(s)
- U H Lerner
- Department of Oral Cell Biology, Umeå University, Umeå SE-901 87, Sweden.
| |
Collapse
|
66
|
Shi Y, Wang F, Tiwari S, Yesilbas M, Steubesand N, Weitkamp JT, Klüter T, Lippross S, Eglin D, Seekamp A, Fuchs S. Role of myeloid early endothelial progenitor cells in bone formation and osteoclast differentiation in tissue construct based on hydroxyapatite poly(ester-urethane) scaffolds. J Orthop Res 2016; 34:1922-1932. [PMID: 26945676 DOI: 10.1002/jor.23222] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 02/23/2016] [Indexed: 02/04/2023]
Abstract
Engineering of a vascularized bone construct is a highly challenging task which needs to take into account the impact of different components on the bone regeneration process. Bone repair influencing factors in such constructs range from the material properties and scaffold design, to the interaction of different cell types contributing to bone formation and remodeling or neovascularization, respectively. In this context, early endothelial progenitor cells (EPC), mononuclear cells isolated from the peripheral blood, express the endothelial marker CD31 but also a series of myeloid markers and have been shown to support the formation of vessel-like structures. These cells are also characterized by a highly adaptable phenotype influenced by other cells creating an instructive niche. The present study was designed to investigate the impact of EPC on bone formation or remodeling using a co-culture system of outgrowth endothelial cells, mature endothelial cells isolated from the peripheral blood cell cultures, and mesenchymal stem cells grown on hydroxyapatite poly(ester-urethane) scaffolds. The formation of vessel-like structures in these constructs was shown by CLSM and immunohistochemistry and further evaluated by real time RT-PCR. Osteogenic differentiation in these constructs was investigated by von Kossa, Alizarin Red, and real time PCR. Data indicated that osteogenic differentiation occurred within the constructs after 14 days of culture but without a direct influence by EPC in this process. Finally, although we observed a series of osteoclast related makers in the constructs when EPC were included, no indications for an increased osteoclast-like activity, which might lead to increased bone resorption, were observed. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1922-1932, 2016.
Collapse
Affiliation(s)
- Yang Shi
- Experimental Trauma Surgery, Arnold-Heller-Straße 3, University Medical Center Schleswig-Holstein, UKSH, 24105, Kiel, Germany
| | - Fanlu Wang
- Experimental Trauma Surgery, Arnold-Heller-Straße 3, University Medical Center Schleswig-Holstein, UKSH, 24105, Kiel, Germany
| | - Sanjay Tiwari
- Molecular Imaging North Competence Center (MOINCC), Kiel, Germany
| | - Meran Yesilbas
- Experimental Trauma Surgery, Arnold-Heller-Straße 3, University Medical Center Schleswig-Holstein, UKSH, 24105, Kiel, Germany
| | - Nadine Steubesand
- Experimental Trauma Surgery, Arnold-Heller-Straße 3, University Medical Center Schleswig-Holstein, UKSH, 24105, Kiel, Germany
| | - Jan-Tobias Weitkamp
- Experimental Trauma Surgery, Arnold-Heller-Straße 3, University Medical Center Schleswig-Holstein, UKSH, 24105, Kiel, Germany
| | - Tim Klüter
- Experimental Trauma Surgery, Arnold-Heller-Straße 3, University Medical Center Schleswig-Holstein, UKSH, 24105, Kiel, Germany
| | - Sebastian Lippross
- Experimental Trauma Surgery, Arnold-Heller-Straße 3, University Medical Center Schleswig-Holstein, UKSH, 24105, Kiel, Germany
| | - David Eglin
- AO Research Institute Davos, Davos, Switzerland
| | - Andreas Seekamp
- Experimental Trauma Surgery, Arnold-Heller-Straße 3, University Medical Center Schleswig-Holstein, UKSH, 24105, Kiel, Germany
| | - Sabine Fuchs
- Experimental Trauma Surgery, Arnold-Heller-Straße 3, University Medical Center Schleswig-Holstein, UKSH, 24105, Kiel, Germany
| |
Collapse
|
67
|
Movila A, Ishii T, Albassam A, Wisitrasameewong W, Howait M, Yamaguchi T, Ruiz-Torruella M, Bahammam L, Nishimura K, Van Dyke T, Kawai T. Macrophage Migration Inhibitory Factor (MIF) Supports Homing of Osteoclast Precursors to Peripheral Osteolytic Lesions. J Bone Miner Res 2016; 31:1688-700. [PMID: 27082509 PMCID: PMC5010512 DOI: 10.1002/jbmr.2854] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 03/28/2016] [Accepted: 04/12/2016] [Indexed: 12/11/2022]
Abstract
By binding to its chemokine receptor CXCR4 on osteoclast precursor cells (OCPs), it is well known that stromal cell-derived factor-1 (SDF-1) promotes the chemotactic recruitment of circulating OCPs to the homeostatic bone remodeling site. However, the engagement of circulating OCPs in pathogenic bone resorption remains to be elucidated. The present study investigated a possible chemoattractant role of macrophage migration inhibitory factor (MIF), another ligand for C-X-C chemokine receptor type 4 (CXCR4), in the recruitment of circulating OCPs to the bone lytic lesion. To accomplish this, we used Csf1r-eGFP-knock-in (KI) mice to establish an animal model of polymethylmethacrylate (PMMA) particle-induced calvarial osteolysis. In the circulating Csf1r-eGFP+ cells of healthy Csf1r-eGFP-KI mice, Csf1r+/CD11b+ cells showed a greater degree of RANKL-induced osteoclastogenesis compared to a subset of Csf1r+/RANK+ cells in vitro. Therefore, Csf1r-eGFP+/CD11b+ cells were targeted as functionally relevant OCPs in the present study. Although expression of the two cognate receptors for MIF, CXCR2 and CXCR4, was elevated on Csf1r+/CD11b+ cells, transmigration of OCPs toward recombinant MIF in vitro was facilitated by ligation with CXCR4, but not CXCR2. Meanwhile, the level of PMMA-induced bone resorption in calvaria was markedly greater in wild-type (WT) mice compared to that detected in MIF-knockout (KO) mice. Interestingly, in contrast to the elevated MIF, diminished SDF-1 was detected in a particle-induced bone lytic lesion of WT mice in conjunction with an increased number of infiltrating CXCR4+ OCPs. However, such diminished SDF-1 was not found in the PMMA-injected calvaria of MIF-KO mice. Furthermore, stimulation of osteoblasts with MIF in vitro suppressed their production of SDF-1, suggesting that MIF can downmodulate SDF-1 production in bone tissue. Systemically administered anti-MIF neutralizing monoclonal antibody (mAb) inhibited the homing of CXCR4+ OCPs, as well as bone resorption, in the PMMA-injected calvaria, while increasing locally produced SDF-1. Collectively, these data suggest that locally produced MIF in the inflammatory bone lytic site is engaged in the chemoattraction of circulating CXCR4+ OCPs. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Alexandru Movila
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, MA, USA
| | - Takenobu Ishii
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, MA, USA.,Department of Orthodontics, Tokyo Dental College, Tokyo, Japan
| | - Abdullah Albassam
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, MA, USA.,School of Dental Medicine, Harvard University, Boston, MA, USA.,Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Wichaya Wisitrasameewong
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, MA, USA.,School of Dental Medicine, Harvard University, Boston, MA, USA.,Department of Periodontology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Mohammed Howait
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, MA, USA.,Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Tsuguno Yamaguchi
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, MA, USA.,Research and Development Headquarters, LION Corporation, Kanagawa, Japan
| | | | - Laila Bahammam
- Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Kazuaki Nishimura
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, MA, USA.,Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Thomas Van Dyke
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, MA, USA
| | - Toshihisa Kawai
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, MA, USA.,School of Dental Medicine, Harvard University, Boston, MA, USA
| |
Collapse
|
68
|
Xu Y, Chu N, Qiu X, Gober HJ, Li D, Wang L. The interconnected role of chemokines and estrogen in bone metabolism. Biosci Trends 2016; 10:433-444. [DOI: 10.5582/bst.2016.01072] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Yingping Xu
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, IBS, Shanghai Medical College, Fudan University
- The Academy of Integrative Medicine of Fudan University
| | - Nan Chu
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, IBS, Shanghai Medical College, Fudan University
| | - Xuemin Qiu
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, IBS, Shanghai Medical College, Fudan University
- The Academy of Integrative Medicine of Fudan University
| | | | - Dajin Li
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, IBS, Shanghai Medical College, Fudan University
- The Academy of Integrative Medicine of Fudan University
| | - Ling Wang
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, IBS, Shanghai Medical College, Fudan University
- The Academy of Integrative Medicine of Fudan University
| |
Collapse
|
69
|
Kanbe K, Chiba J, Inoue Y, Taguchi M, Yabuki A. SDF-1 and CXCR4 in synovium are associated with disease activity and bone and joint destruction in patients with rheumatoid arthritis treated with golimumab. Mod Rheumatol 2015; 26:46-50. [PMID: 25995033 DOI: 10.3109/14397595.2015.1054088] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES The aim of this study was to determine whether the levels of stromal cell-derived factor (SDF)-1 and its receptor C-X-C chemokine receptor 4 (CXCR4) in synovium were correlated with clinical outcome and bone and joint destruction in rheumatoid arthritis (RA) patients being treated with golimumab. METHODS Synovial tissues were obtained from 15 golimumab-treated patients and were assessed for SDF-1 and CXCR4 using a new immunohistological scoring system (IH score). The IH score was used to assess correlations between synovial SDF-1 or CXCR4 and the disease activity score (DAS28 CRP), Rooney score, tumor necrosis factor alpha, interleukin-6 (IL-6), CD4, CD20, CD68 and the Assessment of RA by Scoring of Large-Joint Destruction and Healing in Radiographic Imaging (ARASHI) score. Receiver-operating characteristic (ROC) curves were used to predict ARASHI scores from the CXCR4 IH scores. RESULTS SDF-1 strongly correlated with the DAS28 CRP and serum IL-6. CXCR4 correlated with synovial CD4 and the ARASHI score. ROC analysis of CXCR4 and ARASHI scores >10 indicated a cutoff of 12 points on the IH score for predicting joint destruction during treatment. CONCLUSIONS Synovial SDF-1 correlated with disease activity, and its receptor CXCR4 was related to joint destruction in RA patients treated with golimumab.
Collapse
Affiliation(s)
- Katsuaki Kanbe
- a Department of Orthopaedic Surgery , Tokyo Women's Medical University, Medical Center East , Arakawa , Tokyo , Japan
| | - Junji Chiba
- a Department of Orthopaedic Surgery , Tokyo Women's Medical University, Medical Center East , Arakawa , Tokyo , Japan
| | - Yasuo Inoue
- a Department of Orthopaedic Surgery , Tokyo Women's Medical University, Medical Center East , Arakawa , Tokyo , Japan
| | - Masashi Taguchi
- a Department of Orthopaedic Surgery , Tokyo Women's Medical University, Medical Center East , Arakawa , Tokyo , Japan
| | - Akiko Yabuki
- a Department of Orthopaedic Surgery , Tokyo Women's Medical University, Medical Center East , Arakawa , Tokyo , Japan
| |
Collapse
|
70
|
Zhou Y, Deng HW, Shen H. Circulating monocytes: an appropriate model for bone-related study. Osteoporos Int 2015; 26:2561-72. [PMID: 26194495 DOI: 10.1007/s00198-015-3250-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 07/10/2015] [Indexed: 10/23/2022]
Abstract
Peripheral blood monocytes (PBMs) are an important source of precursors of osteoclasts, the bone-resorbing cells and the cytokines produced by PBMs that have profound effects on osteoclast differentiation, activation, and apoptosis. So PBMs represent a highly valuable and unique working cell model for bone-related study. Finding an appropriate working cell model for clinical and (epi-)genomic studies of human skeletal disorders is a challenge. Peripheral blood monocytes (PBMs) can give rise to osteoclasts, the bone-resorbing cells. Particularly, PBMs provide the sole source of osteoclast precursors for adult peripheral skeleton where the bone marrow is normally hematopoietically inactive. PBMs can secrete potent pro- and anti-inflammatory cytokines, which are important for osteoclast differentiation, activation, and apoptosis. Reduced production of PBM cytokines represents a major mechanism for the inhibitory effects of sex hormones on osteoclastogenesis and bone resorption. Abnormalities in PBMs have been linked to various skeletal disorders/traits, strongly supporting for the biological relevance of PBMs with bone metabolism and disorders. Here, we briefly review the origin and further differentiation of PBMs. In particular, we discuss the close relationship between PBMs and osteoclasts, and highlight the utility of PBMs in study the pathophysiological mechanisms underlying various skeletal disorders.
Collapse
Affiliation(s)
- Y Zhou
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University, New Orleans, LA, 70112, USA
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA, 70118, USA
| | - H-W Deng
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University, New Orleans, LA, 70112, USA
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA, 70118, USA
| | - H Shen
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University, New Orleans, LA, 70112, USA.
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA, 70118, USA.
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine, Tulane University, 1440 Canal St., Suite 2001, New Orleans, LA, 70112, USA.
| |
Collapse
|
71
|
Nevius E, Pinho F, Dhodapkar M, Jin H, Nadrah K, Horowitz MC, Kikuta J, Ishii M, Pereira JP. Oxysterols and EBI2 promote osteoclast precursor migration to bone surfaces and regulate bone mass homeostasis. ACTA ACUST UNITED AC 2015; 212:1931-46. [PMID: 26438360 PMCID: PMC4612084 DOI: 10.1084/jem.20150088] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 08/27/2015] [Indexed: 12/13/2022]
Abstract
The mechanisms guiding cells toward bone surfaces are generally unknown. Here, Nevius et al. show that the Gαi protein–coupled receptor EBI2 is expressed in mouse osteoclast precursors to guide these cells toward bone surfaces. Defective EBI2 signaling increased bone mass in male mice and protected female mice from age- and estrogen deficiency–induced osteoporosis. Bone surfaces attract hematopoietic and nonhematopoietic cells, such as osteoclasts (OCs) and osteoblasts (OBs), and are targeted by bone metastatic cancers. However, the mechanisms guiding cells toward bone surfaces are essentially unknown. Here, we show that the Gαi protein–coupled receptor (GPCR) EBI2 is expressed in mouse monocyte/OC precursors (OCPs) and its oxysterol ligand 7α,25-dihydroxycholesterol (7α,25-OHC) is secreted abundantly by OBs. Using in vitro time-lapse microscopy and intravital two-photon microscopy, we show that EBI2 enhances the development of large OCs by promoting OCP motility, thus facilitating cell–cell interactions and fusion in vitro and in vivo. EBI2 is also necessary and sufficient for guiding OCPs toward bone surfaces. Interestingly, OCPs also secrete 7α,25-OHC, which promotes autocrine EBI2 signaling and reduces OCP migration toward bone surfaces in vivo. Defective EBI2 signaling led to increased bone mass in male mice and protected female mice from age- and estrogen deficiency–induced osteoporosis. This study identifies a novel pathway involved in OCP homing to the bone surface that may have significant therapeutic potential.
Collapse
Affiliation(s)
- Erin Nevius
- Department of Immunobiology and Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT 06510
| | - Flavia Pinho
- Department of Immunobiology and Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT 06510
| | - Meera Dhodapkar
- Department of Immunobiology and Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT 06510
| | - Huiyan Jin
- Department of Immunobiology and Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT 06510
| | - Kristina Nadrah
- Department of Immunobiology and Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT 06510
| | - Mark C Horowitz
- Department of Immunobiology and Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT 06510
| | - Junichi Kikuta
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences and WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences and WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - João P Pereira
- Department of Immunobiology and Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT 06510
| |
Collapse
|
72
|
|
73
|
Osteoprotegerin exposure at different stages of osteoclastogenesis differentially affects osteoclast formation and function. Cytotechnology 2015; 68:1325-35. [PMID: 26044733 DOI: 10.1007/s10616-015-9892-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 05/23/2015] [Indexed: 10/23/2022] Open
Abstract
This study aimed to investigate the effects of osteoprotegerin (OPG), a decoy receptor for receptor activator for nuclear factor κB ligand (RANKL), during the various stages of osteoclast differentiation, and additionally investigate its effects on osteoclast adhesion and activity. RAW264.7 murine monocytic cells were incubated with macrophage colony-stimulating factor and RANKL for 1, 3, 5, or 7 days, followed by an additional 24-h incubation in the presence or absence of OPG (80 ng/mL). We examined osteoclast differentiation and adhesion capacity using the tartrate-resistant acid phosphatase (TRAP) assay and immunofluorescence microscopy, and additionally examined cell growth in real time using the xCELLigence system. Furthermore, the expression levels of TRAP, RANK, integrin β3, matrix metalloproteinase 9, cathepsin K, carbonic anhydrase II, and vesicular-type H(+)-ATPase A1 were examined using western blotting. OPG exposure on day 1 enhanced the osteoclast growth curve as well as adhesion, and increased RANK and integrin β3 expression. In contrast, exposure to OPG at later time points (days 3-7) inhibited osteoclast differentiation, adhesion structure formation, and protease expression. In conclusion, the biological effects of OPG exposure at the various stages of osteoclast differentiation were varied, and included the enhanced adhesion and survival of preosteoclasts, the block of differentiation from the early to the terminal stages of osteoclastogenesis, and suppression of mature osteoclast activation following OPG exposure during the terminal differentiation stage, suggesting that the effects of OPG exposure differ based on the stage of differentiation.
Collapse
|
74
|
Liu H, Li M, Du L, Yang P, Ge S. Local administration of stromal cell-derived factor-1 promotes stem cell recruitment and bone regeneration in a rat periodontal bone defect model. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2015; 53:83-94. [PMID: 26042694 DOI: 10.1016/j.msec.2015.04.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 03/03/2015] [Accepted: 04/01/2015] [Indexed: 01/07/2023]
Abstract
Stromal cell-derived factor-1 (SDF-1) recruits adult stem/progenitor cells via its specific receptor, C-X-C motif receptor 4 (CXCR4), to promote heart, kidney and tendon regeneration, but little is known about the effects of SDF-1 on bone regeneration in periodontal diseases. The objective of this study was to investigate whether local administration of SDF-1 in a collagen membrane scaffold enhanced the recruitment of host stem cells and improved periodontal bone defect repair. To this end, bone defects were established on the buccal side of bilateral mandibles in Wistar rats. After application of collagen membranes loaded with SDF-1 or phosphate-buffered saline (PBS) to the defects, the effects of SDF-1 on stem cell recruitment, inflammatory cell responses, angiogenesis, osteoclastogenesis, scaffold degradation, and bone regeneration were evaluated. It showed that SDF-1 recruited host-derived mesenchymal stem cells and hematopoietic stem cells to the wound area and significantly reduced the CD11b+ inflammatory cell response. Moreover, SDF-1 increased vascular formation, induced early bone osteoclastogenesis, accelerated scaffold degradation, and promoted the quality and quantity of regenerated bone. Our results suggest that this cell-free approach by local administration of SDF-1 may be an effective strategy for development as a simple and safe technique for periodontal bone regeneration.
Collapse
Affiliation(s)
- Hongrui Liu
- Shandong Provincial Key Laboratory of Oral Biomedicine, Department of Periodontology, School of Stomatology, Shandong University, Jinan, Shandong Province, China
| | - Minqi Li
- Shandong Provincial Key Laboratory of Oral Biomedicine, Department of Periodontology, School of Stomatology, Shandong University, Jinan, Shandong Province, China
| | - Lingqian Du
- Shandong Provincial Key Laboratory of Oral Biomedicine, Department of Periodontology, School of Stomatology, Shandong University, Jinan, Shandong Province, China; The Second Hospital of Shandong University, Department of Stomatology, Jinan, Shandong Province, China
| | - Pishan Yang
- Shandong Provincial Key Laboratory of Oral Biomedicine, Department of Periodontology, School of Stomatology, Shandong University, Jinan, Shandong Province, China.
| | - Shaohua Ge
- Shandong Provincial Key Laboratory of Oral Biomedicine, Department of Periodontology, School of Stomatology, Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
75
|
Civriz Bozdag S, Tekgunduz E, Altuntas F. The current status in hematopoietic stem cell mobilization. J Clin Apher 2015; 30:273-80. [PMID: 25790158 DOI: 10.1002/jca.21374] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 11/26/2014] [Indexed: 12/12/2022]
Abstract
Hemotopoietic stem cell mobilization with cytokines alone, has still been widely accepted as the initial attempt for stem cell mobilization. Chemotherapy based mobilization can be preferred as first choice in high risk patients or for remobilization. But mobilization failure still remains to be a problem in one third of patients. Salvage mobilization strategies have been composed to give one more chance to 'poor mobilizers'. Synergistic effect of a reversible inhibitor of CXCR4, plerixafor, with G-CSF has opened a new era for these patients. Preemptive approach in predicted poor mobilizers, immediate salvage approach for patients with suboptimal mobilization or remobilization approach of plerixafor in failed mobilizers have all been demonstrated convincing results in various studies. Alternative CXCR4 inhibitors, VLA4 inhibitors, bortezomib, parathormone have also been emerged as novel agents for mobilization failure.
Collapse
Affiliation(s)
| | - Emre Tekgunduz
- Hematology Department, Ankara Oncology Training Hospital, Ankara, Turkey
| | - Fevzi Altuntas
- Hematology Department, Ankara Oncology Training Hospital, Ankara, Turkey
| |
Collapse
|
76
|
Yang T, Zhang J, Cao Y, Zhang M, Jing L, Jiao K, Yu S, Chang W, Chen D, Wang M. Wnt5a/Ror2 mediates temporomandibular joint subchondral bone remodeling. J Dent Res 2015; 94:803-12. [PMID: 25749876 DOI: 10.1177/0022034515576051] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Increased subchondral trabecular bone turnover due to imbalanced bone-resorbing and bone-forming activities is a hallmark of osteoarthritis (OA). Wnt5a/Ror2 signaling, which can derive from bone marrow stromal cells (BMSCs), takes a role in modulating osteoblast and osteoclast formation. We showed previously that experimentally unilateral anterior crossbites (UACs) elicited OA-like lesions in mice temporomandibular joints (TMJs), displaying as subchondral trabecular bone loss. Herein, we tested the role of BMSC-derived Wnt5a/Ror2 signaling in regulating osteoclast precursor migration and differentiation in this process. The data confirmed the decreased bone mass, increased tartrate-resistant acid phosphatase (TRAP)-positive cell number, and enhanced osteoclast activity in TMJ subchondral trabecular bone of UAC-treated rats. Interestingly, the osteoblast activity in the tissue of TMJ subchondral trabecular bone of these UAC-treated rats was also enhanced, displaying as upregulated expressions of osteoblast markers and increased proliferation, migration, and differentiation capabilities of the locally isolated BMSCs. These BMSCs showed an increased CXCL12 protein expression level and upregulated messenger RNA expressions of Rankl, Wnt5a, and Ror2. Ex vivo data showed that their capacities of inducing migration and differentiation of osteoclast precursors were enhanced, and these enhanced capabilities were restrained after blocking their Ror2 signaling using small interfering RNA (siRNA) assays. Reducing Ror2 expression in the BMSC cell line by siRNA or blocking the downstream signalings with specific inhibitors also demonstrated a suppression of the capacity of the BMSC cell line to promote Wnt5a-dependent migration (including SP600125 and cyclosporine A) and differentiation (cyclosporine A only) of osteoclast precursors. These findings support the idea that Wnt5a/Ror2 signaling in TMJ subchondral BMSCs enhanced by UAC promoted BMSCs to increase Cxcl12 and Rankl expression, in which JNK and/or Ca(2+)/NFAT pathways were involved and therefore were engaged in enhancing the migration and differentiation of osteoclast precursors, leading to increased osteoclast activity and an overall TMJ subchondral trabecular bone loss in the UAC-treated rats.
Collapse
Affiliation(s)
- T Yang
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology, School of Stomatology, the Fourth Military Medical University, Xi'an, Shaanxi, China Department of Stomatology, Chinese PLA General Hospital, Beijing, China
| | - J Zhang
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology, School of Stomatology, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Y Cao
- Department of Cardiac Surgery, Air Force General Hospital, PLA, Beijing, China
| | - M Zhang
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology, School of Stomatology, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - L Jing
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology, School of Stomatology, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - K Jiao
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology, School of Stomatology, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - S Yu
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology, School of Stomatology, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - W Chang
- Endocrine Research Unit, Department of Veterans Affairs Medical Center, Department of Medicine, University of California, San Francisco, CA, USA
| | - D Chen
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - M Wang
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology, School of Stomatology, the Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
77
|
Drummond S, Ramachandran S, Torres E, Huang J, Hehre D, Suguihara C, Young KC. CXCR4 blockade attenuates hyperoxia-induced lung injury in neonatal rats. Neonatology 2015; 107:304-11. [PMID: 25825119 PMCID: PMC4458217 DOI: 10.1159/000371835] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 01/02/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND Lung inflammation is a key factor in the pathogenesis of bronchopulmonary dysplasia (BPD). Stromal-derived factor-1 (SDF-1) and its receptor chemokine receptor 4 (CXCR4) modulate the inflammatory response. It is not known if antagonism of CXCR4 alleviates lung inflammation in neonatal hyperoxia-induced lung injury. OBJECTIVE We aimed to determine whether CXCR4 antagonism would attenuate lung injury in rodents with experimental BPD by decreasing pulmonary inflammation. METHODS Newborn rats exposed to normoxia (room air, RA) or hyperoxia (FiO2 = 0.9) from postnatal day 2 (P2) to P16 were randomized to receive the CXCR4 antagonist, AMD3100 or placebo (PL) from P5 to P15. Lung alveolarization, angiogenesis and inflammation were evaluated at P16. RESULTS Compared to the RA pups, hyperoxic PL pups had a decrease in alveolarization, reduced lung vascular density and increased lung inflammation. In contrast, AMD3100-treated hyperoxic pups had improved alveolarization and increased angiogenesis. This improvement in lung structure was accompanied by a decrease in the macrophage and neutrophil counts in the bronchoalveolar lavage fluid and reduced lung myeloperoxidase activity. CONCLUSION CXCR4 antagonism decreases lung inflammation and improves alveolar and vascular structure in neonatal rats with experimental BPD. These findings suggest a novel therapeutic strategy to alleviate lung injury in preterm infants with BPD.
Collapse
Affiliation(s)
- Shelley Drummond
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Fla., USA
| | | | | | | | | | | | | |
Collapse
|
78
|
Park YJ, Kim JY, Park J, Choi JJ, Kim WU, Cho CS. Bone erosion is associated with reduction of circulating endothelial progenitor cells and endothelial dysfunction in rheumatoid arthritis. Arthritis Rheumatol 2014; 66:1450-60. [PMID: 24991663 DOI: 10.1002/art.38352] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVE To identify factors influencing endothelial progenitor cell (EPC) counts in patients with rheumatoid arthritis (RA). METHODS The number of circulating CD34+/ vascular endothelial growth factor receptor 2-positive EPCs was measured in 126 RA patients and 46 non-RA control patients. Endothelial function was assessed by brachial flow-mediated dilation (FMD). Serum CXCL12 concentrations were determined using an enzyme-linked immunosorbent assay. EPCs and FMD were measured at baseline and after 24 weeks of anti-tumor necrosis factor (TNF) therapy in 29 patients with active RA. RESULTS The numbers of circulating EPCs were significantly lower in the RA patients than in the non-RA controls. In multivariate analysis, older age, reduced levels of high-density lipoprotein cholesterol, and higher bone erosion scores were independent risk factors for reduced EPC counts in RA patients. Serum CXCL12 levels correlated negatively with EPC counts, but positively with bone erosion scores. FMD was impaired in RA patients, and a decreased FMD in RA was closely associated with a higher bone erosion score and a reduced EPC count. In addition, EPC counts were restored by anti-TNF therapy, and this increase was paralleled by improvement in FMD. Interestingly, restoration of EPC counts was attenuated in patients with higher bone erosion scores than in those with lower scores, despite similar levels of improvement in disease activity. CONCLUSION The numbers of circulating EPCs in RA patients are reduced and are inversely correlated with serum levels of CXCL12. Reduced EPC counts are closely associated not only with bone erosion, but also with endothelial dysfunction.
Collapse
|
79
|
Pandey MK, Kale VP, Song C, Sung SS, Sharma AK, Talamo G, Dovat S, Amin SG. Gambogic acid inhibits multiple myeloma mediated osteoclastogenesis through suppression of chemokine receptor CXCR4 signaling pathways. Exp Hematol 2014; 42:883-96. [PMID: 25034231 DOI: 10.1016/j.exphem.2014.07.261] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 06/11/2014] [Accepted: 07/05/2014] [Indexed: 11/17/2022]
Abstract
Bone disease, characterized by the presence of lytic lesions and osteoporosis is the hallmark of multiple myeloma (MM). Stromal cell-derived factor 1α (SDF-1α) and its receptor, CXC chemokine receptor 4 (CXCR4), has been implicated as a regulator of bone resorption, suggesting that agents that can suppress SDF1α/CXCR4 signaling might inhibit osteoclastogenesis, a process closely linked to bone resorption. We, therefore, investigated whether gambogic acid (GA), a xanthone, could inhibit CXCR4 signaling and suppress osteoclastogenesis induced by MM cells. Through docking studies we predicted that GA directly interacts with CXCR4. This xanthone down-regulates the expression of CXCR4 on MM cells in a dose- and time-dependent manner. The down-regulation of CXCR4 was not due to proteolytic degradation, but rather GA suppresses CXCR4 mRNA expression by inhibiting nuclear factor-kappa B (NF-κB) DNA binding. This was further confirmed by quantitative chromatin immunoprecipitation assay, as GA inhibits p65 binding at the CXCR4 promoter. GA suppressed SDF-1α-induced chemotaxis of MM cells and downstream signaling of CXCR4 by inhibiting phosphorylation of Akt, p38, and Erk1/2 in MM cells. GA abrogated the RANKL-induced differentiation of macrophages to osteoclasts in a dose- and time-dependent manner. In addition, we found that MM cells induced differentiation of macrophages to osteoclasts, and that GA suppressed this process. Importantly, suppression of osteoclastogenesis by GA was mediated through IL-6 inhibition. Overall, our results show that GA is a novel inhibitor of CXCR4 expression and has a strong potential to suppress osteoclastogenesis mediated by MM cells.
Collapse
Affiliation(s)
- Manoj K Pandey
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA.
| | - Vijay P Kale
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA
| | - Chunhua Song
- Division of Pediatric Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA
| | - Shen-shu Sung
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA
| | - Arun K Sharma
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA
| | - Giampaolo Talamo
- Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA
| | - Sinisa Dovat
- Division of Pediatric Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA
| | - Shantu G Amin
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA
| |
Collapse
|
80
|
|
81
|
Villalvilla A, Gomez R, Roman-Blas JA, Largo R, Herrero-Beaumont G. SDF-1 signaling: a promising target in rheumatic diseases. Expert Opin Ther Targets 2014; 18:1077-87. [DOI: 10.1517/14728222.2014.930440] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
82
|
Šućur A, Katavić V, Kelava T, Jajić Z, Kovačić N, Grčević D. Induction of osteoclast progenitors in inflammatory conditions: key to bone destruction in arthritis. INTERNATIONAL ORTHOPAEDICS 2014; 38:1893-903. [DOI: 10.1007/s00264-014-2386-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 05/13/2014] [Indexed: 12/14/2022]
|
83
|
Tanaka K, Hashizume M, Mihara M, Yoshida H, Suzuki M, Matsumoto Y. Anti-interleukin-6 receptor antibody prevents systemic bone mass loss via reducing the number of osteoclast precursors in bone marrow in a collagen-induced arthritis model. Clin Exp Immunol 2014; 175:172-80. [PMID: 24028747 DOI: 10.1111/cei.12201] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2013] [Indexed: 11/27/2022] Open
Abstract
Systemic bone loss is a hallmark of rheumatoid arthritis (RA). Inflammatory cytokines such as interleukin (IL)-6 promote bone resorption by osteoclasts. Sphingosine-1-phosphate (S1P) controls the migration of osteoclast precursor cells (OCPs) between the blood and bone marrow, in part via S1P receptors (S1PR1 and S1PR2) expressed on the surface of OCPs. OCPs (CD11b(+) Gr-1(low+med) ) isolated from bone marrow of DBA/1J mice were stimulated with IL-6. S1P-directed chemotaxis of OCPs was evaluated using a transwell plate. mRNA expression of S1PR1 and S1PR2 was measured. DBA/1J mice were immunized with bovine type II collagen (days 0 and 21) and anti-mouse IL-6 receptor antibody (MR16-1) was administered on days 0 and/or 21. Trabecular bone volume was analysed using micro-computed tomography. The percentage of OCPs in tibial bone marrow and S1PR1 and S1PR2 mRNA expression in OCPs were measured. IL-6 stimulation significantly decreased S1P-directed chemotaxis of OCPs. IL-6 induced S1PR2 mRNA expression, but not S1PR1 mRNA expression, in OCPs. Bone volume was significantly lower in arthritic mice than in non-arthritic control mice on day 35. Treatment of immunized mice with MR16-1 significantly inhibited bone loss. In MR16-1-treated mice, the percentage of OCPs and expression of S1PR2 mRNA was each decreased compared with arthritic mice on day 14, but not on day 35. IL-6 increased the number of OCPs in tibial bone marrow via up-regulating S1PR2, thus playing a crucial role in systemic bone loss induced by inflammation.
Collapse
Affiliation(s)
- Keisuke Tanaka
- Product Research Department, Fuji-Gotemba Research Laboratories, Chugai Pharmaceutical Co. Ltd, Gotemba, Shizuoka, Japan
| | | | | | | | | | | |
Collapse
|
84
|
Kim HR, Kim KW, Kim BM, Jung HG, Cho ML, Lee SH. Reciprocal activation of CD4+ T cells and synovial fibroblasts by stromal cell-derived factor 1 promotes RANKL expression and osteoclastogenesis in rheumatoid arthritis. Arthritis Rheumatol 2014; 66:538-48. [PMID: 24574213 DOI: 10.1002/art.38286] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 11/14/2013] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Stromal cell-derived factor 1 (SDF-1) is a chemokine that is involved in the bone-destructive process in rheumatoid arthritis (RA) and bony metastasis in malignancy. This study was undertaken to determine the role and mechanism of SDF-1 in RA-associated osteoclastogenesis. METHODS The expression of SDF-1, tumor necrosis factor α (TNFα), and RANKL in RA synovial tissue was analyzed using confocal microscopy. After synovial fibroblasts and CD4+ T cells were treated with SDF-1, RANKL messenger RNA expression was determined by real-time and reverse transcription polymerase chain reaction. Osteoclastogenesis was assessed by counting tartrate-resistant acid phosphatase-positive multinucleated cells in CD14+ monocytes cultured with SDF-1 in the presence of anticytokine antibodies or signal inhibitors and in monocytes cocultured with SDF-1-pretreated synovial fibroblasts and CD4+ T cells. RESULTS RANKL, TNFα, and SDF-1 were coexpressed in the lining and sublining of RA synovium. SDF-1 stimulated RANKL expression in RA synovial fibroblasts and CD4+ T cells, and TNFα inhibition reduced this stimulation. When monocytes isolated from human peripheral blood were cultured with SDF-1, they were differentiated into osteoclasts in the absence of RANKL. Monocytes were also differentiated into osteoclasts when they were cocultured with SDF-1-pretreated synovial fibroblasts or CD4+T cells; however, this osteoclastogenesis was reduced by TNFα inhibition. CONCLUSION Our findings indicate that SDF-1 induces osteoclastogenesis directly and indirectly via up-regulating RANKL expression in RA synovial fibroblasts and CD4+ T cells, and that this is mediated by TNFα. The axis of SDF-1 and RANKL is a potential therapeutic target for RA-associated bone destruction.
Collapse
Affiliation(s)
- Hae-Rim Kim
- Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, South Korea
| | | | | | | | | | | |
Collapse
|
85
|
The effect of simvastatin on chemotactic capability of SDF-1α and the promotion of bone regeneration. Biomaterials 2014; 35:4489-98. [DOI: 10.1016/j.biomaterials.2014.02.025] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 02/13/2014] [Indexed: 12/29/2022]
|
86
|
C/EBPβ mediates osteoclast recruitment by regulating endothelial progenitor cell expression of SDF-1α. PLoS One 2014; 9:e91217. [PMID: 24618682 PMCID: PMC3949754 DOI: 10.1371/journal.pone.0091217] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 02/10/2014] [Indexed: 12/12/2022] Open
Abstract
Integration of tissue-engineered bone grafts with the host bone is vital for the healing of critical-size bone defects. An important aspect of this process is bone resorption, which must be carried out by osteoclasts derived from the host. However, the mechanism underlying recruitment of host osteoclast precursors to graft sites remains unclear. Endothelial progenitor cells (EPCs) mobilize from the bone marrow into the circulation and home to sites of angiogenesis such as tissue remodeling. Since EPCs express SDF-1, and C/EBPβ is known to regulate SDF-1α expression, we hypothesized that EPCs may recruit CXCR4-expressing host osteoclast precursors to the repair area and that this recruitment may be mediated through C/EBPβ signaling. Using an inflammatory EPC model we showed that EPCs upregulate protein levels of both SDF-1α and C/EBPβ. A luciferase assay confirmed that C/EBPβ acts on the SDF-1α promoter in these cells, and that binding is increased under conditions of inflammation, while silencing of C/EBPβ reduces expression of SDF-1 α and C/EBPβ. Using RAW264.7 cells as a model of osteoclastic monocyte precursors, we investigated their responses to migratory factors in EPC conditioned medium. We showed that RAW264.7 cells migrate towards conditioned medium from EPCs treated with IL-1β, an effect which could be abolished by silencing C/EBPβ in EPCs, and was almost completely blocked by silencing CXCR4 in RAW264.7 cells. These findings show that EPCs respond to inflammatory stimuli by signaling to osteoclast precursors via SDF-1, and that C/EBPβ mediates this response.
Collapse
|
87
|
Dynamic Cross Talk between S1P and CXCL12 Regulates Hematopoietic Stem Cells Migration, Development and Bone Remodeling. Pharmaceuticals (Basel) 2013; 6:1145-69. [PMID: 24276423 PMCID: PMC3818832 DOI: 10.3390/ph6091145] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 08/18/2013] [Accepted: 09/04/2013] [Indexed: 12/23/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are mostly retained in a quiescent non-motile mode in their bone marrow (BM) niches, shifting to a migratory cycling and differentiating state to replenish the blood with mature leukocytes on demand. The balance between the major chemo-attractants CXCL12, predominantly in the BM, and S1P, mainly in the blood, dynamically regulates HSC recruitment to the circulation versus their retention in the BM. During alarm situations, stress-signals induce a decrease in CXCL12 levels in the BM, while S1P levels are rapidly and transiently increased in the circulation, thus favoring mobilization of stem cells as part of host defense and repair mechanisms. Myeloid cytokines, including G-CSF, up-regulate S1P signaling in the BM via the PI3K pathway. Induced CXCL12 secretion from stromal cells via reactive oxygen species (ROS) generation and increased S1P1 expression and ROS signaling in HSCs, all facilitate mobilization. Bone turnover is also modulated by both CXCL12 and S1P, regulating the dynamic BM stromal microenvironment, osteoclasts and stem cell niches which all functionally express CXCL12 and S1P receptors. Overall, CXCL12 and S1P levels in the BM and circulation are synchronized to mutually control HSC motility, leukocyte production and osteoclast/osteoblast bone turnover during homeostasis and stress situations.
Collapse
|
88
|
Dziak R. The role of sphingosine-1-phosphate (S1P) and lysophosphatidic acid (LPA) in regulation of osteoclastic and osteoblastic cells. Immunol Invest 2013; 42:510-8. [DOI: 10.3109/08820139.2013.823804] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
89
|
|
90
|
Guang LG, Boskey AL, Zhu W. Age-related CXC chemokine receptor-4-deficiency impairs osteogenic differentiation potency of mouse bone marrow mesenchymal stromal stem cells. Int J Biochem Cell Biol 2013; 45:1813-20. [DOI: 10.1016/j.biocel.2013.05.034] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 05/16/2013] [Accepted: 05/28/2013] [Indexed: 12/16/2022]
|
91
|
Hou GQ, Guo C, Song GH, Fang N, Fan WJ, Chen XD, Yuan L, Wang ZQ. Lipopolysaccharide (LPS) promotes osteoclast differentiation and activation by enhancing the MAPK pathway and COX-2 expression in RAW264.7 cells. Int J Mol Med 2013; 32:503-10. [PMID: 23740407 DOI: 10.3892/ijmm.2013.1406] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Accepted: 05/29/2013] [Indexed: 11/06/2022] Open
Abstract
Bone degradation is a serious complication of chronic inflammatory diseases such as septic arthritis, osteomyelitis and infected orthopedic implant failure. At present, effective therapeutic treatments for lipopolysaccharide (LPS)-induced bone destruction are limited to antibiotics and surgical repair in chronic inflammatory diseases. The present study aimed to evaluate the mechanism of LPS on osteoclast differentiation and activation. RAW264.7 cells were non-induced, or induced by the receptor activator of nuclear factor-κB (RANK) ligand (RANKL) and macrophage-colony stimulating factor (M-CSF), and then treated with LPS. Following treatment, the number of osteoclasts and cell viability were measured. The expression of osteoclast-related genes including tartrate-resistant acid phosphatase (TRAP), matrix metalloproteinase-9 (MMP-9), cathepsin K (CK), carbonic anhydrase II (CAII) and cyclooxygenase-2 (COX-2) was determined by RT-PCR. Protein levels of RANK, tumor necrosis factor receptor-associated factor 6 (TRAF6), COX-2 and mitogen-activated protein kinases (MAPK) were measured using western blotting assays. LPS promoted osteoclast differentiation of RAW264.7 cells and differentiated osteoclasts. LPS significantly increased mRNA expression of osteoclast-related genes in RAW264.7 cells. Differentiated osteoclasts were treated with LPS (100 ng/ml) and the results showed a significantly increased mRNA expression of osteoclast-related genes and protein levels of RANK, TRAF6 and COX-2. Furthermore, LPS at 100 ng/ml significantly promoted the MAPK pathway including increasing the phosphorylation of c-Jun N-terminal kinases (JNK) and the phosphorylation of the extracellular signal-regulated kinase (ERK1/2). In conclusion, LPS promoted osteoclast differentiation and activation by enhancing RANK signaling and COX-2 expression. LPS also promoted osteoclast differentiation via activation of the JNK and ERK1/2 cell proliferation pathways.
Collapse
Affiliation(s)
- Guo-Qing Hou
- First Affiliated Hospital, Medical College of Shantou University, Shantou, Guangdong 515041, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
92
|
Niwa T, Mizukoshi K, Azuma Y, Kashimata M, Shibutani T. Fundamental study of osteoclast chemotaxis toward chemoattractants expressed in periodontitis. J Periodontal Res 2013; 48:773-80. [PMID: 23586648 DOI: 10.1111/jre.12068] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2013] [Indexed: 01/25/2023]
Abstract
BACKGROUND AND OBJECTIVE Periodontitis is a chronic inflammatory disease that leads to bone resorption by osteoclasts (OCs). Several factors contribute to the differentiation of OCs from hematopoietic precursors. Cellular chemotactic factors are expressed in periodontitis tissue, but the effects of these chemoattractants on OCs are not well understood. Here we examined the effects of chemoattractants produced in inflamed periodontal tissue on OC chemotaxis. MATERIAL AND METHODS Rat bone-marrow OCs were cultured in OC culture medium for 3 or 6 d. Using EZ-TAXIScan™, the chemotactic response of these OCs to several chemoattractants [monocyte chemotactic protein-1; macrophage inflammatory protein 1α; regulated on activation, normal T-cell expressed and secreted; stromal cell-derived factor-1α; and complement activation product 5a (C5a)] was measured. In addition, we measured the effect of C5a-specific inhibitors on chemotactic responses toward C5a. The recorded chemotactic responses were quantitatively analysed using ImageJ software. RESULTS Chemoattractants associated with periodontal disease significantly increased the chemotactic activity of differentiated rat OCs in a concentration-dependent manner, with C5a inducing the highest chemotactic activity of OCs cultured for 3 or 6 d. The C5a-specific inhibitor significantly inhibited chemotaxis toward C5a in a concentration-dependent manner. CONCLUSION We suggest that C5a plays an important role in pathologic bone resorption in periodontal disease by stimulating the chemotaxis of OCs. Therefore, C5a is a potential target for the treatment of periodontal disease.
Collapse
Affiliation(s)
- T Niwa
- Department of Periodontology, Division of Oral Infections and Health Sciences, Asahi University School of Dentistry, Mizuho, Japan
| | | | | | | | | |
Collapse
|
93
|
Yellowley C. CXCL12/CXCR4 signaling and other recruitment and homing pathways in fracture repair. BONEKEY REPORTS 2013; 2:300. [PMID: 24422056 DOI: 10.1038/bonekey.2013.34] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 02/08/2013] [Indexed: 02/06/2023]
Abstract
Cell recruitment, migration and homing to the fracture site are essential for the inflammatory process, neovascularization, chondrogenesis, osteogenesis and ultimately bone remodeling. Mesenchymal stem cells (MSCs) are required to navigate from local sources such as the periosteum and local bone marrow, and may also be recruited from the circulation and distant bone marrow. While the local recruitment process may involve matrix binding and degradation, systemic recruitment may utilize extravasation, a process used by leukocytes to exit the vasculature. CXCL12 (stromal cell-derived factor-1 (SDF-1)), a member of the CXC family of chemokines, is thought to have an important role in cell migration at the fracture site. However, there are many molecules upregulated in the hematoma and callus that have chemotactic potential not only for inflammatory cells but also for endothelial cells and MSCs. Surprisingly, there is little direct data to support their role in cell homing during bone healing. Current therapeutics for bone regeneration utilize local or systemic stem cell transplantation. More recently, a novel strategy that involves mobilization of large numbers of endogenous stem and progenitor cells from bone marrow into the circulation has been shown to have positive effects on bone healing. A more complete understanding of the molecular mechanisms underlying cell recruitment and homing subsequent to fracture will facilitate the fine-tuning of such strategies for bone.
Collapse
Affiliation(s)
- Clare Yellowley
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis , Davis, CA, USA
| |
Collapse
|
94
|
Kikuta J, Ishii M. Osteoclast migration, differentiation and function: novel therapeutic targets for rheumatic diseases. Rheumatology (Oxford) 2012; 52:226-34. [PMID: 23024017 DOI: 10.1093/rheumatology/kes259] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
RA is a chronic autoimmune disease characterized by joint synovial inflammation and progressive cartilage/bone destruction. Although various kinds of RA drug have been developed worldwide, there are currently no established methods for preventing RA-associated bone destruction, the most severe outcome of this disease. One of the major pathogenic factors in arthritic bone destruction is the enhanced activity of osteoclasts at inflammatory sites. Osteoclasts are bone-resorbing giant polykaryons that differentiate from mononuclear macrophage/monocyte-lineage haematopoietic precursors. Upon stimulation by cytokines, such as M-CSF and RANK ligand, osteoclast precursor monocytes migrate and attach onto the bone surface (migration). They then fuse with each other to form giant cells (differentiation) and mediate bone resorption (function). In this review, we summarize the current understanding regarding the mechanisms underlying these three dynamic steps of osteoclastic activity and discuss novel lines of osteoclast-targeted therapies that will impact future treatment of RA.
Collapse
Affiliation(s)
- Junichi Kikuta
- Laboratory of Cellular Dynamics, Immunology Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka, Japan
| | | |
Collapse
|
95
|
Guang LG, Boskey AL, Zhu W. Regulatory role of stromal cell-derived factor-1 in bone morphogenetic protein-2-induced chondrogenic differentiation in vitro. Int J Biochem Cell Biol 2012; 44:1825-33. [PMID: 22771956 DOI: 10.1016/j.biocel.2012.06.033] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 06/22/2012] [Accepted: 06/27/2012] [Indexed: 12/18/2022]
Abstract
Chemokine stromal cell-derived factor-1 (SDF-1) signals via binding to its primary transmembrane receptor, cysteine (C)-X-C chemokine receptor-4 (CXCR4). We previously reported that SDF-1 regulates osteogenic differentiation of mesenchymal stem/progenitor cells (MPCs) induced by bone morphogenetic protein-2 (BMP2). Although BMP2 is also capable of inducing chondrogenic differentiation of MPCs, the involvement of SDF-1 signaling in this function of BMP2 remains unknown. In this study, we aimed to test the role of SDF-1 signaling involved in BMP2-induced chondrogenic differentiation, using ATDC5 chondroprogenitors and mouse bone marrow-derived mesenchymal stromal cells (BMSCs). Our data showed that blocking of the SDF-1/CXCR4 pathway inhibits the differentiation of these cells into the chondrocytic lineages in response to BMP2 stimulation, evidenced by the reduced expression of type II collagen α1 (Col2α1), aggrecan, and type X collagen α1 (Col10α1), markers for chondrogenic differentiation. This effect of blocking SDF-1 signaling on BMP2-chondrogenic differentiation is associated with suppressed Sox9 and Runx2 expression (key transcription factors required for early and late stages of chondrogenic differentiation, respectively) and mediated via inhibiting intracellular Smad and Erk activation. Moreover, we found that addition of exogenous SDF-1 protein synergistically enhances the BMP2-induced chondrogenic differentiation in a dose-dependent manner. Collectively, our results demonstrated a novel role of SDF-1 signaling in regulating BMP2-induced chondrogenic differentiation in vitro. These data provide new insights into molecular mechanisms underlying BMP2-osteo/chondrogenesis, and may lead to the identification of new therapeutic targets and strategies to improve cartilage repair and regeneration in broad orthopaedic situations.
Collapse
Affiliation(s)
- Liang G Guang
- Musculoskeletal Integrity Program, Hospital for Special Surgery, New York, NY 10021, USA
| | | | | |
Collapse
|
96
|
Aggarwal R, Lu J, Kanji S, Joseph M, Das M, Noble GJ, McMichael BK, Agarwal S, Hart RT, Sun Z, Lee BS, Rosol TJ, Jackson R, Mao HQ, Pompili VJ, Das H. Human umbilical cord blood-derived CD34+ cells reverse osteoporosis in NOD/SCID mice by altering osteoblastic and osteoclastic activities. PLoS One 2012; 7:e39365. [PMID: 22724005 PMCID: PMC3377665 DOI: 10.1371/journal.pone.0039365] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 05/23/2012] [Indexed: 12/18/2022] Open
Abstract
Background Osteoporosis is a bone disorder associated with loss of bone mineral density and micro architecture. A balance of osteoblasts and osteoclasts activities maintains bone homeostasis. Increased bone loss due to increased osteoclast and decreased osteoblast activities is considered as an underlying cause of osteoporosis. Methods and Findings The cures for osteoporosis are limited, consequently the potential of CD34+ cell therapies is currently being considered. We developed a nanofiber-based expansion technology to obtain adequate numbers of CD34+ cells isolated from human umbilical cord blood, for therapeutic applications. Herein, we show that CD34+ cells could be differentiated into osteoblastic lineage, in vitro. Systemically delivered CD34+ cells home to the bone marrow and significantly improve bone deposition, bone mineral density and bone micro-architecture in osteoporotic mice. The elevated levels of osteocalcin, IL-10, GM-CSF, and decreased levels of MCP-1 in serum parallel the improvements in bone micro-architecture. Furthermore, CD34+ cells improved osteoblast activity and concurrently impaired osteoclast differentiation, maturation and functionality. Conclusions These findings demonstrate a novel approach utilizing nanofiber-expanded CD34+ cells as a therapeutic application for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Reeva Aggarwal
- Cardiovascular Stem Cell Research Laboratory, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Jingwei Lu
- Cardiovascular Stem Cell Research Laboratory, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Suman Kanji
- Cardiovascular Stem Cell Research Laboratory, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Matthew Joseph
- Cardiovascular Stem Cell Research Laboratory, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Manjusri Das
- Cardiovascular Stem Cell Research Laboratory, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Garrett J. Noble
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, United States of America
| | - Brooke K. McMichael
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Sudha Agarwal
- Division of Oral Biology, Department of Orthopedics, College of Dentistry, The Ohio State University, Columbus, Ohio, United States of America
| | - Richard T. Hart
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, United States of America
| | - Zongyang Sun
- Division of Oral Biology, Department of Orthopedics, College of Dentistry, The Ohio State University, Columbus, Ohio, United States of America
| | - Beth S. Lee
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Thomas J. Rosol
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Rebecca Jackson
- Division of Endocrinology, Diabetes and Metabolism, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Hai-Quan Mao
- Department of Materials Science and Engineering, John's Hopkins University, Baltimore, Maryland, United States of America
| | - Vincent J. Pompili
- Cardiovascular Stem Cell Research Laboratory, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Hiranmoy Das
- Cardiovascular Stem Cell Research Laboratory, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
97
|
Ishii M, Kikuta J. Sphingosine-1-phosphate signaling controlling osteoclasts and bone homeostasis. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1831:223-7. [PMID: 22691949 DOI: 10.1016/j.bbalip.2012.06.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 06/04/2012] [Indexed: 12/31/2022]
Abstract
Bone is a dynamic organ that is continuously turned over during growth, even in adults. During bone remodeling, homeostasis is regulated by the balance between bone formation by osteoblasts and bone resorption by osteoclasts. However, in pathological conditions such as osteoporosis, osteopetrosis, arthritic joint destruction, and bone metastasis, this equilibrium is disrupted. Since osteoclasts are excessively activated in osteolytic diseases, the inhibition of osteoclast function has been a major therapeutic strategy. It has recently been demonstrated that sphingosine-1-phosphate (S1P), a biologically active lysophospholipid that is enriched in blood, controls the trafficking of osteoclast precursors between the circulation and bone marrow cavities via G protein-coupled receptors, S1PRs. While S1PR1 mediates chemoattraction toward S1P in bone marrow, where S1P concentration is low, S1PR2 mediates chemorepulsion in blood, where the S1P concentration is high. The regulation of precursor recruitment may represent a novel therapeutic strategy for controlling osteoclast-dependent bone remodeling. By means of intravital multiphoton imaging of bone tissues, we have recently revealed that the reciprocal action of S1P controls the migration of osteoclast precursors between bone tissues and blood stream. Imaging technologies have enabled us to visualize the in situ behaviors of different cell types in intact tissues. In this review we also discuss future perspectives on this new method in the field of bone biology and medical sciences in general. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.
Collapse
|
98
|
Zoccoli A, Iuliani M, Pantano F, Imperatori M, Intagliata S, Vincenzi B, Marchetti P, Papapietro N, Denaro V, Tonini G, Santini D. Premetastatic niche: ready for new therapeutic interventions? Expert Opin Ther Targets 2012; 16 Suppl 2:S119-29. [PMID: 22443277 DOI: 10.1517/14728222.2012.656092] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Bone marrow-derived cells (BMDC) localize in premetastatic niche through chemokines and integrins signals and establish clusters that precede the arrival of even single metastatic tumor cell at distant site. CSCs demonstrate an increased metastatic propensity and would seem likely candidates for the acquisition of migratory capabilities and propagation of heterogeneous tumor cell populations to different target organs. Sonic Hedgehog (SHH), FOXM1 and Notch pathways and signaling molecules such as integrin and chemokine could dictate their fate. AREAS COVERED In this review, the molecular mechanisms of premetastatic niche onset are summarized. EXPERT OPINION Premetastatic niche is defined as a fertile microenvironment that forms in metastatic target organ and facilitates the invasion, survival and/or proliferation of metastatic tumor cells, providing a novel mechanism for the promotion of metastasis. Drugs targeting premetastatic niche could represent a new promising therapeutic approach in the treatment of bone metastases.
Collapse
Affiliation(s)
- Alice Zoccoli
- Campus Bio-Medico University, Department of Medical Oncology, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Wu SH, Zhong ZM, Chen JT. Low-magnitude high-frequency vibration inhibits RANKL-induced osteoclast differentiation of RAW264.7 cells. Int J Med Sci 2012; 9:801-7. [PMID: 23136544 PMCID: PMC3491440 DOI: 10.7150/ijms.4838] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2012] [Accepted: 10/15/2012] [Indexed: 11/13/2022] Open
Abstract
Osteoclasts are the key participants in regulation of bone mass. Low-magnitude high-frequency vibration (LMHFV) has been found to be anabolic to bone in vivo. This study aimed to investigate the effect of LMHFV on osteoclast differentiation in vitro. Murine monocyte cell line RAW264.7 cells in the presence of receptor activator of nuclear factor-kappaB ligand (RANKL) were treated with or without LMHFV at 45 Hz (0.3 g) for 15 min day(-1). Tartrate resistant acid phosphatase (TRAP)-positive multinucleated cells (MNCs) and actin ring formation were evaluated. Expression of the osteoclast-specific genes, such as cathepsin K, matrix metallopeptidase-9 (MMP-9) and TRAP, were analyzed using real time-PCR. c-Fos, an osteoclast-specific transcription factor, was determined using Western blot. We found that LMHFV significantly decreased the number of RANKL-induced TRAP-positive MNCs (P<0.01), and inhibited the actin ring formation. The mRNA expression of the cathepsin K, MMP-9 and TRAP were down-regulated by LMHFV intervention (all P<0.001). Furthermore, LMHFV also inhibited the expression of c-Fos protein in the RANKL-treated RAW264.7 cells (P<0.05). Our results suggest that LMHFV can inhibit the RANKL-induced osteoclast differentiation of RAW264.7 cells, which give some new insight into the anabolic effects of LMHFV on bone.
Collapse
Affiliation(s)
- Song-Hui Wu
- Department of Orthopedic and Spinal Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | | | | |
Collapse
|
100
|
Ishii T, Kawamura S, Nishiyama I, Kikuta J, Ishii M. Use of intravital microscopy and in vitro chemotaxis assays to study the roles of sphingosine-1-phosphate in bone homeostasis. Methods Mol Biol 2012; 874:129-139. [PMID: 22528444 DOI: 10.1007/978-1-61779-800-9_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
We describe a method to visualize the migration of osteoclast precursors within intact murine bone -marrow in real time using intravital multiphoton microscopy. Conventionally, cell migration has been evaluated using in vitro systems, such as transmigration assays. Although these methods are convenient for quantification and are highly reproducible, these in vitro assay systems may not accurately reflect in vivo cellular behavior. In addition to in vitro analyses, recent technological progress in two-photon excitation-based laser microscopy has enabled the visualization of dynamic cell behavior deep inside intact living organs. Combining this imaging method with in vitro chemoattraction analyses, we have revealed that sphingosine-1-phosphate (S1P), a lipid mediator enriched in blood, bidirectionally controls the trafficking of osteoclast precursors between the circulation and bone marrow cavities via G protein-coupled receptors (GPCRs).
Collapse
Affiliation(s)
- Taeko Ishii
- Laboratory of Cellular Dynamics, WPI-Immunology Frontier Research Center, Osaka University, Suita, Japan
| | | | | | | | | |
Collapse
|