1
|
Teitelbaum M, Culbertson MD, Wetterstrand C, O’Connor JP. Impaired fracture healing is associated with callus chondro-osseous junction abnormalities in periostin-null and osteopontin-null mice. Exp Biol Med (Maywood) 2025; 249:10066. [PMID: 39911634 PMCID: PMC11794000 DOI: 10.3389/ebm.2024.10066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 08/22/2024] [Indexed: 02/07/2025] Open
Abstract
Periostin and osteopontin are matricellular proteins abundantly expressed in bone fracture callus. Null mutation of either the periostin (Postn) gene or the osteopontin (Spp1) gene can impair bone fracture healing. However, the cell and molecular pathways affected by loss of POSTN or SPP1 which lead to impaired fracture healing are not well understood. To identify potential pathways, a detailed radiological, histological, and immunohistochemical analysis of femur fracture healing in Postn-null (PostnKO), Spp1-null (Spp1KO), and normal (WT) mice was performed. Apparent changes in specific protein levels identified by immunohistochemistry were confirmed by mRNA quantitation. Comparisons between the PostnKO and Spp1KO fracture calluses were confounded by interactions between the two genes; loss of Postn reduced Spp1 expression and loss of Spp1 reduced Postn expression. Consequently, alterations in fracture healing between mice heterozygous for the Postn-null allele (PostnHET) as well as the PostnKO and Spp1KO mice were similar. Calluses from PostnHET, PostnKO, and Spp1KO mice all had dysmorphic chondro-osseous junctions and reduced numbers of osteoclasts. The dysmorphic chondro-osseous junctions in the PostnHET, PostnKO, and Spp1KO calluses were associated with reduced numbers of MMP-13 expressing hypertrophic chondrocytes, consistent with delayed cartilage resolution. Unlike collagen X expressing callus chondrocytes, chondrocytes only expressed MMP-13 when localized to the chondro-osseous junction or after traversing the chondro-osseous junction. Cyclooxygenase-2 (COX-2) expression also appeared to be reduced in osteoclasts from the PostnHET, PostnKO, and Spp1KO calluses, including in those osteoclasts localized at the chondro-osseous junction. The results indicate that POSTN and SPP1 are necessary for normal chondro-osseous junction formation and that signaling from the chondro-osseous junction, possibly from COX-2 expressing osteoclasts, regulates callus vasculogenesis and chondrocyte hypertrophy necessary for endochondral ossification during fracture healing.
Collapse
Affiliation(s)
- Marc Teitelbaum
- Department of Orthopaedics, Rutgers-New Jersey Medical School, Newark, NJ, United States
- Rutgers-School of Graduate Studies, Newark Health Sciences Campus, Newark, NJ, United States
| | - Maya D. Culbertson
- Department of Orthopaedics, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Charlene Wetterstrand
- Department of Orthopaedics, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - J. Patrick O’Connor
- Department of Orthopaedics, Rutgers-New Jersey Medical School, Newark, NJ, United States
- Rutgers-School of Graduate Studies, Newark Health Sciences Campus, Newark, NJ, United States
| |
Collapse
|
2
|
Sawauchi K, Fukui T, Oe K, Oda T, Yoshikawa R, Takase K, Inoue S, Nishida R, Kuroda R, Niikura T. Transcutaneous CO 2 application combined with low-intensity pulsed ultrasound accelerates bone fracture healing in rats. BMC Musculoskelet Disord 2024; 25:863. [PMID: 39472824 PMCID: PMC11523825 DOI: 10.1186/s12891-024-07976-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 10/17/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Low-intensity pulsed ultrasound (LIPUS) is a non-invasive therapy that accelerates fracture healing. As a new treatment method for fracture, we recently reported that the transcutaneous application of CO2 accelerated fracture healing in association with promoting angiogenesis, blood flow, and endochondral ossification. We hypothesized that transcutaneous CO2 application, combined with LIPUS, would promote bone fracture healing more than the single treatment with either of them. METHODS Femoral shaft fractures were produced in 12-week-old rats. Animals were randomly divided into four groups: the combination of CO2 and LIPUS, CO2, LIPUS, and control groups. As the transcutaneous CO2 application, the limb was sealed in a CO2-filled bag after applying hydrogel that promotes CO2 absorption. Transcutaneous CO2 application and LIPUS irradiation were performed for 20 min/day, 5 days/week. At weeks 1, 2, 3, and 4 after the fractures, we assessed the fracture healing process using radiography, histology, immunohistochemistry, real-time PCR, and biomechanical assessment. RESULTS The fracture healing score using radiographs in the combination group was significantly higher than that in the control group at all time points and those in both the LIPUS and CO2 groups at weeks 1, 2, and 4. The degree of bone fracture healing in the histological assessment was significantly higher in the combination group than that in the control group at weeks 2, 3, and 4. In the immunohistochemical assessment, the vascular densities of CD31- and endomucin-positive microvessels in the combination group were significantly higher than those in the control and LIPUS groups at week 2. In the gene expression assessment, significant upregulation of runt-related transcription factor 2 (Runx2) and vascular endothelial growth factor (VEGF) was detected in the combination group compared to the LIPUS and CO2 monotherapy groups. In the biomechanical assessment, the ultimate stress was significantly higher in the combination group than in the LIPUS and CO2 groups. CONCLUSION The combination therapy of transcutaneous CO2 application and LIPUS had a superior effect in promoting fracture healing through the promotion of angiogenesis and osteoblast differentiation compared to monotherapy.
Collapse
Affiliation(s)
- Kenichi Sawauchi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Tomoaki Fukui
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Keisuke Oe
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Takahiro Oda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Ryo Yoshikawa
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kyohei Takase
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Shota Inoue
- Department of Rehabilitation Science, Graduate School of Health Sciences, Kobe University, Kobe, Japan
| | - Ryota Nishida
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Takahiro Niikura
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
- Department of Orthopaedic Surgery, Hyogo Prefectural Nishinomiya Hospital, Nishinomiya, Japan.
| |
Collapse
|
3
|
Valeriano AT, Camara LS, Bernardes VDF, Pais FSM, Araújo FMG, Salim ACDM, Fernandes GDR, Stussi F, Gomes CC, de Andrade Santos PP, de Souza LB, Gomez RS, Diniz MG. MMP13 Expression and Activity Suggest Its Role in Bone Resorption in Ameloblastomas. J Oral Pathol Med 2024; 53:577-583. [PMID: 39192690 DOI: 10.1111/jop.13577] [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: 01/25/2024] [Revised: 07/04/2024] [Accepted: 07/17/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Ameloblastoma is a locally destructive benign odontogenic tumor. While the neoplastic cells of conventional ameloblastoma can infiltrate the connective tissue and bone, in unicystic ameloblastoma the epithelium is encapsulated. The mechanisms driving ameloblastoma's bone resorption remains unclear. METHODS RNA sequencing (RNA-seq) was performed in a discovery cohort of conventional ameloblastoma, and pathway enrichment analysis was carried out. mRNA levels of MMP13, a gene associated with bone resorption, were assessed using RT-qPCR in a larger cohort of conventional ameloblastoma and in unicystic ameloblastoma. Zymogram gels and the immunoexpression profile of collagenase 3 (encoded by MMP13 gene) were evaluated as well. RESULTS Enriched pathways related to bone mineralization and upregulation of MMP13 were observed in ameloblastomas. Collagenolytic activity of collagenase 3 was detected in the tumor lysates. Collagenase 3 immunopositivity was observed in ameloblastomatous epithelium infiltrating the fibrous capsule of unicystic ameloblastoma. At the tumor-bone interface, collagenase 3 expression was detected in stromal cells, osteoblasts, and osteocytes. CONCLUSION The results indicate a potential involvement of MMP13 in ameloblastoma-related bone resorption and progression.
Collapse
Affiliation(s)
- Alline Teixeira Valeriano
- Department of Pathology, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Lais Santos Camara
- Department of Pathology, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Vanessa de Fátima Bernardes
- Department of Pathology, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | | | | | | | | | - Fernanda Stussi
- Biological Sciences Institute, Universidade Federal de minas Gerais, Belo Horizonte, MG, Brazil
| | - Carolina Cavalieri Gomes
- Department of Pathology, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | | | - Lélia Batista de Souza
- Department of Oral Pathology, Universidade Federal do Rio Grande do Norte (UFRN), Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil
| | - Ricardo Santiago Gomez
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG) and Medical School, Faculdade Ciências Médicas de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marina Gonçalves Diniz
- Department of Pathology, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| |
Collapse
|
4
|
Angelozzi M, Karvande A, Lefebvre V. SOXC are critical regulators of adult bone mass. Nat Commun 2024; 15:2956. [PMID: 38580651 PMCID: PMC10997656 DOI: 10.1038/s41467-024-47413-2] [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: 09/08/2023] [Accepted: 03/28/2024] [Indexed: 04/07/2024] Open
Abstract
Pivotal in many ways for human health, the control of adult bone mass is governed by complex, incompletely understood crosstalk namely between mesenchymal stem cells, osteoblasts and osteoclasts. The SOX4, SOX11 and SOX12 (SOXC) transcription factors were previously shown to control many developmental processes, including skeletogenesis, and SOX4 was linked to osteoporosis, but how SOXC control adult bone mass remains unknown. Using SOXC loss- and gain-of-function mouse models, we show here that SOXC redundantly promote prepubertal cortical bone mass strengthening whereas only SOX4 mitigates adult trabecular bone mass accrual in early adulthood and subsequent maintenance. SOX4 favors bone resorption over formation by lowering osteoblastogenesis and increasing osteoclastogenesis. Single-cell transcriptomics reveals its prevalent expression in Lepr+ mesenchymal cells and ability to upregulate genes for prominent anti-osteoblastogenic and pro-osteoclastogenic factors, including interferon signaling-related chemokines, contributing to these adult stem cells' secretome. SOXC, with SOX4 predominantly, are thus key regulators of adult bone mass.
Collapse
Affiliation(s)
- Marco Angelozzi
- Department of Surgery, Division of Orthopaedics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Anirudha Karvande
- Department of Surgery, Division of Orthopaedics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Véronique Lefebvre
- Department of Surgery, Division of Orthopaedics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Saranya I, Akshaya R, Gomathi K, Mohanapriya R, He Z, Partridge N, Selvamurugan N. Circ_ST6GAL1-mediated competing endogenous RNA network regulates TGF-β1-stimulated matrix Metalloproteinase-13 expression via Runx2 acetylation in osteoblasts. Noncoding RNA Res 2024; 9:153-164. [PMID: 38035043 PMCID: PMC10686813 DOI: 10.1016/j.ncrna.2023.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 12/02/2023] Open
Abstract
Transforming growth factor-beta1 (TGF-β1) stimulates matrix metalloproteinase-13 (MMP-13, a bone-remodeling gene) expression, and this effect requires p300-mediated Runx2 (Runt-related transcription factor 2) acetylation in osteoblasts. p300 and Runx2 are transcriptional coactivator and bone transcription factor, respectively, which play key roles in the regulation of bone-remodeling genes. Non-coding ribonucleic acids (ncRNAs), such as long ncRNAs (lncRNAs) and microRNAs (miRNAs), have been linked to both physiological and pathological bone states. In this study, we proposed that TGF-β1-mediated stimulation of MMP-13 expression is due to the downregulation of p300 targeting miRNAs in osteoblasts. We identified miR-130b-5p as one of the miRNAs downregulated by TGF-β1 in osteoblasts. Forced expression of miR-130b-5p decreased p300 expression, Runx2 acetylation, and MMP-13 expression in these cells. Furthermore, TGF-β1 upregulated circ_ST6GAL1, (a circular lncRNA) in osteoblasts; circRNA directly targeted miR-130b-5p. Antisense-mediated knockdown of circ_ST6GAL1 restored the function of miR-130b-5p, resulting in downregulation of p300, Runx2, and MMP-13 in these cells. Hence, our results suggest that TGF-β1 influences circ_ST6GAL1 to sponge and degrade miR-130b-5p, thereby promoting p300-mediated Runx2 acetylation for MMP-13 expression in osteoblasts. Thus, the circ_ST6GAL1/miR-130b-5p/p300 axis has potential significance in the treatment of bone and bone-related disorders.
Collapse
Affiliation(s)
- I. Saranya
- Biotechnology, School of Bioengineering, SRMIST, Kattankulathur, India
| | - R.L. Akshaya
- Biotechnology, School of Bioengineering, SRMIST, Kattankulathur, India
| | - K. Gomathi
- Biotechnology, School of Bioengineering, SRMIST, Kattankulathur, India
| | - R. Mohanapriya
- Biotechnology, School of Bioengineering, SRMIST, Kattankulathur, India
| | - Z. He
- Molecular Pathobiology, New York University College of Dentistry, New York, USA
| | - N.C. Partridge
- Molecular Pathobiology, New York University College of Dentistry, New York, USA
| | - N. Selvamurugan
- Biotechnology, School of Bioengineering, SRMIST, Kattankulathur, India
| |
Collapse
|
6
|
Ma C, Tao C, Zhang Z, Zhou H, Fan C, Wang DA. Development of artificial bone graft via in vitro endochondral ossification (ECO) strategy for bone repair. Mater Today Bio 2023; 23:100893. [PMID: 38161510 PMCID: PMC10755541 DOI: 10.1016/j.mtbio.2023.100893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/21/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024] Open
Abstract
Endochondral ossification (ECO) is a form of bone formation whereby the newly deposited bone replaces the cartilage template. A decellularized artificial cartilage graft (dLhCG), which is composed of hyaline cartilage matrixes, has been developed in our previous study. Herein, the osteogenesis of bone marrow-derived MSCs in the dLhCG through chondrogenic differentiation, chondrocyte hypertrophy, and subsequent transdifferentiation induction has been investigated by simulating the physiological processes of ECO for repairing critical-sized bone defects. The MSCs were recellularized into dLhCGs and subsequently allowed to undergo a 14-day proliferation period (mrLhCG). Following this, the mrLhCG constructs were subjected to two distinct differentiation induction protocols to achieve osteogenic differentiation: chondrogenic medium followed by chondrocytes culture medium with a high concentration of fetal bovine serum (CGCC group) and canonical osteogenesis inducing medium (OI group). The formation of a newly developed artificial bone graft, ossified dLhCG (OsLhCG), as well as its capability of aiding bone defect reconstruction were characterized by in vitro and in vivo trials, such as mRNA sequencing, quantitative real-time PCR (qPCR), immunohistochemistry, the greater omentum implantation in nude mice, and repair for the critical-sized femoral defects in rats. The results reveal that the differentiation induction of MSCs in the CGCC group can realize in vitro ECO through chondrogenic differentiation, hypertrophy, and transdifferentiation, while the MSCs in the OI group, as expected, realize ossification through direct osteogenic differentiation. The angiogenesis and osteogenesis of OsLhCG were proved by being implanted into the greater omentum of nude mice. Besides, the OsLhCG exhibits the capability to achieve the repair of critical-size femoral defects.
Collapse
Affiliation(s)
- Cheng Ma
- Department of Biomedical Engineering, College of Engineering, City University of Hong Kong, Hong Kong
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong
| | - Chao Tao
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong
| | - Zhen Zhang
- Department of Biomedical Engineering, College of Engineering, City University of Hong Kong, Hong Kong
| | - Huiqun Zhou
- Department of Biomedical Engineering, College of Engineering, City University of Hong Kong, Hong Kong
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong
| | - Changjiang Fan
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, Shandong, 266071, China
| | - Dong-an Wang
- Department of Biomedical Engineering, College of Engineering, City University of Hong Kong, Hong Kong
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong
| |
Collapse
|
7
|
Javid K, Mourão CF, Mello-Machado RC, Sartoretto SC, Torres M, Stellet Lourenço E, Leite PEC, Granjeiro JM, Alves GG, Calasans-Maia MD. Clinical and Biochemical Evaluation of the Use of Alb-PRF versus L-PRF in Mandibular Third Molar Extractions: A Split-Mouth Randomized Clinical Trial. J Funct Biomater 2023; 14:505. [PMID: 37888172 PMCID: PMC10607814 DOI: 10.3390/jfb14100505] [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: 09/13/2023] [Revised: 09/29/2023] [Accepted: 10/08/2023] [Indexed: 10/28/2023] Open
Abstract
Bone tissue engineering seeks biomaterials that enable cell migration, angiogenesis, matrix deposition, and tissue regeneration. Blood concentrates like platelet-rich fibrin (L-PRF) offer a cost-effective source of cells and growth factors to enhance healing. The present study aimed to evaluate heated serum albumin with liquid PRF (Alb-PRF) and L-PRF clinically and biochemically after placement in dental sockets following mandibular third molar extraction. In a controlled, split-mouth study involving 10 volunteers, 20 extracted molars were treated with either Alb-PRF or L-PRF. Post-extraction, pain, trismus, infection presence, and swelling were measured. The concentrations of different analytes in the surgical sites were also examined. The data were statistically analyzed, with significance defined at p < 0.05 (t-test). No significant difference was noted between the groups for pain and trismus, but Alb-PRF showed a significant reduction in swelling on day seven. The Alb-PRF group showed lower levels of pro-inflammatory cytokines (GM-CSF, IL-1b, IL-6, IFNy, IL-8, IL-15, RANTES, and MIP-1a) after seven days, with only higher expressions of MIP-1b, IL-1b, and MCP-1 found in the L-PRF group. Differences were observed in the release of analytes between L-PRF and Alb-PRF, with Alb-PRF significantly reducing edema after seven days. Alb-PRF reduced edema, while L-PRF increased inflammatory cytokines. When compared to L-PRF, Alb-PRF reduced edema and the release of inflammatory cytokines, suggesting promising effects in socket healing while underscoring the role of growth factors and cytokines in potential applications of blood concentrates.
Collapse
Affiliation(s)
- Kayvon Javid
- Graduate Program in Dentistry, Fluminense Federal University, Niterói 24020-140, Brazil
| | - Carlos Fernando Mourão
- Department of Periodontology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
| | | | | | - Madelaine Torres
- Graduate Program in Dentistry, Fluminense Federal University, Niterói 24020-140, Brazil
| | | | - Paulo Emilio Correa Leite
- Clinical Research Unit, Antonio Pedro Hospital, Fluminense Federal University, Niterói 24033-900, Brazil
| | - José Mauro Granjeiro
- Department of Oral Surgery, Fluminense Federal University, Niterói 24020-140, Brazil
| | - Gutemberg Gomes Alves
- Department of Biotechnology, Fluminense Federal University, Niterói 24033-900, Brazil
| | | |
Collapse
|
8
|
Li YY, Zhang LY, Xiang YH, Li D, Zhang J. Matrix metalloproteinases and tissue inhibitors in multiple myeloma: promote or inhibit? Front Oncol 2023; 13:1127407. [PMID: 37823051 PMCID: PMC10562598 DOI: 10.3389/fonc.2023.1127407] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 09/04/2023] [Indexed: 10/13/2023] Open
Abstract
Matrix metalloproteinases (MMPs) and tissue inhibitor of metalloproteinases (TIMPs) play a vital role in the pathogenesis of multiple myeloma (MM), especially for tumor invasion and osteolytic osteopathy. By breaking down extracellular matrix (ECM) components and releasing the proteins composing the ECM and growth factors, as well as their receptors, MMPs affect tissue integrity and promote cancer cell invasion and metastasis. A vital pathophysiological characteristic of MM is the progress of osteolytic lesions, which are brought on by interactions between myeloma cells and the bone marrow microenvironment. MMPs, certainly, are one of the fundamental causes of myeloma bone disease due to their ability to degrade various types of collagens. TIMPs, as important regulators of MMP hydrolysis or activation, also participate in the occurrence and evolution of MM and the formation of bone disease. This review focuses on the role of MMP-1, MMP-2, MMP-7, MMP-9, MMP-13, MMP-14, and MMP-15 and the four types of TIMPs in the invasion of myeloma cells, angiogenesis, osteolytic osteopathy, to offer some novel perspectives on the clinical diagnostics and therapeutics of MM.
Collapse
Affiliation(s)
- Yan-Ying Li
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Laboratory Medicine, Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Liu-Yun Zhang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Laboratory Medicine, Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yun-Hui Xiang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Laboratory Medicine, Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Dan Li
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Juan Zhang
- Department of Laboratory Medicine, Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
9
|
Varadinkova S, Oralova V, Clarke M, Frampton J, Knopfova L, Lesot H, Bartos P, Matalova E. Expression dynamics of metalloproteinases during mandibular bone formation: association with Myb transcription factor. Front Cell Dev Biol 2023; 11:1168866. [PMID: 37701782 PMCID: PMC10493412 DOI: 10.3389/fcell.2023.1168866] [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: 02/18/2023] [Accepted: 08/18/2023] [Indexed: 09/14/2023] Open
Abstract
As the dentition forms and becomes functional, the alveolar bone is remodelled. Metalloproteinases are known to contribute to this process, but new regulators are emerging and their contextualization is challenging. This applies to Myb, a transcription factor recently reported to be involved in bone development and regeneration. The regulatory effect of Myb on Mmps expression has mostly been investigated in tumorigenesis, where Myb impacted the expression of Mmp1, Mmp2, Mmp7, and Mmp9. The aim of this investigation was to evaluate the regulatory influence of the Myb on Mmps gene expression, impacting osteogenesis and mandibular bone formation. For that purpose, knock-out mouse model was used. Gene expression of bone-related Mmps and the key osteoblastic transcription factors Runx2 and Sp7 was analysed in Myb knock-out mice mandibles at the survival limit. Out of the metalloproteinases under study, Mmp13 was significantly downregulated. The impact of Myb on the expression of Mmp13 was confirmed by the overexpression of Myb in calvarial-derived cells causing upregulation of Mmp13. Expression of Mmp13 in the context of other Mmps during mandibular/alveolar bone development was followed in vivo along with Myb, Sp7 and Runx2. The most significant changes were observed in the expression of Mmp9 and Mmp13. These MMPs and MYB were further localized in situ by immunohistochemistry and were identified in pre/osteoblastic cells as well as in pre/osteocytes. In conclusion, these results provide a comprehensive insight into the expression dynamics of bone related Mmps during mandibular/alveolar bone formation and point to Myb as another potential regulator of Mmp13.
Collapse
Affiliation(s)
- S. Varadinkova
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetics, v.v.i, Academy of Sciences, Brno, Czechia
- Department of Physiology, University of Veterinary Sciences, Brno, Czechia
| | - V. Oralova
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetics, v.v.i, Academy of Sciences, Brno, Czechia
- Department of Physiology, University of Veterinary Sciences, Brno, Czechia
| | - M. Clarke
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - J. Frampton
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - L. Knopfova
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
| | - H. Lesot
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetics, v.v.i, Academy of Sciences, Brno, Czechia
| | - P. Bartos
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetics, v.v.i, Academy of Sciences, Brno, Czechia
| | - E. Matalova
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetics, v.v.i, Academy of Sciences, Brno, Czechia
- Department of Physiology, University of Veterinary Sciences, Brno, Czechia
| |
Collapse
|
10
|
Anwer AH, Ahtesham A, Shoeb M, Mashkoor F, Ansari MZ, Zhu S, Jeong C. State-of-the-art advances in nanocomposite and bio-nanocomposite polymeric materials: A comprehensive review. Adv Colloid Interface Sci 2023; 318:102955. [PMID: 37467558 DOI: 10.1016/j.cis.2023.102955] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/23/2023] [Accepted: 06/20/2023] [Indexed: 07/21/2023]
Abstract
The modern eco-friendly materials used in research and innovation today consist of nanocomposites and bio-nanocomposite polymers. Their unique composite properties make them suitable for various industrial, medicinal, and energy applications. Bio-nanocomposite polymers are made of biopolymer matrices that have nanofillers dispersed throughout them. There are several types of fillers that can be added to polymers to enhance their quality, such as cellulose-based fillers, clay nanomaterials, carbon black, talc, carbon quantum dots, and many others. Biopolymer-based nanocomposites are considered a superior alternative to traditional materials as they reduce reliance on fossil fuels and promote the use of renewable resources. This review covers the current state-of-the-art in nanocomposite and bio-nanocomposite materials, focusing on ways to improve their features and the various applications they can be used for. The review article also investigates the utilization of diverse nanocomposites as a viable approach for developing bio-nanocomposites. It delves into the underlying principles that govern the synthesis of these materials and explores their prospective applications in the biomedical field, food packaging, sensing (Immunosensors), and energy storage devices. Lastly, the review discusses the future outlook and current challenges of these materials, with a focus on sustainability.
Collapse
Affiliation(s)
- Abdul Hakeem Anwer
- School of Mechanical Engineering, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Afreen Ahtesham
- School of Chemical Sciences University Sains Malaysia, Penang, Malaysia
| | - Mohd Shoeb
- School of Mechanical Engineering, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Fouzia Mashkoor
- School of Mechanical Engineering, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Mohd Zahid Ansari
- School of Materials Science and Engineering, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, Republic of Korea
| | - Shushuai Zhu
- School of Mechanical Engineering, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Changyoon Jeong
- School of Mechanical Engineering, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| |
Collapse
|
11
|
Xu X, Zhao L, Terry PD, Chen J. Reciprocal Effect of Environmental Stimuli to Regulate the Adipogenesis and Osteogenesis Fate Decision in Bone Marrow-Derived Mesenchymal Stem Cells (BM-MSCs). Cells 2023; 12:1400. [PMID: 37408234 PMCID: PMC10216952 DOI: 10.3390/cells12101400] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/02/2023] [Accepted: 05/12/2023] [Indexed: 07/07/2023] Open
Abstract
Mesenchymal stem cells derived from bone marrow (BM-MSCs) can differentiate into adipocytes and osteoblasts. Various external stimuli, including environmental contaminants, heavy metals, dietary, and physical factors, are shown to influence the fate decision of BM-MSCs toward adipogenesis or osteogenesis. The balance of osteogenesis and adipogenesis is critical for the maintenance of bone homeostasis, and the interruption of BM-MSCs lineage commitment is associated with human health issues, such as fracture, osteoporosis, osteopenia, and osteonecrosis. This review focuses on how external stimuli shift the fate of BM-MSCs towards adipogenesis or osteogenesis. Future studies are needed to understand the impact of these external stimuli on bone health and elucidate the underlying mechanisms of BM-MSCs differentiation. This knowledge will inform efforts to prevent bone-related diseases and develop therapeutic approaches to treat bone disorders associated with various pathological conditions.
Collapse
Affiliation(s)
- Xinyun Xu
- Department of Nutrition, The University of Tennessee, Knoxville, TN 37996, USA
| | - Ling Zhao
- Department of Nutrition, The University of Tennessee, Knoxville, TN 37996, USA
| | - Paul D. Terry
- Department of Medicine, Graduate School of Medicine, The University of Tennessee, Knoxville, TN 37920, USA;
| | - Jiangang Chen
- Department of Public Health, The University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
12
|
The role of hypertrophic chondrocytes in regulation of the cartilage-to-bone transition in fracture healing. Bone Rep 2022; 17:101616. [PMID: 36105852 PMCID: PMC9465425 DOI: 10.1016/j.bonr.2022.101616] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/05/2022] [Accepted: 06/07/2022] [Indexed: 11/23/2022] Open
Abstract
Endochondral bone formation is an important pathway in fracture healing, involving the formation of a cartilaginous soft callus and the process of cartilage-to-bone transition. Failure or delay in the cartilage-to-bone transition causes an impaired bony union such as nonunion or delayed union. During the healing process, multiple types of cells including chondrocytes, osteoprogenitors, osteoblasts, and endothelial cells coexist in the callus, and inevitably crosstalk with each other. Hypertrophic chondrocytes located between soft cartilaginous callus and bony hard callus mediate the crosstalk regulating cell-matrix degradation, vascularization, osteoclast recruitment, and osteoblast differentiation in autocrine and paracrine manners. Furthermore, hypertrophic chondrocytes can become osteoprogenitors and osteoblasts, and directly contribute to woven bone formation. In this review, we focus on the roles of hypertrophic chondrocytes in fracture healing and dissect the intermingled crosstalk in fracture callus during the cartilage-to-bone transition.
Collapse
|
13
|
Zhao XX, Xie WQ, Xiao WF, Li HZ, Naranmandakh S, Bruyere O, Reginster JY, Li YS. Perlecan: Roles in osteoarthritis and potential treating target. Life Sci 2022; 312:121190. [PMID: 36379311 DOI: 10.1016/j.lfs.2022.121190] [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: 09/10/2022] [Revised: 11/05/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022]
Abstract
Osteoarthritis (OA) is the most common joint disease, affecting hundreds of millions of people globally, which leads to a high cost of treatment and further medical care and an apparent decrease in patient prognosis. The recent view of OA pathogenesis is that increased vascularity, bone remodeling, and disordered turnover are influenced by multivariate risk factors, such as age, obesity, and overloading. The view also reveals the gap between the development of these processes and early stage risk factors. This review presents the latest research on OA-related signaling pathways and analyzes the potential roles of perlecan, a typical component of the well-known protective structure against osteoarthritic pericellular matrix (PCM). Based on the experimental results observed in end-stage OA models, we summarized and analyzed the role of perlecan in the development of OA. In normal cartilage, it plays a protective role by maintaining the integrin of PCM and sequesters growth factors. Second, perlecan in cartilage is required to not only activate vascular epithelium growth factor receptor (VEGFR) signaling of endothelial cells for vascular invasion and catabolic autophagy, but also for different signaling pathways for the catabolic and anabolic actions of chondrocytes. Finally, perlecan may participate in pain sensitization pathways.
Collapse
Affiliation(s)
- Xiao-Xuan Zhao
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Xiangya School of Medicine, Central South University, Changsha 410083, Hunan, China
| | - Wen-Qing Xie
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Wen-Feng Xiao
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Heng-Zhen Li
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Shinen Naranmandakh
- School of Arts and Sciences, National University of Mongolia, Sukhbaatar district, 14201 Ulaanbaatar, Mongolia
| | - Olivier Bruyere
- Department of Public Health, Epidemiology and Health Economics, University of Liège, CHU Sart Tilman B23, 4000 Liège, Belgium
| | - Jean-Yves Reginster
- Department of Public Health, Epidemiology and Health Economics, University of Liège, CHU Sart Tilman B23, 4000 Liège, Belgium.
| | - Yu-Sheng Li
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| |
Collapse
|
14
|
Sandanamsamy L, Harun WSW, Ishak I, Romlay FRM, Kadirgama K, Ramasamy D, Idris SRA, Tsumori F. A comprehensive review on fused deposition modelling of polylactic acid. PROGRESS IN ADDITIVE MANUFACTURING 2022; 8:1-25. [PMID: 38625345 PMCID: PMC9619022 DOI: 10.1007/s40964-022-00356-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 10/15/2022] [Indexed: 05/13/2023]
Abstract
Fused Deposition Modelling (FDM) is one of the additive manufacturing (AM) techniques that have emerged as the most feasible and prevalent approach for generating functional parts due to its ability to produce neat and intricate parts. FDM mainly utilises one of the widely used polymers, polylactic acid, also known as polylactide (PLA). It is an aliphatic polyester material and biocompatible thermoplastic, with the best design prospects due to its eco-friendly properties; when PLA degrades, it breaks down into water and carbon dioxide, neither of which are hazardous to the environment. However, PLA has its limitations of poor mechanical properties. Therefore, a filler reinforcement may enhance the characteristics of PLA and produce higher-quality FDM-printed parts. The processing parameters also play a significant role in the final result of the printed parts. This review aims to study and discover the properties of PLA and the optimum processing parameters. This review covers PLA in FDM, encompassing its mechanical properties, processing parameters, characterisation, and applications. A comprehensive description of FDM processing parameters is outlined as it plays a vital role in determining the quality of a printed product. In addition, PLA polymer is highly desirable for various field industrial applications such as in a medical, automobile, and electronic, given its excellent thermoplastic and biodegradability properties.
Collapse
Affiliation(s)
- L. Sandanamsamy
- Department of Mechanical Engineering, College of Engineering, Universiti Malaysia Pahang, Gambang, 26300 Kuantan, Pahang Malaysia
| | - W. S. W. Harun
- Department of Mechanical Engineering, College of Engineering, Universiti Malaysia Pahang, Gambang, 26300 Kuantan, Pahang Malaysia
| | - I. Ishak
- Faculty of Manufacturing and Mechatronic Engineering Technology, Universiti Malaysia Pahang, 26600 Pekan, Malaysia
| | - F. R. M. Romlay
- Faculty of Mechanical and Automotive Engineering Technology, Universiti Malaysia Pahang, 26600 Pekan, Pahang Malaysia
| | - K. Kadirgama
- Faculty of Mechanical and Automotive Engineering Technology, Universiti Malaysia Pahang, 26600 Pekan, Pahang Malaysia
| | - D. Ramasamy
- Department of Mechanical Engineering, College of Engineering, Universiti Malaysia Pahang, Gambang, 26300 Kuantan, Pahang Malaysia
| | - S. R. A. Idris
- Faculty of Mechanical and Automotive Engineering Technology, Universiti Malaysia Pahang, 26600 Pekan, Pahang Malaysia
| | - F. Tsumori
- Department of Aeronautics and Astronautics, Faculty of Engineering, Kyushu University, 744 Motooka Nishi-Ku, Fukuoka, 819-0395 Japan
| |
Collapse
|
15
|
Tosun B, Wolff LI, Houben A, Nutt S, Hartmann C. Osteoclasts and Macrophages-Their Role in Bone Marrow Cavity Formation During Mouse Embryonic Development. J Bone Miner Res 2022; 37:1761-1774. [PMID: 35689447 DOI: 10.1002/jbmr.4629] [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: 01/20/2022] [Revised: 05/19/2022] [Accepted: 06/04/2022] [Indexed: 11/09/2022]
Abstract
The formation of the bone marrow cavity is a prerequisite for endochondral ossification. In reviews and textbooks, it is occasionally reported that osteoclasts are essential for bone marrow cavity formation removing hypertrophic chondrocytes. Mice lacking osteoclasts or having functionally defective osteoclasts have osteopetrotic bones, yet they still form a bone marrow cavity. Here, we investigated the role of osteoclasts and macrophages in bone marrow cavity formation during embryogenesis. Macrophages can assist osteoclasts in matrix removal by phagocytosing resorption byproducts. Rank-deficient mice, lacking osteoclasts, and Pu.1-deficient mice, lacking monocytes, macrophages, and osteoclasts, displayed a delay in bone marrow cavity formation and a lengthening of the zone of hypertrophic chondrocytes. F4/80-positive monocyte/macrophage numbers increased by about fourfold in the bone marrow cavity of E18.5 Rank-deficient mice. Based on lineage-tracing experiments, the majority of the excess F4/80 cells were derived from definitive hematopoietic precursors of the fetal liver. In long bones of both Rank-/- and Pu.1-/- specimens, Mmp9-positive cells were still present. In addition to monocytes, macrophages, and osteoclasts, Ctsb-positive septoclasts were lost in Pu.1-/- specimens. The mineralization pattern was altered in Rank-/- and Pu.1-/- specimens, revealing a significant rise in transverse-oriented mineralized structures. Taken together, our findings imply that early on during bone marrow cavity formation, osteoclasts facilitate the entry of blood vessels and later the turnover of hypertrophic chondrocytes, whereas macrophages appear to play no major role. Furthermore, the absence of septoclasts in Pu.1-/- specimens suggests that septoclasts are either derived from Pu.1-dependent precursors or require PU.1 activity for their differentiation. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Benjamin Tosun
- Institute of Musculoskeletal Medicine, Department of Bone and Skeletal Research, Medical Faculty of the Westphalian Wilhelms University, Münster, Germany
| | - Lena Ingeborg Wolff
- Institute of Musculoskeletal Medicine, Department of Bone and Skeletal Research, Medical Faculty of the Westphalian Wilhelms University, Münster, Germany
| | - Astrid Houben
- Institute of Musculoskeletal Medicine, Department of Bone and Skeletal Research, Medical Faculty of the Westphalian Wilhelms University, Münster, Germany
| | - Stephen Nutt
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Christine Hartmann
- Institute of Musculoskeletal Medicine, Department of Bone and Skeletal Research, Medical Faculty of the Westphalian Wilhelms University, Münster, Germany
| |
Collapse
|
16
|
Khokhani P, Belluomo R, Croes M, Gawlitta D, Kruyt MC, Weinans H. An in-vitro model to test the influence of immune cell secretome on MSC osteogenic differentiation. Tissue Eng Part C Methods 2022; 28:420-430. [PMID: 35770885 DOI: 10.1089/ten.tec.2022.0086] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Immune cells and their soluble factors have an important role in the bone healing process. Modulation of the immune response, therefore, offers a potential strategy to enhance bone formation. To investigate the influence of the immune system on osteogenesis, we developed and applied an in-vitro model that incorporates both innate and adaptive immune cells. Human peripheral blood mononuclear cells (PBMCs) were isolated and cultured for 24 hours and subsequently stimulated with immune-modulatory agents; C-class CpG oligodeoxynucleotide (CpG ODN C), Polyinosinic acid-polycytidylic acid Poly(I:C), and lipopolysaccharide (LPS); all pathogen recognition receptor agonists, and that target Toll-like receptors TLR9, -3, and -4, respectively. The conditioned medium obtained from PBMCs after 24 hours was used to investigate its effects on the metabolic activity and osteogenic differentiation capacity of human bone marrow-derived mesenchymal stromal cells (MSCs). Conditioned media from unstimulated PBMCs did not affect the metabolic activity and osteogenic differentiation capacity of MSCs. The conditioned medium from CpG ODN C and LPS stimulated PBMCs increased alkaline phosphatase activity of MSCs by approximately 3-fold as compared to the unstimulated control, whereas Poly(I:C) conditioned medium did not enhance ALP activity of MSCs. Moreover, direct stimulation of MSCs with the immune-modulatory stimuli did not result in increased alkaline phosphatase activity. These results demonstrate that soluble factors present in conditioned medium from PBMCs stimulated with immune-modulatory factors enhance osteogenesis of MSCs. This in-vitro model can serve as a tool in screening immune-modulatory stimulants from a broad variety of immune cells for (indirect) effects on osteogenesis and also to identify soluble factors from multiple immune cell types that may modulate bone healing.
Collapse
Affiliation(s)
- Paree Khokhani
- University Medical Centre Utrecht, 8124, Orthopedics , UMC Utrecht, dept. Orthopedics, G5.203, Heidelberglaan 100, Utrecht, Utrecht, Drenthe, Netherlands, 3584CX.,University Medical Centre, Utrecht (UMCU), UMC Utrecht, dept. Orthopedics, G5.203, Heidelberglaan 100, Netherlands;
| | - Ruggero Belluomo
- University Medical Centre Utrecht, 8124, Orthopedics , Utrecht, Utrecht, Netherlands;
| | - Michiel Croes
- University Medical Centre Utrecht, 8124, Orthopedics , Utrecht, Utrecht, Netherlands;
| | - Debby Gawlitta
- University Medical Center Utrecht, Oral and Maxillofacial Surgery, Prosthodontics & Special Dental Care, Heidelberglaan 100, G05.129, PO Box 85500, Utrecht, Netherlands, 3508 GA;
| | - Moyo C Kruyt
- University medical center Utrecht, Orthopedics, HP G 05.228, PO Box 85500, Utrecht, Netherlands, 3508 GA;
| | - Harrie Weinans
- University Medical Centre Utrecht, 8124, Orthopedics, Utrecht, Utrecht, Netherlands;
| |
Collapse
|
17
|
Smith SS, Chu D, Qu T, Aggleton JA, Schneider RA. Species-specific sensitivity to TGFβ signaling and changes to the Mmp13 promoter underlie avian jaw development and evolution. eLife 2022; 11:e66005. [PMID: 35666955 PMCID: PMC9246370 DOI: 10.7554/elife.66005] [Citation(s) in RCA: 3] [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: 12/22/2020] [Accepted: 06/03/2022] [Indexed: 12/02/2022] Open
Abstract
Precise developmental control of jaw length is critical for survival, but underlying molecular mechanisms remain poorly understood. The jaw skeleton arises from neural crest mesenchyme (NCM), and we previously demonstrated that these progenitor cells express more bone-resorbing enzymes including Matrix metalloproteinase 13 (Mmp13) when they generate shorter jaws in quail embryos versus longer jaws in duck. Moreover, if we inhibit bone resorption or Mmp13, we can increase jaw length. In the current study, we uncover mechanisms establishing species-specific levels of Mmp13 and bone resorption. Quail show greater activation of and sensitivity to transforming growth factor beta (TGFβ) signaling than duck; where intracellular mediators like SMADs and targets like Runt-related transcription factor 2 (Runx2), which bind Mmp13, become elevated. Inhibiting TGFβ signaling decreases bone resorption, and overexpressing Mmp13 in NCM shortens the duck lower jaw. To elucidate the basis for this differential regulation, we examine the Mmp13 promoter. We discover a SMAD-binding element and single nucleotide polymorphisms (SNPs) near a RUNX2-binding element that distinguish quail from duck. Altering the SMAD site and switching the SNPs abolish TGFβ sensitivity in the quail Mmp13 promoter but make the duck promoter responsive. Thus, differential regulation of TGFβ signaling and Mmp13 promoter structure underlie avian jaw development and evolution.
Collapse
Affiliation(s)
- Spenser S Smith
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, United States
| | - Daniel Chu
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, United States
| | - Tiange Qu
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, United States
| | - Jessye A Aggleton
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, United States
| | - Richard A Schneider
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
18
|
Duncan HF, Kobayashi Y, Yamauchi Y, Quispe-Salcedo A, Chao Feng Z, Huang J, Partridge NC, Nakatani T, D’Armiento J, Shimizu E. The Critical Role of MMP13 in Regulating Tooth Development and Reactionary Dentinogenesis Repair Through the Wnt Signaling Pathway. Front Cell Dev Biol 2022; 10:883266. [PMID: 35531096 PMCID: PMC9068941 DOI: 10.3389/fcell.2022.883266] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/14/2022] [Indexed: 12/20/2022] Open
Abstract
Matrix-metalloproteinase-13 (MMP13) is important for bone formation and remodeling; however, its role in tooth development remains unknown. To investigate this, MMP13-knockout (Mmp13−/−) mice were used to analyze phenotypic changes in the dentin–pulp complex, mineralization-associated marker-expression, and mechanistic interactions. Immunohistochemistry demonstrated high MMP13-expression in pulp-tissue, ameloblasts, odontoblasts, and dentin in developing WT-molars, which reduced in adults, with human-DPC cultures demonstrating a >2000-fold increase in Mmp13-expression during mineralization. Morphologically, Mmp13−/− molars displayed critical alterations in the dentin-phenotype, affecting dentin-tubule regularity, the odontoblast-palisade and predentin-definition with significantly reduced dentin volume (∼30% incisor; 13% molar), and enamel and dentin mineral-density. Reactionary-tertiary-dentin in response to injury was reduced at Mmp13−/− molar cusp-tips but with significantly more dystrophic pulpal mineralization in MMP13-null samples. Odontoblast differentiation-markers, nestin and DSP, reduced in expression after MMP13-loss in vivo, with reduced calcium deposition in MMP13-null DPC cultures. RNA-sequencing analysis of WT and Mmp13−/− pulp highlighted 5,020 transcripts to have significantly >2.0-fold change, with pathway-analysis indicating downregulation of the Wnt-signaling pathway, supported by reduced in vivo expression of the Wnt-responsive gene Axin2. Mmp13 interaction with Axin2 could be partly responsible for the loss of odontoblastic activity and alteration to the tooth phenotype and volume which is evident in this study. Overall, our novel findings indicate MMP13 as critical for tooth development and mineralization processes, highlighting mechanistic interaction with the Wnt-signaling pathway.
Collapse
Affiliation(s)
- Henry F. Duncan
- Division of Restorative Dentistry & Periodontology, Dublin Dental University Hospital, Trinity College Dublin, Dublin, Ireland
- *Correspondence: Henry F. Duncan, ; Emi Shimizu,
| | - Yoshifumi Kobayashi
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, United States
| | - Yukako Yamauchi
- Division of Restorative Dentistry & Periodontology, Dublin Dental University Hospital, Trinity College Dublin, Dublin, Ireland
| | | | - Zhi Chao Feng
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, United States
| | - Jia Huang
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, United States
| | - Nicola C. Partridge
- Department of Molecular Pathobiology, New York University Dentistry, New York, NY, United States
| | - Teruyo Nakatani
- Department of Molecular Pathobiology, New York University Dentistry, New York, NY, United States
| | - Jeanine D’Armiento
- Department of Physiology and Cellular Biophysics, Columbia University Medical Centre, New York, NY, United States
| | - Emi Shimizu
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, United States
- *Correspondence: Henry F. Duncan, ; Emi Shimizu,
| |
Collapse
|
19
|
Owen-Woods C, Kusumbe A. Fundamentals of bone vasculature: Specialization, interactions and functions. Semin Cell Dev Biol 2022; 123:36-47. [PMID: 34281770 DOI: 10.1016/j.semcdb.2021.06.025] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 02/06/2023]
Abstract
Angiogenesis, hematopoiesis and osteogenesis are fundamental processes mediating complex and essential biological functions. In the bone marrow, endothelial cells (ECs) are a principal mediator of regulatory signals that govern hematopoietic and mesenchymal stem cells. EC and osteoblast interactions and niche functions of ECs are fundamental in maintaining bone health and coordinating repair and regeneration following injury. These cellular interactions are subject to dysregulation and deterioration under stress, aging, chronic disease states and malignancy. Thus, the prospect of manipulating the bone vasculature has tremendous potential to advance therapeutic interventions for the management of bone diseases. This review discusses the current state of vascular-skeletal tissue interactions focusing on osteoblast and hematopoietic stem cells interaction with ECs.
Collapse
Affiliation(s)
- Charlotte Owen-Woods
- Tissue and Tumor Microenvironments Group, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - Anjali Kusumbe
- Tissue and Tumor Microenvironments Group, NDORMS, University of Oxford, Oxford OX3 7FY, UK.
| |
Collapse
|
20
|
Gomathi K, Rohini M, Partridge NC, Selvamurugan N. Regulation of transforming growth factor-β1-stimulation of Runx2 acetylation for matrix metalloproteinase 13 expression in osteoblastic cells. Biol Chem 2022; 403:305-315. [PMID: 34643076 DOI: 10.1515/hsz-2021-0292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 09/30/2021] [Indexed: 01/12/2023]
Abstract
Transforming growth factor beta 1 (TGF-β1) functions as a coupling factor between bone development and resorption. Matrix metalloproteinase 13 (MMP13) is important in bone remodeling, and skeletal dysplasia is caused by a deficiency in MMP13 expre-ssion. Runx2, a transcription factor is essential for bone development, and MMP13 is one of its target genes. TGF-β1 promoted Runx2 phosphorylation, which was necessary for MMP13 production in osteoblastic cells, as we previously shown. Since the phosphorylation of some proteins causes them to be degraded by the ubiquitin/proteasome pathway, we hypothesized that TGF-β1 might stabilize the phosphorylated Runx2 protein for its activity by other post-translational modification (PTM). This study demonstrated that TGF-β1-stimulated Runx2 acetylation in rat osteoblastic cells. p300, a histone acetyltransferase interacted with Runx2, and it promoted Runx2 acetylation upon TGF-β1-treatment in these cells. Knockdown of p300 decreased the TGF-β1-stimulated Runx2 acetylation and MMP13 expression in rat osteoblastic cells. TGF-β1-treatment stimulated the acetylated Runx2 bound at the MMP13 promoter, and knockdown of p300 reduced this effect in these cells. Overall, our studies identified the transcriptional regulation of MMP13 by TGF-β1 via Runx2 acetylation in rat osteoblastic cells, and these findings contribute to the knowledge of events presiding bone metabolism.
Collapse
Affiliation(s)
- Kanagaraj Gomathi
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Muthukumar Rohini
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Nicola C Partridge
- Department of Molecular Pathobiology, New York University College Dentistry, New York, NY, USA
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| |
Collapse
|
21
|
Liu R, Imangali N, Ethiraj LP, Carney TJ, Winkler C. Transcriptome Profiling of Osteoblasts in a Medaka ( Oryzias latipes) Osteoporosis Model Identifies Mmp13b as Crucial for Osteoclast Activation. Front Cell Dev Biol 2022; 10:775512. [PMID: 35281094 PMCID: PMC8911226 DOI: 10.3389/fcell.2022.775512] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 01/14/2022] [Indexed: 11/13/2022] Open
Abstract
Matrix metalloproteases (MMPs) play crucial roles in extracellular matrix (ECM) modulation during osteoclast-driven bone remodeling. In the present study, we used transcriptome profiling of bone cells in a medaka model for osteoporosis and bone regeneration to identify factors critical for bone remodeling and homeostasis. This identified mmp13b, which was strongly expressed in osteoblast progenitors and upregulated under osteoporotic conditions and during regeneration of bony fin rays. To characterize the role of mmp13b in bone remodeling, we generated medaka mmp13b mutants by CRISPR/Cas9. We found that mmp13b mutants form normal numbers of osteoblasts and osteoclasts. However, osteoclast activity was severely impaired under osteoporotic conditions. In mmp13b mutants and embryos treated with the MMP13 inhibitor CL-82198, unmineralized collagens and mineralized bone matrix failed to be degraded. In addition, the dynamic migratory behavior of activated osteoclasts was severely affected in mmp13b mutants. Expression analysis showed that maturation genes were downregulated in mmp13b deficient osteoclasts suggesting that they remain in an immature and non-activated state. We also found that fin regeneration was delayed in mmp13b mutants with a concomitant alteration of the ECM and reduced numbers of osteoblast progenitors in regenerating joint regions. Together, our findings suggest that osteoblast-derived Mmp13b alters the bone ECM to allow the maturation and activation of osteoclasts during bone remodeling in a paracrine manner. Mmp13b-induced ECM alterations are also required to facilitate osteoblast progenitor recruitment and full regeneration of bony fin rays.
Collapse
Affiliation(s)
- Ranran Liu
- Department of Biological Sciences and Centre for Bioimaging Sciences, National University of Singapore, Singapore, Singapore
| | - Nurgul Imangali
- Department of Biological Sciences and Centre for Bioimaging Sciences, National University of Singapore, Singapore, Singapore
| | - Lalith Prabha Ethiraj
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Tom James Carney
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Christoph Winkler
- Department of Biological Sciences and Centre for Bioimaging Sciences, National University of Singapore, Singapore, Singapore
| |
Collapse
|
22
|
Regulation of FGF-2, FGF-18 and Transcription Factor Activity by Perlecan in the Maturational Development of Transitional Rudiment and Growth Plate Cartilages and in the Maintenance of Permanent Cartilage Homeostasis. Int J Mol Sci 2022; 23:ijms23041934. [PMID: 35216048 PMCID: PMC8872392 DOI: 10.3390/ijms23041934] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/24/2022] [Accepted: 02/01/2022] [Indexed: 12/11/2022] Open
Abstract
The aim of this study was to highlight the roles of perlecan in the regulation of the development of the rudiment developmental cartilages and growth plate cartilages, and also to show how perlecan maintains permanent articular cartilage homeostasis. Cartilage rudiments are transient developmental templates containing chondroprogenitor cells that undergo proliferation, matrix deposition, and hypertrophic differentiation. Growth plate cartilage also undergoes similar changes leading to endochondral bone formation, whereas permanent cartilage is maintained as an articular structure and does not undergo maturational changes. Pericellular and extracellular perlecan-HS chains interact with growth factors, morphogens, structural matrix glycoproteins, proteases, and inhibitors to promote matrix stabilization and cellular proliferation, ECM remodelling, and tissue expansion. Perlecan has mechanotransductive roles in cartilage that modulate chondrocyte responses in weight-bearing environments. Nuclear perlecan may modulate chromatin structure and transcription factor access to DNA and gene regulation. Snail-1, a mesenchymal marker and transcription factor, signals through FGFR-3 to promote chondrogenesis and maintain Acan and type II collagen levels in articular cartilage, but prevents further tissue expansion. Pre-hypertrophic growth plate chondrocytes also express high Snail-1 levels, leading to cessation of Acan and CoI2A1 synthesis and appearance of type X collagen. Perlecan differentially regulates FGF-2 and FGF-18 to maintain articular cartilage homeostasis, rudiment and growth plate cartilage growth, and maturational changes including mineralization, contributing to skeletal growth.
Collapse
|
23
|
Erickson CB, Hill R, Pascablo D, Kazakia G, Hansen K, Bahney C. A timeseries analysis of the fracture callus extracellular matrix proteome during bone fracture healing. JOURNAL OF LIFE SCIENCES (WESTLAKE VILLAGE, CALIF.) 2021; 3:1-30. [PMID: 35765657 PMCID: PMC9236279 DOI: 10.36069/jols/20220601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
While most bones fully self-heal, certain diseases require bone allograft to assist with fracture healing. Bone allografts offer promise as treatments for such fractures due to their osteogenic properties. However, current bone allografts made of decellularized bone extracellular matrix (ECM) have high failure rates, and thus grafts which improve fracture healing outcomes are needed. Understanding specific changes to the ECM proteome during normal fracture healing would enable the identification of key proteins that could be used enhance osteogenicity of bone allograft. Here, we performed a timeseries analysis of the fracture callus in mice to investigate proteomic and mineralization changes to the ECM at key stages of fracture healing. We found that changes to the ECM proteome largely coincide with the distinct phases of fracture healing. Basement membrane proteins (AGRN, COL4, LAMA), cartilage proteins (COL2A1, ACAN), and collagen crosslinking enzymes (LOXL, PLOD, ITIH) were initially upregulated, followed by bone specific proteoglycans and collagens (IBSP, COL1A1). Various tissue proteases (MMP2, 9, 13, 14; CTSK, CTSG, ELANE) were expressed at different levels throughout fracture healing. These changes coordinated with mineralization of the fracture callus, which increased steeply during the initial stages of healing. Interestingly the later timepoint was characterized by a response to wound healing and high expression of clotting factors (F2, 7, 9, 10). We identified ELANE and ITIH2 as tissue remodeling enzymes having no prior known involvement with fracture healing. This data can be further mined to identify regenerative proteins for enhanced bone graft design.
Collapse
Affiliation(s)
- Christopher B. Erickson
- Department of Biochemistry and Molecular Genetics,University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Ryan Hill
- Department of Biochemistry and Molecular Genetics,University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Donna Pascablo
- Orthopaedic Trauma Institute, University of California, San Francisco (UCSF), San Francisco, CA
| | - Galateia Kazakia
- Department of Radiology and Biomedical Imaging, University of California, San Francisco (UCSF), San Francisco, CA
| | - Kirk Hansen
- Department of Biochemistry and Molecular Genetics,University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Chelsea Bahney
- Stedman Philippon Research Institute (SPRI), Center for Regenerative and Personalized Medicine. Vail, CO
- Orthopaedic Trauma Institute, University of California, San Francisco (UCSF), San Francisco, CA
| |
Collapse
|
24
|
Li C, Zhao H, Cheng L, Wang B. Allogeneic vs. autologous mesenchymal stem/stromal cells in their medication practice. Cell Biosci 2021; 11:187. [PMID: 34727974 PMCID: PMC8561357 DOI: 10.1186/s13578-021-00698-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/12/2021] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem/stromal cell (MSC)-based therapeutics is already available for treatment of a range of diseases or medical conditions. Autologous or allogeneic MSCs obtained from self or donors have their own advantages and disadvantages in their medical practice. Therapeutic benefits of using autologous vs. allogeneic MSCs are inconclusive. Transplanted MSCs within the body interact with their physical microenvironment or niche, physiologically or pathologically, and such cells in a newly established tissue microenvironment may be impacted by the pathological harmful environmental factors to alter their unique biological behaviors. Meanwhile, a temporary microenvironment/niche may be also altered by the resident or niche-surrounding MSCs. Therefore, the functional plasticity and heterogeneity of MSCs caused by different donors and subpopulations of MSCs may result in potential uncertainty in their safe and efficacious medical practice. Acknowledging a connection between MSCs' biology and their existing microenvironment, donor-controlled clinical practice for the long-term therapeutic benefit is suggested to further consider minimizing MSCs potential harm for MSC-based individual therapies. In this review, we summarize the advantages and disadvantages of autologous vs. allogeneic MSCs in their therapeutic applications. Among other issues, we highlight the importance of better understanding of the various microenvironments that may affect the properties of niche-surrounding MSCs and discuss the clinical applications of MSCs within different contexts for treatment of different diseases including cardiomyopathy, lupus and lupus nephritis, diabetes and diabetic complications, bone and cartilage repair, cancer and tissue fibrosis.
Collapse
Affiliation(s)
- Chenghai Li
- Stem Cell Program of Clinical Research Center, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China.
| | - Hua Zhao
- Institute of Reproductive Medicine, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China
| | - Linna Cheng
- Institute of Hematology, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China
| | - Bin Wang
- Department of Neurosurgery, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China.
| |
Collapse
|
25
|
Valdés-Fernández J, López-Martínez T, Ripalda-Cemboráin P, Calvo IA, Sáez B, Romero-Torrecilla JA, Aldazabal J, Muiños-López E, Montiel V, Orbe J, Rodríguez JA, Páramo JA, Prósper F, Granero-Moltó F. Molecular and Cellular Mechanisms of Delayed Fracture Healing in Mmp10 (Stromelysin 2) Knockout Mice. J Bone Miner Res 2021; 36:2203-2213. [PMID: 34173256 DOI: 10.1002/jbmr.4403] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 06/15/2021] [Accepted: 06/20/2021] [Indexed: 11/08/2022]
Abstract
The remodeling of the extracellular matrix is a central function in endochondral ossification and bone homeostasis. During secondary fracture healing, vascular invasion and bone growth requires the removal of the cartilage intermediate and the coordinate action of the collagenase matrix metalloproteinase (MMP)-13, produced by hypertrophic chondrocytes, and the gelatinase MMP-9, produced by cells of hematopoietic lineage. Interfering with these MMP activities results in impaired fracture healing characterized by cartilage accumulation and delayed vascularization. MMP-10, Stromelysin 2, a matrix metalloproteinase with high homology to MMP-3 (Stromelysin 1), presents a wide range of putative substrates identified in vitro, but its targets and functions in vivo and especially during fracture healing and bone homeostasis are not well defined. Here, we investigated the role of MMP-10 through bone regeneration in C57BL/6 mice. During secondary fracture healing, MMP-10 is expressed by hematopoietic cells and its maximum expression peak is associated with cartilage resorption at 14 days post fracture (dpf). In accordance with this expression pattern, when Mmp10 is globally silenced, we observed an impaired fracture-healing phenotype at 14 dpf, characterized by delayed cartilage resorption and TRAP-positive cell accumulation. This phenotype can be rescued by a non-competitive transplant of wild-type bone marrow, indicating that MMP-10 functions are required only in cells of hematopoietic linage. In addition, we found that this phenotype is a consequence of reduced gelatinase activity and the lack of proMMP-9 processing in macrophages. Our data provide evidence of the in vivo function of MMP-10 during endochondral ossification and defines the macrophages as the lead cell population in cartilage removal and vascular invasion. © 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
| | | | - Purificación Ripalda-Cemboráin
- Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain.,Department of Orthopaedic Surgery and Traumatology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Isabel A Calvo
- Hematology-Oncology Program, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Borja Sáez
- Hematology-Oncology Program, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | | | - Javier Aldazabal
- Tissue Engineering Group, TECNUN-Universidad de Navarra, San Sebastián, Spain
| | | | - Verónica Montiel
- Department of Orthopaedic Surgery and Traumatology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Josune Orbe
- Atherotrombosis, Cardiovascular Disease Program, CIMA, Instituto de Investigación Sanitaria de Navarra (IdiSNA), CIBERCV, Pamplona, Spain
| | - José Antonio Rodríguez
- Atherotrombosis, Cardiovascular Disease Program, CIMA, Instituto de Investigación Sanitaria de Navarra (IdiSNA), CIBERCV, Pamplona, Spain
| | - José Antonio Páramo
- Atherotrombosis, Cardiovascular Disease Program, CIMA, Instituto de Investigación Sanitaria de Navarra (IdiSNA), CIBERCV, Pamplona, Spain.,Department of Hematology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Felipe Prósper
- Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain.,Hematology-Oncology Program, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.,Department of Hematology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Froilán Granero-Moltó
- Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain.,Department of Orthopaedic Surgery and Traumatology, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
26
|
Uda Y, Spatz JM, Hussein A, Garcia JH, Lai F, Dedic C, Fulzele K, Dougherty S, Eberle M, Adamson C, Misener L, Gerstenfeld L, Divieti Pajevic P. Global transcriptomic analysis of a murine osteocytic cell line subjected to spaceflight. FASEB J 2021; 35:e21578. [PMID: 33835498 DOI: 10.1096/fj.202100059r] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/13/2021] [Accepted: 03/22/2021] [Indexed: 01/18/2023]
Abstract
Bone loss is a major health concern for astronauts during long-term spaceflight and for patients during prolonged bed rest or paralysis. Growing evidence suggests that osteocytes, the most abundant cells in the mineralized bone matrix, play a key role in sensing mechanical forces applied to the skeleton and integrating the orchestrated response into subcellular biochemical signals to modulate bone homeostasis. However, the precise molecular mechanisms underlying both mechanosensation and mechanotransduction in late-osteoblast-to-osteocyte cells under microgravity (µG) have yet to be elucidated. To unravel the mechanisms by which late osteoblasts and osteocytes sense and respond to mechanical unloading, we exposed the osteocytic cell line, Ocy454, to 2, 4, or 6 days of µG on the SpaceX Dragon-6 resupply mission to the International Space Station. Our results showed that µG impairs the differentiation of osteocytes, consistent with prior osteoblast spaceflight experiments, which resulted in the downregulation of key osteocytic genes. Importantly, we demonstrate the modulation of critical glycolysis pathways in osteocytes subjected to microgravity and discovered a set of mechanical sensitive genes that are consistently regulated in multiple cell types exposed to microgravity suggesting a common, yet to be fully elucidated, genome-wide response to microgravity. Ground-based simulated microgravity experiments utilizing the NASA rotating-wall-vessel were unable to adequately replicate the changes in microgravity exposure highlighting the importance of spaceflight missions to understand the unique environmental stress that microgravity presents to diverse cell types. In summary, our findings demonstrate that osteocytes respond to µG with an increase in glucose metabolism and oxygen consumption.
Collapse
Affiliation(s)
- Yuhei Uda
- Department of Translational Dental Medicine, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, USA
| | - Jordan M Spatz
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Amira Hussein
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA, USA
| | - Joseph H Garcia
- School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Forest Lai
- Department of Translational Dental Medicine, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, USA
| | - Chris Dedic
- Department of Translational Dental Medicine, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, USA
| | - Keertik Fulzele
- Department of Translational Dental Medicine, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, USA
| | | | | | | | | | - Louis Gerstenfeld
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA, USA
| | - Paola Divieti Pajevic
- Department of Translational Dental Medicine, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, USA.,Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
27
|
Cryptic ligand on collagen matrix unveiled by MMP13 accelerates bone tissue regeneration via MMP13/Integrin α3/RUNX2 feedback loop. Acta Biomater 2021; 125:219-230. [PMID: 33677160 DOI: 10.1016/j.actbio.2021.02.042] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/12/2022]
Abstract
Extracellular matrix (ECM) remodeling is necessary for the development and self-healing of tissue, and the process is tissue specific. Matrix metalloproteinases (MMPs) play a role in ECM remodeling by unwinding and cleaving ECM. We hypothesized that ECM remodeling by MMPs is involved in the differentiation of stem cells into specific lineages during self-healing. To prove the hypothesis, we investigated which MMPs are involved in the osteogenic differentiation of human mesenchymal stem cells (hMSCs) grown on a type I collagen (Col I) matrix, and we found that specifically high expression of MMP13 in hMSCs grown on a Col I matirx during osteogenic differentiation. Moreover, knocking down of MMP13 decreased the osteogenic differentiation of hMSCs grown on a Col I matrix. In addition, pre-treatment of recombinant human MMP13 lead to remodeling of Col I matrix and increased the osteogenic differentiation of hMSCs and in vivo bone formation following the upregulation of the expression of runt-related transcription factor 2 (RUNX2), integrin α3 (ITGA3), and focal adhesion kinase. Furthermore, the transcription factor RUNX2 bound to the MMP13 promoter. These results suggest that growth on a remodeled Col I matrix by MMP13 stimulates osteogenic differentiation of hMSCs and self-healing of bone tissue via an MMP13/ITGA3/RUNX2 positive feedback loop. STATEMENT OF SIGNIFICANCE: Self-healing of tissue could be the key to treating diseases that cannot be overcome by present technology. We investigated the mechanism underlying the self-healing of tissue and we found that the osteogenic differentiation was increased in hMSCs grown on a remodeled Col I matrix by the optimized concentration of MMP13 not in hMSCs grown on a Col I fragments cleaved by a high concentration of MMP13. In addition, we found the remodeled Col I matrix by MMP13 increased the osteogenic capacity through a MMP13/integrin α3/RUNX2 positive feedback loop. This result would be able to not only provide a strategy for bone tissue-specific functional materials following strong evidence about the self-healing mechanism of bone through the interaction between stem cells and the ECM matrix. As such, we strongly believe our finding will be of interest to researchers studying biomaterials, stem cell biology and matrix interaction for regenerative medicine and therapy.
Collapse
|
28
|
Bonatto Paese CL, Brooks EC, Aarnio-Peterson M, Brugmann SA. Ciliopathic micrognathia is caused by aberrant skeletal differentiation and remodeling. Development 2021; 148:148/4/dev194175. [PMID: 33589509 DOI: 10.1242/dev.194175] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 01/13/2021] [Indexed: 12/16/2022]
Abstract
Ciliopathies represent a growing class of diseases caused by defects in microtubule-based organelles called primary cilia. Approximately 30% of ciliopathies are characterized by craniofacial phenotypes such as craniosynostosis, cleft lip/palate and micrognathia. Patients with ciliopathic micrognathia experience a particular set of difficulties, including impaired feeding and breathing, and have extremely limited treatment options. To understand the cellular and molecular basis for ciliopathic micrognathia, we used the talpid2 (ta2 ), a bona fide avian model for the human ciliopathy oral-facial-digital syndrome subtype 14. Histological analyses revealed that the onset of ciliopathic micrognathia in ta2 embryos occurred at the earliest stages of mandibular development. Neural crest-derived skeletal progenitor cells were particularly sensitive to a ciliopathic insult, undergoing unchecked passage through the cell cycle and subsequent increased proliferation. Furthermore, whereas neural crest-derived skeletal differentiation was initiated, osteoblast maturation failed to progress to completion. Additional molecular analyses revealed that an imbalance in the ratio of bone deposition and resorption also contributed to ciliopathic micrognathia in ta2 embryos. Thus, our results suggest that ciliopathic micrognathia is a consequence of multiple aberrant cellular processes necessary for skeletal development, and provide potential avenues for future therapeutic treatments.
Collapse
Affiliation(s)
- Christian Louis Bonatto Paese
- Division of Developmental Biology, Department of Pediatrics Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Division of Plastic Surgery, Department of Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Evan C Brooks
- Division of Developmental Biology, Department of Pediatrics Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Division of Plastic Surgery, Department of Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Megan Aarnio-Peterson
- Division of Developmental Biology, Department of Pediatrics Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Division of Plastic Surgery, Department of Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Samantha A Brugmann
- Division of Developmental Biology, Department of Pediatrics Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA .,Division of Plastic Surgery, Department of Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Shriners Children's Hospital, Cincinnati, OH 45229, USA
| |
Collapse
|
29
|
McClurg O, Tinson R, Troeberg L. Targeting Cartilage Degradation in Osteoarthritis. Pharmaceuticals (Basel) 2021; 14:ph14020126. [PMID: 33562742 PMCID: PMC7916085 DOI: 10.3390/ph14020126] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis is a common, degenerative joint disease with significant socio-economic impact worldwide. There are currently no disease-modifying drugs available to treat the disease, making this an important area of pharmaceutical research. In this review, we assessed approaches being explored to directly inhibit metalloproteinase-mediated cartilage degradation and to counteract cartilage damage by promoting growth factor-driven repair. Metalloproteinase-blocking antibodies are discussed, along with recent clinical trials on FGF18 and Wnt pathway inhibitors. We also considered dendrimer-based approaches being developed to deliver and retain such therapeutics in the joint environment. These may reduce systemic side effects while improving local half-life and concentration. Development of such targeted anabolic therapies would be of great benefit in the osteoarthritis field.
Collapse
|
30
|
Cassuto J, Folestad A, Göthlin J, Malchau H, Kärrholm J. Concerted actions by MMPs, ADAMTS and serine proteases during remodeling of the cartilage callus into bone during osseointegration of hip implants. Bone Rep 2020; 13:100715. [PMID: 32995386 PMCID: PMC7509196 DOI: 10.1016/j.bonr.2020.100715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 09/01/2020] [Accepted: 09/07/2020] [Indexed: 11/09/2022] Open
Abstract
INTRODUCTION Although the number of patients undergoing total hip arthroplasty is constantly on the rise, we only have limited knowledge of the molecular mechanisms necessary for successful osseointegration of implants or the reasons why some fail. Understanding the spatiotemporal characteristics of signaling pathways involved in bone healing of implants is therefore of particular importance for our ability to identify factors causing implants to fail. The current study investigated the role of three families of proteases, i.e. MMPs (matrix metalloproteinases), ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) and serine proteases, as well as their endogenous inhibitors during osseointegration of hip implants that have endured two decades of use without clinical or radiological signs of loosening. MATERIALS AND METHODS Twenty-four patients that had undergone primary THA due to one-sided osteoarthritis (OA) were monitored during 18 years (Y) with repeated measurements of plasma biomarkers, clinical variables and radiographs. All implants were clinically and radiographically well-fixed throughout the follow-up. Eighty-one healthy donors divided in three gender and age-matched groups and twenty OA patients awaiting THA, served as controls. Plasma was analyzed for MMP-1, -2, -3, -8, -9, -10, -13, -14, tissue inhibitor of metalloproteinase (TIMP)-1, -2, -3, ADAMTS4, ADAMTS5, the serine proteases neutrophil elastase (NE), proteinase 3 (PR3) and their endogenous inhibitors, secretory leucocyte proteinase inhibitor (SLPI), trappin-2/elafin and serpina1 (α-1 antitrypsin). Cartilage turnover was monitored using two markers of cartilage synthesis, type II procollagen and PIICP (procollagen II C-terminal propeptide), and two markers of cartilage degradation, CTX-II (C-terminal telopeptide fragments of type II collagen) and split products of aggrecan (G1-IGD-G2). RESULTS MMP-1, MMP-9, ADAMTS4, NE and PR3 were above healthy in presurgery OA patients but returned to the level of healthy within 6 weeks (W) after surgery. MMPs and serine proteases were counter-regulated during this phase by TIMP-1, SLPI and trappin-2/elafin. Type II procollagen, PIICP and CTX-II increased to a peak 6 W after surgery with a gradual return to the level of controls within weeks. Significant increases by MMP-8, MMP-9, ADAMTS4, ADAMTS5, NE, PR3 and the protease inhibitors, TIMP-3 and serpina1, were seen 5 Y after hip arthroplasty paralleled by a sharp increase in the levels of the cartilage degradation markers, CTX-II and G1-IGD-G2. All the above mediators were normalized before 18 Y, except MMP-1 and MMP-9 that remained above healthy at 18 Y. MMP-14 increased immediately after surgery and remained elevated until 5 Y postsurgery before returning to the level of controls at 7 Y. CONCLUSION Notwithstanding temporal differences, the molecular processes of bone repair in arthroplasty patients show great spatial similarities with the classical phases of fracture repair as previously shown in animal models. Cartilagenous callus, produced and remodeled early after hip arthroplasty, is replaced with bone 5 Y to7 Y after surgery by the concerted actions of MMP-8, MMP-9, ADAMTS4, ADAMTS5, NE and PR3, thus suggesting that a complex regulatory cross-talk may exist between different families of proteases during this transitional phase of cartilage degradation. Regulation and fine-tuning of cartilage remodeling by MMPs and ADAMTS is controlled by TIMP-3 whereas serine proteases are regulated by serpina1. Increased MMP-1 and MMP-9 beyond 10Y post-THA support a role during coupled bone remodeling.
Collapse
Affiliation(s)
- Jean Cassuto
- Orthopedic Research Unit, Department of Orthopedic Surgery, Sahlgrenska University Hospital, Mölndal, Sweden
- Institution of Clinical Sciences, Göteborg University, Göteborg, Sweden
| | - Agnetha Folestad
- Department of Orthopedics, CapioLundby Hospital, Göteborg, Sweden
| | - Jan Göthlin
- Department of Radiology, Sahlgrenska University Hospital, Mölndal, Sweden
- Institution of Clinical Sciences, Göteborg University, Göteborg, Sweden
| | - Henrik Malchau
- Orthopedic Research Unit, Department of Orthopedic Surgery, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Orthopedic Surgery, Harvard Medical School, Boston, USA
| | - Johan Kärrholm
- Orthopedic Research Unit, Department of Orthopedic Surgery, Sahlgrenska University Hospital, Mölndal, Sweden
- Institution of Clinical Sciences, Göteborg University, Göteborg, Sweden
| |
Collapse
|
31
|
Hendriks M, Ramasamy SK. Blood Vessels and Vascular Niches in Bone Development and Physiological Remodeling. Front Cell Dev Biol 2020; 8:602278. [PMID: 33330496 PMCID: PMC7729063 DOI: 10.3389/fcell.2020.602278] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/11/2020] [Indexed: 12/12/2022] Open
Abstract
Recent advances in our understanding of blood vessels and vascular niches in bone convey their critical importance in regulating bone development and physiology. The contribution of blood vessels in bone functions and remodeling has recently gained enormous interest because of their therapeutic potential. The mammalian skeletal system performs multiple functions in the body to regulate growth, homeostasis and metabolism. Blood vessels provide support to various cell types in bone and maintain functional niches in the bone marrow microenvironment. Heterogeneity within blood vessels and niches indicate the importance of specialized vascular niches in regulating skeletal functions. In this review, we discuss physiology of bone vasculature and their specialized niches for hematopoietic stem cells and mesenchymal progenitor cells. We provide clinical and experimental information available on blood vessels during physiological bone remodeling.
Collapse
Affiliation(s)
- Michelle Hendriks
- Institute of Clinical Sciences, Imperial College London, London, United Kingdom
- MRC London Institute of Medical Sciences, Imperial College London, London, United Kingdom
| | - Saravana K. Ramasamy
- Institute of Clinical Sciences, Imperial College London, London, United Kingdom
- MRC London Institute of Medical Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
32
|
Stucker S, Chen J, Watt FE, Kusumbe AP. Bone Angiogenesis and Vascular Niche Remodeling in Stress, Aging, and Diseases. Front Cell Dev Biol 2020; 8:602269. [PMID: 33324652 PMCID: PMC7726257 DOI: 10.3389/fcell.2020.602269] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/05/2020] [Indexed: 02/05/2023] Open
Abstract
The bone marrow (BM) vascular niche microenvironments harbor stem and progenitor cells of various lineages. Bone angiogenesis is distinct and involves tissue-specific signals. The nurturing vascular niches in the BM are complex and heterogenous consisting of distinct vascular and perivascular cell types that provide crucial signals for the maintenance of stem and progenitor cells. Growing evidence suggests that the BM niche is highly sensitive to stress. Aging, inflammation and other stress factors induce changes in BM niche cells and their crosstalk with tissue cells leading to perturbed hematopoiesis, bone angiogenesis and bone formation. Defining vascular niche remodeling under stress conditions will improve our understanding of the BM vascular niche and its role in homeostasis and disease. Therefore, this review provides an overview of the current understanding of the BM vascular niches for hematopoietic stem cells and their malfunction during aging, bone loss diseases, arthritis and metastasis.
Collapse
Affiliation(s)
- Sina Stucker
- Tissue and Tumor Microenvironments Group, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Junyu Chen
- Tissue and Tumor Microenvironments Group, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fiona E. Watt
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Anjali P. Kusumbe
- Tissue and Tumor Microenvironments Group, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
33
|
Huang S, Jin M, Su N, Chen L. New insights on the reparative cells in bone regeneration and repair. Biol Rev Camb Philos Soc 2020; 96:357-375. [PMID: 33051970 DOI: 10.1111/brv.12659] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022]
Abstract
Bone possesses a remarkable repair capacity to regenerate completely without scar tissue formation. This unique characteristic, expressed during bone development, maintenance and injury (fracture) healing, is performed by the reparative cells including skeletal stem cells (SSCs) and their descendants. However, the identity and functional roles of SSCs remain controversial due to technological difficulties and the heterogeneity and plasticity of SSCs. Moreover, for many years, there has been a biased view that bone marrow is the main cell source for bone repair. Together, these limitations have greatly hampered our understanding of these important cell populations and their potential applications in the treatment of fractures and skeletal diseases. Here, we reanalyse and summarize current understanding of the reparative cells in bone regeneration and repair and outline recent progress in this area, with a particular emphasis on the temporal and spatial process of fracture healing, the sources of reparative cells, an updated definition of SSCs, and markers of skeletal stem/progenitor cells contributing to the repair of craniofacial and long bones, as well as the debate between SSCs and pericytes. Finally, we also discuss the existing problems, emerging novel technologies and future research directions in this field.
Collapse
Affiliation(s)
- Shuo Huang
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| | - Min Jin
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| | - Nan Su
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| | - Lin Chen
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| |
Collapse
|
34
|
Wang H, Ni Z, Yang J, Li M, Liu L, Pan X, Xu L, Wang X, Fang S. IL-1β promotes osteogenic differentiation of mouse bone marrow mesenchymal stem cells via the BMP/Smad pathway within a certain concentration range. Exp Ther Med 2020; 20:3001-3008. [PMID: 32855666 PMCID: PMC7444350 DOI: 10.3892/etm.2020.9065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 03/24/2020] [Indexed: 12/21/2022] Open
Abstract
Inflammatory factors play an important role in the process of fracture healing. The influence of interleukin (IL)-1β, a key inflammatory factory, on new bone formation has been controversial. The aim of the present study was to investigate whether IL-1β affects the osteogenic differentiation of mouse bone marrow mesenchymal stem cells (MBMMSCs), and examined its effective concentration range and molecular mechanism of action. MBMMSC proliferation in the presence of IL-1β was observed using a Cell-Counting Kit-8 assay, and the effect of IL-1β on MBMMSC apoptosis was examined via flow cytometry. Alkaline phosphatase assay, Alizarin Red staining and quantitative assays were performed to evaluate the osteogenic differentiation of MBMMSCs. The expression levels of osteogenic differentiation markers were detected using reverse transcription-quantitative PCR (RT-qPCR). It was demonstrated that within a concentration range of 0.01-1 ng/ml, IL-1β promoted osteogenic differentiation of MBMMSCs and did not induce apoptosis. Furthermore, RT-qPCR results indicated that IL-1β increased osteogenic gene expression within this concentration range. Moreover, Western blotting results identified that the bone morphogenetic protein/Smad (BMP/Smad) signaling pathway was significantly activated by IL-1β under osteogenic conditions. Therefore, the present results suggested that within a certain concentration range, IL-1β promoted osteogenic differentiation and function of MBMMSCs via the BMP/Smad signaling pathway.
Collapse
Affiliation(s)
- Hao Wang
- Department of Orthopedics, Huainan First People's Hospital, Anhui University of Science and Technology, Huainan, Anhui 232000, P.R. China.,Department of Orthopedics, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230000, P.R. China
| | - Zhihao Ni
- Department of Orthopedics, Hefei First People's Hospital, Anhui Medical University, Hefei, Anhui 230000, P.R. China
| | - Jiazhao Yang
- Department of Orthopedics, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230000, P.R. China
| | - Meng Li
- Department of Orthopedics, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230000, P.R. China
| | - Lei Liu
- Department of Orthopedics, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230000, P.R. China
| | - Xuejie Pan
- Department of Orthopedics, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230000, P.R. China
| | - Lei Xu
- Department of Orthopedics, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230000, P.R. China
| | - Xujin Wang
- Department of Orthopedics, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230000, P.R. China
| | - Shiyuan Fang
- Department of Orthopedics, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230000, P.R. China
| |
Collapse
|
35
|
Dai J, Dong R, Han X, Li J, Gong X, Bai Y, Kang F, Liang M, Zeng F, Hou Z, Dong S. Osteoclast-derived exosomal let-7a-5p targets Smad2 to promote the hypertrophic differentiation of chondrocytes. Am J Physiol Cell Physiol 2020; 319:C21-C33. [PMID: 32374679 DOI: 10.1152/ajpcell.00039.2020] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The invasion of osteoclasts into the cartilage via blood vessels advances the process of endochondral ossification, and dysregulation of dynamic intercellular interactions results in skeletal dysplasias. Although the regulation of osteoclasts by growth plate chondrocytes has been reported in detail, the effect of osteoclasts on chondrocytes remains to be determined. In this study, ATDC5 cells and bone marrow mesenchymal stem cells were differentiated into chondrocytes and treated with conditioned medium obtained from bone marrow macrophages differentiated to osteoclast precursors and osteoclasts. Exosomes were inhibited in conditioned medium or isolated directly from osteoclasts to further determine whether osteoclast-derived exosomes play an important role in chondrocyte hypertrophy. Additionally, exosomal miRNAs were detected, and let-7a-5p was selected as an miRNA with significantly increased expression in osteoclast-derived exosomes. Experiments were performed to verify the potential target Smad2 and investigate how let-7a-5p affected chondrocytes. The results suggest that both osteoclast precursors and osteoclasts promote chondrocyte hypertrophy and that the promotive effect of osteoclasts is more significant than that of osteoclast precursors. Osteoclast-derived exosomes promote the hypertrophic differentiation of chondrocytes. Moreover, osteoclast-derived exosomal let-7a-5p inhibits Smad2 to decrease the transforming growth factor-β-induced inhibition of chondrocyte hypertrophy. Our research reveals the role of osteoclasts in the regulation of chondrocytes and provides insights into the highly coordinated intercellular process of endochondral ossification.
Collapse
Affiliation(s)
- Jingjin Dai
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China
| | - Rui Dong
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xinyun Han
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jianmei Li
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiaoshan Gong
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yun Bai
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fei Kang
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China
| | - Mengmeng Liang
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fanchun Zeng
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhiyong Hou
- Department of Orthopedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
36
|
Morse A, McDonald MM, Mikulec K, Schindeler A, Munns CF, Little DG. Pretreatment with Pamidronate Decreases Bone Formation but Increases Callus Bone Volume in a Rat Closed Fracture Model. Calcif Tissue Int 2020; 106:172-179. [PMID: 31578632 DOI: 10.1007/s00223-019-00615-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 08/05/2019] [Indexed: 12/18/2022]
Abstract
Clinical concerns have been raised over prior exposure to bisphosphonates impairing fracture healing. To model this, groups of male Wistar rats were assigned to saline control or treatment groups receiving 0.15 mg/kg (low dose), 0.5 mg/kg (medium dose), and 5 mg/kg (high dose) Pamidronate (PAM) twice weekly for 4 weeks. At this point, closed fractures were made using an Einhorn apparatus, and bisphosphonate dosing was continued until the experimental endpoint. Specimens were analyzed at 2 and 6 weeks (N = 8 per group per time point). Twice weekly PAM dosing was found to have no effect on early soft callus remodeling at 2 weeks post fracture. At this time point, the highest dose PAM group gave significant increases in bone volume (+ 10%, p < 0.05), bone mineral content (+ 30%, p < 0.01), and bone mineral density (+ 10%, p < 0.01). This PAM dosing regimen showed more substantive effects on hard callus at 6 weeks post fracture, with PAM treatment groups showing + 46-79% increased bone volume. Dynamic bone labeling showed reduced calcein signal in the PAM-treated calluses (38-63%, p < 0.01) and reduced MAR (32-49%, p < 0.01), suggesting a compensatory reduction in bone anabolism. These data support the concept that bisphosphonates lead to profound decreases in bone turnover in fracture repair, however, this does not affect soft callus remodeling.
Collapse
Affiliation(s)
- Alyson Morse
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW, 2145, Australia
- Discipline of Child and Adolescent Health, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Michelle M McDonald
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW, 2145, Australia
- Bone Biology Division, The Garvan Institute for Medical Research, Sydney, Australia
| | - Kathy Mikulec
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW, 2145, Australia
| | - Aaron Schindeler
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW, 2145, Australia
- Discipline of Child and Adolescent Health, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Craig F Munns
- Discipline of Child and Adolescent Health, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Department for Endocrinology and Diabetes, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - David G Little
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW, 2145, Australia.
- Discipline of Child and Adolescent Health, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
37
|
Gu XD, Wei L, Li PC, Che XD, Zhao RP, Han PF, Lu JG, Wei XC. Adenovirus-mediated transduction with Histone Deacetylase 4 ameliorates disease progression in an osteoarthritis rat model. Int Immunopharmacol 2019; 75:105752. [PMID: 31310910 DOI: 10.1016/j.intimp.2019.105752] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/21/2019] [Accepted: 07/10/2019] [Indexed: 01/15/2023]
Abstract
BACKGROUND Downregulation of histone deacetylase-4 (HDAC4) contributes to cartilage degeneration in osteoarthritis (OA) because it promotes upregulation of runt-related transcription factor-2 (Runx-2) and osteoarthritis-related genes. The effect of HDAC4 upregulation on cartilage damage in OA remains unknown. METHODS Rat chondrocytes were infected with Ad-GFP or Ad-HDAC4-GFP for 48 h, stimulated with interleukin-1β (IL-1β, 10 ng/mL) for 24 h, and then harvested for RT-qPCR. Male Sprague-Dawley rats in 3 groups were given anterior cruciate ligament transection (ACLT) or sham operation, and knee injections with different adenovirus (Ad) vectors at 48 h after surgery and every 3 weeks thereafter: ACLT+Ad-GFP (n = 17); ACLT+Ad-HDAC4-GFP (n = 20); and sham+Ad-GFP (n = 15). Three ACLT-Ad-HDAC4-GFP rats were sacrificed at different times to examine the expression of HDAC4. Two ACLT-Ad-GFP rats and two ACLT-Ad-HDAC4-GFP rats were euthanized at week-2; articular cartilage was harvested and expression of HDAC4 was determined by RT-qPCR. All other rats were euthanized at week-8. Cartilage damage and OA progression was assessed using radiography, fluorescence molecular tomography (FMT), histology, immunohistochemistry (IHC), ELISA, and RT-qPCR. RESULTS Overexpression of HDAC4 in chondrocytes stimulated by IL-1β reduced the levels of Runx-2, MMP-13, and Collagen X, but increased the levels of Collagen II and Aggrecan. Upregulation of HDAC4 reduced osteophyte formation and cartilage damage, and increased articular cartilage anabolism. CONCLUSION HDAC4 attenuated articular cartilage damage by repression of Runx-2, MMP-13, and collagen X and induction of collagen II and ACAN in this rat model of OA. Upregulation of HDAC4 may provide chondroprotection in OA patients.
Collapse
Affiliation(s)
- Xiao-Dong Gu
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, PR China
| | - Lei Wei
- Department of Orthopedics, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Peng-Cui Li
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, PR China
| | - Xian-da Che
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, PR China
| | - Rui-Peng Zhao
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, PR China
| | - Peng-Fei Han
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, PR China
| | - Jian-Gong Lu
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, PR China
| | - Xiao-Chun Wei
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, PR China.
| |
Collapse
|
38
|
Saiganesh S, Saathvika R, Arumugam B, Vishal M, Udhaya V, Ilangovan R, Selvamurugan N. TGF-β1-stimulation of matrix metalloproteinase-13 expression by down-regulation of miR-203a-5p in rat osteoblasts. Int J Biol Macromol 2019; 132:541-549. [PMID: 30951775 DOI: 10.1016/j.ijbiomac.2019.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/01/2019] [Accepted: 04/01/2019] [Indexed: 02/06/2023]
Abstract
Transforming growth factor-beta1 (TGF-β1) is a pleiotropic and ubiquitous cytokine involved in bone development and bone remodeling. Matrix metalloproteinase-13 (MMP13) plays a role in the degradation of the extracellular matrix (ECM), and the regulation of this gene is critical in bone remodeling. We previously reported that TGF-β1 stimulates MMP13 expression in rat osteoblasts. Recently, studies have examined the regulation of bone metabolism by microRNAs (miRNAs) to determine their therapeutic potential in osteogenesis. Here, we assessed the effect of TGF-β1 on down-regulation of miRNAs that target MMP13 and stimulation of MMP13 expression in osteoblasts. We used in silico analysis and identified 11 specific miRNAs which directly target rat MMP13. Among these miRNAs, miR-203a-5p expression was significantly decreased by TGF-β1-treatment in rat osteoblasts. Transient transfection of a miR-203a-5p mimic into rat osteoblasts reduced MMP13 expression. A luciferase reporter assay confirmed a direct targeting of miR-miR-203a-5p with the 3' untranslated regions of the MMP13 gene. Hence, we suggest that TGF-β1 stimulated down-regulation of miR-203a-5p, resulting in the stimulation of MMP13 expression in rat osteoblasts. Thus, identification of the role of miR-203a-5p via TGF-β1 and MMP13 in bone remodeling indicated its potential as a biomarker or therapeutic agent for treating bone and bone-related diseases.
Collapse
Affiliation(s)
- S Saiganesh
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - R Saathvika
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - B Arumugam
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - M Vishal
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - V Udhaya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - R Ilangovan
- Department of Endocrinology, Dr. A.L.M. PG Institute of Basic Medical Sciences, University of Madras, Taramani, Chennai 600 113, Tamil Nadu, India
| | - N Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India.
| |
Collapse
|
39
|
Singhvi MS, Zinjarde SS, Gokhale DV. Polylactic acid: synthesis and biomedical applications. J Appl Microbiol 2019; 127:1612-1626. [PMID: 31021482 DOI: 10.1111/jam.14290] [Citation(s) in RCA: 354] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 03/29/2019] [Accepted: 04/16/2019] [Indexed: 12/13/2022]
Abstract
Social and economic development has driven considerable scientific and engineering efforts on the discovery, development and utilization of polymers. Polylactic acid (PLA) is one of the most promising biopolymers as it can be produced from nontoxic renewable feedstock. PLA has emerged as an important polymeric material for biomedical applications on account of its properties such as biocompatibility, biodegradability, mechanical strength and process ability. Lactic acid (LA) can be obtained by fermentation of sugars derived from renewable resources such as corn and sugarcane. PLA is thus an eco-friendly nontoxic polymer with features that permit use in the human body. Although PLA has a wide spectrum of applications, there are certain limitations such as slow degradation rate, hydrophobicity and low impact toughness associated with its use. Blending PLA with other polymers offers convenient options to improve associated properties or to generate novel PLA polymers/blends for target applications. A variety of PLA blends have been explored for various biomedical applications such as drug delivery, implants, sutures and tissue engineering. PLA and their copolymers are becoming widely used in tissue engineering for function restoration of impaired tissues due to their excellent biocompatibility and mechanical properties. The relationship between PLA material properties, manufacturing processes and development of products with desirable characteristics is described in this article. LA production, PLA synthesis and their applications in the biomedical field are also discussed.
Collapse
Affiliation(s)
- M S Singhvi
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| | - S S Zinjarde
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| | - D V Gokhale
- CSIR-National Chemical Laboratory, NCIM Resource Centre, Pune, India
| |
Collapse
|
40
|
Mikael PE, Golebiowska AA, Xin X, Rowe DW, Nukavarapu SP. Evaluation of an Engineered Hybrid Matrix for Bone Regeneration via Endochondral Ossification. Ann Biomed Eng 2019; 48:992-1005. [PMID: 31037444 DOI: 10.1007/s10439-019-02279-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 04/24/2019] [Indexed: 12/28/2022]
Abstract
Despite its regenerative ability, long and segmental bone defect repair remains a significant orthopedic challenge. Conventional tissue engineering efforts induce bone formation through intramembranous ossification (IO) which limits vascular formation and leads to poor bone regeneration. To overcome this challenge, a novel hybrid matrix comprised of a load-bearing polymer template and a gel phase is designed and assessed for bone regeneration. Our previous studies developed a synthetic ECM, hyaluronan (HA)-fibrin (FB), that is able to mimic cartilage-mediated bone formation in vitro. In this study, the well-characterized HA-FB hydrogel is combined with a biodegradable polymer template to form a hybrid matrix. In vitro evaluation of the matrix showed cartilage template formation, cell recruitment and recruited cell osteogenesis, essential stages in endochondral ossification. A transgenic reporter-mouse critical-defect model was used to evaluate the bone healing potential of the hybrid matrix in vivo. The results demonstrated host cell recruitment into the hybrid matrix that led to new bone formation and subsequent remodeling of the mineralization. Overall, the study developed and evaluated a novel load-bearing graft system for bone regeneration via endochondral ossification.
Collapse
Affiliation(s)
- Paiyz E Mikael
- Department of Materials Science, & Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Aleksandra A Golebiowska
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, CT, 06269, USA
| | - Xiaonan Xin
- Center for Regenerative Medicine and Skeletal Development, University of Connecticut Health, Farmington, CT, 06032, USA
| | - David W Rowe
- Center for Regenerative Medicine and Skeletal Development, University of Connecticut Health, Farmington, CT, 06032, USA
| | - Syam P Nukavarapu
- Department of Materials Science, & Engineering, University of Connecticut, Storrs, CT, 06269, USA. .,Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, CT, 06269, USA. .,Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA.
| |
Collapse
|
41
|
Dong Y, Yang L, Luo W, Zhu T, Yan W, Kong J, Yuan Z, Zhao Q. Mannose receptor C type 2 mediates 1,25(OH) 2D 3/vitamin D receptor-regulated collagen metabolism through collagen type 5, alpha 2 chain and matrix metalloproteinase 13 in murine MC3T3-E1 cells. Mol Cell Endocrinol 2019; 483:74-86. [PMID: 30641101 DOI: 10.1016/j.mce.2019.01.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/27/2018] [Accepted: 01/09/2019] [Indexed: 11/24/2022]
Abstract
Vitamin D plays an important role in maintaining skeletal development and bone homeostasis. Although vitamin D has been extensively researched, the direct effect of 1,25(OH)2D3 on osteoblasts is unclear. To explore the 1,25(OH)2D3 action on murine osteoblasts, we performed tandem mass tag experiments on MC3T3-E1 cells treated with and without 1,25(OH)2D3. Three up-regulated proteins (MRC2, WWTR1 and RASSF2) related to bone metabolism were confirmed in this study. 1,25(OH)2D3 up-regulated the expression of MRC2 through vitamin D receptor. MRC2 affects collagen metabolism in osteoblasts. Combined with bioinformatics and parallel reaction monitoring analysis, we inhibited the expression of MRC2 to explore the relationship between MRC2 and collagens. Then we found MRC2 down-regulated COL5A2 and up-regulated MMP13. This study provides a protein profile of 1,25(OH)2D3-treated murine osteoblasts, reveals a newly discovered signaling axis (1,25(OH)2D3/VDR/MRC2/COL5A2 and MMP13), and explains the effect of 1,25(OH)2D3 on bone metabolism from a new perspective.
Collapse
Affiliation(s)
- Yaping Dong
- Department of Pediatric Orthopedic, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China; Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Benxi, Liaoning, 117004, China
| | - Liping Yang
- Department of Pediatric Orthopedic, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China; Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Benxi, Liaoning, 117004, China
| | - Wenting Luo
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Benxi, Liaoning, 117004, China
| | - Tong Zhu
- Department of Pediatric Orthopedic, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Wei Yan
- Department of Pediatric Orthopedic, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Juan Kong
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Benxi, Liaoning, 117004, China; Nutrition Department, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Zhengwei Yuan
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Benxi, Liaoning, 117004, China
| | - Qun Zhao
- Department of Pediatric Orthopedic, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China; Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Benxi, Liaoning, 117004, China.
| |
Collapse
|
42
|
Bahney CS, Zondervan RL, Allison P, Theologis A, Ashley JW, Ahn J, Miclau T, Marcucio RS, Hankenson KD. Cellular biology of fracture healing. J Orthop Res 2019; 37:35-50. [PMID: 30370699 PMCID: PMC6542569 DOI: 10.1002/jor.24170] [Citation(s) in RCA: 339] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/27/2018] [Indexed: 02/04/2023]
Abstract
The biology of bone healing is a rapidly developing science. Advances in transgenic and gene-targeted mice have enabled tissue and cell-specific investigations of skeletal regeneration. As an example, only recently has it been recognized that chondrocytes convert to osteoblasts during healing bone, and only several years prior, seminal publications reported definitively that the primary tissues contributing bone forming cells during regeneration were the periosteum and endosteum. While genetically modified animals offer incredible insights into the temporal and spatial importance of various gene products, the complexity and rapidity of healing-coupled with the heterogeneity of animal models-renders studies of regenerative biology challenging. Herein, cells that play a key role in bone healing will be reviewed and extracellular mediators regulating their behavior discussed. We will focus on recent studies that explore novel roles of inflammation in bone healing, and the origins and fates of various cells in the fracture environment. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
- Chelsea S. Bahney
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California
| | - Robert L. Zondervan
- Department of Physiology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan
| | - Patrick Allison
- Department of Physiology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan
| | - Alekos Theologis
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California
| | - Jason W. Ashley
- Department of Biology, Eastern Washington University, Cheney, Washington
| | - Jaimo Ahn
- Department of Biology, Eastern Washington University, Cheney, Washington
| | - Theodore Miclau
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California
| | - Ralph S. Marcucio
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California
| | - Kurt D. Hankenson
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
43
|
Duchamp de Lageneste O, Colnot C. Periostin in Bone Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1132:49-61. [PMID: 31037624 DOI: 10.1007/978-981-13-6657-4_6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Bone regeneration is an efficient regenerative process depending on the recruitment and activation of skeletal stem cells that allow cartilage and bone formation leading to fracture consolidation. Periosteum, the tissue located at the outer surface of bone is now recognized as an essential player in the bone repair process and contains skeletal stem cells with high regenerative potential. The matrix composition of the periosteum defines its roles in bone growth, in cortical bone modeling and remodeling in response to mechanical strain, and in bone repair. Periostin is a key extracellular matrix component of the periosteum involved in periosteum functions. In this chapter, we summarize the current knowledge on the bone regeneration process, the role of the periosteum and skeletal stem cells, and Periostin functions in this context. The matricellular protein Periostin has several roles through all stages of bone repair: in the early days of repair during the initial activation of stem cells within periosteum, in the active phase of cartilage and bone deposition in the facture callus, and in the final phase of bone bridging and reconstitution of the stem cell pool within periosteum.
Collapse
Affiliation(s)
| | - Céline Colnot
- INSERM UMR1163, Imagine Institute, Paris Descartes University, Paris, France.
| |
Collapse
|
44
|
Su YW, Chim SM, Zhou L, Hassanshahi M, Chung R, Fan C, Song Y, Foster BK, Prestidge CA, Peymanfar Y, Tang Q, Butler LM, Gronthos S, Chen D, Xie Y, Chen L, Zhou XF, Xu J, Xian CJ. Osteoblast derived-neurotrophin‑3 induces cartilage removal proteases and osteoclast-mediated function at injured growth plate in rats. Bone 2018; 116:232-247. [PMID: 30125729 PMCID: PMC6550307 DOI: 10.1016/j.bone.2018.08.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 07/25/2018] [Accepted: 08/14/2018] [Indexed: 01/08/2023]
Abstract
Faulty bony repair causes dysrepair of injured growth plate cartilage and bone growth defects in children; however, the underlying mechanisms are unclear. Recently, we observed the prominent induction of neurotrophin‑3 (NT-3) and its important roles as an osteogenic and angiogenic factor promoting the bony repair. The current study investigated its roles in regulating injury site remodelling. In a rat tibial growth plate drill-hole injury repair model, NT-3 was expressed prominently in osteoblasts at the injury site. Recombinant NT-3 (rhNT-3) systemic treatment enhanced, but NT-3 immunoneutralization attenuated, expression of cartilage-removal proteases (MMP-9 and MMP-13), presence of bone-resorbing osteoclasts and expression of osteoclast protease cathepsin K, and remodelling at the injury site. NT-3 was also highly induced in cultured mineralizing rat bone marrow stromal cells, and the conditioned medium augmented osteoclast formation and resorptive activity, an ability that was blocked by presence of anti-NT-3 antibody. Moreover, NT-3 and receptor TrkC were induced during osteoclastogenesis, and rhNT-3 treatment activated TrkC downstream kinase Erk1/2 in differentiating osteoclasts although rhNT-3 alone did not affect activation of osteoclastogenic transcription factors NF-κB or NFAT in RAW264.7 osteoclast precursor cells. Furthermore, rhNT-3 treatment increased, but NT-3 neutralization reduced, expression of osteoclastogenic cytokines (RANKL, TNF-α, and IL-1) in mineralizing osteoblasts and in growth plate injury site, and rhNT-3 augmented the induction of these cytokines caused by RANKL treatment in RAW264.7 cells. Thus, injury site osteoblast-derived NT-3 is important in promoting growth plate injury site remodelling, as it induces cartilage proteases for cartilage removal and augments osteoclastogenesis and resorption both directly (involving activing Erk1/2 and substantiating RANKL-induced increased expression of osteoclastogenic signals in differentiating osteoclasts) and indirectly (inducing osteoclastogenic signals in osteoblasts).
Collapse
Affiliation(s)
- Yu-Wen Su
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5001, Australia.
| | - Shek Man Chim
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, WA 6009, Australia.
| | - Lin Zhou
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, WA 6009, Australia.
| | - Mohammadhossein Hassanshahi
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5001, Australia.
| | - Rosa Chung
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5001, Australia.
| | - Chiaming Fan
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5001, Australia
| | - Yunmei Song
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5001, Australia.
| | - Bruce K Foster
- Department of Orthopaedic Surgery, Women's and Children's Hospital, North Adelaide, SA 5006, Australia.
| | - Clive A Prestidge
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5001, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of South Australia, Mawson Lakes Campus, Mawson Lakes 5095, Australia.
| | - Yaser Peymanfar
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5001, Australia.
| | - Qian Tang
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5001, Australia.
| | - Lisa M Butler
- University of Adelaide Schools of Medicine and Medical Sciences, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
| | - Stan Gronthos
- University of Adelaide Schools of Medicine and Medical Sciences, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
| | - Di Chen
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Yangli Xie
- State Key Laboratory of Trauma, Burns and Combined Injury, Center of Bone Metabolism and Repair, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Lin Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Center of Bone Metabolism and Repair, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5001, Australia.
| | - Jiake Xu
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, WA 6009, Australia.
| | - Cory J Xian
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5001, Australia.
| |
Collapse
|
45
|
Stegen S, Carmeliet G. The skeletal vascular system - Breathing life into bone tissue. Bone 2018; 115:50-58. [PMID: 28844835 DOI: 10.1016/j.bone.2017.08.022] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 08/23/2017] [Indexed: 11/30/2022]
Abstract
During bone development, homeostasis and repair, a dense vascular system provides oxygen and nutrients to highly anabolic skeletal cells. Characteristic for the vascular system in bone is the serial organization of two capillary systems, each typified by specific morphological and physiological features. Especially the arterial capillaries mediate the growth of the bone vascular system, serve as a niche for skeletal and hematopoietic progenitors and couple angiogenesis to osteogenesis. Endothelial cells and osteoprogenitor cells interact not only physically, but also communicate to each other by secretion of growth factors. A vital angiogenic growth factor is vascular endothelial growth factor and its expression in skeletal cells is controlled by osteogenic transcription factors and hypoxia signaling, whereas the secretion of angiocrine factors by endothelial cells is regulated by Notch signaling, blood flow and possibly hypoxia. Bone loss and impaired fracture repair are often associated with reduced and disorganized blood vessel network and therapeutic targeting of the angiogenic response may contribute to enhanced bone regeneration.
Collapse
Affiliation(s)
- Steve Stegen
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
46
|
Schneider RA. Neural crest and the origin of species-specific pattern. Genesis 2018; 56:e23219. [PMID: 30134069 PMCID: PMC6108449 DOI: 10.1002/dvg.23219] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 12/20/2022]
Abstract
For well over half of the 150 years since the discovery of the neural crest, the special ability of these cells to function as a source of species-specific pattern has been clearly recognized. Initially, this observation arose in association with chimeric transplant experiments among differentially pigmented amphibians, where the neural crest origin for melanocytes had been duly noted. Shortly thereafter, the role of cranial neural crest cells in transmitting species-specific information on size and shape to the pharyngeal arch skeleton as well as in regulating the timing of its differentiation became readily apparent. Since then, what has emerged is a deeper understanding of how the neural crest accomplishes such a presumably difficult mission, and this includes a more complete picture of the molecular and cellular programs whereby neural crest shapes the face of each species. This review covers studies on a broad range of vertebrates and describes neural-crest-mediated mechanisms that endow the craniofacial complex with species-specific pattern. A major focus is on experiments in quail and duck embryos that reveal a hierarchy of cell-autonomous and non-autonomous signaling interactions through which neural crest generates species-specific pattern in the craniofacial integument, skeleton, and musculature. By controlling size and shape throughout the development of these systems, the neural crest underlies the structural and functional integration of the craniofacial complex during evolution.
Collapse
Affiliation(s)
- Richard A. Schneider
- Department of Orthopedic SurgeryUniversity of California at San Francisco, 513 Parnassus AvenueS‐1161San Francisco, California
| |
Collapse
|
47
|
Matrix metalloproteinase-13: A special focus on its regulation by signaling cascades and microRNAs in bone. Int J Biol Macromol 2018; 109:338-349. [DOI: 10.1016/j.ijbiomac.2017.12.091] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 12/15/2017] [Accepted: 12/17/2017] [Indexed: 01/03/2023]
|
48
|
Javaheri B, Caetano-Silva SP, Kanakis I, Bou-Gharios G, Pitsillides AA. The Chondro-Osseous Continuum: Is It Possible to Unlock the Potential Assigned Within? Front Bioeng Biotechnol 2018; 6:28. [PMID: 29619368 PMCID: PMC5871702 DOI: 10.3389/fbioe.2018.00028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 03/08/2018] [Indexed: 01/08/2023] Open
Abstract
Endochondral ossification (EO), by which long bones of the axial skeleton form, is a tightly regulated process involving chondrocyte maturation with successive stages of proliferation, maturation, and hypertrophy, accompanied by cartilage matrix synthesis, calcification, and angiogenesis, followed by osteoblast-mediated ossification. This developmental sequence reappears during fracture repair and in osteoarthritic etiopathology. These similarities suggest that EO, and the cells involved, are of great clinical importance for bone regeneration as it could provide novel targeted approaches to increase specific signaling to promote fracture healing, and if regulated appropriately in the treatment of osteoarthritis. The long-held accepted dogma states that hypertrophic chondrocytes are terminally differentiated and will eventually undergo apoptosis. In this mini review, we will explore recent evidence from experiments that revisit the idea that hypertrophic chondrocytes have pluripotent capacity and may instead transdifferentiate into a specific sub-population of osteoblast cells. There are multiple lines of evidence, including our own, showing that local, selective alterations in cartilage extracellular matrix (ECM) remodeling also indelibly alter bone quality. This would be consistent with the hypothesis that osteoblast behavior in long bones is regulated by a combination of their lineage origins and the epigenetic effects of chondrocyte-derived ECM which they encounter during their recruitment. Further exploration of these processes could help to unlock potential novel targets for bone repair and regeneration and in the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Behzad Javaheri
- Skeletal Biology Group, Comparative Biomedical Sciences, The Royal Veterinary College, London, United Kingdom
| | - Soraia P Caetano-Silva
- Skeletal Biology Group, Comparative Biomedical Sciences, The Royal Veterinary College, London, United Kingdom
| | - Ioannis Kanakis
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - George Bou-Gharios
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Andrew A Pitsillides
- Skeletal Biology Group, Comparative Biomedical Sciences, The Royal Veterinary College, London, United Kingdom
| |
Collapse
|
49
|
Santos Kotake BG, Gonzaga MG, Coutinho-Netto J, Ervolino E, de Figueiredo FAT, Issa JPM. Bone repair of critical-sized defects in Wistar rats treated with autogenic, allogenic or xenogenic bone grafts alone or in combination with natural latex fraction F1. Biomed Mater 2018; 13:025022. [DOI: 10.1088/1748-605x/aa9504] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
50
|
Giannoni P, Muraglia A, Giordano C, Narcisi R, Cancedda R, Quarto R, Chiesa R. Osteogenic Differentiation of Human Mesenchymal Stromal Cells on Surface-Modified Titanium Alloys for Orthopedic and Dental Implants. Int J Artif Organs 2018; 32:811-20. [DOI: 10.1177/039139880903201107] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Purpose Surface properties of titanium alloys, used for orthopedic and dental applications, are known to affect implant interactions with host tissues. Osteointegration, bone growth and remodeling in the area surrounding the implants can be implemented by specific biomimetic treatments; these allow the preparation of micro/nanostructured titanium surfaces with a thickened oxide layer, doped with calcium and phosphorus ions. We have challenged these experimental titanium alloys with primary human bone marrow stromal cells to compare the osteogenic differentiation outcomes of the cells once they are seeded onto the modified surfaces, thus simulating a prosthetic device-biological interface of clinical relevance. Methods A specific anodic spark discharge was the biomimetic treatment of choice, providing experimental titanium disks treated with different alkali etching approaches. The disks, checked by electron microscopy and spectroscopy, were subsequently used as substrates for the proliferation and osteogenic differentiation of human cells. Expression of markers of the osteogenic lineage was assessed by means of qualitative and quantitative PCR, by cytochemistry, immunohistochemistry Western blot and matrix metalloprotease activity analyses. Results Metal surfaces were initially less permissive for cell growth. Untreated control substrates were less efficient in sustaining mineralized matrix deposition upon osteogenic induction of the cells. Interestingly, bone sialo protein and matrix metalloprotease 2 levels were enhanced on experimental metals compared to control surfaces, particularly for titanium oxide coatings etched with KOH. Discussion As a whole, the KOH-modification of titanium surfaces seems to allow the best osteogenic differentiation of human mesenchymal stromal cells, representing a possible plus for future clinical prosthetic applications.
Collapse
Affiliation(s)
- Paolo Giannoni
- Stem Cell Laboratory, Advanced Biotechnology Center, Genoa - Italy
| | | | - Carmen Giordano
- Giulio Natta Department of Industrial Chemistry and Chemical Engineering, Milan Polytechnic University, Milan - Italy
| | - Roberto Narcisi
- Stem Cell Laboratory, Advanced Biotechnology Center, Genoa - Italy
| | - Ranieri Cancedda
- Laboratory of Regenerative Medicine, National Cancer Research Institute, University of Genoa - Italy
| | - Rodolfo Quarto
- Stem Cell Laboratory, Advanced Biotechnology Center, Genoa - Italy
| | - Roberto Chiesa
- Giulio Natta Department of Industrial Chemistry and Chemical Engineering, Milan Polytechnic University, Milan - Italy
| |
Collapse
|