1
|
Wang H, Qi LL, Shema C, Jiang KY, Ren P, Wang H, Wang L. Advances in the role and mechanism of fibroblasts in fracture healing. Front Endocrinol (Lausanne) 2024; 15:1350958. [PMID: 38469138 PMCID: PMC10925620 DOI: 10.3389/fendo.2024.1350958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/01/2024] [Indexed: 03/13/2024] Open
Abstract
With the development of social population ageing, bone fracture has become a global public health problem due to its high morbidity, disability and mortality. Fracture healing is a complex phenomenon involving the coordinated participation of immigration, differentiation and proliferation of inflammatory cells, angioblasts, fibroblasts, chondroblasts and osteoblasts which synthesize and release bioactive substances of extracellular matrix components, Mortality caused by age-related bone fractures or osteoporosis is steadily increasing worldwide as the population ages. Fibroblasts play an important role in the process of fracture healing. However, it is not clear how the growth factors and extracellular matrix stiffness of the bone-regeneration microenvironment affects the function of osteoblasts and fibroblasts in healing process. Therefore, this article focuses on the role of fibroblasts in the process of fracture healing and mechanisms of research progress.
Collapse
Affiliation(s)
- Hui Wang
- Department of Orthopedics, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Li-li Qi
- Experimental Center for Teaching of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Clement Shema
- Department of Orthopedic Research Center, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- International Education College of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Kui-ying Jiang
- National Demonstration Center for Experimental Basic Medical Education, Capital Medical University, Beijing, China
| | - Ping Ren
- Experimental Center for Teaching of Hebei Medical University, Shijiazhuang, Hebei, China
| | - He Wang
- Department of Pathogenic Biology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lei Wang
- Department of Human Anatomy, Institute of Medicine and Health, Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, Hebei, China
| |
Collapse
|
2
|
Chen Y, Mehmood K, Chang YF, Tang Z, Li Y, Zhang H. The molecular mechanisms of glycosaminoglycan biosynthesis regulating chondrogenesis and endochondral ossification. Life Sci 2023; 335:122243. [PMID: 37949211 DOI: 10.1016/j.lfs.2023.122243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/23/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023]
Abstract
Disorders of chondrocyte differentiation and endochondral osteogenesis are major underlying factors in skeletal developmental disorders, including tibial dysplasia (TD), osteoarthritis (OA), chondrodysplasia (ACH), and multiple epiphyseal dysplasia (MED). Understanding the cellular and molecular pathogenesis of these disorders is crucial for addressing orthopedic diseases resulting from impaired glycosaminoglycan synthesis. Glycosaminoglycan is a broad term that refers to the glycan component of proteoglycan macromolecules. It is an essential component of the cartilage extracellular matrix and plays a vital role in various biological processes, including gene transcription, signal transduction, and chondrocyte differentiation. Recent studies have demonstrated that glycosaminoglycan biosynthesis plays a regulatory role in chondrocyte differentiation and endochondral osteogenesis by modulating various growth factors and signaling molecules. For instance, glycosaminoglycan is involved in mediating pathways such as Wnt, TGF-β, FGF, Ihh-PTHrP, and O-GlcNAc glycosylation, interacting with transcription factors SOX9, BMPs, TGF-β, and Runx2 to regulate chondrocyte differentiation and endochondral osteogenesis. To propose innovative approaches for addressing orthopedic diseases caused by impaired glycosaminoglycan biosynthesis, we conducted a comprehensive review of the molecular mechanisms underlying chondrocyte glycosaminoglycan biosynthesis, which regulates chondrocyte differentiation and endochondral osteogenesis. Our analysis considers the role of genes, glycoproteins, and associated signaling pathways during chondrogenesis and endochondral ossification.
Collapse
Affiliation(s)
- Yongjian Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Khalid Mehmood
- Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, 63100, Pakistan
| | - Yung-Fu Chang
- College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Hui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
3
|
Zhou Y, Zhu P, Shen S, Wang Y, Li B, Guo B, Li H. Overexpression of fibroblast growth factor receptor 2 in bone marrow mesenchymal stem cells enhances osteogenesis and promotes critical cranial bone defect regeneration. Front Cell Dev Biol 2023; 11:1208239. [PMID: 37266455 PMCID: PMC10229770 DOI: 10.3389/fcell.2023.1208239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 05/09/2023] [Indexed: 06/03/2023] Open
Abstract
Background: Reconstruction of cranial bone defects is one of the most challenging problems in reconstructive surgery, and several biological tissue engineering methods have been used to promote bone repair, such as genetic engineering of bone marrow mesenchymal stem cells (BMSCs). Fibroblast growth factor receptor 2 (Fgfr2) is an important regulator of bone construction and can be used as a potential gene editing site. However, its role in the osteogenesis process of BMSCs remains unclear. This article clarifies the function of Fgfr2 in BMSCs and explores the role of Fgfr2-overexpressed BMSCs carried by light-induced porous hydrogel (GelMA) in the repair of cranial bone defects. Methods: Lenti-virus was used to overexpress Fgfr2 in BMSCs, and cell counting kit-8, transwell, and flow cytometry assays were conducted to investigate the proliferation, migration, and characteristics. After 0, 3, 7, and 10 days of osteogenic or chondrogenic induction, the changes in osteogenic and chondrogenic ability were detected by real-time PCR, western blot, alkaline phosphatase staining, alizarin Red staining, and alcian blue staining. To investigate the viability of BMSCs carried by GelMA, calcein and propyl iodide staining were carried out as well. Finally, a critical cranial bone defect model was established in 6-week-old male mice and micro-computerized tomography, masson staining, and immunohistochemistry of OCN were conducted to test the bone regeneration properties of implanting Fgfr2-overexpressed BMSCs with GelMA in cranial bone defects over 6 weeks. Results: Overexpression of Fgfr2 in BMSCs significantly promoted cell proliferation and migration and increased the percentage of CD200+CD105+ cells. After osteogenic and chondrogenic induction, Fgfr2 overexpression enhanced both osteogenic and chondrogenic ability. Furthermore, in cranial bone defect regeneration, BMSCs carried by light-induced GelMA showed favorable biocompatibility, and Fgfr2-overexpressed BMSCs induced superior cranial bone regeneration compared to a normal BMSCs group and an untreated blank group. Conclusion: In vitro, Fgfr2 enhanced the proliferation, migration, and stemness of BMSCs and promoted osteogenesis and chondrogenesis after parallel induction. In vivo, BMSCs with Fgfr2 overexpression carried by GelMA showed favorable performance in treating critical cranial bone defects. This study clarifies the multiple functions of Fgfr2 in BMSCs and provides a new method for future tissue engineering.
Collapse
Affiliation(s)
- Yiwen Zhou
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
- Medical School of Nanjing University, Nanjing, China
| | - Peixiang Zhu
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
- Medical School of Nanjing University, Nanjing, China
| | - Siyu Shen
- Medical School of Nanjing University, Nanjing, China
| | - Yanyi Wang
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
- Medical School of Nanjing University, Nanjing, China
| | - Baochao Li
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
- Medical School of Nanjing University, Nanjing, China
| | - Baosheng Guo
- Medical School of Nanjing University, Nanjing, China
| | - Huang Li
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
- Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
4
|
Song K, Yang GM, Han J, Gil M, Dayem AA, Kim K, Lim KM, Kang GH, Kim S, Jang SB, Vellingiri B, Cho SG. Modulation of Osteogenic Differentiation of Adipose-Derived Stromal Cells by Co-Treatment with 3, 4'-Dihydroxyflavone, U0126, and N-Acetyl Cysteine. Int J Stem Cells 2022; 15:334-345. [PMID: 35769058 PMCID: PMC9396012 DOI: 10.15283/ijsc22044] [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: 03/18/2022] [Revised: 04/20/2022] [Accepted: 04/20/2022] [Indexed: 11/30/2022] Open
Abstract
Background and Objectives Flavonoids form the largest group of plant phenols and have various biological and pharmacological activities. In this study, we investigated the effect of a flavonoid, 3, 4’-dihydroxyflavone (3, 4’-DHF) on osteogenic differentiation of equine adipose-derived stromal cells (eADSCs). Methods and Results Treatment of 3, 4’-DHF led to increased osteogenic differentiation of eADSCs by increasing phosphorylation of ERK and modulating Reactive Oxygen Species (ROS) generation. Although PD98059, an ERK inhibitor, suppressed osteogenic differentiation, another ERK inhibitor, U0126, apparently increased osteogenic differentiation of the 3, 4’-DHF-treated eADSCs, which may indicate that the effect of U0126 on bone morphogenetic protein signaling is involved in the regulation of 3, 4’-DHF in osteogenic differentiation of eADSCs. We revealed that 3, 4’-DHF could induce osteogenic differentiation of eADSCs by suppressing ROS generation and co-treatment of 3, 4’-DHF, U0126, and/or N-acetyl cysteine (NAC) resulted in the additive enhancement of osteogenic differentiation of eADSCs. Conclusions Our results showed that co-treatment of 3, 4’-DHF, U0126, and/or NAC cumulatively regulated osteogenesis in eADSCs, suggesting that 3, 4’-DHF, a flavonoid, can provide a novel approach to the treatment of osteoporosis and can provide potential therapeutic applications in therapeutics and regenerative medicine for human and companion animals.
Collapse
Affiliation(s)
- Kwonwoo Song
- Department of Stem Cell and Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul, Korea
| | - Gwang-Mo Yang
- Department of Stem Cell and Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul, Korea
| | - Jihae Han
- Department of Stem Cell and Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul, Korea
| | - Minchan Gil
- Department of Stem Cell and Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul, Korea
| | - Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul, Korea
| | - Kyeongseok Kim
- Department of Stem Cell and Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul, Korea
| | - Kyung Min Lim
- Department of Stem Cell and Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul, Korea
| | - Geun-Ho Kang
- Department of Stem Cell and Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul, Korea
| | - Sejong Kim
- Department of Stem Cell and Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul, Korea
| | - Soo Bin Jang
- Department of Stem Cell and Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul, Korea
| | - Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, India
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul, Korea
| |
Collapse
|
5
|
Regulation of biomineralization by proteoglycans: From mechanisms to application. Carbohydr Polym 2022; 294:119773. [DOI: 10.1016/j.carbpol.2022.119773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/20/2022] [Accepted: 06/20/2022] [Indexed: 11/17/2022]
|
6
|
Cho S, Lee H, Lee HY, Kim SJ, Song W. The effect of fibroblast growth factor receptor inhibition on resistance exercise training-induced adaptation of bone and muscle quality in mice. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY 2022; 26:207-218. [PMID: 35477548 PMCID: PMC9046891 DOI: 10.4196/kjpp.2022.26.3.207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/19/2022] [Accepted: 03/24/2022] [Indexed: 11/15/2022]
Affiliation(s)
- Suhan Cho
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hojun Lee
- Department of Sport and Exercise Science, Seoul Women's University, Seoul 01797, Korea
| | - Ho-Young Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea
| | - Sung Joon Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Wook Song
- Health and Exercise Science Laboratory, Institute of Sport Science, Seoul 08826, Korea
- Department of Physical Education, Seoul National University, Seoul 08826, Korea
- Institute on Aging, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
7
|
Usategui-Martín R, Rigual R, Ruiz-Mambrilla M, Fernández-Gómez JM, Dueñas A, Pérez-Castrillón JL. Molecular Mechanisms Involved in Hypoxia-Induced Alterations in Bone Remodeling. Int J Mol Sci 2022; 23:ijms23063233. [PMID: 35328654 PMCID: PMC8953213 DOI: 10.3390/ijms23063233] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 12/31/2022] Open
Abstract
Bone is crucial for the support of muscles and the protection of vital organs, and as a reservoir of calcium and phosphorus. Bone is one of the most metabolically active tissues and is continuously renewed to adapt to the changes required for healthy functioning. To maintain normal cellular and physiological bone functions sufficient oxygen is required, as evidence has shown that hypoxia may influence bone health. In this scenario, this review aimed to analyze the molecular mechanisms involved in hypoxia-induced bone remodeling alterations and their possible clinical consequences. Hypoxia has been associated with reduced bone formation and reduced osteoblast matrix mineralization due to the hypoxia environment inhibiting osteoblast differentiation. A hypoxic environment is involved with increased osteoclastogenesis and increased bone resorptive capacity of the osteoclasts. Clinical studies, although with contradictory results, have shown that hypoxia can modify bone remodeling.
Collapse
Affiliation(s)
- Ricardo Usategui-Martín
- Department of Cell Biology, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, 47003 Valladolid, Spain;
- IOBA, University of Valladolid, 47011 Valladolid, Spain
- Correspondence: (R.U.-M.); (J.L.P.-C.)
| | - Ricardo Rigual
- Department of Biochemistry, Molecular Biology and Physiology, Faculty of Medicine, University of Valladolid, 47003 Valladolid, Spain;
- IBGM, University of Valladolid, 47003 Valladolid, Spain
| | - Marta Ruiz-Mambrilla
- Department of Surgery, Faculty of Medicine, University of Valladolid, 47003 Valladolid, Spain;
| | - José-María Fernández-Gómez
- Department of Cell Biology, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, 47003 Valladolid, Spain;
| | - Antonio Dueñas
- Department of Medicine, Faculty of Medicine, University of Valladolid, 47003 Valladolid, Spain;
- Department of Toxicology, Río Hortega University Hospital, 47012 Valladolid, Spain
| | - José Luis Pérez-Castrillón
- Department of Medicine, Faculty of Medicine, University of Valladolid, 47003 Valladolid, Spain;
- Department of Internal Medicine, Río Hortega University Hospital, 47012 Valladolid, Spain
- Correspondence: (R.U.-M.); (J.L.P.-C.)
| |
Collapse
|
8
|
Chang MC, Chen NY, Chen JH, Huang WL, Chen CY, Huang CC, Pan YH, Chang HH, Jeng JH. bFGF stimulated plasminogen activation factors, but inhibited alkaline phosphatase and SPARC in stem cells from apical Papilla: Involvement of MEK/ERK, TAK1 and p38 signaling. J Adv Res 2021; 40:95-107. [PMID: 36100336 PMCID: PMC9481946 DOI: 10.1016/j.jare.2021.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/04/2021] [Accepted: 12/14/2021] [Indexed: 11/25/2022] Open
Abstract
bFGF induced uPA, uPAR, PAI-1 production/expression in SCAP → bFGF induced decline of ALP and SPARC of SCAP → The effects of bFGF are regulated by ERK, p38, TAK1 and Akt signaling → Crucial for SCAP proliferation, matrix turnover and differentiation → These events are important for revascularization/root apexogenesis
Introduction Objectives Methods Results Conclusion
Collapse
|
9
|
Chen X, Wang Y, Chen R, Qu N, Zhang B, Xia C. Suppressing PLCγ1 enhances osteogenic and chondrogenic potential of BMSCs. Biochem Biophys Res Commun 2020; 532:292-299. [PMID: 32868075 DOI: 10.1016/j.bbrc.2020.08.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/14/2020] [Indexed: 11/28/2022]
Abstract
Phosphatidylcholine-specific phospholipase Cγ1 (PLCγ1) is involved in regulating cell metabolism. However, little is known how PLCγ1 directs BMSC differentiation. Here, we investigated the role of PLCγ1 in rat BMSC differentiation into osteoblasts and chondrocytes. The results of Alizarin red and Alcian blue staining showed that PLCγ1 inhibitor U73122 significantly enhanced the mineralization capacity and proteoglycan deposition of BMSCs. The results of qPCR technique and Western blot analysis showed that long-term treatment of U73122 enhanced COL1A1 and OPG mRNA levels and Collagen 1A1, BMP2, and p-Smad1/5/9 protein levels and that short-term treatment of U73122 enhanced COL2A1 and SOX9 mRNA levels and Collagen 2, SOX9, Aggrecan, TGF-β3, and p-Smad2/3 protein levels. Decreased p-mTOR and p-P38 contributed to enhanced osteogenic potentials of BMSCs and increased p-P38 contributed to enhanced chondrogenic potentials of BMSCs. The scaffold transplantation with U73122+BMSC was more efficacious than BMSC alone for osteochondral defect repair in a rat model. Therefore, suppressing PLCγ1 could improve the capacity to effectively use BMSCs for cell therapy of osteochondral defect.
Collapse
Affiliation(s)
- Xiaolei Chen
- Bone & Joint Research Institute, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, 361004, China
| | - Yue Wang
- Bone & Joint Research Institute, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, 361004, China
| | - Ri Chen
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Ning Qu
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Bing Zhang
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Chun Xia
- Bone & Joint Research Institute, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, 361004, China.
| |
Collapse
|
10
|
Hefka Blahnova V, Dankova J, Rampichova M, Filova E. Combinations of growth factors for human mesenchymal stem cell proliferation and osteogenic differentiation. Bone Joint Res 2020; 9:412-420. [PMID: 32864112 PMCID: PMC7437520 DOI: 10.1302/2046-3758.97.bjr-2019-0183.r2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aims Here we introduce a wide and complex study comparing effects of growth factors used alone and in combinations on human mesenchymal stem cell (hMSC) proliferation and osteogenic differentiation. Certain ways of cell behaviour can be triggered by specific peptides – growth factors, influencing cell fate through surface cellular receptors. Methods In our study transforming growth factor β (TGF-β), basic fibroblast growth factor (bFGF), hepatocyte growth factor (HGF), insulin-like growth factor 1 (IGF-1), and vascular endothelial growth factor (VEGF) were used in order to induce osteogenesis and proliferation of hMSCs from bone marrow. These cells are naturally able to differentiate into various mesodermal cell lines. Effect of each factor itself is pretty well known. We designed experimental groups where two and more growth factors were combined. We supposed cumulative effect would appear when more growth factors with the same effect were combined. The cellular metabolism was evaluated using MTS assay and double-stranded DNA (dsDNA) amount using PicoGreen assay. Alkaline phosphatase (ALP) activity, as early osteogenesis marker, was observed. Phase contrast microscopy was used for cell morphology evaluation. Results TGF-β and bFGF were shown to significantly enhance cell proliferation. VEGF and IGF-1 supported ALP activity. Light microscopy showed initial extracellular matrix mineralization after VEGF/IGF-1 supply. Conclusion A combination of more than two growth factors did not support the cellular metabolism level and ALP activity even though the growth factor itself had a positive effect. This is probably caused by interplay of various messengers shared by more growth factor signalling cascades. Cite this article: Bone Joint Res 2020;9(7):412–420.
Collapse
Affiliation(s)
- Veronika Hefka Blahnova
- Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic.,Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jana Dankova
- Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Michala Rampichova
- Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Eva Filova
- Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic.,Second Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
11
|
Vital Pulp Therapy an Insight Over the Available Literature and Future Expectations. Eur Endod J 2020; 5:46-53. [PMID: 32342038 PMCID: PMC7183799 DOI: 10.14744/eej.2019.44154] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 09/29/2019] [Indexed: 12/16/2022] Open
Abstract
Vital pulp therapy (VPT) defined as “treatment which aims at preserving and maintaining the pulp tissue that has been compromised but not destroyed by extensive dental caries, dental trauma, and restorative procedures or for iatrogenic reasons”, offers some beneficial advantages over the conventional root canal treatment such as protective resistance for mastication forces or to prevent the loss of environmental changes sensation ability, which can lead to unnoticeable progression of caries and later fracture. A wide range of materials are suggested in the literature to be used as pulp capping protective dressing materials that varies from ready-made synthetic materials to biological based scaffolds and composites. The aim of the present review is to provide a full understanding of currently used materials to clinicians in order to help in their decision-making process delivering the best available evidence-based treatments to their patients. An extensive search for recent available data regarding direct pulp capping materials and potential suggestions for future use have been made. Newly developed biological based scaffolds showed promising results in dentine regeneration therefore strengthening the tooth structure and overcoming potential drawbacks of use of currently available recommended materials.
Collapse
|
12
|
Chang MC, Chen CY, Chang YC, Zhong BH, Wang YL, Yeung SY, Chang HH, Jeng JH. Effect of bFGF on the growth and matrix turnover of stem cells from human apical papilla: Role of MEK/ERK signaling. J Formos Med Assoc 2020; 119:1666-1672. [PMID: 31932202 DOI: 10.1016/j.jfma.2019.12.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/11/2019] [Accepted: 12/23/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND/PURPOSE Basic fibroblast growth factor (bFGF) exhibits multiple biological functions in various tissues. Stem cells from apical papilla (SCAP) can be isolated from human apical papilla tissues in developmental teeth of children. The purposes of this study were to investigate the expression of FGF receptors (FGFRs) and the effects of bFGF on SCAP and related MEK/ERK signaling. METHODS SCAP cells were treated under different concentrations of bFGF with or without U0126 (an inhibitor of MEK/ERK). Expression of FGFR1 and FGFR2 in SCAP was analyzed by RT-PCR. Cell proliferation was measured by MTT assay. The expressions of type I collagen, cdc 2, cyclin B1, TIMP-1 and p-ERK proteins were examined by Western blot. RESULTS SCAP cells expressed FGFR1 and FGFR2. Exposure of SCAP to bFGF enhanced cell proliferation, and the expression cyclinB1, cdc 2, and TIMP-1, but not type I collagen. U0126 pretreatment and co-incubation attenuated the bFGF-induced proliferation, cdc2, cyclin B1 and TIMP-1 proteins' expression, but not type I collagen in SCAP. CONCLUSION SCAP cells express FGFRs. bFGF may stimulate proliferation and affect the matrix turnover of SCAP cells, possibly via stimulation of FGFRs and MEK/ERK signaling pathway. These results are useful for clinical therapies for apexogenesis and regeneration of pulpo-dentin complex.
Collapse
Affiliation(s)
- Mei-Chi Chang
- Biomedical Science Team, Chang Gung University of Science and Technology, Taoyuan, Taiwan; Department of Dentistry, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Chih-Yu Chen
- School of Dentistry, National Taiwan University Medical College, Taipei, Taiwan; Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan
| | - Ya-Ching Chang
- Department of Dentistry, MacKay Memorial Hospital, Taipei, Taiwan
| | - Bo-Hao Zhong
- School of Dentistry, National Taiwan University Medical College, Taipei, Taiwan; Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan
| | - Yin-Lin Wang
- School of Dentistry, National Taiwan University Medical College, Taipei, Taiwan; Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan
| | - Sin-Yuet Yeung
- Department of Dentistry, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Hsiao-Hua Chang
- School of Dentistry, National Taiwan University Medical College, Taipei, Taiwan; Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan.
| | - Jiiang-Huei Jeng
- School of Dentistry, National Taiwan University Medical College, Taipei, Taiwan; Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
13
|
Zhao SJ, Kong FQ, Jie J, Li Q, Liu H, Xu AD, Yang YQ, Jiang B, Wang DD, Zhou ZQ, Tang PY, Chen J, Wang Q, Zhou Z, Chen Q, Yin GY, Zhang HW, Fan J. Macrophage MSR1 promotes BMSC osteogenic differentiation and M2-like polarization by activating PI3K/AKT/GSK3β/β-catenin pathway. Theranostics 2020; 10:17-35. [PMID: 31903103 PMCID: PMC6929615 DOI: 10.7150/thno.36930] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 09/21/2019] [Indexed: 12/18/2022] Open
Abstract
Approximately 10% of bone fractures do not heal satisfactorily, leading to significant clinical and socioeconomic implications. Recently, the role of macrophages in regulating bone marrow stem cell (BMSC) differentiation through the osteogenic pathway during fracture healing has attracted much attention. Methods: The tibial monocortical defect model was employed to determine the critical role of macrophage scavenger receptor 1 (MSR1) during intramembranous ossification (IO) in vivo. The potential functions and mechanisms of MSR1 were explored in a co-culture system of bone marrow-derived macrophages (BMDMs), RAW264.7 cells, and BMSCs using qPCR, Western blotting, immunofluorescence, and RNA sequencing. Results: In this study, using the tibial monocortical defect model, we observed delayed IO in MSR1 knockout (KO) mice compared to MSR1 wild-type (WT) mice. Furthermore, macrophage MSR1 mediated PI3K/AKT/GSK3β/β-catenin signaling increased ability to promote osteogenic differentiation of BMSCs in the co-culture system. We also identified proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) as the target gene for macrophage MSR1-activated PI3K/AKT/GSK3β/β-catenin pathway in the co-culture system that facilitated M2-like polarization by enhancing mitochondrial oxidative phosphorylation. Conclusion: Our findings revealed a previously unrecognized function of MSR1 in macrophages during fracture repair. Targeting MSR1 might, therefore, be a new therapeutic strategy for fracture repair.
Collapse
|
14
|
Kuroda Y, Kawai T, Goto K, Matsuda S. Clinical application of injectable growth factor for bone regeneration: a systematic review. Inflamm Regen 2019; 39:20. [PMID: 31660090 PMCID: PMC6805537 DOI: 10.1186/s41232-019-0109-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 09/25/2019] [Indexed: 12/04/2022] Open
Abstract
Bone regeneration has been the ultimate goal in the field of bone and joint medicine and has been evaluated through various basic research studies to date. Translational research of regenerative medicine has focused on three primary approaches, which are expected to increase in popularity: cell therapy, proteins, and artificial materials. Among these, the local injection of a gelatin hydrogel impregnated with the protein fibroblast growth factor (FGF)-2 is a biomaterial technique that has been developed in Japan. We have previously reported the efficacy of gelatin hydrogel containing injectable FGF-2 for the regenerative treatment of osteonecrosis of the femoral head. Injectable growth factors will probably be developed in the future and gain popularity as a medical approach in various fields as well as orthopedics. Several clinical trials have already been conducted and have focused on this technique, reporting its efficacy and safety. To date, reports of the clinical application of FGF-2 in revascularization for critical limb ischemia, treatment of periodontal disease, early bone union for lower limb fracture and knee osteotomy, and bone regeneration for osteonecrosis of the femoral head have been based on basic research conducted in Japan. In the present report, we present an extensive review of clinical applications using injectable growth factors and discuss the associated efficacy and safety of their administration.
Collapse
Affiliation(s)
- Yutaka Kuroda
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Shogoin, Kawahara-cho 54, Sakyo-ku, Kyoto, 606-8507 Japan
| | - Toshiyuki Kawai
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Shogoin, Kawahara-cho 54, Sakyo-ku, Kyoto, 606-8507 Japan
| | - Koji Goto
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Shogoin, Kawahara-cho 54, Sakyo-ku, Kyoto, 606-8507 Japan
| | - Shuichi Matsuda
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Shogoin, Kawahara-cho 54, Sakyo-ku, Kyoto, 606-8507 Japan
| |
Collapse
|
15
|
Gebuijs IGE, Raterman ST, Metz JR, Swanenberg L, Zethof J, Van den Bos R, Carels CEL, Wagener FADTG, Von den Hoff JW. Fgf8a mutation affects craniofacial development and skeletal gene expression in zebrafish larvae. Biol Open 2019; 8:bio.039834. [PMID: 31471293 PMCID: PMC6777363 DOI: 10.1242/bio.039834] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Craniofacial development is tightly regulated and therefore highly vulnerable to disturbance by genetic and environmental factors. Fibroblast growth factors (FGFs) direct migration, proliferation and survival of cranial neural crest cells (CNCCs) forming the human face. In this study, we analyzed bone and cartilage formation in the head of five dpf fgf8ati282 zebrafish larvae and assessed gene expression levels for 11 genes involved in these processes. In addition, in situ hybridization was performed on 8 and 24 hours post fertilization (hpf) larvae (fgf8a, dlx2a, runx2a, col2a1a). A significant size reduction of eight out of nine craniofacial cartilage structures was found in homozygous mutant (6–36%, P<0.01) and heterozygous (7–24%, P<0.01) larvae. Also, nine mineralized structures were not observed in all or part of the homozygous (0–71%, P<0.0001) and heterozygous (33–100%, P<0.0001) larvae. In homozygote mutants, runx2a and sp7 expression was upregulated compared to wild type, presumably to compensate for the reduced bone formation. Decreased col9a1b expression may compromise cartilage formation. Upregulated dlx2a in homozygotes indicates impaired CNCC function. Dlx2a expression was reduced in the first and second stream of CNCCs in homozygous mutants at 24 hpf, as shown by in situ hybridization. This indicates an impairment of CNCC migration and survival by fgf8 mutation. Summary: A function-blocking mutation in fgf8a causes craniofacial malformations in zebrafish larvae due to impaired cranial neural crest cell migration and survival.
Collapse
Affiliation(s)
- I G E Gebuijs
- Department of Orthodontics and Craniofacial Biology, Radboudumc, Nijmegen, The Netherlands.,Department of Orthodontics and Craniofacial Biology, Radboud Institute of Molecular Life Sciences, Nijmegen, The Netherlands.,Department of Animal Ecology and Physiology, Radboud University, Nijmegen, The Netherlands
| | - S T Raterman
- Department of Orthodontics and Craniofacial Biology, Radboudumc, Nijmegen, The Netherlands.,Department of Orthodontics and Craniofacial Biology, Radboud Institute of Molecular Life Sciences, Nijmegen, The Netherlands.,Department of Animal Ecology and Physiology, Radboud University, Nijmegen, The Netherlands
| | - J R Metz
- Department of Animal Ecology and Physiology, Radboud University, Nijmegen, The Netherlands
| | - L Swanenberg
- Department of Orthodontics and Craniofacial Biology, Radboudumc, Nijmegen, The Netherlands.,Department of Animal Ecology and Physiology, Radboud University, Nijmegen, The Netherlands
| | - J Zethof
- Department of Animal Ecology and Physiology, Radboud University, Nijmegen, The Netherlands
| | - R Van den Bos
- Department of Animal Ecology and Physiology, Radboud University, Nijmegen, The Netherlands
| | - C E L Carels
- Department of Orthodontics and Craniofacial Biology, Radboud Institute of Molecular Life Sciences, Nijmegen, The Netherlands.,Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands.,Department of Oral Health Sciences and Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - F A D T G Wagener
- Department of Orthodontics and Craniofacial Biology, Radboudumc, Nijmegen, The Netherlands.,Department of Orthodontics and Craniofacial Biology, Radboud Institute of Molecular Life Sciences, Nijmegen, The Netherlands
| | - J W Von den Hoff
- Department of Orthodontics and Craniofacial Biology, Radboudumc, Nijmegen, The Netherlands .,Department of Orthodontics and Craniofacial Biology, Radboud Institute of Molecular Life Sciences, Nijmegen, The Netherlands
| |
Collapse
|
16
|
Review of the Pathways Involved in the Osteogenic Differentiation of Adipose-Derived Stem Cells. J Craniofac Surg 2019; 30:703-708. [PMID: 30839467 DOI: 10.1097/scs.0000000000005447] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Grafts and prosthetic materials used for the repair of bone defects are often accompanied by comorbidity and rejection. Therefore, there is an immense need for novel approaches to combating the issues surrounding such defects. Because of their accessibility, substantial proportion, and osteogenic differentiation potential, adipose-derived stem cells (ASCs) make for an ideal source of bone tissue in regenerative medicine. However, efficient induction of ASCs toward an osteoblastic lineage in vivo is met with challenges, and many signaling pathways must come together to secure osteoblastogenesis. Among them are bone morphogenic protein, wingless-related integration site protein, Notch, Hedgehog, fibroblast growth factor, vascular endothelial growth factor, and extracellular regulated-signal kinase. The goal of this literature review is to conglomerate the present research on these pathways to formulate a better understanding of how ASCs are most effectively transformed into bone in the context of tissue engineering.
Collapse
|
17
|
Cheng CH, Chen YW, Kai-Xing Lee A, Yao CH, Shie MY. Development of mussel-inspired 3D-printed poly (lactic acid) scaffold grafted with bone morphogenetic protein-2 for stimulating osteogenesis. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2019; 30:78. [PMID: 31222566 DOI: 10.1007/s10856-019-6279-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/07/2019] [Indexed: 06/09/2023]
Abstract
3D printing is a versatile technique widely applied in tissue engineering due to its ability to manufacture large quantities of scaffolds or constructs with various desired architectures. In this study, we demonstrated that poly (lactic acid) (PLA) scaffolds fabricated via fused deposition not only retained the original interconnected microporous architectures, the scaffolds also exhibited lower lactic acid dissolution as compared to the freeze-PLA scaffold. The 3D-printed scaffolds were then grafted with human bone morphogenetic protein-2 (BMP-2) via the actions of polydopamine (PDA) coatings. The loading and release rate of BMP-2 were monitored for a period of 35 days. Cellular behaviors and osteogenic activities of co-cultured human mesenchymal stem cells (hMSCs) were assessed to determine for efficacies of scaffolds. In addition, we demonstrated that our fabricated scaffolds were homogenously coated with PDA and well grafted with BMP-2 (219.1 ± 20.4 ng) when treated with 250 ng/mL of BMP-2 and 741.4 ± 127.3 ng when treated with 1000 ng/mL of BMP-2. This grafting enables BMP-2 to be released in a sustained profile. From the osteogenic assay, it was shown that the ALP activity and osteocalcin of hMSCs cultured on BMP-2/PDA/PLA were significantly higher when compared with PLA and PDA/PLA scaffolds. The methodology of PDA coating employed in this study can be used as a simple model to immobilize multiple growth factors onto different 3D-printed scaffold substrates. Therefore, there is potential for generation of scaffolds with different unique modifications with different capabilities in regulating physiochemical and biological properties for future applications in bone tissue engineering.
Collapse
Affiliation(s)
- Cheng-Hsin Cheng
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Department of Neurosurgery, Tainan Municipal An-Nan Hospital-China Medical University, Tainan, Taiwan
- Department of Neurosurgery, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Yi-Wen Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- 3D Printing Medical Research Institute, Asia University, Taichung, Taiwan
| | - Alvin Kai-Xing Lee
- 3D Printing Medical Research Center, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Chun-Hsu Yao
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan.
- School of Chinese Medicine, China Medical University, Taichung, Taiwan.
- Biomaterials Translational Research Center, China Medical University Hospital, Taichung, Taiwan.
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan.
| | - Ming-You Shie
- 3D Printing Medical Research Center, China Medical University Hospital, Taichung, Taiwan.
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan.
- School of Dentistry, China Medical University, Taichung, Taiwan.
| |
Collapse
|
18
|
Ji L, Song Z, Zeng F, Hu M, Chen S, Qin Z, Xia D. [Research progress on controlled release of various growth factors in bone regeneration]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2019; 33:750-755. [PMID: 31198005 PMCID: PMC8355764 DOI: 10.7507/1002-1892.201901116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/28/2019] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To summarize the research progress of controlled release of angiogenic factors and osteogenic factors in bone tissue engineering. METHODS The domestic and abroad literature on the controlled release structure of growth factors during bone regeneration in recent years was extensively reviewed and summarized. RESULTS The sustained-release structure includes direct binding, microsphere-three-dimensional scaffold structure, core-shell structure, layer self-assembly, hydrogel, and gene carrier. A sustained-release system composed of different sustained-release structures combined with different growth factors can promote bone regeneration and angiogenesis. CONCLUSION Due to its controllability and persistence, the growth factor sustained-release system has become a research hotspot in bone tissue engineering and has broad application prospects.
Collapse
Affiliation(s)
- Lin Ji
- Department of Burn and Plastic Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Ziwei Song
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Fuhai Zeng
- Department of Burn and Plastic Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Ming Hu
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Siqi Chen
- Department of Burn and Plastic Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Zhongjie Qin
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Delin Xia
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000,
| |
Collapse
|
19
|
Abstract
Fibroblast growth factors (FGFs) and their receptors (FGFRs) are expressed throughout all stages of skeletal development. In the limb bud and in cranial mesenchyme, FGF signaling is important for formation of mesenchymal condensations that give rise to bone. Once skeletal elements are initiated and patterned, FGFs regulate both endochondral and intramembranous ossification programs. In this chapter, we review functions of the FGF signaling pathway during these critical stages of skeletogenesis, and explore skeletal malformations in humans that are caused by mutations in FGF signaling molecules.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States.
| | - Pierre J Marie
- UMR-1132 Inserm (Institut national de la Santé et de la Recherche Médicale) and University Paris Diderot, Sorbonne Paris Cité, Hôpital Lariboisière, Paris, France
| |
Collapse
|
20
|
Somm E, Henry H, Bruce SJ, Bonnet N, Montandon SA, Niederländer NJ, Messina A, Aeby S, Rosikiewicz M, Fajas L, Sempoux C, Ferrari SL, Greub G, Pitteloud N. β-Klotho deficiency shifts the gut-liver bile acid axis and induces hepatic alterations in mice. Am J Physiol Endocrinol Metab 2018; 315:E833-E847. [PMID: 29944388 DOI: 10.1152/ajpendo.00182.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
β-Klotho (encoded by Klb) is an obligate coreceptor, mediating both fibroblast growth factor (FGF)15 and FGF21 signaling. Klb-/- mice are refractory to metabolic FGF15 and FGF21 action and exhibit derepressed (increased) bile acid (BA) synthesis. Here, we deeply phenotyped male Klb-/- mice on a pure C57BL/6J genetic background, fed a chow diet focusing on metabolic aspects. This aims to better understand the physiological consequences of concomitant FGF15 and FGF21 signaling deficiency, in particular on the gut-liver axis. Klb-/- mice present permanent growth restriction independent of adiposity and energy balance. Klb-/- mice also exhibit few changes in carbohydrate metabolism, combining normal gluco-tolerance, insulin sensitivity, and fasting response with increased gluconeogenic capacity and decreased glycogen mobilization. Livers of Klb-/- mice reveal pathologic features, including a proinflammatory status and initiation of fibrosis. These defects are associated to a massive shift in BA composition in the enterohepatic system and blood circulation featured by a large excess of microbiota-derived deoxycholic acid, classically known for its genotoxicity in the gastrointestinal tract. In conclusion, β-Klotho is a gatekeeper of hepatic integrity through direct action (mediating FGF21 anti-inflammatory signaling) and indirect mechanisms (mediating FGF15 signaling that maintains BA level and composition).
Collapse
Affiliation(s)
- Emmanuel Somm
- Service of Endocrinology, Diabetology, and Metabolism, Department of Physiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Hugues Henry
- Clinical Chemistry Laboratory, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Stephen J Bruce
- Clinical Chemistry Laboratory, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Nicolas Bonnet
- Division of Bone Diseases, Department of Internal Medicine Specialties, Geneva University Hospital and Faculty of Medicine , Geneva , Switzerland
| | - Sophie A Montandon
- Service of Endocrinology, Diabetes, Hypertension, and Nutrition, Geneva University Hospital and Faculty of Medicine , Geneva , Switzerland
| | - Nicolas J Niederländer
- Service of Endocrinology, Diabetology, and Metabolism, Department of Physiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Andrea Messina
- Service of Endocrinology, Diabetology, and Metabolism, Department of Physiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Sébastien Aeby
- Institute of Microbiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Marta Rosikiewicz
- Institute of Microbiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Lluis Fajas
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne , Lausanne , Switzerland
| | - Christine Sempoux
- Institute of Pathology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Serge L Ferrari
- Division of Bone Diseases, Department of Internal Medicine Specialties, Geneva University Hospital and Faculty of Medicine , Geneva , Switzerland
| | - Gilbert Greub
- Institute of Microbiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Nelly Pitteloud
- Service of Endocrinology, Diabetology, and Metabolism, Department of Physiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| |
Collapse
|
21
|
Deubiquitinating Enzymes and Bone Remodeling. Stem Cells Int 2018; 2018:3712083. [PMID: 30123285 PMCID: PMC6079350 DOI: 10.1155/2018/3712083] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 05/29/2018] [Indexed: 02/05/2023] Open
Abstract
Bone remodeling, which is essential for bone homeostasis, is controlled by multiple factors and mechanisms. In the past few years, studies have emphasized the role of the ubiquitin-dependent proteolysis system in regulating bone remodeling. Deubiquitinases, which are grouped into five families, remove ubiquitin from target proteins and are involved in several cell functions. Importantly, a number of deubiquitinases mediate bone remodeling through regulating differentiation and/or function of osteoblast and osteoclasts. In this review, we review the functions and mechanisms of deubiquitinases in mediating bone remodeling.
Collapse
|
22
|
Making Them Commit: Strategies to Influence Phenotypic Differentiation in Mesenchymal Stem Cells. Sports Med Arthrosc Rev 2018; 26:64-69. [DOI: 10.1097/jsa.0000000000000187] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
23
|
Toosi S, Behravan N, Behravan J. Nonunion fractures, mesenchymal stem cells and bone tissue engineering. J Biomed Mater Res A 2018; 106:2552-2562. [PMID: 29689623 DOI: 10.1002/jbm.a.36433] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 03/22/2018] [Accepted: 04/10/2018] [Indexed: 12/15/2022]
Abstract
Depending on the duration of healing process, 5-10% of bone fractures may result in either nonunion or delayed union. Because nonunions remain a clinically important problem, there is interest in the utilization of tissue engineering strategies to augment bone fracture repair. Three basic biologic elements that are required for bone regeneration include cells, extracellular matrix scaffolds and biological adjuvants for growth, differentiation and angiogenesis. Mesenchymal stem cells (MSCs) are capable to differentiate into various types of the cells including chondrocytes, myoblasts, osteoblasts, and adipocytes. Due to their potential for multilineage differentiation, MSCs are considered important contributors in bone tissue engineering research. In this review we highlight the progress in the application of biomaterials, stem cells and tissue engineering in promoting nonunion bone fracture healing. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A:2551-2561, 2018.
Collapse
Affiliation(s)
- Shirin Toosi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nima Behravan
- Exceptionally Talented Students Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Behravan
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
24
|
Human Spinal Bone Dust as a Potential Local Autograft: In Vitro Potent Anabolic Effect on Human Osteoblasts. Spine (Phila Pa 1976) 2018; 43:E193-E199. [PMID: 28723877 DOI: 10.1097/brs.0000000000002331] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN In vitro Study. OBJECTIVE To evaluate the effect that factors released from human posterior spinal bone dust have on primary human osteoblast growth and maturation. SUMMARY OF BACKGROUND DATA Bone dust, created during spinal fusion surgeries, has the potential to be used as an autologous bone graft by providing a source of viable autologous osteoblasts and mesenchymal stem cells with osteogenic potential. Till date, no information is available on whether bone dust also provides a source of anabolic factors with the potential to enhance osteoblast proliferation and maturation, which would enhance its therapeutic potential. METHODS Bone dust was collected from consenting patients undergoing elective posterior spinal fusion surgeries, and primary human osteoblasts were cultured from patients undergoing elective hip or knee arthroplasty. Growth factors and cytokines released by bone dust were quantified using enzyme-linked immunosorbent assay. Primary human osteoblast proliferation and gene expression in response to bone dust were assessed using H-thymidine incorporation and real-time polymerase chain reaction, respectively. RESULTS Human bone dust released anabolic cytokines (IL-1β and IL-6) and growth factors (TGF-β, VEGF, FGF-Basic, and PDGF-BB) in increasing concentrations over a 7-day period. In vitro, the anabolic factors released by bone dust increased osteoblast proliferation by 7-fold, compared with osteoblasts cultured alone. In addition, the factors released from bone dust up-regulated a number of osteoblastic genes integral to osteoblast differentiation, maturation, and angiogenesis. CONCLUSION This study is the first to demonstrate that human posterior spinal bone dust released anabolic factors that potently enhance osteoblast proliferation and the expression of genes that favor bone healing and bone union. As bone dust is anabolic and its harvest is fast, simple, and safe to perform, spinal surgeons should be encouraged to 'recycle' bone dust and harness the regenerative potential of this free autologous bone graft. LEVEL OF EVIDENCE N/A.
Collapse
|
25
|
Kähkönen TE, Ivaska KK, Jiang M, Büki KG, Väänänen HK, Härkönen PL. Role of fibroblast growth factor receptors (FGFR) and FGFR like-1 (FGFRL1) in mesenchymal stromal cell differentiation to osteoblasts and adipocytes. Mol Cell Endocrinol 2018; 461:194-204. [PMID: 28923346 DOI: 10.1016/j.mce.2017.09.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 08/16/2017] [Accepted: 09/13/2017] [Indexed: 12/13/2022]
Abstract
Fibroblast growth factors (FGF) and their receptors (FGFRs) regulate many developmental processes including differentiation of mesenchymal stromal cells (MSC). We developed two MSC lines capable of differentiating to osteoblasts and adipocytes and studied the role of FGFRs in this process. We identified FGFR2 and fibroblast growth factor receptor like-1 (FGFRL1) as possible actors in MSC differentiation with gene microarray and qRT-PCR. FGFR2 and FGFRL1 mRNA expression strongly increased during MSC differentiation to osteoblasts. FGF2 treatment, resulting in downregulation of FGFR2, or silencing FGFR2 expression with siRNAs inhibited osteoblast differentiation. During adipocyte differentiation expression of FGFR1 and FGFRL1 increased and was down-regulated by FGF2. FGFR1 knockdown inhibited adipocyte differentiation. Silencing FGFR2 and FGFR1 in MSCs was associated with decreased FGFRL1 expression in osteoblasts and adipocytes, respectively. Our results suggest that FGFR1 and FGFR2 regulate FGFRL1 expression. FGFRL1 may mediate or modulate FGFR regulation of MSC differentiation together with FGFR2 in osteoblastic and FGFR1 in adipocytic lineage.
Collapse
Affiliation(s)
- T E Kähkönen
- University of Turku, Institute of Biomedicine, Turku, Finland.
| | - K K Ivaska
- University of Turku, Institute of Biomedicine, Turku, Finland
| | - M Jiang
- University of Turku, Institute of Biomedicine, Turku, Finland
| | - K G Büki
- University of Turku, Institute of Biomedicine, Turku, Finland
| | - H K Väänänen
- University of Turku, Institute of Biomedicine, Turku, Finland
| | - P L Härkönen
- University of Turku, Institute of Biomedicine, Turku, Finland
| |
Collapse
|
26
|
Sobacchi C, Palagano E, Villa A, Menale C. Soluble Factors on Stage to Direct Mesenchymal Stem Cells Fate. Front Bioeng Biotechnol 2017; 5:32. [PMID: 28567372 PMCID: PMC5434159 DOI: 10.3389/fbioe.2017.00032] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 04/27/2017] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells that are identified by in vitro plastic adherence, colony-forming capacity, expression of a panel of surface molecules, and ability to differentiate at least toward osteogenic, adipogenic, and chondrogenic lineages. They also produce trophic factors with immunomodulatory, proangiogenic, and antiapoptotic functions influencing the behavior of neighboring cells. On the other hand, a reciprocal regulation takes place; in fact, MSCs can be isolated from several tissues, and depending on the original microenvironment and the range of stimuli received from there, they can display differences in their essential characteristics. Here, we focus mainly on the bone tissue and how soluble factors, such as growth factors, cytokines, and hormones, present in this microenvironment can orchestrate bone marrow-derived MSCs fate. We also briefly describe the alteration of MSCs behavior in pathological settings such as hematological cancer, bone metastasis, and bone marrow failure syndromes. Overall, the possibility to modulate MSCs plasticity makes them an attractive tool for diverse applications of tissue regeneration in cell therapy. Therefore, the comprehensive understanding of the microenvironment characteristics and components better suited to obtain a specific MSCs response can be extremely useful for clinical use.
Collapse
Affiliation(s)
- Cristina Sobacchi
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Milan Unit, Milan, Italy.,Human Genome Laboratory, Humanitas Clinical and Research Institute, Rozzano, Milan, Italy
| | - Eleonora Palagano
- Human Genome Laboratory, Humanitas Clinical and Research Institute, Rozzano, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Anna Villa
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Milan Unit, Milan, Italy.,Human Genome Laboratory, Humanitas Clinical and Research Institute, Rozzano, Milan, Italy
| | - Ciro Menale
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Milan Unit, Milan, Italy.,Human Genome Laboratory, Humanitas Clinical and Research Institute, Rozzano, Milan, Italy
| |
Collapse
|
27
|
Huang CY, Huang TH, Kao CT, Wu YH, Chen WC, Shie MY. Mesoporous Calcium Silicate Nanoparticles with Drug Delivery and Odontogenesis Properties. J Endod 2017; 43:69-76. [DOI: 10.1016/j.joen.2016.09.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 09/08/2016] [Accepted: 09/13/2016] [Indexed: 02/08/2023]
|
28
|
Blair HC, Larrouture QC, Li Y, Lin H, Beer-Stoltz D, Liu L, Tuan RS, Robinson LJ, Schlesinger PH, Nelson DJ. Osteoblast Differentiation and Bone Matrix Formation In Vivo and In Vitro. TISSUE ENGINEERING PART B-REVIEWS 2016; 23:268-280. [PMID: 27846781 DOI: 10.1089/ten.teb.2016.0454] [Citation(s) in RCA: 314] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We review the characteristics of osteoblast differentiation and bone matrix synthesis. Bone in air breathing vertebrates is a specialized tissue that developmentally replaces simpler solid tissues, usually cartilage. Bone is a living organ bounded by a layer of osteoblasts that, because of transport and compartmentalization requirements, produce bone matrix exclusively as an organized tight epithelium. With matrix growth, osteoblasts are reorganized and incorporated into the matrix as living cells, osteocytes, which communicate with each other and surface epithelium by cell processes within canaliculi in the matrix. The osteoblasts secrete the organic matrix, which are dense collagen layers that alternate parallel and orthogonal to the axis of stress loading. Into this matrix is deposited extremely dense hydroxyapatite-based mineral driven by both active and passive transport and pH control. As the matrix matures, hydroxyapatite microcrystals are organized into a sophisticated composite in the collagen layer by nucleation in the protein lattice. Recent studies on differentiating osteoblast precursors revealed a sophisticated proton export network driving mineralization, a gene expression program organized with the compartmentalization of the osteoblast epithelium that produces the mature bone matrix composite, despite varying serum calcium and phosphate. Key issues not well defined include how new osteoblasts are incorporated in the epithelial layer, replacing those incorporated in the accumulating matrix. Development of bone in vitro is the subject of numerous projects using various matrices and mesenchymal stem cell-derived preparations in bioreactors. These preparations reflect the structure of bone to variable extents, and include cells at many different stages of differentiation. Major challenges are production of bone matrix approaching the in vivo density and support for trabecular bone formation. In vitro differentiation is limited by the organization and density of osteoblasts and by endogenous and exogenous inhibitors.
Collapse
Affiliation(s)
- Harry C Blair
- 1 Veteran's Affairs Medical Center , Pittsburgh, Pennsylvania.,2 Department of Pathology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | | | - Yanan Li
- 3 Department of Stomatology, Chinese PLA General Hospital , Beijing, China
| | - Hang Lin
- 4 Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Donna Beer-Stoltz
- 2 Department of Pathology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Li Liu
- 2 Department of Pathology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Rocky S Tuan
- 4 Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Lisa J Robinson
- 5 Department of Pathology, West Virginia University School of Medicine , Morgantown, West Virginia.,6 Department of Microbiology, Immunology & Cell Biology, West Virginia University School of Medicine , Morgantown, West Virginia
| | - Paul H Schlesinger
- 7 Department of Cell Biology, Washington University , Saint Louis, Missouri
| | - Deborah J Nelson
- 8 Department of Neurobiology, Pharmacology & Physiology, University of Chicago , Chicago, Illinois
| |
Collapse
|
29
|
Moghaddam A, Yildirim TM, Westhauser F, Danner W, Swing T, Bruckner T, Biglari B. Low intensity pulsed ultrasound in the treatment of long bone nonunions: Evaluation of cytokine expression as a tool for objectifying nonunion therapy. J Orthop 2016; 13:306-12. [PMID: 27408510 DOI: 10.1016/j.jor.2016.06.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 06/17/2016] [Accepted: 06/27/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Nonunion is one of the most common complications in bone healing. After several clinical studies, the effect of low intensity pulsed ultrasound (LIPUS) in the treatment of nonunions remains unclear because of the difficulty in evaluating its effect on bone healing. In former studies, the analysis of serum cytokine expression patterns over time has proven to be an objective method for showing the bone healing process and evaluating nonunion therapies. This study evaluates LIPUS as a treatment option for patients with nonunions based on the analysis of serum cytokine expression. METHODS In this prospective, single institution study, venous blood samples from 23 patients were taken from October 2012 to October 2013 before starting LIPUS therapy and at the end of week 1 and 2 and after 1, 2, and 3 months. Patients attended clinical and radiological follow-up examinations at the same intervals. After treating all patients according to the LIPUS therapy protocol, we divided them into two groups: Group 1 consisted of patients with healing at the nonunion site, and Group 2 consisted of patients with failed nonunion therapy. We measured transforming growth factor-β1 (TGF-β1), platelet-derived growth factor (PDGF), and basic fibroblastic growth factor (bFGF) at all time-points. RESULTS The TGF-β1 serum concentration increased from the pre-treatment value to 1 week within the unsuccessful group. Otherwise, no significant differences between groups in measured cytokines during LIPUS therapy could be detected. CONCLUSION Our findings suggest that LIPUS does not lead to a significant increase in cytokine levels in patients with nonunions. It is likely that "successful" treatment can be attributed to spontaneous healing. Our results suggest that LIPUS is not a proper treatment for long bone nonunions.
Collapse
Affiliation(s)
- Arash Moghaddam
- HTRG - Heidelberg Trauma Research Group, Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Heidelberg University Hospital, Schlierbacher Landstraße 200a, 69118 Heidelberg, Baden-Württemberg, Germany
| | - Timur Mert Yildirim
- HTRG - Heidelberg Trauma Research Group, Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Heidelberg University Hospital, Schlierbacher Landstraße 200a, 69118 Heidelberg, Baden-Württemberg, Germany
| | - Fabian Westhauser
- HTRG - Heidelberg Trauma Research Group, Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Heidelberg University Hospital, Schlierbacher Landstraße 200a, 69118 Heidelberg, Baden-Württemberg, Germany
| | - Wolfgang Danner
- BG Trauma Centre Ludwigshafen, Ludwig-Guttmann-Str. 13, 67071 Ludwigshafen, Rheinland-Pfalz, Germany
| | - Tyler Swing
- HTRG - Heidelberg Trauma Research Group, Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Heidelberg University Hospital, Schlierbacher Landstraße 200a, 69118 Heidelberg, Baden-Württemberg, Germany
| | - Thomas Bruckner
- Institute of Medical Biometry and Informatics, University of Heidelberg, Im Neuenheimer Feld 305, 69120 Heidelberg, Baden-Württemberg, Germany
| | - Bahram Biglari
- BG Trauma Centre Ludwigshafen, Ludwig-Guttmann-Str. 13, 67071 Ludwigshafen, Rheinland-Pfalz, Germany
| |
Collapse
|
30
|
Simann M, Le Blanc S, Schneider V, Zehe V, Lüdemann M, Schütze N, Jakob F, Schilling T. Canonical FGFs Prevent Osteogenic Lineage Commitment and Differentiation of Human Bone Marrow Stromal Cells Via ERK1/2 Signaling. J Cell Biochem 2016; 118:263-275. [PMID: 27305863 DOI: 10.1002/jcb.25631] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 06/14/2016] [Indexed: 12/21/2022]
Abstract
Controlling the adipo-osteogenic lineage decision of trabecular human bone marrow stromal cells (hBMSCs) in favor of osteogenesis represents a promising approach for osteoporosis therapy and prevention. Previously, Fibroblast Growth Factor 1 (FGF1) and its subfamily member FGF2 were scored as leading candidates to exercise control over skeletal precursor commitment and lineage decision albeit literature results are highly inconsistent. We show here that FGF1 and 2 strongly prevent the osteogenic commitment and differentiation of hBMSCs. Mineralization of extracellular matrix (ECM) and mRNA expression of osteogenic marker genes Alkaline Phosphatase (ALP), Collagen 1A1 (COL1A1), and Integrin-Binding Sialoprotein (IBSP) were significantly reduced. Furthermore, master regulators of osteogenic commitment like Runt-Related Transcription Factor 2 (RUNX2) and Bone Morphogenetic Protein 4 (BMP4) were downregulated. When administered under adipogenic culture conditions, canonical FGFs did not support osteogenic marker expression. Moreover despite the presence of osteogenic differentiation factors, FGFs even disabled the pro-osteogenic lineage decision of pre-differentiated adipocytic cells. In contrast to FGF Receptor 2 (FGFR2), FGFR1 was stably expressed throughout osteogenic and adipogenic differentiation and FGF addition. Moreover, FGFR1 and Extracellular Signal-Regulated Kinases 1 and 2 (ERK1/2) were found to be responsible for underlying signal transduction using respective inhibitors. Taken together, we present new findings indicating that canonical FGFR-ERK1/2 signaling entrapped hBMSCs in a pre-committed state and arrested further maturation of committed precursors. Our results might aid in unraveling and controlling check points relevant for ageing-associated aberrant adipogenesis with consequences for the treatment of degenerative diseases such as osteoporosis and for skeletal tissue engineering strategies. J. Cell. Biochem. 118: 263-275, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Meike Simann
- Department of Orthopedics, Orthopedic Center for Musculoskeletal Research, University of Würzburg, Würzburg, Germany
| | - Solange Le Blanc
- Department of Orthopedics, Orthopedic Center for Musculoskeletal Research, University of Würzburg, Würzburg, Germany
| | - Verena Schneider
- Chair Tissue Engineering & Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Viola Zehe
- Department of Orthopedics, Orthopedic Center for Musculoskeletal Research, University of Würzburg, Würzburg, Germany
| | - Martin Lüdemann
- Orthopedic Department König-Ludwig-Haus, Center for Musculoskeletal Research, University of Würzburg, Würzburg, Germany
| | - Norbert Schütze
- Department of Orthopedics, Orthopedic Center for Musculoskeletal Research, University of Würzburg, Würzburg, Germany
| | - Franz Jakob
- Department of Orthopedics, Orthopedic Center for Musculoskeletal Research, University of Würzburg, Würzburg, Germany
| | - Tatjana Schilling
- Department of Orthopedics, Orthopedic Center for Musculoskeletal Research, University of Würzburg, Würzburg, Germany
| |
Collapse
|
31
|
Chen T, Gomez AW, Zuo Y, Li X, Zhang Z, Li Y, Hu J, Li J. Osteogenic potential and synergistic effects of growth factors delivered from a bionic composite system. J Biomed Mater Res A 2015; 104:659-668. [PMID: 26514654 DOI: 10.1002/jbm.a.35605] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 10/15/2015] [Accepted: 10/28/2015] [Indexed: 01/18/2023]
Affiliation(s)
- Tao Chen
- State Key Laboratory of Oral Diseases and Department of Oral and Maxillofacial Surgery, West China College of Stomatology; Sichuan University; Chengdu 610041 China
| | - Alan W. Gomez
- Department of Surgery, Division of Plastic and Reconstructive Surgery; Stanford School of Medicine, Stanford University; Stanford California 94305
| | - Yi Zuo
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University; Chengdu 610064 China
| | - Xiang Li
- State Key Laboratory of Oral Diseases and Department of Oral and Maxillofacial Surgery, West China College of Stomatology; Sichuan University; Chengdu 610041 China
| | - Zhen Zhang
- State Key Laboratory of Oral Diseases and Department of Oral and Maxillofacial Surgery, West China College of Stomatology; Sichuan University; Chengdu 610041 China
| | - Yubao Li
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University; Chengdu 610064 China
| | - Jing Hu
- State Key Laboratory of Oral Diseases and Department of Oral and Maxillofacial Surgery, West China College of Stomatology; Sichuan University; Chengdu 610041 China
| | - Jihua Li
- State Key Laboratory of Oral Diseases and Department of Oral and Maxillofacial Surgery, West China College of Stomatology; Sichuan University; Chengdu 610041 China
| |
Collapse
|
32
|
Pawaputanon Na Mahasarakham C, Ezura Y, Kawasaki M, Smriti A, Moriya S, Yamada T, Izu Y, Nifuji A, Nishimori K, Izumi Y, Noda M. BMP-2 Enhances Lgr4 Gene Expression in Osteoblastic Cells. J Cell Physiol 2015; 231:887-95. [PMID: 26332449 DOI: 10.1002/jcp.25180] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 08/31/2015] [Indexed: 01/21/2023]
Abstract
Osteoporosis is one of the most prevalent diseases and the number of patients suffering from this disease is soaring due to the increase in the aged population in the world. The severity of bone loss in osteoporosis is based on the levels of impairment in the balance between bone formation and bone resorption, two arms of the bone metabolism, and bone remodeling. However, determination of bone formation levels is under many layers of control that are as yet fully defined. Bone morphogenetic protein (BMP) plays a key role in regulation of bone formation while its downstream targets are still incompletely understood. Lgr4 gene encodes an orphan receptor and has been identified as a genetic determinant for bone mass in osteoporotic patients. Here, we examine the effects of BMP on the expression of Lgr4 in osteoblastic cells. Lgr4 gene is expressed in an osteoblastic cell line, MC3T3E1 in a time dependent manner during the culture. BMP treatment enhances Lgr4 mRNA expression at least in part via transcriptional event. When Lgr4 mRNA is knocked down, the levels of BMP-induced increase in alkaline phosphatase (Alp) activity and Alp mRNA are suppressed. BMP enhancement of Lgr4 gene expression is suppressed by FGF and reversed by dexamethasone. BMP also enhances Lgr4 expression in primary cultures of calvarial osteoblasts. These data indicate that Lgr4 gene is regulated by BMP and is required for BMP effects on osteoblastic differentiation. J. Cell. Physiol. 231: 887-895, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Chantida Pawaputanon Na Mahasarakham
- Department of Molecular Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Japan.,Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| | - Yoichi Ezura
- Department of Molecular Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Japan
| | - Makiri Kawasaki
- Department of Molecular Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Japan
| | - Arayal Smriti
- Department of Molecular Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Japan
| | - Shuichi Moriya
- Department of Molecular Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Japan
| | - Takayuki Yamada
- Department of Molecular Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Japan
| | - Yayoi Izu
- Department of Molecular Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Japan
| | - Akira Nifuji
- Department of Pharmacology, Tsurumi University, School of Dental Medicine, Japan
| | - Katsuhiko Nishimori
- Laboratory of Molecular Biology, Department of Molecular and Cell Biology, Graduate School of Agricultural Science, Tohoku University, Japan
| | - Yuichi Izumi
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| | - Masaki Noda
- Department of Molecular Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Japan
| |
Collapse
|
33
|
Sagomonyants K, Kalajzic I, Maye P, Mina M. Enhanced Dentinogenesis of Pulp Progenitors by Early Exposure to FGF2. J Dent Res 2015; 94:1582-90. [PMID: 26276371 DOI: 10.1177/0022034515599768] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Members of the fibroblast growth factor (FGF) family play essential and important roles in primary and reparative dentinogenesis. Although there appears to be a general agreement on the effects of FGF signaling on the proliferation of pulp cells, there are conflicting results regarding its effects on odontoblast differentiation. We recently examined the effects of continuous exposure of dental pulp cells to FGF2 and showed that the effects of FGF2 on differentiation of progenitor cells into odontoblasts were stage specific and dependent on the stage of cell maturity. The purpose of this study was to gain further insight into cellular and molecular mechanisms regulating the stimulatory effects of FGF2 on odontoblast differentiation. To do so, we examined the effects of early and limited exposure of pulp cells from a series of green fluorescent protein (GFP) reporter transgenic mice that display stage-specific activation of transgenes during odontoblast differentiation to FGF2. Our results showed that early and limited exposure of pulp cells to FGF2 did not have significant effects on the extent of mineralization but induced significant increases in the expression of Dmp1 and Dspp and the number of DMP1-GFP(+) and DSPP-Cerulean(+) odontoblasts. Our results also showed that the stimulatory effects of FGF2 on odontoblast differentiation were mediated through FGFR/MEK/Erk1/2 signaling, increases in Bmp2, and activation of the BMP/BMPR signaling pathway. These observations show that early and limited exposure of pulp cells to FGF2 alone promotes odontoblast differentiation and provides critical insight for applications of FGF2 in dentin regeneration.
Collapse
Affiliation(s)
- K Sagomonyants
- Department of Craniofacial Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - I Kalajzic
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - P Maye
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - M Mina
- Department of Craniofacial Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
34
|
Bayer EA, Gottardi R, Fedorchak MV, Little SR. The scope and sequence of growth factor delivery for vascularized bone tissue regeneration. J Control Release 2015; 219:129-140. [PMID: 26264834 DOI: 10.1016/j.jconrel.2015.08.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/01/2015] [Accepted: 08/03/2015] [Indexed: 12/21/2022]
Abstract
Bone regeneration is a complex process, that in vivo, requires the highly coordinated presentation of biochemical cues to promote the various stages of angiogenesis and osteogenesis. Taking inspiration from the natural healing process, a wide variety of growth factors are currently being released within next generation tissue engineered scaffolds (in a variety of ways) in order to heal non-union fractures and bone defects. This review will focus on the delivery of multiple growth factors to the bone regeneration niche, specifically 1) dual growth factor delivery signaling and crosstalk, 2) the importance of growth factor timing and temporal separation, and 3) the engineering of delivery systems that allow for temporal control over presentation of soluble growth factors. Alternative methods for growth factor presentation, including the use of gene therapy and platelet-rich plasma scaffolds, are also discussed.
Collapse
Affiliation(s)
- E A Bayer
- The University of Pittsburgh, Department of Bioengineering, USA; The University of Pittsburgh, The McGowan Institute for Regenerative Medicine, USA
| | - R Gottardi
- The University of Pittsburgh, Department of Chemical Engineering, USA; The University of Pittsburgh, Department of Orthopedic Surgery, USA; The University of Pittsburgh, The McGowan Institute for Regenerative Medicine, USA; RiMED Foundation, Palermo, Italy
| | - M V Fedorchak
- The University of Pittsburgh, Department of Bioengineering, USA; The University of Pittsburgh, Department of Chemical Engineering, USA; The University of Pittsburgh, Department of Ophthalmology, USA; The University of Pittsburgh, The McGowan Institute for Regenerative Medicine, USA
| | - S R Little
- The University of Pittsburgh, Department of Bioengineering, USA; The University of Pittsburgh, Department of Chemical Engineering, USA; The University of Pittsburgh, Department of Immunology, USA; The University of Pittsburgh, The McGowan Institute for Regenerative Medicine, USA.
| |
Collapse
|
35
|
Wu BC, Youn SC, Kao CT, Huang SC, Hung CJ, Chou MY, Huang TH, Shie MY. The effects of calcium silicate cement/fibroblast growth factor-2 composite on osteogenesis accelerator in human dental pulp cells. J Dent Sci 2015. [DOI: 10.1016/j.jds.2013.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
36
|
Han X, Xiao Z, Quarles LD. Membrane and integrative nuclear fibroblastic growth factor receptor (FGFR) regulation of FGF-23. J Biol Chem 2015; 290:10447-59. [PMID: 25752607 PMCID: PMC4400353 DOI: 10.1074/jbc.m114.609230] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 02/27/2015] [Indexed: 01/15/2023] Open
Abstract
Fibroblastic growth factor receptor 1 (FGFR1) signaling pathways are implicated in the regulation of FGF-23 gene transcription, but the molecular pathways remain poorly defined. We used low molecular weight (LMW, 18 kDa) FGF-2 and high molecular weight (HMW) FGF-2 isoforms, which, respectively, activate cell surface FGF receptors and intranuclear FGFR1, to determine the roles of membrane FGFRs and integrative nuclear FGFR1 signaling (INFS) in the regulation of FGF-23 gene transcription in osteoblasts. We found that LMW-FGF-2 induced NFAT and Ets1 binding to conserved cis-elements in the proximal FGF-23 promoter and stimulated FGF-23 promoter activity through PLCγ/calcineurin/NFAT and MAPK pathways in SaOS-2 and MC3T3-E1 osteoblasts. In contrast, HMW-FGF-2 stimulated FGF-23 promoter activity in osteoblasts through a cAMP-dependent binding of FGFR1 and cAMP-response element-binding protein (CREB) to a conserved cAMP response element (CRE) contiguous with the NFAT binding site in the FGF-23 promoter. Mutagenesis of the NFAT and CRE binding sites, respectively, inhibited the effects of LMW-FGF-2 and HMW-FGF-23 to stimulate FGF-23 promoter activity. FGF-2 activation of both membrane FGFRs and INFS-dependent FGFR1 pathways may provide a means to integrate systemic and local regulation of FGF-23 transcription under diverse physiological and pathological conditions.
Collapse
Affiliation(s)
- Xiaobin Han
- From the Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Zhousheng Xiao
- From the Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - L Darryl Quarles
- From the Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| |
Collapse
|
37
|
Marie PJ. Osteoblast dysfunctions in bone diseases: from cellular and molecular mechanisms to therapeutic strategies. Cell Mol Life Sci 2015; 72:1347-61. [PMID: 25487608 PMCID: PMC11113967 DOI: 10.1007/s00018-014-1801-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 11/13/2014] [Accepted: 12/01/2014] [Indexed: 12/27/2022]
Abstract
Several metabolic, genetic and oncogenic bone diseases are characterized by defective or excessive bone formation. These abnormalities are caused by dysfunctions in the commitment, differentiation or survival of cells of the osteoblast lineage. During the recent years, significant advances have been made in our understanding of the cellular and molecular mechanisms underlying the osteoblast dysfunctions in osteoporosis, skeletal dysplasias and primary bone tumors. This led to suggest novel therapeutic approaches to correct these abnormalities such as the modulation of WNT signaling, the pharmacological modulation of proteasome-mediated protein degradation, the induction of osteoprogenitor cell differentiation, the repression of cancer cell proliferation and the manipulation of epigenetic mechanisms. This article reviews our current understanding of the major cellular and molecular mechanisms inducing osteoblastic cell abnormalities in age-related bone loss, genetic skeletal dysplasias and primary bone tumors, and discusses emerging therapeutic strategies to counteract the osteoblast abnormalities in these disorders of bone formation.
Collapse
Affiliation(s)
- Pierre J Marie
- INSERM UMR-1132, Hôpital Lariboisière, 2 rue Ambroise Paré, 75475, Paris Cedex 10, France,
| |
Collapse
|
38
|
Sagomonyants K, Mina M. Stage-specific effects of fibroblast growth factor 2 on the differentiation of dental pulp cells. Cells Tissues Organs 2015; 199:311-28. [PMID: 25823776 DOI: 10.1159/000371343] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2014] [Indexed: 12/31/2022] Open
Abstract
Dentinogenesis is a complex and multistep process, which is regulated by various growth factors, including members of the fibroblast growth factor (FGF) family. Both positive and negative effects of FGFs on dentinogenesis have been reported, but the underlying mechanisms of these conflicting results are still unclear. To gain a better insight into the role of FGF2 in dentinogenesis, we used dental pulp cells from various transgenic mice, in which fluorescent protein expression identifies cells at different stages of odontoblast differentiation. Our results showed that the continuous exposure of pulp cells to FGF2 inhibited mineralization and revealed both the stimulatory and inhibitory effects of FGF2 on the expression of markers of dentinogenesis and various transgenes. During the proliferation phase of in vitro growth, FGF2 increased the expression of markers of dentinogenesis and the percentages of dentin matrix protein 1/green fluorescent protein (DMP1-GFP)-positive functional odontoblasts and dentin sialophosphoprotein (DSPP)-Cerulean-positive odontoblasts. Additional exposure to FGF2 during the differentiation/mineralization phase of in vitro growth decreased the extent of mineralization and the expression of markers of dentinogenesis and of the DMP1-GFP and DSPP-Cerulean transgenes. Recovery experiments showed that the inhibitory effects of FGF2 on dentinogenesis were related to the blocking of the differentiation of cells into mature odontoblasts. These observations together showed the stage-specific effects of FGF2 on dentinogenesis by dental pulp cells, and they provide critical information for the development of improved treatments for vital pulp therapy and dentin regeneration.
Collapse
Affiliation(s)
- Karen Sagomonyants
- Department of Craniofacial Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, Conn., USA
| | | |
Collapse
|
39
|
Ségaliny AI, Tellez-Gabriel M, Heymann MF, Heymann D. Receptor tyrosine kinases: Characterisation, mechanism of action and therapeutic interests for bone cancers. J Bone Oncol 2015; 4:1-12. [PMID: 26579483 PMCID: PMC4620971 DOI: 10.1016/j.jbo.2015.01.001] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 01/18/2015] [Indexed: 01/13/2023] Open
Abstract
Bone cancers are characterised by the development of tumour cells in bone sites, associated with a dysregulation of their environment. In the last two decades, numerous therapeutic strategies have been developed to target the cancer cells or tumour niche. As the crosstalk between these two entities is tightly controlled by the release of polypeptide mediators activating signalling pathways through several receptor tyrosine kinases (RTKs), RTK inhibitors have been designed. These inhibitors have shown exciting clinical impacts, such as imatinib mesylate, which has become a reference treatment for chronic myeloid leukaemia and gastrointestinal tumours. The present review gives an overview of the main molecular and functional characteristics of RTKs, and focuses on the clinical applications that are envisaged and already assessed for the treatment of bone sarcomas and bone metastases.
Collapse
Affiliation(s)
- Aude I Ségaliny
- INSERM, UMR 957, Equipe LIGUE Nationale Contre le Cancer 2012, Nantes 44035, France ; Université de Nantes, Nantes atlantique universités, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, Nantes, France
| | - Marta Tellez-Gabriel
- INSERM, UMR 957, Equipe LIGUE Nationale Contre le Cancer 2012, Nantes 44035, France ; Université de Nantes, Nantes atlantique universités, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, Nantes, France
| | - Marie-Françoise Heymann
- INSERM, UMR 957, Equipe LIGUE Nationale Contre le Cancer 2012, Nantes 44035, France ; Université de Nantes, Nantes atlantique universités, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, Nantes, France ; CHU de Nantes, France
| | - Dominique Heymann
- INSERM, UMR 957, Equipe LIGUE Nationale Contre le Cancer 2012, Nantes 44035, France ; Université de Nantes, Nantes atlantique universités, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, Nantes, France ; CHU de Nantes, France
| |
Collapse
|
40
|
Stegen S, van Gastel N, Carmeliet G. Bringing new life to damaged bone: the importance of angiogenesis in bone repair and regeneration. Bone 2015; 70:19-27. [PMID: 25263520 DOI: 10.1016/j.bone.2014.09.017] [Citation(s) in RCA: 316] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 09/14/2014] [Accepted: 09/18/2014] [Indexed: 12/26/2022]
Abstract
Bone has the unique capacity to heal without the formation of a fibrous scar, likely because several of the cellular and molecular processes governing bone healing recapitulate the events during skeletal development. A critical component in bone healing is the timely appearance of blood vessels in the fracture callus. Angiogenesis, the formation of new blood vessels from pre-existing ones, is stimulated after fracture by the local production of numerous angiogenic growth factors. The fracture vasculature not only supplies oxygen and nutrients, but also stem cells able to differentiate into osteoblasts and in a later phase also the ions necessary for mineralization. This review provides a concise report of the regulation of angiogenesis by bone cells, its importance during bone healing and its possible therapeutic applications in bone tissue engineering. This article is part of a Special Issue entitled "Stem Cells and Bone".
Collapse
Affiliation(s)
- Steve Stegen
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium
| | - Nick van Gastel
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
41
|
Zhang W, Xue D, Hu D, Xie T, Tao Y, Zhu T, Chen E, Pan Z. Secreted klotho protein attenuates osteogenic differentiation of human bone marrow mesenchymal stem cells in vitro via inactivation of the FGFR1/ERK signaling pathway. Growth Factors 2015; 33:356-65. [PMID: 26607681 DOI: 10.3109/08977194.2015.1108313] [Citation(s) in RCA: 28] [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] [Indexed: 01/03/2023]
Abstract
Increasing evidence indicates that the osteogenic differentiation of mesenchymal stem cells (MSCs) is related to bone formation, heterotopic ossification, and even vascular calcification. Therefore, it is essential to understand the microenvironment that regulates these processes. The Klotho gene plays an important role in tissue mineralization, and its secreted protein functions as a hormone. We investigated the effects of secreted Klotho protein on the osteogenesis of human bone marrow MSC (hBMSCs). To this end, the cells received osteogenic medium with or without Klotho protein. The results showed that osteoblast-specific gene expression and mineral deposition were decreased when MSCs were incubated with Klotho. Klotho reduced the expression of fibroblast growth factor receptor 1 (FGFR1) and phosphorylated extracellular signal-regulated kinase 1/2. However, both MEK and FGFR1 inhibitors delayed bone mineral formation more than Klotho. These data suggest that secreted Klotho protein attenuates the osteogenic differentiation of hBMSCs in vitro through FGFR1/ERK signaling.
Collapse
Affiliation(s)
- Wei Zhang
- a Department of Orthopedics , Second Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou , People's Republic of China
| | - Deting Xue
- a Department of Orthopedics , Second Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou , People's Republic of China
| | - Dongcai Hu
- a Department of Orthopedics , Second Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou , People's Republic of China
| | - Tao Xie
- a Department of Orthopedics , Second Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou , People's Republic of China
| | - Yiqing Tao
- a Department of Orthopedics , Second Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou , People's Republic of China
| | - Ting Zhu
- a Department of Orthopedics , Second Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou , People's Republic of China
| | - Erman Chen
- a Department of Orthopedics , Second Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou , People's Republic of China
| | - Zhijun Pan
- a Department of Orthopedics , Second Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou , People's Republic of China
| |
Collapse
|
42
|
Non-apoptotic functions of caspase-7 during osteogenesis. Cell Death Dis 2014; 5:e1366. [PMID: 25118926 PMCID: PMC4454305 DOI: 10.1038/cddis.2014.330] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 06/12/2014] [Accepted: 06/19/2014] [Indexed: 11/09/2022]
Abstract
Caspase-3 and -7 are generally known for their central role in the execution of apoptosis. However, their function is not limited to apoptosis and under specific conditions activation has been linked to proliferation or differentiation of specialised cell types. In the present study, we followed the localisation of the activated form of caspase-7 during intramembranous (alveolar and mandibular bones) and endochondral (long bones of limbs) ossification in mice. In both bone types, the activated form of caspase-7 was detected from the beginning of ossification during embryonic development and persisted postnatally. The bone status was investigated by microCT in both wild-type and caspase-7-deficient adult mice. Intramembranous bone in mutant mice displayed a statistically significant decrease in volume while the mineral density was not altered. Conversely, endochondral bone showed constant volume but a significant decrease in mineral density in caspase-7 knock-out mice. Cleaved caspase-7 was present in a number of cells that did not show signs of apoptosis. PCR array analysis of the mandibular bone of caspase-7-deficient versus wild-type mice pointed to a significant decrease in mRNA levels for Msx1 and Smad1 in early bone formation. These observations might explain the decrease in the alveolar bone volume of adult knock-out mice. In conclusion, this study is the first to report a non-apoptotic function of caspase-7 in osteogenesis and also demonstrates further specificities in endochondral versus intramembranous ossification.
Collapse
|
43
|
Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) Signalling Enhances Osteogenesis in UMR-106 Cell Line. J Mol Neurosci 2014; 54:555-73. [DOI: 10.1007/s12031-014-0389-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 07/22/2014] [Indexed: 01/14/2023]
|
44
|
Liu CH, Hung CJ, Huang TH, Lin CC, Kao CT, Shie MY. Odontogenic differentiation of human dental pulp cells by calcium silicate materials stimulating via FGFR/ERK signaling pathway. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2014; 43:359-66. [PMID: 25175224 DOI: 10.1016/j.msec.2014.06.025] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Revised: 05/07/2014] [Accepted: 06/30/2014] [Indexed: 10/25/2022]
Abstract
Bone healing needs a complex interaction of growth factors that establishes an environment for efficient bone formation. We examine how calcium silicate (CS) and tricalcium phosphate (β-TCP) cements influence the behavior of human dental pulp cells (hDPCs) through fibroblast growth factor receptor (FGFR) and active MAPK pathways, in particular ERK. The hDPCs are cultured with β-TCP and CS, after which the cells' viability and odontogenic differentiation markers are determined by using PrestoBlue® assay and western blot, respectively. The effect of small interfering RNA (siRNA) transfection targeting FGFR was also evaluated. The results showed that CS promoted cell proliferation and enhances FGFR expression. It was also found that CS increases ERK and p38 activity in hDPCs, and furthermore, raises the expression and secretion of DSP, and DMP-1. Additionally, statistically significant differences (p<0.05) have been found in the calcium deposition in si-FGFR transfection and ERK inhibitor between CS and β-TCP; these variations indicated that ERK/MAPK signaling is involved in the silicon-induced odontogenic differentiation of hDPCs. The current study shows that CS substrates play a key role in odontoblastic differentiation of hDPCs through FGFR and modulate ERK/MAPK activation.
Collapse
Affiliation(s)
- Chao-Hsin Liu
- School of Dentistry, Chung Shan Medical University, Taichung City, Taiwan
| | - Chi-Jr Hung
- School of Dentistry, Chung Shan Medical University, Taichung City, Taiwan; Department of Dentistry, Chung Shan Medical University Hospital, Taichung City, Taiwan
| | - Tsui-Hsien Huang
- School of Dentistry, Chung Shan Medical University, Taichung City, Taiwan; Department of Dentistry, Chung Shan Medical University Hospital, Taichung City, Taiwan
| | - Chi-Chang Lin
- Department of Chemical and Materials Engineering, Tunghai University, Taichung City, Taiwan
| | - Chia-Tze Kao
- School of Dentistry, Chung Shan Medical University, Taichung City, Taiwan; Department of Dentistry, Chung Shan Medical University Hospital, Taichung City, Taiwan
| | - Ming-You Shie
- Department of Chemical and Materials Engineering, Tunghai University, Taichung City, Taiwan.
| |
Collapse
|
45
|
Padmashali RM, Mistriotis P, Liang MS, Andreadis ST. Lentiviral arrays for live-cell dynamic monitoring of gene and pathway activity during stem cell differentiation. Mol Ther 2014; 22:1971-82. [PMID: 24895998 DOI: 10.1038/mt.2014.103] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 05/02/2014] [Indexed: 02/07/2023] Open
Abstract
Uncovering the complexity of mesenchymal stem cell (MSC) differentiation requires novel methods to capture the dynamics of the process in a quantitative and high-throughput manner. To this end, we developed a lentiviral array (LVA) of reporters to capture the dynamics of gene and pathway activity during MSC differentiation into adipogenic, chondrogenic, and osteogenic lineages. Our results identified signature promoters and pathways with unique activation profile for each MSC lineage. In combination with chemical inhibitors, lineage-specific reporters predicted the effects of signaling pathway perturbations on MSC differentiation. Interestingly, some pathways were critical for differentiation into all lineages, while others had differential effects on each lineage. Our study suggests that when combined with large chemical or siRNA libraries, the reporter LVA can be used to uncover novel genes and signaling pathways affecting complex biological processes such as stem cell differentiation or reprogramming.
Collapse
Affiliation(s)
- Roshan M Padmashali
- Department of Chemical and Biological Engineering, Bioengineering Laboratory, University at Buffalo, The State University of New York, Amherst, New York, USA
| | - Panagiotis Mistriotis
- Department of Chemical and Biological Engineering, Bioengineering Laboratory, University at Buffalo, The State University of New York, Amherst, New York, USA
| | - Mao-shih Liang
- Department of Chemical and Biological Engineering, Bioengineering Laboratory, University at Buffalo, The State University of New York, Amherst, New York, USA
| | - Stelios T Andreadis
- 1] Department of Chemical and Biological Engineering, Bioengineering Laboratory, University at Buffalo, The State University of New York, Amherst, New York, USA [2] Department of Biomedical Engineering, University at Buffalo, The State University of New York, New York, Amherst, USA [3] Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York, USA
| |
Collapse
|
46
|
Senarath-Yapa K, McArdle A, Renda A, Longaker MT, Quarto N. Adipose-derived stem cells: a review of signaling networks governing cell fate and regenerative potential in the context of craniofacial and long bone skeletal repair. Int J Mol Sci 2014; 15:9314-30. [PMID: 24865492 PMCID: PMC4100096 DOI: 10.3390/ijms15069314] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 05/16/2014] [Accepted: 05/20/2014] [Indexed: 02/07/2023] Open
Abstract
Improvements in medical care, nutrition and social care are resulting in a commendable change in world population demographics with an ever increasing skew towards an aging population. As the proportion of the world's population that is considered elderly increases, so does the incidence of osteodegenerative disease and the resultant burden on healthcare. The increasing demand coupled with the limitations of contemporary approaches, have provided the impetus to develop novel tissue regeneration therapies. The use of stem cells, with their potential for self-renewal and differentiation, is one potential solution. Adipose-derived stem cells (ASCs), which are relatively easy to harvest and readily available have emerged as an ideal candidate. In this review, we explore the potential for ASCs to provide tangible therapies for craniofacial and long bone skeletal defects, outline key signaling pathways that direct these cells and describe how the developmental signaling program may provide clues on how to guide these cells in vivo. This review also provides an overview of the importance of establishing an osteogenic microniche using appropriately customized scaffolds and delineates some of the key challenges that still need to be overcome for adult stem cell skeletal regenerative therapy to become a clinical reality.
Collapse
Affiliation(s)
- Kshemendra Senarath-Yapa
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, School of Medicine, Stanford University, Stanford, CA 94305-2200, USA; E-Mails: (K.S.-Y.); (A.M.)
| | - Adrian McArdle
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, School of Medicine, Stanford University, Stanford, CA 94305-2200, USA; E-Mails: (K.S.-Y.); (A.M.)
| | - Andrea Renda
- Dipartimento di Scienze Biomediche Avanzate, Universita’ degli Studi di Napoli Federico II, Napoli 80131, Italy; E-Mail:
| | - Michael T. Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, School of Medicine, Stanford University, Stanford, CA 94305-2200, USA; E-Mails: (K.S.-Y.); (A.M.)
| | - Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, School of Medicine, Stanford University, Stanford, CA 94305-2200, USA; E-Mails: (K.S.-Y.); (A.M.)
- Dipartimento di Scienze Biomediche Avanzate, Universita’ degli Studi di Napoli Federico II, Napoli 80131, Italy; E-Mail:
| |
Collapse
|
47
|
Osteoblast and osteocyte: games without frontiers. Arch Biochem Biophys 2014; 561:3-12. [PMID: 24832390 DOI: 10.1016/j.abb.2014.05.003] [Citation(s) in RCA: 234] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 04/30/2014] [Accepted: 05/03/2014] [Indexed: 01/23/2023]
Abstract
The portrait of osteoblasts and osteocytes has been subjected to a revision, since a large body of evidence is attributing these cells amazing roles both inside and outside the bone. The osteoblast, long confined to its bone building function, is actually a very eclectic cell, actively regulating osteoclast formation and function as well as hematopoietic stem cells homeostasis. It is also an endocrine cell, affecting energy metabolism, male fertility and cognition through the release of osteocalcin, a perfect definition-fitting hormone in its uncarboxylated state. As for the osteocytes, many evidence shows that they do not merely represent the final destination of the osteoblasts, but they are instead very active cells that, besides a mechanosensorial function, actively contribute to the bone remodelling by regulating bone formation and resorption. The regulation is exerted by the production of sclerostin (SOST), which in turn inhibits osteoblast differentiation by blocking Wnt/beta-catenin pathway. At the same time, osteocytes influence bone resorption both indirectly, by producing RANKL, which stimulates osteoclastogenesis, and directly by means of a local osteolysis, which is observed especially under pathological conditions. The great versatility of both these cells reflects the complexity of the bone tissue, which has not only a structural role, but influences and is influenced by different organs, taking part in homeostatic and adaptive responses affecting the whole organism.
Collapse
|
48
|
Hu X, Zhang P, Xu Z, Chen H, Xie X. GPNMB enhances bone regeneration by promoting angiogenesis and osteogenesis: potential role for tissue engineering bone. J Cell Biochem 2014; 114:2729-37. [PMID: 23794283 DOI: 10.1002/jcb.24621] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 06/18/2013] [Indexed: 12/13/2022]
Abstract
Bone regeneration is a coordinated process involving the connection between blood vessels and bone cells. Glycoprotein non-metastatic melanoma protein B (GPNMB) is known to be vital in bone formation. However, the effect of GPNMB on bone regeneration and the underlying molecular mechanism are still undefined. Fibroblast growth factor receptor (FGFR)-mediating signaling is pivotal in bone formation and angiogenesis. Therefore, we assessed GPNMB function as a communicating molecule between osteoblasts and angiogenesis, and the possible correlation with FGFR-1 signaling. Recombinant GPNMB dose-dependently increased the differentiation of human bone marrow stromal cells (hBMSCs) into osteoblasts, as well as the mRNA levels of osteoblasts marker alkaline phosphatase (ALP) and osteocalcin (OCN). Furthermore, these increases depended on the activation of FGFR-1 signaling, as pretreatment with FGFR-1 siRNA or its inhibitor SU5402 dramatically dampened GPNMB-induced osteogenesis. Additionally, GPNMB triggered dose-dependently the proliferation and migration of human umbilical vein endothelial cells (hUVECs), FGFR-1 phosphorylation, as well as capillary tube and vessels formation in vitro and in vivo. Blocking FGFR-1 signaling dampened GPNMB-induced angiogenic activity. Following construction of a rodent cranial defect model, scaffolds delivering GPNMB resulted in an evident increase in blood vessels and new bone formation; however, combined delivery of GPNMB and SU5402 abated these increase in defect sites. Taken together, these results suggest that GPNMB stimulates bone regeneration by inducing osteogenesis and angiogenesis via regulating FGFR-1 signaling. Consequently, our findings will clarify a new explanation about how GPNMB induces bone repair, and provide a potential target for bone regeneration therapeutics and bone engineering.
Collapse
Affiliation(s)
- Xuefeng Hu
- Department of Orthopedics, Chinese PLA 171 Hospital, Jiangxi, 332000, China
| | | | | | | | | |
Collapse
|
49
|
Liu CH, Huang TH, Hung CJ, Lai WY, Kao CT, Shie MY. The synergistic effects of fibroblast growth factor-2 and mineral trioxide aggregate on an osteogenic acceleratorin vitro. Int Endod J 2014; 47:843-53. [DOI: 10.1111/iej.12227] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 12/02/2013] [Indexed: 11/28/2022]
Affiliation(s)
- C.-H. Liu
- School of Dentistry; Chung Shan Medical University; Taichung City Taiwan
| | - T.-H. Huang
- School of Dentistry; Chung Shan Medical University; Taichung City Taiwan
- Department of Dentistry; Chung Shan Medical University Hospital; Taichung City Taiwan
| | - C.-J. Hung
- School of Dentistry; Chung Shan Medical University; Taichung City Taiwan
- Department of Dentistry; Chung Shan Medical University Hospital; Taichung City Taiwan
| | - W.-Y. Lai
- School of Dentistry; Chung Shan Medical University; Taichung City Taiwan
- Department of Dentistry; Chung Shan Medical University Hospital; Taichung City Taiwan
| | - C.-T. Kao
- School of Dentistry; Chung Shan Medical University; Taichung City Taiwan
- Department of Dentistry; Chung Shan Medical University Hospital; Taichung City Taiwan
| | - M.-Y. Shie
- Institute of Oral Science; Chung Shan Medical University; Taichung City Taiwan
| |
Collapse
|
50
|
Byun MR, Kim AR, Hwang JH, Kim KM, Hwang ES, Hong JH. FGF2 stimulates osteogenic differentiation through ERK induced TAZ expression. Bone 2014; 58:72-80. [PMID: 24125755 DOI: 10.1016/j.bone.2013.09.024] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 09/17/2013] [Accepted: 09/21/2013] [Indexed: 11/25/2022]
Abstract
TAZ (transcriptional coactivator with PDZ-binding motif) is a transcriptional modulator that regulates mesenchymal stem cell differentiation. It stimulates osteogenic differentiation while inhibiting adipocyte differentiation. FGFs (fibroblast growth factors) stimulate several signaling proteins to regulate their target genes, which are involved in cell proliferation, differentiation, and cell survival. Within this family, FGF2 stimulates osteoblast differentiation though a mechanism that is largely unknown. In this report, we show that TAZ mediates FGF2 signaling in osteogenesis. We observed that FGF2 increases TAZ expression by stimulating its mRNA expression. Depletion of TAZ using small hairpin RNA blocked FGF2-mediated osteogenic differentiation. FGF2 induced TAZ expression was stimulated by ERK (extracellular signal-regulated kinase) activation and the inhibition of ERK blocked TAZ expression. FGF2 increased nuclear localization of TAZ and, thus, facilitated the interaction of TAZ and Runx2, activating Runx2-mediated gene transcription. Taken together, these results suggest that TAZ is an important mediator of FGF2 signaling in osteoblast differentiation.
Collapse
Affiliation(s)
- Mi Ran Byun
- Department of Life Sciences, Korea University, Seoul 136-701, Republic of Korea
| | | | | | | | | | | |
Collapse
|