1
|
Foster BL. The role of bone sialoprotein in bone healing. J Struct Biol 2024; 216:108132. [PMID: 39369971 DOI: 10.1016/j.jsb.2024.108132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Bone sialoprotein (BSP) is a multi-functional extracellular matrix (ECM) protein associated with mineralized tissues, particularly bone and cementum. The amino acid sequence of BSP includes three evolutionarily conserved sequences which contribute to functions of the protein: an N-terminal collagen-binding domain, polyglutamic acid (polyE) sequences involved in hydroxyapatite nucleation and crystal growth, and a C-terminal arginine-glycine-aspartic acid (RGD) integrin-binding domain. BSP promotes attachment and differentiation of osteogenic and osteoclastic cells. Genetic ablation of BSP in mice results in skeletal and dental developmental defects and impaired bone healing in both appendicular bone and alveolar bone of the jaw. Several studies demonstrated positive effects of BSP on bone healing in rodent models, though other experiments show negligible results. Native (harvested from rat bones) BSP cross-linked to collagen induced slight improvements in calvarial bone healing in rats. Recombinant BSP and collagen delivered in a polylactide (PLA) cylinder improved bone defect healing in rat femurs. Both native and recombinant BSP delivered in a collagen gel improved alveolar bone healing in wild-type and BSP-deficient mice. These advances suggest BSP is a new player in bone healing that has potential to be an alternative or complimentary to other bioactive factors. Future studies are necessary to understand mechanisms of how BSP influences bone healing and optimize delivery and dose in different types of bone defects and injuries.
Collapse
Affiliation(s)
- B L Foster
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
2
|
Ferrari M, Taboni S, Chan HHL, Townson J, Gualtieri T, Franz L, Ruaro A, Mathews S, Daly MJ, Douglas CM, Eu D, Sahovaler A, Muhanna N, Ventura M, Dey K, Pandini S, Pasini C, Re F, Bernardi S, Bosio K, Mattavelli D, Doglietto F, Joshi S, Gilbert RW, Nicolai P, Viswanathan S, Sartore L, Russo D, Irish JC. Hydrogel-chitosan and polylactic acid-polycaprolactone bioengineered scaffolds for reconstruction of mandibular defects: a preclinical in vivo study with assessment of translationally relevant aspects. Front Bioeng Biotechnol 2024; 12:1353523. [PMID: 39076208 PMCID: PMC11284118 DOI: 10.3389/fbioe.2024.1353523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 06/10/2024] [Indexed: 07/31/2024] Open
Abstract
Background: Reconstruction of mandibular bone defects is a surgical challenge, and microvascular reconstruction is the current gold standard. The field of tissue bioengineering has been providing an increasing number of alternative strategies for bone reconstruction. Methods: In this preclinical study, the performance of two bioengineered scaffolds, a hydrogel made of polyethylene glycol-chitosan (HyCh) and a hybrid core-shell combination of poly (L-lactic acid)/poly ( ε -caprolactone) and HyCh (PLA-PCL-HyCh), seeded with different concentrations of human mesenchymal stromal cells (hMSCs), has been explored in non-critical size mandibular defects in a rabbit model. The bone regenerative properties of the bioengineered scaffolds were analyzed by in vivo radiological examinations and ex vivo radiological, histomorphological, and immunohistochemical analyses. Results: The relative density increase (RDI) was significantly more pronounced in defects where a scaffold was placed, particularly if seeded with hMSCs. The immunohistochemical profile showed significantly higher expression of both VEGF-A and osteopontin in defects reconstructed with scaffolds. Native microarchitectural characteristics were not demonstrated in any experimental group. Conclusion: Herein, we demonstrate that bone regeneration can be boosted by scaffold- and seeded scaffold-reconstruction, achieving, respectively, 50% and 70% restoration of presurgical bone density in 120 days, compared to 40% restoration seen in spontaneous regeneration. Although optimization of the regenerative performance is needed, these results will help to establish a baseline reference for future experiments.
Collapse
Affiliation(s)
- Marco Ferrari
- Guided Therapeutics (GTx) Program International Scholarship, University Health Network (UHN), Toronto, ON, Canada
- Section of Otorhinolaryngology-Head and Neck Surgery, Department of Neurosciences, University of Padua, Padua, Italy
- Unit of Otorhinolaryngology-Head and Neck Surgery, Azienda Ospedale-Università di Padova, Padova, Italy
| | - Stefano Taboni
- Guided Therapeutics (GTx) Program International Scholarship, University Health Network (UHN), Toronto, ON, Canada
- Section of Otorhinolaryngology-Head and Neck Surgery, Department of Neurosciences, University of Padua, Padua, Italy
- Unit of Otorhinolaryngology-Head and Neck Surgery, Azienda Ospedale-Università di Padova, Padova, Italy
- Artificial Intelligence in Medicine and Innovation in Clinical Research and Methodology (PhD Program), Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Harley H. L. Chan
- Guided Therapeutics (GTx) Program, Techna Institute, University Health Network, Toronto, ON, Canada
| | - Jason Townson
- Guided Therapeutics (GTx) Program, Techna Institute, University Health Network, Toronto, ON, Canada
| | - Tommaso Gualtieri
- Guided Therapeutics (GTx) Program International Scholarship, University Health Network (UHN), Toronto, ON, Canada
- Department of Otorhinolaryngology, Head & Neck Surgery, Nuovo Santo Stefano Civil Hospital, Prato, Italy
| | - Leonardo Franz
- Guided Therapeutics (GTx) Program International Scholarship, University Health Network (UHN), Toronto, ON, Canada
- Section of Otorhinolaryngology-Head and Neck Surgery, Department of Neurosciences, University of Padua, Padua, Italy
| | - Alessandra Ruaro
- Guided Therapeutics (GTx) Program International Scholarship, University Health Network (UHN), Toronto, ON, Canada
- Section of Otorhinolaryngology-Head and Neck Surgery, Department of Neurosciences, University of Padua, Padua, Italy
- Unit of Otorhinolaryngology-Head and Neck Surgery, Azienda Ospedale-Università di Padova, Padova, Italy
| | - Smitha Mathews
- Osteoarthritis Program, Schroeder Arthritis Institute, Krembil Research Institute, Institute of Biomedical Engineering, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Michael J. Daly
- Guided Therapeutics (GTx) Program, Techna Institute, University Health Network, Toronto, ON, Canada
| | - Catriona M. Douglas
- Guided Therapeutics (GTx) Program, Techna Institute, University Health Network, Toronto, ON, Canada
- Princess Margaret Cancer Centre, Toronto General Hospital, Department of Otolaryngology-Head and Neck Surgery/Surgical Oncology, University Health Network, Toronto, ON, Canada
- Department of Otolaryngology, Head and Neck Surgery, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Donovan Eu
- Guided Therapeutics (GTx) Program, Techna Institute, University Health Network, Toronto, ON, Canada
- Princess Margaret Cancer Centre, Toronto General Hospital, Department of Otolaryngology-Head and Neck Surgery/Surgical Oncology, University Health Network, Toronto, ON, Canada
- Department of Otolaryngology-Head and Neck Surgery, National University Hospital, Singapore, Singapore
| | - Axel Sahovaler
- Guided Therapeutics (GTx) Program, Techna Institute, University Health Network, Toronto, ON, Canada
- Princess Margaret Cancer Centre, Toronto General Hospital, Department of Otolaryngology-Head and Neck Surgery/Surgical Oncology, University Health Network, Toronto, ON, Canada
- Head & Neck Surgery Unit, University College London Hospitals, London, United Kingdom
| | - Nidal Muhanna
- Guided Therapeutics (GTx) Program, Techna Institute, University Health Network, Toronto, ON, Canada
- Department of Otolaryngology-Head and Neck Surgery, Tel Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Manuela Ventura
- STTARR Innovation Centre, University Health Network, Toronto, ON, Canada
- Human Technopole Foundation, Milan, Italy
| | - Kamol Dey
- Department of Mechanical and Industrial Engineering, University of Brescia Via Branze, Brescia, Italy
- Department of Applied Chemistry and Chemical Engineering, Faculty of Science, University of Chittagong, Chittagong, Bangladesh
| | - Stefano Pandini
- Department of Mechanical and Industrial Engineering, University of Brescia Via Branze, Brescia, Italy
| | - Chiara Pasini
- Department of Mechanical and Industrial Engineering, University of Brescia Via Branze, Brescia, Italy
| | - Federica Re
- Unit of Blood Diseases and Bone Marrow Transplantation, Department of Clinical and Experimental Sciences, ASST Spedali Civili, University of Brescia, Brescia, Italy
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, Brescia, Italy
| | - Simona Bernardi
- Unit of Blood Diseases and Bone Marrow Transplantation, Department of Clinical and Experimental Sciences, ASST Spedali Civili, University of Brescia, Brescia, Italy
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, Brescia, Italy
| | - Katia Bosio
- Unit of Blood Diseases and Bone Marrow Transplantation, Department of Clinical and Experimental Sciences, ASST Spedali Civili, University of Brescia, Brescia, Italy
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, Brescia, Italy
| | - Davide Mattavelli
- Unit of Otorhinolaryngology-Head and Neck Surgery, ASST Spedali Civili of Brescia, Brescia, Italy
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy
| | - Francesco Doglietto
- Neurosurgery Unit, Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy
- Catholic University School of Medicine, Rome, Italy
| | - Shrinidh Joshi
- Osteoarthritis Program, Schroeder Arthritis Institute, Krembil Research Institute, Institute of Biomedical Engineering, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Ralph W. Gilbert
- Princess Margaret Cancer Centre, Toronto General Hospital, Department of Otolaryngology-Head and Neck Surgery/Surgical Oncology, University Health Network, Toronto, ON, Canada
| | - Piero Nicolai
- Section of Otorhinolaryngology-Head and Neck Surgery, Department of Neurosciences, University of Padua, Padua, Italy
- Unit of Otorhinolaryngology-Head and Neck Surgery, Azienda Ospedale-Università di Padova, Padova, Italy
| | - Sowmya Viswanathan
- Osteoarthritis Program, Schroeder Arthritis Institute, Krembil Research Institute, Institute of Biomedical Engineering, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Luciana Sartore
- Department of Mechanical and Industrial Engineering, University of Brescia Via Branze, Brescia, Italy
| | - Domenico Russo
- Unit of Blood Diseases and Bone Marrow Transplantation, Department of Clinical and Experimental Sciences, ASST Spedali Civili, University of Brescia, Brescia, Italy
| | - Jonathan C. Irish
- Guided Therapeutics (GTx) Program, Techna Institute, University Health Network, Toronto, ON, Canada
- Princess Margaret Cancer Centre, Toronto General Hospital, Department of Otolaryngology-Head and Neck Surgery/Surgical Oncology, University Health Network, Toronto, ON, Canada
| |
Collapse
|
3
|
Arambula-Maldonado R, Liu Y, Xing M, Mequanint K. Bioactive and electrically conductive GelMA-BG-MWCNT nanocomposite hydrogel bone biomaterials. BIOMATERIALS ADVANCES 2023; 154:213616. [PMID: 37708668 DOI: 10.1016/j.bioadv.2023.213616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 08/17/2023] [Accepted: 09/01/2023] [Indexed: 09/16/2023]
Abstract
Natural bone is a complex organic-inorganic composite tissue that possesses endogenous electrically conductive properties in response to mechanical forces. Mimicking these unique properties collectively in a single synthetic biomaterial has so far remained a formidable task. In this study, we report a synthesis strategy that comprised gelatin methacryloyl (GelMA), sol-gel derived tertiary bioactive glass (BG), and uniformly dispersed multiwall carbon nanotubes (MWCNTs) to create nanocomposite hydrogels that mimic the organic-inorganic composition of bone. Using this strategy, biomaterials that are electrically conductive and possess electro-mechanical properties similar to endogenous bone were prepared without affecting their biocompatibility. Nanocomposite hydrogel biomaterials were biodegradable and promoted biomineralization, and supported multipotent mesenchymal progenitor cell (10T1/2) cell interactions and differentiation into an osteogenic lineage. To the best of our knowledge, this work presents the first study to functionally characterize suitable electro-mechanical responses in nanocomposite hydrogels, a key process that occurs in the natural bone to drive its repair and regeneration. Overall, the results demonstrated GelMA-BG-MWCNT nanocomposite hydrogels have the potential to become promising bioactive biomaterials for use in bone repair and regeneration.
Collapse
Affiliation(s)
- Rebeca Arambula-Maldonado
- School of Biomedical Engineering, University of Western Ontario, 1151 Richmond Street, London N6A 5B9, Canada
| | - Yuqing Liu
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Kibret Mequanint
- School of Biomedical Engineering, University of Western Ontario, 1151 Richmond Street, London N6A 5B9, Canada; Department of Chemical and Biochemical Engineering, University of Western Ontario, 1151 Richmond Street, London N6A 5B9, Canada.
| |
Collapse
|
4
|
Solorzano A, Brady M, Bhatt N, Johnson A, Burgess B, Leyva H, Puangmalai N, Jerez C, Wood R, Kayed R, Deane R. Central and peripheral tau retention modulated by an anti-tau antibody. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.17.553682. [PMID: 37645819 PMCID: PMC10462070 DOI: 10.1101/2023.08.17.553682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Tau protein blood levels dependent on its distribution to peripheral organs and possible elimination from the body. Thus, the peripheral distribution of CSF-derived tau protein was explored, especially since there is a transition to blood-based biomarkers and the emerging idea that tau pathology may spread beyond brain. Near infrared fluorescence (NIRF) was mainly used to analyze tau (tau-NIRF) distribution after its intracisternal or intravenous injection. There was a striking uptake of blood- or CSF-derived tau-NIRF protein by the skeletal structures, liver, small intestine (duodenum), gall bladder, kidneys, urinary bladder, lymph nodes, heart, and spleen. In aging and in older APP/PS1 mice, tau uptake in regions, such as the brain, liver, and skeleton, was increased. In bone (femur) injected tau protein was associated with integrin-binding sialoprotein (IBSP), a major non-collagenous glycoprotein that is associated with mineralization. Tau-NIRF was cleared slowly from CSF via mainly across the cribriform plate, and cervical lymph nodes. In brain, some of the CSF injected tau protein was associated with NeuN-positive and PDGFRý-positive cells, which may explain its retention. The presence of tau in the bladders suggested excretion routes of tau. CSF anti-tau antibody increased CSF tau clearance, while blood anti-tau antibody decreased tau accumulation in the femur but not in liver, kidney, and spleen. Thus, the data show a body-wide distribution and retention of CSF-derived tau protein, which increased with aging and in older APP/PS1 mice. Further work is needed to elucidate the relevance of tau accumulation in each organ to tauopathy.
Collapse
|
5
|
Roth CE, Niederau C, Radermacher C, Rizk M, Neuss S, Jankowski J, Apel C, Craveiro RB, Wolf M. Knockout of Bone Sialoprotein in Cementoblasts Cell Lines Affects Specific Gene Expression in Unstimulated and Mechanically Stimulated Conditions. Ann Anat 2023; 249:152102. [PMID: 37150306 DOI: 10.1016/j.aanat.2023.152102] [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: 12/22/2022] [Revised: 02/28/2023] [Accepted: 04/17/2023] [Indexed: 05/09/2023]
Abstract
One of the major components in cementum extracellular matrix is bone sialoprotein (BSP). BSP knockout (Ibsp) mice were reported to have a nonfunctional hypo-mineralized cementum, as well as detachment and disorganization of the periodontal ligament tissue. However, studies investigating the influence of Ibsp in cementoblasts are missing yet. This study investigates the influences of Bsp in three cementoblasts cell lines (OCCM.30-WT,IbspΔNterm, and IbspKAE). The mRNA expression of cementoblast and osteoclast markers (Col1a1, Alpl, Ocn, Runx2, Ctsk, Rankl and Opg) and the cell morphology were compared. Additionally, a functional monocyte adhesion assay was performed. To understand the influence of external stimuli, the effect of Ibsp was investigated under static compressive force, mimicking the compression side of orthodontic tooth movement. Cementoblasts with genotype IbspΔNterm and IbspKAE showed slight differences in cell morphology compared to OCCM.30-WT, as well as different gene expression. Under compressive force, the Ibsp cell lines presented expression pattern markers similar to the OCCM.30-WT cell line. However, Cathepsin K was strongly upregulated in IbspΔNterm cementoblasts under compressive force. This study provides insight into the role of BSP in cementoblasts and explores the influence of BSP on periodontal ligament tissues. BSP markers in cementoblasts seem to be involved in the regulation of cementum organization as an important factor for a functional periodontium. In summary, our findings provide a basis for investigations regarding molecular biology interactions of BSP in cementoblasts, and a supporting input for understanding the periodontal and cellular cementum remodeling.
Collapse
Affiliation(s)
- Charlotte E Roth
- Department of Orthodontics, Dental Clinic, University of Aachen, 52074 Aachen, Germany; Correspondence to: Pauwelsstr. 30, 52074 Aachen, Germany. E-mail:
| | - Christian Niederau
- Department of Orthodontics, Dental Clinic, University of Aachen, 52074 Aachen, Germany
| | - Chloé Radermacher
- Department of Orthodontics, Dental Clinic, University of Aachen, 52074 Aachen, Germany; Helmholtz Institute for Biomedical Engineering, Bionterface Group, RWTH Aachen University, 52056 Aachen, Germany; Institute of Pathology, RWTH Aachen University Hospital, 52074, Germany
| | - Marta Rizk
- Department of Orthodontics, Dental Clinic, University of Aachen, 52074 Aachen, Germany
| | - Sabine Neuss
- Helmholtz Institute for Biomedical Engineering, Bionterface Group, RWTH Aachen University, 52056 Aachen, Germany; Institute of Pathology, RWTH Aachen University Hospital, 52074, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research, University Hospital RWTH Aachen, Germany
| | - Christian Apel
- Department of Biohybrid & Medical Textiles (BioTex), Institut of Applied Medical Engineering, RWTH Aachen University & Hospital, Germany
| | - Rogerio B Craveiro
- Department of Orthodontics, Dental Clinic, University of Aachen, 52074 Aachen, Germany; Correspondence to: Pauwelsstr. 30, 52074 Aachen, Germany. E-mail:
| | - Michael Wolf
- Department of Orthodontics, Dental Clinic, University of Aachen, 52074 Aachen, Germany
| |
Collapse
|
6
|
Bone Sialoprotein Immobilized in Collagen Type I Enhances Angiogenesis In Vitro and In Ovo. Polymers (Basel) 2023; 15:polym15041007. [PMID: 36850289 PMCID: PMC9968013 DOI: 10.3390/polym15041007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/24/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
Bone fracture healing is a multistep process, including early immunological reactions, osteogenesis, and as a key factor, angiogenesis. Molecules inducing osteogenesis as well as angiogenesis are rare, but hold promise to be employed in bone tissue engineering. It has been demonstrated that the bone sialoprotein (BSP) can induce bone formation when immobilized in collagen type I, but its effect on angiogenesis still has to be characterized in detail. Therefore, the aim of this study was to analyse the effects of BSP immobilized in a collagen type I gel on angiogenesis. First, in vitro analyses with endothelial cells (HUVECs) were performed detecting enhancing effects of BSP on proliferation and gene expression of endothelial markers. A spheroid model was employed confirming these results. Finally, the inducing impact of BSP-collagen on vascular density was proved in a yolk sac membrane assay. Our results demonstrate that BSP is capable of inducing angiogenesis and confirm that collagen type I is the optimal carrier for this protein. Taking into account former results, and literature showing that BSP also induces osteogenesis, one can hypothesize that BSP couples angiogenesis and osteogenesis, making it a promising molecule to be used in bone tissue regeneration.
Collapse
|
7
|
Osteogenesis in human periodontal ligament stem cell sheets is enhanced by the protease-activated receptor 1 (PAR1) in vivo. Sci Rep 2022; 12:15637. [PMID: 36117187 PMCID: PMC9482923 DOI: 10.1038/s41598-022-19520-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/30/2022] [Indexed: 11/24/2022] Open
Abstract
Human periodontal ligament stem cells (PDLSCs) have been studied as a promising strategy in regenerative approaches. The protease-activated receptor 1 (PAR1) plays a key role in osteogenesis and has been shown to induce osteogenesis and increase bone formation in PDLSCs. However, little is known about its effects when activated in PDLSCs as a cell sheet construct and how it would impact bone formation as a graft in vivo. Here, PDLSCs were obtained from 3 patients. Groups were divided into control, osteogenic medium and osteogenic medium + PAR1 activation by TFLLR-NH2 peptide. Cell phenotype was determined by flow cytometry and immunofluorescence. Calcium deposition was quantified by Alizarin Red Staining. Cell sheet microstructure was analyzed through light, scanning electron microscopy and histology and transplanted to Balb/c nude mice. Immunohistochemistry for bone sialoprotein (BSP), integrin β1 and collagen type 1 and histological stains (H&E, Van Giesson, Masson’s Trichrome and Von Kossa) were performed on the ex-vivo mineralized tissue after 60 days of implantation in vivo. Ectopic bone formation was evaluated through micro-CT. PAR1 activation increased calcium deposition in vitro as well as BSP, collagen type 1 and integrin β1 protein expression and higher ectopic bone formation (micro-CT) in vivo.
Collapse
|
8
|
Nagasaki K, Chavez M, Nagasaki A, Taylor J, Tan M, Ma M, Ralston E, Thew M, Kim DG, Somerman M, Foster B. The Bone Sialoprotein RGD Domain Modulates and Maintains Periodontal Development. J Dent Res 2022; 101:1238-1247. [PMID: 35686360 PMCID: PMC9403724 DOI: 10.1177/00220345221100794] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Bone sialoprotein (gene: Ibsp; protein: BSP) is a multifunctional extracellular matrix protein present in bone, cementum, and dentin. Accumulating evidence supports BSP as a key regulator of mineralized tissue formation via evolutionarily conserved functional domains, including a C-terminal integrin-binding Arg-Gly-Asp (RGD) domain implicated in extracellular matrix-cell signaling. Ablation of Ibsp in mice (Ibsp-/-) results in impaired bone growth and mineralization and defective osteoclastogenesis, with effects in the craniofacial region including reduced acellular cementum formation, detachment of the periodontal ligament (PDL), alveolar bone hypomineralization, and severe periodontal breakdown. We hypothesized that BSP-RGD plays an important role in cementum and alveolar bone formation and mineralization, as well as periodontal function. This hypothesis was tested by replacing the RGD motif with a nonfunctional Lys-Ala-Glu (KAE) sequence in (IbspKAE/KAE) mice and OCCM.30 murine (IbspKAE) cementoblasts. The RGD domain was not critical for acellular or cellular cementum formation in IbspKAE/KAE mice. However, PDL volume and thickness were increased, and significantly more tartrate-resistant acid phosphatase-positive osteoclasts were found on alveolar bone surfaces of IbspKAE/KAE mice versus wild type mice. PDL organization was disrupted as indicated by picrosirius red stain, second harmonic generation imaging, dynamic mechanical analysis, and decreased asporin proteoglycan localization. In vitro studies implicated RGD functions in cell migration, adhesion, and mineralization, and this was confirmed by an ossicle implant model where cells lacking BSP-RGD showed substantial defects as compared with controls. In total, the BSP-RGD domain is implicated in periodontal development, though the scale and scope of changes indicated by in vitro studies indicate that other factors may partially compensate for and reduce the phenotypic severity of mice lacking BSP-RGD in vivo.
Collapse
Affiliation(s)
- K. Nagasaki
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - M.B. Chavez
- Biosciences Division, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - A. Nagasaki
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - J.M. Taylor
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - M.H. Tan
- Biosciences Division, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - M. Ma
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - E. Ralston
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - M.E. Thew
- Division of Orthodontics, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - D.-G. Kim
- Division of Orthodontics, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - M.J. Somerman
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - B.L. Foster
- Biosciences Division, College of Dentistry, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
9
|
Li Z, Cong X, Kong W. Matricellular proteins: Potential biomarkers and mechanistic factors in aortic aneurysms. J Mol Cell Cardiol 2022; 169:41-56. [DOI: 10.1016/j.yjmcc.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 03/30/2022] [Accepted: 05/03/2022] [Indexed: 10/18/2022]
|
10
|
Nagasaki K, Gavrilova O, Hajishengallis G, Somerman MJ. Does the RGD region of certain proteins affect metabolic activity? FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2022.974862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A better understanding of the role of mineralized tissues and their associated factors in governing whole-body metabolism should be of value toward informing clinical strategies to treat mineralized tissue and metabolic disorders, such as diabetes and obesity. This perspective provides evidence suggesting a role for the arginine-glycine-aspartic acid (RGD) region, a sequence identified in several proteins secreted by bone cells, as well as other cells, in modulating systemic metabolic activity. We focus on (a) two of the SIBLING (small integrin-binding ligand, N-linked glycoprotein) family genes/proteins, bone sialoprotein (BSP) and osteopontin (OPN), (b) insulin-like growth factor-binding protein-1 & 2 (IGFBP-1, IGFBP-2) and (c) developmental endothelial locus 1 (DEL1) and milk fat globule–EGF factor-8 (MFG-E8). In addition, for our readers to appreciate the mounting evidence that a multitude of bone secreted factors affect the activity of other tissues, we provide a brief overview of other proteins, to include fibroblast growth factor 23 (FGF23), phosphatase orphan 1 (PHOSPHO1), osteocalcin (OCN/BGLAP), tissue non-specific alkaline phosphatase (TNAP) and acidic serine aspartic-rich MEPE-associated motif (ASARM), along with known/suggested functions of these factors in influencing energy metabolism.
Collapse
|
11
|
Mizuguchi Y, Mashimo Y, Mie M, Kobatake E. Temperature-Responsive Multifunctional Protein Hydrogels with Elastin-like Polypeptides for 3-D Angiogenesis. Biomacromolecules 2020; 21:1126-1135. [PMID: 32003967 DOI: 10.1021/acs.biomac.9b01496] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Supramolecular protein hydrogels with tunable properties represent promising candidates for advanced designer extracellular matrices (ECMs). To control cellular functions, ECMs should be able to spatiotemporally regulate synergistic signaling between transmembrane receptors and growth factor (GF) receptors. In this study, we developed genetically engineered temperature-responsive multifunctional protein hydrogels. The designed hydrogel was fabricated by combining the following four peptide blocks: thermosensitive elastin-like polypeptides (ELPs), a polyaspartic acid (polyD) chain to control aggregation and delivery of GFs, a de novo-designed helix peptide that forms antiparallel homotetrameric coiled-coils, and a biofunctional peptide. The resultant coiled-coil unit bound ELPs (CUBEs) exhibit a controllable sol-gel transition with tunable mechanical properties. CUBEs were functionalized with bone sialoprotein-derived RGD (bRGD), and human umbilical vein endothelial cells (HUVECs) were three-dimensionally cultured in bRGD-modified CUBE (bRGD-CUBE) hydrogels. Proangiogenic activity of HUVECs was promoted by bRGD. Moreover, heparin-binding angiogenic GFs were immobilized to bRGD-CUBEs via electrostatic interactions. HUVECs cultured in GF-tethered bRGD-CUBE hydrogels formed three-dimensional (3-D) tubulelike structures. The designed CUBE hydrogels may demonstrate utility as advanced smart biomaterials for biomedical applications. Further, the protein hydrogel design strategy may provide a novel platform for constructing designer 3-D microenvironments for specific cell types.
Collapse
Affiliation(s)
- Yoshinori Mizuguchi
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta, Midori-ku, Yokohama 226-8502, Japan
| | - Yasumasa Mashimo
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta, Midori-ku, Yokohama 226-8502, Japan
| | - Masayasu Mie
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta, Midori-ku, Yokohama 226-8502, Japan
| | - Eiry Kobatake
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta, Midori-ku, Yokohama 226-8502, Japan
| |
Collapse
|
12
|
Chakraborty S, Banerjee S, Raina M, Haldar S. Force-Directed “Mechanointeractome” of Talin–Integrin. Biochemistry 2019; 58:4677-4695. [DOI: 10.1021/acs.biochem.9b00442] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Soham Chakraborty
- Department of Biological Sciences, Ashoka University, Sonepat, Haryana 131029, India
| | - Souradeep Banerjee
- Department of Biological Sciences, Ashoka University, Sonepat, Haryana 131029, India
| | - Manasven Raina
- Department of Biological Sciences, Ashoka University, Sonepat, Haryana 131029, India
| | - Shubhasis Haldar
- Department of Biological Sciences, Ashoka University, Sonepat, Haryana 131029, India
| |
Collapse
|
13
|
Klein A, Baranowski A, Ritz U, Mack C, Götz H, Langendorf E, Al-Nawas B, Drees P, Rommens PM, Hofmann A. Effect of bone sialoprotein coating on progression of bone formation in a femoral defect model in rats. Eur J Trauma Emerg Surg 2019; 46:277-286. [PMID: 31139842 DOI: 10.1007/s00068-019-01159-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 05/21/2019] [Indexed: 11/26/2022]
Abstract
PURPOSE In orthopedic and trauma surgery, calcium phosphate cement (CPC) scaffolds are widely used as substitute for autologous bone grafts. The purpose of this study was to evaluate bone formation in a femoral condyle defect model in rats after scaffold-coating with bioactive bone sialoprotein (BSP). Our hypothesis was that BSP-coating results in additional bone formation. METHODS In 20 Wistar rats, defects of 3.0 mm diameter were drilled into the lateral femoral condyles of both legs. BSP-coated scaffolds or uncoated control scaffolds were implanted into the defects. After 4 and 8 weeks, five rats of each group were euthanized, respectively. µCT scans and histological analyses were performed. The ratio of bone volume-total volume (BV/TV) was analyzed and histological sections were evaluated. RESULTS At week four, bone fraction reached 5.2 ± 1.7% in BSP-coated scaffolds and 4.5 ± 3.2% in the control (p = 0.06). While bone fraction of the BSP-group did not change much between week four and eight [week eight: 5.4 ± 3.8% (p = 0.53)], there was a tendency towards an increase in the control [week eight: 7.0 ± 2.2% (p = 0.08)]. No significant difference in bone fraction were observable between BSP-coated and uncoated scaffolds at week eight (p = 0.08). CONCLUSIONS A significant superiority of BSP-coated scaffolds over uncoated scaffolds could not be proven. However, BSP-coating showed a tendency towards improving bone ingrowth in the scaffolds 4 weeks after implantation. This effect was only short-lived: bone growth in the control scaffolds tended to outpace that of the BSP-group at week eight.
Collapse
Affiliation(s)
- Anja Klein
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Andreas Baranowski
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany.
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Christiane Mack
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Hermann Götz
- Platform for Biomaterial Research, Biomatics Group, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Eva Langendorf
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Bilal Al-Nawas
- Department of Oral and Maxillofacial Surgery, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Philipp Drees
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Pol M Rommens
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Alexander Hofmann
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
- Department of Traumatology and Orthopaedics 1, Westpfalz-Medical Centre Kaiserslautern, Kaiserslautern, Germany
| |
Collapse
|
14
|
Veltrini VC, Figueira JA, Santin GC, de Sousa SCOM, de Araújo NS. Can non-collagenous proteins be employed for the differential diagnosis among fibrous dysplasia, cemento-osseous dysplasia and cemento-ossifying fibroma? Pathol Res Pract 2019; 215:152450. [PMID: 31109869 DOI: 10.1016/j.prp.2019.152450] [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: 01/09/2019] [Revised: 04/27/2019] [Accepted: 05/12/2019] [Indexed: 11/20/2022]
Abstract
Differential diagnosis among fibrous dysplasias, cemento-ossifying fibromas and cemento-osseous dysplasias is difficult, since there is considerable overlap of histologic features, but also extremely important, since they differ greatly in etiology, clinical behaviour, prognosis and terapeuthic approach. There is no data about the use of immunohistochemistry, a viable and accessible technique, for this purpose. The objective of this study was to investigate, comparatively, the immunohistochemical expression of major non-collagenous proteins (osteonectin [ON], osteopontin [OP], bone sialoprotein [BSP] and osteocalcin [OC]) of mineralized tissue extracellular matrix in 22 cases of fibrous dysplasias, 16 of cemento-ossifying fibromas and 16 of cemento-osseous dysplasias. ON maintained the same expression profile in all cases; the staining for OP was negative in fusiform cells producing cementoid globules and weak, as well as heterogeneous, in high mineralized matrixes; there was negativity for BSP in cementoid globules and in the fusiform cells that produce them, differently from the strong positive expression found in the majority of bone trabeculae and their peripheral cuboidal osteoblasts; and finally, the immuno-reactivity for OC was weak, except in cuboidal osteoblasts and osteocytes. We can conclude that the nature of mineralized structure and the cellular phenotype are much more responsible for variability in immunohistochemical profile than the type of lesion (fibrous dysplasias, cemento-ossifying fibromas and cemento-osseous dysplasias) which makes difficult, at least for a while, the use of these proteins with diagnosis purpose.
Collapse
Affiliation(s)
- Vanessa Cristina Veltrini
- Oral Pathology Discipline, Dentistry Department, State University of Maringa, Av. Mandacaru, 1550, CEP 87080-000, Maringa, PR, Brazil
| | - Jéssica Araújo Figueira
- Oral Oncology Center, São Paulo State University (UNESP), School of Dentistry, Rua José Bonifácio, 1193, CEP 16015-050, Araçatuba, SP, Brazil.
| | - Gabriela Cristina Santin
- Pediatric Dentistry Discipline, Dentistry Department, State University of Maringa, Av. Mandacaru, 1550, CEP 87080-000, Maringa, PR, Brazil
| | | | - Ney Soares de Araújo
- Oral Pathology Department, School of Dentistry, University of São Paulo, Av. Prof. Lineu Prestes, 2227, CEP 05508-000, São Paulo, SP, Brazil
| |
Collapse
|
15
|
Liu B, Xu M, Guo Z, Liu J, Chu X, Jiang H. Interleukin-8 promotes prostate cancer bone metastasis through upregulation of bone sialoprotein. Oncol Lett 2019; 17:4607-4613. [PMID: 30988819 PMCID: PMC6447917 DOI: 10.3892/ol.2019.10138] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 02/18/2019] [Indexed: 11/25/2022] Open
Abstract
The aim of the present study was to investigate whether interleukin-8 (IL-8) enhances the ability of prostate cancer bone metastasis by influencing the coding level of bone sialoprotein (BSP). Cultured prostate cancer cell lines LNCaP (androgen dependent) and DU145 (androgen independent) were divided into three groups: IL-8 treatment group; IL-8 receptor inhibitor (SB225002) treatment group; and control group. Western blotting and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were used to detect BSP protein and mRNA expression levels. Matrigel and bone adhesion experiments were used to detect the invasiveness of cancer cells and bone adhesion changes. Compared with the control group, western blotting and RT-qPCR results indicated that BSP protein and mRNA levels in LNCaP and DU145 were significantly upregulated following IL-8 treatment. Matrigel experiments indicated that following IL-8 treatment, the invasiveness of LNCaP and DU145 cells was significantly increased. The results of bone adhesion experiments indicated that following IL-8 treatment, the number of DU145 cells adhered to the surface of the bone was increased, compared with the control group. Following treatment of both cell lines with SB225002, western blotting and RT-qPCR results indicated that the expression levels of BSP protein and mRNA were significantly downregulated. Matrigel experiments indicated that following SB225002 treatment, the invasiveness of LNCaP and DU145 cells was significantly reduced. The number of DU145 cells adhered to the surface of the bone was reduced, compared with the untreated group. Therefore, IL-8 may promote prostate cancer bone metastasis by enhancing BSP regulation.
Collapse
Affiliation(s)
- Baohao Liu
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Meng Xu
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Zhongqing Guo
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Jiajie Liu
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Xu Chu
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Huamao Jiang
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| |
Collapse
|
16
|
Lopes D, Martins-Cruz C, Oliveira MB, Mano JF. Bone physiology as inspiration for tissue regenerative therapies. Biomaterials 2018; 185:240-275. [PMID: 30261426 PMCID: PMC6445367 DOI: 10.1016/j.biomaterials.2018.09.028] [Citation(s) in RCA: 209] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 09/15/2018] [Accepted: 09/17/2018] [Indexed: 12/14/2022]
Abstract
The development, maintenance of healthy bone and regeneration of injured tissue in the human body comprise a set of intricate and finely coordinated processes. However, an analysis of current bone regeneration strategies shows that only a small fraction of well-reported bone biology aspects has been used as inspiration and transposed into the development of therapeutic products. Specific topics that include inter-scale bone structural organization, developmental aspects of bone morphogenesis, bone repair mechanisms, role of specific cells and heterotypic cell contact in the bone niche (including vascularization networks and immune system cells), cell-cell direct and soluble-mediated contact, extracellular matrix composition (with particular focus on the non-soluble fraction of proteins), as well as mechanical aspects of native bone will be the main reviewed topics. In this Review we suggest a systematic parallelization of (i) fundamental well-established biology of bone, (ii) updated and recent advances on the understanding of biological phenomena occurring in native and injured tissue, and (iii) critical discussion of how those individual aspects have been translated into tissue regeneration strategies using biomaterials and other tissue engineering approaches. We aim at presenting a perspective on unexplored aspects of bone physiology and how they could be translated into innovative regeneration-driven concepts.
Collapse
Affiliation(s)
- Diana Lopes
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago,, 3810 193 Aveiro, Portugal
| | - Cláudia Martins-Cruz
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago,, 3810 193 Aveiro, Portugal
| | - Mariana B Oliveira
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago,, 3810 193 Aveiro, Portugal.
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago,, 3810 193 Aveiro, Portugal.
| |
Collapse
|
17
|
Evaluation of Bone Sialoprotein Coating of Three-Dimensional Printed Calcium Phosphate Scaffolds in a Calvarial Defect Model in Mice. MATERIALS 2018; 11:ma11112336. [PMID: 30469365 PMCID: PMC6267578 DOI: 10.3390/ma11112336] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 11/15/2018] [Accepted: 11/16/2018] [Indexed: 01/31/2023]
Abstract
The bioactive coating of calcium phosphate cement (CPC) is a promising approach to enhance the bone-healing properties of bone substitutes. The purpose of this study was to evaluate whether coating CPCs with bone sialoprotein (BSP) results in increased bone formation. Forty-five female C57BL/6NRj mice with an average age of six weeks were divided into three groups. Either a BSP-coated or an uncoated three-dimensional plotted scaffold was implanted into a drilled 2.7-mm diameter calvarial defect, or the defect was left empty (control group; no CPC). Histological analyses revealed that BSP-coated scaffolds were better integrated into the local bone stock eight weeks after implantation. Bone volume/total volume (BV/TV) ratios and bone thickness at the bone–implant contact were analyzed via micro computed tomography (µCT) after eight weeks. BSP-coated scaffolds and uncoated CPC scaffolds increased bone thickness in comparison to the control (CPC + BSP: 691.1 ± 253.5 µm, CPC: 603.1 ± 164.4 µm, no CPC: 261.7 ± 37.8 µm, p < 0.01). Accordingly, BV/TV was enhanced in both scaffold groups (CPC + BSP: 1.3 ± 0.5%, CPC: 0.9 ± 0.5%, no CPC: 0.2 ± 0.3%, p < 0.01). The BSP coating showed a tendency towards an increased bone thickness (p = 0.18) and BV/TV (p = 0.18) in comparison to uncoated CPC scaffolds. However, a significant increase in bone formation through BSP coating was not found.
Collapse
|
18
|
Klein A, Baranowski A, Ritz U, Götz H, Heinemann S, Mattyasovszky S, Rommens PM, Hofmann A. Effect of bone sialoprotein coated three-dimensional printed calcium phosphate scaffolds on primary human osteoblasts. J Biomed Mater Res B Appl Biomater 2018; 106:2565-2575. [PMID: 29316208 DOI: 10.1002/jbm.b.34073] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/29/2017] [Accepted: 12/22/2017] [Indexed: 01/14/2023]
Abstract
The combination of the two techniques of rapid prototyping 3D-plotting and bioactive surface functionalization is presented, with emphasis on the in vitro effect of Bone Sialoprotein (BSP) on primary human osteoblasts (hOBs). Our primary objective was to demonstrate the BSP influence on the expression of distinctive osteoblast markers in hOBs. Secondary objectives included examinations of the scaffolds' surface and the stability of BSP-coating as well as investigations of cell viability and proliferation. 3D-plotted calcium phosphate cement (CPC) scaffolds were coated with BSP via physisorption. hOBs were seeded on the coated scaffolds, followed by cell viability measurements, gene expression analysis and visualization. Physisorption is an effective method for BSP-coating. Coating with higher BSP concentrations leads to enhanced BSP release. Two BSP concentrations (50 and 200 μg/mL) were examined in this study. The lower BSP concentration (50 µg/mL) decreased ALP and SPARC expression, whereas the higher BSP concentration (200 μg/mL) did not change gene marker expression. Enhanced cell viability was observed on BSP-coated scaffolds on day 3. hOBs developed a polygonal shape and connected in an intercellular network under BSP influence. Quantitative cell morphology analyses demonstrated for BSP-coated CPCs an enhanced cell area and reduced circularity. The strength of the above-mentioned effects of BSP-coated scaffolds in vivo is unknown, and future work is focusing on bone ingrowth and vascularization in vivo. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 2565-2575, 2018.
Collapse
Affiliation(s)
- Anja Klein
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Andreas Baranowski
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Hermann Götz
- Platform for Biomaterial Research, Biomatics Group, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | | | - Stefan Mattyasovszky
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Pol M Rommens
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Alexander Hofmann
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
19
|
Bouleftour W, Granito RN, Vanden-Bossche A, Sabido O, Roche B, Thomas M, Linossier MT, Aubin JE, Lafage-Proust MH, Vico L, Malaval L. Bone Shaft Revascularization After Marrow Ablation Is Dramatically Accelerated in BSP-/- Mice, Along With Faster Hematopoietic Recolonization. J Cell Physiol 2016; 232:2528-2537. [PMID: 27704558 DOI: 10.1002/jcp.25630] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 10/03/2016] [Indexed: 12/20/2022]
Abstract
The bone organ integrates the activity of bone tissue, bone marrow, and blood vessels and the factors ensuring this coordination remain ill defined. Bone sialoprotein (BSP) is with osteopontin (OPN) a member of the small integrin binding ligand N-linked glycoprotein (SIBLING) family, involved in bone formation, hematopoiesis and angiogenesis. In rodents, bone marrow ablation induces a rapid formation of medullary bone which peaks by ∼8 days (d8) and is blunted in BSP-/- mice. We investigated the coordinate hematopoietic and vascular recolonization of the bone shaft after marrow ablation of 2 month old BSP+/+ and BSP-/- mice. At d3, the ablated area in BSP-/- femurs showed higher vessel density (×4) and vascular volume (×7) than BSP+/+. Vessel numbers in the shaft of ablated BSP+/+ mice reached BSP-/- values only by d8, but with a vascular volume which was twice the value in BSP-/-, reflecting smaller vessel size in ablated mutants. At d6, a much higher number of Lin- (×3) as well as LSK (Lin- IL-7Rα- Sca-1hi c-Kithi , ×2) and hematopoietic stem cells (HSC: Flt3- LSK, ×2) were counted in BSP-/- marrow, indicating a faster recolonization. However, the proportion of LSK and HSC within the Lin- was lower in BSP-/- and more differentiated stages were more abundant, as also observed in unablated bone, suggesting that hematopoietic differentiation is favored in the absence of BSP. Interestingly, unablated BSP-/- femur marrow also contains more blood vessels than BSP+/+, and in both intact and ablated shafts expression of VEGF and OPN are higher, and DMP1 lower in the mutants. In conclusion, bone marrow ablation in BSP-/- mice is followed by a faster vascular and hematopoietic recolonization, along with lower medullary bone formation. Thus, lack of BSP affects the interplay between hematopoiesis, angiogenesis, and osteogenesis, maybe in part through higher expression of VEGF and the angiogenic SIBLING, OPN. J. Cell. Physiol. 232: 2528-2537, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Wafa Bouleftour
- Laboratoire de Biologie des Tissus Ostéoarticulaires, INSERM, U1059 Sainbiose, Université de Lyon, Université Jean Monnet, Faculté de Médecine, Campus Santé Innovation, Saint-Étienne, France
| | - Renata Neves Granito
- Laboratoire de Biologie des Tissus Ostéoarticulaires, INSERM, U1059 Sainbiose, Université de Lyon, Université Jean Monnet, Faculté de Médecine, Campus Santé Innovation, Saint-Étienne, France
| | - Arnaud Vanden-Bossche
- Laboratoire de Biologie des Tissus Ostéoarticulaires, INSERM, U1059 Sainbiose, Université de Lyon, Université Jean Monnet, Faculté de Médecine, Campus Santé Innovation, Saint-Étienne, France
| | - Odile Sabido
- Laboratoire de Biologie des Tissus Ostéoarticulaires, INSERM, U1059 Sainbiose, Université de Lyon, Université Jean Monnet, Faculté de Médecine, Campus Santé Innovation, Saint-Étienne, France.,Flow Cytometry Facility, Université de Lyon, Université Jean Monnet, Faculté de Médecine, Campus Santé Innovation, Saint-Étienne, France
| | - Bernard Roche
- Laboratoire de Biologie des Tissus Ostéoarticulaires, INSERM, U1059 Sainbiose, Université de Lyon, Université Jean Monnet, Faculté de Médecine, Campus Santé Innovation, Saint-Étienne, France
| | - Mireille Thomas
- Laboratoire de Biologie des Tissus Ostéoarticulaires, INSERM, U1059 Sainbiose, Université de Lyon, Université Jean Monnet, Faculté de Médecine, Campus Santé Innovation, Saint-Étienne, France
| | - Marie Thérèse Linossier
- Laboratoire de Biologie des Tissus Ostéoarticulaires, INSERM, U1059 Sainbiose, Université de Lyon, Université Jean Monnet, Faculté de Médecine, Campus Santé Innovation, Saint-Étienne, France
| | - Jane E Aubin
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Marie-Hélène Lafage-Proust
- Laboratoire de Biologie des Tissus Ostéoarticulaires, INSERM, U1059 Sainbiose, Université de Lyon, Université Jean Monnet, Faculté de Médecine, Campus Santé Innovation, Saint-Étienne, France
| | - Laurence Vico
- Laboratoire de Biologie des Tissus Ostéoarticulaires, INSERM, U1059 Sainbiose, Université de Lyon, Université Jean Monnet, Faculté de Médecine, Campus Santé Innovation, Saint-Étienne, France
| | - Luc Malaval
- Laboratoire de Biologie des Tissus Ostéoarticulaires, INSERM, U1059 Sainbiose, Université de Lyon, Université Jean Monnet, Faculté de Médecine, Campus Santé Innovation, Saint-Étienne, France
| |
Collapse
|
20
|
Schweighofer N, Aigelsreiter A, Trummer O, Graf-Rechberger M, Hacker N, Kniepeiss D, Wagner D, Stiegler P, Trummer C, Pieber T, Obermayer-Pietsch B, Müller H. Direct comparison of regulators of calcification between bone and vessels in humans. Bone 2016; 88:31-38. [PMID: 27108945 DOI: 10.1016/j.bone.2016.04.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 11/30/2015] [Accepted: 04/16/2016] [Indexed: 01/16/2023]
Abstract
Calcification is not only physiologically present in bone but is a main pathophysiological process in vasculature, favouring cardiovascular diseases. Our aim was to investigate changes in the expression of calcification regulators during vascular calcification in bone and vasculature. Levels of gene expression of osteoprotegerin (OPG), receptor activator of NF-κB ligand (RANKL), osteopontin (OPN), matrix gla protein (MGP), bone sialoprotein (BSP), SMAD6, and runt-related transcription factor 2 (RUNX2) were determined in bone, aorta, and external iliac artery tissue samples of transplant donors. Histological stages of atherosclerosis (AS) in vessels are defined as "no changes", "intima thickening", or "intima calcification". Patients' bone samples were subgrouped accordingly. We demonstrate that in vessels BSP and OPN expression significantly increased during intima thickening and decreased during intima calcification, whereas the expression of regulators of calcification did not significantly change in bone during intima thickening and intima calcification. At the stage of intima thickening, MGP, OPG, and SMAD6 expression and at stage of intima calcification only MGP expression was lower in bone than in vessel. The expression of BSP and RANKL was regulated in opposite ways in bone and vessels, whereas the expression of MGP, OC, RUNX2, and OPN was regulated in a tissue-specific manner. Our study is the first direct comparison of gene expression changes during AS progression in bone and vessels. Our results indicate that changes in the expression of regulators of calcification in the vessel wall as well as in bone occur early in the calcification process, even prior to deposition of calcium/phosphate precipitation.
Collapse
Affiliation(s)
- N Schweighofer
- Department of Internal Medicine, Divison of Endocrinology and Diabetology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - A Aigelsreiter
- Institute of Pathology, Medical University of Graz, Auenbruggerplatz 25, 8036 Graz, Austria
| | - O Trummer
- Department of Internal Medicine, Divison of Endocrinology and Diabetology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - M Graf-Rechberger
- Institute of Pathology, Medical University of Graz, Auenbruggerplatz 25, 8036 Graz, Austria
| | - N Hacker
- Department of Internal Medicine, Divison of Endocrinology and Diabetology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - D Kniepeiss
- Department of Surgery, Division of Transplantation Surgery, Medical University of Graz, Auenbruggerplatz 29, 8036 Graz, Austria
| | - D Wagner
- Department of Surgery, Division of Transplantation Surgery, Medical University of Graz, Auenbruggerplatz 29, 8036 Graz, Austria
| | - P Stiegler
- Department of Surgery, Division of Transplantation Surgery, Medical University of Graz, Auenbruggerplatz 29, 8036 Graz, Austria
| | - C Trummer
- Department of Internal Medicine, Divison of Endocrinology and Diabetology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - T Pieber
- Department of Internal Medicine, Divison of Endocrinology and Diabetology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria; Joanneum Research Health, Elisabethstrasse 5, 8010 Graz, Austria
| | - B Obermayer-Pietsch
- Department of Internal Medicine, Divison of Endocrinology and Diabetology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria.
| | - H Müller
- Department of Surgery, Division of Transplantation Surgery, Medical University of Graz, Auenbruggerplatz 29, 8036 Graz, Austria
| |
Collapse
|
21
|
Yang Y, Huang Y, Zhang L, Zhang C. Transcriptional regulation of bone sialoprotein gene expression by Osx. Biochem Biophys Res Commun 2016; 476:574-579. [PMID: 27261434 DOI: 10.1016/j.bbrc.2016.05.164] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 05/30/2016] [Indexed: 10/21/2022]
Abstract
Osteoporosis is the most common metabolic bone disease characterized by decreased bone mass, decreased bone strength, and increased risk of fracture. It is due to unbalance between bone formation and bone resorption. Bone formation is a complex process which involves the differentiation of mesenchymal stem cells to osteoblasts. Osteoblasts produce a characteristic extracellular collagenous matrix that subsequently becomes mineralized. Osterix (Osx) is an osteoblast-specific transcription factor required for osteoblast differentiation. Bone sialoprotein (Bsp) is a member of the SIBLING gene family. Expression of Bsp correlates with the differentiation of osteoblasts and the onset of mineralization. Our preliminary data showed that Bsp was abolished in Osx-null mice; however, the detailed mechanism of Osx regulation on Bsp is not fully understood. In this study, regulation of Bsp expression by Osx was further characterized. It was shown that overexpression of Osx led to Bsp upregulation. Inhibition of Osx by small interfering RNA resulted in Bsp downregulation in osteoblast. Transfection assay demonstrated that Osx was able to activate Bsp promoter reporter in a dose-dependent manner. To define minimal region of Bsp promoter activated by Osx, a series of deletion mutants of Bsp promoter were generated, and the minimal region was narrowed down to the proximal 100 bp. Point-mutagenesis studies showed that one GC-rich site was required for Bsp promoter activation by Osx. ChIP assays demonstrated that endogenous Osx associated with native Bsp promoter in primary osteoblasts. Our observations provide evidence that Osx targets Bsp expression directly.
Collapse
Affiliation(s)
- Ya Yang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Yehong Huang
- Department of Research, Daobio, Inc., Dallas, TX 75093, USA
| | - Li Zhang
- Department of Research, Daobio, Inc., Dallas, TX 75093, USA
| | - Chi Zhang
- Department of Research, Daobio, Inc., Dallas, TX 75093, USA; Bone Research Laboratory, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
22
|
Baranowski A, Klein A, Ritz U, Ackermann A, Anthonissen J, Kaufmann KB, Brendel C, Götz H, Rommens PM, Hofmann A. Surface Functionalization of Orthopedic Titanium Implants with Bone Sialoprotein. PLoS One 2016; 11:e0153978. [PMID: 27111551 PMCID: PMC4844107 DOI: 10.1371/journal.pone.0153978] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 04/06/2016] [Indexed: 12/22/2022] Open
Abstract
Orthopedic implant failure due to aseptic loosening and mechanical instability remains a major problem in total joint replacement. Improving osseointegration at the bone-implant interface may reduce micromotion and loosening. Bone sialoprotein (BSP) has been shown to enhance bone formation when coated onto titanium femoral implants and in rat calvarial defect models. However, the most appropriate method of BSP coating, the necessary level of BSP coating, and the effect of BSP coating on cell behavior remain largely unknown. In this study, BSP was covalently coupled to titanium surfaces via an aminosilane linker (APTES), and its properties were compared to BSP applied to titanium via physisorption and untreated titanium. Cell functions were examined using primary human osteoblasts (hOBs) and L929 mouse fibroblasts. Gene expression of specific bone turnover markers at the RNA level was detected at different intervals. Cell adhesion to titanium surfaces treated with BSP via physisorption was not significantly different from that of untreated titanium at any time point, whereas BSP application via covalent coupling caused reduced cell adhesion during the first few hours in culture. Cell migration was increased on titanium disks that were treated with higher concentrations of BSP solution, independent of the coating method. During the early phases of hOB proliferation, a suppressive effect of BSP was observed independent of its concentration, particularly when BSP was applied to the titanium surface via physisorption. Although alkaline phosphatase activity was reduced in the BSP-coated titanium groups after 4 days in culture, increased calcium deposition was observed after 21 days. In particular, the gene expression level of RUNX2 was upregulated by BSP. The increase in calcium deposition and the stimulation of cell differentiation induced by BSP highlight its potential as a surface modifier that could enhance the osseointegration of orthopedic implants. Both physisorption and covalent coupling of BSP are similarly effective, feasible methods, although a higher BSP concentration is recommended.
Collapse
Affiliation(s)
- Andreas Baranowski
- Department of Orthopedics and Traumatology, University Medical Centre, Johannes Gutenberg University, Mainz, Germany
| | - Anja Klein
- Department of Orthopedics and Traumatology, University Medical Centre, Johannes Gutenberg University, Mainz, Germany
| | - Ulrike Ritz
- Department of Orthopedics and Traumatology, University Medical Centre, Johannes Gutenberg University, Mainz, Germany
| | - Angelika Ackermann
- Department of Orthopedics and Traumatology, University Medical Centre, Johannes Gutenberg University, Mainz, Germany
| | - Joris Anthonissen
- Department of Orthopedics and Traumatology, University Medical Centre, Johannes Gutenberg University, Mainz, Germany
| | - Kerstin B. Kaufmann
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Christian Brendel
- Division of Pediatric Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Hermann Götz
- Platform for Biomaterial Research, University Medical Centre, Johannes Gutenberg University, Mainz, Germany
| | - Pol M. Rommens
- Department of Orthopedics and Traumatology, University Medical Centre, Johannes Gutenberg University, Mainz, Germany
| | - Alexander Hofmann
- Department of Orthopedics and Traumatology, University Medical Centre, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
23
|
Tharp KM, Jha AK, Kraiczy J, Yesian A, Karateev G, Sinisi R, Dubikovskaya EA, Healy KE, Stahl A. Matrix-Assisted Transplantation of Functional Beige Adipose Tissue. Diabetes 2015; 64:3713-24. [PMID: 26293504 PMCID: PMC4613967 DOI: 10.2337/db15-0728] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 07/21/2015] [Indexed: 12/14/2022]
Abstract
Novel, clinically relevant, approaches to shift energy balance are urgently needed to combat metabolic disorders such as obesity and diabetes. One promising approach has been the expansion of brown adipose tissues that express uncoupling protein (UCP) 1 and thus can uncouple mitochondrial respiration from ATP synthesis. While expansion of UCP1-expressing adipose depots may be achieved in rodents via genetic and pharmacological manipulations or the transplantation of brown fat depots, these methods are difficult to use for human clinical intervention. We present a novel cell scaffold technology optimized to establish functional brown fat-like depots in vivo. We adapted the biophysical properties of hyaluronic acid-based hydrogels to support the differentiation of white adipose tissue-derived multipotent stem cells (ADMSCs) into lipid-accumulating, UCP1-expressing beige adipose tissue. Subcutaneous implantation of ADMSCs within optimized hydrogels resulted in the establishment of distinct UCP1-expressing implants that successfully attracted host vasculature and persisted for several weeks. Importantly, implant recipients demonstrated elevated core body temperature during cold challenges, enhanced respiration rates, improved glucose homeostasis, and reduced weight gain, demonstrating the therapeutic merit of this highly translatable approach. This novel approach is the first truly clinically translatable system to unlock the therapeutic potential of brown fat-like tissue expansion.
Collapse
Affiliation(s)
- Kevin M Tharp
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences & Toxicology, University of California, Berkeley, Berkeley, CA Department of Bioengineering, University of California, Berkeley, Berkeley, CA
| | - Amit K Jha
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA
| | - Judith Kraiczy
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences & Toxicology, University of California, Berkeley, Berkeley, CA
| | - Alexandra Yesian
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences & Toxicology, University of California, Berkeley, Berkeley, CA
| | - Grigory Karateev
- Institute of Chemical Sciences and Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Riccardo Sinisi
- Institute of Chemical Sciences and Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Elena A Dubikovskaya
- Institute of Chemical Sciences and Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Kevin E Healy
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA Department of Materials Science & Engineering, University of California, Berkeley, Berkeley, CA
| | - Andreas Stahl
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences & Toxicology, University of California, Berkeley, Berkeley, CA
| |
Collapse
|
24
|
Molecular weight and concentration of heparin in hyaluronic acid-based matrices modulates growth factor retention kinetics and stem cell fate. J Control Release 2015; 209:308-16. [PMID: 25931306 DOI: 10.1016/j.jconrel.2015.04.034] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 04/20/2015] [Accepted: 04/25/2015] [Indexed: 12/22/2022]
Abstract
Growth factors are critical for regulating and inducing various stem cell functions. To study the effects of growth factor delivery kinetics and presentation on stem cell fate, we developed a series of heparin-containing hyaluronic acid (HyA)-based hydrogels with various degrees of growth factor affinity and retention. To characterize this system, we investigated the effect of heparin molecular weight, fractionation, and relative concentration on the loading efficiency and retention kinetics of TGFβ1 as a model growth factor. At equal concentrations, high MW heparin both loaded and retained the greatest amount of TGFβ1, and had the slowest release kinetics, primarily due to the higher affinity with TGFβ1 compared to low MW or unfractionated heparin. Subsequently, we tested the effect of TGFβ1, presented from various heparin-containing matrices, to differentiate a versatile population of Sca-1(+)/CD45(-) cardiac progenitor cells (CPCs) into endothelial cells and form vascular-like networks in vitro. High MW heparin HyA hydrogels stimulated more robust differentiation of CPCs into endothelial cells, which formed vascular-like networks within the hydrogel. This observation was attributed to the ability of high MW heparin HyA hydrogels to sequester endogenously synthesized angiogenic factors within the matrix. These results demonstrate the importance of molecular weight, fractionation, and concentration of heparin on presentation of heparin-binding growth factors and their effect on stem cell differentiation and lineage specification.
Collapse
|
25
|
Holm E, Aubin JE, Hunter GK, Beier F, Goldberg HA. Loss of bone sialoprotein leads to impaired endochondral bone development and mineralization. Bone 2015; 71:145-54. [PMID: 25464126 DOI: 10.1016/j.bone.2014.10.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 10/07/2014] [Accepted: 10/14/2014] [Indexed: 10/24/2022]
Abstract
Bone sialoprotein (BSP) is an anionic phosphoprotein in the extracellular matrix of mineralized tissues, and a promoter of biomineralization and osteoblast development. Previous studies on the Bsp-deficient mouse (Bsp(-/-)) have demonstrated a significant bone and periodontal tissue phenotype in adulthood. However, the role of BSP during early long bone development is not known. To address this, early endochondral ossification in the Bsp(-/-) mouse was studied. Embryonic day 15.5 (E15.5) wild-type (WT) tibiae showed early stages of ossification that were absent in Bsp(-/-) mice. At E16.5, mineralization had commenced in the Bsp(-/-) mice, but staining for mineral was less intense and more dispersed compared with that in WT controls. Tibiae from Bsp(-/-) mice also demonstrated decreased mineralization and shortened length at postnatal day 0.5 (P0.5) compared to WT bones. There was no detectable difference in the number of tartrate-resistant acid phosphatase-positive foci at P0.5, although the P0.5 Bsp(-/-) tibiae had decreased Vegfα expression compared with WT tissue. Due to the shortened tibiae the growth plates were examined and determined to be of normal overall length. However, the length of the resting zone was increased in P0.5 Bsp(-/-) tibiae whereas that of the proliferative zone was decreased, with no change in the hypertrophic zone length of Bsp(-/-) mice. A reduction in cells positive for Ki-67, an S-phase cell-cycle marker, was noted in the proliferative zone. Decreased numbers of TUNEL-positive hypertrophic chondrocytes were also apparent in the Bsp(-/-) tibial growth plates, suggesting decreased apoptosis. Expression of the osteogenic markers Alp1, Col1a1, Sp7, Runx2, and Bglap was reduced in the endochondral bone of the neonatal Bsp(-/-) compared to WT tibiae. These results suggest that BSP is an important and multifaceted protein that regulates both chondrocyte proliferation and apoptosis as well as transition from cartilage to bone during development of endochondral bone.
Collapse
Affiliation(s)
- Erik Holm
- Department of Biochemistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1.
| | - Jane E Aubin
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5S 1A8.
| | - Graeme K Hunter
- Department of Biochemistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1; School of Dentistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1.
| | - Frank Beier
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada, N6A 5C1.
| | - Harvey A Goldberg
- Department of Biochemistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1; School of Dentistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1.
| |
Collapse
|
26
|
Bouleftour W, Boudiffa M, Wade-Gueye NM, Bouët G, Cardelli M, Laroche N, Vanden-Bossche A, Thomas M, Bonnelye E, Aubin JE, Vico L, Lafage-Proust MH, Malaval L. Skeletal development of mice lacking bone sialoprotein (BSP)--impairment of long bone growth and progressive establishment of high trabecular bone mass. PLoS One 2014; 9:e95144. [PMID: 24816232 PMCID: PMC4015893 DOI: 10.1371/journal.pone.0095144] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 03/24/2014] [Indexed: 01/28/2023] Open
Abstract
Adult Ibsp-knockout mice (BSP−/−) display shorter stature, lower bone turnover and higher trabecular bone mass than wild type, the latter resulting from impaired bone resorption. Unexpectedly, BSP knockout also affects reproductive behavior, as female mice do not construct a proper "nest" for their offsprings. Multiple crossing experiments nonetheless indicated that the shorter stature and lower weight of BSP−/− mice, since birth and throughout life, as well as their shorter femur and tibia bones are independent of the genotype of the mothers, and thus reflect genetic inheritance. In BSP−/− newborns, µCT analysis revealed a delay in membranous primary ossification, with wider cranial sutures, as well as thinner femoral cortical bone and lower tissue mineral density, reflected in lower expression of bone formation markers. However, trabecular bone volume and osteoclast parameters of long bones do not differ between genotypes. Three weeks after birth, osteoclast number and surface drop in the mutants, concomitant with trabecular bone accumulation. The growth plates present a thinner hypertrophic zone in newborns with lower whole bone expression of IGF-1 and higher IHH in 6 days old BSP−/− mice. At 3 weeks the proliferating zone is thinner and the hypertrophic zone thicker in BSP−/− than in BSP+/+ mice of either sex, maybe reflecting a combination of lower chondrocyte proliferation and impaired cartilage resorption. Six days old BSP−/− mice display lower osteoblast marker expression but higher MEPE and higher osteopontin(Opn)/Runx2 ratio. Serum Opn is higher in mutants at day 6 and in adults. Thus, lack of BSP alters long bone growth and membranous/cortical primary bone formation and mineralization. Endochondral development is however normal in mutant mice and the accumulation of trabecular bone observed in adults develops progressively in the weeks following birth. Compensatory high Opn may allow normal endochondral development in BSP−/− mice, while impairing primary mineralization.
Collapse
Affiliation(s)
- Wafa Bouleftour
- Laboratoire de Biologie du Tissu Osseux and Institut National de la Santé et de la Recherche Médicale - U1059, Université de Lyon - Université Jean Monnet, Saint-Etienne, France
| | - Maya Boudiffa
- Laboratoire de Biologie du Tissu Osseux and Institut National de la Santé et de la Recherche Médicale - U1059, Université de Lyon - Université Jean Monnet, Saint-Etienne, France
| | - Ndeye Marième Wade-Gueye
- Laboratoire de Biologie du Tissu Osseux and Institut National de la Santé et de la Recherche Médicale - U1059, Université de Lyon - Université Jean Monnet, Saint-Etienne, France
| | - Guénaëlle Bouët
- Laboratoire de Biologie du Tissu Osseux and Institut National de la Santé et de la Recherche Médicale - U1059, Université de Lyon - Université Jean Monnet, Saint-Etienne, France
| | - Marco Cardelli
- Dept. of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Norbert Laroche
- Laboratoire de Biologie du Tissu Osseux and Institut National de la Santé et de la Recherche Médicale - U1059, Université de Lyon - Université Jean Monnet, Saint-Etienne, France
| | - Arnaud Vanden-Bossche
- Laboratoire de Biologie du Tissu Osseux and Institut National de la Santé et de la Recherche Médicale - U1059, Université de Lyon - Université Jean Monnet, Saint-Etienne, France
| | - Mireille Thomas
- Laboratoire de Biologie du Tissu Osseux and Institut National de la Santé et de la Recherche Médicale - U1059, Université de Lyon - Université Jean Monnet, Saint-Etienne, France
| | - Edith Bonnelye
- Institut National de la Santé et de la Recherche Médicale - U1033, Université de Lyon - Université Claude Bernard, Lyon, France
| | - Jane E. Aubin
- Dept. of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Laurence Vico
- Laboratoire de Biologie du Tissu Osseux and Institut National de la Santé et de la Recherche Médicale - U1059, Université de Lyon - Université Jean Monnet, Saint-Etienne, France
| | - Marie Hélène Lafage-Proust
- Laboratoire de Biologie du Tissu Osseux and Institut National de la Santé et de la Recherche Médicale - U1059, Université de Lyon - Université Jean Monnet, Saint-Etienne, France
| | - Luc Malaval
- Laboratoire de Biologie du Tissu Osseux and Institut National de la Santé et de la Recherche Médicale - U1059, Université de Lyon - Université Jean Monnet, Saint-Etienne, France
- * E-mail:
| |
Collapse
|
27
|
Pesesse L, Sanchez C, Walsh DA, Delcour JP, Baudouin C, Msika P, Henrotin Y. Bone sialoprotein as a potential key factor implicated in the pathophysiology of osteoarthritis. Osteoarthritis Cartilage 2014; 22:547-56. [PMID: 24530278 DOI: 10.1016/j.joca.2014.01.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 01/21/2014] [Accepted: 01/27/2014] [Indexed: 02/02/2023]
Abstract
OBJECTIVE We previously identified an association between bone sialoprotein (BSP) and osteoarthritic (OA) chondrocyte hypertrophy but the precise role of BSP in ostearthritis (OA) has not been extensively studied. This study aimed to confirm the association between BSP and OA chondrocyte hypertrophy, to define its effect on molecules produced by chondrocytes and to analyse its association with cartilage degradation and vascular density at the osteochondral junction. METHOD Human OA chondrocytes were cultivated in order to increase hypertrophic differentiation. The effect of parathyroid hormone-related peptide (PTHrP), interleukin (IL)-1β or tumour necrosis factor (TNF)-α on BSP was analysed by real-time reverse transcription polymerase chain reaction (RT-PCR) and western blot. The effects of BSP on OA chondrocytes production of inflammatory response mediators (IL-6, nitric oxide), major matrix molecule (aggrecan), matrix metalloprotease-3 and angiogenic factors (vascular endothelial growth factor, basic fibroblast growth factor, IL-8, and thrombospondin-1) were investigated. BSP was detected by immunohistochemistry and was associated with cartilage lesions severity and vascular density. RESULTS PTHrP significantly decreased BSP, confirming its association with chondrocyte hypertrophy. In presence of IL-1β, BSP stimulated IL-8 synthesis, a pro-angiogenic cytokine but decreased the production of TSP-1, an angiogenesis inhibitor. The presence of BSP-immunoreactive chondrocytes in cartilage was associated with the severity of histological cartilage lesions and with vascular density at the osteochondral junction. CONCLUSION This study supports the implication of BSP in the pathology of OA and suggests that it could be a key mediator of the hypertrophic chondrocytes-induced angiogenesis. To control chondrocyte hypertrophic differentiation is promising in the treatment of OA.
Collapse
Affiliation(s)
- L Pesesse
- Bone and Cartilage Research Unit, University of Liege, Liege, Belgium.
| | - C Sanchez
- Bone and Cartilage Research Unit, University of Liege, Liege, Belgium.
| | - D A Walsh
- Arthritis Research UK Pain Centre, Academic Rheumatology, University of Nottingham, Nottingham, United Kingdom.
| | - J-P Delcour
- Bois de l'Abbaye Hospital, Seraing, Belgium.
| | - C Baudouin
- Expanscience Laboratories, IRD Direction, Epernon, France.
| | - P Msika
- Expanscience Laboratories, IRD Direction, Epernon, France.
| | - Y Henrotin
- Bone and Cartilage Research Unit, University of Liege, Liege, Belgium; Princess Paola Hospital, Vivalia, Marche-en-Famenne, Belgium.
| |
Collapse
|
28
|
Xie X, Ma S, Li C, Liu P, Wang H, Chen L, Qin C. Expression of Small Integrin-Binding LIgand N-linked Glycoproteins (SIBLINGs) in the reparative dentin of rat molars. Dent Traumatol 2014; 30:285-95. [PMID: 24502800 DOI: 10.1111/edt.12093] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2013] [Indexed: 12/27/2022]
Abstract
AIM To analyze the expression and distribution of Small Integrin-Binding LIgand N-linked Glycoproteins (SIBLINGs) in reparative dentin (RepD). METHODOLOGY Cavities on the mesial surfaces of rat molars were prepared to expose the pulp, and a calcium hydroxide agent was applied to cap the exposed pulp. The molars with pulp capping were extracted at postoperative 1, 2, and 4 weeks. The immunolocalization of four SIBLINGs, dentin matrix protein 1 (DMP1), dentin sialoprotein (DSP), bone sialoprotein (BSP), and osteopontin (OPN) in RepD, was analyzed in comparison with reactionary dentin (ReaD) and primary dentin (PD). RESULTS At two weeks after operation, the region of the exposed pulp formed a layer of reparative dentin bridge sealing the communication between the cavity and pulp chamber. Dentinal tubules in RepD were more irregular in shape and fewer in number than PD. At postoperative 2 and 4 weeks, RepD had lower levels of DMP1 and DSP than PD. BSP and OPN were present in RepD, but not in PD. RepD showed certain similarities to ReaD in the expression of SIBLINGs. CONCLUSIONS The reduced levels of DMP1 and DSP may be associated with the decreased number of dentinal tubules in RepD. The expression of BSP and OPN in RepD indicates that the odontoblast-like cells were attempting to produce a hard tissue at a very rapid pace. These findings suggest that in response to the surgical injury, the newly differentiated odontoblast-like cells altered their synthesis of the dentinogenesis-related proteins and produced a hard tissue that is an intermediate between dentin and bone.
Collapse
Affiliation(s)
- Xiaohua Xie
- Longjiang Scholar Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Endodontics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | | | | | | | | | | | | |
Collapse
|
29
|
Kruger TE, Miller AH, Godwin AK, Wang J. Bone sialoprotein and osteopontin in bone metastasis of osteotropic cancers. Crit Rev Oncol Hematol 2014; 89:330-41. [PMID: 24071501 PMCID: PMC3946954 DOI: 10.1016/j.critrevonc.2013.08.013] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 08/23/2013] [Accepted: 08/28/2013] [Indexed: 02/06/2023] Open
Abstract
The mechanisms underlying malignant cell metastasis to secondary sites such as bone are complex and no doubt multifactorial. Members of the small integrin-binding ligand N-linked glycoproteins (SIBLINGs) family, particularly bone sialoprotein (BSP) and osteopontin (OPN), exhibit multiple activities known to promote malignant cell proliferation, detachment, invasion, and metastasis of several osteotropic cancers. The expression level of BSP and OPN is elevated in a variety of human cancers, particularly those that metastasize preferentially to the skeleton. Recent studies suggest that the "osteomimicry" of malignant cells is not only conferred by transmembrane receptors bound by BSP and OPN, but includes the "switch" in gene expression repertoire typically expressed in cells of skeletal lineage. Understanding the role of BSP and OPN in tumor progression, altered pathophysiology of bone microenvironment, and tumor metastasis to bone will likely result in development of better diagnostic approaches and therapeutic regimens for osteotropic malignant diseases.
Collapse
Affiliation(s)
- Thomas E Kruger
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Andrew H Miller
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; University of Kansas Cancer Center, Kansas City, KS 66160, USA
| | - Jinxi Wang
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, KS 66160, USA; Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
30
|
Pesesse L, Sanchez C, Delcour JP, Bellahcène A, Baudouin C, Msika P, Henrotin Y. Consequences of chondrocyte hypertrophy on osteoarthritic cartilage: potential effect on angiogenesis. Osteoarthritis Cartilage 2013; 21:1913-23. [PMID: 23973427 DOI: 10.1016/j.joca.2013.08.018] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 08/09/2013] [Accepted: 08/14/2013] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The aim of this study was to investigate the link between the hypertrophic phenotype of chondrocytes and angiogenesis in osteoarthritis (OA) and more particularly to demonstrate that OA hypertrophic chondrocytes potentially express a phenotype promoting angiogenesis through the expression of factors controlling endothelial cells migration, invasion and adhesion. METHOD Human OA chondrocytes were cultivated in alginate beads in medium supplemented with 10% fetal bovine serum (FBS) to induce chondrocyte hypertrophy. The hypertrophic phenotype was characterized throughout 28 days of culture by measuring the expression of specific genes and by a microscopic observation of cellular morphology. The effect of media conditioned by OA hypertrophic chondrocyte on endothelial cells migration, invasion and adhesion was evaluated in functional assays. Moreover, hypertrophic OA chondrocytes were tested for the expression of angiogenic factors by real-time RT-PCR. RESULTS Specific markers of hypertrophy and observation of cellular morphology attested of the hypertrophic phenotype of chondrocytes in our culture model. Functional angiogenesis assays showed that factors produced by hypertrophic chondrocytes stimulated migration, invasion and adhesion of endothelial cells. Among the evaluated angiogenic factors, bone sialoprotein (BSP) was the most highly upregulated in hypertrophic chondrocytes. The inhibition of endothelial cell adhesion by a GRGDS peptide confirmed the implication of RGD domain proteins, like BSP, in hypertrophic chondrocyte-induced adhesion of endothelial cells. CONCLUSION Hypertrophic differentiation of chondrocyte may promote angiogenesis. Our findings established the relation of BSP with OA chondrocyte hypertrophy and suggested that this factor could constitute a potential target to control cartilage neovascularisation in OA.
Collapse
Affiliation(s)
- L Pesesse
- Bone and Cartilage Research Unit, University of Liège, Liège, Belgium.
| | | | | | | | | | | | | |
Collapse
|
31
|
Lamour V, Nokin MJ, Henry A, Castronovo V, Bellahcène A. [SIBLING proteins: molecular tools for tumor progression and angiogenesis]. Med Sci (Paris) 2013; 29:1018-25. [PMID: 24280506 DOI: 10.1051/medsci/20132911019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The small integrin-binding ligand N-linked glycoprotein (SIBLING) family consists of osteopontin (OPN), bonesialoprotein (BSP), dentin matrix protein 1 (DMP1), dentin sialophosphoprotein (DSPP) and matrix extracellular phosphoglycoprotein (MEPE). These proteins, initially identified in bone and teeth, share many structural characteristics. It is now well established that they are over expressed in many tumors and play a critical role at different steps of cancer development. In this review, we describe the roles of SIBLING proteins at different stages of cancer progression including cancer cell adhesion, proliferation, migration, invasion, metastasis and angiogenesis.
Collapse
Affiliation(s)
- Virginie Lamour
- Laboratoire de recherche sur les métastases, GIGA (groupe interdisciplinaire de génoprotéomique appliquée)-Cancer, Université de Liège, Building 23, Sart Tilman, 4000 Liège, Belgique
| | | | | | | | | |
Collapse
|
32
|
Xu T, Qin R, Zhou J, Yan Y, Lu Y, Zhang X, Fu D, Lv Z, Li W, Xia C, Hu G, Ding X, Chen J. High bone sialoprotein (BSP) expression correlates with increased tumor grade and predicts a poorer prognosis of high-grade glioma patients. PLoS One 2012; 7:e48415. [PMID: 23119009 PMCID: PMC3485236 DOI: 10.1371/journal.pone.0048415] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Accepted: 09/25/2012] [Indexed: 01/02/2023] Open
Abstract
Objectives To investigate the expression and prognostic value of bone sialoprotein (BSP) in glioma patients. Methods We determined the expression of BSP using real-time RT-PCR and immunohistochemistry in tissue microarrays containing 15 normal brain and 270 glioma samples. Cumulative survival was calculated by the Kaplan-Meier method and analyzed by the log-rank test. Univariate and multivariate analyses were performed by the stepwise forward Cox regression model. Results Both BSP mRNA and protein levels were significantly elevated in high-grade glioma tissues compared with those of normal brain and low-grade glioma tissues, and BSP expression positively correlated with tumor grade (P<0.001). Univariate and multivariate analysis showed high BSP expression was an independent prognostic factor for a shorter progression-free survival (PFS) and overall survival (OS) in both grade III and grade IV glioma patients [hazard ratio (HR) = 2.549 and 3.154 for grade III glioma, and HR = 1.637 and 1.574 for grade IV glioma, respectively]. Patients with low BSP expression had a significantly longer median OS and PFS than those with high BSP expression. Small extent of resection and lineage of astrocyte served as independent risk factors of both shorter PFS and OS in grade III glioma patients; GBM patients without O6-methylguanine (O6-meG) DNA methyltransferase (MGMT) methylation and Karnofsky performance score (KPS) less than 70 points were related to poor prognosis. Lack of radiotherapy related to shorter OS but not affect PFS in both grade III and grade IV glioma patients. Conclusion High BSP expression occurs in a significant subset of high-grade glioma patients and predicts a poorer outcome. The study identifies a potentially useful molecular marker for the categorization and targeted therapy of gliomas.
Collapse
Affiliation(s)
- Tao Xu
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Rong Qin
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jinxu Zhou
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yong Yan
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yicheng Lu
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xiaoping Zhang
- Department of Nuclear Medicine, Shanghai 10th People’s Hospital, Tongji University School of Medicine, Shanghai, China
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Da Fu
- Department of Nuclear Medicine, Shanghai 10th People’s Hospital, Tongji University School of Medicine, Shanghai, China
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Zhongwei Lv
- Department of Nuclear Medicine, Shanghai 10th People’s Hospital, Tongji University School of Medicine, Shanghai, China
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Weiqing Li
- Department of Pathology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Chunyan Xia
- Department of Pathology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Guohan Hu
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xuehua Ding
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Juxiang Chen
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
- * E-mail:
| |
Collapse
|
33
|
Wohlrab S, Müller S, Schmidt A, Neubauer S, Kessler H, Leal-Egaña A, Scheibel T. Cell adhesion and proliferation on RGD-modified recombinant spider silk proteins. Biomaterials 2012; 33:6650-9. [DOI: 10.1016/j.biomaterials.2012.05.069] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 05/30/2012] [Indexed: 10/28/2022]
|
34
|
Mosig RA, Martignetti JA. Loss of MMP-2 in murine osteoblasts upregulates osteopontin and bone sialoprotein expression in a circuit regulating bone homeostasis. Dis Model Mech 2012; 6:397-403. [PMID: 22917927 PMCID: PMC3597021 DOI: 10.1242/dmm.007914] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Multicentric osteolysis with arthropathy (MOA; MIM 605156) is an inherited osteolyses and arthritis syndrome resulting from loss of matrix metalloproteinase 2 (MMP-2). We recently demonstrated that Mmp2–/– mice represent a unique model for the study of the human disease, sharing many features of the human syndrome including skeletal dysplasia and defects in osteoblast behavior. We therefore sought to explore the secondary molecular effects of MMP-2 loss, which coexist with the underlying skeletal and osteoblast phenotypes. We used quantitative real-time RT-PCR (qRT-PCR) to measure osteoblast-related gene expression through ex vivo osteoblast differentiation of bone marrow stromal cells (BMSC) from Mmp2−/− and Mmp2+/+ mice. We used western blot to measure osteopontin (OPN) serum levels and immunohistochemical staining to examine bone expression. MMP-2 expression was inhibited in SaOS2 cells using siRNA, and decreased MMP-2 expression at both RNA and protein levels was confirmed by qRT-PCR and western blot, respectively. Mmp2−/− BMSC induced to differentiate into osteoblasts were shown to significantly upregulate OPN and bone sialoprotein (BSP) expression levels compared with controls. Transcriptional upregulation was maintained in vivo, as demonstrated by increased levels of OPN in serum and bone in Mmp2−/− mice. These effects are generalizable because siRNA-mediated inhibition in cultured cells also upregulated OPN and BSP. OPN and BSP are known to affect MMP-2 expression and activity but have not previously been shown to be regulated by MMP-2. Identification of this newly defined circuitry provides insight into the potential molecular landscape underlying the MOA phenotype and highlights a pathway that might play a role in normal bone homeostasis.
Collapse
Affiliation(s)
- Rebecca A Mosig
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | |
Collapse
|
35
|
Mittag F, Hennenlotter J, Minkley L, Ipach I, Schilling D, Scharpf M, Stenzl A, Leichtle U, Kluba T. Alteration of bone sialoprotein expression in osseous metastasized renal cell carcinomas and the tumor surrounding tissue. Clin Exp Metastasis 2011; 29:179-83. [PMID: 22130963 DOI: 10.1007/s10585-011-9441-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Accepted: 11/24/2011] [Indexed: 11/25/2022]
Abstract
Bone sialoprotein (BSP) regulates bone metabolism by directly influencing the activity of osteoblasts and osteoclasts. A significant correlation between the tissue expression of BSP in tumors and the occurrence of bone metastases was found in different cancers. Aim of this study was to identify the BSP expression in renal cell carcinomas (RCC) according to their stage of metastatic disease. Tissue samples of patients with RCC who underwent partial resection or nephrectomy were separated into three groups, each with 10 patients showing either no metastases (group I), only soft tissue metastases (group II) or bone metastases (group III) at date of surgery. Immunohistochemical analysis of BSP expression in tumor tissue and corresponding renal parenchyma was performed and evaluated with an established semiquantitative scoring system. BSP expression was detected both in tumor tissue and renal parenchyma. Concerning the expression in malignant tissue, no significant difference could be found between the three groups whereas the corresponding renal parenchyma showed a staining score of 164, 198 and 224 for group I, II and III (P = 0.07). RCC staged T3 showed only a little higher BSP expression than those staged T1/2 (P < 0.21), while the corresponding parenchyma of T3 tumors showed significantly higher expressions (P = 0.02). This pilot study revealed a correlation between expression of BSP and tumor staging and type of metastases, especially for osseous metastases in RCC. Alternation of BSP expression could be detected particularly in renal parenchyma and linked to the type of metastases.
Collapse
Affiliation(s)
- F Mittag
- Department of Orthopedics, Eberhard-Karls-University, Tuebingen, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
The metastasis is the spread of cancer from one part of the body to another. Two-thirds of patients with cancer will develop bone metastasis. Breast, prostate and lung cancer are responsible for more than 80% of cases of metastatic bone disease. The spine is the most common site of bone metastasis. A spinal metastasis may cause pain, instability and neurological injuries. The diffusion through Batson venous system is the principal process of spinal metastasis, but the dissemination is possible also through arterial and lymphatic system or by contiguity. Once cancer cells have invaded the bone, they produce growth factors that stimulate osteoblastic or osteolytic activity resulting in bone remodeling with release of other growth factors that lead to a vicious cycle of bone destruction and growth of local tumour.
Collapse
|
37
|
Bidirectional interactions between bone metabolism and hematopoiesis. Exp Hematol 2011; 39:809-16. [PMID: 21609752 DOI: 10.1016/j.exphem.2011.04.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 04/19/2011] [Accepted: 04/30/2011] [Indexed: 01/04/2023]
Abstract
Interactions between hematopoiesis and bone metabolism have been described in various developmental and pathological situations. Here we review this evidence from the literature with a focus on microenvironmental regulation of hematopoiesis and bone metabolism. Our hypothesis is that this process occurs by bidirectional signaling between hematopoietic and mesenchymal cells through cell adhesion molecules, membrane-bound growth factors, and secreted matrix proteins. Examples of steady-state hematopoiesis and pathologies are presented and support our view that hematopoietic and mesenchymal cell functions are modulated by specific microenvironments in the bone marrow.
Collapse
|
38
|
Dentin matrix protein 1 induces membrane expression of VE-cadherin on endothelial cells and inhibits VEGF-induced angiogenesis by blocking VEGFR-2 phosphorylation. Blood 2010; 117:2515-26. [PMID: 21190990 DOI: 10.1182/blood-2010-08-298810] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Dentin matrix protein 1 (DMP1) is a member of the small integrin-binding ligand N-linked glycoprotein (SIBLING) family, a group of proteins initially described as mineralized extracellular matrices components. More recently, SIBLINGs have been implicated in several key steps of cancer progression, including angiogenesis. Although proangiogenic activities have been demonstrated for 2 SIBLINGs, the role of DMP1 in angiogenesis has not yet been addressed. We demonstrate that this extracellular matrix protein induced the expression of vascular endothelial cadherin (VE-cadherin), a key regulator of intercellular junctions and contact inhibition of growth of endothelial cells that is also known to modulate vascular endothelial growth factor receptor 2 (VEGFR-2) activity, the major high-affinity receptor for VEGF. DMP1 induced VE-cadherin and p27(Kip1) expression followed by cell-cycle arrest in human umbilical vein endothelial cells (HUVECs) in a CD44-dependent manner. VEGF-induced proliferation, migration, and tubulogenesis responses were specifically blocked on DMP1 pretreatment of HUVECs. Indeed, after VE-cadherin induction, DMP1 inhibited VEGFR-2 phosphorylation and Src-mediated signaling. However, DMP1 did not interfere with basic fibroblast growth factor-induced angiogenesis. In vivo, DMP1 significantly reduced laser-induced choroidal neovascularization lesions and tumor-associated angiogenesis. These data enable us to put DMP1 on the angiogenic chessboard for the first time and to identify this protein as a new specific inhibitor of VEGF-induced angiogenesis.
Collapse
|
39
|
Angiogenic activity of an enamel matrix derivative (EMD) and EMD-derived proteins: an experimental study in mice. J Clin Periodontol 2010; 38:253-60. [DOI: 10.1111/j.1600-051x.2010.01656.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
40
|
Wall ST, Yeh CC, Tu RYK, Mann MJ, Healy KE. Biomimetic matrices for myocardial stabilization and stem cell transplantation. J Biomed Mater Res A 2010; 95:1055-66. [PMID: 20878934 DOI: 10.1002/jbm.a.32904] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Revised: 02/09/2010] [Accepted: 02/09/2010] [Indexed: 01/07/2023]
Abstract
Although natural biological matrices have demonstrated modest improvement in the survival of cells transplanted into the infarcted myocardium, these materials have not been amenable to systematic optimization and therefore have limited potential to treat postinfarct cardiac injuries. Here we have developed tunable bioactive semi-interpenetrating polymer network (sIPN) hydrogels with matrix metalloproteinase (MMP) labile crosslinkers to be used as an assistive microenvironment for transplantation of bone marrow-derived mesenchymal stem cells (BMSCs) into the infarcted myocardium. Injectable sIPN hydrogels were designed with a range of mechanical and biological properties that yielded material-dependent BMSC proliferation in vitro. Five groups were evaluated to treat myocardial infarction (MI) in adult mice: saline injection; green fluorescent protein (GFP)(+)-BMSCs delivered in saline; a sIPN matrix; a sIPN + GFP(+)-BMSCs; and Matrigel™ + GFP(+)-BMSCs. Injection of cells alone created a transient improvement in LV function that declined over time, and the synthetic hydrogel without cells resulted in the highest LV function at 6 weeks. Donor GFP-positive cells were detected after matrix-enhanced transplantation, but not without matrix support. Biomimetic sIPN hydrogel matrices succeeded both in mechanically supporting the injured myocardium and modestly enhancing donor cell survival. These matrices provide a foundation for systematic development of "pro-survival" microenvironments, and improvement in the long-term results of cardiac stem cell transplantation therapies.
Collapse
Affiliation(s)
- Samuel T Wall
- Department of Bioengineering, University of California at Berkeley, Berkeley, California, USA
| | | | | | | | | |
Collapse
|
41
|
Pesesse L, Sanchez C, Henrotin Y. Osteochondral plate angiogenesis: a new treatment target in osteoarthritis. Joint Bone Spine 2010; 78:144-9. [PMID: 20851653 DOI: 10.1016/j.jbspin.2010.07.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2010] [Indexed: 01/05/2023]
Abstract
Healthy adult joint cartilage contains neither blood vessels nor nerves. Osteoarthritic cartilage, in contrast, may be invaded by blood vessels from the subchondral bone. The mechanisms underlying cartilage angiogenesis in osteoarthritis are unclear but may involve hypertrophic chondrocyte differentiation. Active research is under way to identify the factors involved in cartilage angiogenesis. Here, we discuss the pathophysiological mechanisms of osteoarthritic cartilage angiogenesis based on evidence from a systematic literature review of articles retrieved via PubMed and ISI Web of Knowledge. Our conclusions suggest new research perspectives and treatment options.
Collapse
Affiliation(s)
- Laurence Pesesse
- Unité de Recherche Sur l'Os et le Cartilage, Institut de Pathologie, Université de Liège, CHU Sart-Tilman, 4000 Liège, Belgium
| | | | | |
Collapse
|
42
|
Su J, Wall ST, Healy KE, Wildsoet CF. Scleral reinforcement through host tissue integration with biomimetic enzymatically degradable semi-interpenetrating polymer network. Tissue Eng Part A 2010; 16:905-16. [PMID: 19814587 DOI: 10.1089/ten.tea.2009.0488] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Enzymatically degradable semi-interpenetrating polymer networks (edsIPNs) were explored for their biocompatibility and ability to promote new scleral tissue growth, as a means of reinforcing the posterior wall of the eye. The edsIPNs comprised thermoresponsive poly(N-isopropylacrylamide-co-acrylic acid), customizable peptide crosslinkers cleavable by matrix metalloproteinases, and interpenetrating linear poly(acrylic acid)-graft-peptide chains to engage with cell surface receptors. Rheological studies revealed an increase in stiffness at body temperature; the complex shear modulus |G*| was 14.13 +/- 6.13 Pa at 22 degrees C and 63.18 +/- 12.24 Pa at 37 degrees C, compatible with injection at room temperature. Primary chick scleral fibroblasts and chondrocytes cultured on edsIPN increased by 15.1- and 11.1-fold, respectively, over 11 days; both exhibited delayed onset of exponential growth compared with the cells plated on tissue culture polystyrene. The edsIPN was delivered by retrobulbar injection (100 microL) to nine 2-week-old chicks to assess biocompatibility in vivo. Ocular axial dimensions were assessed using A-scan ultrasonography over 28 days, after which eyes were processed for histological analysis. Although edsIPN injections did not affect the rate of ocular elongation, the outer fibrous sclera showed significant thickening. The demonstration that injectable biomimetic edsIPNs stimulate scleral fibrous tissue growth represents proof-of-principle for a novel approach for scleral reinforcement and a potential therapy for high myopia.
Collapse
Affiliation(s)
- James Su
- Vision Science Group, School of Optometry, University of California, Berkeley, CA 94720-2020, USA
| | | | | | | |
Collapse
|
43
|
Neville-Webbe HL, Coleman RE. Bisphosphonates and RANK ligand inhibitors for the treatment and prevention of metastatic bone disease. Eur J Cancer 2010; 46:1211-22. [PMID: 20347292 DOI: 10.1016/j.ejca.2010.02.041] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Accepted: 02/23/2010] [Indexed: 11/19/2022]
Abstract
Bisphosphonates (BPs) are potent inhibitors of osteoclast-mediated bone resorption, which is increased when cancer cells invade bone. BPs are an established treatment for cancer that has spread to bone, and effectively reduce pain and other skeletal-related events. New directions in metastatic bone disease (MBD) include personalised BP therapy, such as using bone markers to guide frequency of BP administration and bone targeting agents such as denosumab (AMG 162). Clinical trials strongly suggest that denosumab might play a defined role in the future management of MBD. In terms of potential anti-cancer activity, early data tentatively suggest that zoledronic acid might have a role to play in the prevention of metastatic disease, though whether this is a direct effect on cancer cells, or indirect via the bone marrow micro-environment, or both, is as yet undiscovered. The definitive answer as to the role of adjuvant BP in early cancer is being addressed, with over 20,000 patients with breast, prostate or lung cancer currently participating in adjuvant BP randomised trials. The results of these trials should be available in the next few years, and this will establish whether BPs given early in the course of cancer will be able to prevent the formation of metastases, bone or otherwise.
Collapse
Affiliation(s)
- H L Neville-Webbe
- Academic Department of Clinical Oncology, Weston Park Hospital, Sheffield S10 2SJ, UK.
| | | |
Collapse
|
44
|
Organic/Inorganic bioactive materials Part II: in vitro bioactivity of Collagen-Calcium Phosphate Silicate/Wollastonite hybrids. OPEN CHEM 2009. [DOI: 10.2478/s11532-009-0076-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AbstractIn the present study, novel hybrid materials of Collagen (C) and Calcium Phosphate Silicate/Wollastonite (CPS/W) were synthesized. The CPS/W ceramic was prepared via polystep sol-gel method. The dissolution test of CPS/W ceramic was filled with TRIS-HCl buffer. FTIR depicts that hydroxyl carbonate apatite (OHCO3HA) was observed after 3 days of immersion in TRIS-HCl buffer. Biohybrids of C-CPS/W were produced from diluted hydrochloric acid collagen type I and ceramic powder with different ratios of C and CPS/W equal to 25:75 and 75:25 wt.%. The synthesized hybrids were characterized by FTIR, XRD and SEM. FTIR depicts a “red shift” if amide I could be attributed to the fact that the collagen prefers to chelate Ca2+ from partial dissolution of CPS/W ceramic. The growth of B-type carbonate containing hydroxyapatite (B-CO3HA) on the C-CPS/W hybrids soaked in 1.5SBF was observed. The negatively charged carboxylate groups from the collagen may be responsible for hydroxyapatite (HA) deposition. This fact was confirmed by the “red shift” of carboxylate groups of collagen in FTIR spectra. The formation of HA was observed by FTIR, XRD and SEM.
Collapse
|
45
|
Malaval L, Monfoulet L, Fabre T, Pothuaud L, Bareille R, Miraux S, Thiaudiere E, Raffard G, Franconi JM, Lafage-Proust MH, Aubin JE, Vico L, Amédée J. Absence of bone sialoprotein (BSP) impairs cortical defect repair in mouse long bone. Bone 2009; 45:853-61. [PMID: 19524706 DOI: 10.1016/j.bone.2009.06.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2008] [Revised: 05/13/2009] [Accepted: 06/02/2009] [Indexed: 01/25/2023]
Abstract
Matrix proteins of the SIBLING family interact with bone cells and with bone mineral and are thus in a key position to regulate bone development, remodeling and repair. Within this family, bone sialoprotein (BSP) is highly expressed by osteoblasts, hypertrophic chondrocytes and osteoclasts. We recently reported that mice lacking BSP (BSP-/-) have very low trabecular bone turnover. In the present study, we set up an experimental model of bone repair by drilling a 1 mm diameter hole in the cortical bone of femurs in both BSP-/- and +/+ mice. A non-invasive MRI imaging and bone quantification procedure was designed to follow bone regeneration, and these data were extended by microCT imaging and histomorphometry on undecalcified sections for analysis at cellular level. These combined approaches revealed that the repair process as reflected in defect-refilling in the cortical area was significantly delayed in BSP-/- mice compared to +/+ mice. Concomitantly, histomorphometry showed that formation, mineralization and remodeling of repair (primary) bone in the medulla were delayed in BSP-/- mice, with lower osteoid and osteoclast surfaces at day 15. In conclusion, the absence of BSP delays bone repair at least in part by impairing both new bone formation and osteoclast activity.
Collapse
Affiliation(s)
- Luc Malaval
- Université de Lyon, Saint-Etienne, F42023, France.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Bellahcène A, Chaplet M, Bonjean K, Castronovo V. Zoledronate Inhibits αvβ3 and αvβ5 Integrin Cell Surface Expression in Endothelial Cells. ACTA ACUST UNITED AC 2009; 14:123-30. [PMID: 17497369 DOI: 10.1080/10623320701347187] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Zoledronate exhibits antiangiogenic properties in vitro and in vivo. Integrins alphavbeta3 and alphavbeta5 are involved in angiogenesis. Because zoledronate inhibits endothelial cell adhesion, the authors explored the hypothesis that it could alter these integrins recruitment to focal adhesion sites. Human umbilical vein endothelial cells (HUVECs) were treated with zoledronate or with mevalonate pathway intermediates geranylgeraniol (GGOH) and farnesol (FOH). Zoledronate generated a significant decrease in alphavbeta3 and alphavbeta5 expression at HUVEC cell surface using flow cytometry and immunofluorescence. This inhibition was reversed by GGOH but not by FOH. Cells cotreated with zoledronate and GGOH were able to attach to vitronectin through alphavbeta3 and alphavbeta5, as confirmed by the use of specific function-blocking antibodies. The authors showed that zoledronate alters endothelial cell integrin-mediated adhesion. This effect is likely to contribute to the previously demonstrated antiangiogenic effect of zoledronate. Whether this mechanism of action also applies to metastatic tumor cells is under investigation.
Collapse
Affiliation(s)
- A Bellahcène
- Metastasis Research Laboratory, Center of Experimental Cancer Research, University of Liège, 4000 Liège, Belgium.
| | | | | | | |
Collapse
|
47
|
Wohl GR, Towler DA, Silva MJ. Stress fracture healing: fatigue loading of the rat ulna induces upregulation in expression of osteogenic and angiogenic genes that mimic the intramembranous portion of fracture repair. Bone 2009; 44:320-30. [PMID: 18950737 PMCID: PMC2759644 DOI: 10.1016/j.bone.2008.09.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Revised: 08/29/2008] [Accepted: 09/08/2008] [Indexed: 11/21/2022]
Abstract
Woven bone is formed in response to fatigue-induced stress fractures and is associated with increased local angiogenesis. The molecular mechanisms that regulate this woven bone formation are unknown. Our objective was to measure the temporal and spatial expression of osteo- and angiogenic genes in woven bone formation in response to increasing levels of fatigue-induced damage. We used the rat forelimb compression model to produce four discrete levels of fatigue damage in the right ulna of 115 male Fischer rats. Rats were killed at 0 (1 h), 1, 3 and 7 days after loading. Using qRT-PCR, we quantified gene expression associated with osteogenesis (BMP2, Msx2, Runx2, Osx, BSP, Osc), cell proliferation (Hist4), and angiogenesis (VEGF, PECAM-1) from the central half of the ulna. The spatial distribution of BMP2, BSP and PCNA was assessed by immunohistochemistry or in situ hybridization in transverse histological sections 1, 4, and 7 mm distal to the ulnar mid-diaphysis. One hour after loading, BMP2 was significantly upregulated in neurovascular structures in the medial ulnar periosteum. Expression of angiogenic markers (VEGF, PECAM-1) increased significantly between Day 0 and 1 and, as with BMP2 expression, remained upregulated through Day 7. While Osx and BSP were upregulated on Day 1, the other osteogenic genes (Msx2, Runx2, Osx, BSP and Osc) were induced on Day 3 in association with the initiation of periosteal woven bone formation and continued through Day 7. The magnitude of osteogenic gene expression, particularly matrix genes (BSP, Osc) was significantly proportional the level of fatigue damage. The woven bone response to fatigue injury is remarkably similar to the "intramembranous" portion of fracture repair - rapid formation of periosteal woven bone characterized by early BMP2 expression, cell proliferation, and upregulation of osteogenic genes. We speculate that woven bone repair of fatigue damage may be an abbreviated fracture response without the requirement for endochondral repair. We conclude that bone fatigue repair is a process similar to intramembranous fracture repair characterized by increases in the expression of genes associated with angiogenesis, cell proliferation and osteoblastogenesis, and that the response from the local vasculature precedes the osteogenic response to fatigue loading.
Collapse
Affiliation(s)
- Gregory R Wohl
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, USA.
| | | | | |
Collapse
|
48
|
Tu Q, Zhang J, Fix A, Brewer E, Li YP, Zhang ZY, Chen J. Targeted overexpression of BSP in osteoclasts promotes bone metastasis of breast cancer cells. J Cell Physiol 2008; 218:135-45. [PMID: 18756497 DOI: 10.1002/jcp.21576] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Bone is one of the most common sites of breast cancer metastasis while bone sialoprotein (BSP) is thought to play an important role in bone metastasis of malignant tumors. The objective of this study is to determine the role of BSP overexpression in osteolytic metastasis using two homozygous transgenic mouse lines in which BSP expression is elevated either in all the tissues (CMV-BSP mice) or only in the osteoclasts (CtpsK-BSP mice). The results showed that skeletal as well as systemic metastases of 4T1 murine breast cancer cells were dramatically increased in CMV-BSP mice. In CtpsK-BSP mice, it was found that targeted BSP overexpression in osteoclasts promoted in vitro osteoclastogenesis and activated osteoclastic differentiation markers such as Cathepsin K, TRAP and NFAT2. MicroCT scan demonstrated that CtpsK/BSP mice had reduced trabecular bone volume and bone mineral density (BMD). The real-time IVIS Imaging System showed that targeted BSP overexpression in osteoclasts promoted bone metastasis of breast cancer cells. The osteolytic lesion area was significantly larger in CtpsK/BSP mice than in the controls as demonstrated by both radiographic and histomorphometric analyses. TRAP staining demonstrated a twofold increase in the number of osteoclasts in the bone lesion area from CtpsK/BSP mice compared with that from wild type mice. We conclude that host tissue-derived BSP also plays important roles in breast cancer metastasis through inducing tumor cell seeding into the remote host tissues. Furthermore, osteoclast-derived BSP promotes osteoclast differentiation in an autocrine manner and consequently promotes osteolytic bone metastasis of breast cancer.
Collapse
Affiliation(s)
- Qisheng Tu
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, Massachusetts, USA.
| | | | | | | | | | | | | |
Collapse
|
49
|
Pohjolainen V, Taskinen P, Soini Y, Rysä J, Ilves M, Juvonen T, Ruskoaho H, Leskinen H, Satta J. Noncollagenous bone matrix proteins as a part of calcific aortic valve disease regulation. Hum Pathol 2008; 39:1695-701. [PMID: 18701137 DOI: 10.1016/j.humpath.2008.04.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Revised: 04/01/2008] [Accepted: 04/01/2008] [Indexed: 10/21/2022]
Abstract
Clinically, calcific aortic valve disease is a progressive continuum from obstructive fibro(sclero)tic valve thickening to aortic stenosis. Recent evidence suggests that, in addition to nonbone miscellaneous mineralization, calcified valves present distinct signs of active bone remodeling; and in this context, noncollagenous bone-associated proteins are assumed to have a critical role. The expression of 5 bone matrix proteins-bone morphogenetic protein-2 and -4, bone sialoprotein II, osteopontin, and osteoprotegerin-was examined by reverse transcriptase polymerase chain reaction (n = 31) and immunolabeling (n = 83) in the clinical continuum from healthy pliable valves to heavily calcified ones. As a known structural pathologic sign, the extent of neovascularization was also examined. We observed progressive increase in the gene expression of osteopontin (7.4-fold elevation, P < .001) and bone sialoprotein II (5.8-fold elevation, P < .05), and also 1.7-fold elevation (P < .05) in osteoprotegerin gene expression during the disease course. These findings were congruent with that of immunohistochemical analysis. Surprisingly, bone morphogenetic protein-2 and -4 showed a comparable significant decrease in messenger RNA levels in calcified valves (P < .01 and P < .05, respectively). Our results support the view that aortic valve calcification is an actively regulated process. Furthermore, the results suggest that the expression of pro- and anticalcific noncollagenous bone-associated matrix proteins is altered during the disease continuum and that this imbalance may contribute to the pathology of calcific aortic valve disease.
Collapse
Affiliation(s)
- Virva Pohjolainen
- Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Jain A, Fisher LW, Fedarko NS. Bone sialoprotein binding to matrix metalloproteinase-2 alters enzyme inhibition kinetics. Biochemistry 2008; 47:5986-95. [PMID: 18465841 DOI: 10.1021/bi800133n] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Bone sialoprotein (BSP) is a secreted glycophosphoprotein normally restricted in expression to skeletal tissue that is also induced by multiple neoplasms in vivo. Previous work has shown that BSP can bind to matrix metalloproteinase-2 (MMP-2). Because of MMP-2 activity in promoting tumor progression, potential therapeutic inhibitors were developed, but clinical trials have been disappointing. The effect of BSP on MMP-2 modulation by inhibitors was determined with purified components and in cell culture. Enzyme inhibition kinetics were studied using a low-molecular weight freely diffusable substrate and purified MMP-2, BSP, and natural (tissue inhibitor of matrix metalloproteinase-2) and synthetic (ilomastat and oleoyl- N-hydroxylamide) inhibitors. We determined parameters of enzyme kinetics by varying substrate concentrations at different fixed inhibitor concentrations added to MMP-2 alone, MMP-2 and BSP, or preformed MMP-2-BSP complexes and solving a general linear mixed inhibition rate equation with a global curve fitting program. Two in vitro angiogenesis model systems employing human umbilical vein endothelial cells (HUVECs) were used to follow BSP modulation of MMP-2 inhibition and tubule formation. The presence of BSP increased the competitive K I values between 15- and 47-fold for natural and synthetic inhibitors. The extent of tubule formation by HUVECs cocultured with dermal fibroblasts was reduced in the presence of inhibitors, while the addition of BSP restored vessel formation. A second HUVEC culture system demonstrated that tubule formation by cells expressing BSP could be inhibited by an activity blocking antibody against MMP-2. BSP modulation of MMP-2 activity and inhibition may define its biological role in promoting tumor progression.
Collapse
Affiliation(s)
- Alka Jain
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21224, USA
| | | | | |
Collapse
|