1
|
Castoldi G, Mauri M, D’Aliberti D, Spinelli S, Testa L, Gaverina F, Rubinacci A, Villa I, Bellelli G, Zerbini G, Piazza R, Zatti G. Transcriptomic Profile of Human Osteoblast-like Cells Grown on Trabecular Titanium. Int J Mol Sci 2025; 26:3598. [PMID: 40332083 PMCID: PMC12026869 DOI: 10.3390/ijms26083598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Trabecular titanium implants are widely used in orthopedic surgery and are known to promote osseointegration. In this study, we investigated whether primary human osteoblast-like cells grown inside a 3D trabecular titanium scaffold undergo changes in migration capacity, transcriptomic profile, and cellular phenotype as compared to the same osteoblasts not grown inside the scaffold. Scratch tests have shown that primary human osteoblast-like cells grown inside the 3D trabecular titanium scaffold promote the migration of cells from the external environment into the scaffold. Next generation sequencing analysis demonstrated that primary human osteoblast-like cells grown inside the 3D trabecular titanium scaffold modified the expression of genes involved in cell cycle and extracellular matrix remodeling, while maintaining a normal expression of the specific osteoblast markers, such as osteocalcin and osterix, as well as a comparable mineralization capacity. These data demonstrate that primary human osteoblast-like cells grown inside the titanium scaffold in a 3D environment acquire specific features favoring osseointegration.
Collapse
Affiliation(s)
- Giovanna Castoldi
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Milano-Bicocca, 20900 Monza, Italy; (M.M.); (D.D.); (S.S.); (L.T.); (F.G.); (G.B.); (R.P.); (G.Z.)
| | - Mario Mauri
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Milano-Bicocca, 20900 Monza, Italy; (M.M.); (D.D.); (S.S.); (L.T.); (F.G.); (G.B.); (R.P.); (G.Z.)
| | - Deborah D’Aliberti
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Milano-Bicocca, 20900 Monza, Italy; (M.M.); (D.D.); (S.S.); (L.T.); (F.G.); (G.B.); (R.P.); (G.Z.)
| | - Silvia Spinelli
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Milano-Bicocca, 20900 Monza, Italy; (M.M.); (D.D.); (S.S.); (L.T.); (F.G.); (G.B.); (R.P.); (G.Z.)
| | - Leonardo Testa
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Milano-Bicocca, 20900 Monza, Italy; (M.M.); (D.D.); (S.S.); (L.T.); (F.G.); (G.B.); (R.P.); (G.Z.)
- Clinica Ortopedica, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Federico Gaverina
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Milano-Bicocca, 20900 Monza, Italy; (M.M.); (D.D.); (S.S.); (L.T.); (F.G.); (G.B.); (R.P.); (G.Z.)
- Clinica Ortopedica, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | | | - Isabella Villa
- Laboratorio di Endocrinologia e Metabolismo Osseo, Istituto di Scienze Endocrine e Metaboliche, IRCCS Ospedale San Raffaele, 20132 Milano, Italy;
| | - Giuseppe Bellelli
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Milano-Bicocca, 20900 Monza, Italy; (M.M.); (D.D.); (S.S.); (L.T.); (F.G.); (G.B.); (R.P.); (G.Z.)
- Acute Geriatric Unit, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Gianpaolo Zerbini
- Unità Complicanze del Diabete, Diabetes Research Institute, IRCCS Istituto Scientifico San Raffaele, 20132 Milano, Italy;
| | - Rocco Piazza
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Milano-Bicocca, 20900 Monza, Italy; (M.M.); (D.D.); (S.S.); (L.T.); (F.G.); (G.B.); (R.P.); (G.Z.)
- Divisione di Ematologia, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Giovanni Zatti
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Milano-Bicocca, 20900 Monza, Italy; (M.M.); (D.D.); (S.S.); (L.T.); (F.G.); (G.B.); (R.P.); (G.Z.)
- Clinica Ortopedica, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| |
Collapse
|
2
|
Beltrán‐Hernández NE, Cardenas L, Jimenez‐Jacinto V, Vega‐Alvarado L, Rivera HM. Biological Activity of Biomarkers Associated With Metastasis in Osteosarcoma Cell Lines. Cancer Med 2025; 14:e70391. [PMID: 40079158 PMCID: PMC11904427 DOI: 10.1002/cam4.70391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/26/2024] [Accepted: 10/20/2024] [Indexed: 03/14/2025] Open
Abstract
INTRODUCTION Osteosarcoma, a highly aggressive bone cancer primarily affecting children and young adults, remains a significant challenge in clinical oncology. Metastasis stands as the primary cause of mortality in osteosarcoma patients. However, the mechanisms driving this process remain incompletely understood. Clarifying the molecular pathways involved in metastasis is essential for enhancing patient prognoses and facilitating the development of targeted therapeutic strategies. METHODS RNA sequencing (RNA-Seq) analysis was employed to compare three conditions, hFOB1.19 versus Saos-2, hFOB1.19 versus SJSA-1, and Saos-2 versus SJSA-1, involving non-cancer osteoblasts (hFOB1.19) and highly metastatic osteosarcoma cell lines (Saos-2 and SJSA-1). Additionally, ENA datasets of RNA-Seq from osteosarcoma biopsies were included. Differentially expressed genes (DEGs) were identified and analyzed through enrichment pathway analysis and protein-protein interaction (PPI) networks. Additionally, for gene candidates, a biochemical evaluation was performed. RESULTS DEGs associated with biological functions pertinent to migration, invasion, and metastasis in osteosarcoma were identified. Notably, matrix metalloproteinase-2 (MMP-2) emerged as a promising candidate. Both canonical or full-length (FL-mmp-2) and N-terminal truncated (NTT-mmp-2) isoforms were discerned in biopsies. Moreover, MMP-2's activity was characterized in cell lines. Additionally, mRNA expression of voltage-gated sodium channels (NaVs) and voltage-gated potassium channels (KVs) was detected, and their functional expression was validated using patch clamp techniques. Evaluation of cell line migration and invasion capacities revealed their reduction in the presence of ion channel blockers (TTX and TEA) and MMP inhibitor (GM6001). CONCLUSIONS The gene functional enrichment analysis of DEGs enabled the identification of interaction networks in osteosarcoma, thereby revealing potential biomarkers. Moreover, the elucidated co-participation of TTX-sensitive NaVs and MMP-2 in facilitating migration and invasion suggests their suitability as novel prognostic biomarkers for osteosarcoma. Additionally, this study introduces a model delineating the potential interaction mechanism among ion channels, MMP-2, and other crucial factors in the metastatic cascade of osteosarcoma.
Collapse
Affiliation(s)
| | - Luis Cardenas
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de MéxicoCuernavacaMorelosMexico
| | - Verónica Jimenez‐Jacinto
- Unidad Universitaria de Secuenciación Masiva y Bioinformática, Instituto de Biotecnología, Universidad Nacional Autónoma de MéxicoCuernavacaMorelosMexico
| | - Leticia Vega‐Alvarado
- Instituto de Ciencias Aplicadas y Tecnología, Universidad Nacional Autónoma de MéxicoCoyoacán Ciudad de MéxicoMexico
| | | |
Collapse
|
3
|
Perepletchikova D, Kuchur P, Basovich L, Khvorova I, Lobov A, Azarkina K, Aksenov N, Bozhkova S, Karelkin V, Malashicheva A. Endothelial-mesenchymal crosstalk drives osteogenic differentiation of human osteoblasts through Notch signaling. Cell Commun Signal 2025; 23:100. [PMID: 39972367 PMCID: PMC11841332 DOI: 10.1186/s12964-025-02096-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 02/08/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Angiogenesis and osteogenesis are closely interrelated. The interaction between endothelial and bone-forming cells, such as osteoblasts, is crucial for normal bone development and repair. Juxtacrine and paracrine mechanisms play key roles in cell differentiation towards the osteogenic direction, assuming the direct effect of endothelium on osteogenic differentiation. However, the mechanisms of this interplay have yet to be thoroughly studied. METHODS Isolated endothelial cells (EC) from human umbilical vein and human osteoblasts (OB) from the epiphysis of the femur or tibia were cultured in direct and indirect (separated by membrane) contact in vitro under the osteogenic differentiation conditions. Osteogenic differentiation was verified by RT-PCR, and alizarin red staining. Shotgun proteomics and RNA-sequencing were used to compare both EC and OB under different co-culture conditions to assess the mechanisms of EC-OB interplay. To verify the role of Notch signaling, experiments with Notch modulation in EC were performed by EC lentiviral transduction with further co-cultivation with OB. Additionally, the effect of Notch modulation in EC was assessed by RNA-sequencing. RESULTS EC have opposite effects on osteogenic differentiation depending on the co-culture conditions with OB. In direct contact, EC enhance osteogenic differentiation, but in indirect cultures, EC suppress it. Our proteotranscriptomic analysis revealed that the osteosuppressive effect is related to the action of paracrine factors secreted by EC, while the osteoinductive properties of EC are mediated by the Notch signaling pathway, which can be activated only upon a physical contact of EC with OB. Indeed, in the direct co-culture, the knockdown of Notch1 and Notch3 receptors in EC has an inhibitory effect on the OB osteogenic differentiation, whereas activation of Notch by intracellular domain of either Notch1 or Notch3 in EC has an inductive effect on the OB osteogenic differentiation. CONCLUSION The data indicate the dual role of the endothelium in regulating osteogenic differentiation and highlight the unique role of the Notch signaling pathway in inducing osteogenic differentiation during cell-to-cell interactions. The findings of the study emphasize the importance of intercellular communication in the regulation of osteoblast differentiation during bone development and maintenance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Svetlana Bozhkova
- Vreden National Medical Research Center of Traumatology and Orthopedics, Saint- Petersburg, Russia
| | - Vitaliy Karelkin
- Vreden National Medical Research Center of Traumatology and Orthopedics, Saint- Petersburg, Russia
| | | |
Collapse
|
4
|
Hojo H, Tani S, Ohba S. Modeling of skeletal development and diseases using human pluripotent stem cells. J Bone Miner Res 2024; 40:5-19. [PMID: 39498496 DOI: 10.1093/jbmr/zjae178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/28/2024] [Accepted: 11/02/2024] [Indexed: 01/07/2025]
Abstract
Human skeletal elements are formed from distinct origins at distinct positions of the embryo. For example, the neural crest produces the facial bones, the paraxial mesoderm produces the axial skeleton, and the lateral plate mesoderm produces the appendicular skeleton. During skeletal development, different combinations of signaling pathways are coordinated from distinct origins during the sequential developmental stages. Models for human skeletal development have been established using human pluripotent stem cells (hPSCs) and by exploiting our understanding of skeletal development. Stepwise protocols for generating skeletal cells from different origins have been designed to mimic developmental trails. Recently, organoid methods have allowed the multicellular organization of skeletal cell types to recapitulate complicated skeletal development and metabolism. Similarly, several genetic diseases of the skeleton have been modeled using patient-derived induced pluripotent stem cells and genome-editing technologies. Model-based drug screening is a powerful tool for identifying drug candidates. This review briefly summarizes our current understanding of the embryonic development of skeletal tissues and introduces the current state-of-the-art hPSC methods for recapitulating skeletal development, metabolism, and diseases. We also discuss the current limitations and future perspectives for applications of the hPSC-based modeling system in precision medicine in this research field.
Collapse
Affiliation(s)
- Hironori Hojo
- Division of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8655, Japan
| | - Shoichiro Tani
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Shinsuke Ohba
- Department of Tissue and Developmental Biology, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
5
|
Hamai R, Tsuchiya K, Suzuki O. Enhancement of the Cytokine Adsorption Capacity of Octacalcium Phosphate by Interaction-Controlled Glycosaminoglycan Modification to Promote Osteoblastic Differentiation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:27253-27269. [PMID: 39694685 DOI: 10.1021/acs.langmuir.4c03067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
This study was designed to investigate how the strength of the interaction between octacalcium phosphate (OCP) and modified chondroitin-A sulfate (CS-A), a glycosaminoglycan, regulates the adsorption-desorption of cytokines and subsequently affects the osteoblastic differentiation of mesenchymal stem cells (MSCs) in vitro. The utilization of cytokines produced by cells, such as macrophages, stimulated by the hydrolysis of OCP, is expected to enhance the bone regeneration capacity of the OCP. CS-Na was used to modify CS-A on the OCP immobilized with the amino group through electrostatic interactions. On the other hand, N-hydroxysuccinimide (NHS)-esterified CS-A was used to form the covalent bond between CS-A and the amino group on the surface of OCP. X-ray diffraction and Raman spectroscopy indicated that the CS-A-modified OCP maintained its structure, regardless of the modification process. The remaining ratio of the modified CS-A in the buffer suggests that increasing the immobilized density of the amino group and the modification using NHS ester may enhance interaction strength between OCP and CS-A. The adsorption amount and retention rate of recombinant human bone morphological protein-2 (rhBMP-2, an endogenous cytokine model) increased onto CS-A-modified OCP under physiological conditions when the interaction strength between OCP and the protein was stronger. The higher interaction strength between the OCP and CS-A could be associated with the enhanced adsorption affinity for the lower-molecular-weight basic protein. The alkaline phosphatase activity of MSCs increased depending on the remaining rate of rhBMP-2 adsorption on the CS-A-modified-OCP. Fourier transform infrared spectroscopy and chemical analysis indicated that the hydrolysis of the OCP progressed regardless of CS-A modification after MSC incubation. The present study suggests that stronger interactions between the OCP and glycosaminoglycans could contribute to the capture and retention of endogenous cytokines on the surface to further promote osteoblastic differentiation under the chemical environment induced by the hydrolysis of the OCP.
Collapse
Affiliation(s)
- Ryo Hamai
- Division of Craniofacial Function Engineering (Division of Biomaterials Science and Engineering), Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Kaori Tsuchiya
- Division of Craniofacial Function Engineering (Division of Biomaterials Science and Engineering), Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Osamu Suzuki
- Division of Craniofacial Function Engineering (Division of Biomaterials Science and Engineering), Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| |
Collapse
|
6
|
Gu C, Tang Q, Li L, Chen Y. Optimization and Implication of Adipose-Derived Stem Cells in Craniofacial Bone Regeneration and Repair. Bioengineering (Basel) 2024; 11:1100. [PMID: 39593759 PMCID: PMC11592193 DOI: 10.3390/bioengineering11111100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/17/2024] [Accepted: 10/27/2024] [Indexed: 11/28/2024] Open
Abstract
Adipose-derived stem cells (ADSCs) have emerged as a promising resource for craniofacial bone regeneration due to their high abundance and easy accessibility, significant osteogenic potential, versatile applications, and potential for personalized medicine, which underscore their importance in this field. This article reviews the current progress of preclinical studies that describe the careful selection of specific ADSC subpopulations, key signaling pathways involved, and usage of various strategies to enhance the osteogenic potential of ADSCs. Additionally, clinical case reports regarding the application of ADSCs in the repair of calvarial defects, cranio-maxillofacial defects, and alveolar bone defects are also discussed.
Collapse
Affiliation(s)
- Cong Gu
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA; (Q.T.); (L.L.); (Y.C.)
| | - Qinghuang Tang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA; (Q.T.); (L.L.); (Y.C.)
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY 14214, USA
| | - Liwen Li
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA; (Q.T.); (L.L.); (Y.C.)
- Department of Biological Sciences, University at Buffalo, Buffalo, NY 14260, USA
| | - YiPing Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA; (Q.T.); (L.L.); (Y.C.)
| |
Collapse
|
7
|
Sexton B, Han Y, Dal-Fabbro R, Xu J, Kaigler D, Bottino MC. The role of fibroblast growth factor-2 in modulating the differentiation of periodontal ligament and alveolar bone-derived stem cells. Arch Oral Biol 2024; 165:106027. [PMID: 38870610 DOI: 10.1016/j.archoralbio.2024.106027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/15/2024]
Abstract
OBJECTIVE This study examined how range concentrations of Fibroblast Growth Factor-2 (FGF-2) influence the differentiation and activity of human-derived periodontal ligament (hPDLSCs) and alveolar bone-derived stem cells (haBMSCs). DESIGN hPDLSCs and haBMSCs were cultured with varying concentrations of FGF-2 (0, 1, 2.5, 5, 10, 20 ng/mL) and monitored for osteogenic differentiation through alkaline phosphatase (ALP) activity and quantification of gene expression (qRT-PCR) for osteogenesis markers. Additionally, alizarin red staining and a hydroxyproline colorimetric assay evaluated and quantified osteogenic matrix mineralization and collagen deposition. Statistical analyses were performed using one-way ANOVA or two-way ANOVA for multiple comparisons between groups. RESULTS At low FGF-2 concentrations, hPDLSCs differentiated toward an osteogenic lineage, whereas higher concentrations of FGF-2 inhibited osteogenesis and promoted fibroblastic differentiation. The effect of FGF-2 at the lowest concentration tested (1 ng/mL) led to significantly higher ALP activity than osteogenically induced positive controls at early time points and equivalent RUNX2 expression at early and later time points. FGF-2 supplementation of haBMSC cultures was sufficient, at all concentrations, to increase ALP activity at an earlier time point. Mineralization of haBMSC cultures increased significantly within 5-20 ng/mL FGF-2 concentrations under basal growth media conditions (α-minimal essential medium supplemented with 15 % fetal bovine serum and 1 % penicillin/streptomycin). CONCLUSIONS FGF-2 has a dual capacity in promoting osteogenic and fibroblastic differentiation within hPDLSCs contingent upon the dosage and timing of administration, alongside supporting osteogenic differentiation in haBMSCs. These findings underscore the need for precision growth factors dosing when considering the design of biomaterials for periodontal regeneration.
Collapse
Affiliation(s)
- Benjamin Sexton
- Department of Biologic and Materials Science, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Yuanyuan Han
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Renan Dal-Fabbro
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Jinping Xu
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Darnell Kaigler
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, United States; Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Marco C Bottino
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States; Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
8
|
Dong Y, Yuan H, Ma G, Cao H. Bone-muscle crosstalk under physiological and pathological conditions. Cell Mol Life Sci 2024; 81:310. [PMID: 39066929 PMCID: PMC11335237 DOI: 10.1007/s00018-024-05331-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/30/2024]
Abstract
Anatomically connected bones and muscles determine movement of the body. Forces exerted on muscles are then turned to bones to promote osteogenesis. The crosstalk between muscle and bone has been identified as mechanotransduction previously. In addition to the mechanical features, bones and muscles are also secretory organs which interact closely with one another through producing myokines and osteokines. Moreover, besides the mechanical features, other factors, such as nutrition metabolism, physiological rhythm, age, etc., also affect bone-muscle crosstalk. What's more, osteogenesis and myogenesis within motor system occur almost in parallel. Pathologically, defective muscles are always detected in bone associated diseases and induce the osteopenia, inflammation and abnormal bone metabolism, etc., through biomechanical or biochemical coupling. Hence, we summarize the study findings of bone-muscle crosstalk and propose potential strategies to improve the skeletal or muscular symptoms of certain diseases. Altogether, functional improvement of bones or muscles is beneficial to each other within motor system.
Collapse
Affiliation(s)
- Yuechao Dong
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Hongyan Yuan
- Shenzhen Key Laboratory of Soft Mechanics & Smart Manufacturing, Department of Mechanics and Aerospace Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Guixing Ma
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Huiling Cao
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
9
|
Zhu S, Chen W, Masson A, Li YP. Cell signaling and transcriptional regulation of osteoblast lineage commitment, differentiation, bone formation, and homeostasis. Cell Discov 2024; 10:71. [PMID: 38956429 PMCID: PMC11219878 DOI: 10.1038/s41421-024-00689-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 05/04/2024] [Indexed: 07/04/2024] Open
Abstract
The initiation of osteogenesis primarily occurs as mesenchymal stem cells undergo differentiation into osteoblasts. This differentiation process plays a crucial role in bone formation and homeostasis and is regulated by two intricate processes: cell signal transduction and transcriptional gene expression. Various essential cell signaling pathways, including Wnt, BMP, TGF-β, Hedgehog, PTH, FGF, Ephrin, Notch, Hippo, and Piezo1/2, play a critical role in facilitating osteoblast differentiation, bone formation, and bone homeostasis. Key transcriptional factors in this differentiation process include Runx2, Cbfβ, Runx1, Osterix, ATF4, SATB2, and TAZ/YAP. Furthermore, a diverse array of epigenetic factors also plays critical roles in osteoblast differentiation, bone formation, and homeostasis at the transcriptional level. This review provides an overview of the latest developments and current comprehension concerning the pathways of cell signaling, regulation of hormones, and transcriptional regulation of genes involved in the commitment and differentiation of osteoblast lineage, as well as in bone formation and maintenance of homeostasis. The paper also reviews epigenetic regulation of osteoblast differentiation via mechanisms, such as histone and DNA modifications. Additionally, we summarize the latest developments in osteoblast biology spurred by recent advancements in various modern technologies and bioinformatics. By synthesizing these insights into a comprehensive understanding of osteoblast differentiation, this review provides further clarification of the mechanisms underlying osteoblast lineage commitment, differentiation, and bone formation, and highlights potential new therapeutic applications for the treatment of bone diseases.
Collapse
Affiliation(s)
- Siyu Zhu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| | - Alasdair Masson
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
10
|
Salamanna F, Faldini C, Veronesi F, Borsari V, Ruffilli A, Manzetti M, Viroli G, Traversari M, Marchese L, Fini M, Giavaresi G. A Pilot Study on Circulating, Cellular, and Tissue Biomarkers in Osteosarcopenic Patients. Int J Mol Sci 2024; 25:5879. [PMID: 38892069 PMCID: PMC11172451 DOI: 10.3390/ijms25115879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/23/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
Aging comes with the loss of muscle and bone mass, leading to a condition known as osteosarcopenia. Circulating, cellular, and tissue biomarkers research for osteosarcopenia is relatively scarce and, currently, no established biomarkers exist. Here we find that osteosarcopenic patients exhibited elevated basophils and TNFα levels, along with decreased aPPT, PT/INR, IL15, alpha-Klotho, DHEA-S, and FGF-2 expression and distinctive bone and muscle tissue micro-architecture and biomarker expressions. They also displayed an increase in osteoclast precursors with a concomitant imbalance towards spontaneous osteoclastogenesis. Similarities were noted with osteopenic and sarcopenic patients, including a lower neutrophil percentage and altered cytokine expression. A linear discriminant analysis (LDA) on models based on selected biomarkers showed a classification accuracy in the range of 61-78%. Collectively, our data provide compelling evidence for novel biomarkers for osteosarcopenia that may hold potential as diagnostic tools to promote healthy aging.
Collapse
Affiliation(s)
- Francesca Salamanna
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (F.S.); (V.B.); (L.M.); (G.G.)
| | - Cesare Faldini
- 1st Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (C.F.); (A.R.); (M.M.); (G.V.); (M.T.)
- Department of Biomedical and Neuromotor Science (DIBINEM), University of Bologna, Via Zamboni 33, 40126 Bologna, Italy
| | - Francesca Veronesi
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (F.S.); (V.B.); (L.M.); (G.G.)
| | - Veronica Borsari
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (F.S.); (V.B.); (L.M.); (G.G.)
| | - Alberto Ruffilli
- 1st Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (C.F.); (A.R.); (M.M.); (G.V.); (M.T.)
- Department of Biomedical and Neuromotor Science (DIBINEM), University of Bologna, Via Zamboni 33, 40126 Bologna, Italy
| | - Marco Manzetti
- 1st Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (C.F.); (A.R.); (M.M.); (G.V.); (M.T.)
- Department of Biomedical and Neuromotor Science (DIBINEM), University of Bologna, Via Zamboni 33, 40126 Bologna, Italy
| | - Giovanni Viroli
- 1st Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (C.F.); (A.R.); (M.M.); (G.V.); (M.T.)
- Department of Biomedical and Neuromotor Science (DIBINEM), University of Bologna, Via Zamboni 33, 40126 Bologna, Italy
| | - Matteo Traversari
- 1st Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (C.F.); (A.R.); (M.M.); (G.V.); (M.T.)
| | - Laura Marchese
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (F.S.); (V.B.); (L.M.); (G.G.)
| | - Milena Fini
- Scientific Direction, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
| | - Gianluca Giavaresi
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (F.S.); (V.B.); (L.M.); (G.G.)
| |
Collapse
|
11
|
Rifai A, Weerasinghe DK, Tilaye GA, Nisbet D, Hodge JM, Pasco JA, Williams LJ, Samarasinghe RM, Williams RJ. Biofabrication of functional bone tissue: defining tissue-engineered scaffolds from nature. Front Bioeng Biotechnol 2023; 11:1185841. [PMID: 37614632 PMCID: PMC10444209 DOI: 10.3389/fbioe.2023.1185841] [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: 03/14/2023] [Accepted: 07/24/2023] [Indexed: 08/25/2023] Open
Abstract
Damage to bone leads to pain and loss of movement in the musculoskeletal system. Although bone can regenerate, sometimes it is damaged beyond its innate capacity. Research interest is increasingly turning to tissue engineering (TE) processes to provide a clinical solution for bone defects. Despite the increasing biomimicry of tissue-engineered scaffolds, significant gaps remain in creating the complex bone substitutes, which include the biochemical and physical conditions required to recapitulate bone cells' natural growth, differentiation and maturation. Combining advanced biomaterials with new additive manufacturing technologies allows the development of 3D tissue, capable of forming cell aggregates and organoids based on natural and stimulated cues. Here, we provide an overview of the structure and mechanical properties of natural bone, the role of bone cells, the remodelling process, cytokines and signalling pathways, causes of bone defects and typical treatments and new TE strategies. We highlight processes of selecting biomaterials, cells and growth factors. Finally, we discuss innovative tissue-engineered models that have physiological and anatomical relevance for cancer treatments, injectable stimuli gels, and other therapeutic drug delivery systems. We also review current challenges and prospects of bone TE. Overall, this review serves as guide to understand and develop better tissue-engineered bone designs.
Collapse
Affiliation(s)
- Aaqil Rifai
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - D. Kavindi Weerasinghe
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Gebreselassie Addisu Tilaye
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - David Nisbet
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC, Australia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Melbourne, VIC, Australia
- Melbourne Medical School, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Melbourne, VIC, Australia
- Laboratory of Advanced Biomaterials, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Aikenhead Centre for Medical Discovery, St. Vincent’s Hospital, Melbourne, VIC, Australia
| | - Jason M. Hodge
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- Barwon Health, Geelong, VIC, Australia
| | - Julie A. Pasco
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- Barwon Health, Geelong, VIC, Australia
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia
- Department of Medicine-Western Health, The University of Melbourne, St Albans, VIC, Australia
| | - Lana J. Williams
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- Barwon Health, Geelong, VIC, Australia
| | - Rasika M. Samarasinghe
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Richard J. Williams
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC, Australia
- Aikenhead Centre for Medical Discovery, St. Vincent’s Hospital, Melbourne, VIC, Australia
| |
Collapse
|
12
|
Takayama T, Imamura K, Yamano S. Growth Factor Delivery Using a Collagen Membrane for Bone Tissue Regeneration. Biomolecules 2023; 13:biom13050809. [PMID: 37238679 DOI: 10.3390/biom13050809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/01/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
The use of biomaterials and bioactive agents has shown promise in bone defect repair, leading to the development of strategies for bone regeneration. Various artificial membranes, especially collagen membranes (CMs) that are widely used for periodontal therapy and provide an extracellular matrix-simulating environment, play a significant role in promoting bone regeneration. In addition, numerous growth factors (GFs) have been used as clinical applications in regenerative therapy. However, it has been established that the unregulated administration of these factors may not work to their full regenerative potential and could also trigger unfavorable side effects. The utilization of these factors in clinical settings is still restricted due to the lack of effective delivery systems and biomaterial carriers. Hence, considering the efficiency of bone regeneration, both spaces maintained using CMs and GFs can synergistically create successful outcomes in bone tissue engineering. Therefore, recent studies have demonstrated a significant interest in the potential of combining CMs and GFs to effectively promote bone repair. This approach holds great promise and has become a focal point in our research. The purpose of this review is to highlight the role of CMs containing GFs in the regeneration of bone tissue, and to discuss their use in preclinical animal models of regeneration. Additionally, the review addresses potential concerns and suggests future research directions for growth factor therapy in the field of regenerative science.
Collapse
Affiliation(s)
- Tadahiro Takayama
- Department of Periodontology, Nihon University School of Dentistry, Tokyo 101-8310, Japan
- Division of Advanced Dental Treatment, Dental Research Center, Nihon University School of Dentistry, Tokyo 101-8310, Japan
| | - Kentaro Imamura
- Department of Periodontology, Tokyo Dental College, Tokyo 101-0061, Japan
- Oral Health Science Center, Tokyo Dental College, Tokyo 101-0061, Japan
| | - Seiichi Yamano
- Department of Prosthodontics, New York University College of Dentistry, New York, NY 10010, USA
| |
Collapse
|
13
|
Xiao L, Shiwaku Y, Hamai R, Baba K, Tsuchiya K, Imazato S, Sasaki K, Suzuki O. Osteogenic capacity of octacalcium phosphate involving macrophage polarization. J Biomed Mater Res A 2022; 111:1006-1020. [PMID: 36573692 DOI: 10.1002/jbm.a.37484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 11/16/2022] [Accepted: 12/08/2022] [Indexed: 12/28/2022]
Abstract
Previous research has found that octacalcium phosphate (OCP) increases macrophage accumulation and alters the initial inflammatory response. However, the role of the immune response induced by OCP in osteogenesis remains unknown. This study investigated the behavior of macrophages and bone regeneration capacity during the early inflammatory stage of OCP-mediated osteogenesis. To assess the change in macrophage polarization and osteogenic capacity, we used a standardized rat defect model filled with OCP or calcium-deficient hydroxyapatite (CDHA)-a material obtained through the hydrolysis of the original OCP. OCP or CDHA granules were incubated with RAW264 cells for 5 days to investigate the effect of physicochemical characteristics on macrophage cytokine/chemokine expression in vitro. Our in vivo results show that due to the OCP implantation, macrophages in the rat tibial defect area tend to polarize to the M2 phenotype (anti-inflammatory) and inhibit the formation of the M1 phenotype (pro-inflammatory). In comparison to CDHA, OCP exhibited superior bone regeneration potential due to its rapid promotion of cortical bone healing and stimulation of macrophage-related growth factors. Furthermore, our in vitro results have shown that OCP regulates the expression of macrophage chemokines over time. Compared to incubation with CDHA, incubation with OCP caused changes in the ionic microenvironment. These findings suggest that the OCP-mediated macrophage polarization and secretion profile not only regulate immune function but also positively affect osteogenesis.
Collapse
Affiliation(s)
- Linghao Xiao
- Division of Craniofacial Function Engineering Tohoku University Graduate School of Dentistry Sendai Japan
- Division of Advanced Prosthetic Dentistry Tohoku University Graduate School of Dentistry Sendai Japan
- Department of Advanced Functional Materials Science Osaka University Graduate School of Dentistry Suita Japan
| | - Yukari Shiwaku
- Division of Craniofacial Function Engineering Tohoku University Graduate School of Dentistry Sendai Japan
| | - Ryo Hamai
- Division of Craniofacial Function Engineering Tohoku University Graduate School of Dentistry Sendai Japan
| | - Kazuyoshi Baba
- Department of Orthopedic Surgery Tohoku University Graduate School of Medicine Sendai Japan
| | - Kaori Tsuchiya
- Division of Craniofacial Function Engineering Tohoku University Graduate School of Dentistry Sendai Japan
| | - Satoshi Imazato
- Department of Biomaterials Science Osaka University Graduate School of Dentistry Suita Japan
| | - Keiichi Sasaki
- Division of Advanced Prosthetic Dentistry Tohoku University Graduate School of Dentistry Sendai Japan
| | - Osamu Suzuki
- Division of Craniofacial Function Engineering Tohoku University Graduate School of Dentistry Sendai Japan
| |
Collapse
|
14
|
Vlashi R, Zhang X, Wu M, Chen G. Wnt signaling: essential roles in osteoblast differentiation, bone metabolism and therapeutic implications for bone and skeletal disorders. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
|
15
|
Becerikli M, Reinkemeier F, Dadras M, Wallner C, Wagner JM, Drysch M, Sogorski A, von Glinski M, Lehnhardt M, Hahn SA, Behr B. TGF-beta pathway inhibition as the therapeutic acceleration of diabetic bone regeneration. J Orthop Res 2022; 40:1810-1826. [PMID: 34775640 DOI: 10.1002/jor.25212] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/10/2021] [Accepted: 10/30/2021] [Indexed: 02/04/2023]
Abstract
Bone regeneration and fracture healing are impaired in diabetic patients due to defective functions of associated cells. Thus, the search for molecular causes and new treatment strategies are of particular clinical relevance. We investigated the gene expression profile of bones from type 2 diabetic (db- /db- ) mice and wild-type (wt) mice by comparative microarray analyses before and after placing tibial defects and examined the expression of several osteogenesis- and osteoclastogenesis-related markers by quantitative real-time polymerase chain reaction. In regenerating wt bones, pathways related to, for example, inhibition of matrix metalloproteases were activated, whereas in db- /db- bones activation of pathways related to, for example, osteoarthritis, transforming growth factor-beta (Tgfb), or hypoxia-inducible factor 1a were detected during regeneration. We defined the Tgfb pathway as a potential therapeutic target and locally applied a single dose (0.5 µg) of the Tgfb 1, 2, and 3 neutralizing antibody 1D11 on tibial defects in db- /db- mice (n = 7). Seven days postoperation, histological and immunohistochemical stainings were performed. Decreased bone regeneration, osteogenic differentiation, osteoclast invasion, and angiogenesis in db- /db- mice were significantly restored by local 1D11 application in comparison to the phosphate-buffered saline controls. Thus, local treatment of db- /db- bony defects with Tgfb neutralizing antibody 1D11 might be considered a good candidate for the successful acceleration of bone regeneration.
Collapse
Affiliation(s)
- Mustafa Becerikli
- Department of Plastic and Reconstructive Surgery, BG University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Felix Reinkemeier
- Department of Plastic and Reconstructive Surgery, BG University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Mehran Dadras
- Department of Plastic and Reconstructive Surgery, BG University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Christoph Wallner
- Department of Plastic and Reconstructive Surgery, BG University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Johannes M Wagner
- Department of Plastic and Reconstructive Surgery, BG University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Marius Drysch
- Department of Plastic and Reconstructive Surgery, BG University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Alexander Sogorski
- Department of Plastic and Reconstructive Surgery, BG University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Maxi von Glinski
- Department of Plastic and Reconstructive Surgery, BG University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Marcus Lehnhardt
- Department of Plastic and Reconstructive Surgery, BG University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Stephan A Hahn
- Department of Molecular GI-Oncology (MGO), Clinical Research Center (ZKF), Ruhr-University Bochum, Bochum, Germany
| | - Björn Behr
- Department of Plastic and Reconstructive Surgery, BG University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
16
|
Guo X, Lu H, Liu C, Zhang Y, Bi L. Effects of Super-Activated Platelet Lysate on Early Healing of Tooth Extraction Sockets in Rats. Drug Des Devel Ther 2022; 16:2213-2227. [PMID: 35860524 PMCID: PMC9289175 DOI: 10.2147/dddt.s363766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/30/2022] [Indexed: 12/28/2022] Open
Abstract
Purpose To evaluate the effect of super-activated platelet lysate (sPL) on wound healing of tooth extraction sockets in rats. Methods Rat models of the tooth extraction socket were established. Thirty-six rats were divided into control and sPL groups and sacrificed on days 7, 14, and 28 after tooth extraction. Bone formation in tooth extraction sockets were observed by microscopic computed tomography (micro-CT) and hematoxylin and eosin (HE) staining; osteoprotegerin (OPG), receptor activator of nuclear factor kappa-Β ligand (RANKL), interleukin 6(IL-6), and tumor necrosis factor-alpha (TNF-α) proteins were detected by immunohistochemistry; and chemokine and osteogenic gene expressions were detected by polymerase chain reaction (PCR). Results sPL accelerated soft tissue wound healing in the extraction socket of rats. Micro-CT showed that the amount of bone formation and bone volume fraction were higher in the sPL group than the control 14 days after extraction. HE staining showed promotion of the formation of bony trabeculae by sPL in the apical third of the extraction socket 7 days after extraction and more mature and organized bony trabeculae in the sPL group than the control 14 days after extraction; mature bony trabeculae filling most of the fossa with lesser bone porosity in the socket in the sPL group than the control 28 days after extraction. Immunohistochemistry showed that sPL induced OPG expressions 7 and 14 days after tooth extraction but did not affect the RANKL expression while transiently promoting the IL-6 expression 7 days after extraction. PCR showed that sPL promoted chemokine expressions 7 and 14 days after extraction. The expressions of osteogenesis-related factors were higher in the sPL group than the control 7 and 28 days after extraction, while the opposite trend was observed 14 days after extraction. Conclusion sPL has a transient pro-inflammatory effect and promotes soft tissue healing and bone formation during early wound healing of extraction sockets in rats.
Collapse
Affiliation(s)
- Xiaorui Guo
- Department of Stomatology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, 150001, People's Republic of China.,State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of Oral Diseases, Department of Operative Dentistry and Endodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Huiying Lu
- National and Local Joint Stem Cell Research & Engineering Center for Aging Diseases, Tian Qing Stem Cell Co., Ltd, Harbin, 150028, People's Republic of China
| | - Chunxiang Liu
- National and Local Joint Stem Cell Research & Engineering Center for Aging Diseases, Tian Qing Stem Cell Co., Ltd, Harbin, 150028, People's Republic of China
| | - Yi Zhang
- National and Local Joint Stem Cell Research & Engineering Center for Aging Diseases, Tian Qing Stem Cell Co., Ltd, Harbin, 150028, People's Republic of China
| | - Liangjia Bi
- Department of Stomatology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, 150001, People's Republic of China
| |
Collapse
|
17
|
Li J, Ma J, Zhang Q, Gong H, Gao D, Wang Y, Li B, Li X, Zheng H, Wu Z, Zhu Y, Leng L. Spatially resolved proteomic map shows that extracellular matrix regulates epidermal growth. Nat Commun 2022; 13:4012. [PMID: 35817779 PMCID: PMC9273758 DOI: 10.1038/s41467-022-31659-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 06/28/2022] [Indexed: 02/07/2023] Open
Abstract
Human skin comprises stratified squamous epithelium and dermis with various stromal cells and the extracellular matrix (ECM). The basement membrane (BM), a thin layer at the top of the dermis, serves as a unique niche for determining the fate of epidermal stem cells (EpSCs) by transmitting physical and biochemical signals to establish epidermal cell polarity and maintain the hierarchical structure and function of skin tissue. However, how stem cell niches maintain tissue homeostasis and control wound healing by regulating the behavior of EpSCs is still not completely understood. In this study, a hierarchical skin proteome map is constructed using spatial quantitative proteomics combined with decellularization, laser capture microdissection, and mass spectrometry. The specific functions of different structures of normal native skin tissues or tissues with a dermatologic disease are analyzed in situ. Transforming growth factor-beta (TGFβ)-induced protein ig-h3 (TGFBI), an ECM glycoprotein, in the BM is identified that could enhance the growth and function of EpSCs and promote wound healing. Our results provide insights into the way in which ECM proteins facilitate the growth and function of EpSCs as part of an important niche. The results may benefit the clinical treatment of skin ulcers or diseases with refractory lesions that involve epidermal cell dysfunction and re-epithelialization block in the future. Ling Leng et al. construct a hierarchical skin proteome map and identify an extracellular matrix glycoprotein TGFBI, which is located in basement membrane and could enhance the growth and function of epidermal stem cells and promote wound healing.
Collapse
Affiliation(s)
- Jun Li
- Department of Dermatology and Venereology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jie Ma
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Qiyu Zhang
- Department of Dermatology and Venereology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Stem Cell and Regenerative Medicine Lab, State Key Laboratory of Complex Severe and Rare Diseases, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huizi Gong
- Department of Dermatology and Venereology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Stem Cell and Regenerative Medicine Lab, State Key Laboratory of Complex Severe and Rare Diseases, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dunqin Gao
- Stem Cell and Regenerative Medicine Lab, State Key Laboratory of Complex Severe and Rare Diseases, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yujie Wang
- Stem Cell and Regenerative Medicine Lab, State Key Laboratory of Complex Severe and Rare Diseases, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Biyou Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.,Basic Medical School, Anhui Medical University, Anhui, China
| | - Xiao Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Heyi Zheng
- Department of Dermatology and Venereology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhihong Wu
- Stem Cell and Regenerative Medicine Lab, State Key Laboratory of Complex Severe and Rare Diseases, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yunping Zhu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China. .,Basic Medical School, Anhui Medical University, Anhui, China.
| | - Ling Leng
- Stem Cell and Regenerative Medicine Lab, State Key Laboratory of Complex Severe and Rare Diseases, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
18
|
Humphreys PA, Mancini FE, Ferreira MJS, Woods S, Ogene L, Kimber SJ. Developmental principles informing human pluripotent stem cell differentiation to cartilage and bone. Semin Cell Dev Biol 2022; 127:17-36. [PMID: 34949507 DOI: 10.1016/j.semcdb.2021.11.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 12/14/2022]
Abstract
Human pluripotent stem cells can differentiate into any cell type given appropriate signals and hence have been used to research early human development of many tissues and diseases. Here, we review the major biological factors that regulate cartilage and bone development through the three main routes of neural crest, lateral plate mesoderm and paraxial mesoderm. We examine how these routes have been used in differentiation protocols that replicate skeletal development using human pluripotent stem cells and how these methods have been refined and improved over time. Finally, we discuss how pluripotent stem cells can be employed to understand human skeletal genetic diseases with a developmental origin and phenotype, and how developmental protocols have been applied to gain a better understanding of these conditions.
Collapse
Affiliation(s)
- Paul A Humphreys
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK; Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering & Henry Royce Institute, University of Manchester, UK
| | - Fabrizio E Mancini
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK
| | - Miguel J S Ferreira
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK; Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering & Henry Royce Institute, University of Manchester, UK
| | - Steven Woods
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK
| | - Leona Ogene
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK
| | - Susan J Kimber
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK
| |
Collapse
|
19
|
Cell-Based Double-Screening Method to Identify a Reliable Candidate for Osteogenesis-Targeting Compounds. Biomedicines 2022; 10:biomedicines10020426. [PMID: 35203635 PMCID: PMC8962348 DOI: 10.3390/biomedicines10020426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 02/05/2022] [Accepted: 02/07/2022] [Indexed: 01/15/2023] Open
Abstract
Small-molecule compounds strongly affecting osteogenesis can form the basis of effective therapeutic strategies in bone regenerative medicine. A cell-based high-throughput screening system might be a powerful tool for identifying osteoblast-targeting candidates; however, this approach is generally limited with using only one molecule as a cell-based sensor that does not always reflect the activation of the osteogenic phenotype. In the present study, we used the MC3T3-E1 cell line stably transfected with the green fluorescent protein (GFP) reporter gene driven by a fragment of type I collagen promoter (Col-1a1GFP-MC3T3-E1) to evaluate a double-screening system to identify osteogenic inducible compounds using a combination of a cell-based reporter assay and detection of alkaline phosphatase (ALP) activity. Col-1a1GFP-MC3T3-E1 cells were cultured in an osteogenic induction medium after library screening of 1280 pharmacologically active compounds (Lopack1280). After 7 days, GFP fluorescence was measured using a microplate reader. After 14 days of osteogenic induction, the cells were stained with ALP. Library screening using the Col-1a1/GFP reporter and ALP staining assay detected three candidates with significant osteogenic induction ability. Furthermore, leflunomide, one of the three detected candidates, significantly promoted new bone formation in vivo. Therefore, this double-screening method could identify candidates for osteogenesis-targeting compounds more reliably than conventional methods.
Collapse
|
20
|
Chen Y, Zhou Y, Lin J, Zhang S. Challenges to Improve Bone Healing Under Diabetic Conditions. Front Endocrinol (Lausanne) 2022; 13:861878. [PMID: 35418946 PMCID: PMC8996179 DOI: 10.3389/fendo.2022.861878] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/02/2022] [Indexed: 12/17/2022] Open
Abstract
Diabetes mellitus (DM) can affect bone metabolism and the bone microenvironment, resulting in impaired bone healing. The mechanisms include oxidative stress, inflammation, the production of advanced glycation end products (AGEs), etc. Improving bone healing in diabetic patients has important clinical significance in promoting fracture healing and improving bone integration. In this paper, we reviewed the methods of improving bone healing under diabetic conditions, including drug therapy, biochemical cues, hyperbaric oxygen, ultrasound, laser and pulsed electromagnetic fields, although most studies are in preclinical stages. Meanwhile, we also pointed out some shortcomings and challenges, hoping to provide a potential therapeutic strategy for accelerating bone healing in patients with diabetes.
Collapse
Affiliation(s)
- Yiling Chen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yue Zhou
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jie Lin
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Jie Lin, ; Shiwen Zhang,
| | - Shiwen Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Jie Lin, ; Shiwen Zhang,
| |
Collapse
|
21
|
Nirwana I, Munadziroh E, Yuliati A, Fadhila AI, Nurliana, Wardhana AS, Shariff KA, Surboyo MDC. Ellagic acid and hydroxyapatite promote angiogenesis marker in bone defect. J Oral Biol Craniofac Res 2022; 12:116-120. [PMID: 34840942 PMCID: PMC8605383 DOI: 10.1016/j.jobcr.2021.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/19/2021] [Accepted: 11/08/2021] [Indexed: 11/20/2022] Open
Abstract
The combination of hydroxyapatite and the herbal extract ellagic acid is expected to accelerate the bone healing process (osteogenesis) due to the extract's anti-inflammatory and antioxidant properties. The osteogenesis process is closely associated with angiogenesis markers, such as fibroblast growth factor 2 (FGF-2), vascular endothelial growth factor (VEGF) and alkali phosphatase (ALP). The objective of this study is to analyse the combination of ellagic acid and hydroxyapatite to promote FGF-2, VEGF and ALP expression as angiogenesis markers in a bone defect model. The research sample comprised 30 male Wistar rats with a defect introduced on the left femur; these were divided into three groups for treatment with ellagic acid and hydroxyapatite, hydroxyapatite and polyethylene glycol (PEG) (control). On days 7 and 14 days after treatment, the Wistar rats were euthanised, and the femoral bone tissue was removed for the immunohistochemical analysis of FGF-2, VEGF and ALP expression. FGF-2 and ALP expression increased in the group treated with ellagic acid and hydroxyapatite on days 7 and 14 post treatment (p < 0.05), and there was an increase in VEGF expression on day 7 post treatment (p < 0.05). The combination of ellagic acid and hydroxyapatite promoted FGF-2, VEGF and ALP expression as angiogenesis markers in the bone defect model.
Collapse
Affiliation(s)
- Intan Nirwana
- Department of Dental Material, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, 60132, Indonesia
| | - Elly Munadziroh
- Department of Dental Material, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, 60132, Indonesia
| | - Anita Yuliati
- Department of Dental Material, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, 60132, Indonesia
| | - Azalia Izzah Fadhila
- Bachelor of Dental Science, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, 60132, Indonesia
| | - Nurliana
- Bachelor of Dental Science, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, 60132, Indonesia
| | - Agung Satria Wardhana
- Department of Dental Material, Faculty of Dentistry, Universitas Lambung Mangkurat, Banjarmasin, Indonesia
| | - Khairul Anuar Shariff
- Department of Dental Material, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, 60132, Indonesia
- Biomaterial Niche Area, School of Material and Mineral Resource Enginering, Universiti Sains Malaysia, Engineering Campus, 14300, Nibong Tebal, Pulang Pinang, Malaysia
| | | |
Collapse
|
22
|
Furuhata M, Takayama T, Yamamoto T, Ozawa Y, Senoo M, Ozaki M, Yamano S, Sato S. Real-time assessment of guided bone regeneration in critical size mandibular bone defects in rats using collagen membranes with adjunct fibroblast growth factor-2. J Dent Sci 2021; 16:1170-1181. [PMID: 34484585 PMCID: PMC8403809 DOI: 10.1016/j.jds.2021.03.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/14/2021] [Indexed: 10/27/2022] Open
Abstract
Background/purpose Fibroblast growth factor-2 (FGF-2) regulates bone formation. The concept of guided bone regeneration using a resorbable collagen membrane (RCM) is generally accepted in implant dentistry. This study aimed to investigate the bone healing pattern in rat mandibular bone defects in real-time with and without RCM containing FGF-2 (RCM/FGF-2). Materials and methods Critical-size circular bone defects (4.0 mm diameter) were created on both sides of the rat mandibular bone. The defects were randomly divided into the following groups: control, RCM alone, RCM containing low (0.5 μg) or high (2.0 μg) concentration of FGF-2. We performed real-time in vivo micro-computerized tomography scans at the baseline and at 2, 4, and 6 weeks, and measured the volume of newly formed bone (NFB), bone mineral density (BMD) of NFB, and the closure percentage of the NFB area. At 6 weeks, the mandibular specimens were assessed histologically and histomorphometrically to evaluate the area of new bone regeneration. Results Real-time assessment revealed a significant increase in the volume, BMD, and closure percentage of the NFB area in the RCM/FGF-2-treated groups than that in the control and RCM groups. In the H-FGF-2 group, the volume and BMD of NFB exhibited a significant increase at 6 weeks than that at the baseline. Histological evaluation revealed the presence of osteoblasts, osteocytes, and blood vessels within the NFB. Conclusion The real-time in vivo experiment demonstrated that RCM/FGF-2 effectively promoted bone regeneration within the critical-size mandibular defects in rats and verified new bone formation starting in the early postoperative phase.
Collapse
Affiliation(s)
- Mitsuaki Furuhata
- Division of Applied Oral Sciences, Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Tadahiro Takayama
- Department of Periodontology, Nihon University School of Dentistry, Tokyo, Japan.,Division of Advanced Dental Treatment, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| | - Takanobu Yamamoto
- Department of Periodontology, Nihon University School of Dentistry, Tokyo, Japan
| | - Yasumasa Ozawa
- Department of Periodontology, Nihon University School of Dentistry, Tokyo, Japan
| | - Motoki Senoo
- Division of Applied Oral Sciences, Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Manami Ozaki
- Department of Oral Health Sciences, Nihon University School of Dentistry, Tokyo, Japan.,Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| | - Seiichi Yamano
- Department of Prosthodontics, New York University College of Dentistry, NY, USA
| | - Shuichi Sato
- Department of Periodontology, Nihon University School of Dentistry, Tokyo, Japan.,Division of Advanced Dental Treatment, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| |
Collapse
|
23
|
Shoushrah SH, Transfeld JL, Tonk CH, Büchner D, Witzleben S, Sieber MA, Schulze M, Tobiasch E. Sinking Our Teeth in Getting Dental Stem Cells to Clinics for Bone Regeneration. Int J Mol Sci 2021; 22:6387. [PMID: 34203719 PMCID: PMC8232184 DOI: 10.3390/ijms22126387] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/27/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022] Open
Abstract
Dental stem cells have been isolated from the medical waste of various dental tissues. They have been characterized by numerous markers, which are evaluated herein and differentiated into multiple cell types. They can also be used to generate cell lines and iPSCs for long-term in vitro research. Methods for utilizing these stem cells including cellular systems such as organoids or cell sheets, cell-free systems such as exosomes, and scaffold-based approaches with and without drug release concepts are reported in this review and presented with new pictures for clarification. These in vitro applications can be deployed in disease modeling and subsequent pharmaceutical research and also pave the way for tissue regeneration. The main focus herein is on the potential of dental stem cells for hard tissue regeneration, especially bone, by evaluating their potential for osteogenesis and angiogenesis, and the regulation of these two processes by growth factors and environmental stimulators. Current in vitro and in vivo publications show numerous benefits of using dental stem cells for research purposes and hard tissue regeneration. However, only a few clinical trials currently exist. The goal of this review is to pinpoint this imbalance and encourage scientists to pick up this research and proceed one step further to translation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Edda Tobiasch
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig- Strasse. 20, 53359 Rheinbach, Germany; (S.H.S.); (J.L.T.); (C.H.T.); (D.B.); (S.W.); (M.A.S.); (M.S.)
| |
Collapse
|
24
|
Polyelectrolyte multilayer composite coating on 316 L stainless steel for controlled release of dual growth factors accelerating restoration of bone defects. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 126:112187. [PMID: 34082986 DOI: 10.1016/j.msec.2021.112187] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/08/2021] [Accepted: 05/11/2021] [Indexed: 11/21/2022]
Abstract
A composite coating of polyelectrolyte multilayers (PEMs) consisting of collagen, a chitosan barrier, and poly-γ-glutamic acid was fabricated using a spin coating technique to investigate and overcome the limited osseointegration capacity of 316 L stainless steel (316 L SS). To further enhance the biocompatibility, bone morphogenetic protein 2 (BMP-2) and basic fibroblast growth factor-2 (FGF-2) were loaded separately as dual growth factors, allowing for progressive drug release following the natural process of bone regeneration. The first burst release of FGF-2 triggered the proliferation of surrounding cells, and the subsequent release of BMP-2 stimulated their differentiation. The microstructure, surface potential, hardness, reduced Young's modulus, and wettability were assessed using scanning electron microscopy, nanoindentation, and water contact angle. The formation of apatite layers after immersion in simulated body fluid confirmed the bioactivity of this PEM. PEMs loaded with BMP-2 and FGF-2 showed a long sustained release of growth factors for up to 48 days. The biological properties were studied in vitro with rat bone mesenchymal stem cells (rBMSCs) and in vivo using a rat critical-sized calvarial defect model. PEMs loaded with growth factors further stimulated the proliferation and osteogenic differentiation of rBMSCs and the histology results indicated that new bone tissues could directly grow onto the PEMs. These findings suggest that PEM composite coating possesses significant potential for surface modification and long-term drug release of metallic implants to assist with bone restoration.
Collapse
|
25
|
Cook B, Walker N, Zhang Q, Chen S, Evans T. The small molecule DIPQUO promotes osteogenic differentiation via inhibition of glycogen synthase kinase 3-beta signaling. J Biol Chem 2021; 296:100696. [PMID: 33895139 PMCID: PMC8138761 DOI: 10.1016/j.jbc.2021.100696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/12/2021] [Accepted: 04/21/2021] [Indexed: 10/24/2022] Open
Abstract
Bone fractures are common impact injuries typically resolved through natural processes of osteogenic regeneration and bone remodeling, restoring the biological and mechanical function. However, dysfunctionality in bone healing and repair often arises in the context of aging-related chronic disorders, such as Alzheimer's disease (AD). There is unmet need for effective pharmacological modulators of osteogenic differentiation and an opportunity to probe the complex links between bone biology and cognitive disorders. We previously discovered the small molecule DIPQUO, which promotes osteoblast differentiation and bone mineralization in mouse and human cell culture models, and in zebrafish developmental and regenerative models. Here, we examined the detailed function of this molecule. First, we used kinase profiling, cellular thermal shift assays, and functional studies to identify glycogen synthase kinase 3-beta (GSK3-β) inhibition as a mechanism of DIPQUO action. Treatment of mouse C2C12 myoblasts with DIPQUO promoted alkaline phosphatase expression and activity, which could be enhanced synergistically by treatment with other GSK3-β inhibitors. Suppression of the expression or function of GSK3-β attenuated DIPQUO-dependent osteogenic differentiation. In addition, DIPQUO synergized with GSK3-β inhibitors to stimulate expression of osteoblast genes in human multipotent progenitors. Accordingly, DIPQUO promoted accumulation and activation of β-catenin. Moreover, DIPQUO suppressed activation of tau microtubule-associated protein, an AD-related effector of GSK3-β signaling. Therefore, DIPQUO has potential as both a lead candidate for bone therapeutic development and a pharmacological modulator of GSK3-β signaling in cell culture and animal models of disorders including AD.
Collapse
Affiliation(s)
- Brandoch Cook
- Department of Surgery, Weill Cornell Medicine, New York, New York, USA.
| | - Nicholas Walker
- Department of Surgery, Weill Cornell Medicine, New York, New York, USA; Program in Physiology, Biophysics & Systems Biology, Weill Cornell Medicine, New York, New York, USA
| | - Qisheng Zhang
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, New York, New York, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
26
|
Novais A, Chatzopoulou E, Chaussain C, Gorin C. The Potential of FGF-2 in Craniofacial Bone Tissue Engineering: A Review. Cells 2021; 10:932. [PMID: 33920587 PMCID: PMC8073160 DOI: 10.3390/cells10040932] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/10/2021] [Accepted: 04/15/2021] [Indexed: 12/21/2022] Open
Abstract
Bone is a hard-vascularized tissue, which renews itself continuously to adapt to the mechanical and metabolic demands of the body. The craniofacial area is prone to trauma and pathologies that often result in large bone damage, these leading to both aesthetic and functional complications for patients. The "gold standard" for treating these large defects is autologous bone grafting, which has some drawbacks including the requirement for a second surgical site with quantity of bone limitations, pain and other surgical complications. Indeed, tissue engineering combining a biomaterial with the appropriate cells and molecules of interest would allow a new therapeutic approach to treat large bone defects while avoiding complications associated with a second surgical site. This review first outlines the current knowledge of bone remodeling and the different signaling pathways involved seeking to improve our understanding of the roles of each to be able to stimulate or inhibit them. Secondly, it highlights the interesting characteristics of one growth factor in particular, FGF-2, and its role in bone homeostasis, before then analyzing its potential usefulness in craniofacial bone tissue engineering because of its proliferative, pro-angiogenic and pro-osteogenic effects depending on its spatial-temporal use, dose and mode of administration.
Collapse
Affiliation(s)
- Anita Novais
- Pathologies, Imagerie et Biothérapies Orofaciales, Université de Paris, URP2496, 1 rue Maurice Arnoux, 92120 Montrouge, France; (A.N.); (E.C.); (C.C.)
- AP-HP Département d’Odontologie, Services d’odontologie, GH Pitié Salpêtrière, Henri Mondor, Paris Nord, Hôpital Rothschild, Paris, France
| | - Eirini Chatzopoulou
- Pathologies, Imagerie et Biothérapies Orofaciales, Université de Paris, URP2496, 1 rue Maurice Arnoux, 92120 Montrouge, France; (A.N.); (E.C.); (C.C.)
- AP-HP Département d’Odontologie, Services d’odontologie, GH Pitié Salpêtrière, Henri Mondor, Paris Nord, Hôpital Rothschild, Paris, France
- Département de Parodontologie, Université de Paris, UFR Odontologie-Garancière, 75006 Paris, France
| | - Catherine Chaussain
- Pathologies, Imagerie et Biothérapies Orofaciales, Université de Paris, URP2496, 1 rue Maurice Arnoux, 92120 Montrouge, France; (A.N.); (E.C.); (C.C.)
- AP-HP Département d’Odontologie, Services d’odontologie, GH Pitié Salpêtrière, Henri Mondor, Paris Nord, Hôpital Rothschild, Paris, France
| | - Caroline Gorin
- Pathologies, Imagerie et Biothérapies Orofaciales, Université de Paris, URP2496, 1 rue Maurice Arnoux, 92120 Montrouge, France; (A.N.); (E.C.); (C.C.)
- AP-HP Département d’Odontologie, Services d’odontologie, GH Pitié Salpêtrière, Henri Mondor, Paris Nord, Hôpital Rothschild, Paris, France
| |
Collapse
|
27
|
Abstract
The aim of this article was to synopsize platelet-rich plasma (PRP) use in musculoskeletal pathologies through evidence-based assessment of the preparation, classification, mechanism of action and applications of PRP, thereby answering which PRP type is best for each clinical indication. The literature search was performed using Medline, EMBASE and Cochrane Reviews databases for papers containing the key terms “platelet-rich plasma” AND “orthopaedics” AND (“classification” OR “mechanism of action” OR “preparation” OR “clinical application”). Generated papers were evaluated for pertinence in following areas: preparation, classification, mechanism of action, clinical application within orthopaedics. Non-English papers were excluded. Included studies were evaluated for quality. Sixty studies were included in our review. There are many commercial PRP preparation kits with differing component concentrations. There is no consensus on optimal component concentrations. Multiple PRP classifications exist but none have been validated. Platelet-rich plasma acts via growth factors (GFs) released from α-granules within platelets. Growth factors have been shown to be beneficial in healing. Grossly elevated concentrations of GFs may have inhibitory effects on healing. Multiple systematic reviews show efficacy of PRP in tendinopathies, early osteoarthritis, acute muscle injuries and in combination with rotator cuff repair and anterior cruciate ligament reconstruction. The literature suggests leukocyte-rich PRP (L-PRP) is more beneficial in tendinopathies and pure PRP (P-PRP) is more beneficial in cartilage pathology. However, different PRP preparations have not been directly compared in any pathology. Classification of PRP type is frequently not stated in research. Standardization of PRP research parameters is needed to streamline findings and generate clear indications for PRP types to yield maximum clinical benefit.
Cite this article: EFORT Open Rev 2021;6:225-235. DOI: 10.1302/2058-5241.6.200017
Collapse
Affiliation(s)
- Thomas Collins
- Trauma & Orthopaedics, Wythenshawe Hospital, Wythenshawe, UK
| | | | | |
Collapse
|
28
|
Narmada IB, Putri PD, Lucynda L, Triwardhani A, Ardani IGAW, Nugraha AP. Effect of Caffeic Acid Phenethyl Ester Provision on Fibroblast Growth Factor-2, Matrix Metalloproteinase-9 Expression, Osteoclast and Osteoblast Numbers during Experimental Tooth Movement in Wistar Rats (Rattus norvegicus). Eur J Dent 2021; 15:295-301. [PMID: 33511599 PMCID: PMC8184315 DOI: 10.1055/s-0040-1718640] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Objectives
To investigate the effect of caffeic acid phenethyl ester (CAPE) provision on matrix metalloproteinase-9 (MMP-9), fibroblast growth factor-2 (FGF-2) expression, osteoclast and osteoblast numbers during experimental orthodontic tooth movement (OTM) in male Wistar rats (
Rattus norvegicus
).
Materials and Methods
Forty-eight healthy male Wistar rats (
R. norvegicus
), 16 to 20 weeks old with 200 to 250 g body weight (bw) were divided into several groups as follows: K1: OTM for 3 days; K2: OTM for 7 days; K3: OTM for 14 days; KP1: OTM and CAPE for 3 days; KP2: OTM and CAPE for 7 days; and KP3: OTM and CAPE for 14 days. A nickel titanium closed coil spring 8.0 mm long with 10 g/mm
2
was installed between the upper left first molar and upper central incisor to move molar mesially. CAPE provision with a dose of 20 mg/kg bw of animal studies was done per orally. Immunohistochemistry was done to examine MMP-9 expression and osteoclast number in compression side as well as FGF-2 expression and osteoblast number in tensile side of the OTM.
Statistical Analysis
One-way analysis of variance test and Tukey’s honest significant difference test were performed to determine the difference between the groups (
p
< 0.05).
Results
MMP-9 expression and osteoclast numbers in the compression side were significantly different between the groups. Similarly, FGF-2 expression and osteoclast numbers in the tensile side were significantly different between the groups.
Conclusions
CAPE provision during OTM increases the number of osteoblasts and the FGF-2 expression significantly in the tensile side. Osteoclast numbers and MMP-9 expression significantly decrease in the compression side.
Collapse
Affiliation(s)
- Ida Bagus Narmada
- Department of Orthodontics, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Paristyawati Dwi Putri
- Department of Orthodontics, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Lucky Lucynda
- Department of Orthodontics, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Ari Triwardhani
- Department of Orthodontics, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - I Gusti Aju Wahju Ardani
- Department of Orthodontics, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Alexander Patera Nugraha
- Department of Orthodontics, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
29
|
Kawakami S, Shiota M, Kon K, Shimogishi M, Iijima H, Kasugai S. Autologous micrografts from the palatal mucosa for bone regeneration in calvarial defects in rats: a radiological and histological analysis. Int J Implant Dent 2021; 7:6. [PMID: 33491155 PMCID: PMC7829308 DOI: 10.1186/s40729-020-00288-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 12/28/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The application of dental implants is often restricted by bone volume. In such cases, bone grafts are required, although bone graft materials have some disadvantages. Therefore, other effective approaches are needed. Our previous study showed that the autologous micrograft, a dissociated cell suspension made out of palatal connective tissue grafts, promoted bone-marrow cell proliferation and differentiation under osteogenic conditions. In this study, we aimed to evaluate the effects of dissociated soft-tissue suspensions relevant to bone regeneration in animal model. MATERIAL AND METHODS Twelve-week-old male Wistar rats were used in the study. Defects were created in rat calvaria, and were filled with hydrogel containing either dissociated soft-tissue suspension (test) or sucrose (control). The new bone formation was evaluated at 1 and 2 weeks after surgery (n = 16) by radiological and histological analysis. RESULTS The conducted radiological analysis showed that the new bone volume was significantly greater in the dissociated soft-tissue suspension group. This finding was further confirmed by the conducted histological analysis. CONCLUSIONS The dissociated mucosa tissue suspension enhanced bone regeneration in vivo; thus, it is a promising potential method to aid the successful application for bone augmentation in the implant practice.
Collapse
Affiliation(s)
- Sawako Kawakami
- Department of Oral Implantology and Regenerative Dental Medicine, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan.
| | - Makoto Shiota
- Department of Oral Implantology and Regenerative Dental Medicine, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Kazuhiro Kon
- Department of Oral Implantology and Regenerative Dental Medicine, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Masahiro Shimogishi
- Department of Oral Implantology and Regenerative Dental Medicine, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Hajime Iijima
- Department of Oral Implantology and Regenerative Dental Medicine, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Shohei Kasugai
- Department of Oral Implantology and Regenerative Dental Medicine, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| |
Collapse
|
30
|
Abstract
This chapter provides an overview of the growth factors active in bone regeneration and healing. Both normal and impaired bone healing are discussed, with a focus on the spatiotemporal activity of the various growth factors known to be involved in the healing response. The review highlights the activities of most important growth factors impacting bone regeneration, with a particular emphasis on those being pursued for clinical translation or which have already been marketed as components of bone regenerative materials. Current approaches the use of bone grafts in clinical settings of bone repair (including bone grafts) are summarized, and carrier systems (scaffolds) for bone tissue engineering via localized growth factor delivery are reviewed. The chapter concludes with a consideration of how bone repair might be improved in the future.
Collapse
|
31
|
Chang M, Lin H, Fu H, Wang J, Yang Y, Wan Z, Han G. CREB activation affects mesenchymal stem cell migration and differentiation in periodontal tissues due to orthodontic force. Int J Biochem Cell Biol 2020; 129:105862. [PMID: 33045372 DOI: 10.1016/j.biocel.2020.105862] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 12/31/2022]
Abstract
During the orthodontic tooth movement, cells in periodontal ligament could differentiate into osteoblasts to synthesize alveolar bone as well as affect the proliferation, migration and differentiation of mesenchymal stem cells, which also contribute to bone remodeling. However, the mechanism is still largely elusive. Here, we evaluated the expression of CREB at the tension site of mouse periodontal ligament under orthodontic mechanical strain and in the cyclic tension strain treated human periodontal ligament cells. Then, through gain and loss of function analysis, we revealed that CREB in PDLCs promotes SDF-1 and FGF2 secretion, which enhance the migration and osteoblastic differentiation of BMSCs. We further discovered that CREB transcriptionally activates FGF2 and SDF-1 expressions by binding to the promoter regions.In conclusion, this study confirms that CREB is an upregulated gene in periodontal ligament under orthodontic tension strain stimulation and plays an important role in regulating BMSCs' physiological activity in orthodontic tension strain-induced bone formation.
Collapse
Affiliation(s)
- Maolin Chang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Heng Lin
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Haidi Fu
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jie Wang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yang Yang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ziqiu Wan
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Guangli Han
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
32
|
Leng L, Ma J, Lv L, Wang W, Gao D, Zhu Y, Wu Z. Both Wnt signaling and epidermal stem cell-derived extracellular vesicles are involved in epidermal cell growth. Stem Cell Res Ther 2020; 11:415. [PMID: 32967725 PMCID: PMC7510321 DOI: 10.1186/s13287-020-01933-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/29/2020] [Accepted: 09/10/2020] [Indexed: 11/10/2022] Open
Abstract
Millions suffer from skin diseases. Functional interfollicular epidermal stem cells are needed in skin therapy or drug screening in vitro. We obtained functional interfollicular epidermal stem cells with intact stemness and cell junctions by treating them with Wnt3a. Moreover, epidermal stem cell-derived extracellular vesicles were useful in epidermal cell growth. Finally, functional epidermal 3D organoids with polarity were cultured using Wnt3a and the supernatant derived from interfollicular epidermal stem cells and fresh medium in a 1:1 ratio. These results provide novel directions for the improvement of skin organoids and their potential in clinical application.
Collapse
Affiliation(s)
- Ling Leng
- Stem Cell and Regenerative Medicine Lab, Department of Medical Science Research Center, Translational Medicine Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| | - Jie Ma
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, China
| | - Luye Lv
- Institute of NBC Defense, Beijing, China
| | - Wenjuan Wang
- Department of Dermatology, Chinese PLA General Hospital, Beijing, China
| | - Dunqin Gao
- Stem Cell and Regenerative Medicine Lab, Department of Medical Science Research Center, Translational Medicine Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yunping Zhu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, China.,Basic Medical School, Anhui Medical University, Hefei, Anhui, China
| | - Zhihong Wu
- Stem Cell and Regenerative Medicine Lab, Department of Medical Science Research Center, Translational Medicine Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
33
|
Min Swe NM, Kobayashi Y, Kamimoto H, Moriyama K. Aberrantly activated Wnt/β-catenin pathway co-receptors LRP5 and LRP6 regulate osteoblast differentiation in the developing coronal sutures of an Apert syndrome (Fgfr2 S252W /+ ) mouse model. Dev Dyn 2020; 250:465-476. [PMID: 32822074 DOI: 10.1002/dvdy.239] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 08/13/2020] [Accepted: 08/13/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Apert syndrome is an autosomal, dominant inherited disorder characterized by craniosynostosis and syndactyly caused by gain-of-function mutations in the fibroblast growth factor receptor 2 (FGFR2) gene. Wnt/β-catenin signaling plays critical roles in regulating the skeletal development. Here, we analyzed the role of this pathway in the developing coronal sutures (CS) of a murine Apert syndrome model (Fgfr2S252W/+ ). RESULTS We observed aberrantly increased mRNA expression of Lrp5 and Lrp6 in CS of Fgfr2S252W/+ mice, whereas both wild type (WT) and Fgfr2S252W/+ mice showed similar expression of other Wnt/β-catenin-related genes, such as Wnt3, Wnt3a, Fzd4, Fzd6, Axin2, and Dkk1 as evidenced by in situ hybridization. Significantly increased Lrp5 and Lrp6 mRNA expression was observed by quantitative PCR analysis of cultured cells isolated from CS of Fgfr2S252W/+ mice. Phospho-LRP5, phospho-LRP6, and non-phospho-β-catenin were upregulated in Fgfr2S252W/+ CS compared with that in WT CS. Short-interfering RNA targeting Lrp5 and Lrp6 significantly reduced runt-related transcription factor 2, collagen type 1 alpha 1, and osteocalcin mRNA expression, and alkaline phosphatase activity in cultured cells. CONCLUSIONS The Wnt/β-catenin pathway was activated in the CS of Fgfr2S252W/+ mice during craniofacial development, suggesting the involvement of the Wnt/β-catenin pathway in the pathogenesis of CS synostosis in Fgfr2S252W/+ mice.
Collapse
Affiliation(s)
- Nay Myo Min Swe
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Yukiho Kobayashi
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Hiroyuki Kamimoto
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Keiji Moriyama
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
34
|
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
|
35
|
Sanjeev G, Sidharthan DS, Pranavkrishna S, Pranavadithya S, Abhinandan R, Akshaya RL, Balagangadharan K, Siddabathuni N, Srinivasan S, Selvamurugan N. An osteoinductive effect of phytol on mouse mesenchymal stem cells (C3H10T1/2) towards osteoblasts. Bioorg Med Chem Lett 2020; 30:127137. [PMID: 32245598 DOI: 10.1016/j.bmcl.2020.127137] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/16/2020] [Accepted: 03/20/2020] [Indexed: 01/11/2023]
Abstract
In recent years, phytochemicals have been widely researched and utilized for the treatment of various medical conditions such as cancer, cardiovascular diseases, age-related problems and are also said to have bone regenerative effects. In this study, phytol (3,7,11,15-tetramethylhexadec-2-en-1-ol), an acyclic unsaturated diterpene alcohol and a secondary metabolite derived from aromatic plants was investigated for its effect on osteogenesis. Phytol was found to be nontoxic in mouse mesenchymal stem cells (C3H10T1/2). At the cellular level, phytol-treatment promoted osteoblast differentiation, as seen by the increased calcium deposits. At the molecular level, phytol-treatment stimulated the expression of Runx2 (a bone-related transcription factor) and other osteogenic marker genes. MicroRNAs (miRNAs) play an essential role in controlling bone metabolism by targeting genes at the post-transcriptional level. Upon phytol-treatment in C3H10T1/2 cells, mir-21a and Smad7 levels were increased and decreased, respectively. It was previously reported that mir-21a targets Smad7 (an antagonist of TGF-beta1 signaling) and thus, protects Runx2 from its degradation. Thus, based on our results, we suggest that phytol-treatment promoted osteoblast differentiation in C3H10T1/2 cells via Runx2 due to downregulation of Smad7 by mir-21a. Henceforth, phytol was identified to bolster osteoblast differentiation, which in turn may be used for bone regeneration.
Collapse
Affiliation(s)
- Ganesh Sanjeev
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - D Saleth Sidharthan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - S Pranavkrishna
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - S Pranavadithya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - R Abhinandan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - R L Akshaya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - K Balagangadharan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Nishitha Siddabathuni
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Swathi Srinivasan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - N Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW The goal of this review is to highlight the deficits in muscle and bone in children with cerebral palsy (CP), discuss the muscle-bone relationship in the CP population, and identify muscle-based intervention strategies that may stimulate an improvement in their bone development. RECENT FINDINGS The latest research suggests that muscle and bone are both severely underdeveloped and weak in children with CP, even in ambulatory children with mild forms of the disorder. The small and low-performing muscles and limited participation in physical activity are likely the major contributors to the poor bone development in children with CP. However, the muscle-bone relationship may be complicated by other factors, such as a high degree of fat and collagen infiltration of muscle, atypical muscle activation, and muscle spasticity. Muscle-based interventions, such as resistance training, vibration, and nutritional supplementation, have the potential to improve bone development in children with CP, especially if they are initiated before puberty. Studies are needed to identify the muscle-related factors with the greatest influence on bone development in children with CP. Identifying treatment strategies that capitalize on the relationship between muscle and bone, while also improving balance, coordination, and physical activity participation, is an important step toward increasing bone strength and minimizing fractures in children with CP.
Collapse
Affiliation(s)
- Christopher M Modlesky
- Department of Kinesiology, University of Georgia, 330 River Road, Room 353, Athens, GA, 30602, USA.
| | - Chuan Zhang
- Department of Kinesiology, University of Georgia, 330 River Road, Room 353, Athens, GA, 30602, USA
| |
Collapse
|
37
|
Wang Q, Shi D, Geng Y, Huang Q, Xiang L. Baicalin augments the differentiation of osteoblasts via enhancement of microRNA-217. Mol Cell Biochem 2020; 463:91-100. [PMID: 31606864 DOI: 10.1007/s11010-019-03632-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 09/25/2019] [Indexed: 12/17/2022]
Abstract
Baicalin (BAI), a sort of flavonoid monomer, acquires from Scutellaria baicalensis Georgi, which was forcefully reported in diversified ailments due to the pleiotropic properties. But, the functions of BAI in osteoblast differentiation have not been addressed. The intentions of this study are to attest the influences of BAI in the differentiation of osteoblasts. MC3T3-E1 cells or rat primary osteoblasts were exposed to BAI, and then cell viability, ALP activity, mineralization process, and Runx2 and Ocn expression were appraised through implementing CCK-8, p-nitrophenyl phosphate (pNPP), Alizarin red staining, western blot, and RT-qPCR assays. The microRNA-217 (miR-217) expression was evaluated in MC3T3-E1 cells or rat primary osteoblasts after BAI disposition; meanwhile, the functions of miR-217 in BAI-administrated MC3T3-E1 cells were estimated after miR-217 inhibitor transfection. The impacts of BAI and miR-217 inhibition on Wnt/β-catenin and MEK/ERK pathways were probed to verify the involvements in BAI-regulated the differentiation of osteoblasts. BAI accelerated cell viability, osteoblast activity, and Runx2 and Ocn expression in MC3T3-E1 cells or rat primary osteoblasts, and the phenomena were mediated via activations of Wnt/β-catenin and MEK/ERK pathways. Elevation of miR-217 was observed in BAI-disposed MC3T3-E1 cells or rat primary osteoblasts, and miR-217 repression annulled the functions of BAI in MC3T3-E1 cell viability and differentiation. Additionally, the activations of Wnt/β-catenin and MEK/ERK pathways evoked by BAI were both restrained by repression of miR-217. These explorations uncovered that BAI augmented the differentiation of osteoblasts via activations of Wnt/β-catenin and MEK/ERK pathways by ascending miR-217 expression.
Collapse
Affiliation(s)
- Qi Wang
- Department of Orthopaedics, Heze Municipal Hospital, No. 2888 Caozhou Road, Heze, 274031, China
| | - Donglei Shi
- Department of Orthopaedics, Heze Municipal Hospital, No. 2888 Caozhou Road, Heze, 274031, China
| | - Yuanyuan Geng
- Department of Comprehensive Medical, Heze Infectious Disease Hospital, No. 298 Juyang Road, Heze, 274029, China
| | - Qishan Huang
- Department of Orthopaedics, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan West Road, Wenzhou, 325000, China
| | - Longzhan Xiang
- Department of Orthopaedics, Heze Municipal Hospital, No. 2888 Caozhou Road, Heze, 274031, China.
| |
Collapse
|
38
|
Current and Future Concepts for the Treatment of Impaired Fracture Healing. Int J Mol Sci 2019; 20:ijms20225805. [PMID: 31752267 PMCID: PMC6888215 DOI: 10.3390/ijms20225805] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/15/2019] [Accepted: 11/15/2019] [Indexed: 02/06/2023] Open
Abstract
Bone regeneration represents a complex process, of which basic biologic principles have been evolutionarily conserved over a broad range of different species. Bone represents one of few tissues that can heal without forming a fibrous scar and, as such, resembles a unique form of tissue regeneration. Despite a tremendous improvement in surgical techniques in the past decades, impaired bone regeneration including non-unions still affect a significant number of patients with fractures. As impaired bone regeneration is associated with high socio-economic implications, it is an essential clinical need to gain a full understanding of the pathophysiology and identify novel treatment approaches. This review focuses on the clinical implications of impaired bone regeneration, including currently available treatment options. Moreover, recent advances in the understanding of fracture healing are discussed, which have resulted in the identification and development of novel therapeutic approaches for affected patients.
Collapse
|
39
|
Adhikary S, Choudhary D, Tripathi AK, Karvande A, Ahmad N, Kothari P, Trivedi R. FGF-2 targets sclerostin in bone and myostatin in skeletal muscle to mitigate the deleterious effects of glucocorticoid on musculoskeletal degradation. Life Sci 2019; 229:261-276. [PMID: 31082400 DOI: 10.1016/j.lfs.2019.05.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/03/2019] [Accepted: 05/09/2019] [Indexed: 10/26/2022]
Abstract
AIM Myokines are associated with regulation of bone and muscle mass. However, limited information is available regarding the impact of myokines on glucocorticoid (GC) mediated adverse effects on the musculoskeletal system. This study investigates the role of myokine fibroblast growth factor-2 (FGF-2) in regulating GC-induced deleterious effects on bone and skeletal muscle. METHODS Primary osteoblast cells and C2C12 myoblast cell line were treated with FGF-2 and then exposed to dexamethasone (GC). FGF-2 mediated attenuation of the inhibitory effect of GC on osteoblast and myoblast differentiation and muscle atrophy was assessed through quantitative PCR and western blot analysis. Further, FGF-2 was administered subcutaneously to dexamethasone treated mice to collect bone and skeletal muscle tissue for in vivo analysis of bone microarchitecture, mechanical strength, histomorphometry and for histological alterations in treated tissue samples. KEY FINDINGS FGF-2 abrogated the dexamethasone induced inhibitory effect on osteoblast differentiation by modulating BMP-2 pathway and inhibiting Wnt antagonist sclerostin. Further, dexamethasone induced atrophy in C2C12 cells was mitigated by FGF-2 as evident from down regulation of atrogenes expression. FGF-2 prevented GC-induced impairment of mineral density, biomechanical strength, trabecular bone volume, cortical thickness and bone formation rate in mice. Additionally, skeletal muscle tissue from GC treated mice displayed weak myostatin immunostaining and reduced expression of atrogenes following FGF-2 treatment. SIGNIFICANCE FGF-2 mitigated GC induced effects through inhibition of sclerostin and myostatin expression in bone and muscle respectively. Taken together, this study exhibited the role of exogenous FGF-2 in sustaining osteoblastogenesis and inhibiting muscle atrophy in presence of glucocorticoid.
Collapse
Affiliation(s)
- Sulekha Adhikary
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Dharmendra Choudhary
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ashish Kumar Tripathi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Anirudha Karvande
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Naseer Ahmad
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Priyanka Kothari
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ritu Trivedi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India.
| |
Collapse
|
40
|
Balagangadharan K, Trivedi R, Vairamani M, Selvamurugan N. Sinapic acid-loaded chitosan nanoparticles in polycaprolactone electrospun fibers for bone regeneration in vitro and in vivo. Carbohydr Polym 2019; 216:1-16. [PMID: 31047045 DOI: 10.1016/j.carbpol.2019.04.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 03/29/2019] [Accepted: 04/01/2019] [Indexed: 12/23/2022]
Abstract
Sinapic acid (SA) is a plant-derived phenolic compound known for its multiple biological properties, but its role in the promotion of bone formation is not yet well-studied. Moreover, the delivery of SA is hindered by its complex hydrophobic nature, limiting its bioavailability. In this study, we fabricated a drug delivery system using chitosan nanoparticles (nCS) loaded with SA at different concentrations. These were incorporated into polycaprolactone (PCL) fibers via an electrospinning method. nCS loaded with 50 μM SA in PCL fibers promoted osteoblast differentiation. Furthermore, SA treatment activated the osteogenesis signaling pathways in mouse mesenchymal stem cells. A critical-sized rat calvarial bone defect model system identified that the inclusion of SA into PCL/nCS fibers accelerated bone formation. Collectively, these data suggest that SA promoted osteoblast differentiation in vitro and bone formation in vivo, possibly by activating the TGF-β1/BMP/Smads/Runx2 signaling pathways, suggesting SA might have therapeutic benefits in bone regeneration.
Collapse
Affiliation(s)
- Kalimuthu Balagangadharan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Ritu Trivedi
- Division of Endocrinology, Central Drug Research Institute (Council of Scientific and Industrial Research), Lucknow 226031, Uttar Pradesh, India
| | - Mariappanadar Vairamani
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India.
| |
Collapse
|
41
|
The Molecular Mechanism of Vitamin E as a Bone-Protecting Agent: A Review on Current Evidence. Int J Mol Sci 2019; 20:ijms20061453. [PMID: 30909398 PMCID: PMC6471965 DOI: 10.3390/ijms20061453] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 03/19/2019] [Accepted: 03/20/2019] [Indexed: 01/16/2023] Open
Abstract
Bone remodelling is a tightly-coordinated and lifelong process of replacing old damaged bone with newly-synthesized healthy bone. In the bone remodelling cycle, bone resorption is coupled with bone formation to maintain the bone volume and microarchitecture. This process is a result of communication between bone cells (osteoclasts, osteoblasts, and osteocytes) with paracrine and endocrine regulators, such as cytokines, reactive oxygen species, growth factors, and hormones. The essential signalling pathways responsible for osteoclastic bone resorption and osteoblastic bone formation include the receptor activator of nuclear factor kappa-B (RANK)/receptor activator of nuclear factor kappa-B ligand (RANKL)/osteoprotegerin (OPG), Wnt/β-catenin, and oxidative stress signalling. The imbalance between bone formation and degradation, in favour of resorption, leads to the occurrence of osteoporosis. Intriguingly, vitamin E has been extensively reported for its anti-osteoporotic properties using various male and female animal models. Thus, understanding the underlying cellular and molecular mechanisms contributing to the skeletal action of vitamin E is vital to promote its use as a potential bone-protecting agent. This review aims to summarize the current evidence elucidating the molecular actions of vitamin E in regulating the bone remodelling cycle.
Collapse
|
42
|
Cai Q, Zheng P, Ma F, Zhang H, Li Z, Fu Q, Han C, Sun Y. MicroRNA-224 enhances the osteoblastic differentiation of hMSCs via Rac1. Cell Biochem Funct 2019; 37:62-71. [PMID: 30773655 DOI: 10.1002/cbf.3373] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/07/2018] [Accepted: 12/10/2018] [Indexed: 12/30/2022]
Abstract
Osteogenesis is the differentiation of mesenchymal stem cells (MSCs) into osteoblasts. MicroRNAs (miRNAs) are short noncoding RNAs that target specific genes to mediate translational activities. In this study, we investigated how miR-224 regulates the osteoblastic differentiation of human MSCs (hMSCs) as well as the underlying mechanism. The results revealed the upregulation of miR-224 during hMSC differentiation. In vitro experiments showed that the downregulation of miR-224 suppressed the differentiation of hMSCs into osteoblasts. However, upregulation of miR-224 was concomitant with increased expression of relevant genes and augmented activity of alkaline phosphatase. Furthermore, the results indicated that Rac1 acted as the bona fide target of miR-224 and that Rac1 depletion promoted osteogenic differentiation in miR-224-silenced hMSCs. In addition, we found that both JAK/STAT3 and Wnt/β-catenin pathways were repressed by Rac1 depletion using quantitative reverse transcription polymerase chain reaction (qRT-PCR), western blotting, and immunofluorescence. Our data indicate a novel molecular mechanism in relation to hMSCs differentiation into osteoblasts, which may facilitate bone anabolism via miR-224. SIGNIFICANCE OF THE STUDY: In this study, we mainly explored the effects of miR-224 on hMSCs differentiation into osteoblasts. We find that induced miR-224 expression in hMSCs is considered closely associated with specific osteogenesis-related genes, alkaline phosphatase activity, and matrix mineralization, indicating that miR-224 may serve as a promising biomarker for osteogenic differentiation. Our data indicate a novel molecular mechanism in relation to hMSCs differentiation into osteoblasts, which may facilitate bone anabolism via miR-224.
Collapse
Affiliation(s)
- Qing Cai
- Department of Dental Implantology, School and Hospital of Stomotology, Jinlin University, Changchun, China.,Jinlin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Peng Zheng
- Department of Endodontics, School and Hospital of Stomotology, Jinlin University, Changchun, China
| | - Fuzhe Ma
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Huiyan Zhang
- Department of Dental Implantology, School and Hospital of Stomotology, Jinlin University, Changchun, China
| | - Zuntai Li
- Department of Dental Implantology, School and Hospital of Stomotology, Jinlin University, Changchun, China
| | - Qiyue Fu
- Department of Dental Implantology, School and Hospital of Stomotology, Jinlin University, Changchun, China
| | - Chunyu Han
- Department of Dental Implantology, School and Hospital of Stomotology, Jinlin University, Changchun, China
| | - Yingying Sun
- Department of Stomatology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
43
|
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
|
44
|
Abstract
Wnt/β-catenin signaling pathway is essential for embryo development and adult tissue homeostasis and regeneration, abnormal regulation of the pathway is tightly associated with many disease types, suggesting that Wnt/β-catenin signaling pathway is an attractive target for disease therapy. While the Wnt inhibitors have been extensively reviewed, small molecules activating Wnt/β-catenin signaling were rarely addressed. In this article, we firstly reviewed the diseases that were associated with disruption of Wnt/β-catenin signaling pathway, including hair loss, pigmentary disorders, wound healing, bone diseases, neurodegenerative diseases and chronic obstructive pulmonary diseases, etc. We also comprehensively summarized small molecules that activated Wnt/β-catenin signaling pathway in various models in vitro and in vivo. To evaluate the therapeutic potential of Wnt activation, we focused on the discovery strategies, phenotypic characterization, and target identification of the Wnt activators. Finally, we proposed the challenges and opportunities in development of Wnt activators for pharmacological agents in term of targeting safety and selectivity.
Collapse
|
45
|
Xiao L, Fei Y, Hurley MM. FGF2 crosstalk with Wnt signaling in mediating the anabolic action of PTH on bone formation. Bone Rep 2018; 9:136-144. [PMID: 30258857 PMCID: PMC6152810 DOI: 10.1016/j.bonr.2018.09.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/30/2018] [Accepted: 09/20/2018] [Indexed: 12/20/2022] Open
Abstract
The mechanisms of the anabolic effect of parathyroid hormone (PTH) in bone are not fully defined. The bone anabolic effects of PTH require fibroblast growth factor 2 (FGF2) as well as Wnt signaling and FGF2 modulates Wnt signaling in osteoblasts. In vivo PTH administration differentially modulated Wnt signaling in bones of wild type (WT) and in mice that Fgf2 was knocked out (Fgf2KO). PTH increased Wnt10b mRNA and protein in WT but not in KO mice. Wnt antagonist SOST mRNA and protein was significantly higher in KO group. However, PTH decreased Sost mRNA significantly in WT as well as in Fgf2KO mice, but to a lesser extent in Fgf2KO. Dickhopf 2 (DKK2) is critical for osteoblast mineralization. PTH increased Dkk2 mRNA in WT mice but the response was impaired in Fgf2KO mice. PTH significantly increased Lrp5 mRNA and phosphorylation of Lrp6 in WT but the increase was markedly attenuated in Fgf2KO mice. PTH increased β-catenin expression and Wnt/β-catenin transcriptional activity significantly in WT but not in Fgf2KO mice. These data suggest that the impaired bone anabolic response to PTH in Fgf2KO mice is partially mediated by attenuated Wnt signaling. In vivo PTH administration differentially modulated Wnt signaling in bones of WT and Fgf2KO mice. PTH treatment increased WNT10b and DKK2 expression in WT mice but the increase was blunted in Fgf2KO mice PTH increased Lrp5 mRNA and phosphorylation of Lrp6 in WT but the increase was markedly attenuated in Fgf2KO mice. PTH treatment increased β-catenin protein level and Wnt/β-catenin transcriptional activity in WT but not in Fgf2KO mice The impaired bone anabolic response to PTH in Fgf2KO mice is partially mediated by attenuated Wnt signaling.
Collapse
Affiliation(s)
| | | | - Marja M. Hurley
- Corresponding author at: Department of Medicine, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT 06030, USA.
| |
Collapse
|
46
|
Alhamdi J, Jacobs E, Gronowicz G, Benkirane-Jessel N, Hurley M, Kuhn L. Cell Type Influences Local Delivery of Biomolecules from a Bioinspired Apatite Drug Delivery System. MATERIALS 2018; 11:ma11091703. [PMID: 30217000 PMCID: PMC6163578 DOI: 10.3390/ma11091703] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/04/2018] [Accepted: 09/07/2018] [Indexed: 01/08/2023]
Abstract
Recently, the benefit of step-wise sequential delivery of fibroblast growth factor-2 (FGF-2) and bone morphogenetic protein-2 from a bioinspired apatite drug delivery system on mouse calvarial bone repair was demonstrated. The thicknesses of the nanostructured poly-l-Lysine/poly-l-Glutamic acid polyelectrolyte multilayer (PEM) and the bone-like apatite barrier layer that make up the delivery system, were varied. The effects of the structural variations of the coating on the kinetics of cell access to a cytotoxic factor delivered by the layered structure were evaluated. FGF-2 was adsorbed into the outer PEM, and cytotoxic antimycin-A (AntiA) was adsorbed to the substrate below the barrier layer to detect the timing of the cell access. While MC3T3-E1 osteoprogenitor cells accessed AntiA after three days, the RAW 264.7 macrophage access occurred within 4 h, unless the PEM layer was removed, in which case the results were reversed. Pits were created in the coating by the RAW 264.7 macrophages and initiated delivery, while the osteoprogenitor cell access to drugs occurred through a solution-mediated coating dissolution, at junctions between the islands of crystals. Macrophage-mediated degradation is therefore a mechanism that controls drug release from coatings containing bioinspired apatite.
Collapse
Affiliation(s)
- Jumana Alhamdi
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - Emily Jacobs
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - Gloria Gronowicz
- Department of Surgery, University of Connecticut Health, Farmington, CT 06030, USA.
| | - Nadia Benkirane-Jessel
- French National Institute of Health and Medical Research (INSERM), UMR 1260, Faculté de Médecine, University of Strasbourg, 67085 Strasbourg, France.
| | - Marja Hurley
- Department of Medicine, University of Connecticut Health, Farmington, CT 06030, USA.
| | - Liisa Kuhn
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA.
| |
Collapse
|
47
|
Aziz K, Sieben CJ, Jeganathan KB, Hamada M, Davies BA, Velasco ROF, Rahman N, Katzmann DJ, van Deursen JM. Mosaic-variegated aneuploidy syndrome mutation or haploinsufficiency in Cep57 impairs tumor suppression. J Clin Invest 2018; 128:3517-3534. [PMID: 30035751 PMCID: PMC6063474 DOI: 10.1172/jci120316] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 06/06/2018] [Indexed: 12/29/2022] Open
Abstract
A homozygous truncating frameshift mutation in CEP57 (CEP57T/T) has been identified in a subset of mosaic-variegated aneuploidy (MVA) patients; however, the physiological roles of the centrosome-associated protein CEP57 that contribute to disease are unknown. To investigate these, we have generated a mouse model mimicking this disease mutation. Cep57T/T mice died within 24 hours after birth with short, curly tails and severely impaired vertebral ossification. Osteoblasts in lumbosacral vertebrae of Cep57T/T mice were deficient for Fgf2, a Cep57 binding partner implicated in diverse biological processes, including bone formation. Furthermore, a broad spectrum of tissues of Cep57T/T mice had severe aneuploidy at birth, consistent with the MVA patient phenotype. Cep57T/T mouse embryonic fibroblasts and patient-derived skin fibroblasts failed to undergo centrosome maturation in G2 phase, causing premature centriole disjunction, centrosome amplification, aberrant spindle formation, and high rates of chromosome missegregation. Mice heterozygous for the truncating frameshift mutation or a Cep57-null allele were overtly indistinguishable from WT mice despite reduced Cep57 protein levels, yet prone to aneuploidization and cancer, with tumors lacking evidence for loss of heterozygosity. This study identifies Cep57 as a haploinsufficient tumor suppressor with biologically diverse roles in centrosome maturation and Fgf2-mediated bone formation.
Collapse
Affiliation(s)
- Khaled Aziz
- Department of Biochemistry and Molecular Biology and
| | | | - Karthik B. Jeganathan
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Masakazu Hamada
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Nazneen Rahman
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | | | - Jan M. van Deursen
- Department of Biochemistry and Molecular Biology and
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
48
|
MicroRNA-378 Promotes Osteogenesis-Angiogenesis Coupling in BMMSCs for Potential Bone Regeneration. Anal Cell Pathol (Amst) 2018; 2018:8402390. [PMID: 29686962 PMCID: PMC5852880 DOI: 10.1155/2018/8402390] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/02/2017] [Accepted: 11/08/2017] [Indexed: 01/25/2023] Open
Abstract
Bone tissue regeneration was closely associated with osteogenesis and angiogenesis. The harmonious regulation of osteogenetic and angiogenic growth factors would enhance bone regeneration, while the imbalance of that would lead to local excessive bone formation or vascular mass due to exogenous delivery. Therefore, microRNA is believed to regulate multiple metabolism progress through endogenous signaling pathways on the gene level. In this work, we identified microRNA 378 as a positive regulator of osteogenesis and angiogenesis simultaneously and also observed an increase of microRNA 378 than control in human bone marrow mesenchymal stem cells (hBMMSCs) after osteoblast induction. Besides, osteogenetic and angiogenic gene expression increased simultaneously after overexpression of microRNA 378. Moreover, alizarin red staining and alkaline phosphatase (ALP) staining enhanced, and secretion of vascular endothelial growth factor (VEGF) increased. In this way, we believed miR378 was an ideal target to osteogenesis-angiogenesis coupling for bone regeneration, which provides a potential tool for the gene therapy of bone regeneration.
Collapse
|
49
|
García-Giménez JL, Rubio-Belmar PA, Peiró-Chova L, Hervás D, González-Rodríguez D, Ibañez-Cabellos JS, Bas-Hermida P, Mena-Mollá S, García-López EM, Pallardó FV, Bas T. Circulating miRNAs as diagnostic biomarkers for adolescent idiopathic scoliosis. Sci Rep 2018; 8:2646. [PMID: 29422531 PMCID: PMC5805715 DOI: 10.1038/s41598-018-21146-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 01/31/2018] [Indexed: 11/09/2022] Open
Abstract
The aetiology of adolescent idiopathic scoliosis (AIS) has been linked to many factors, such as asymmetric growth, neuromuscular condition, bone strength and genetic background. Recently, epigenetic factors have been proposed as contributors of AIS physiopathology, but information about the molecular mechanisms and pathways involved is scarce. Regarding epigenetic factors, microRNAs (miRNAs) are molecules that contribute to gene expression modulation by regulating important cellular pathways. We herein used Next-Generation Sequencing to discover a series of circulating miRNAs detected in the blood samples of AIS patients, which yielded a unique miRNA biomarker signature that diagnoses AIS with high sensitivity and specificity. We propose that these miRNAs participate in the epigenetic control of signalling pathways by regulating osteoblast and osteoclast differentiation, thus modulating the genetic background of AIS patients. Our study yielded two relevant results: 1) evidence for the deregulated miRNAs that participate in osteoblast/osteoclast differentiation mechanisms in AIS; 2) this miRNA-signature can be potentially used as a clinical tool for molecular AIS diagnosis. Using miRNAs as biomarkers for AIS diagnostics is especially relevant since miRNAs can serve for early diagnoses and for evaluating the positive effects of applied therapies to therefore reduce the need of high-risk surgical interventions.
Collapse
Affiliation(s)
- José Luis García-Giménez
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain.
- Instituto de Investigación Sanitaria INCLIVA, Avenida de Menéndez y Pelayo, 4, 46010, Valencia, Spain.
- Dept. Physiology. Faculty of Medicine and Dentistry, University of Valencia, Av/Blasco Ibañez, 15, 46010, Valencia, Spain.
| | - Pedro Antonio Rubio-Belmar
- Instituto de Investigación Sanitaria IISLAFE, Av/Fernando Abril Martorell, 106. Torre A 7, 46026, Valencia, Spain
- Unidad de Raquis. Hospital Universitari i Politècnic La Fe, Av/Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Lorena Peiró-Chova
- Instituto de Investigación Sanitaria INCLIVA, Avenida de Menéndez y Pelayo, 4, 46010, Valencia, Spain
| | - David Hervás
- Unidad de Bioestadística, Instituto de Investigación Sanitaria IISLAFE, Av/Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Daymé González-Rodríguez
- Instituto de Investigación Sanitaria INCLIVA, Avenida de Menéndez y Pelayo, 4, 46010, Valencia, Spain
| | - José Santiago Ibañez-Cabellos
- Instituto de Investigación Sanitaria INCLIVA, Avenida de Menéndez y Pelayo, 4, 46010, Valencia, Spain
- Dept. Physiology. Faculty of Medicine and Dentistry, University of Valencia, Av/Blasco Ibañez, 15, 46010, Valencia, Spain
| | - Paloma Bas-Hermida
- Instituto de Investigación Sanitaria IISLAFE, Av/Fernando Abril Martorell, 106. Torre A 7, 46026, Valencia, Spain
- Unidad de Raquis. Hospital Universitari i Politècnic La Fe, Av/Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Salvador Mena-Mollá
- Dept. Physiology. Faculty of Medicine and Dentistry, University of Valencia, Av/Blasco Ibañez, 15, 46010, Valencia, Spain
| | - Eva María García-López
- Instituto de Investigación Sanitaria INCLIVA, Avenida de Menéndez y Pelayo, 4, 46010, Valencia, Spain
- Dept. Physiology. Faculty of Medicine and Dentistry, University of Valencia, Av/Blasco Ibañez, 15, 46010, Valencia, Spain
| | - Federico V Pallardó
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain
- Instituto de Investigación Sanitaria INCLIVA, Avenida de Menéndez y Pelayo, 4, 46010, Valencia, Spain
- Dept. Physiology. Faculty of Medicine and Dentistry, University of Valencia, Av/Blasco Ibañez, 15, 46010, Valencia, Spain
| | - Teresa Bas
- Instituto de Investigación Sanitaria IISLAFE, Av/Fernando Abril Martorell, 106. Torre A 7, 46026, Valencia, Spain
- Unidad de Raquis. Hospital Universitari i Politècnic La Fe, Av/Fernando Abril Martorell, 106, 46026, Valencia, Spain
| |
Collapse
|
50
|
Chen X, Wang L, Zhao K, Wang H. Osteocytogenesis: Roles of Physicochemical Factors, Collagen Cleavage, and Exogenous Molecules. TISSUE ENGINEERING PART B-REVIEWS 2018; 24:215-225. [PMID: 29304315 DOI: 10.1089/ten.teb.2017.0378] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Osteocytes, the most abundant cell type in mammalian bone, are generally considered as the terminally differentiated cells of osteoblasts that are progressively self-buried or passively embedded in bone matrix. Emerging evidence reveals the essential functions of osteocytes in bone homeostasis and mechanotransduction. However, our knowledge on osteocytes, especially their formation, remains scarce. In this regard, the current review mainly focuses on several key factors that drive the osteocytic differentiation of osteoblasts, that is, osteocytogenesis. Available literature has demonstrated the involvement of physicochemical factors such as matrix composition, oxygen tension, and mechanical stress in the osteoblast-to-osteocyte transition. During cell migration and matrix remodeling, the matrix metalloproteinase-dependent collagen cleavage would play an "active" role in maturation and maintenance of the osteocytes. Besides, some in vitro methodologies are also established to induce the transformation of osteoblastic cell lines and primary mesenchymal cells to preosteocytes through cell transfection or addition of exogenous molecules (e.g., fibroblast growth factor-2, retinoic acid), which could potentiate the effort to form functional bone substitutes through elevated osteocytogenesis. Thus, advances of new technologies would enable comprehensive and in-depth understanding of osteocytes and their development, which in turn help promote the research on osteocyte biology and osteopathology.
Collapse
Affiliation(s)
- Xuening Chen
- 1 National Engineering Research Center for Biomaterials, Sichuan University , Chengdu, China
| | - Lichen Wang
- 2 Department of Biomedical Engineering, Chemistry and Biological Sciences, Stevens Institute of Technology , Hoboken, New Jersey
| | - Kaitao Zhao
- 2 Department of Biomedical Engineering, Chemistry and Biological Sciences, Stevens Institute of Technology , Hoboken, New Jersey
| | - Hongjun Wang
- 2 Department of Biomedical Engineering, Chemistry and Biological Sciences, Stevens Institute of Technology , Hoboken, New Jersey
| |
Collapse
|