101
|
Javed S, Sohail A, Nutini A. Integrative modeling of drug therapy and the bone turnover. Clin Biomech (Bristol, Avon) 2018; 60:141-148. [PMID: 30359867 DOI: 10.1016/j.clinbiomech.2018.10.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 10/05/2018] [Accepted: 10/12/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Denosumab has been successfully used for the postmenopausal osteoporosis treatment. This research is focused on the computational analysis of the effect of denosumab on bone remodeling. METHODS Inspired by the advancement in the field of multiscale modeling , this research encompasses on the cellular and molecular bone remodeling key players. The model is designed to cover all the dominant interacting factors and their respective gradients. During this research, we have performed numerical experiments to validate our mathematical model, by interfacing it with the parametric values available in the literature. FINDINGS The novelty of our work relies in the fact that we have considered the effect of estrogen, sclerostin and NFATc1 during osteoporosis and their combined effect with the variable effect of denosumab during therapy. INTERPRETATIONS From our analysis, we have concluded that denosumab suppresses osteoclast differentiation, that results in reduced bone resorption. These results are in agreement with the experimental findings.
Collapse
Affiliation(s)
- Sana Javed
- Department of Mathematics, Comsats University Islamabad, Lahore Campus, 54000, Pakistan
| | - Ayesha Sohail
- Department of Mathematics, Comsats University Islamabad, Lahore Campus, 54000, Pakistan.
| | - Alessandro Nutini
- Center for Study in Motor Science, 94 via di Tiglio, loc. Arancio, 55100 Lucca, Italy
| |
Collapse
|
102
|
Sato M, Asano T, Hosomichi J, Ono T, Nakata T. Optogenetic manipulation of intracellular calcium by BACCS promotes differentiation of MC3T3-E1 cells. Biochem Biophys Res Commun 2018; 506:716-722. [PMID: 30376992 DOI: 10.1016/j.bbrc.2018.10.107] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 10/17/2018] [Indexed: 10/28/2022]
Abstract
Bone remodeling is maintained through the balance between bone formation by osteoblasts and bone resorption by osteoclasts. Previous studies suggested that intracellular Ca2+ signaling plays an important role in the differentiation of osteoblasts; however, the molecular mechanism of Ca2+ signaling in the differentiation of osteoblasts remains unclear. To elucidate the effect of Ca2+ signaling in osteoblasts, we employed an optogenetic tool, blue light-activated Ca2+ channel switch (BACCS). BACCS was used to spatiotemporally control intracellular Ca2+ with blue light stimulation. MC3T3-E1 cells, which have been used as a model of differentiation from preosteoblast to osteoblast, were promoted to differentiate by BACCS expression and rhythmical blue light stimulation. The results indicated that intracellular Ca2+ change from the outside of the cells can regulate signaling for differentiation of MC3T3-E1 cells. Our findings provide evidence that Ca2+ could cause osteoblast differentiation.
Collapse
Affiliation(s)
- Moe Sato
- Department of Orthodontic Science, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan; Department of Cell Biology, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan; The Center for Brain Integration Research (CBIR), Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - Toshifumi Asano
- Department of Cell Biology, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan; The Center for Brain Integration Research (CBIR), Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - Jun Hosomichi
- Department of Orthodontic Science, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - Takashi Ono
- Department of Orthodontic Science, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - Takao Nakata
- Department of Cell Biology, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan; The Center for Brain Integration Research (CBIR), Tokyo Medical and Dental University, Tokyo, 113-8510, Japan.
| |
Collapse
|
103
|
Bai H, Zhu H, Yan Q, Shen X, Lu X, Wang J, Li J, Chen L. TRPV2-induced Ca 2+-calcineurin-NFAT signaling regulates differentiation of osteoclast in multiple myeloma. Cell Commun Signal 2018; 16:68. [PMID: 30326911 PMCID: PMC6191893 DOI: 10.1186/s12964-018-0280-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/05/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Myeloma bone disease (MBD) can cause bone destruction and increase the level of Ca2+ concentration in the bone marrow microenvironment by stimulating osteoclastic differentiation. Nevertheless, the relationships between MBD and highly efficient stimuli of Ca2+ in multiple myeloma (MM) progression, and possible regulatory mechanisms are poorly defined. Here, we reported that the nonselective cation channel transient receptor potential vanilloid 2 (TRPV2) plays a functional role in Ca2+ oscillations and osteoclastogenesis. METHODS To investigate the expression of TRPV2 in MM, we analyzed publicly available MM data sets and performed immunohistochemistry in MM patients. The correlations between TRPV2 expression levels and osteoclast-related cytokines were analyzed. Fluo-4 staining and ELISA assays were used to assess the regulated function of TRPV2 in intracellular Ca2+ and cytokines. Western blotting and Chromatin immunoprecipitation (ChIP) assays were performed to explore the signaling pathway of TRPV2-induced osteoclastic differentiation. Real-time PCR, Western blotting, ELISA and tartrate-resistant acid phosphatase (TRAP) staining were performed to detect the biological effects of TRPV2 inhibitor on osteoclastogenesis. RESULTS The functional expression of TRPV2, involved in the osteolysis through gating the calcium influx, was changed in the MM cells cultured in a high Ca2+ environment. Mechanistically, TRPV2 modulates nuclear factor-κB ligand (RANKL)-dependent osteoclastic differentiation through the Ca2+-calcineurin-NFAT signaling pathway. Of clinical relevance, systemic administration with SKF96365 could attenuate the MM-induced osteoclast formation in vitro. CONCLUSIONS Our study uncovers the possible roles of TRPV2, which enhances MBD, suggesting that targeting osteocyte-MM cells interactions through blockade of TRPV2 channel may provide a promising treatment strategy in MM.
Collapse
Affiliation(s)
- Hua Bai
- Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Huayuan Zhu
- Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Qing Yan
- Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Xuxing Shen
- Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Xiupan Lu
- Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Juejin Wang
- Department of Physiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Jianyong Li
- Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Lijuan Chen
- Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
104
|
Son A, Kang N, Kang JY, Kim KW, Yang YM, Shin DM. TRPM3/TRPV4 regulates Ca2+-mediated RANKL/NFATc1 expression in osteoblasts. J Mol Endocrinol 2018; 61:207-218. [PMID: 30328352 DOI: 10.1530/jme-18-0051] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Mechanical stress plays an important role in the regulation of bone turnover. However, the mechanism underlying hypo-osmotic stress-induced cellular response in osteoblasts remains poorly understood. In this study, we investigated the effect of hypotonic stress on the expression of bone remodeling factors, including the receptor activator of nuclear factor-kappa B ligand (RANKL) and the nuclear factor of activated T cells type c1 (NFATc1) in primary mouse osteoblasts and MC3T3-E1 cells. Hypo-osmotic stress induced significant increases in RANKL mRNA expression and intracellular Ca2+ concentration ([Ca2+]i) from the extracellular space. Hypo-osmotic stress-induced effects on [Ca2+]i and RANKL and NFATc1 protein expression were decreased by antagonists of transient receptor potential melastatin 3 (TRPM3) and vanilloid 4 (TRPV4). Agonists of TRPM3 and TRPV4 activated [Ca2+]i and RANKL and NFATc1 protein expression. Furthermore, genetic suppression of Trpm3 and Trpv4 reduced hypo-osmotic stress-induced effects in mouse osteoblasts. These results suggest that hypo-osmotic stress induces increases in [Ca2+]i through TRPM3 and TRPV4 to regulate RANKL and NFATc1 expression in mouse osteoblastic cells and that mechanical stress-activated TRP channels may play a critical role in bone remodeling.
Collapse
Affiliation(s)
- Aran Son
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Korea
| | - Namju Kang
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Korea
- BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea
| | - Jung Yun Kang
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Korea
- BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea
| | - Ki Woo Kim
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Korea
| | - Yu-Mi Yang
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Korea
| | - Dong Min Shin
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Korea
- BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea
| |
Collapse
|
105
|
Park KL, Oh DG, Kim YO, Song KS, Ahn DW. Rosiglitazone suppresses RANKL-induced NFATc1 autoamplification by disrupting the physical interaction between NFATc1 and PPARγ. FEBS Open Bio 2018; 8:1584-1593. [PMID: 30338210 PMCID: PMC6168694 DOI: 10.1002/2211-5463.12513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 02/20/2018] [Accepted: 03/15/2018] [Indexed: 11/09/2022] Open
Abstract
Receptor activator of nuclear factor-κB ligand (RANKL) is required for initiation of osteoclastogenesis, with the signaling pathway including the NF-kB, c-Fos, and nuclear factor of activated T cells, cytoplasmic 1 (NFATc1) transcription factors. Because NFATc1 expression is autoamplified, we investigated the molecular mechanism by which peroxisome proliferator-activated receptor gamma (PPARγ) activation by the thiazolidinedione drug rosiglitazone decreases NFATc1 expression during RANKL stimulation. Western blotting demonstrated that rosiglitazone attenuated the increase in NFATc1 protein level induced by RANKL without affecting that of PPARγ. Immunofluorescence data indicated that rosiglitazone tended to suppress RANKL-induced NFATc1 nuclear translocation, partly by reducing calcineurin activity, as reflected by the observed decrease in nuclear NFATc1 abundance. On coimmunoprecipitation, the intensity of the physical interaction between NFATc1 and PPARγ was unexpectedly higher in the RANKL-stimulated group than in the control, but rosiglitazone reduced this to basal levels. Furthermore, RANKL failed to elevate mRNA expression of NFATc1 after PPARγ knockdown. ChIP assay indicated that rosiglitazone significantly reduced the binding of NFATc1 to its own promoter despite RANKL stimulation. These findings suggest that PPARγ activation by rosiglitazone blocks NFATc1 from binding to its own promoter, thereby reducing RANKL-induced NFATc1 autoamplification.
Collapse
Affiliation(s)
- Kyeong-Lok Park
- Department of Dentistry Kosin University Gospel Hospital Seo-gu Korea
| | - Da-Gyo Oh
- Department of Physiology Kosin University College of Medicine Seo-gu Korea
| | - Young-Ok Kim
- Department of Pathology Kosin University College of Medicine Seo-gu Korea
| | - Kyeong-Seob Song
- Department of Physiology Kosin University College of Medicine Seo-gu Korea
| | - Do-Whan Ahn
- Department of Physiology Kosin University College of Medicine Seo-gu Korea
| |
Collapse
|
106
|
Thu HE, Hussain Z, Mohamed IN, Shuid AN. Eurycoma longifolia, a promising suppressor of RANKL-induced differentiation and activation of osteoclasts: An in vitro mechanistic evaluation. J Ayurveda Integr Med 2018; 10:102-110. [PMID: 30120052 PMCID: PMC6598823 DOI: 10.1016/j.jaim.2017.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 06/26/2017] [Accepted: 07/09/2017] [Indexed: 11/10/2022] Open
Abstract
Background Eurycoma longifolia (E. longifolia) has gained remarkable recognition due to its promising efficacy of stimulating bone formation in androgen-deficient osteoporosis. Numerous in vivo studies have explored the effects of E. longifolia on osteoporosis; however, the in vitro cellular mechanism was not discovered yet. Objectives The present study was aimed to investigate the effect of E. longifolia on the proliferation, differentiation and maturation of osteoclasts and the translational mechanism of inhibition of osteoclastogenesis using RAW 264.7 cells as an in vitro osteoclastic model. Materials and methods Having assessed cytotoxicity, the cell viability, cell proliferation rate and osteoclastic differentiation capacity of E. longifolia was investigated by evaluating the tartrate-resistant acid phosphatase (TRAP) activity in receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL)-induced osteoclasts. Taken together, the time-mannered expression of osteoclast-related protein biomarkers such as matrix metallopeptidase-9 (MMP-9), cathepsin-K, TRAP, nuclear factor of activated T-cells cytoplasmic 1 (NFATc1), superoxide (free radicals) generation and superoxide dismutase activity were also measured to comprehend the mechanism of osteoclastogenesis. Results E. longifolia did not show significant effects on cytotoxicity and cell proliferation of RAW 264.7 cells; however, a significant inhibition of cells differentiation and maturation of osteoclasts was observed. Moreover, a significant down-regulation of RANKL-induced TRAP activity and expression of MMP-9, cathepsin-K, TRAP, NFATc1 and generation of superoxide and enhanced superoxide dismutase activity was observed in E. longifolia treated cell cultures. Conclusion We anticipated that E. longifolia that enhances bone regeneration on the one hand and suppresses osteoclast’s maturation on the other hand may have great therapeutic value in treating osteoporosis and other bone-erosive diseases such as rheumatoid arthritis and metastasis associated with bone loss.
Collapse
Affiliation(s)
- Hnin Ei Thu
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia (The National University of Malaysia), Jalan Yaacob Latif 56000, Cheras, Malaysia
| | - Zahid Hussain
- Department of Pharmaceutics, Faculty of Pharmacy, Universiti Teknologi MARA, Puncak Alam Campus, Bandar Puncak Alam 42300, Selangor, Malaysia
| | - Isa Naina Mohamed
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia (The National University of Malaysia), Jalan Yaacob Latif 56000, Cheras, Malaysia
| | - Ahmad Nazrun Shuid
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia (The National University of Malaysia), Jalan Yaacob Latif 56000, Cheras, Malaysia.
| |
Collapse
|
107
|
Qiu C, Shen H, Fu X, Xu C, Deng H. Meta-Analysis of Genome-Wide Association Studies Identifies Novel Functional CpG-SNPs Associated with Bone Mineral Density at Lumbar Spine. Int J Genomics 2018; 2018:6407257. [PMID: 30159320 PMCID: PMC6109501 DOI: 10.1155/2018/6407257] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/19/2018] [Indexed: 12/16/2022] Open
Abstract
Osteoporosis is a serious public health issue, which is mostly characterized by low bone mineral density (BMD). To search for additional genetic susceptibility loci underlying BMD variation, an effective strategy is to focus on testing of specific variants with high potential of functional effects. Single nucleotide polymorphisms (SNPs) that introduce or disrupt CpG dinucleotides (CpG-SNPs) may alter DNA methylation levels and thus represent strong candidate functional variants. Here, we performed a targeted GWAS for 63,627 potential functional CpG-SNPs that may affect DNA methylation in bone-related cells, in five independent cohorts (n = 5905). By meta-analysis, 9 CpG-SNPs achieved a genome-wide significance level (p < 7.86 × 10-7) for association with lumbar spine BMD and additional 15 CpG-SNPs showed suggestive significant (p < 5.00 × 10-5) association, of which 2 novel SNPs rs7231498 (NFATC1) and rs7455028 (ESR1) also reached a genome-wide significance level in the joint analysis. Several identified CpG-SNPs were mapped to genes that have not been reported for association with BMD in previous GWAS, such as NEK3 and NFATC1 genes, highlighting the enhanced power of targeted association analysis for identification of novel associations that were missed by traditional GWAS. Interestingly, several genomic regions, such as NEK3 and LRP5 regions, contained multiple significant/suggestive CpG-SNPs for lumbar spine BMD, suggesting that multiple neighboring CpG-SNPs may synergistically mediate the DNA methylation level and gene expression pattern of target genes. Furthermore, functional annotation analyses suggested a strong regulatory potential of the identified BMD-associated CpG-SNPs and a significant enrichment in biological processes associated with protein localization and protein signal transduction. Our results provided novel insights into the genetic basis of BMD variation and highlighted the close connections between genetic and epigenetic mechanisms of complex disease.
Collapse
Affiliation(s)
- Chuan Qiu
- Department of Global Biostatistics and Data Science, Center for Bioinformatics and Genomics, School of Public Health and Tropical Medicine, New Orleans 70112, USA
| | - Hui Shen
- Department of Global Biostatistics and Data Science, Center for Bioinformatics and Genomics, School of Public Health and Tropical Medicine, New Orleans 70112, USA
| | - Xiaoying Fu
- Department of Global Biostatistics and Data Science, Center for Bioinformatics and Genomics, School of Public Health and Tropical Medicine, New Orleans 70112, USA
| | - Chao Xu
- Department of Global Biostatistics and Data Science, Center for Bioinformatics and Genomics, School of Public Health and Tropical Medicine, New Orleans 70112, USA
| | - Hongwen Deng
- Department of Global Biostatistics and Data Science, Center for Bioinformatics and Genomics, School of Public Health and Tropical Medicine, New Orleans 70112, USA
- School of Basic Medical Science, Central South University, Changsha 410013, China
| |
Collapse
|
108
|
T'Jonck W, Guilliams M, Bonnardel J. Niche signals and transcription factors involved in tissue-resident macrophage development. Cell Immunol 2018; 330:43-53. [PMID: 29463401 PMCID: PMC6108424 DOI: 10.1016/j.cellimm.2018.02.005] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 02/07/2018] [Accepted: 02/10/2018] [Indexed: 12/25/2022]
Abstract
Tissue-resident macrophages form an essential part of the first line of defense in all tissues of the body. Next to their immunological role, they play an important role in maintaining tissue homeostasis. Recently, it was shown that they are primarily of embryonic origin. During embryogenesis, precursors originating in the yolk sac and fetal liver colonize the embryonal tissues where they develop into mature tissue-resident macrophages. Their development is governed by two distinct sets of transcription factors. First, in the pre-macrophage stage, a core macrophage program is established by lineage-determining transcription factors. Under the influence of tissue-specific signals, this core program is refined by signal-dependent transcription factors. This nurturing by the niche allows the macrophages to perform tissue-specific functions. In the last 15 years, some of these niche signals and transcription factors have been identified. However, detailed insight in the exact mechanism of development is still lacking.
Collapse
Affiliation(s)
- Wouter T'Jonck
- Laboratory of Myeloid Cell Ontogeny and Functional Specialization, VIB-UGent Center for Inflammation Research, Technologiepark 927, 9052 Gent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052 Gent, Belgium.
| | - Martin Guilliams
- Laboratory of Myeloid Cell Ontogeny and Functional Specialization, VIB-UGent Center for Inflammation Research, Technologiepark 927, 9052 Gent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052 Gent, Belgium
| | - Johnny Bonnardel
- Laboratory of Myeloid Cell Ontogeny and Functional Specialization, VIB-UGent Center for Inflammation Research, Technologiepark 927, 9052 Gent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052 Gent, Belgium.
| |
Collapse
|
109
|
Yu J, Zanotti S, Schilling L, Canalis E. Nuclear factor of activated T cells 2 is required for osteoclast differentiation and function in vitro but not in vivo. J Cell Biochem 2018; 119:9334-9345. [PMID: 30010214 DOI: 10.1002/jcb.27212] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 05/30/2018] [Indexed: 01/09/2023]
Abstract
Nuclear factor of activated T cells (NFAT) c2 is important for the immune response and it compensates for NFATc1 for its effects on osteoclastogenesis, but its role in this process is not established. To study the function of NFATc2 in the skeleton, Nfatc2loxP/loxP mice, where the Nfact2 exon 2 is flanked by loxP sequences, were created and mated with mice expressing the Cre recombinase under the control of the Lyz2 promoter. Bone marrow-derived macrophage (BMM) from Lyz2Cre/WT ;Nfatc2Δ/Δ mice cultured in the presence of macrophage-colony stimulating factor and receptor activator of NF-κB ligand exhibited a decrease in the number and size of osteoclasts and a smaller sealing zone when compared to BMMs from Nfatc2loxP/loxP littermate controls. Bone resorption was decreased in osteoclasts from Lyz2Cre/WT ;Nfatc2Δ/Δ mice. This demonstrates that NFATc2 is necessary for optimal osteoclast maturation and function in vitro. Male and female Lyz2Cre/WT ;Nfatc2Δ/Δ mice did not exhibit an obvious skeletal phenotype by microcomputed tomography (μCT) at either 1 or 4 months of age when compared to Nfatc2loxP/loxP sex-matched littermates. Bone histomorphometry confirmed the μCT results, and conditional 4-month-old Lyz2Cre/WT ;Nfatc2Δ/Δ mice did not exhibit changes in parameters of bone histomorphometry. In conclusion, NFATc2 is necessary for optimal osteoclastogenesis in vitro, but its downregulation in the myeloid lineage has no consequences in skeletal remodeling in vivo.
Collapse
Affiliation(s)
- Jungeun Yu
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut
| | - Stefano Zanotti
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut
- Department of Medicine, UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut
| | - Lauren Schilling
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut
| | - Ernesto Canalis
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut
- Department of Medicine, UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut
| |
Collapse
|
110
|
Abstract
PURPOSE OF REVIEW Bone is constantly being remodeled throughout adult life through constant anabolic and catabolic actions that maintain tissue homeostasis. A number of hormones, cytokines growth factors, and the proximity of various cells to bone surfaces influence this process. Inflammatory changes at the bone microenvironment result in alterations leading to both excessive bone loss and bone formation. Detailed understanding of the physiological and pathological mechanisms that dictate these changes will allow us to harness inflammatory signals in bone regeneration. RECENT FINDINGS Recent reports have suggested that inflammatory signals are able to stimulate transcription factors that regulate osteoblast differentiation from their precursors. SUMMARY In this review, we summarized current understanding of the roles of inflammation in bone resorption and bone formation, which give rise to different disorders and discuss the huge potential of harnessing these inflammatory signals to achieve bone regeneration.
Collapse
Affiliation(s)
- Iannis E Adamopoulos
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis.,Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children-Northern California, Sacramento, California, USA
| |
Collapse
|
111
|
Sandker MJ, Duque LF, Redout EM, Klijnstra EC, Steendam R, Kops N, Waarsing JH, van Weeren R, Hennink WE, Weinans H. Degradation, Intra-Articular Biocompatibility, Drug Release, and Bioactivity of Tacrolimus-Loaded Poly(d-l-lactide-PEG)-b-poly(l-lactide) Multiblock Copolymer-Based Monospheres. ACS Biomater Sci Eng 2018; 4:2390-2403. [DOI: 10.1021/acsbiomaterials.8b00116] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Maria J. Sandker
- Department of Orthopaedics, Erasmus Medical Centre, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
- Department of Orthopaedics, UMC Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Luisa F. Duque
- InnoCore Pharmaceuticals, L.J. Zielstraweg 1, 9713 GX Groningen, The Netherlands
| | - Everaldo M. Redout
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | - Evelien C. Klijnstra
- InnoCore Pharmaceuticals, L.J. Zielstraweg 1, 9713 GX Groningen, The Netherlands
| | - Rob Steendam
- InnoCore Pharmaceuticals, L.J. Zielstraweg 1, 9713 GX Groningen, The Netherlands
| | - Nicole Kops
- Department of Orthopaedics, Erasmus Medical Centre, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Jan H. Waarsing
- Department of Orthopaedics, Erasmus Medical Centre, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Rene van Weeren
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | - Wim E. Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3512 JE Utrecht, The Netherlands
| | - Harrie Weinans
- Department of Orthopaedics and Department of Rheumatology, UMC Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
- Department of Biomechanical Engineering, TUDelft, Mekelweg 2, 2628 CD Delft, The Netherlands
| |
Collapse
|
112
|
Mizuguchi T, Nakashima M, Kato M, Okamoto N, Kurahashi H, Ekhilevitch N, Shiina M, Nishimura G, Shibata T, Matsuo M, Ikeda T, Ogata K, Tsuchida N, Mitsuhashi S, Miyatake S, Takata A, Miyake N, Hata K, Kaname T, Matsubara Y, Saitsu H, Matsumoto N. Loss-of-function and gain-of-function mutations in PPP3CA cause two distinct disorders. Hum Mol Genet 2018; 27:1421-1433. [PMID: 29432562 DOI: 10.1093/hmg/ddy052] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 02/05/2018] [Indexed: 02/07/2023] Open
Abstract
Calcineurin is a calcium (Ca2+)/calmodulin-regulated protein phosphatase that mediates Ca2+-dependent signal transduction. Here, we report six heterozygous mutations in a gene encoding the alpha isoform of the calcineurin catalytic subunit (PPP3CA). Notably, mutations were observed in different functional domains: in addition to three catalytic domain mutations, two missense mutations were found in the auto-inhibitory (AI) domain. One additional frameshift insertion that caused premature termination was also identified. Detailed clinical evaluation of the six individuals revealed clinically unexpected consequences of the PPP3CA mutations. First, the catalytic domain mutations and frameshift mutation were consistently found in patients with nonsyndromic early onset epileptic encephalopathy. In contrast, the AI domain mutations were associated with multiple congenital abnormalities including craniofacial dysmorphism, arthrogryposis and short stature. In addition, one individual showed severe skeletal developmental defects, namely, severe craniosynostosis and gracile bones (severe bone slenderness and perinatal fractures). Using a yeast model system, we showed that the catalytic and AI domain mutations visibly result in decreased and increased calcineurin signaling, respectively. These findings indicate that different functional effects of PPP3CA mutations are associated with two distinct disorders and suggest that functional approaches using a simple cellular system provide a tool for resolving complex genotype-phenotype correlations.
Collapse
Affiliation(s)
- Takeshi Mizuguchi
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Mitsuko Nakashima
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | - Mitsuhiro Kato
- Department of Pediatrics, Showa University School of Medicine, Tokyo 142-8666, Japan
| | - Nobuhiko Okamoto
- Department of Medical Genetics, Osaka Women's and Children's Hospital, Osaka 594-1101, Japan
| | - Hirokazu Kurahashi
- Department of Pediatrics, Aichi Medical University, Aichi 480-1195, Japan
| | - Nina Ekhilevitch
- The Genetics Institute, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Masaaki Shiina
- Department of Biochemistry, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Gen Nishimura
- Center for Intractable Diseases, Saitama Medical University Hospital, Saitama 350-0495, Japan
| | - Takashi Shibata
- Department of Child Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Muneaki Matsuo
- Department of Pediatrics, Saga University Faculty of Medicine, Saga 849-8501, Japan
| | - Tae Ikeda
- Department of Pediatric Neurology, Osaka Women's and Children's Hospital, Osaka 594-1101, Japan
| | - Kazuhiro Ogata
- Department of Biochemistry, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Naomi Tsuchida
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Satomi Mitsuhashi
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Satoko Miyatake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
- Clinical Genetics Department, Yokohama City University Hospital, Yokohama 236-0004, Japan
| | - Atsushi Takata
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Noriko Miyake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Kenichiro Hata
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Tadashi Kaname
- Department of Genome Medicine, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Yoichi Matsubara
- Department of Medical Genetics, Tohoku University School of Medicine, Sendai 980-8574, Japan
- National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Hirotomo Saitsu
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| |
Collapse
|
113
|
Choi H, Srikanth S, Atti E, Pirih FQ, Nervina JM, Gwack Y, Tetradis S. Deletion of Orai1 leads to bone loss aggravated with aging and impairs function of osteoblast lineage cells. Bone Rep 2018; 8:147-155. [PMID: 29955633 PMCID: PMC6020256 DOI: 10.1016/j.bonr.2018.03.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 02/02/2023] Open
Abstract
Osteoblast lineage cells, a group of cells including mesenchymal progenitors, osteoblasts, and osteocytes, are tightly controlled for differentiation, proliferation and stage-specific functions in processes of skeletal development, growth and maintenance. Recently, the plasma membrane calcium channel Orai1 was highlighted for its role in skeletal development and osteoblast differentiation. Yet the roles of Orai1 in osteoblast lineage cells at various stages of maturation have not been investigated. Herein we report the severe bone loss that occurred in Orai1−/− mice, aggravated by aging, as shown by the microcomputed tomography (mCT) and bone histomorphometry analysis of 8-week and 12-week old Orai1−/− mice and sex-matched WT littermates. We also report that Orai1 deficiency affected the differentiation, proliferation, and type I collagen secretion of primary calvarial osteoblasts, mesenchymal progenitors, and osteocytes in Orai1−/− mice; specifically, our study revealed a significant decrease in the expression of osteocytic genes Fgf23, DMP1 and Phex in the cortical long bone of Orai1−/− mice; a defective cellular and nuclear morphology of Orai1−/− osteocytes; and defective osteogenic differentiation of Orai1−/− primary calvarial osteoblasts (pOBs), including a decrease in extracellular-secretion of type I collagen. An increase in the mesenchymal progenitor population of Orai1−/− bone marrow cells was indicated by a colony forming unit-fibroblasts (CFU-F) assay, and the increased proliferation of Orai1−/− pOBs was indicated by an MTT assay. Notably, Orai1 deficiency reduced the nuclear localization and transcription activity of the Nuclear Factor of Activated T-cell c1 (NFATc1), a calcium-regulated transcription factor, in pOBs. Altogether, our study demonstrated the crucial role of Orai1 in bone development and maintenance, via its diverse effects on osteoblast lineage cells from mesenchymal progenitors to osteocytes. Severe bone loss in adult Orai1-/- mice was aggravated by aging. Orai1 deficiency affected function, differentiation and proliferation of osteoblast lineage cells, from mesenchymal progenitors to and osteocytes. Orai1 deficiency reduced the nuclear localization and transcription activity of NFATc1, a calcium-regulated transcription factor, in primary calvarial osteoblasts.
Collapse
Affiliation(s)
- Hyewon Choi
- Division of Oral Biology and Medicine, School of Dentistry, University of California at Los Angeles, 10833 Le Conte Ave., Los Angeles, CA 90095-1668, United States
| | - Sonal Srikanth
- Department of Physiology, David Geffen School of Medicine, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095-1751, United States
| | - Elisa Atti
- Division of Oral Biology and Medicine, School of Dentistry, University of California at Los Angeles, 10833 Le Conte Ave., Los Angeles, CA 90095-1668, United States
| | - Flavia Q Pirih
- Section of Periodontics, School of Dentistry, University of California at Los Angeles, 10833 Le Conte Ave., Los Angeles, CA 90095-1668, United States
| | - Jeanne M Nervina
- Section of Orthodontics, School of Dentistry, University of California at Los Angeles, 10833 Le Conte Ave., Los Angeles, CA 90095-1668, United States
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095-1751, United States
| | - Sotirios Tetradis
- Division of Diagnostic and Surgical Sciences, School of Dentistry, University of California at Los Angeles, 10833 Le Conte Ave., Los Angeles, CA 90095-1668, United States
| |
Collapse
|
114
|
Marballi KK, Gallitano AL. Immediate Early Genes Anchor a Biological Pathway of Proteins Required for Memory Formation, Long-Term Depression and Risk for Schizophrenia. Front Behav Neurosci 2018; 12:23. [PMID: 29520222 PMCID: PMC5827560 DOI: 10.3389/fnbeh.2018.00023] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 01/29/2018] [Indexed: 01/02/2023] Open
Abstract
While the causes of myriad medical and infectious illnesses have been identified, the etiologies of neuropsychiatric illnesses remain elusive. This is due to two major obstacles. First, the risk for neuropsychiatric disorders, such as schizophrenia, is determined by both genetic and environmental factors. Second, numerous genes influence susceptibility for these illnesses. Genome-wide association studies have identified at least 108 genomic loci for schizophrenia, and more are expected to be published shortly. In addition, numerous biological processes contribute to the neuropathology underlying schizophrenia. These include immune dysfunction, synaptic and myelination deficits, vascular abnormalities, growth factor disruption, and N-methyl-D-aspartate receptor (NMDAR) hypofunction. However, the field of psychiatric genetics lacks a unifying model to explain how environment may interact with numerous genes to influence these various biological processes and cause schizophrenia. Here we describe a biological cascade of proteins that are activated in response to environmental stimuli such as stress, a schizophrenia risk factor. The central proteins in this pathway are critical mediators of memory formation and a particular form of hippocampal synaptic plasticity, long-term depression (LTD). Each of these proteins is also implicated in schizophrenia risk. In fact, the pathway includes four genes that map to the 108 loci associated with schizophrenia: GRIN2A, nuclear factor of activated T-cells (NFATc3), early growth response 1 (EGR1) and NGFI-A Binding Protein 2 (NAB2); each of which contains the "Index single nucleotide polymorphism (SNP)" (most SNP) at its respective locus. Environmental stimuli activate this biological pathway in neurons, resulting in induction of EGR immediate early genes: EGR1, EGR3 and NAB2. We hypothesize that dysfunction in any of the genes in this pathway disrupts the normal activation of Egrs in response to stress. This may result in insufficient electrophysiologic, immunologic, and neuroprotective, processes that these genes normally mediate. Continued adverse environmental experiences, over time, may thereby result in neuropathology that gives rise to the symptoms of schizophrenia. By combining multiple genes associated with schizophrenia susceptibility, in a functional cascade triggered by neuronal activity, the proposed biological pathway provides an explanation for both the polygenic and environmental influences that determine the complex etiology of this mental illness.
Collapse
Affiliation(s)
- Ketan K. Marballi
- Department of Basic Medical Sciences and Psychiatry, University of Arizona College of Medicine—Phoenix, Phoenix, AZ, United States
| | - Amelia L. Gallitano
- Department of Basic Medical Sciences and Psychiatry, University of Arizona College of Medicine—Phoenix, Phoenix, AZ, United States
| |
Collapse
|
115
|
Son HE, Kim KM, Kim EJ, Jang WG. Kisspeptin-10 (KP-10) stimulates osteoblast differentiation through GPR54-mediated regulation of BMP2 expression and activation. Sci Rep 2018; 8:2134. [PMID: 29391507 PMCID: PMC5794871 DOI: 10.1038/s41598-018-20571-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 01/15/2018] [Indexed: 11/09/2022] Open
Abstract
Kisspeptin-10 (KP-10) acts as a tumor metastasis suppressor via its receptor, G-protein-coupled receptor 54 (GPR54). The KP-10-GPR54 system plays an important role in embryonic kidney development. However, its function in osteoblast differentiation is unknown. Osteoblast differentiation is controlled by a range of hormones and cytokines, such as bone morphogenetic protein (BMPs), and multiple transcription factors, such as Runt-related transcription factor 2 (Runx2), alkaline phosphatase (ALP), and Distal-less homeobox 5 (Dlx5). In the present study, KP-10-treatment significantly increased the expression of osteogenic genes, including mRNA and protein levels of BMP2, in C3H10T1/2 cells. Moreover, KP-10 induced BMP2-luc activity and increased phosphorylation of Smad1/5/9. In addition, NFATc4 specifically mediated KP-10-induced BMP2 gene expression. However, KP-10 treatment did not induce expression of the BMP2 and Runx2 genes in GPR54-/- cells. To examine whether KP-10 induced secretion of BMP2 to the culture medium, we used the conditioned-medium (C.M) of KP-10 treated medium on C3H10T1/2 cells. Dlx5 and Runx2 expressions were higher in GPR54-/- cells treated with C.M than in those treated with KP-10. These results demonstrate that BMP2 protein has an autocrine effect upon KP-10 treatment. Taken together, these findings suggest that KP-10/GPR54 signaling induces osteoblast differentiation via NFATc4-mediated BMP2 expression.
Collapse
Affiliation(s)
- Hyo-Eun Son
- Department of Biotechnology, School of Engineering, Daegu University, Gyeongbuk, 38453, Republic of Korea.,Research Institute of Anti-Aging, Daegu University, Gyeongbuk, 38453, Republic of Korea
| | - Kyeong-Min Kim
- Department of Biotechnology, School of Engineering, Daegu University, Gyeongbuk, 38453, Republic of Korea.,Research Institute of Anti-Aging, Daegu University, Gyeongbuk, 38453, Republic of Korea
| | - Eun-Jung Kim
- Research Institute of Anti-Aging, Daegu University, Gyeongbuk, 38453, Republic of Korea. .,Department of Immunology, Kyungpook National University School of Medicine, Daegu, 41944, Republic of Korea.
| | - Won-Gu Jang
- Department of Biotechnology, School of Engineering, Daegu University, Gyeongbuk, 38453, Republic of Korea. .,Research Institute of Anti-Aging, Daegu University, Gyeongbuk, 38453, Republic of Korea.
| |
Collapse
|
116
|
Kim RY, Seong Y, Cho TH, Lee B, Kim IS, Hwang SJ. Local administration of nuclear factor of activated T cells (NFAT) c1 inhibitor to suppress early resorption and inflammation induced by bone morphogenetic protein-2. J Biomed Mater Res A 2018; 106:1299-1310. [PMID: 29316218 DOI: 10.1002/jbm.a.36332] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/15/2017] [Accepted: 01/05/2018] [Indexed: 11/05/2022]
Abstract
Nuclear factor of activated T cells (NFAT)-c1 is known as a key regulator in osteoclast differentiation and immune response. This study is a follow-up to our previous study showing the antiresorptive activity of VIVIT, a peptide type NFATc1 inhibitor, using absorbable collagen sponge (ACS). This study aimed to investigate the effective concentration range of local VIVIT that suppresses early excessive osteoclast activation and inflammation induced by high-dose recombinant human bone morphogenetic protein (rhBMP)-2 and concomitantly enhances bone healing in a rat critical-sized calvaria defect model. High-dose rhBMP-2 (40 μg/defect) alone significantly increased in vivo osteoclast activation and expression of the inflammatory cytokines interleukin-1β and transforming necrosis factor-α on the scaffold at 7 days after surgery. However, rhBMP-2 had no direct effect on osteoclast activation in vitro. Osteoclast activation by rhBMP-2 was significantly suppressed by combined treatment with VIVIT at concentrations of 75 and 150 μM, but not at 15 μM, whereas suppression of inflammation occurred at all doses of VIVIT. Microcomputed tomography at 4 and 8 weeks after implantation revealed that the combination of rhBMP-2 and VIVIT at 75 μM VIVIT led to a greater bone fraction at the initial defect area, compared with rhBMP-2 alone. These findings revealed that local administration of VIVIT at certain concentrations has multiple positive effects that weaken early excessive osteoimmunological responses and enhance bone healing after rhBMP-2 administration. VIVIT has the potential to expand the therapeutic area of high-dose rhBMP-2 therapy to inflammatory bone loss. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 1299-1310, 2018.
Collapse
Affiliation(s)
- Ri Youn Kim
- Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 110-749, Republic of Korea
| | - Yeju Seong
- Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 110-749, Republic of Korea
| | - Tae Hyung Cho
- Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 110-749, Republic of Korea
| | - Beomseok Lee
- Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 110-749, Republic of Korea
| | - In Sook Kim
- Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 110-749, Republic of Korea
| | - Soon Jung Hwang
- Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 110-749, Republic of Korea.,Department of Oral and Maxillofacial Surgery, School of Dentistry, Seoul National University, Seoul, 110-749, Republic of Korea
| |
Collapse
|
117
|
Liu Z, Dong L, Wang L, Wang X, Cheng K, Luo Z, Weng W. Mediation of cellular osteogenic differentiation through daily stimulation time based on polypyrrole planar electrodes. Sci Rep 2017; 7:17926. [PMID: 29263335 PMCID: PMC5738366 DOI: 10.1038/s41598-017-17120-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 11/22/2017] [Indexed: 12/12/2022] Open
Abstract
In electrical stimulation (ES), daily stimulation time means the interacting duration with cells per day, and is a vital factor for mediating cellular function. In the present study, the effect of stimulation time on osteogenic differentiation of MC3T3-E1 cells was investigated under ES on polypyrrole (Ppy) planar interdigitated electrodes (IDE). The results demonstrated that only a suitable daily stimulation time supported to obviously upregulate the expression of ALP protein and osteogenesis-related genes (ALP, Col-I, Runx2 and OCN), while a short or long daily stimulation time showed no significant outcomes. These might be attributed to the mechanism that an ES induced transient change in intracellular calcium ion concentration, which was responsible for activating calcium ion signaling pathway to enhance cellular osteogenic differentiation. A shorter daily time could lead to insufficient duration for the transient change in intracellular calcium ion concentration, and a longer daily time could give rise to cellular fatigue with no transient change. This work therefore provides new insights into the fundamental understanding of cell responses to ES and will have an impact on further designing materials to mediate cell behaviors.
Collapse
Affiliation(s)
- Zongguang Liu
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou, 310027, China
| | - Lingqing Dong
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou, 310027, China
| | - Liming Wang
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou, 310027, China
| | - Xiaozhao Wang
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou, 310027, China
| | - Kui Cheng
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou, 310027, China
| | - Zhongkuan Luo
- Zhejiang-California International NanoSystems Institute, Hangzhou, 310058, China
| | - Wenjian Weng
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou, 310027, China.
| |
Collapse
|
118
|
Wang J, Fu B, Lu F, Hu X, Tang J, Huang L. Inhibitory activity of linarin on osteoclastogenesis through receptor activator of nuclear factor κB ligand-induced NF-κB pathway. Biochem Biophys Res Commun 2017; 495:2133-2138. [PMID: 29269297 DOI: 10.1016/j.bbrc.2017.12.091] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 12/16/2017] [Indexed: 12/17/2022]
Abstract
Linarin, a natural flavonoid glycoside widely found in plants, has been reported to possess anti-inflammation, neuroprotection and osteogenic properties. However, its impact on osteoclast remains unclear. In the present study, the effects of linarin on osteoclastogenesis and its underlying molecular mechanisms of action were investigated. Using the culture systems of osteoclasts derived from bone marrow macrophages (BMMs), we found that linarin dose-dependently inhibited osteoclasts formation and bone resorptive activity. The Cell Counting Kit-8 test displayed that the viability of cells was not influenced by linarin at doses up to 10 μg/mL. In addition, linarin downregulated osteoclast-related genes expression, including nuclear factor of activated T cells cytoplasmic 1 (NFATc1), tartrate resistant acid phosphatase (TRAP), osteoclast-associated receptor (OSCAR) and c-Fos, as shown by quantitative real time polymerase chain reaction (RT-qPCR). Western blot analysis further showed that linarin inhibited receptor activator of nuclear factor κB ligand (RANKL)-induced nuclear factor kappa B (NF-κB) p65 and NFATc1 activity. The present findings show that linarin exerted a potent inhibitory effect on osteoclastogenesis through RANKL-induced NF-κB signaling pathway. In conclusion, the results suggest that linarin has anti-osteoclastic effects and may serve as potential modulatory agents for the prevention and treatment of bone loss-associated diseases.
Collapse
Affiliation(s)
- Junsheng Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; Department of Orthopaedic Surgery, Huai'an Second People's Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huai'an 223000, China
| | - Bin Fu
- Department of Orthopaedic Surgery, Changzhou Wujin People's Hospital, Changzhou 213100, China
| | - Fuchun Lu
- Department of Orthopaedic Surgery, Huai'an Second People's Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huai'an 223000, China
| | - Xiaowu Hu
- Department of Orthopaedic Surgery, Huai'an Second People's Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huai'an 223000, China
| | - Jinshan Tang
- Department of Orthopaedic Surgery, Huai'an Second People's Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huai'an 223000, China
| | - Lixin Huang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| |
Collapse
|
119
|
Song J, Jing Z, Hu W, Yu J, Cui X. α-Linolenic Acid Inhibits Receptor Activator of NF-κB Ligand Induced (RANKL-Induced) Osteoclastogenesis and Prevents Inflammatory Bone Loss via Downregulation of Nuclear Factor-KappaB-Inducible Nitric Oxide Synthases (NF-κB-iNOS) Signaling Pathways. Med Sci Monit 2017; 23:5056-5069. [PMID: 29061958 PMCID: PMC5665607 DOI: 10.12659/msm.904795] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Inflammation is a major cellular strain causing increased risk of osteo-degenerative diseases. Omega-3 fatty acids have been great source in suppressing inflammation. We investigated the effect of α-linolenic acid (ALA) on RANKL-stimulated osteoclast differentiation, LPS-induced and ovariectomized bone loss in mice models. Material/Methods The bone marrow macrophages (BMMs) were isolated from femurs of ICR mice, stimulated with RANKL, and treated with ALA (100, 200, 300 μM). Major analytical methods include histological analysis, osteoclasts viability assay, serum cytokines and chemokines ELISA, and gene expression by qPCR. Results ALA intervention inhibited RANKL-induced osteoclasts proliferation and differentiation. ALA inhibited bone resorption activity as measured by materialization of F-actin ring structures as well. ALA suppressed the RANKL-induced osteoclast markers c-Fos, c-Jun and NFATc1 together with transcription factor proteins TRAP, OSCAR, cathepsin K and β3-integrin. ALA also suppressed the RANKL-stimulated phosphorylation of JNK, ERK, and AKT as well as NF-κB and BCL-2 proteins. ALA intervention (100 and 300 mg/kg) to LPS-challenged mice showed annulled morphometric changes induced by LPS by suppressing the levels of proinflammatory cytokines and chemokines. ALA (100 and 300 mg/kg) intervention to estrogen-deficiency induced bone loss mice (ovariectomized) showed reductions in TRAP+ osteoclasts count, CTX-I expression, levels of IL-1β, IL-2, IL-6, IL10, TNF-α and MCP-1 and iNOS and COX-2. Conclusions ALA suppresses RANKL-induced osteoclast differentiation and prevents inflammatory bone loss via downregulation of NF-κB-iNOS-COX-2 signaling. ALA is suggested to be a preventive herbal medicine against inflammatory bone disorders.
Collapse
Affiliation(s)
- Jiefu Song
- Department of Orthopedics, Shan Xi Provincial People's Hospital, Taiyuan, Shanxi, China (mainland)
| | - Zhizhen Jing
- Department of Orthopedics, Shan Xi Provincial People's Hospital, Taiyuan, Shanxi, China (mainland)
| | - Wei Hu
- Department of Orthopedics, Shan Xi Provincial People's Hospital, Taiyuan, Shanxi, China (mainland)
| | - Jianping Yu
- Department of Orthopedics, Shan Xi Provincial People's Hospital, Taiyuan, Shanxi, China (mainland)
| | - Xiaoping Cui
- Department of Orthopedics, Shan Xi Provincial People's Hospital, Taiyuan, Shanxi, China (mainland)
| |
Collapse
|
120
|
Zhang H, Liang S, Du Y, Li R, He C, Wang W, Liu S, Ye Z, Liang X, Shi W, Zhang B. Inducible ATF3-NFAT axis aggravates podocyte injury. J Mol Med (Berl) 2017; 96:53-64. [PMID: 29038896 PMCID: PMC5760612 DOI: 10.1007/s00109-017-1601-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/12/2017] [Accepted: 10/05/2017] [Indexed: 01/25/2023]
Abstract
Abstract Podocyte injury and loss contribute to proteinuria, glomerulosclerosis, and eventually kidney failure. Activating transcription factor 3 (ATF3) is a stress inducible transcription factor that is transiently expressed following stimulation. However, we show for the first time an induction of ATF3 in podocytes from patients with chronic kidney disease, including minimal change disease, focal segmental glomerulosclerosis, and diabetic nephropathy. The role of ATF3 induction in podocytes under chronic conditions is currently unknown. Compared with the control (C57 or BKS), ATF3 expression was elevated in animal model of proteinuria (LPS-treated C57 mice) and the model of diabetic nephropathy (db/db mice). Similarly, ATF3 was increased in high glucose (HG)-treated, lipopolysaccharide (LPS)-treated, or Ionomycin-treated podocytes in vitro. Overexpression of ATF3 increased podocyte apoptosis and decreased expression of podocin, the cell marker of podocyte; in contrast, ATF3–small interfering RNA knockdown reduced podocyte apoptosis and increased podocin expression. The translocation of ATF3 to the nucleus was increased upon stimulation. ATF3 directly modulates the regulation of NFATc1 gene promoter activity and alters the expression of Wnt6 and Fzd9, direct target genes of NFATc1 signaling. The ATF3 binding site of NFATc1 gene promoter is located at the region 671–775 base pairs upstream of the transcription start site. These results indicate a novel inducible axis of ATF3–NFAT in podocyte injury and loss. Key messages • The stress factor ATF3 is induced in podocytes from proteinuric patients, including diabetes. • ATF3 increased podocyte apoptosis and injury. • ATF3 directly modulates the regulation of NFATc1 gene promoter activity.
Collapse
Affiliation(s)
- Hong Zhang
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106# Zhongshan No. 2 Road, Guangzhou, 510080, China.,Southern Medical University, Guangzhou, 510515, China
| | - Shun Liang
- Department of Nephrology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Yue Du
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106# Zhongshan No. 2 Road, Guangzhou, 510080, China.,School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Ruizhao Li
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106# Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Chaosheng He
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106# Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Wenjian Wang
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106# Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Shuangxin Liu
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106# Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Zhiming Ye
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106# Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Xinling Liang
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106# Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Wei Shi
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106# Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Bin Zhang
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106# Zhongshan No. 2 Road, Guangzhou, 510080, China. .,Southern Medical University, Guangzhou, 510515, China. .,School of Medicine, South China University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
121
|
NUMBL Interacts with TAK1, TRAF6 and NEMO to Negatively Regulate NF-κB Signaling During Osteoclastogenesis. Sci Rep 2017; 7:12600. [PMID: 28974699 PMCID: PMC5626749 DOI: 10.1038/s41598-017-12707-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/13/2017] [Indexed: 11/09/2022] Open
Abstract
NF-κB signaling is essential for osteoclast differentiation and skeletal homeostasis. We have reported recently that NUMB-like (NUMBL) protein modulates osteoclastogenesis by down regulating NF-κB activation. Herein, we decipher the mechanism underlying this phenomenon. We found that whereas NUMBL mRNA expression decreases upon stimulation of wild type (WT) bone marrow macrophages (BMMs) with RANKL, TAK1 deficiency in these cells leads to increased NUMBL and decreased TRAF6 and NEMO expression. These changes were restored upon WT-TAK1 expression, but not with catalytically inactive TAK1-K63W, suggesting that TAK1 enzymatic activity is required for these events. Forced expression of NUMBL inhibits osteoclast differentiation and function as evident by reduction in all hallmarks of osteoclastogenesis. Conversely, NUMBL-null BMMs, show increased osteoclast differentiation and mRNA expression of osteoclast marker genes. Post-translationally, K48-linked poly-ubiquitination of NUMBL is diminished in TAK1-null BMMs compared to elevated K48-poly-ubiquitination in WT cells, indicating increased stability of NUMBL in TAK1-null conditions. Further, our studies show that NUMBL directly interacts with TRAF6 and NEMO, and induces their K48-poly-ubiquitination mediated proteasomal degradation. Collectively, our data suggest that NUMBL and TAK1 are reciprocally regulated and that NUMBL acts as an endogenous regulator of NF-κB signaling and osteoclastogenesis by targeting the TAK1-TRAF6-NEMO axis.
Collapse
|
122
|
Okamoto K, Nakashima T, Shinohara M, Negishi-Koga T, Komatsu N, Terashima A, Sawa S, Nitta T, Takayanagi H. Osteoimmunology: The Conceptual Framework Unifying the Immune and Skeletal Systems. Physiol Rev 2017; 97:1295-1349. [DOI: 10.1152/physrev.00036.2016] [Citation(s) in RCA: 369] [Impact Index Per Article: 46.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 03/29/2017] [Accepted: 04/04/2017] [Indexed: 12/13/2022] Open
Abstract
The immune and skeletal systems share a variety of molecules, including cytokines, chemokines, hormones, receptors, and transcription factors. Bone cells interact with immune cells under physiological and pathological conditions. Osteoimmunology was created as a new interdisciplinary field in large part to highlight the shared molecules and reciprocal interactions between the two systems in both heath and disease. Receptor activator of NF-κB ligand (RANKL) plays an essential role not only in the development of immune organs and bones, but also in autoimmune diseases affecting bone, thus effectively comprising the molecule that links the two systems. Here we review the function, gene regulation, and signal transduction of osteoimmune molecules, including RANKL, in the context of osteoclastogenesis as well as multiple other regulatory functions. Osteoimmunology has become indispensable for understanding the pathogenesis of a number of diseases such as rheumatoid arthritis (RA). We review the various osteoimmune pathologies, including the bone destruction in RA, in which pathogenic helper T cell subsets [such as IL-17-expressing helper T (Th17) cells] induce bone erosion through aberrant RANKL expression. We also focus on cellular interactions and the identification of the communication factors in the bone marrow, discussing the contribution of bone cells to the maintenance and regulation of hematopoietic stem and progenitors cells. Thus the time has come for a basic reappraisal of the framework for understanding both the immune and bone systems. The concept of a unified osteoimmune system will be absolutely indispensable for basic and translational approaches to diseases related to bone and/or the immune system.
Collapse
Affiliation(s)
- Kazuo Okamoto
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Tomoki Nakashima
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Masahiro Shinohara
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Takako Negishi-Koga
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Noriko Komatsu
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Asuka Terashima
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Shinichiro Sawa
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Takeshi Nitta
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Hiroshi Takayanagi
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| |
Collapse
|
123
|
Abstract
PURPOSE OF REVIEW Bone remodeling is a diverse field of study with many direct clinical applications; past studies have implicated epigenetic alterations as key factors of both normal bone tissue development and function and diseases of pathologic bone remodeling. The purpose of this article is to review the most important recent advances that link epigenetic changes to the bone remodeling field. RECENT FINDINGS Epigenetics describes three major phenomena: DNA modification via methylation, histone side chain modifications, and short non-coding RNA sequences which work in concert to regulate gene transcription in a heritable fashion. Recent findings include the role of DNA methylation changes of Wnt, RANK/RANKL, and other key signaling pathways, epigenetic regulation of osteoblast and osteoclast differentiation, and others. Although much work has been done, much is still unknown. Future epigenome-wide studies should focus on extending the tissue coverage, integrating multiple epigenetic analyses with transcriptome data, and working to uncover epigenetic changes linked with early events in aberrant bone remodeling.
Collapse
Affiliation(s)
- Ali Husain
- Division of Rheumatology, Immunology, and Allergy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Matlock A Jeffries
- Division of Rheumatology, Immunology, and Allergy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma Medical Research Foundation, Arthritis and Clinical Immunology Program, 825 NE 13th St., Laboratory MC400, Oklahoma City, OK, USA.
| |
Collapse
|
124
|
Niu C, Xiao F, Yuan K, Hu X, Lin W, Ma R, Zhang X, Huang Z. Nardosinone Suppresses RANKL-Induced Osteoclastogenesis and Attenuates Lipopolysaccharide-Induced Alveolar Bone Resorption. Front Pharmacol 2017; 8:626. [PMID: 28955231 PMCID: PMC5601052 DOI: 10.3389/fphar.2017.00626] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 08/25/2017] [Indexed: 12/30/2022] Open
Abstract
Periodontitis is a chronic inflammatory disease that damages the integrity of the tooth-supporting tissues, known as the periodontium, and comprising the gingiva, periodontal ligament and alveolar bone. In this study, the effects of nardosinone (Nd) on bone were tested in a model of lipopolysaccharide (LPS)-induced alveolar bone loss, and the associated mechanisms were elucidated. Nd effectively suppressed LPS-induced alveolar bone loss and reduced osteoclast (OC) numbers in vivo. Nd suppressed receptor activator of nuclear factor-κB ligand (RANKL)-mediated OC differentiation, bone resorption, and F-actin ring formation in a dose-dependent manner. Further investigation revealed that Nd suppressed osteoclastogenesis by suppressing the ERK and JNK signaling pathways, scavenging reactive oxygen species, and suppressing the activation of PLCγ2 that consequently affects the expression and/or activity of the OC-specific transcription factors, c-Fos and nuclear factor of activated T-cells cytoplasmic 1 (NFATc1). In addition, Nd significantly reduced the expression of OC-specific markers in mouse bone marrow-derived pre-OCs, including c-Fos, cathepsin K (Ctsk), VATPase d2, and Nfatc1. Collectively, these findings suggest that Nd has beneficial effects on bone, and the suppression of OC number implies that the effect is exerted directly on osteoclastogenesis.
Collapse
Affiliation(s)
- Chenguang Niu
- Shanghai Key Laboratory of Stomatology, Department of Endodontics, Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Fei Xiao
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Keyong Yuan
- Shanghai Key Laboratory of Stomatology, Department of Endodontics, Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - XuChen Hu
- Shanghai Key Laboratory of Stomatology, Department of Endodontics, Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Wenzhen Lin
- Shanghai Key Laboratory of Stomatology, Department of Endodontics, Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Rui Ma
- Shanghai Key Laboratory of Stomatology, Department of Endodontics, Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Xiaoling Zhang
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Zhengwei Huang
- Shanghai Key Laboratory of Stomatology, Department of Endodontics, Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| |
Collapse
|
125
|
Osteoporosis following heart transplantation and immunosuppressive therapy. Transplant Rev (Orlando) 2017; 31:232-239. [PMID: 28865930 DOI: 10.1016/j.trre.2017.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 07/21/2017] [Accepted: 08/03/2017] [Indexed: 12/20/2022]
Abstract
Heart transplantation (HT) remains the ultimate final therapy for patients with end-stage heart failure, who despite optimal medical and surgical treatments exhibit severe symptoms. To prevent rejection of the transplanted organ, HT patients require life-long immunosuppressive therapy. The goal of the immunosuppression is to minimise the risk of immune-mediated graft rejection, while avoiding clinical side-effects. Current immunosuppressive agents have yielded good survival outcome, however, complications of the immunosuppressive therapy, such as impaired bone strength and increased fracture risk, are common among HT patients rendering increased morbidity and mortality rates. The main aim of the present review was to summarise current knowledge on bone strength impairment after HT and concomitant immunosuppressive therapy.
Collapse
|
126
|
Calcineurin inhibitors regulate fibroblast growth factor 23 (FGF23) synthesis. Naunyn Schmiedebergs Arch Pharmacol 2017; 390:1117-1123. [DOI: 10.1007/s00210-017-1411-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 07/21/2017] [Indexed: 01/06/2023]
|
127
|
Hu K, Sun H, Gui B, Sui C. TRPV4 functions in flow shear stress induced early osteogenic differentiation of human bone marrow mesenchymal stem cells. Biomed Pharmacother 2017; 91:841-848. [DOI: 10.1016/j.biopha.2017.04.094] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 04/12/2017] [Accepted: 04/20/2017] [Indexed: 12/25/2022] Open
|
128
|
Fierro FA, Nolta JA, Adamopoulos IE. Concise Review: Stem Cells in Osteoimmunology. Stem Cells 2017; 35:1461-1467. [PMID: 28390147 DOI: 10.1002/stem.2625] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 02/06/2017] [Accepted: 03/16/2017] [Indexed: 12/11/2022]
Abstract
Bone remodeling is a lifelong process in which mature bone tissue is removed from the skeleton by bone resorption and is replenished by new during ossification or bone formation. The remodeling cycle requires both the differentiation and activation of two cell types with opposing functions; the osteoclast, which orchestrates bone resorption, and the osteoblast, which orchestrates bone formation. The differentiation of these cells from their respective precursors is a process which has been overshadowed by enigma, particularly because the precise osteoclast precursor has not been identified and because the identification of skeletal stem cells, which give rise to osteoblasts, is very recent. Latest advances in the area of stem cell biology have enabled us to gain a better understanding of how these differentiation processes occur in physiological and pathological conditions. In this review we postulate that modulation of stem cells during inflammatory conditions is a necessary prerequisite of bone remodeling and therefore an essential new component to the field of osteoimmunology. In this context, we highlight the role of transcription factor nuclear factor of activated T cells cytoplasmic 1 (NFATc1), because it directly links inflammation with differentiation of osteoclasts and osteoblasts. Stem Cells 2017;35:1461-1467.
Collapse
Affiliation(s)
- Fernando A Fierro
- Stem Cell Program, University of California at Davis, Sacramento, California, USA.,Department of Cell Biology and Human Anatomy, University of California at Davis, Sacramento, California, USA
| | - Jan A Nolta
- Stem Cell Program, University of California at Davis, Sacramento, California, USA.,Department of Cell Biology and Human Anatomy, University of California at Davis, Sacramento, California, USA.,Department of Internal Medicine, University of California at Davis, Sacramento, California, USA
| | - Iannis E Adamopoulos
- Institute for Pediatric Regenerative Medicine, University of California at Davis, Sacramento, California, USA.,Department of Rheumatology, University of California at Davis, Sacramento, California, USA
| |
Collapse
|
129
|
Park HJ, Baek K, Baek JH, Kim HR. TNFα Increases RANKL Expression via PGE₂-Induced Activation of NFATc1. Int J Mol Sci 2017; 18:ijms18030495. [PMID: 28245593 PMCID: PMC5372511 DOI: 10.3390/ijms18030495] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 02/12/2017] [Accepted: 02/20/2017] [Indexed: 01/07/2023] Open
Abstract
Tumor necrosis factor α (TNFα) is known to upregulate the expression of receptor activator of NF-κB ligand (RANKL). We investigated the role of the calcineurin/nuclear factor of activated T-cells (NFAT) signaling pathway in TNFα-induced RANKL expression in C2C12 and primary cultured mouse calvarial cells. TNFα-induced RANKL expression was blocked by the calcineurin/NFAT pathway inhibitors. TNFα increased NFAT transcriptional activity and subsequent RANKL promoter binding. Mutations in the NFAT-binding element (MT(N)) suppressed TNFα-induced RANKL promoter activity. TNFα increased prostaglandin E2 (PGE2) production, which in turn enhanced NFAT transcriptional activity and binding to the RANKL promoter. MT(N) suppressed PGE2-induced RANKL promoter activity. TNFα and PGE2 increased the expression of RANKL, NFAT cytoplasmic-1 (NFATc1), cAMP response element-binding protein (CREB), and cyclooxygenase 2 (COX2); which increment was suppressed by indomethacin, a COX inhibitor. Mutations in the CRE-like element blocked PGE2-induced RANKL promoter activity. PGE2 induced the binding of CREB to the RANKL promoter, whereas TNFα increased the binding of both CREB and NFATc1 to this promoter through a process blocked by indomethacin. The PGE2 receptor antagonists AH6809 and AH23848 blocked TNFα-induced expression of RANKL, NFATc1, and CREB; transcriptional activity of NFAT; and binding of NFATc1 or CREB to the RANKL promoter. These results suggest that TNFα-induced RANKL expression depends on PGE2 production and subsequent transcriptional activation/enhanced binding of NFATc1 and CREB to the RANKL promoter.
Collapse
Affiliation(s)
- Hyun-Jung Park
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Korea.
| | - Kyunghwa Baek
- Department of Pharmacology, College of Dentistry and Research Institute of Oral Science, Gangneung-Wonju National University, Gangwon-do 25457, Korea.
| | - Jeong-Hwa Baek
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Korea.
| | | |
Collapse
|
130
|
He LH, Liu M, He Y, Xiao E, Zhao L, Zhang T, Yang HQ, Zhang Y. TRPV1 deletion impaired fracture healing and inhibited osteoclast and osteoblast differentiation. Sci Rep 2017; 7:42385. [PMID: 28225019 PMCID: PMC5320507 DOI: 10.1038/srep42385] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 01/09/2017] [Indexed: 12/24/2022] Open
Abstract
Fracture healing, in which osteoclasts and osteoblasts play important roles, has drawn much clinical attention. Osteoclast deficiency or decreased osteoblast activity will impair fracture healing. TRPV1 is a member of the Ca2+ permeable cation channel subfamily, and pharmacological inhibition of TRPV1 prevents ovariectomy-induced bone loss, which makes TRPV1 a potential target for osteoporosis. However, whether long term TRPV1 inhibition or TRPV1 deletion will affect the fracture healing process is unclear. In this study, we found that the wild-type mice showed a well-remodeled fracture callus, whereas TRPV1 knockout mice still had an obvious fracture gap with unresorbed soft-callus 4 weeks post-fracture. The number of osteoclasts was reduced in the TRPV1 knockout fracture callus, and osteoclast formation and resorption activity were also impaired in vitro. TRPV1 deletion decreased the calcium oscillation frequency and peak cytoplasmic concentration in osteoclast precursors, subsequently reducing the expression and nuclear translocation of NFATc1 and downregulating DC-stamp, cathepsin K, and ATP6V. In addition, TRPV1 deletion caused reduced mRNA and protein expression of Runx2 and ALP in bone marrow stromal cells (BMSCs) and reduced calcium deposition in vitro. Our results suggest that TRPV1 deletion impairs fracture healing, and inhibited osteoclastogenesis and osteogenesis.
Collapse
Affiliation(s)
- Lin-Hai He
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology; National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of digital Stomatology, Beijing, China
| | - Meng Liu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology; National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of digital Stomatology, Beijing, China
| | - Yang He
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology; National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of digital Stomatology, Beijing, China
| | - E. Xiao
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology; National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of digital Stomatology, Beijing, China
| | - Lu Zhao
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology; National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of digital Stomatology, Beijing, China
| | - Ting Zhang
- Center for Craniofacial Stem Cell Research and Regeneration, Department of Orthodontics, Peking University School and Hospital of Stomatology; National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of digital Stomatology, Beijing, China
| | - Hua-Qian Yang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing, China
| | - Yi Zhang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology; National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of digital Stomatology, Beijing, China
| |
Collapse
|
131
|
Zhang J, Xu H, Han Z, Chen P, Yu Q, Lei Y, Li Z, Zhao M, Tian J. Pulsed electromagnetic field inhibits RANKL-dependent osteoclastic differentiation in RAW264.7 cells through the Ca 2+ -calcineurin-NFATc1 signaling pathway. Biochem Biophys Res Commun 2017; 482:289-295. [DOI: 10.1016/j.bbrc.2016.11.056] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 11/11/2016] [Indexed: 10/20/2022]
|
132
|
RCANs regulate the convergent roles of NFATc1 in bone homeostasis. Sci Rep 2016; 6:38526. [PMID: 27917924 PMCID: PMC5137032 DOI: 10.1038/srep38526] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 11/11/2016] [Indexed: 01/04/2023] Open
Abstract
Activation of calcineurin-dependent nuclear factor of activated T cells c1 (NFATc1) is convergent for normal bone homeostasis. NFATc1 regulates both osteoclastogenesis and osteoblastogenesis. Here we investigated the roles of regulator of calcineurin (RCAN) genes in bone homeostasis. RCANs function as potent physiological inhibitors of calcineurin. Overexpression of RCANs in osteoclast precursor cells attenuated osteoclast differentiation, while their overexpression in osteoblasts enhanced osteoblast differentiation and function. Intriguingly, opposing effects of RCANs in both cell types were shown by blocking activation of the calcineurin-NFATc1 pathway. Moreover, the disruption of RCAN1 or RCAN2 in mice resulted in reduced bone mass, which is associated with strongly increased osteoclast function and mildly reduced osteoblast function. Taken together, RCANs play critical roles in bone homeostasis by regulating both osteoclastogenesis and osteoblastogenesis, and they serve as inhibitors for calcineurin-NFATc1 signaling both in vivo and in vitro.
Collapse
|
133
|
Dou XW, Park W, Lee S, Zhang QZ, Carrasco LR, Le AD. Loss of Notch3 Signaling Enhances Osteogenesis of Mesenchymal Stem Cells from Mandibular Torus. J Dent Res 2016; 96:347-354. [PMID: 27879421 DOI: 10.1177/0022034516680349] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Mandibular torus (MT) is a common intraoral osseous outgrowth located on the lingual surface of the mandible. Histologic features include hyperplastic bone consisting of mature cortical and trabecular bone. Some theories on the etiology of MT have been postulated, such as genetic factors, masticatory hyperfunction, trauma, and continued growth, but the underlying mechanism remains largely unknown. In this study, we investigated the potential role of mesenchymal stem cells (MSCs) derived from human MT in the pathogenesis of bone outgrowth. We demonstrated that MT harbored a distinct subpopulation of MSCs, with enhanced osteogenic and decreased adipogenic differentiation capacities, as compared with their counterparts from normal jaw bone. The increased osteogenic differentiation of mandibular torus MSCs was associated with the suppression of Notch3 signaling and its downstream target genes, Jag1 and Hey1, and a reciprocal increase in the transcriptional activation of ATF4 and NFATc1 genes. Targeted knockdown of Notch3 expression by transient siRNA transfection promoted the expression of osteogenic transcription factors in normal jaw bone MSCs. Our data suggest that the loss of Notch3 signaling may contribute partly to bone outgrowth in MT, as mediated by enhanced MSC-driven osteogenic differentiation in the jaw bone.
Collapse
Affiliation(s)
- X W Dou
- 1 Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
| | - W Park
- 1 Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA.,2 Department of Advanced General Dentistry, College of Dentistry, Yonsei University, Seoul, South Korea
| | - S Lee
- 3 Department of Endodontics, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
| | - Q Z Zhang
- 1 Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
| | - L R Carrasco
- 1 Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA.,4 Department of Oral and Maxillofacial Surgery, Penn Medicine Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - A D Le
- 1 Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA.,4 Department of Oral and Maxillofacial Surgery, Penn Medicine Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
134
|
Protein/amino-acid modulation of bone cell function. BONEKEY REPORTS 2016; 5:827. [PMID: 28149508 PMCID: PMC5238414 DOI: 10.1038/bonekey.2016.58] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 06/24/2016] [Indexed: 01/07/2023]
Abstract
Nutrients (protein, carbohydrates and fats) have traditionally been thought of as fuels simply providing the energy for cellular metabolic activity. According to the classic view, if nutrients are available, then anabolic pathways are activated, and if nutrients are not available, catabolic pathways are activated. However, it is becoming increasingly clear that nutrient effects on bone cells (stem cells, osteoblasts and osteoclasts) are complex, some nutrients promote bone formation, whereas others interfere with bone formation or actually promote bone break down. At an organ level, nutrient intake can suppress bone breakdown and modulate the activity of the calcium/vitamin D/parathyroid hormone axis. At a cellular level, nutrient intake can impact cellular energetics either through a direct mechanism (binding or uptake of the nutrient into the cell) or indirect (by elevating nutrient-related hormones such as insulin, insulin-like growth factor 1 or incretin hormones). It is also becoming clear that within a nutrient class (for example, protein), individual components (that is, amino acids) can have markedly different effects on cell function and impact bone formation. The focus of this review will be on one nutrient class in particular, dietary protein. As the prevalence of inadequate dietary protein intake increases with age, these findings may have translational implications as to the optimal dietary protein content in the setting of age-associated bone loss.
Collapse
|
135
|
ATF3 controls proliferation of osteoclast precursor and bone remodeling. Sci Rep 2016; 6:30918. [PMID: 27480204 PMCID: PMC4969588 DOI: 10.1038/srep30918] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 07/11/2016] [Indexed: 12/21/2022] Open
Abstract
Bone homeostasis is maintained by the sophisticated coupled actions of bone-resorbing osteoclasts and bone-forming osteoblasts. Here we identify activating transcription factor 3 (ATF3) as a pivotal transcription factor for the regulation of bone resorption and bone remodeling under a pathological condition through modulating the proliferation of osteoclast precursors. The osteoclast precursor-specific deletion of ATF3 in mice led to the prevention of receptor activator of nuclear factor-κB (RANK) ligand (RANKL)-induced bone resorption and bone loss, although neither bone volume nor osteoclastic parameter were markedly altered in these knockout mice under the physiological condition. RANKL-dependent osteoclastogenesis was impaired in vitro in ATF3-deleted bone marrow macrophages (BMM). Mechanistically, the deficiency of ATF3 impaired the RANKL-induced transient increase in cell proliferation of osteoclast precursors in bone marrow in vivo as well as of BMM in vitro. Moreover, ATF3 regulated cyclin D1 mRNA expression though modulating activator protein-1-dependent transcription in the osteoclast precursor, and the introduction of cyclin D1 significantly rescued the impairment of osteoclastogenesis in ATF3-deleted BMM. Therefore, these findings suggest that ATF3 could have a pivotal role in osteoclastogenesis and bone homeostasis though modulating cell proliferation under pathological conditions, thereby providing a target for bone diseases.
Collapse
|
136
|
Pedigo CE, Ducasa GM, Leclercq F, Sloan A, Mitrofanova A, Hashmi T, Molina-David J, Ge M, Lassenius MI, Forsblom C, Lehto M, Groop PH, Kretzler M, Eddy S, Martini S, Reich H, Wahl P, Ghiggeri G, Faul C, Burke GW, Kretz O, Huber TB, Mendez AJ, Merscher S, Fornoni A. Local TNF causes NFATc1-dependent cholesterol-mediated podocyte injury. J Clin Invest 2016; 126:3336-50. [PMID: 27482889 DOI: 10.1172/jci85939] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 05/26/2016] [Indexed: 12/14/2022] Open
Abstract
High levels of circulating TNF and its receptors, TNFR1 and TNFR2, predict the progression of diabetic kidney disease (DKD), but their contribution to organ damage in DKD remains largely unknown. Here, we investigated the function of local and systemic TNF in podocyte injury. We cultured human podocytes with sera collected from DKD patients, who displayed elevated TNF levels, and focal segmental glomerulosclerosis (FSGS) patients, whose TNF levels resembled those of healthy patients. Exogenous TNF administration or local TNF expression was equally sufficient to cause free cholesterol-dependent apoptosis in podocytes by acting through a dual mechanism that required a reduction in ATP-binding cassette transporter A1-mediated (ABCA1-mediated) cholesterol efflux and reduced cholesterol esterification by sterol-O-acyltransferase 1 (SOAT1). TNF-induced albuminuria was aggravated in mice with podocyte-specific ABCA1 deficiency and was partially prevented by cholesterol depletion with cyclodextrin. TNF-stimulated free cholesterol-dependent apoptosis in podocytes was mediated by nuclear factor of activated T cells 1 (NFATc1). ABCA1 overexpression or cholesterol depletion was sufficient to reduce albuminuria in mice with podocyte-specific NFATc1 activation. Our data implicate an NFATc1/ABCA1-dependent mechanism in which local TNF is sufficient to cause free cholesterol-dependent podocyte injury irrespective of TNF, TNFR1, or TNFR2 serum levels.
Collapse
|
137
|
Toll-Like Receptor 2 Stimulation of Osteoblasts Mediates Staphylococcus Aureus Induced Bone Resorption and Osteoclastogenesis through Enhanced RANKL. PLoS One 2016; 11:e0156708. [PMID: 27311019 PMCID: PMC4911171 DOI: 10.1371/journal.pone.0156708] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 04/28/2016] [Indexed: 11/22/2022] Open
Abstract
Severe Staphylococcus aureus (S. aureus) infections pose an immense threat to population health and constitute a great burden for the health care worldwide. Inter alia, S. aureus septic arthritis is a disease with high mortality and morbidity caused by destruction of the infected joints and systemic bone loss, osteoporosis. Toll-Like receptors (TLRs) are innate immune cell receptors recognizing a variety of microbial molecules and structures. S. aureus recognition via TLR2 initiates a signaling cascade resulting in production of various cytokines, but the mechanisms by which S. aureus causes rapid and excessive bone loss are still unclear. We, therefore, investigated how S. aureus regulates periosteal/endosteal osteoclast formation and bone resorption. S. aureus stimulation of neonatal mouse parietal bone induced ex vivo bone resorption and osteoclastic gene expression. This effect was associated with increased mRNA and protein expression of receptor activator of NF-kB ligand (RANKL) without significant change in osteoprotegerin (OPG) expression. Bone resorption induced by S. aureus was abolished by OPG. S. aureus increased the expression of osteoclastogenic cytokines and prostaglandins in the parietal bones but the stimulatory effect of S. aureus on bone resorption and Tnfsf11 mRNA expression was independent of these cytokines and prostaglandins. Stimulation of isolated periosteal osteoblasts with S. aureus also resulted in increased expression of Tnfsf11 mRNA, an effect lost in osteoblasts from Tlr2 knockout mice. S. aureus stimulated osteoclastogenesis in isolated periosteal cells without affecting RANKL-stimulated resorption. In contrast, S. aureus inhibited RANKL-induced osteoclast formation in bone marrow macrophages. These data show that S. aureus enhances bone resorption and periosteal osteoclast formation by increasing osteoblast RANKL production through TLR2. Our study indicates the importance of using different in vitro approaches for studies of how S. aureus regulates osteoclastogenesis to obtain better understanding of the complex mechanisms of S. aureus induced bone destruction in vivo.
Collapse
|
138
|
Chiu YH, Ritchlin CT. DC-STAMP: A Key Regulator in Osteoclast Differentiation. J Cell Physiol 2016; 231:2402-7. [PMID: 27018136 DOI: 10.1002/jcp.25389] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 03/24/2016] [Indexed: 01/09/2023]
Abstract
Osteoimmunology research is a new emerging research field that investigates the links between the bone and immune responses. Results from osteoimmunology studies suggest that bone is not only an essential component of the musculoskeletal system, but is also actively involved in immune regulation. Many important factors involved in immune regulation also participate in bone homeostasis. Bone homeostasis is achieved by a coordinated action between bone-synthesizing osteoblasts and bone-degrading osteoclasts. An imbalanced action between osteoblasts and osteoclasts often results in pathological bone diseases: osteoporosis is caused by an excessive osteoclast activity, whereas osteopetrosis results from an increased osteoblast activity. This review focuses on dendritic cell-specific transmembrane protein (DC-STAMP), an important protein currently considered as a master regulator of osteoclastogenesis. Of clinical relevance, the frequency of circulating DC-STAMP+ cells is elevated during the pathogenesis of psoriatic diseases. Intriguingly, recent results suggest that DC-STAMP also plays an imperative role in bone homeostasis by regulating the differentiation of both osteoclasts and osteoblasts. This article summarizes our current knowledge on DC-STAMP by focusing on its interacting proteins, its regulation on osteoclastogenesis-related genes, its possible involvement in immunoreceptor tyrosine-based inhibitory motif (ITIM)-mediated signaling cascade, and its potential of developing therapeutics for clinical applications. J. Cell. Physiol. 231: 2402-2407, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ya-Hui Chiu
- Division of Allergy, Immunology and Rheumatology, School of Medicine, University of Rochester, Rochester, New York
| | - Christopher T Ritchlin
- Division of Allergy, Immunology and Rheumatology, School of Medicine, University of Rochester, Rochester, New York
| |
Collapse
|
139
|
Szabo-Rogers H, Yakob W, Liu KJ. Frontal Bone Insufficiency in Gsk3β Mutant Mice. PLoS One 2016; 11:e0149604. [PMID: 26886780 PMCID: PMC4757545 DOI: 10.1371/journal.pone.0149604] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 02/03/2016] [Indexed: 12/17/2022] Open
Abstract
The development of the mammalian skull is a complex process that requires multiple tissue interactions and a balance of growth and differentiation. Disrupting this balance can lead to changes in the shape and size of skull bones, which can have serious clinical implications. For example, insufficient ossification of the bony elements leads to enlarged anterior fontanelles and reduced mechanical protection of the brain. In this report, we find that loss of Gsk3β leads to a fully penetrant reduction of frontal bone size and subsequent enlarged frontal fontanelle. In the absence of Gsk3β the frontal bone primordium undergoes increased cell death and reduced proliferation with a concomitant increase in Fgfr2-IIIc and Twist1 expression. This leads to a smaller condensation and premature differentiation. This phenotype appears to be Wnt-independent and is not rescued by decreasing the genetic dose of β-catenin/Ctnnb1. Taken together, our work defines a novel role for Gsk3β in skull development.
Collapse
Affiliation(s)
- Heather Szabo-Rogers
- Craniofacial Development and Stem Cell Biology, Floor 27, Tower Wing, Guy’s Campus, King’s College London, London, United Kingdom SE1 9RT
| | - Wardati Yakob
- Craniofacial Development and Stem Cell Biology, Floor 27, Tower Wing, Guy’s Campus, King’s College London, London, United Kingdom SE1 9RT
| | - Karen J. Liu
- Craniofacial Development and Stem Cell Biology, Floor 27, Tower Wing, Guy’s Campus, King’s College London, London, United Kingdom SE1 9RT
- * E-mail:
| |
Collapse
|
140
|
Kim JH, Kim EY, Lee B, Min JH, Song DU, Lim JM, Eom JW, Yeom M, Jung HS, Sohn Y. The effects of Lycii Radicis Cortex on RANKL-induced osteoclast differentiation and activation in RAW 264.7 cells. Int J Mol Med 2016; 37:649-58. [PMID: 26848104 PMCID: PMC4771095 DOI: 10.3892/ijmm.2016.2477] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 01/25/2016] [Indexed: 01/16/2023] Open
Abstract
Post-menopausal osteoporosis is a serious age-related disease. After the menopause, estrogen deficiency is common, and excessive osteoclast activity causes osteoporosis. Osteoclasts are multinucleated cells generated from the differentiation of monocyte/macrophage precursor cells such as RAW 264.7 cells. The water extract of Lycii Radicis Cortex (LRC) is made from the dried root bark of Lycium chinense Mill. and is termed 'Jigolpi' in Korea. Its effects on osteoclastogenesis and post‑menopausal osteoporosis had not previously been tested. In the present study, the effect of LRC on receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL)-induced osteoclast differentiation was demonstrated using a tartrate-resistant acid phosphatase (TRAP) assay and pit formation assay. Moreover, in order to analyze molecular mechanisms, we studied osteoclastogenesis-related markers such as nuclear factor of activated T-cells cytoplasmic 1 (NFATc1), c-Fos, receptor activator of NF-κB (RANK), TRAP, cathepsin K (CTK), matrix metallopeptidase-9 (MMP-9), calcitonin receptor (CTR) and carbonic anhydrase Ⅱ (CAII) using RT-qPCR and western blot analysis. Additionally, we also determined the effect of LRC on an ovariectomized (OVX) rat model. We noted that LRC inhibited RANKL-induced osteoclast differentiation via suppressing osteoclastogenesis-related markers. It also inhibited osteoporosis in the OVX rat model by decreasing loss of bone density and trabecular area. These results suggest that LRC exerts a positive effect on menopausal osteoporosis.
Collapse
Affiliation(s)
- Jae-Hyun Kim
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Eun-Young Kim
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Bina Lee
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Ju-Hee Min
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Dea-Uk Song
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Jeong-Min Lim
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Ji Whan Eom
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Mijung Yeom
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Hyuk-Sang Jung
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Youngjoo Sohn
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| |
Collapse
|
141
|
Ding W, Tong Y, Zhang X, Pan M, Chen S. Study of Arsenic Sulfide in Solid Tumor Cells Reveals Regulation of Nuclear Factors of Activated T-cells by PML and p53. Sci Rep 2016; 6:19793. [PMID: 26795951 PMCID: PMC4726130 DOI: 10.1038/srep19793] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 12/18/2015] [Indexed: 11/09/2022] Open
Abstract
Arsenic sulfide (AS) has excellent cytotoxic activity in acute promyelocytic leukemia (APL) but its activity in solid tumors remains to be explored. Here we show that AS and cyclosporine A (CsA) exerted synergistic inhibitory effect on cell growth and c-Myc expression in HCT116 cells. AS inhibited the expression of PML, c-Myc, NFATc1, NFATc3, and NFATc4, while stimulating the expression of p53 and NFATc2. Knockdown of PML reduced NFATc1, NFATc2, NFATc3 and NFATc4 expression while overexpression of p53 stimulated NFATc2-luciferase activity that was further augmented by AS by binding to a set of p53 responsive elements (PREs) on the NFATc2 promoter. Additionally, overexpression of p53 suppressed NFATc3 and NFATc4. Reciprocally, NFATc3 knockdown enhanced p53 while reducing MDM2 expression indicating that NFATc3 is a negative regulator of p53 while a positive regulator of MDM2, consistent with its tumor-promoting property as knockdown of NFATc3 retarded cell growth in vitro and tumor growth in xenograft. In patients with colon cancer, tumor expression of NFATc2 correlated with superior survival, while nuclear NFATc1 with inferior survival. These results indicate that AS differentially regulates NFAT pathway through PML and p53 and reveal an intricate reciprocal regulatory relationship between NFAT proteins and p53 pathway.
Collapse
Affiliation(s)
- Wenping Ding
- Department of Oncology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingying Tong
- Department of Oncology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiuli Zhang
- Department of Oncology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minggui Pan
- Department of Oncology and Hematology, Kaiser Permanente Medical Center, Santa Clara, CA.,Kaiser Permanente Division of Research, Oakland, CA, USA
| | - Siyu Chen
- Department of Oncology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
142
|
Luthringer BJ, Willumeit-Römer R. Effects of magnesium degradation products on mesenchymal stem cell fate and osteoblastogenesis. Gene 2016; 575:9-20. [DOI: 10.1016/j.gene.2015.08.028] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 08/10/2015] [Accepted: 08/13/2015] [Indexed: 01/02/2023]
|
143
|
Algate K, Haynes DR, Bartold PM, Crotti TN, Cantley MD. The effects of tumour necrosis factor-α on bone cells involved in periodontal alveolar bone loss; osteoclasts, osteoblasts and osteocytes. J Periodontal Res 2015; 51:549-66. [DOI: 10.1111/jre.12339] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2015] [Indexed: 12/22/2022]
Affiliation(s)
- K. Algate
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
| | - D. R. Haynes
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
| | - P. M. Bartold
- School of Dentistry; University of Adelaide; Adelaide SA Australia
| | - T. N. Crotti
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
| | - M. D. Cantley
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
- Myeloma Research Laboratory; University of Adelaide; Adelaide SA Australia
| |
Collapse
|
144
|
Regulation of transcriptional network system during bone and cartilage development. J Oral Biosci 2015. [DOI: 10.1016/j.job.2015.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
145
|
Mandal CC, Das F, Ganapathy S, Harris SE, Choudhury GG, Ghosh-Choudhury N. Bone Morphogenetic Protein-2 (BMP-2) Activates NFATc1 Transcription Factor via an Autoregulatory Loop Involving Smad/Akt/Ca2+ Signaling. J Biol Chem 2015; 291:1148-61. [PMID: 26472929 DOI: 10.1074/jbc.m115.668939] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Indexed: 11/06/2022] Open
Abstract
Bone remodeling is controlled by dual actions of osteoclasts (OCs) and osteoblasts (OBs). The calcium-sensitive nuclear factor of activated T cells (NFAT) c1 transcription factor, as an OC signature gene, regulates differentiation of OCs downstream of bone morphogenetic protein-2 (BMP-2)-stimulated osteoblast-coded factors. To analyze a functional link between BMP-2 and NFATc1, we analyzed bones from OB-specific BMP-2 knock-out mice for NFATc1 expression by immunohistochemical staining and found significant reduction in NFATc1 expression. This indicated a requirement of BMP-2 for NFATc1 expression in OBs. We showed that BMP-2, via the receptor-specific Smad pathway, regulates expression of NFATc1 in OBs. Phosphatidylinositol 3-kinase/Akt signaling acting downstream of BMP-2 also drives NFATc1 expression and transcriptional activation. Under the basal condition, NFATc1 is phosphorylated. Activation of NFAT requires dephosphorylation by the calcium-dependent serine/threonine phosphatase calcineurin. We examined the role of calcium in BMP-2-stimulated regulation of NFATc1 in osteoblasts. 1,2Bis(2aminophenoxy)ethaneN,N,N',N'-tetraacetic acid acetoxymethyl ester, an inhibitor of intracellular calcium abundance, blocked BMP-2-induced transcription of NFATc1. Interestingly, BMP-2 induced calcium release from intracellular stores and increased calcineurin phosphatase activity, resulting in NFATc1 nuclear translocation. Cyclosporin A, which inhibits calcineurin upstream of NFATc1, blocked BMP-2-induced NFATc1 mRNA and protein expression. Expression of NFATc1 directly increased its transcription and VIVIT peptide, an inhibitor of NFATc1, suppressed BMP-2-stimulated NFATc1 transcription, confirming its autoregulation. Together, these data show a role of NFATc1 downstream of BMP-2 in mouse bone development and provide novel evidence for the presence of a cross-talk among Smad, phosphatidylinositol 3-kinase/Akt, and Ca(2+) signaling for BMP-2-induced NFATc1 expression through an autoregulatory loop.
Collapse
Affiliation(s)
| | | | | | - Stephen E Harris
- Periodontics, University of Texas Health Science Center, San Antonio, Texas 78229
| | - Goutam Ghosh Choudhury
- Medicine, and From Veterans Affairs Research and Geriatric Research Education and Clinical Center, South Texas Veterans Health Care System and
| | | |
Collapse
|
146
|
Kim JY, Park SH, Baek JM, Erkhembaatar M, Kim MS, Yoon KH, Oh J, Lee MS. Harpagoside Inhibits RANKL-Induced Osteoclastogenesis via Syk-Btk-PLCγ2-Ca(2+) Signaling Pathway and Prevents Inflammation-Mediated Bone Loss. JOURNAL OF NATURAL PRODUCTS 2015; 78:2167-2174. [PMID: 26308264 DOI: 10.1021/acs.jnatprod.5b00233] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Harpagoside (HAR) is a natural compound isolated from Harpagophytum procumbens (devil's claw) that is reported to have anti-inflammatory effects; however, these effects have not been investigated in the context of bone development. The current study describes for the first time that HAR inhibits receptor activator of nuclear factor κB ligand (RANKL)-induced osteoclastogenesis in vitro and suppresses inflammation-induced bone loss in a mouse model. HAR also inhibited the formation of osteoclasts from mouse bone marrow macrophages (BMMs) in a dose-dependent manner as well as the activity of mature osteoclasts, including filamentous actin (F-actin) ring formation and bone matrix breakdown. This involved a HAR-induced decrease in extracellular signal-regulated kinase (ERK) and c-jun N-terminal kinase (JNK) phosphorylation, leading to the inhibition of Syk-Btk-PLCγ2-Ca(2+) in RANKL-dependent early signaling, as well as the activation of c-Fos and nuclear factor of activated T cells cytoplasmic 1 (NFATc1), which resulted in the down-regulation of various target genes. Consistent with these in vitro results, HAR blocked lipopolysaccharide (LPS)-induced bone loss in an inflammatory osteoporosis model. However, HAR did not prevent ovariectomy-mediated bone erosion in a postmenopausal osteoporosis model. These results suggest that HAR is a valuable agent against inflammation-related bone disorders but not osteoporosis induced by hormonal abnormalities.
Collapse
Affiliation(s)
- Ju-Young Kim
- Imaging Science-Based Lung and Bone Diseases Research Center, ‡Department of Anatomy, School of Medicine, §Department of Oral Physiology, School of Dentistry, ⊥Department of Radiology, School of Medicine, ∥Institute for Skeletal Disease, and ▽Division of Rheumatology, Department of Internal Medicine, Wonkwang University , Iksan, Jeonbuk 570-749, Korea
| | - Sun-Hyang Park
- Imaging Science-Based Lung and Bone Diseases Research Center, ‡Department of Anatomy, School of Medicine, §Department of Oral Physiology, School of Dentistry, ⊥Department of Radiology, School of Medicine, ∥Institute for Skeletal Disease, and ▽Division of Rheumatology, Department of Internal Medicine, Wonkwang University , Iksan, Jeonbuk 570-749, Korea
| | - Jong Min Baek
- Imaging Science-Based Lung and Bone Diseases Research Center, ‡Department of Anatomy, School of Medicine, §Department of Oral Physiology, School of Dentistry, ⊥Department of Radiology, School of Medicine, ∥Institute for Skeletal Disease, and ▽Division of Rheumatology, Department of Internal Medicine, Wonkwang University , Iksan, Jeonbuk 570-749, Korea
| | - Munkhsoyol Erkhembaatar
- Imaging Science-Based Lung and Bone Diseases Research Center, ‡Department of Anatomy, School of Medicine, §Department of Oral Physiology, School of Dentistry, ⊥Department of Radiology, School of Medicine, ∥Institute for Skeletal Disease, and ▽Division of Rheumatology, Department of Internal Medicine, Wonkwang University , Iksan, Jeonbuk 570-749, Korea
| | - Min Seuk Kim
- Imaging Science-Based Lung and Bone Diseases Research Center, ‡Department of Anatomy, School of Medicine, §Department of Oral Physiology, School of Dentistry, ⊥Department of Radiology, School of Medicine, ∥Institute for Skeletal Disease, and ▽Division of Rheumatology, Department of Internal Medicine, Wonkwang University , Iksan, Jeonbuk 570-749, Korea
| | - Kwon-Ha Yoon
- Imaging Science-Based Lung and Bone Diseases Research Center, ‡Department of Anatomy, School of Medicine, §Department of Oral Physiology, School of Dentistry, ⊥Department of Radiology, School of Medicine, ∥Institute for Skeletal Disease, and ▽Division of Rheumatology, Department of Internal Medicine, Wonkwang University , Iksan, Jeonbuk 570-749, Korea
| | - Jaemin Oh
- Imaging Science-Based Lung and Bone Diseases Research Center, ‡Department of Anatomy, School of Medicine, §Department of Oral Physiology, School of Dentistry, ⊥Department of Radiology, School of Medicine, ∥Institute for Skeletal Disease, and ▽Division of Rheumatology, Department of Internal Medicine, Wonkwang University , Iksan, Jeonbuk 570-749, Korea
| | - Myeung Su Lee
- Imaging Science-Based Lung and Bone Diseases Research Center, ‡Department of Anatomy, School of Medicine, §Department of Oral Physiology, School of Dentistry, ⊥Department of Radiology, School of Medicine, ∥Institute for Skeletal Disease, and ▽Division of Rheumatology, Department of Internal Medicine, Wonkwang University , Iksan, Jeonbuk 570-749, Korea
| |
Collapse
|
147
|
Xiao E, Yang HQ, Gan YH, Duan DH, He LH, Guo Y, Wang SQ, Zhang Y. Brief reports: TRPM7 Senses mechanical stimulation inducing osteogenesis in human bone marrow mesenchymal stem cells. Stem Cells 2015; 33:615-21. [PMID: 25263397 DOI: 10.1002/stem.1858] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 07/24/2014] [Accepted: 08/24/2014] [Indexed: 01/13/2023]
Abstract
Mesenchymal stem cells (MSCs) are multipotential stem cells residing in the bone marrow. Several studies have shown that mechanical stimulation modulates MSC differentiation through mobilization of second messengers, but the mechanism of mechanotransduction remains poorly understood. In this study, using fluorescence and laser confocal microcopy as well as patch-clamp techniques, we identified the transient receptor potential melastatin type 7 (TRPM7) channel as the key channel involved in mechanotransduction in bone marrow MSCs. TRPM7 knockdown completely abolished the pressure-induced cytosolic Ca(2+) increase and pressure-induced osteogenesis. TRPM7 directly sensed membrane tension, independent of the cytoplasm and the integrity of cytoskeleton. Ca(2+) influx through TRPM7 further triggered Ca(2+) release from the inositol trisphosphate receptor type 2 on the endoplasmic reticulum and promoted NFATc1 nuclear localization and osteogenesis. These results identified a central role of TRPM7 in MSC mechanical stimulation-induced osteogenesis.
Collapse
Affiliation(s)
- E Xiao
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
148
|
Expression of thymosin beta-4 in human periodontal ligament cells and mouse periodontal tissue and its role in osteoblastic/cementoblastic differentiation. Differentiation 2015; 90:16-26. [DOI: 10.1016/j.diff.2015.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 06/30/2015] [Accepted: 08/30/2015] [Indexed: 11/21/2022]
|
149
|
Zanotti S, Canalis E. Activation of Nfatc2 in osteoblasts causes osteopenia. J Cell Physiol 2015; 230:1689-95. [PMID: 25573264 DOI: 10.1002/jcp.24928] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 01/07/2015] [Indexed: 01/08/2023]
Abstract
Nuclear factor of activated T-cells (Nfat) c1 to c4 are transcription factors that play an undisputable role in osteoclastogenesis. However, Nfat function in osteoblastic cells is controversial. Constitutive activation of Nfatc1 and c2 in osteoblasts suppresses cell function, although the study of Nfat in vivo has yielded conflicting results. To establish the consequences of Nfatc2 activation in osteoblasts, we generated transgenic mice where a 3.6 kb fragment of the collagen type I α1 promoter directs expression of a constitutively active Nfatc2 mutant (Col3.6-Nfatc2). The skeletal phenotype of Col3.6-Nfatc2 mice of both sexes and of sex-matched littermate controls was investigated by microcomputed tomography and histomorphometry. Col3.6- Nfatc2 mice were born at the expected Mendelian ratio and appeared normal. Nfatc2 expression was confirmed in parietal bones from 1 and 3 month old transgenic mice. One month old Col3.6-Nfatc2 female mice exhibited cancellous bone compartment osteopenia secondary to a 30% reduction in bone formation. In contrast, cancellous femoral bone volume and bone formation were not altered in male transgenics, whereas osteoblast number was higher, suggesting incomplete osteoblast maturation. Indices of bone resorption were not affected in either sex. At 3 months of age, the skeletal phenotype evolved; and Col3.6-Nfatc2 male mice exhibited vertebral osteopenia, whereas femoral cancellous bone was not affected in either sex. Nfatc2 activation in osteoblasts had no impact on cortical bone structure. Nfatc2 activation inhibited alkaline phosphatase activity and mineralized nodule formation in bone marrow stromal cell cultures. In conclusion, Nfatc2 activation in osteoblasts inhibits bone formation and causes cancellous bone osteopenia.
Collapse
Affiliation(s)
- Stefano Zanotti
- Departments of Orthopaedic Surgery and Medicine, UConn Health Center, Farmington, Connecticut
| | | |
Collapse
|
150
|
Guo Q, Tripathi P, Manson SR, Austin PF, Chen F. Transcriptional dysregulation in the ureteric bud causes multicystic dysplastic kidney by branching morphogenesis defect. J Urol 2015; 193:1784-90. [PMID: 25301096 PMCID: PMC4504205 DOI: 10.1016/j.juro.2014.08.122] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2014] [Indexed: 11/25/2022]
Abstract
PURPOSE The calcineurin-NFAT signaling pathway regulates the transcription of genes important for development. It is impacted by various genetic and environmental factors. We investigated the potential role of NFAT induced transcriptional dysregulation in the pathogenesis of congenital abnormalities of the kidneys and urinary tract. MATERIALS AND METHODS A murine model of conditional NFATc1 activation in the ureteric bud was generated and examined for histopathological changes. Metanephroi were also cultured in vitro to analyze branching morphogenesis in real time. RESULTS NFATc1 activation led to defects resembling multicystic dysplastic kidney. These mutants showed severe disorganization of branching morphogenesis characterized by decreased ureteric bud branching and the disconnection of ureteric bud derivatives from the main collecting system. The orphan ureteric bud derivatives may have continued to induce nephrogenesis and likely contributed to the subsequent formation of blunt ended filtration units and cysts. The ureter also showed irregularities consistent with impaired epithelial-mesenchymal interaction. CONCLUSIONS This study reveals the profound effects of NFAT signaling dysregulation on the ureteric bud and provides insight into the pathogenesis of multicystic dysplastic kidney. Our results suggest that the obstruction hypothesis and the bud theory may not be mutually exclusive to explain the pathogenesis of multicystic dysplastic kidney. Ureteric bud dysfunction such as that induced by NFAT activation can disrupt ureteric bud-metanephric mesenchyma interaction, causing primary defects in branching morphogenesis, subsequent dysplasia and cyst formation. Obstruction of the main collecting system can further enhance these defects, producing the pathological changes associated with multicystic dysplastic kidney.
Collapse
Affiliation(s)
- Qiusha Guo
- Washington University School of Medicine, St. Louis, Missouri
| | - Piyush Tripathi
- Renal Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Scott R Manson
- Division of Urology, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Paul F Austin
- Division of Urology, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Feng Chen
- Renal Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|