1
|
Hirose H, Fujimasa S, Kanemaru S, Yoshimoto S, Matsumoto N, Anan H, Matsuzaki E. Sphingosine-1-phosphate receptor 1-mediated odontogenic differentiation of mouse apical papilla-derived stem cells. J Dent Sci 2024; 19:2323-2331. [PMID: 39347102 PMCID: PMC11437261 DOI: 10.1016/j.jds.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/05/2024] [Indexed: 10/01/2024] Open
Abstract
Background/purpose Sphingosine-1-phosphate (S1P) exhibits receptor-mediated physiological effects by facilitating the differentiation of mesenchymal stem cells toward the osteoblast lineage. This study aimed to determine the effect of S1P on odontogenic differentiation of mouse immortalized stem cells of dental apical papilla (iSCAP) and assess the distribution of the S1P receptor 1 (S1PR1) in the apical papilla and the root canal wall of immature rat molars. Materials and methods Immunostaining for S1PR1 was conducted at the apex of the rat mandibular first molar and within the root canal wall. The iSCAP was treated with S1P and bone morphogenetic protein (BMP)-9 (for comparison), and the expression levels of the odontogenic differentiation marker were evaluated via real-time reverse-transcriptase quantitative polymerase chain reaction and enzyme-linked immunosorbent assay. Mineralization and lipid droplet formation were evaluated via Alizarin red and Oil red O staining. Results S1PR1-positive cells were expressed in areas of both apical papilla and dentin-pulp interface of root canal wall. During the odontogenic differentiation of iSCAP, S1P and BMP-9 increased the expression of the differentiation marker mRNA and secreted proteins including dentin sialophosphoprotein, dentin matrix phosphoprotein 1, and matrix extracellular phosphoglycoprotein. The S1PR1 signaling pathway is involved in the action of S1P, but not that of BMP-9. S1PR1 signaling also facilitated mineralization in iSCAP and suppressed the differentiation of these cells into adipocytes. Conclusion S1P induced odontogenic differentiation of iSCAP through S1PR1. Furthermore, S1PR1-positive cells were expressed in the apical papilla of immature rat molars and in the dentin-pulp interface where odontoblast-like cells exist.
Collapse
Affiliation(s)
- Haruna Hirose
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan
| | - Seishiro Fujimasa
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan
| | - Shingo Kanemaru
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan
| | - Shohei Yoshimoto
- Section of Pathology, Department of Morphological Biology, Division of Biomedical Sciences, Fukuoka Dental College, Fukuoka, Japan
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Noriyoshi Matsumoto
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan
| | | | - Etsuko Matsuzaki
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
| |
Collapse
|
2
|
Tangporncharoen R, Phanus-Umporn C, Prachayasittikul S, Nantasenamat C, Prachayasittikul V, Supokawej A. Computer-guided design of novel nitrogen-based heterocyclic sphingosine-1-phosphate (S1P) activators as osteoanabolic agents. EXCLI JOURNAL 2024; 23:818-832. [PMID: 39574964 PMCID: PMC11579520 DOI: 10.17179/excli2024-7214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 04/17/2024] [Indexed: 11/24/2024]
Abstract
Osteoanabolic agents, or drugs that promote bone formation, have gained considerable attention for osteoporosis management due to their curative and preventive potentials. Sphingosine-1-phosphate receptor 2 (S1PR2) is an attractive drug target, in which its activation leads to osteogenesis-promoting effect. Nitrogen-containing heterocyclic scaffolds (i.e., quinoxaline and indole) are promising pharmacophores possessing diverse bioactivities and were reported as S1PR2 activators. Quantitative structure-activity relationship (QSAR) modeling is a computational approach well-known as a fundamental tool for facilitating successful drug development. This study demonstrates the discovery of new S1PR2 activators using computational-driven rational design. Herein, an original dataset of nitrogen-containing S1PR2 activators was collected from ChEMBL database. The retrieved dataset was separated into two datasets according to their core scaffolds (i.e., quinoxaline and indole). QSAR modeling was performed using multiple linear regression (MLR) algorithm to successfully obtain two models with good predictive performance. The constructed models also revealed key properties playing essential roles for potent S1PR2 activation, such as Van der Waals volume (R2v+ and E3v), mass (MATS5m and Km), electronegativity (H3e), and number of 5-membered rings (nR05). Subsequently, the constructed models were further employed to guide rational design and predict S1PR2 activating effects of an additional set of 752 structurally modified compounds. Most of the modified compounds were predicted to have higher potency than their parents, and a set of promising potent newly designed compounds was highlighted. Additionally, drug-likeness prediction was performed to reveal that most of the newly designed compounds are druggable compounds with possibility for further development.
Collapse
Affiliation(s)
- Rattanawan Tangporncharoen
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| | - Chuleeporn Phanus-Umporn
- Department of Community Medical Technology, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| | - Supaluk Prachayasittikul
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| | | | - Veda Prachayasittikul
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| | - Aungkura Supokawej
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
3
|
Frost K, Lewis JW, Jones SW, Edwards JR, Naylor AJ, McGettrick HM. The Species Effect: Differential Sphingosine-1-Phosphate Responses in the Bone in Human Versus Mouse. Int J Mol Sci 2024; 25:5118. [PMID: 38791156 PMCID: PMC11121697 DOI: 10.3390/ijms25105118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/29/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
The deterioration of osteoblast-led bone formation and the upregulation of osteoclast-regulated bone resorption are the primary causes of bone diseases, including osteoporosis. Numerous circulating factors play a role in bone homeostasis by regulating osteoblast and osteoclast activity, including the sphingolipid-sphingosine-1-phosphate (S1P). However, to date no comprehensive studies have investigated the impact of S1P activity on human and murine osteoblasts and osteoclasts. We observed species-specific responses to S1P in both osteoblasts and osteoclasts, where S1P stimulated human osteoblast mineralisation and reduced human pre-osteoclast differentiation and mineral resorption, thereby favouring bone formation. The opposite was true for murine osteoblasts and osteoclasts, resulting in more mineral resorption and less mineral deposition. Species-specific differences in osteoblast responses to S1P were potentially explained by differential expression of S1P receptor 1. By contrast, human and murine osteoclasts expressed comparable levels of S1P receptors but showed differential expression patterns of the two sphingosine kinase enzymes responsible for S1P production. Ultimately, we reveal that murine models may not accurately represent how human bone cells will respond to S1P, and thus are not a suitable model for exploring S1P physiology or potential therapeutic agents.
Collapse
Affiliation(s)
- Kathryn Frost
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (K.F.); (J.W.L.); (S.W.J.); (A.J.N.)
| | - Jonathan W. Lewis
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (K.F.); (J.W.L.); (S.W.J.); (A.J.N.)
| | - Simon W. Jones
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (K.F.); (J.W.L.); (S.W.J.); (A.J.N.)
| | - James R. Edwards
- Botnar Research Centre, University of Oxford, Oxford OX3 7LD, UK;
| | - Amy J. Naylor
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (K.F.); (J.W.L.); (S.W.J.); (A.J.N.)
| | - Helen M. McGettrick
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (K.F.); (J.W.L.); (S.W.J.); (A.J.N.)
| |
Collapse
|
4
|
Al Saedi A, Yacoub AS, Awad K, Karasik D, Brotto M, Duque G. The Interplay of Lipid Signaling in Musculoskeletal Cross Talk: Implications for Health and Disease. Methods Mol Biol 2024; 2816:1-11. [PMID: 38977583 DOI: 10.1007/978-1-0716-3902-3_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The intricate interplay between the muscle and bone tissues is a fundamental aspect of musculoskeletal physiology. Over the past decades, emerging research has highlighted the pivotal role of lipid signaling in mediating communication between these tissues. This chapter delves into the multifaceted mechanisms through which lipids, particularly phospholipids, sphingolipids, and eicosanoids, participate in orchestrating cellular responses and metabolic pathways in both muscle and bone. Additionally, we examine the clinical implications of disrupted lipid signaling in musculoskeletal disorders, offering insights into potential therapeutic avenues. This chapter aims to shed light on the complex lipid-driven interactions between the muscle and bone tissues, paving the way for a deeper understanding of musculoskeletal health and disease.
Collapse
Affiliation(s)
- Ahmed Al Saedi
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| | - Ahmed S Yacoub
- Bone-Muscle Research Center, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX, USA
| | - Kamal Awad
- Bone-Muscle Research Center, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX, USA
| | - David Karasik
- The Musculoskeletal Genetics Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Marco Brotto
- Bone-Muscle Research Center, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX, USA
| | - Gustavo Duque
- Research Institute of McGill University Health Center, Department of Medicine, McGill University, Québec, Canada
| |
Collapse
|
5
|
Xu X, Han Y, Zhu T, Fan F, Wang X, Liu Y, Luo D. The role of SphK/S1P/S1PR signaling pathway in bone metabolism. Biomed Pharmacother 2023; 169:115838. [PMID: 37944444 DOI: 10.1016/j.biopha.2023.115838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023] Open
Abstract
There are a large number of people worldwide who suffer from osteoporosis, which imposes a huge economic burden, so it is necessary to explore the underlying mechanisms to achieve better supportive and curative care outcomes. Sphingosine kinase (SphK) is an enzyme that plays a crucial role in the synthesis of sphingosine-1-phosphate (S1P). S1P with paracrine and autocrine activities that act through its cell surface S1P receptors (S1PRs) and intracellular signals. In osteoporosis, S1P is indispensable for both normal and disease conditions. S1P has complicated roles in regulating osteoblast and osteoclast, respectively, and there have been exciting developments in understanding how SphK/S1P/S1PR signaling regulates these processes in response to osteoporosis therapy. Here, we review the proliferation, differentiation, apoptosis, and functions of S1P, specifically detailing the roles of S1P and S1PRs in osteoblasts and osteoclasts. Finally, we focus on the S1P-based therapeutic approaches in bone metabolism, which may provide valuable insights into potential therapeutic strategies for osteoporosis.
Collapse
Affiliation(s)
- Xuefeng Xu
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, China
| | - Yi Han
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, China
| | - Tianxin Zhu
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, China
| | - Faxin Fan
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, China
| | - Xin Wang
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, China
| | - Yuqing Liu
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, China
| | - Duosheng Luo
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, China.
| |
Collapse
|
6
|
Wagner JM, Wille A, Fueth M, Weske S, Lotzien S, Reinkemeier F, Wallner C, Sogorski A, Dittfeld S, Becerikli M, Schildhauer TA, Lehnhardt M, Levkau B, Behr B. Pharmacological elevation of sphingosine-1-phosphate by S1P lyase inhibition accelerates bone regeneration after post-traumatic osteomyelitis. J Cell Mol Med 2023; 27:3786-3795. [PMID: 37710406 PMCID: PMC10718149 DOI: 10.1111/jcmm.17952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/15/2023] [Accepted: 09/01/2023] [Indexed: 09/16/2023] Open
Abstract
Posttraumatic osteomyelitis and the ensuing bone defects are a debilitating complication after open fractures with little therapeutic options. We have recently identified potent osteoanabolic effects of sphingosine-1-phosphate (S1P) signalling and have now tested whether it may beneficially affect bone regeneration after infection. We employed pharmacological S1P lyase inhibition by 4-deoxypyrodoxin (DOP) to raise S1P levels in vivo in an unicortical long bone defect model of posttraumatic osteomyelitis in mice. In a translational approach, human bone specimens of clinical osteomyelitis patients were treated in organ culture in vitro with DOP. Bone regeneration was assessed by μCT, histomorphometry, immunohistology and gene expression analysis. The role of S1P receptors was addressed using S1PR3 deficient mice. Here, we present data that DOP treatment markedly enhanced osteogenesis in posttraumatic osteomyelitis. This was accompanied by greatly improved osteoblastogenesis and enhanced angiogenesis in the callus accompanied by osteoclast-mediated bone remodelling. We also identified the target of increased S1P to be the S1PR3 as S1PR3-/- mice showed no improvement of bone regeneration by DOP. In the human bone explants, bone mass significantly increased along with enhanced osteoblastogenesis and angiogenesis. Our data suggest that enhancement of S1P/S1PR3 signalling may be a promising therapeutic target for bone regeneration in posttraumatic osteomyelitis.
Collapse
Affiliation(s)
- Johannes M. Wagner
- Department of Plastic SurgeryBG University Hospital Bergmannsheil BochumBochumGermany
- Department of Trauma Surgery and General SurgeryBG University Hospital Bergmannsheil BochumBochumGermany
| | - Annalena Wille
- Institute of Molecular Medicine IIIUniversity Hospital Düsseldorf and Heinrich Heine Universität DüsseldorfDüsseldorfGermany
| | - Maria Fueth
- Department of Plastic SurgeryBG University Hospital Bergmannsheil BochumBochumGermany
| | - Sarah Weske
- Institute of Molecular Medicine IIIUniversity Hospital Düsseldorf and Heinrich Heine Universität DüsseldorfDüsseldorfGermany
| | - Sebastian Lotzien
- Department of Trauma Surgery and General SurgeryBG University Hospital Bergmannsheil BochumBochumGermany
| | - Felix Reinkemeier
- Department of Plastic SurgeryBG University Hospital Bergmannsheil BochumBochumGermany
| | - Christoph Wallner
- Department of Plastic SurgeryBG University Hospital Bergmannsheil BochumBochumGermany
| | - Alexander Sogorski
- Department of Plastic SurgeryBG University Hospital Bergmannsheil BochumBochumGermany
| | - Stephanie Dittfeld
- Department of Plastic SurgeryBG University Hospital Bergmannsheil BochumBochumGermany
| | - Mustafa Becerikli
- Department of Plastic SurgeryBG University Hospital Bergmannsheil BochumBochumGermany
| | - Thomas A. Schildhauer
- Department of Trauma Surgery and General SurgeryBG University Hospital Bergmannsheil BochumBochumGermany
| | - Marcus Lehnhardt
- Department of Plastic SurgeryBG University Hospital Bergmannsheil BochumBochumGermany
| | - Bodo Levkau
- Institute of Molecular Medicine IIIUniversity Hospital Düsseldorf and Heinrich Heine Universität DüsseldorfDüsseldorfGermany
| | - Björn Behr
- Department of Plastic SurgeryBG University Hospital Bergmannsheil BochumBochumGermany
| |
Collapse
|
7
|
Zhang F, Xu J, Hu Y, Fang J, Yang M, Huang K, Xu W, He X. Diallyl trisulfide ameliorates bone loss and alters specific gut microbiota and serum metabolites in natural aging mice. Food Funct 2023; 14:7642-7653. [PMID: 37540026 DOI: 10.1039/d3fo01840g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
Aging is a major cause of bone loss and osteoporosis. Diallyl trisulfide (DATS), one of the main organic sulfides in garlic oil, has been shown to alleviate arthritis in mice. However, further research is still needed to determine how DATS affects bone formation and bone loss in aging mice. Here, we established a mouse model of natural aging for dietary DATS intervention. DATS treatment improved the bone microstructure, including the disorganized arrangement of bone trabeculae and promoted collagen synthesis, as confirmed by micro-CT and histological analyses. The abundance of beneficial bacteria for bone formation, such as Clostridiaceae and Erysipelotrichaceae, and the microbial diversity and community richness were all altered by DATS, according to 16S rRNA sequencing data. 14 potential biomarkers and 9 important metabolic pathways were examined using serum metabolomics analysis. Additionally, there has been a significant reduction in sphingosine, which is directly associated with bone metabolism. The level of sphingosine and relative abundance of Clostridium were found to be negatively correlated by correlation analysis, indicating that bacteria may regulate bone reconstruction via influencing metabolites. Furthermore, Runx2 and β-catenin gene expression levels increased in bones, which may be related to the ameliorative mechanism of DATS. Our results suggested that DATS may prevent age-related bone loss by upregulating osteogenic gene expression through altering gut microbes and serum metabolism.
Collapse
Affiliation(s)
- Feng Zhang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; College of Food Science and Nutritional Engineering; China Agricultural University, Beijing 100083, China.
| | - Jia Xu
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; College of Food Science and Nutritional Engineering; China Agricultural University, Beijing 100083, China.
| | - Yanzhou Hu
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; College of Food Science and Nutritional Engineering; China Agricultural University, Beijing 100083, China.
| | - Jingjing Fang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; College of Food Science and Nutritional Engineering; China Agricultural University, Beijing 100083, China.
| | - Minglan Yang
- Department of Clinical Nutrition, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kunlun Huang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; College of Food Science and Nutritional Engineering; China Agricultural University, Beijing 100083, China.
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), the Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing, 100083, China
| | - Wentao Xu
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), the Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing, 100083, China
- Department of Nutrition and Health, China Agricultural University, Beijing 100191, China
| | - Xiaoyun He
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; College of Food Science and Nutritional Engineering; China Agricultural University, Beijing 100083, China.
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), the Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing, 100083, China
| |
Collapse
|
8
|
Frost K, Naylor AJ, McGettrick HM. The Ying and Yang of Sphingosine-1-Phosphate Signalling within the Bone. Int J Mol Sci 2023; 24:6935. [PMID: 37108099 PMCID: PMC10139073 DOI: 10.3390/ijms24086935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/28/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Bone remodelling is a highly active and dynamic process that involves the tight regulation of osteoblasts, osteoclasts, and their progenitors to allow for a balance of bone resorption and formation to be maintained. Ageing and inflammation are risk factors for the dysregulation of bone remodelling. Once the balance between bone formation and resorption is lost, bone mass becomes compromised, resulting in disorders such as osteoporosis and Paget's disease. Key molecules in the sphingosine-1-phosphate signalling pathway have been identified for their role in regulating bone remodelling, in addition to its more recognised role in inflammatory responses. This review discusses the accumulating evidence for the different, and, in certain circumstances, opposing, roles of S1P in bone homeostasis and disease, including osteoporosis, Paget's disease, and inflammatory bone loss. Specifically, we describe the current, often conflicting, evidence surrounding S1P function in osteoblasts, osteoclasts, and their precursors in health and disease, concluding that S1P may be an effective biomarker of bone disease and also an attractive therapeutic target for disease.
Collapse
Affiliation(s)
| | - Amy J. Naylor
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | | |
Collapse
|
9
|
Krishnan RH, Sadu L, Akshaya RL, Gomathi K, Saranya I, Das UR, Satishkumar S, Selvamurugan N. Circ_CUX1/miR-130b-5p/p300 axis for parathyroid hormone-stimulation of Runx2 activity in rat osteoblasts: A combined bioinformatic and experimental approach. Int J Biol Macromol 2023; 225:1152-1163. [PMID: 36427609 DOI: 10.1016/j.ijbiomac.2022.11.176] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/31/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022]
Abstract
Parathyroid hormone (PTH) regulates the expression of bone remodeling genes by enhancing the activity of Runx2 in osteoblasts. p300, a histone acetyltransferase, acetylated Runx2 to activate the expression of its target genes. PTH stimulated the expression of p300 in rat osteoblastic cells. Increasing studies suggested the potential of non-coding RNAs (ncRNAs), such as microRNAs (miRNAs) and circular RNAs (circRNAs), in regulating gene expression under both physiological and pathological conditions. In this study, we hypothesized that PTH regulates Runx2 activity via ncRNAs-mediated p300 expression in rat osteoblastic cells. Bioinformatics and experimental approaches identified PTH-upregulation of miR-130b-5p and circ_CUX1 that putatively target p300 and miR-130b-5p, respectively. An antisense-mediated knockdown of circ_CUX1 was performed to determine the sponging activity of circ_CUX1. Knockdown of circ_CUX1 promoted miR-130b-5p activity and reduced p300 expression, resulting in decreased Runx2 acetylation in rat osteoblastic cells. Further, bioinformatics analysis identified the possible signaling pathways that regulate Runx2 activity and osteoblast differentiation via circ_CUX1/miR-130b-5p/p300 axis. The predicted circ_CUX1/miR-130b-5p/p300 axis might pave the way for better diagnostic and therapeutic approaches for bone-related diseases.
Collapse
Affiliation(s)
- R Hari Krishnan
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Lakshana Sadu
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - R L Akshaya
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - K Gomathi
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - I Saranya
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Udipt Ranjan Das
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Sneha Satishkumar
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - N Selvamurugan
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| |
Collapse
|
10
|
Zheng Z, Wu L, Li Z, Tang R, Li H, Huang Y, Wang T, Xu S, Cheng H, Ye Z, Xiao D, Lin X, Wu G, Jaspers RT, Pathak JL. Mir155 regulates osteogenesis and bone mass phenotype via targeting S1pr1 gene. eLife 2023; 12:77742. [PMID: 36598122 PMCID: PMC9839347 DOI: 10.7554/elife.77742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 01/03/2023] [Indexed: 01/05/2023] Open
Abstract
MicroRNA-155 (miR155) is overexpressed in various inflammatory diseases and cancer, in which bone resorption and osteolysis are frequently observed. However, the role of miR155 on osteogenesis and bone mass phenotype is still unknown. Here, we report a low bone mass phenotype in the long bone of Mir155-Tg mice compared with wild-type mice. In contrast, Mir155-KO mice showed a high bone mass phenotype and protective effect against inflammation-induced bone loss. Mir155-KO mice showed robust bone regeneration in the ectopic and orthotopic model, but Mir155-Tg mice showed compromised bone regeneration compared with the wild-type mice. Similarly, the osteogenic differentiation potential of bone marrow stromal stem cells (BMSCs) from Mir155-KO mice was robust and Mir155-Tg was compromised compared with that of wild-type mice. Moreover, Mir155 knockdown in BMSCs from wild-type mice showed higher osteogenic differentiation potential, supporting the results from Mir155-KO mice. TargetScan analysis predicted sphingosine 1-phosphate receptor-1 (S1pr1) as a target gene of Mir155, which was further confirmed by luciferase assay and Mir155 knockdown. S1pr1 overexpression in BMSCs robustly promoted osteogenic differentiation without affecting cell viability and proliferation. Furthermore, osteoclastogenic differentiation of Mir155-Tg bone marrow-derived macrophages was inhibited compared with that of wild-type mice. Thus, Mir155 showed a catabolic effect on osteogenesis and bone mass phenotype via interaction with the S1pr1 gene, suggesting inhibition of Mir155 as a potential strategy for bone regeneration and bone defect healing.
Collapse
Affiliation(s)
- Zhichao Zheng
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative MedicineGuangzhouChina,Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamNetherlands
| | - Lihong Wu
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative MedicineGuangzhouChina
| | - Zhicong Li
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative MedicineGuangzhouChina
| | - Ruoshu Tang
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative MedicineGuangzhouChina
| | - Hongtao Li
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Yinyin Huang
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative MedicineGuangzhouChina
| | - Tianqi Wang
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative MedicineGuangzhouChina
| | - Shaofen Xu
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative MedicineGuangzhouChina
| | - Haoyu Cheng
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative MedicineGuangzhouChina
| | - Zhitong Ye
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative MedicineGuangzhouChina
| | - Dong Xiao
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumour Immunology Research, Cancer Research Institute, School of Basic Medical Science, Southern Medical UniversityGuangzhouChina,Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical UniversityGuangzhouChina
| | - Xiaolin Lin
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumour Immunology Research, Cancer Research Institute, School of Basic Medical Science, Southern Medical UniversityGuangzhouChina,Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical UniversityGuangzhouChina
| | - Gang Wu
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Amsterdam Movement Science, Vrije Universiteit AmsterdamAmsterdamNetherlands,Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit AmsterdamAmsterdamNetherlands
| | - Richard T Jaspers
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative MedicineGuangzhouChina,Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamNetherlands
| | - Janak L Pathak
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative MedicineGuangzhouChina
| |
Collapse
|
11
|
Yan H, Yu T, Li J, Zhang T, Li Q, Zhou Y, Liu D. Kartogenin Improves Osteogenesis of Bone Marrow Mesenchymal Stem Cells via Autophagy. Stem Cells Int 2022; 2022:1278921. [PMID: 36591373 PMCID: PMC9800103 DOI: 10.1155/2022/1278921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/27/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
Kartogenin (KGN), a novel small-molecule compound, has been considered a promising chondrogenic promoter in cartilage regeneration. However, whether KGN also participates in osteogenesis and bone regeneration remains unclear. This research was designed to explore the roles of KGN on osteogenic differentiation in bone marrow mesenchymal stem cells (BMMSCs) as well as determine the possible mechanism of osteogenesis. We revealed that KGN enhanced the osteogenic differentiation capacity of BMMSCs without affecting cell proliferation, during which autophagic activities and the expression of autophagy-related genes were promoted. Moreover, KGN upregulated the phosphorylation level of the Smad1/5/9 signaling, and inhibition and activation of Smad signaling were also applied to validate the involvement of Smad in BMMSCs during KGN treatment. In summary, this study shows that KGN promotes osteogenic differentiation of BMMSCs through enhancing autophagic levels and upregulating Smad1/5/9 signaling mechanically.
Collapse
Affiliation(s)
- Huichun Yan
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Tingting Yu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Jing Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Ting Zhang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Qian Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Yanheng Zhou
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Dawei Liu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
- State Key Laboratory of Military Stomatology, Xi'an 710000, China
| |
Collapse
|
12
|
Yang C, Liu Y, Wang Z, Lin M, Liu C. Controlled mechanical loading improves bone regeneration by regulating type H vessels in a S1Pr1-dependent manner. FASEB J 2022; 36:e22530. [PMID: 36063128 DOI: 10.1096/fj.202200339rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 08/04/2022] [Accepted: 08/22/2022] [Indexed: 11/11/2022]
Abstract
Despite the best treatment, approximately 10% of fractures still face undesirable repair and result in delayed unions or non-unions. Dynamic mechanical stimulation promotes bone formation, when applied at the correct time frame, with optimal loading magnitude, frequency, and repetition. Controlled mechanical loading significantly increases osteogenic cells during the matrix deposition phase of bone repair. In the bone defect, the blood vessel network guides the initial bone formation activities. A unique blood vessel subtype (Type H) exists in bone, which expresses high levels of CD31 and endomucin, and functions to couple angiogenesis and osteogenesis. However, how this form of controlled mechanical loading regulates the Type H vessels and promotes bone formation is still not clear. Sphingosine 1-phosphate (S1P) participates in the bone anabolic process and is a key regulator of the blood vessel. Its receptor, sphingosine 1-phosphate receptor 1 (S1Pr1), is a mechanosensitive protein that regulates vascular integrity. Therefore, we hypothesis that controlled anabolic mechanical loading promotes bone repair by acting on Type H vessels. To study the effect of S1Pr1 on loading induced-bone repair, we utilized a stabilized tibial defect model, which allows for the application of anabolic mechanical loading. Mechanical loading upregulated S1Pr1 within the entire defect, with up to 80% expressed in blood vessels, as observed by deep tissue imaging. Additionally, S1Pr1 antagonism by W146 inhibited the anabolic effects of mechanical loading. We showed that mechanical loading or activating S1Pr1 could induce YAP nuclear translocation, a key regulator in the cell's mechanical response, in endothelial cells (ECs) in vitro. Inhibition of S1Pr1 in endothelial cells by siRNA reduced loading-induced YAP nuclear translocation and expressions of angiogenic genes. In vivo, YAP nuclear translocation in Type H vessels was up-regulated after mechanical loading but was inhibited by antagonizing S1Pr1. S1Pr1 agonist, FTY720, increased bone volume and Type H vessel volume, similar to that of mechanical stimulation. In conclusion, controlled anabolic mechanical loading enhanced bone formation mainly through Type H vessels in a S1Pr1-dependent manner.
Collapse
Affiliation(s)
- Chengyu Yang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Yang Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Ziyan Wang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Minmin Lin
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Chao Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
13
|
Yan J, Chen Y, Wu Q, Shao L, Zhou X. Expression of sphingosine‑1‑phosphate receptor 2 is correlated with migration and invasion of human colon cancer cells: A preliminary clinical study. Oncol Lett 2022; 24:241. [PMID: 35720474 PMCID: PMC9185157 DOI: 10.3892/ol.2022.13361] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 03/15/2022] [Indexed: 11/27/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive phospholipid that serves as a potent mediator of cell proliferation, differentiation and apoptosis by binding to S1P receptors (S1PRs). S1P signalling is involved in the pathogenesis of numerous types of disease, including cancer. To the best of our knowledge, however, little is known about the expression patterns of S1PRs and their role in human colorectal cancer (CRC) cell migration and invasion. The aim of the present study was to investigate the role of S1P signalling in the metastasis of colon cancer cells and the expression of S1PRs in patients with CRC. The protein and mRNA expression levels of S1PRs and sphingosine kinases (SPHKs) in 55 patients with CRC were detected by western blotting (WB), immunohistochemical (IHC) analysis and reverse transcription-quantitative PCR. The levels of S1P in serum from patients and healthy individuals were quantified by ELISA. S1PRs antagonists JTE013, FTY720 and S1PR2-small interfering (si)RNA were used to determine the role of S1PR2 in human CRC LOVO and SW480 cell lines. Migration and invasion assays were performed for functional analysis. The levels of S1P in serum were significantly increased in patients with CRC compared with healthy individuals. The relative mRNA expression levels of S1PR2 were significantly downregulated in tumour compared with normal tissue, whereas S1PR1 and SPHK1 were upregulated. WB showed that 58% (32/55 cases) of patients presented downregulated S1PR2 protein expression. IHC analysis indicated that expression of S1PR2 was lower in tumour than in normal tissue in 65.5% (36/55 cases) of patients. Exogenous addition of S1P promoted migration and invasion in the different cell types. S1P stimulated the migration and invasion of SW480 cells. The inhibition of S1PR2 by JTE013 or S1PR2-siRNA significantly promoted the migration and invasion of SW480 cells, while FTY720 reversed these effects. The present study indicated that expression levels of S1PRs, particularly S1PR2, were associated with migration and invasion of CRC cells. The present findings revealed a novel mechanism by which S1P inhibited tumour cell migration and invasion via a S1PR2-dependent pathway, suggesting that S1PR2 may be a therapeutic target for treatment of colon cancer.
Collapse
Affiliation(s)
- Junjun Yan
- Department of Gastroenterology, The First People's Hospital of Jiujiang, Jiujiang, Jiangxi 332000, P.R. China
| | - Yi Chen
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Qibiao Wu
- State Key Laboratory of Quality Research in Chinese Medicines and Faculty of Chinese Medicine, Macau University of Science and Technology (MUST), Taipa, Macau 999078, P.R. China
| | - Le Shao
- Center for Medical Research and Innovation, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410000, P.R. China
| | - Xiqiao Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
14
|
Choi B, Kim JE, Park SO, Kim EY, Oh S, Choi H, Yoon D, Min HJ, Kim HR, Chang EJ. Sphingosine-1-phosphate hinders the osteogenic differentiation of dental pulp stem cells in association with AKT signaling pathways. Int J Oral Sci 2022; 14:21. [PMID: 35459199 PMCID: PMC9033766 DOI: 10.1038/s41368-022-00173-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/24/2022] [Accepted: 02/24/2022] [Indexed: 11/16/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is an important lipid mediator that regulates a diverse range of intracellular cell signaling pathways that are relevant to tissue engineering and regenerative medicine. However, the precise function of S1P in dental pulp stem cells (DPSCs) and its osteogenic differentiation remains unclear. We here investigated the function of S1P/S1P receptor (S1PR)-mediated cellular signaling in the osteogenic differentiation of DPSCs and clarified the fundamental signaling pathway. Our results showed that S1P-treated DPSCs exhibited a low rate of differentiation toward the osteogenic phenotype in association with a marked reduction in osteogenesis-related gene expression and AKT activation. Of note, both S1PR1/S1PR3 and S1PR2 agonists significantly downregulated the expression of osteogenic genes and suppressed AKT activation, resulting in an attenuated osteogenic capacity of DPSCs. Most importantly, an AKT activator completely abrogated the S1P-mediated downregulation of osteoblastic markers and partially prevented S1P-mediated attenuation effects during osteogenesis. Intriguingly, the pro-inflammatory TNF-α cytokine promoted the infiltration of macrophages toward DPSCs and induced S1P production in both DPSCs and macrophages. Our findings indicate that the elevation of S1P under inflammatory conditions suppresses the osteogenic capacity of the DPSCs responsible for regenerative endodontics.
Collapse
Affiliation(s)
- Bongkun Choi
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ji-Eun Kim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Si-On Park
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Eun-Young Kim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Soyoon Oh
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyuksu Choi
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Dohee Yoon
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyo-Jin Min
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyung-Ryong Kim
- Department of Pharmacology, College of Dentistry, Jeonbuk National University, Jeonju, Korea.
| | - Eun-Ju Chang
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea. .,Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea. .,Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
15
|
Ahmadzadeh K, Vanoppen M, Rose CD, Matthys P, Wouters CH. Multinucleated Giant Cells: Current Insights in Phenotype, Biological Activities, and Mechanism of Formation. Front Cell Dev Biol 2022; 10:873226. [PMID: 35478968 PMCID: PMC9035892 DOI: 10.3389/fcell.2022.873226] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/17/2022] [Indexed: 12/21/2022] Open
Abstract
Monocytes and macrophages are innate immune cells with diverse functions ranging from phagocytosis of microorganisms to forming a bridge with the adaptive immune system. A lesser-known attribute of macrophages is their ability to fuse with each other to form multinucleated giant cells. Based on their morphology and functional characteristics, there are in general three types of multinucleated giant cells including osteoclasts, foreign body giant cells and Langhans giant cells. Osteoclasts are bone resorbing cells and under physiological conditions they participate in bone remodeling. However, under pathological conditions such as rheumatoid arthritis and osteoporosis, osteoclasts are responsible for bone destruction and bone loss. Foreign body giant cells and Langhans giant cells appear only under pathological conditions. While foreign body giant cells are found in immune reactions against foreign material, including implants, Langhans giant cells are associated with granulomas in infectious and non-infectious diseases. The functionality and fusion mechanism of osteoclasts are being elucidated, however, our knowledge on the functions of foreign body giant cells and Langhans giant cells is limited. In this review, we describe and compare the phenotypic aspects, biological and functional activities of the three types of multinucleated giant cells. Furthermore, we provide an overview of the multinucleation process and highlight key molecules in the different phases of macrophage fusion.
Collapse
Affiliation(s)
- Kourosh Ahmadzadeh
- Laboratory of Immunobiology, Department Microbiology and Immunology, Rega Institute, KU Leuven – University of Leuven, Leuven, Belgium
- *Correspondence: Kourosh Ahmadzadeh, ; Carine Helena Wouters,
| | - Margot Vanoppen
- Laboratory of Immunobiology, Department Microbiology and Immunology, Rega Institute, KU Leuven – University of Leuven, Leuven, Belgium
| | - Carlos D. Rose
- Division of Pediatric Rheumatology Nemours Children’s Hospital, Thomas Jefferson University, Philadelphia, PA, United States
| | - Patrick Matthys
- Laboratory of Immunobiology, Department Microbiology and Immunology, Rega Institute, KU Leuven – University of Leuven, Leuven, Belgium
| | - Carine Helena Wouters
- Laboratory of Immunobiology, Department Microbiology and Immunology, Rega Institute, KU Leuven – University of Leuven, Leuven, Belgium
- Division Pediatric Rheumatology, UZ Leuven, Leuven, Belgium
- European Reference Network for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) at University Hospital Leuven, Leuven, Belgium
- *Correspondence: Kourosh Ahmadzadeh, ; Carine Helena Wouters,
| |
Collapse
|
16
|
Matsuzaki E, Hirose H, Fujimasa S, Yoshimoto S, Yanagi T, Matsumoto K, Nikaido M, Minakami M, Matsumoto N, Anan H. Sphingosine-1-phosphate receptor 2 agonist induces bone formation in rat apicoectomy and alveolar bone defect model. J Dent Sci 2022; 17:787-794. [PMID: 35756763 PMCID: PMC9201516 DOI: 10.1016/j.jds.2021.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/04/2021] [Indexed: 01/01/2023] Open
Affiliation(s)
- Etsuko Matsuzaki
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
- Corresponding author. Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka, 814-0193, Japan.
| | - Haruna Hirose
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan
| | - Seishiro Fujimasa
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan
| | - Shohei Yoshimoto
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
- Section of Pathology, Department of Morphological Biology, Division of Biomedical Sciences, Fukuoka Dental College, Fukuoka, Japan
| | - Tsukasa Yanagi
- Section of Oral Implantology, Department of Oral Rehabilitation, Fukuoka Dental College, Fukuoka, Japan
| | - Kazuma Matsumoto
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan
| | - Misaki Nikaido
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan
| | - Masahiko Minakami
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan
| | - Noriyoshi Matsumoto
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan
| | - Hisashi Anan
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan
| |
Collapse
|
17
|
Lin S, Pandruvada S, Yu H. Inhibition of Sphingosine-1-Phosphate Receptor 2 by JTE013 Promoted Osteogenesis by Increasing Vesicle Trafficking, Wnt/Ca 2+, and BMP/Smad Signaling. Int J Mol Sci 2021; 22:ijms222112060. [PMID: 34769490 PMCID: PMC8584480 DOI: 10.3390/ijms222112060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/25/2021] [Accepted: 11/05/2021] [Indexed: 12/31/2022] Open
Abstract
Sphingosine-1-phosphate receptor 2 (S1PR2) is a G protein-coupled receptor that regulates various immune responses. Herein, we determine the effects of a S1PR2 antagonist (JTE013) or a S1PR2 shRNA on osteogenesis by culturing murine bone marrow stromal cells (BMSCs) in osteogenic media with JTE013, dimethylsulfoxide (DMSO), a S1PR2 shRNA, or a control shRNA. Treatment with JTE013 or the S1PR2 shRNA increased alkaline phosphatase and alizarin red s staining, and enhanced alkaline phosphatase, RUNX2, osteocalcin, and osterix mRNA levels in BMSCs compared with the controls. Protein analysis revealed that a high dose of JTE013 (4 or 8 μM) increased vesicle trafficking-associated proteins (F-actin, clathrin, Early Endosome Antigen 1 (EEA1), and syntaxin 6) and Wnt/Ca2+ signaling. On the other hand, a low dose of JTE013 (1 to 2 μM) increased BMP/Smad signaling. In contrast, the S1PR2 shRNA reduced vesicle trafficking-associated proteins and attenuated Wnts and BMP/Smad signaling, but enhanced p-CaMKII compared with the control, suggesting that the S1PR2 shRNA influenced osteogenesis via different signaling pathways. Moreover, inhibiting protein trafficking by brefeldin A in BMSCs suppressed Wnts and BMPRs expressions. These data supported that enhanced osteogenesis in JTE013-treated BMSCs is associated with increased vesicle trafficking, which promotes the synthesis and transport of osteogenic protein and matrix vesicles and enhances matrix mineralization.
Collapse
|
18
|
Mechanisms of sphingosine-1-phosphate (S1P) signaling on excessive stress-induced root resorption during orthodontic molar intrusion. Clin Oral Investig 2021; 26:1003-1016. [PMID: 34363103 DOI: 10.1007/s00784-021-04084-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 07/15/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVES The aim of this study was to investigate cementocyte mechanotransduction during excessive orthodontic intrusive force-induced root resorption and the role of S1P signaling in this process. MATERIALS AND METHODS Fifty-four 12-week-old male Wistar rats were randomly divided into 3 groups: control group (Control), intrusive stress application group (Stress), and intrusive stress together with S1PR2-specific antagonist injection group (Stress + JTE). A rat molar intrusion model was established on animals in the Stress and the Stress + JTE groups. The animals in the Stress + JTE group received daily intraperitoneal (i.p.) injection of S1PR2 antagonist JTE-013, while the Control and Stress groups received only the vehicle. Histomorphometric, immunohistochemical, quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analyses were performed after euthanizing of the rats. RESULTS Root resorption was promoted in the Stress group with increased volumes of resorption pits and amounts of molar intrusion compared with the Control group. The expression levels of cementogenic- and cementoclastic-related factors were affected under excessive intrusive force. Immunohistochemical staining and qRT-PCR analysis showed promoted S1P signaling activities during molar intrusion. Western blot analysis indicated decreased nuclear translocation of β-catenin under excessive intrusive force. Through the administration of JTE-013, S1P signaling activity was suppressed and excessive intrusive force-induced root resorption was reversed. The regulation of S1P signaling could also influence the nuclear translocation of β-catenin and the expressions of cementogenic- and cementoclastic-related factors. CONCLUSIONS Root resorption was promoted under excessive orthodontic intrusive force due to the disruption of cementum homeostasis. S1P signaling pathway might play an important role in cementocyte mechanotransduction in this process. CLINICAL RELEVANCE The S1P signaling might be a promising therapeutic target for novel therapeutic approaches to prevent external root resorption caused by excessive orthodontic intrusive force.
Collapse
|
19
|
Kalinkovich A, Livshits G. Biased and allosteric modulation of bone cell-expressing G protein-coupled receptors as a novel approach to osteoporosis therapy. Pharmacol Res 2021; 171:105794. [PMID: 34329703 DOI: 10.1016/j.phrs.2021.105794] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/20/2021] [Accepted: 07/25/2021] [Indexed: 12/16/2022]
Abstract
On the cellular level, osteoporosis (OP) is a result of imbalanced bone remodeling, in which osteoclastic bone resorption outcompetes osteoblastic bone formation. Currently available OP medications include both antiresorptive and bone-forming drugs. However, their long-term use in OP patients, mainly in postmenopausal women, is accompanied by severe side effects. Notably, the fundamental coupling between bone resorption and formation processes underlies the existence of an undesirable secondary outcome that bone anabolic or anti-resorptive drugs also reduce bone formation. This drawback requires the development of anti-OP drugs capable of selectively stimulating osteoblastogenesis and concomitantly reducing osteoclastogenesis. We propose that the application of small synthetic biased and allosteric modulators of bone cell receptors, which belong to the G-protein coupled receptors (GPCR) family, could be the key to resolving the undesired anti-OP drug selectivity. This approach is based on the capacity of these GPCR modulators, unlike the natural ligands, to trigger signaling pathways that promote beneficial effects on bone remodeling while blocking potentially deleterious effects. Under the settings of OP, an optimal anti-OP drug should provide fine-tuned regulation of downstream effects, for example, intermittent cyclic AMP (cAMP) elevation, preservation of Ca2+ balance, stimulation of osteoprotegerin (OPG) and estrogen production, suppression of sclerostin secretion, and/or preserved/enhanced canonical β-catenin/Wnt signaling pathway. As such, selective modulation of GPCRs involved in bone remodeling presents a promising approach in OP treatment. This review focuses on the evidence for the validity of our hypothesis.
Collapse
Affiliation(s)
- Alexander Kalinkovich
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6905126, Israel
| | - Gregory Livshits
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6905126, Israel; Adelson School of Medicine, Ariel University, Ariel 4077625, Israel.
| |
Collapse
|
20
|
Jang JH, Kim MS, Antao AM, Jo WJ, Kim HJ, Kim SJ, Choi MJ, Ramakrishna S, Kim KS. Bioactive Lipid O-cyclic phytosphingosine-1-phosphate Promotes Differentiation of Human Embryonic Stem Cells into Cardiomyocytes via ALK3/BMPR Signaling. Int J Mol Sci 2021; 22:ijms22137015. [PMID: 34209900 PMCID: PMC8267745 DOI: 10.3390/ijms22137015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 12/04/2022] Open
Abstract
Adult human cardiomyocytes have an extremely limited proliferative capacity, which poses a great barrier to regenerative medicine and research. Human embryonic stem cells (hESCs) have been proposed as an alternative source to generate large numbers of clinical grade cardiomyocytes (CMs) that can have potential therapeutic applications to treat cardiac diseases. Previous studies have shown that bioactive lipids are involved in diverse cellular responses including cardiogenesis. In this study, we explored the novel function of the chemically synthesized bioactive lipid O-cyclic phytosphingosine-1-phosphate (cP1P) as an inducer of cardiac differentiation. Here, we identified cP1P as a novel factor that significantly enhances the differentiation potential of hESCs into cardiomyocytes. Treatment with cP1P augments the beating colony number and contracting area of CMs. Furthermore, we elucidated the molecular mechanism of cP1P regulating SMAD1/5/8 signaling via the ALK3/BMP receptor cascade during cardiac differentiation. Our result provides a new insight for cP1P usage to improve the quality of CM differentiation for regenerative therapies.
Collapse
Affiliation(s)
- Ji-Hye Jang
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (J.-H.J.); (M.-S.K.); (A.M.A.); (W.-J.J.); (H.-J.K.)
| | - Min-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (J.-H.J.); (M.-S.K.); (A.M.A.); (W.-J.J.); (H.-J.K.)
| | - Ainsley Mike Antao
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (J.-H.J.); (M.-S.K.); (A.M.A.); (W.-J.J.); (H.-J.K.)
| | - Won-Jun Jo
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (J.-H.J.); (M.-S.K.); (A.M.A.); (W.-J.J.); (H.-J.K.)
| | - Hyung-Joon Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (J.-H.J.); (M.-S.K.); (A.M.A.); (W.-J.J.); (H.-J.K.)
| | - Su-Jin Kim
- Axceso Biopharma Co., Ltd., Yongin 16914, Korea; (S.-J.K.); (M.-J.C.)
| | - Myeong-Jun Choi
- Axceso Biopharma Co., Ltd., Yongin 16914, Korea; (S.-J.K.); (M.-J.C.)
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (J.-H.J.); (M.-S.K.); (A.M.A.); (W.-J.J.); (H.-J.K.)
- College of Medicine, Hanyang University, Seoul 04763, Korea
- Correspondence: or (S.R.); (K.-S.K.)
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (J.-H.J.); (M.-S.K.); (A.M.A.); (W.-J.J.); (H.-J.K.)
- College of Medicine, Hanyang University, Seoul 04763, Korea
- Correspondence: or (S.R.); (K.-S.K.)
| |
Collapse
|
21
|
Matsuzaki E, Minakami M, Matsumoto N, Anan H. Dental regenerative therapy targeting sphingosine-1-phosphate (S1P) signaling pathway in endodontics. JAPANESE DENTAL SCIENCE REVIEW 2020; 56:127-134. [PMID: 33088365 PMCID: PMC7567953 DOI: 10.1016/j.jdsr.2020.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/02/2020] [Accepted: 09/18/2020] [Indexed: 12/31/2022] Open
Abstract
The establishment of regenerative therapy in endodontics targeting the dentin-pulp complex, cementum, periodontal ligament tissue, and alveolar bone will provide valuable information to preserve teeth. It is well known that the application of stem cells such as induced pluripotent stem cells, embryonic stem cells, and somatic stem cells is effective in regenerative medicine. There are many somatic stem cells in teeth and periodontal tissues including dental pulp stem cells (DPSCs), stem cells from the apical papilla, and periodontal ligament stem cells. Particularly, several studies have reported the regeneration of clinical pulp tissue and alveolar bone by DPSCs transplantation. However, further scientific issues for practical implementation remain to be addressed. Sphingosine-1-phosphate (S1P) acts as a bioactive signaling molecule that has multiple biological functions including cellular differentiation, and has been shown to be responsible for bone resorption and formation. Here we discuss a strategy for bone regeneration and a possibility for regenerative endodontics targeting S1P signaling pathway as one of approaches for induction of regeneration by improving the regenerative capacity of endogenous cells. SCIENTIFIC FIELD OF DENTAL SCIENCE Endodontology.
Collapse
Affiliation(s)
- Etsuko Matsuzaki
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Masahiko Minakami
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan
| | - Noriyoshi Matsumoto
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan
| | - Hisashi Anan
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan
| |
Collapse
|
22
|
Kang X, Qian Z, Liu J, Feng D, Li H, Zhang Z, Jin X, Ma Z, Xu M, Li F, Zhang Y, Gao X, Sun H, Wu S. Neuropeptide Y Acts Directly on Cartilage Homeostasis and Exacerbates Progression of Osteoarthritis Through NPY2R. J Bone Miner Res 2020; 35:1375-1384. [PMID: 32101625 DOI: 10.1002/jbmr.3991] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/20/2020] [Accepted: 02/22/2020] [Indexed: 12/22/2022]
Abstract
Neuropeptide Y (NPY) is known to regulate bone homeostasis; however, its functional role as a risk factor during osteoarthritis (OA) remains elusive. In this study, we aim to investigate the direct effect of NPY on degradation of cartilage and progression of OA and explore the molecular events involved. NPY was overexpressed in human OA cartilage accompanied with increased expression of NPY1 receptor (NPY1R) and NPY2 receptor (NPY2R). Stressors such as cold exposure resulted in the peripheral release of NPY from sympathetic nerves, which in turn promoted upregulation of NPY and NPY2R in articular cartilage in vivo. Intra-articular administration of NPY significantly promoted chondrocyte hypertrophy and cartilage matrix degradation, with a higher OARSI score than that of control mice, whereas inhibition of NPY2R but not NPY1R with its specific antagonist remarkably ameliorated NPY-mediated effects. Moreover, NPY activated mTORC1 pathway in articular chondrocytes, whereas the administration of rapamycin (an mTORC1 inhibitor) in vitro abrogated NPY-mediated effects. Mechanistically, mTORC1 downstream kinase S6K1 interacted with and phosphorylated SMAD1/5/8 and promoted SMAD4 nuclear translocation, resulting in upregulation of Runx2 expression to promote chondrocyte hypertrophy and cartilage degradation. In conclusion, our findings provided the direct evidence and the crucial role of NPY in cartilage homeostasis. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Xiaomin Kang
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, PR China
| | - Zhuang Qian
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, PR China
| | - Jiali Liu
- Department of Clinical Laboratory, the Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, PR China
| | - Dongxu Feng
- Hong Hui Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, PR China
| | - Huixia Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, PR China
| | - Zhuanmin Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, PR China
| | - Xinxin Jin
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, PR China
| | - Zhengmin Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, PR China
| | - Mao Xu
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, PR China
| | - Fang Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, PR China
| | - Ying Zhang
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, PR China
| | - Xin Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, PR China
| | - Hongzhi Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, PR China
| | - Shufang Wu
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, PR China
| |
Collapse
|
23
|
Wang Z, Liu Q, Liu C, Tan W, Tang M, Zhou X, Sun T, Deng Y. Mg 2+ in β-TCP/Mg-Zn composite enhances the differentiation of human bone marrow stromal cells into osteoblasts through MAPK-regulated Runx2/Osx. J Cell Physiol 2020; 235:5182-5191. [PMID: 31742679 DOI: 10.1002/jcp.29395] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/27/2019] [Indexed: 12/15/2022]
Abstract
Inducing the osteogenic differentiation from bone marrow stromal cells (BMSCs) might be a potent strategy for treating bone loss and nonunion during fracture and improving fracture healing. Among several signaling pathways involved, mitogen-activated protein kinases (MAPKs) have been reported to play a critical role. Magnesium (Mg)-based alloys, including Mg-Zn alloy, have been used clinically as implants in the musculoskeletal field and could promote BMSC osteogenic differentiation. However, the underlying mechanisms remain unclear. In this study, we produced Mg-Zn alloy consists of Mg and low concentrations of Zn, calcium carbonate, and β-tricalcium phosphate (β-TCP; manifesting process not shown), prepared Mg, Zn, and Mg-Zn extracts, and investigated the specific effects of these extracts on human BMSC (hBMSC) osteogenic differentiation and MAPK signaling. Mg extracts and Mg-Zn extracts could significantly promote the osteogenic differentiation of hBMSCs as manifested as increased alkaline phosphatase levels, enhanced calcium nodules formation, and increased messenger RNA expression and protein levels of osteogenesis markers, including BMPs, Col-I, Runx2, and Osx; in the meantime, Mg culture medium (CM) and Mg-Zn CM both significantly enhanced the activation of MAPK signaling in hBMSCs. By adding ERK1/2 signaling, p38 signaling, or JNK signaling inhibitor to Mg-Zn CM, or conducting p38 MAPK silence in hBMSCs, we revealed that these extracts might promote hBMSC osteogenic differentiation via p38 MAPK signaling and MAPK-regulated Runx2/Osx. In conclusion, Mg2+ in β-TCP/Mg-Zn extract promotes the osteogenic differentiation of hBMSCs via MAPK-regulated Runx2/Osx interaction.
Collapse
Affiliation(s)
- Zhenting Wang
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Urology Surgery, Haikou People's Hospital/Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, China
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan
| | - Qing Liu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan
| | - Congcong Liu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan
| | - Wei Tan
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan
| | - Mingying Tang
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaohua Zhou
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tianshi Sun
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Youwen Deng
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan
| |
Collapse
|
24
|
Zhang L, Dong Y, Wang Y, Hu W, Dong S, Chen Y. Sphingosine-1-phosphate (S1P) receptors: Promising drug targets for treating bone-related diseases. J Cell Mol Med 2020; 24:4389-4401. [PMID: 32155312 PMCID: PMC7176849 DOI: 10.1111/jcmm.15155] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/22/2020] [Accepted: 02/01/2020] [Indexed: 12/20/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a natural bioactive lipid molecule and a common first or second messenger in the cardiovascular and immune systems. By binding with its receptors, S1P can serve as mediator of signalling during cell migration, differentiation, proliferation and apoptosis. Although the predominant role of S1P in bone regeneration has been noted in many studies, this role is not as well-known as its roles in the cardiovascular and immune systems. In this review, we summarize previous research on the role of S1P receptors (S1PRs) in osteoblasts and osteoclasts. In addition, S1P is regarded as a bridge between bone resorption and formation, which brings hope to patients with bone-related diseases. Finally, we discuss S1P and its receptors as therapeutic targets for treating osteoporosis, inflammatory osteolysis and bone metastasis based on the biological effects of S1P in osteoclastic/osteoblastic cells, immune cells and tumour cells.
Collapse
Affiliation(s)
- Lincheng Zhang
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China.,Battalion One of Basic Medical Sciences, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yutong Dong
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China.,Battalion One of Basic Medical Sciences, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yiran Wang
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China
| | - Wenhui Hu
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, China.,Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yueqi Chen
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China.,Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
25
|
El Jamal A, Briolay A, Mebarek S, Le Goff B, Blanchard F, Magne D, Brizuela L, Bougault C. Cytokine-Induced and Stretch-Induced Sphingosine 1-Phosphate Production by Enthesis Cells Could Favor Abnormal Ossification in Spondyloarthritis. J Bone Miner Res 2019; 34:2264-2276. [PMID: 31373726 DOI: 10.1002/jbmr.3844] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 07/11/2019] [Accepted: 07/19/2019] [Indexed: 12/12/2022]
Abstract
Spondyloarthritis (SpA) is a common rheumatic disease characterized by enthesis inflammation (enthesitis) and ectopic ossification (enthesophytes). The current pathogenesis model suggests that inflammation and mechanical stress are both strongly involved in SpA pathophysiology. We have previously observed that the levels of sphingosine 1-phosphate (S1P), a bone anabolic molecule, were particularly high in SpA patients' serum compared to healthy donors. Therefore, we wondered how this deregulation was related to SpA molecular mechanisms. Mouse primary osteoblasts, chondrocytes, and tenocytes were used as cell culture models. The sphingosine kinase 1 (Sphk1) gene expression and S1P secretion were significantly enhanced by cyclic stretch in osteoblasts and chondrocytes. Further, TNF-α and IL-17, cytokines implicated in enthesitis, increased Sphk1 mRNA in chondrocytes in an additive manner when combined to stretch. The immunochemistry on mouse ankles showed that sphingosine kinase 1 (SK1) was localized in some chondrocytes; the addition of a pro-inflammatory cocktail augmented Sphk1 expression in cultured ankles. Subsequently, fingolimod was used to block S1P metabolism in cell cultures. It inhibited S1P receptors (S1PRs) signaling and SK1 and SK2 activity in both osteoblasts and chondrocytes. Fingolimod also reduced S1PR-induced activation by SpA patients' synovial fluid (SF), demonstrating that the stimulation of chondrocytes by SFs from SpA patients involves S1P. In addition, when the osteogenic culture medium was supplemented with fingolimod, alkaline phosphatase activity, matrix mineralization, and bone formation markers were significantly reduced in osteoblasts and hypertrophic chondrocytes. Osteogenic differentiation was accompanied by an increase in S1prs mRNA, especially S1P1/3 , but their contribution to S1P-impact on mineralization seemed limited. Our results suggest that S1P might be overproduced in SpA enthesis in response to cytokines and mechanical stress, most likely by chondrocytes. Moreover, S1P could locally favor the abnormal ossification of the enthesis; therefore, blocking the S1P metabolic pathway could be a potential therapeutic approach for the treatment of SpA. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Alaeddine El Jamal
- Univ Lyon, Univ Claude Bernard Lyon 1 (UCBL), CNRS, UMR5246, Lyon, France
| | - Anne Briolay
- Univ Lyon, Univ Claude Bernard Lyon 1 (UCBL), CNRS, UMR5246, Lyon, France
| | - Saida Mebarek
- Univ Lyon, Univ Claude Bernard Lyon 1 (UCBL), CNRS, UMR5246, Lyon, France
| | - Benoit Le Goff
- INSERM UMR1238, Nantes University, Nantes, France.,Rheumatology Department, Nantes University, Nantes, France
| | | | - David Magne
- Univ Lyon, Univ Claude Bernard Lyon 1 (UCBL), CNRS, UMR5246, Lyon, France
| | - Leyre Brizuela
- Univ Lyon, Univ Claude Bernard Lyon 1 (UCBL), CNRS, UMR5246, Lyon, France
| | - Carole Bougault
- Univ Lyon, Univ Claude Bernard Lyon 1 (UCBL), CNRS, UMR5246, Lyon, France
| |
Collapse
|
26
|
Lu XD, Han WX, Liu YX. Suppression of miR-451a accelerates osteogenic differentiation and inhibits bone loss via Bmp6 signaling during osteoporosis. Biomed Pharmacother 2019; 120:109378. [PMID: 31541885 DOI: 10.1016/j.biopha.2019.109378] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 08/16/2019] [Accepted: 08/22/2019] [Indexed: 11/15/2022] Open
Abstract
Bone homeostasis is known as a dynamic balance, including bone formation through osteoblasts and bone resorption by osteoclasts. MicroRNAs (miRs) play a critical role in regulating bone formation and homeostasis. In the study, the effects of miR-451a on bone homeostasis were investigated. The results indicated that the primary osteoblasts and mesenchymal stem cells (MSCs), as the main source of osteoblasts, isolated from miR-451a-knockout (KO) mice showed promoted osteogenesis. in vivo, an ovariectomized (OVX) animal model was used to further explore the effect of miR-451a on osteoporosis. Micro-computed tomography (μCT) indicated a promoted bone volume in miR-451a-KO mice compared to wild-type (WT) mice after OVX operation, demonstrating a redundant bone formation after the knockout of miR-451a. Importantly, we for the first time found that bone morphogenetic protein 6 (Bmp6) was a direct target of miR-451a, elevating bone formation through regulating SMAD1/5/8 expression. In conclusion, reducing miR-451a expression levels could enhance bone formation during the progression of osteoporosis, which might be at least partly via the meditation of Bmp6 expression.
Collapse
Affiliation(s)
- Xiang-Dong Lu
- Department of Orthopedics, The Second Hospital of Shan Xi Medical University, Taiyuan City, Shanxi Province, 030001, China
| | - Wen-Xing Han
- Department of Orthopedics Dept. Unit 6, The Seventh Medical Center of PLA General Hospital, Beijing, 100700, China
| | - Yan-Xiong Liu
- Departmentof Spinal Surgery, Affiliated Hospital of Yan'anUniversity, Yan'anCity, Shaanxi Province, 716000, China.
| |
Collapse
|
27
|
Weske S, Vaidya M, von Wnuck Lipinski K, Keul P, Manthe K, Burkhart C, Haberhauer G, Heusch G, Levkau B. Agonist-induced activation of the S1P receptor 2 constitutes a novel osteoanabolic therapy for the treatment of osteoporosis in mice. Bone 2019; 125:1-7. [PMID: 31028959 DOI: 10.1016/j.bone.2019.04.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/09/2019] [Accepted: 04/23/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND PURPOSE Osteoporosis is a worldwide epidemic but pharmacological agents to stimulate new bone formation are scarce. We have shown that increasing tissue levels of sphingosine-1-phosphate (S1P) by blocking its degradation by the S1P lyase has pronounced osteoanabolic effect in mouse osteoporosis models by stimulating osteoblast differentiation through the S1P receptor 2 (S1P2). However, S1P lyase inhibitors have side effects complicating potential clinical use. Here, we tested whether direct S1P2 engagement by the S1P2 agonist CYM5520 exerted osteoanabolic potential in estrogen deficiency-induced osteopenia in mice. We compared its efficacy to LX2931, a novel S1P lyase inhibitor currently tested in rheumatoid arthritis. EXPERIMENTAL APPROACH CYM5520, LX2931 or vehicle were administered to ovariectomized mice for 6 weeks beginning 5 weeks after ovariectomy, Bone mass, cellular composition and mechanical strength were assessed by microCT, histomorphometry and three point bending tests. Plasma markers of bone metabolism were analyzed by ELISA. KEY RESULTS Therapeutic treatment with CYM5520 and LX2931 clearly increased long bone and vertebral bone mass to impressive 3-5 fold over vehicle in osteopenic ovariectomized mice. As expected, lymphopenia was a side effect of LX2931, whereas none occurred with CYM5520. Consistent with an osteoanabolic effect, CYM5520 increased osteoblast number, osteoid surface and alkaline phosphatase area 2-3 fold over vehicle. Plasma concentrations of the osteoanabolic marker procollagen I C-terminal propeptide were also elevated by CYM5520 and LX2931. LX2931 but not yet CYM5520 increased cortical thickness and mechanical strength without affecting mineral density. CONCLUSION AND IMPLICATIONS Treatment with a pharmacological S1P2 agonist corrected ovariectomy-induced osteopenia in mice by inducing new bone formation thus constituting a novel osteoanabolic approach to osteoporosis.
Collapse
Affiliation(s)
- Sarah Weske
- Institute for Pathophysiology, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Mithila Vaidya
- Institute for Pathophysiology, University Hospital Essen, University of Duisburg-Essen, Germany
| | | | - Petra Keul
- Institute for Pathophysiology, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Kristina Manthe
- Institute for Pathophysiology, University Hospital Essen, University of Duisburg-Essen, Germany
| | | | | | - Gerd Heusch
- Institute for Pathophysiology, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Bodo Levkau
- Institute for Pathophysiology, University Hospital Essen, University of Duisburg-Essen, Germany.
| |
Collapse
|
28
|
Peng XY, Hu M, Liao F, Yang F, Ke QF, Guo YP, Zhu ZH. La-Doped mesoporous calcium silicate/chitosan scaffolds for bone tissue engineering. Biomater Sci 2019; 7:1565-1573. [PMID: 30688345 DOI: 10.1039/c8bm01498a] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Trace rare earth elements such as lanthanum (La) regulated effectively bone tissue performances; however, the underlying mechanism remains unknown. In order to accelerate bone defects especially in patients with osteoporosis or other metabolic diseases, we firstly constructed lanthanum-doped mesoporous calcium silicate/chitosan (La-MCS/CTS) scaffolds by freeze-drying technology. During the freeze-drying procedure, three-dimensional macropores were produced within the La-MCS/CTS scaffolds by using ice crystals as templates, and the La-MCS nanoparticles were distributed on the macropore walls. The hierarchically porous structures and biocompatible components contributed to the adhesion, spreading and proliferation of rat bone marrow-derived mesenchymal stem cells (rBMSCs), and accelerated the in-growth of new bone tissues. Particularly, the La3+ ions in the bone scaffolds remarkably induced the osteogenic differentiation of rBMSCs via the activation of the TGF signal pathway. A critical-sized calvarial-defect rat model further revealed that the La-MCS/CTS scaffolds significantly promoted new bone regeneration as compared with pure MCS/CTS scaffolds. In conclusion, the La-MCS/CTS scaffold showed the prominent ability in osteogenesis and bone regeneration, which showed its application potential for bone defect therapy.
Collapse
Affiliation(s)
- Xiao-Yuan Peng
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P. R. China.
| | | | | | | | | | | | | |
Collapse
|
29
|
Xiao L, Zhou Y, Friis T, Beagley K, Xiao Y. S1P-S1PR1 Signaling: the "Sphinx" in Osteoimmunology. Front Immunol 2019; 10:1409. [PMID: 31293578 PMCID: PMC6603153 DOI: 10.3389/fimmu.2019.01409] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 06/04/2019] [Indexed: 12/24/2022] Open
Abstract
The fundamental interaction between the immune and skeletal systems, termed as osteoimmunology, has been demonstrated to play indispensable roles in the maintenance of balance between bone resorption and formation. The pleiotropic sphingolipid metabolite, sphingosine 1-phosphate (S1P), together with its cognate receptor, sphingosine-1-phosphate receptor-1 (S1PR1), are known as key players in osteoimmunology due to the regulation on both immune system and bone remodeling. The role of S1P-S1PR1 signaling in bone remodeling can be directly targeting both osteoclastogenesis and osteogenesis. Meanwhile, inflammatory cell function and polarization in both adaptive immune (T cell subsets) and innate immune cells (macrophages) are also regulated by this signaling axis, suggesting that S1P-S1PR1 signaling could aslo indirectly regulate bone remodeling via modulating the immune system. Therefore, it could be likely that S1P-S1PR1 signaling might take part in the maintenance of continuous bone turnover under physiological conditions, while lead to the pathogenesis of bone deformities during inflammation. In this review, we summarized the immunological regulation of S1P-S1PR1 signal axis during bone remodeling with an emphasis on how osteo-immune regulators are affected by inflammation, an issue with relevance to chronical bone disorders such as rheumatoid arthritis, spondyloarthritis and periodontitis.
Collapse
Affiliation(s)
- Lan Xiao
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia.,The Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, QLD, Australia
| | - Yinghong Zhou
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia.,The Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, QLD, Australia.,Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Thor Friis
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Kenneth Beagley
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia.,The Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, QLD, Australia
| | - Yin Xiao
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia.,The Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, QLD, Australia.,Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
30
|
Peltier L, Bendavid C, Cavey T, Island ML, Doyard M, Leroyer P, Allain C, De Tayrac M, Ropert M, Loréal O, Guggenbuhl P. Iron excess upregulates SPNS2 mRNA levels but reduces sphingosine-1-phosphate export in human osteoblastic MG-63 cells. Osteoporos Int 2018; 29:1905-1915. [PMID: 29721575 DOI: 10.1007/s00198-018-4531-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 04/11/2018] [Indexed: 02/06/2023]
Abstract
UNLABELLED We aimed to study the mechanisms involved in bone-related iron impairment by using the osteoblast-like MG-63 cell line. Our results indicate that iron impact the S1P/S1PR signalizing axis and suggest that iron can affect the S1P process and favor the occurrence of osteoporosis during chronic iron overload. INTRODUCTION Systemic iron excess favors the development of osteoporosis, especially during genetic hemochromatosis. The cellular mechanisms involved are still unclear despite numerous data supporting a direct effect of iron on bone biology. Therefore, the aim of this study was to characterize mechanisms involved in the iron-related osteoblast impairment. METHODS We studied, by using the MG-63 cell lines, the effect of iron excess on SPNS2 gene expression which was previously identified by us as potentially iron-regulated. Cell-type specificity was investigated with hepatoma HepG2 and enterocyte-like Caco-2 cell lines as well as in iron-overloaded mouse liver. The SPNS2-associated function was also investigated in MG-63 cells by fluxomic strategy which led us to determinate the S1P efflux in iron excess condition. RESULTS We showed in MG-63 cells that iron exposure strongly increased the mRNA level of the SPNS2 gene. This was not observed in HepG2, in Caco-2 cells, and in mouse livers. Fluxomic study performed concomitantly on MG-63 cells revealed an unexpected decrease in the cellular capacity to export S1P. Iron excess did not modulate SPHK1, SPHK2, SGPL1, or SGPP1 gene expression, but decreased COL1A1 and S1PR1 mRNA levels, suggesting a functional implication of low extracellular S1P concentration on the S1P/S1PR signalizing axis. CONCLUSIONS Our results indicate that iron impacts the S1P/S1PR signalizing axis in the MG-63 cell line and suggest that iron can affect the bone-associated S1P pathway and favor the occurrence of osteoporosis during chronic iron overload.
Collapse
Affiliation(s)
- L Peltier
- Service de Biochimie - Toxicologie, CHU Rennes, F-35033, Rennes, France
- INSERM, INRA, Univ Rennes1, Univ Bretagne Loire, Nutrition, Metabolism, and Cancer, Rennes, France
- Faculté de Médecine, Université Rennes 1, F-35043, Rennes, France
| | - C Bendavid
- Service de Biochimie - Toxicologie, CHU Rennes, F-35033, Rennes, France
- INSERM, INRA, Univ Rennes1, Univ Bretagne Loire, Nutrition, Metabolism, and Cancer, Rennes, France
- Faculté de Médecine, Université Rennes 1, F-35043, Rennes, France
| | - T Cavey
- Service de Biochimie - Toxicologie, CHU Rennes, F-35033, Rennes, France
- INSERM, INRA, Univ Rennes1, Univ Bretagne Loire, Nutrition, Metabolism, and Cancer, Rennes, France
- Faculté de Médecine, Université Rennes 1, F-35043, Rennes, France
| | - M-L Island
- INSERM, INRA, Univ Rennes1, Univ Bretagne Loire, Nutrition, Metabolism, and Cancer, Rennes, France
| | - M Doyard
- INSERM, INRA, Univ Rennes1, Univ Bretagne Loire, Nutrition, Metabolism, and Cancer, Rennes, France
| | - P Leroyer
- INSERM, INRA, Univ Rennes1, Univ Bretagne Loire, Nutrition, Metabolism, and Cancer, Rennes, France
| | - C Allain
- INSERM, INRA, Univ Rennes1, Univ Bretagne Loire, Nutrition, Metabolism, and Cancer, Rennes, France
| | - M De Tayrac
- Faculté de Médecine, Université Rennes 1, F-35043, Rennes, France
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes (IGdR), F-35043, Rennes, France
- Service de Génétique Moléculaire et Génomique, CHU Rennes, F-35033, Rennes, France
| | - M Ropert
- Service de Biochimie - Toxicologie, CHU Rennes, F-35033, Rennes, France
- INSERM, INRA, Univ Rennes1, Univ Bretagne Loire, Nutrition, Metabolism, and Cancer, Rennes, France
| | - O Loréal
- INSERM, INRA, Univ Rennes1, Univ Bretagne Loire, Nutrition, Metabolism, and Cancer, Rennes, France
| | - P Guggenbuhl
- INSERM, INRA, Univ Rennes1, Univ Bretagne Loire, Nutrition, Metabolism, and Cancer, Rennes, France.
- Faculté de Médecine, Université Rennes 1, F-35043, Rennes, France.
- Service de Rhumatologie, CHU Rennes, F-35203, Rennes, France.
| |
Collapse
|
31
|
Hofrichter MAH, Mojarad M, Doll J, Grimm C, Eslahi A, Hosseini NS, Rajati M, Müller T, Dittrich M, Maroofian R, Haaf T, Vona B. The conserved p.Arg108 residue in S1PR2 (DFNB68) is fundamental for proper hearing: evidence from a consanguineous Iranian family. BMC MEDICAL GENETICS 2018; 19:81. [PMID: 29776397 PMCID: PMC5960148 DOI: 10.1186/s12881-018-0598-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/01/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND Genetic heterogeneity and consanguineous marriages make recessive inherited hearing loss in Iran the second most common genetic disorder. Only two reported pathogenic variants (c.323G>C, p.Arg108Pro and c.419A>G, p.Tyr140Cys) in the S1PR2 gene have previously been linked to autosomal recessive hearing loss (DFNB68) in two Pakistani families. We describe a segregating novel homozygous c.323G>A, p.Arg108Gln pathogenic variant in S1PR2 that was identified in four affected individuals from a consanguineous five generation Iranian family. METHODS Whole exome sequencing and bioinformatics analysis of 116 hearing loss-associated genes was performed in an affected individual from a five generation Iranian family. Segregation analysis and 3D protein modeling of the p.Arg108 exchange was performed. RESULTS The two Pakistani families previously identified with S1PR2 pathogenic variants presented profound hearing loss that is also observed in the affected Iranian individuals described in the current study. Interestingly, we confirmed mixed hearing loss in one affected individual. 3D protein modeling suggests that the p.Arg108 position plays a key role in ligand receptor interaction, which is disturbed by the p.Arg108Gln change. CONCLUSION In summary, we report the third overall mutation in S1PR2 and the first report outside the Pakistani population. Furthermore, we describe a novel variant that causes an amino acid exchange (p.Arg108Gln) in the same amino acid residue as one of the previously reported Pakistani families (p.Arg108Pro). This finding emphasizes the importance of the p.Arg108 amino acid in normal hearing and confirms and consolidates the role of S1PR2 in autosomal recessive hearing loss.
Collapse
Affiliation(s)
| | - Majid Mojarad
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Julia Doll
- Institute of Human Genetics, Julius Maximilians University, Würzburg, Germany
| | - Clemens Grimm
- Department of Biochemistry, Biocenter, Julius Maximilians University, Würzburg, Germany
| | - Atiye Eslahi
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Neda Sadat Hosseini
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Rajati
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine, Ghaem Educational Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Tobias Müller
- Institute of Bioinformatics, Julius Maximilians University, Würzburg, Germany
| | - Marcus Dittrich
- Institute of Human Genetics, Julius Maximilians University, Würzburg, Germany
- Institute of Bioinformatics, Julius Maximilians University, Würzburg, Germany
| | - Reza Maroofian
- Genetics and Molecular Cell Sciences Research Centre, St George’s, University of London, Cranmer Terrace, London, SW17 0RE UK
| | - Thomas Haaf
- Institute of Human Genetics, Julius Maximilians University, Würzburg, Germany
| | - Barbara Vona
- Institute of Human Genetics, Julius Maximilians University, Würzburg, Germany
| |
Collapse
|
32
|
Tantikanlayaporn D, Tourkova IL, Larrouture Q, Luo J, Piyachaturawat P, Witt MR, Blair HC, Robinson LJ. Sphingosine-1-Phosphate Modulates the Effect of Estrogen in Human Osteoblasts. JBMR Plus 2018; 2:217-226. [PMID: 30123862 PMCID: PMC6095197 DOI: 10.1002/jbm4.10037] [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] [Indexed: 01/22/2023] Open
Abstract
Production of sphingosine‐1‐phosphate (S1P) is linked to 17β‐estradiol (E2) activity in many estrogen‐responsive cells; in bone development, the role of S1P is unclear. We studied effects of S1P on proliferation and differentiation of human osteoblasts (hOB). Ten nM E2, 1 μM S1P, or 1 μM of the S1P receptor 1 (S1PR1) agonist SEW2871 increased hOB proliferation at 24 hours. S1PR 1, 2, and 3 mRNAs are expressed by hOB but not S1PR4 or S1PR5. Expression of S1PR2 was increased at 7 and 14 days of differentiation, in correspondence with osteoblast‐related mRNAs. Expression of S1PR1 was increased by E2 or S1P in proliferating hOB, whereas S1PR2 mRNA was unaffected in proliferating cells; S1PR3 was not affected by E2 or S1P. Inhibiting sphingosine kinase (SPHK) activity with sphingosine kinase inhibitor (Ski) greatly reduced the E2 proliferative effect. Both E2 and S1P increased SPHK mRNA at 24 hours in hOB. S1P promoted osteoblast proliferation via activating MAP kinase activity. Either E2 or S1P increased S1P synthesis in a fluorescent S1P assay. Interaction of E2 and S1P signaling was indicated by upregulation of E2 receptor mRNA after S1P treatment. E2 and S1P also promoted alkaline phosphatase expression. During osteoblast differentiation, S1P increased bone‐specific mRNAs, similarly to the effects of E2. However, E2 and S1P showed differences in the activation of some osteoblast pathways. Pathway analysis by gene expression arrays was consistent with regulation of pathways of osteoblast differentiation; collagen and cell adhesion proteins centered on Rho/Rac small GTPase signaling and Map kinase or signal transducer and activator of transcription (Stat) intermediates. Transcriptional activation also included significant increases in superoxide dismutase 1 and 2 transcription by either S1P or E2. We demonstrate that the SPHK system is a co‐mediator for osteoblast proliferation and differentiation, which is mainly, but not entirely, complementary to E2, whose effects are mediated by S1PR1 and S1PR2. © 2018 The Authors JBMR Plus is published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
| | - Irina L Tourkova
- Veterans Affairs Medical Center, Pittsburgh, PA, USA.,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Jianhua Luo
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Michelle R Witt
- Departments of Pathology and of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Harry C Blair
- Veterans Affairs Medical Center, Pittsburgh, PA, USA.,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lisa J Robinson
- Departments of Pathology and of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
33
|
Bougault C, El Jamal A, Briolay A, Mebarek S, Boutet MA, Garraud T, Le Goff B, Blanchard F, Magne D, Brizuela L. Involvement of sphingosine kinase/sphingosine 1-phosphate metabolic pathway in spondyloarthritis. Bone 2017; 103:150-158. [PMID: 28684192 DOI: 10.1016/j.bone.2017.07.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 06/23/2017] [Accepted: 07/01/2017] [Indexed: 12/13/2022]
Abstract
Spondyloarthritis (SpA) is a relatively common chronic inflammatory joint disorder, with a prevalence of about 0.2-0.5% worldwide. The primary target of the pathological process is the enthesis, where tendons and ligaments attach to underlying bone. These insertion sites are hotspots of bone formation (enthesophytes), which can lead to ankylosis. Unfortunately, the mechanisms causing the onset and progression of entheseal ossification remain largely unknown. Sphingosine 1-phosphate (S1P), a lipid generated after sphingosine phosphorylation by sphingosine kinases 1 and 2 (SK1/2), plays important roles in cell proliferation, differentiation and survival. S1P regulates fundamental biological processes such as cell cycle, inflammatory response or bone homeostasis. Indeed, S1P has been involved in some of most-spread skeletal diseases such as rheumatoid arthritis or osteoarthritis. On the other hand, the implication of S1P in SpA has not been explored yet. In the present work, we observed by ELISA that S1P content was significantly increased in the serum of SpA patients (6.1±4.2μM, n=21) compared to healthy donors (1.6±0.9μM, n=12). In vitro, gene expression of SK1 and SK2 as well as their activity were increased during differentiation of primary murine chondrocytes and osteoblasts into mineralizing cells. In addition, mRNA of the S1P-specific transporter Spns2 and S1P secretion were augmented. Using the pharmacological drugs SKi (SK pan-inhibitor), PF-543 (SK1 specific inhibitor) or K-145 (SK2 specific inhibitor), we showed that the inhibition of SK1 and/or SK2 decreased matrix mineralization, alkaline phosphatase activity and the mRNA expression of Runx2 and Bglap in chondrocytes and osteoblasts. To our knowledge, this is the first study indicating that S1P levels are significantly increased in serum from SpA patients. Moreover, we showed in vitro that SK activity was involved in the mineralization capacity of osteoblasts and chondrocytes. S1P metabolic pathway may represent an ingenious therapeutic target for SpA in the future.
Collapse
Affiliation(s)
- Carole Bougault
- Univ Lyon, Univ Claude Bernard Lyon 1, CNRS UMR 5246, ICBMS, F-69622 Lyon, France
| | - Alaeddine El Jamal
- Univ Lyon, Univ Claude Bernard Lyon 1, CNRS UMR 5246, ICBMS, F-69622 Lyon, France
| | - Anne Briolay
- Univ Lyon, Univ Claude Bernard Lyon 1, CNRS UMR 5246, ICBMS, F-69622 Lyon, France
| | - Saida Mebarek
- Univ Lyon, Univ Claude Bernard Lyon 1, CNRS UMR 5246, ICBMS, F-69622 Lyon, France
| | | | | | | | | | - David Magne
- Univ Lyon, Univ Claude Bernard Lyon 1, CNRS UMR 5246, ICBMS, F-69622 Lyon, France
| | - Leyre Brizuela
- Univ Lyon, Univ Claude Bernard Lyon 1, CNRS UMR 5246, ICBMS, F-69622 Lyon, France.
| |
Collapse
|
34
|
Sphingosine 1-phosphate (S1P) signalling: Role in bone biology and potential therapeutic target for bone repair. Pharmacol Res 2017; 125:232-245. [PMID: 28855094 DOI: 10.1016/j.phrs.2017.08.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 12/30/2022]
Abstract
The lipid mediator sphingosine 1-phosphate (S1P) affects cellular functions in most systems. Interest in its therapeutic potential has increased following the discovery of its G protein-coupled receptors and the recent availability of agents that can be safely administered in humans. Although the role of S1P in bone biology has been the focus of much less research than its role in the nervous, cardiovascular and immune systems, it is becoming clear that this lipid influences many of the functions, pathways and cell types that play a key role in bone maintenance and repair. Indeed, S1P is implicated in many osteogenesis-related processes including stem cell recruitment and subsequent differentiation, differentiation and survival of osteoblasts, and coupling of the latter cell type with osteoclasts. In addition, S1P's role in promoting angiogenesis is well-established. The pleiotropic effects of S1P on bone and blood vessels have significant potential therapeutic implications, as current therapeutic approaches for critical bone defects show significant limitations. Because of the complex effects of S1P on bone, the pharmacology of S1P-like agents and their physico-chemical properties, it is likely that therapeutic delivery of S1P agents will offer significant advantages compared to larger molecular weight factors. Hence, it is important to explore novel methods of utilizing S1P agents therapeutically, and improve our understanding of how S1P and its receptors modulate bone physiology and repair.
Collapse
|