1
|
Rodríguez-Sosa MR, Del Castillo LM, Belarra A, Zapata AG, Alfaro D. The lack of EphB3 receptor prevents bone loss in mouse models of osteoporosis. J Bone Miner Res 2024; 39:1008-1024. [PMID: 38739682 DOI: 10.1093/jbmr/zjae075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 03/21/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
Bone homeostasis is a complex process in which some Eph kinase receptors and their ephrin ligands appear to be involved. In the present study, we address this issue by examining, both in vitro and in vivo, the role of EphB2 and EphB3 in mesenchymal stromal/stem cell (MSC) differentiation into bone tissue. This was first evaluated by quantitative reverse transcription PCR (RT-qPCR) and histological staining in MSCs cultured in specific mediums revealing that although EphB2-/- MSCs mainly expressed pro-adipogenic transcription factors, EphB3-/- MSCs showed abundant osteogenic transcripts, such as Runx2, Msx2, and Sp7. To clarify the underlying molecular mechanisms, we found that the lack of EphB3 signaling alters the genetic profile of differentiating MSCs, reducing the expression of many inhibitory molecules and antagonists of the BMP signaling pathway, and increasing Bmp7 expression, a robust bone inductor. Then, to confirm the osteogenic role of EphB3 in vivo, we studied the condition of 2 mouse models of induced bone loss (ovariectomy or long-term glucocorticoid treatment). Interestingly, in both models, both WT and EphB2-/- mice equally developed the disease but EphB3-/- mice did not exhibit the typical bone loss, nor an increase in urine Ca2+ or blood serum CTX-1. This phenotype in EphB3-KO mice could be due to their significantly higher proportions of osteoprogenitor cells and preosteoblasts, and their lower number of osteoclasts, as compared with WT and EphB2-KO mice. Thus, we conclude that EphB3 acts as a negative regulator of the osteogenic differentiation, and its absence prevents bone loss in mice subjected to ovariectomy or dexamethasone treatment.
Collapse
Affiliation(s)
- Mariano R Rodríguez-Sosa
- Department of Cell Biology, Faculty of Biological Sciences, Complutense University of Madrid, C.P. 28040, Madrid, Spain
- Research Institute Hospital "12 de Octubre" (imas12), C.P. 28041, Madrid, Spain
| | - Luis M Del Castillo
- Reproductive Medicine Research Group, IVI Foundation, Health Research Institute Hospital La Fe (IIS La Fe), C.P. 46026, Valencia, Spain
| | - Adrián Belarra
- Micro-CT Laboratory, Central Radioactive Facility, Department of Radiology, Rehabilitation and Physiotherapy, Faculty of Medicine, Complutense University of Madrid, C.P. 28040, Madrid, Spain
| | - Agustín G Zapata
- Department of Cell Biology, Faculty of Biological Sciences, Complutense University of Madrid, C.P. 28040, Madrid, Spain
| | - David Alfaro
- Department of Cell Biology, Faculty of Biological Sciences, Complutense University of Madrid, C.P. 28040, Madrid, Spain
| |
Collapse
|
2
|
Volova LT, Kotelnikov GP, Shishkovsky I, Volov DB, Ossina N, Ryabov NA, Komyagin AV, Kim YH, Alekseev DG. 3D Bioprinting of Hyaline Articular Cartilage: Biopolymers, Hydrogels, and Bioinks. Polymers (Basel) 2023; 15:2695. [PMID: 37376340 DOI: 10.3390/polym15122695] [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: 04/06/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
The musculoskeletal system, consisting of bones and cartilage of various types, muscles, ligaments, and tendons, is the basis of the human body. However, many pathological conditions caused by aging, lifestyle, disease, or trauma can damage its elements and lead to severe disfunction and significant worsening in the quality of life. Due to its structure and function, articular (hyaline) cartilage is the most susceptible to damage. Articular cartilage is a non-vascular tissue with constrained self-regeneration capabilities. Additionally, treatment methods, which have proven efficacy in stopping its degradation and promoting regeneration, still do not exist. Conservative treatment and physical therapy only relieve the symptoms associated with cartilage destruction, and traditional surgical interventions to repair defects or endoprosthetics are not without serious drawbacks. Thus, articular cartilage damage remains an urgent and actual problem requiring the development of new treatment approaches. The emergence of biofabrication technologies, including three-dimensional (3D) bioprinting, at the end of the 20th century, allowed reconstructive interventions to get a second wind. Three-dimensional bioprinting creates volume constraints that mimic the structure and function of natural tissue due to the combinations of biomaterials, living cells, and signal molecules to create. In our case-hyaline cartilage. Several approaches to articular cartilage biofabrication have been developed to date, including the promising technology of 3D bioprinting. This review represents the main achievements of such research direction and describes the technological processes and the necessary biomaterials, cell cultures, and signal molecules. Special attention is given to the basic materials for 3D bioprinting-hydrogels and bioinks, as well as the biopolymers underlying the indicated products.
Collapse
Affiliation(s)
- Larisa T Volova
- Research and Development Institute of Biotechnologies, Samara State Medical University, Chapayevskaya St. 89, 443099 Samara, Russia
| | - Gennadiy P Kotelnikov
- Research and Development Institute of Biotechnologies, Samara State Medical University, Chapayevskaya St. 89, 443099 Samara, Russia
| | - Igor Shishkovsky
- Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Dmitriy B Volov
- Research and Development Institute of Biotechnologies, Samara State Medical University, Chapayevskaya St. 89, 443099 Samara, Russia
| | - Natalya Ossina
- Research and Development Institute of Biotechnologies, Samara State Medical University, Chapayevskaya St. 89, 443099 Samara, Russia
| | - Nikolay A Ryabov
- Research and Development Institute of Biotechnologies, Samara State Medical University, Chapayevskaya St. 89, 443099 Samara, Russia
| | - Aleksey V Komyagin
- Research and Development Institute of Biotechnologies, Samara State Medical University, Chapayevskaya St. 89, 443099 Samara, Russia
| | - Yeon Ho Kim
- RokitHealth Care Ltd., 9, Digital-ro 10-gil, Geumcheon-gu, Seoul 08514, Republic of Korea
| | - Denis G Alekseev
- Research and Development Institute of Biotechnologies, Samara State Medical University, Chapayevskaya St. 89, 443099 Samara, Russia
| |
Collapse
|
3
|
Gipson GR, Nolan K, Kattamuri C, Kenny AP, Agricola Z, Edwards NA, Zinski J, Czepnik M, Mullins MC, Zorn AM, Thompson TB. Formation and characterization of BMP2/GDF5 and BMP4/GDF5 heterodimers. BMC Biol 2023; 21:16. [PMID: 36726183 PMCID: PMC9893541 DOI: 10.1186/s12915-023-01522-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 01/19/2023] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Proteins of the TGFβ family, which are largely studied as homodimers, are also known to form heterodimers with biological activity distinct from their component homodimers. For instance, heterodimers of bone morphogenetic proteins, including BMP2/BMP7, BMP2/BMP6, and BMP9/BMP10, among others, have illustrated the importance of these heterodimeric proteins within the context of TGFβ signaling. RESULTS In this study, we have determined that mature GDF5 can be combined with mature BMP2 or BMP4 to form BMP2/GDF5 and BMP4/GDF5 heterodimer. Intriguingly, this combination of a BMP2 or BMP4 monomer, which exhibit high affinity to heparan sulfate characteristic to the BMP class, with a GDF5 monomer with low heparan sulfate affinity produces a heterodimer with an intermediate affinity. Using heparin affinity chromatography to purify the heterodimeric proteins, we then determined that both the BMP2/GDF5 and BMP4/GDF5 heterodimers consistently signaled potently across an array of cellular and in vivo systems, while the activities of their homodimeric counterparts were more context dependent. These differences were likely driven by an increase in the combined affinities for the type 1 receptors, Alk3 and Alk6. Furthermore, the X-ray crystal structure of BMP2/GDF5 heterodimer was determined, highlighting the formation of two asymmetric type 1 receptor binding sites that are both unique relative to the homodimers. CONCLUSIONS Ultimately, this method of heterodimer production yielded a signaling molecule with unique properties relative to the homodimeric ligands, including high affinity to multiple type 1 and moderate heparan binding affinity.
Collapse
Affiliation(s)
- Gregory R Gipson
- Department of Molecular & Cellular Biosciences, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kristof Nolan
- Department of Biochemistry and Molecular Biophysics, University of Chicago, Chicago, IL, USA
| | - Chandramohan Kattamuri
- Department of Molecular & Cellular Biosciences, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Alan P Kenny
- Perinatal Institute, Divisions of Developmental Biology and Neonatology & Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Zachary Agricola
- Perinatal Institute, Divisions of Developmental Biology and Neonatology & Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Nicole A Edwards
- Perinatal Institute, Divisions of Developmental Biology and Neonatology & Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Joseph Zinski
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Magdalena Czepnik
- Department of Molecular & Cellular Biosciences, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Mary C Mullins
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Aaron M Zorn
- Perinatal Institute, Divisions of Developmental Biology and Neonatology & Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Thomas B Thompson
- Department of Molecular & Cellular Biosciences, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
4
|
Discovery of Some Heterocyclic Molecules as Bone Morphogenetic Protein 2 (BMP-2)-Inducible Kinase Inhibitors: Virtual Screening, ADME Properties, and Molecular Docking Simulations. Molecules 2022; 27:molecules27175571. [PMID: 36080338 PMCID: PMC9457949 DOI: 10.3390/molecules27175571] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/13/2022] [Accepted: 08/18/2022] [Indexed: 12/02/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are growth factors that have a vital role in the production of bone, cartilage, ligaments, and tendons. Tumors’ upregulation of bone morphogenetic proteins (BMPs) and their receptors are key features of cancer progression. Regulation of the BMP kinase system is a new promising strategy for the development of anti-cancer drugs. In this work, based on a careful literature study, a library of benzothiophene and benzofuran derivatives was subjected to different computational techniques to study the effect of chemical structure changes on the ability of these two scaffolds to target BMP-2 inducible kinase, and to reach promising candidates with proposed activity against BMP-2 inducible kinase. The results of screening against Lipinski’s and Veber’s Rules produced twenty-one outside eighty-four compounds having drug-like molecular nature. Computational ADMET studies favored ten compounds (11, 26, 27, 29, 30, 31, 34, 35, 65, and 72) with good pharmacokinetic profile. Computational toxicity studies excluded compound 34 to elect nine compounds for molecular docking studies which displayed eight compounds (26, 27, 29, 30, 31, 35, 65, and 72) as promising BMP-2 inducible kinase inhibitors. The nine fascinating compounds will be subjected to extensive screening against serine/threonine kinases to explore their potential against these critical proteins. These promising candidates based on benzothiophene and benzofuran scaffolds deserve further clinical investigation as BMP-2 kinase inhibitors for the treatment of cancer.
Collapse
|
5
|
Nezu S, Saito T, Yoshida A, Narazaki S, Shimamura Y, Furumatsu T, Ozaki T. Effect of difference in fixation methods of tendon graft and the microfracture procedure on tendon-bone junction healing. JSES Int 2021; 6:155-166. [PMID: 35141691 PMCID: PMC8811408 DOI: 10.1016/j.jseint.2021.10.001] [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] [Indexed: 12/02/2022] Open
Abstract
Background There are generally two methods of fixation for tendon grafts used in ligament reconstruction: bone tunnel fixation and anchor fixation. The microfracture (Mf) procedure is a technique to induce bleeding from the bone marrow, and the bleeding may contain cells with differentiation potential. However, few studies have compared the effects of the Mf procedure with those of the fixation methods. This study aimed to evaluate the effectiveness of the Mf procedure on two tendon graft fixation methods: histological, gene expression, tendon graft thickness, and mechanical. We especially focused our investigation on junction healing of tendon grafts and bone in the two fixation methods. Methods We used 20 rabbits to evaluate tendon and bone healing in a peroneal tendon graft model. The rabbit models were divided into five groups according to the combination of peroneal tendon graft fixation method and Mf technique as follows: control group (C, n = 4), bone tunnel fixation without Mf procedure group (BT − Mf, n = 4), bone tunnel fixation with Mf procedure group (BT + Mf, n = 4), anchor fixation without Mf procedure group (A − Mf, n = 4), and anchor fixation with Mf procedure group (A + Mf, n = 4). All animals were sacrificed at 4 weeks postoperatively. The specimens underwent histological evaluation, mRNA analysis, tendon graft thickness at the tendon-bone junction, and biomechanical testing. Results Histological evaluation of the BT + Mf and A + Mf groups showed healing with fibrocartilage formation at the tendon graft-bone junction. The mRNA expression showed significant increase in type 2 collagen, Scleraxis, and SRY-box9 in the BT + Mf and A + Mf groups. In biomechanical tests, the BT + Mf and A + Mf groups showed significantly increased tensile strength compared with the BT − Mf and A − Mf groups (BT + Mf group, 21.6 ± 1.7 N; A + Mf group, 22.5 ± 2.3 N vs. BT − Mf group, 12.3 ± 2.4 N; A − Mf group, 11 ± 2.3 N). Conclusion The Mf procedure resulted in fibrocartilage formation at the tendon-bone junction in the BT and anchor fixation and improved the fixation strength at 4 weeks.
Collapse
Affiliation(s)
- Satoshi Nezu
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Taichi Saito
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Corresponding author: Taichi Saito, MD, PhD, Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine Dentistry, and Pharmaceutical Sciences, Okayama, Japan, 2-5-1, Shikatacho, Kitaku, Okayama City, 700-8558.
| | - Aki Yoshida
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinji Narazaki
- Department of Orthopaedic Surgery, Okayama Saiseikai General Hospital, Okayama, Okayama, Japan
| | - Yasunori Shimamura
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takayuki Furumatsu
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toshifumi Ozaki
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
6
|
Karimi Ghahfarrokhi E, Meimandi-Parizi A, Oryan A, Ahmadi N. Effects of Combination of BMP7, PFG, and Autograft on Healing of the Experimental Critical Radial Bone Defect by Induced Membrane (Masquelet) Technique in Rabbit. THE ARCHIVES OF BONE AND JOINT SURGERY 2021; 9:585-597. [PMID: 34692943 DOI: 10.22038/abjs.2020.50852.2532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/02/2020] [Indexed: 01/08/2023]
Abstract
Background Healing of large segmental bone defects can be challenging for orthopedic surgeons. This research was conducted to provide further insight into the effects of BMP7 in combination with autograft and platelet fibrin glue (PFG) on bone regeneration by Masquelet technique (MT). Methods Twenty five domestic male rabbits, more than 6 months old, weighing 2.00±0.25 kg were randomly divided into five equal groups as follows: MT-blank cavity (without any biological or synthetic materials) (1), blank cavity (2), MT-autograft (3), MT-autograft-BMP7 (4), and MT-BMP7-PFG (5). A 20 mm segmental defect was made in radial bone in both forelimbs. The Masquelet technique was done in all groups except group 2. The study was evaluated by radiology, biomechanics, histopathology and scanning electron microscopy. Results The results showed that Masquelet technique enhanced the healing process, as, the structural and functional criteria of the injured bone showed significantly improved bone healing (P<0.05). Treatment by PFG-BMP7, Autograft-BMP7, and autograft demonstrated beneficial effects on bone healing. However, Autograft-BMP7 was more effective than autograft in healing of the radial defect in rabbits. Conclusion Our findings introduce the osteogenic materials in combination with Masquelet technique as an alternative for reconstruction of the big diaphyseal defects in the long bones in animal models. Our findings may be useful for clinical application in future.
Collapse
Affiliation(s)
| | | | - Ahmad Oryan
- Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Nasrollah Ahmadi
- Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
7
|
Zheng W, Li H, Hu K, Li L, Bei M. Chondromalacia patellae: current options and emerging cell therapies. Stem Cell Res Ther 2021; 12:412. [PMID: 34275494 PMCID: PMC8287755 DOI: 10.1186/s13287-021-02478-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 06/22/2021] [Indexed: 01/08/2023] Open
Abstract
Chondromalacia patellae (CMP), also known as runner’s knee, typically occurs in young patients, which is characterized by anterior knee pain (AKP) that is associated with visible changes in patellar cartilage. The initial pathological changes include cartilage softening, swelling, and edema. CMP is caused by several factors, including trauma, increased cartilage vulnerability, patellofemoral instability, bony anatomic variations, abnormal patellar kinematics, and occupation hazards. CMP may be reversible or may progress to develop patellofemoral osteoarthritis. Quadriceps wasting, patellofemoral crepitus, and effusion are obvious clinical indications. Additionally, radiological examinations are also necessary for diagnosis. Magnetic resonance imaging (MRI) is a non-invasive diagnostic method, which holds a promise in having the unique ability to potentially identify cartilage lesions. Modalities are conventionally proposed to treat cartilage lesions in the PF joint, but none have emerged as a gold standard, neither to alleviated symptoms and function nor to prevent OA degeneration. Recently, researchers have been focused on cartilage-targeted therapy. Various efforts including cell therapy and tissue emerge for cartilage regeneration exhibit as the promising regime, especially in the application of mesenchymal stem cells (MSCs). Intra-articular injections of variously sourced MSC are found safe and beneficial for treating CMP with improved clinical parameters, less invasiveness, symptomatic relief, and reduced inflammation. The mechanism of MSC injection remains further clinical investigation and is tremendously promising for CMP treatment. In this short review, etiology, MRI diagnosis, and treatment in CMP, especially the treatment of the cell-based therapies, are reviewed.
Collapse
Affiliation(s)
- Weitao Zheng
- Hubei Provincial Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, 430068, Hubei Province, China.,Shanxi Yinmei Technology Co., Taiyuan Economic and Technological Development Zone, Room 301, No. 8, East Street, Taiyuan, China
| | - Hanluo Li
- Hubei Provincial Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, 430068, Hubei Province, China
| | - Kanghong Hu
- Hubei Provincial Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, 430068, Hubei Province, China
| | - Liming Li
- Shanxi Yinmei Technology Co., Taiyuan Economic and Technological Development Zone, Room 301, No. 8, East Street, Taiyuan, China
| | - Mingjian Bei
- Department of Orthopedic Surgery, Emergency General Hospital, Xibahenanli29, Chaoyang dis, Beijing, 100028, China.
| |
Collapse
|
8
|
Postradiation Fractures after Combined Modality Treatment in Extremity Soft Tissue Sarcomas. Sarcoma 2021; 2021:8877567. [PMID: 33790687 PMCID: PMC7984930 DOI: 10.1155/2021/8877567] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 01/20/2021] [Accepted: 02/27/2021] [Indexed: 12/26/2022] Open
Abstract
Soft tissue sarcoma (STS) of the extremities is typically treated with limb-sparing surgery and radiation therapy; with this treatment approach, high local control rates can be achieved. However, postradiation bone fractures, fractures occurring in the prior radiation field with minimal or no trauma, are a serious late complication that occurs in 2–22% of patients who receive surgery and radiation for STS. Multiple risk factors for sustaining a postradiation fracture exist, including high radiation dose, female sex, periosteal stripping, older age, femur location, and chemotherapy administration. The treatment of these pathological fractures can be difficult, with complications including delayed union, nonunion, and infection posing particular challenges. Here, we review the mechanisms, risk factors, and treatment challenges associated with postradiation fractures in STS patients.
Collapse
|
9
|
Characterization of the different oligomeric states of the DAN family antagonists SOSTDC1 and SOST. Biochem J 2021; 477:3167-3182. [PMID: 32779697 PMCID: PMC7473711 DOI: 10.1042/bcj20200552] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/07/2020] [Accepted: 08/11/2020] [Indexed: 12/16/2022]
Abstract
The DAN (differential screening-selected gene aberrative in neuroblastoma) family are a group of secreted extracellular proteins which typically bind to and antagonize BMP (bone morphogenetic protein) ligands. Previous studies have revealed discrepancies between the oligomerization state of certain DAN family members, with SOST (a poor antagonist of BMP signaling) forming a monomer while Grem1, Grem2, and NBL1 (more potent BMP antagonists) form non-disulfide linked dimers. The protein SOSTDC1 (Sclerostin domain containing protein 1) is sequentially similar to SOST, but has been shown to be a better BMP inhibitor. In order to determine the oligomerization state of SOSTDC1 and determine what effect dimerization might have on the mechanism of DAN family antagonism of BMP signaling, we isolated the SOSTDC1 protein and, using a battery of biophysical, biochemical, and structural techniques, showed that SOSTDC1 forms a highly stable non-covalent dimer. Additionally, this SOSTDC1 dimer was shown, using an in vitro cell based assay system, to be an inhibitor of multiple BMP signaling growth factors, including GDF5, while monomeric SOST was a very poor antagonist. These results demonstrate that SOSTDC1 is distinct from paralogue SOST in terms of both oligomerization and strength of BMP inhibition.
Collapse
|
10
|
Lawrence EA, Aggleton J, van Loon J, Godivier J, Harniman R, Pei J, Nowlan N, Hammond C. Exposure to hypergravity during zebrafish development alters cartilage material properties and strain distribution. Bone Joint Res 2021; 10:137-148. [PMID: 33560137 DOI: 10.1302/2046-3758.102.bjr-2020-0239.r1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
AIMS Vertebrates have adapted to life on Earth and its constant gravitational field, which exerts load on the body and influences the structure and function of tissues. While the effects of microgravity on muscle and bone homeostasis are well described, with sarcopenia and osteoporosis observed in astronauts returning from space, the effects of shorter exposures to increased gravitational fields are less well characterized. We aimed to test how hypergravity affects early cartilage and skeletal development in a zebrafish model. METHODS We exposed zebrafish to 3 g and 6 g hypergravity from three to five days post-fertilization, when key events in jaw cartilage morphogenesis occur. Following this exposure, we performed immunostaining along with a range of histological stains and transmission electron microscopy (TEM) to examine cartilage morphology and structure, atomic force microscopy (AFM) and nanoindentation experiments to investigate the cartilage material properties, and finite element modelling to map the pattern of strain and stress in the skeletal rudiments. RESULTS We did not observe changes to larval growth, or morphology of cartilage or muscle. However, we observed altered mechanical properties of jaw cartilages, and in these regions we saw changes to chondrocyte morphology and extracellular matrix (ECM) composition. These areas also correspond to places where strain and stress distribution are predicted to be most different following hypergravity exposure. CONCLUSION Our results suggest that altered mechanical loading, through hypergravity exposure, affects chondrocyte maturation and ECM components, ultimately leading to changes to cartilage structure and function. Cite this article: Bone Joint Res 2021;10(2):137-148.
Collapse
Affiliation(s)
| | - Jessye Aggleton
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK.,School of Anthropology and Archaeology, University of Bristol, Bristol, UK
| | - Jack van Loon
- European Space Agency (ESA) Technology Center (ESTEC), TEC-MMG, Noordwijk, The Netherlands.,Department Oral & Maxillofacial Surgery/Pathology, Amsterdam Movement Sciences & Amsterdam Bone Center (ABC), Amsterdam University Medical Center Location VUmc & Academic Center for Dentistry Amsterdam (ACTA), Amsterdam, The Netherlands
| | - Josepha Godivier
- Department of Bioengineering, Imperial College London, London, UK
| | | | - Jiaxin Pei
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Niamh Nowlan
- Department of Bioengineering, Imperial College London, London, UK
| | - Chrissy Hammond
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| |
Collapse
|
11
|
Gipson GR, Goebel EJ, Hart KN, Kappes EC, Kattamuri C, McCoy JC, Thompson TB. Structural perspective of BMP ligands and signaling. Bone 2020; 140:115549. [PMID: 32730927 PMCID: PMC7502536 DOI: 10.1016/j.bone.2020.115549] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 06/30/2020] [Accepted: 07/03/2020] [Indexed: 12/13/2022]
Abstract
The Bone Morphogenetic Proteins (BMPs) are the largest class signaling molecules within the greater Transforming Growth Factor Beta (TGFβ) family, and are responsible for a wide array of biological functions, including dorsal-ventral patterning, skeletal development and maintenance, as well as cell homeostasis. As such, dysregulation of BMPs results in a number of diseases, including fibrodysplasia ossificans progressiva (FOP) and pulmonary arterial hypertension (PAH). Therefore, understanding BMP signaling and regulation at the molecular level is essential for targeted therapeutic intervention. This review discusses the recent advances in the structural and biochemical characterization of BMPs, from canonical ligand-receptor interactions to co-receptors and antagonists. This work aims to highlight how BMPs differ from other members of the TGFβ family, and how that information can be used to further advance the field. Lastly, this review discusses several gaps in the current understanding of BMP structures, with the aim that discussion of these gaps will lead to advancements in the field.
Collapse
Affiliation(s)
- Gregory R Gipson
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Medical Sciences Building, Cincinnati, OH 45267, USA
| | - Erich J Goebel
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Medical Sciences Building, Cincinnati, OH 45267, USA
| | - Kaitlin N Hart
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Medical Sciences Building, Cincinnati, OH 45267, USA
| | - Emily C Kappes
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Medical Sciences Building, Cincinnati, OH 45267, USA
| | - Chandramohan Kattamuri
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Medical Sciences Building, Cincinnati, OH 45267, USA
| | - Jason C McCoy
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Medical Sciences Building, Cincinnati, OH 45267, USA
| | - Thomas B Thompson
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Medical Sciences Building, Cincinnati, OH 45267, USA.
| |
Collapse
|
12
|
Lee EJ, Jain M, Alimperti S. Bone Microvasculature: Stimulus for Tissue Function and Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:313-329. [PMID: 32940150 DOI: 10.1089/ten.teb.2020.0154] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bone is a highly vascularized organ, providing structural support to the body, and its development, regeneration, and remodeling depend on the microvascular homeostasis. Loss or impairment of vascular function can develop diseases, such as large bone defects, avascular necrosis, osteoporosis, osteoarthritis, and osteopetrosis. In this review, we summarize how vasculature controls bone development and homeostasis in normal and disease cases. A better understanding of this process will facilitate the development of novel disease treatments that promote bone regeneration and remodeling. Specifically, approaches based on tissue engineering components, such as stem cells and growth factors, have demonstrated the capacity to induce bone microvasculature regeneration and mineralization. This knowledge will have relevant clinical implications for the treatment of bone disorders by developing novel pharmaceutical approaches and bone grafts. Finally, the tissue engineering approaches incorporating vascular components may widely be applied to treat other organ diseases by enhancing their regeneration capacity. Impact statement Bone vasculature is imperative in the process of bone development, regeneration, and remodeling. Alterations or disruption of the bone vasculature leads to loss of bone homeostasis and the development of bone diseases. In this study, we review the role of vasculature on bone diseases and how vascular tissue engineering strategies, with a detailed emphasis on the role of stem cells and growth factors, will contribute to bone therapeutics.
Collapse
Affiliation(s)
- Eun-Jin Lee
- American Dental Association Science and Research Institute, Gaithersburg, Maryland, USA
| | - Mahim Jain
- Kennedy Krieger Institute, John Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Stella Alimperti
- American Dental Association Science and Research Institute, Gaithersburg, Maryland, USA
| |
Collapse
|
13
|
Potential roles of bone morphogenetic protein-9 in glucose and lipid homeostasis. J Physiol Biochem 2020; 76:503-512. [PMID: 32808114 DOI: 10.1007/s13105-020-00763-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/09/2020] [Indexed: 12/20/2022]
Abstract
Bone morphogenetic protein-9 (BMP-9) is a novel cytokine which is cloned from the fetal mouse liver cDNA library and belongs to the member of the transforming growth factor-β (TGF-β) superfamily. BMP-9 is mainly secreted by the liver and exerts a variety of physiological functions. In this review, we present the latest knowledge on the biochemistry of BMP-9 and its role in glucose metabolism and lipid homeostasis. We introduced the expression site, structure, synthesis, and secretion of BMP-9, as well as BMP-9 signaling pathway. We also discuss the effects of BMP-9 on glucose metabolism and lipid metabolism in different organs. BMP-9 can regulate glucose and lipid homeostasis in the body by inhibiting liver gluconeogenesis, transforming white adipose tissue to brown adipose tissue, promoting muscle glycogen synthesis, increasing the uptake and utilization of glucose by muscle tissue, increasing liver and adipose tissue insulin sensitivity, promoting insulin synthesis and secretion, inhibiting liver lipid deposition, and playing a leptin-like role. Finally, through the results of animal intervention studies and human clinical studies in the review, we deeply understand the association of BMP-9 with obesity, insulin resistance (IR), type 2 diabetes, and non-alcoholic fatty liver disease (NAFLD), which provides new ideas for the prevention and treatment of diseases.
Collapse
|
14
|
Wang T, Hill RC, Dzieciatkowska M, Zhu L, Infante AM, Hu G, Hansen KC, Pei M. Site-Dependent Lineage Preference of Adipose Stem Cells. Front Cell Dev Biol 2020; 8:237. [PMID: 32351957 PMCID: PMC7174673 DOI: 10.3389/fcell.2020.00237] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/20/2020] [Indexed: 12/15/2022] Open
Abstract
Adult stem cells have unique properties in both proliferation and differentiation preference. In this study, we hypothesized that adipose stem cells have a depot-dependent lineage preference. Four rabbits were used to provide donor-matched adipose stem cells from either subcutaneous adipose tissue (ScAT) or infrapatellar fat pad (IPFP). Proliferation and multi-lineage differentiation were evaluated in adipose stem cells from donor-matched ScAT and IPFP. RNA sequencing (RNA-seq) and proteomics were conducted to uncover potential molecular discrepancy in adipose stem cells and their corresponding matrix microenvironments. We found that stem cells from ScAT exhibited significantly higher proliferation and adipogenic capacity compared to those from donor-matched IPFP while stem cells from IPFP displayed significantly higher chondrogenic potential compared to those from donor-matched ScAT. Our findings are strongly endorsed by supportive data from transcriptome and proteomics analyses, indicating a site-dependent lineage preference of adipose stem cells.
Collapse
Affiliation(s)
- Tingliang Wang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopedics, West Virginia University, Morgantown, WV, United States
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ryan C. Hill
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, United States
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, United States
| | - Lian Zhu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aniello M. Infante
- Bioinformatics Core Facility, West Virginia University, Morgantown, WV, United States
| | - Gangqing Hu
- Bioinformatics Core Facility, West Virginia University, Morgantown, WV, United States
- Department of Microbiology, Immunology and Cell Biology, School of Medicine, West Virginia University, Morgantown, WV, United States
| | - Kirk C. Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, United States
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopedics, West Virginia University, Morgantown, WV, United States
- WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
15
|
Aluganti Narasimhulu C, Singla DK. The Role of Bone Morphogenetic Protein 7 (BMP-7) in Inflammation in Heart Diseases. Cells 2020; 9:cells9020280. [PMID: 31979268 PMCID: PMC7073173 DOI: 10.3390/cells9020280] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/31/2022] Open
Abstract
Bone morphogenetic protein-7 is (BMP-7) is a potent anti-inflammatory growth factor belonging to the Transforming Growth Factor Beta (TGF-β) superfamily. It plays an important role in various biological processes, including embryogenesis, hematopoiesis, neurogenesis and skeletal morphogenesis. BMP-7 stimulates the target cells by binding to specific membrane-bound receptor BMPR 2 and transduces signals through mothers against decapentaplegic (Smads) and mitogen activated protein kinase (MAPK) pathways. To date, rhBMP-7 has been used clinically to induce the differentiation of mesenchymal stem cells bordering the bone fracture site into chondrocytes, osteoclasts, the formation of new bone via calcium deposition and to stimulate the repair of bone fracture. However, its use in cardiovascular diseases, such as atherosclerosis, myocardial infarction, and diabetic cardiomyopathy is currently being explored. More importantly, these cardiovascular diseases are associated with inflammation and infiltrated monocytes where BMP-7 has been demonstrated to be a key player in the differentiation of pro-inflammatory monocytes, or M1 macrophages, into anti-inflammatory M2 macrophages, which reduces developed cardiac dysfunction. Therefore, this review focuses on the molecular mechanisms of BMP-7 treatment in cardiovascular disease and its role as an anti-fibrotic, anti-apoptotic and anti-inflammatory growth factor, which emphasizes its potential therapeutic significance in heart diseases.
Collapse
|
16
|
Zhang J, Zhu X, Kong Y, Huang Y, Dang X, Mei L, Zhao B, Lin Q, Wang J. Strontium stimulates alkaline phosphatase and bone morphogenetic protein-4 expression in rat chondrocytes cultured in vitro. J Trace Elem Med Biol 2019; 55:15-19. [PMID: 31345353 DOI: 10.1016/j.jtemb.2019.05.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/14/2019] [Accepted: 05/17/2019] [Indexed: 12/22/2022]
Abstract
The trace element strontium has a significant impact on cartilage metabolism. However, the direct effects of strontium on alkaline phosphatase (ALP), a marker of bone growth, and bone morphogenetic protein-4 (BMP-4), which plays a key role in the regulation of bone and cartilage development, are not entirely clear. In order to understand the mechanisms involved in these processes, the chondrocytes were isolated from Wistar rat articular cartilage by enzymatic digestion and cultured under standard conditions. They were then treated with strontium at 0.5, 1.0, 2.0, 5.0, 20.0 and 100.0 μg/mL for 72 h. The mRNA abundance and protein expression levels of ALP and BMP-4 were measured using real-time polymerase chain reaction (real-time PCR) and Western blot analysis. The results showed that the levels of expression of ALP and BMP-4 in chondrocytes increased as the concentration of strontium increased relative to the control group, and the difference became significant at 1.0 μg/mL strontium (P<0.05). These results indicated that strontium could be involved in cartilage development via regulating ALP and BMP-4 expression.
Collapse
Affiliation(s)
- Jinfeng Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China; State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, Qinghai, China
| | - Xiaoyan Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Yezi Kong
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Yan Huang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xukun Dang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Linshan Mei
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Baoyu Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Qing Lin
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China; State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, Qinghai, China.
| | - Jianguo Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China.
| |
Collapse
|
17
|
Kakuta A, Tanaka T, Chazono M, Komaki H, Kitasato S, Inagaki N, Akiyama S, Marumo K. Effects of micro-porosity and local BMP-2 administration on bioresorption of β-TCP and new bone formation. Biomater Res 2019; 23:12. [PMID: 31372237 PMCID: PMC6660686 DOI: 10.1186/s40824-019-0161-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/17/2019] [Indexed: 01/19/2023] Open
Abstract
Background It has been reported that the microporous structure of calcium phosphate (CaP) ceramics is important to osteoconduction. Bone morphogenetic protein-2 (BMP-2) has been shown to be a promising alternative to bone grafting and a therapeutic agent promoting bone regeneration when delivered locally. The aim of this study was to evaluate the effects of micro-porosity within beta-tricalcium phosphate (β-TCP) cylinders and local BMP-2 administration on β-TCP resorption and new bone formation. Methods Bilateral cylindrical bone defects were created in rabbit distal femora, and the defects were filled with β-TCP. Rabbits were divided into 3 groups; defects were filled with a β-TCP cylinder with a total of approximately 60% porosity (Group A: 13.4% micro- and 46.9% macropore, Group B: 38.5% micro- and 20.3% macropore, Group C: the same micro- and macro-porosity as in group B supplemented with BMP-2). Rabbits were sacrificed 4, 8, 12, and 24 weeks postoperatively. Results The number of TRAP-positive cells and new bone formation in group B were significantly greater than those in group A at every period. The amount of residual β-TCP in group C was less than that in group B at all time periods, resulting in significantly more new bone formation in group C at 8 and 12 weeks. The number of TRAP-positive cells in group C was maximum at 4 weeks. Conclusions These results suggest that the amount of submicron microporous structure and local BMP-2 administration accelerated both osteoclastic resorption of β-TCP and new bone formation, probably through a coupling-like phenomenon between resorption and new bone formation.
Collapse
Affiliation(s)
- Atsuhito Kakuta
- 1Department of Orthopaedic Surgery, Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Minato-ku, Tokyo, 105-0003 Japan
| | - Takaaki Tanaka
- 1Department of Orthopaedic Surgery, Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Minato-ku, Tokyo, 105-0003 Japan.,Department of Orthopaedic Surgery, NHO Utsunomiya National Hospital, 2160 Shimo-Okamoto, Utsunomiya City, Tochigi 329-1193 Japan
| | - Masaaki Chazono
- 1Department of Orthopaedic Surgery, Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Minato-ku, Tokyo, 105-0003 Japan.,Department of Orthopaedic Surgery, NHO Utsunomiya National Hospital, 2160 Shimo-Okamoto, Utsunomiya City, Tochigi 329-1193 Japan
| | - Hirokazu Komaki
- 1Department of Orthopaedic Surgery, Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Minato-ku, Tokyo, 105-0003 Japan
| | - Seiichiro Kitasato
- 1Department of Orthopaedic Surgery, Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Minato-ku, Tokyo, 105-0003 Japan
| | - Naoya Inagaki
- 1Department of Orthopaedic Surgery, Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Minato-ku, Tokyo, 105-0003 Japan
| | - Shoshi Akiyama
- 1Department of Orthopaedic Surgery, Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Minato-ku, Tokyo, 105-0003 Japan
| | - Keishi Marumo
- 1Department of Orthopaedic Surgery, Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Minato-ku, Tokyo, 105-0003 Japan
| |
Collapse
|
18
|
Convergence of TGFβ and BMP signaling in regulating human bone marrow stromal cell differentiation. Sci Rep 2019; 9:4977. [PMID: 30899078 PMCID: PMC6428815 DOI: 10.1038/s41598-019-41543-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 02/26/2019] [Indexed: 01/11/2023] Open
Abstract
Targeting regulatory signaling pathways that control human bone marrow stromal (skeletal or mesenchymal) stem cell (hBMSC) differentiation and lineage fate determination is gaining momentum in the regenerative medicine field. Therefore, to identify the central regulatory mechanism of osteoblast differentiation of hBMSCs, the molecular phenotypes of two clonal hBMSC lines exhibiting opposite in vivo phenotypes, namely, bone forming (hBMSC+bone) and non-bone forming (hBMSC−Bone) cells, were studied. Global transcriptome analysis revealed significant downregulation of several TGFβ responsive genes, namely, TAGLN, TMP1, ACTA2, TGFβ2, SMAD6, SMAD9, BMP2, and BMP4 in hBMSC−Bone cells and upregulation on SERPINB2 and NOG. Transcriptomic data was associated with marked reduction in SMAD2 protein phosphorylation, which thereby implies the inactivation of TGFβ and BMP signaling in those cells. Concordantly, activation of TGFβ signaling in hBMSC−Bone cells using either recombinant TGFβ1 protein or knockdown of SERPINB2 TGFβ-responsive gene partially restored their osteoblastic differentiation potential. Similarly, the activation of BMP signaling using exogenous BMP4 or via siRNA-mediated knockdown of NOG partially restored the differentiation phenotype of hBMSC−Bone cells. Concordantly, recombinant NOG impaired ex vivo osteoblastic differentiation of hBMSC+Bone cells, which was associated with SERBINB2 upregulation. Our data suggests the existence of reciprocal relationship between TGFB and BMP signaling that regulates hBMSC lineage commitment and differentiation, whilst provide a plausible strategy for generating osteoblastic committed cells from hBMSCs for clinical applications.
Collapse
|
19
|
Rausch V, Seybold D, Königshausen M, Köller M, Schildhauer TA, Geßmann J. [Basic principles of fracture healing]. DER ORTHOPADE 2018; 46:640-647. [PMID: 28718007 DOI: 10.1007/s00132-017-3449-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND In contrast to other tissues, bone has the remarkable ability to heal without scarring. After union of the fracture, the remodeled bone ideally does not differ from the original bone, especially in terms of biomechanical properties. The healing of a fracture resembles the embryonic development of bone. Depending on the biomechanical properties of the fracture, bone heals directly or indirectly, which refers to the formation of cartilage as a step before new bone appears. Currently, treatment of the patient is often limited to anatomical reduction and optimization of the fracture environment with respect to biomechanics. PROSPECTS Future treatment strategies, however, could include systemic medication that could be especially beneficial for patients at risk of complications in fracture healing. The aim of this review is to provide an overview on the process of fracture healing and to depict possibilities for current and future treatment strategies.
Collapse
Affiliation(s)
- Valentin Rausch
- Chirurgische Universitätsklinik und Poliklinik, Berufsgenossenschaftliches Universitätsklinikum Bergmannsheil, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Deutschland
| | - Dominik Seybold
- Chirurgische Universitätsklinik und Poliklinik, Berufsgenossenschaftliches Universitätsklinikum Bergmannsheil, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Deutschland.
| | - Matthias Königshausen
- Chirurgische Universitätsklinik und Poliklinik, Berufsgenossenschaftliches Universitätsklinikum Bergmannsheil, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Deutschland
| | - Manfred Köller
- Chirurgische Universitätsklinik und Poliklinik, Berufsgenossenschaftliches Universitätsklinikum Bergmannsheil, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Deutschland
| | - Thomas A Schildhauer
- Chirurgische Universitätsklinik und Poliklinik, Berufsgenossenschaftliches Universitätsklinikum Bergmannsheil, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Deutschland
| | - Jan Geßmann
- Chirurgische Universitätsklinik und Poliklinik, Berufsgenossenschaftliches Universitätsklinikum Bergmannsheil, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Deutschland
| |
Collapse
|
20
|
Smith RAA, Murali S, Rai B, Lu X, Lim ZXH, Lee JJL, Nurcombe V, Cool SM. Minimum structural requirements for BMP-2-binding of heparin oligosaccharides. Biomaterials 2018; 184:41-55. [PMID: 30205243 DOI: 10.1016/j.biomaterials.2018.08.056] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/24/2018] [Accepted: 08/27/2018] [Indexed: 12/27/2022]
Abstract
Bone morphogenetic proteins (BMPs) are essential during tissue repair and remodeling after injury. Glycosaminoglycan (GAG) sugars are known to enhance BMP activity in vitro and in vivo; here the interactions of BMP-2 with various glycosaminoglycan classes were compared and shown to be selective for heparin over other comparable saccharides. The minimal chain lengths and specific sulfate moieties required for heparin-derived oligosaccharide binding to BMP-2, and the ability of such oligosaccharides to promote BMP-2-induced osteogenic differentiation in vitro were then determined. BMP-2 could bind to heparin hexasaccharides (dp6) and octasaccharides (dp8), but decasaccharides (dp10) were the minimum chain length required for both efficient binding of BMP-2 and consequent heparin-dependent cell responses. N-sulfation is the most important, and 6-O-sulfation moderately important for BMP-2 binding and activity, whereas 2-O-sulfation was much less critical. Bone formation assays in vivo further confirmed that dp10, N-sulfated heparin oligosaccharides were the minimal requirement for effective enhancement of BMP-2-induced bone formation. Such information is necessary for the rational design of the next generations of heparan-based devices for bone tissue repair.
Collapse
Affiliation(s)
- Raymond A A Smith
- Glycotherapeutics Group, Institute of Medical Biology, 8A Biomedical Grove, #06-06 Immunos, 138648, Singapore
| | - Sadasivam Murali
- Glycotherapeutics Group, Institute of Medical Biology, 8A Biomedical Grove, #06-06 Immunos, 138648, Singapore
| | - Bina Rai
- Glycotherapeutics Group, Institute of Medical Biology, 8A Biomedical Grove, #06-06 Immunos, 138648, Singapore
| | - Xiaohua Lu
- Glycotherapeutics Group, Institute of Medical Biology, 8A Biomedical Grove, #06-06 Immunos, 138648, Singapore
| | - Zophia Xue Hui Lim
- Glycotherapeutics Group, Institute of Medical Biology, 8A Biomedical Grove, #06-06 Immunos, 138648, Singapore
| | - Jaslyn J L Lee
- Glycotherapeutics Group, Institute of Medical Biology, 8A Biomedical Grove, #06-06 Immunos, 138648, Singapore
| | - Victor Nurcombe
- Glycotherapeutics Group, Institute of Medical Biology, 8A Biomedical Grove, #06-06 Immunos, 138648, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University-Imperial College London, Singapore
| | - Simon M Cool
- Glycotherapeutics Group, Institute of Medical Biology, 8A Biomedical Grove, #06-06 Immunos, 138648, Singapore; Dept. of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119074, Singapore.
| |
Collapse
|
21
|
Stranger Things: A Whimsical Account of a Demineralized Bone Matrix Study With Unexpected Results. J Craniofac Surg 2018; 29:1107-1109. [DOI: 10.1097/scs.0000000000004520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
22
|
Pacelli S, Basu S, Berkland C, Wang J, Paul A. Design of a cytocompatible hydrogel coating to modulate properties of ceramic-based scaffolds for bone repair. Cell Mol Bioeng 2018; 11:211-217. [PMID: 30338007 DOI: 10.1007/s12195-018-0521-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Introduction Physical and mechanical properties of ceramic-based scaffolds can be modulated by introducing hydrogel coatings on their surface. For instance, hydrogels can be used as elastic layers to overcome the brittleness of synthetic ceramic materials or to control the delivery of essential osteogenic factors. In this work, we aimed to achieve both goals by fabricating a novel cytocompatible hydrogel made of gelatin-alginate as a coating for beta-tricalcium phosphate (β-TCP) scaffolds. Methods The hydrogel synthesis was optimized by varying the concentration of the crosslinkers N-hydroxysuccinimide and N-Ethyl-N'-(3-dimethyl aminopropyl) carbodiimide (NHS/EDC). Swelling, degradability and mechanical studies were carried out to identify the suitable hydrogel coating formulation for the β-TCP scaffolds. The cytocompatibility of the coated ceramic was assessed in vitro by testing the proliferation and the osteogenic differentiation of human adipose stem cell (hASCs) for two weeks. Results The designed hydrogel layer could withstand cyclic compression and protected the brittle internal core of the ceramic. The hydrogel coating modulated the diffusion of the model protein BSA according to the degree of crosslinking of the hydrogel layer. Additionally, the polymeric network was able to retain positively charged proteins such as lysozyme due to the strong electrostatic interactions with carboxylic groups of alginate. A higher expression of alkaline phosphates activity was found on hASCs seeded on the coated scaffolds compared to the hydrogels without any β-TCP. Conclusion Overall, the hydrogel coating characterized in this study represents a valid strategy to overcome limitations of brittle ceramic-based materials used as scaffolds for bone tissue engineering applications.
Collapse
Affiliation(s)
- Settimio Pacelli
- BioIntel Research Laboratory, Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, School of Engineering, University of Kansas, Lawrence, KS 66045 USA
| | - Sayantani Basu
- BioIntel Research Laboratory, Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, School of Engineering, University of Kansas, Lawrence, KS 66045 USA
| | - Cory Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047 USA.,Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, University of Kansas, Lawrence, KS 66045 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 & Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160 USA
| | - Arghya Paul
- BioIntel Research Laboratory, Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, School of Engineering, University of Kansas, Lawrence, KS 66045 USA
| |
Collapse
|
23
|
Park SY, Kim KH, Park CH, Shin SY, Rhyu IC, Lee YM, Seol YJ. Enhanced Bone Regeneration by Diabetic Cell-Based Adenoviral BMP-2 Gene Therapy in Diabetic Animals. Tissue Eng Part A 2018; 24:930-942. [PMID: 29160182 DOI: 10.1089/ten.tea.2017.0101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The application of bone morphogenetic protein 2 (BMP-2) has been extensively investigated to improve diabetes-impaired bone healing; however, the delivery of BMP-2 by gene therapy for bone regeneration has rarely been investigated in diabetic animals. In this study, we aimed to evaluate which cells induce more new bone formation in diabetic animals when cell-based BMP2 gene therapy is applied. For this purpose, we harvested bone marrow stromal cells (BMSCs) twice in the same animal before (non-diabetic BMSCs; nBMSCs) and after diabetes induction (diabetic BMSCs; dBMSCs) using modified bone marrow ablation methods. And then, cells were transduced by adenoviral vectors carrying the BMP2 gene (AdBMP2). In in vitro, AdBMP2-transfected dBMSCs (B2/dBMSCs) produced higher BMP-2 mRNA levels over 48 h, whereas AdBMP2-transfected nBMSCs (B2/nBMSCs) exhibited a transient increase in BMP-2 mRNA followed by a decrease to the baseline level within 48 h. Both B2/dBMSCs and B2/nBMSCs induced secretion of BMP-2 for 3 weeks. However, B2/dBMSC BMP-2 secretion peaked from day 3 to 10, whereas B2/nBMSC BMP-2 secretion peaked from day 1 to 7. The analysis of osteogenic activity revealed that mineralization nodule formation and the expression levels of osteogenic genes were significantly higher in B2/dBMSCs than B2/nBMSCs and were accompanied by upregulation of canonical Wnt/β-catenin and Smad signaling. AdBMP2-transfected autologous cells were implanted into critical-sized calvarial defects in diabetic animals and induced significantly more bone regeneration than non-AdBMP2-transfected cells. In addition, B2/dBMSCs led to significantly more new bone formation than B2/nBMSCs. Thus, BMP2 gene therapy using diabetic cells effectively supported diabetic bone healing and it was related to the enhanced responses to AdBMP2 of dBMSCs.
Collapse
Affiliation(s)
- Shin-Young Park
- 1 Department of Periodontology and Dental Research Institute, School of Dentistry, Seoul National University , Seoul, Korea.,2 Section of Dentistry, Department of Periodontology, Seoul National University Bundang Hospital , Seongnam, Gyeonggi-do, Korea
| | - Kyoung-Hwa Kim
- 1 Department of Periodontology and Dental Research Institute, School of Dentistry, Seoul National University , Seoul, Korea
| | - Chan-Ho Park
- 3 Dental Research Institute, Seoul National University , Seoul, Korea
| | - Seung-Yun Shin
- 4 Department of Periodontology, Institute of Oral Biology, School of Dentistry, Kyung Hee University , Seoul, Korea
| | - In-Chul Rhyu
- 1 Department of Periodontology and Dental Research Institute, School of Dentistry, Seoul National University , Seoul, Korea
| | - Yong-Moo Lee
- 1 Department of Periodontology and Dental Research Institute, School of Dentistry, Seoul National University , Seoul, Korea
| | - Yang-Jo Seol
- 1 Department of Periodontology and Dental Research Institute, School of Dentistry, Seoul National University , Seoul, Korea
| |
Collapse
|
24
|
Jung HD, Kim SY, Jung HS, Park HS, Jung YS. Immunohistochemical Analysis on Cortex-to-Cortex Healing After Mandibular Vertical Ramus Osteotomy: A Preliminary Study. J Oral Maxillofac Surg 2017; 76:437.e1-437.e8. [PMID: 29112826 DOI: 10.1016/j.joms.2017.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 10/05/2017] [Accepted: 10/05/2017] [Indexed: 10/18/2022]
Abstract
PURPOSE The present study analyzed the expression of specific cytokines in the transforming growth factor (TGF)-β superfamily postoperatively after mandibular vertical ramus osteotomy (VRO). MATERIALS AND METHODS Four beagle dogs were enrolled and euthanized at 1, 2, 4, and 8 weeks postoperatively for immunohistochemical analysis using 6 specific antibodies (bone morphogenetic protein [BMP]-2/4, BMP-7, TGF-β2, TGF-β3, matrix metalloproteinase-3, and vascular endothelial growth factor [VEGF]). The results from the surgical site and control (adjacent area) were compared. RESULTS Generalized upregulation of BMP-2/4 was observed in all healing periods, and the strongest expression of BMP-7 was observed at 1 week postoperatively. The strongest expression of TGF-β2 was observed at 8 weeks with increasing pattern. The strong expression of TGF-β3 was observed at 1 and 4 weeks, with the strongest expression of VEGF at 1 week, with a decreasing pattern. No notable uptake was detected with the 6 specific antibodies in the adjacent bone (control). CONCLUSIONS The absence of internal fixation after VRO led to dynamic healing with a specific expression pattern of BMP-7 and TGF-β2. The anatomic factors, including sufficient preexisting vascularity, led to the earlier expression pattern of VEGF.
Collapse
Affiliation(s)
- Hwi-Dong Jung
- Assistant Professor, Department of Oral and Maxillofacial Surgery, Oral Science Research Institute, Yonsei University College of Dentistry, Seoul, Korea
| | - Sang Yoon Kim
- Private Practice, McLean, VA; Former Resident, Harvard Oral and Maxillofacial Surgery, Boston, MA
| | - Han-Sung Jung
- Professor, Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea
| | - Hyung-Sik Park
- Professor, Department of Oral and Maxillofacial Surgery, Oral Science Research Institute, Yonsei University College of Dentistry, Seoul, Korea
| | - Young-Soo Jung
- Emeritus Professor, Department of Oral and Maxillofacial Surgery, Oral Science Research Institute, Yonsei University College of Dentistry, Seoul, Korea.
| |
Collapse
|
25
|
Osseoconductive and Corrosion-Inhibiting Plasma-Sprayed Calcium Phosphate Coatings for Metallic Medical Implants. METALS 2017. [DOI: 10.3390/met7110468] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
26
|
The Elution Kinetics of BMP-2, BMP-4, and BMP-7 From a Commercial Human Demineralized Bone Matrix Putty. J Craniofac Surg 2017; 28:2183-2188. [DOI: 10.1097/scs.0000000000004016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
27
|
Lykissas M, Gkiatas I. Use of recombinant human bone morphogenetic protein-2 in spine surgery. World J Orthop 2017; 8:531-535. [PMID: 28808623 PMCID: PMC5534401 DOI: 10.5312/wjo.v8.i7.531] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 02/05/2017] [Accepted: 04/20/2017] [Indexed: 02/06/2023] Open
Abstract
Bone morphogenetic proteins are osteoinductive factors which have gained popularity in orthopaedic surgery and especially in spine surgery. The use of recombinant human bone morphogenetic protein-2 has been officially approved by the United States Food and Drug Administration only for single level anterior lumbar interbody fusion, nevertheless it is widely used by many surgeons with off-label indications. Despite advantages in bone formation, its use still remains a controversial issue and several complications have been described by authors who oppose their wide use.
Collapse
|
28
|
Lee CS, Bishop ES, Zhang R, Yu X, Farina EM, Yan S, Zhao C, Zeng Z, Shu Y, Wu X, Lei J, Li Y, Zhang W, Yang C, Wu K, Wu Y, Ho S, Athiviraham A, Lee MJ, Wolf JM, Reid RR, He TC. Adenovirus-Mediated Gene Delivery: Potential Applications for Gene and Cell-Based Therapies in the New Era of Personalized Medicine. Genes Dis 2017; 4:43-63. [PMID: 28944281 PMCID: PMC5609467 DOI: 10.1016/j.gendis.2017.04.001] [Citation(s) in RCA: 450] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 04/19/2017] [Indexed: 12/12/2022] Open
Abstract
With rapid advances in understanding molecular pathogenesis of human diseases in the era of genome sciences and systems biology, it is anticipated that increasing numbers of therapeutic genes or targets will become available for targeted therapies. Despite numerous setbacks, efficacious gene and/or cell-based therapies still hold the great promise to revolutionize the clinical management of human diseases. It is wildly recognized that poor gene delivery is the limiting factor for most in vivo gene therapies. There has been a long-lasting interest in using viral vectors, especially adenoviral vectors, to deliver therapeutic genes for the past two decades. Among all currently available viral vectors, adenovirus is the most efficient gene delivery system in a broad range of cell and tissue types. The applications of adenoviral vectors in gene delivery have greatly increased in number and efficiency since their initial development. In fact, among over 2,000 gene therapy clinical trials approved worldwide since 1989, a significant portion of the trials have utilized adenoviral vectors. This review aims to provide a comprehensive overview on the characteristics of adenoviral vectors, including adenoviral biology, approaches to engineering adenoviral vectors, and their applications in clinical and pre-clinical studies with an emphasis in the areas of cancer treatment, vaccination and regenerative medicine. Current challenges and future directions regarding the use of adenoviral vectors are also discussed. It is expected that the continued improvements in adenoviral vectors should provide great opportunities for cell and gene therapies to live up to its enormous potential in personalized medicine.
Collapse
Affiliation(s)
- Cody S. Lee
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Elliot S. Bishop
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Ruyi Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Xinyi Yu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Evan M. Farina
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Shujuan Yan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Chen Zhao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Zongyue Zeng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Yi Shu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Xingye Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Jiayan Lei
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Yasha Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Wenwen Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Laboratory Medicine and Clinical Diagnostics, The Affiliated Yantai Hospital, Binzhou Medical University, Yantai 264100, China
| | - Chao Yang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Ke Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Ying Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Immunology and Microbiology, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Sherwin Ho
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Aravind Athiviraham
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jennifer Moriatis Wolf
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Russell R. Reid
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| |
Collapse
|
29
|
Histologic and Histomorphometric Comparison of Bone Regeneration Between Bone Morphogenetic Protein-2 and Platelet-Derived Growth Factor-BB in Experimental Groups. J Craniofac Surg 2017; 27:805-9. [PMID: 27092911 DOI: 10.1097/scs.0000000000002560] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Efficacy of recombinant human bone morphogenetic protein-2 (rhBMP-2) and recombinant human platelet-derived growth factor-BB (rhPDGF-BB) delivered via absorbable collagen sponge (ACS) on bone formation was evaluated in guinea pig tibias. Three-millimeter-circular bone tibia defects were created in 24 guinea pigs assigned randomly to 4 groups according to the following defect filling materials: ACS only, rhBMP-2+ACS, rhPDGF-BB+ACS, or empty. New bone formation was evaluated histologically and histomorphometrically at 15 (early healing) and 45 days (late healing). Mean new bone per total defect area ratio was 0.73, 0.57, 0.43, and 0.42 in rhBMP-2+ACS, rhPDGF-BB+ACS, ACS only, and empty groups at early healing, respectively. During early healing, significantly more new bone formation was observed in rhBMP-2+ACS and rhPDGF-BB+ACS groups than in the control groups. New bone formation was significantly higher with rhBMP-2+ACS than with rhPDGF-BB+ACS. Mean new bone per total defect area ratio was 0.81, 0.86, 0.74, and 0.75 in the rhBMP-2+ACS, rhPDGF-BB+ACS, ACS only, and empty groups at late healing, respectively. During late healing, new bone formation was significantly higher in the rhPDGF-BB+ACS group relative to both control groups, but the results did not differ significantly from those in the rhBMP-2+ACS group. New bone formation in the rhBMP-2+ACS group did not change significantly between the healing periods. In the rhPDGF-BB+ACS group, however, new bone formation was significantly higher in the late healing period. Both growth factors accelerated new bone formation in the early healing period. Although rhBMP-2 was more effective in the early healing period, the effects of rhPDGF-BB were longer lasting.
Collapse
|
30
|
Stavropoulos A, Sculean A, Bosshardt DD, Buser D, Klinge B. Pre-clinical in vivo models for the screening of bone biomaterials for oral/craniofacial indications: focus on small-animal models. Periodontol 2000 2017; 68:55-65. [PMID: 25867979 DOI: 10.1111/prd.12065] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2014] [Indexed: 11/26/2022]
Abstract
Preclinical in vivo experimental studies are performed for evaluating proof-of-principle concepts, safety and possible unwanted reactions of candidate bone biomaterials before proceeding to clinical testing. Specifically, models involving small animals have been developed for screening bone biomaterials for their potential to enhance bone formation. No single model can completely recreate the anatomic, physiologic, biomechanic and functional environment of the human mouth and jaws. Relevant aspects regarding physiology, anatomy, dimensions and handling are discussed in this paper to elucidate the advantages and disadvantages of small-animal models. Model selection should be based not on the 'expertise' or capacities of the team, but rather on a scientifically solid rationale, and the animal model selected should reflect the question for which an answer is sought. The rationale for using heterotopic or orthotopic testing sites, and intraosseous, periosseous or extraskeletal defect models, is discussed. The paper also discusses the relevance of critical size defect modeling, with focus on calvarial defects in rodents. In addition, the rabbit sinus model and the capsule model in the rat mandible are presented and discussed in detail. All animal experiments should be designed with care and include sample-size and study-power calculations, thus allowing generation of meaningful data. Moreover, animal experiments are subject to ethical approval by the relevant authority. All procedures and the postoperative handling and care, including postoperative analgesics, should follow best practice.
Collapse
|
31
|
Landis WJ, Chubinskaya S, Tokui T, Wada Y, Isogai N, Jacquet R. Tissue engineering a human phalanx. J Tissue Eng Regen Med 2016; 11:2373-2387. [PMID: 26999523 DOI: 10.1002/term.2137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 11/11/2015] [Accepted: 12/10/2015] [Indexed: 12/31/2022]
Abstract
A principal purpose of tissue engineering is the augmentation, repair or replacement of diseased or injured human tissue. This study was undertaken to determine whether human biopsies as a cell source could be utilized for successful engineering of human phalanges consisting of both bone and cartilage. This paper reports the use of cadaveric human chondrocytes and periosteum as a model for the development of phalanx constructs. Two factors, osteogenic protein-1 [OP-1/bone morphogenetic protein-7 (BMP7)], alone or combined with insulin-like growth factor (IGF-1), were examined for their potential enhancement of chondrocytes and their secreted extracellular matrices. Design of the study included culture of chondrocytes and periosteum on biodegradable polyglycolic acid (PGA) and poly-l-lactic acid (PLLA)-poly-ε-caprolactone (PCL) scaffolds and subsequent implantation in athymic nu/nu (nude) mice for 5, 20, 40 and 60 weeks. Engineered constructs retrieved from mice were characterized with regard to genotype and phenotype as a function of developmental (implantation) time. Assessments included gross observation, X-ray radiography or microcomputed tomography, histology and gene expression. The resulting data showed that human cell-scaffold constructs could be successfully developed over 60 weeks, despite variability in donor age. Cartilage formation of the distal phalanx models enhanced with both OP-1 and IGF-1 yielded more cells and extracellular matrix (collagen and proteoglycans) than control chondrocytes without added factors. Summary data demonstrated that human distal phalanx models utilizing cadaveric chondrocytes and periosteum were successfully fabricated and OP-1 and OP-1/IGF-1 accelerated construct development and mineralization. The results suggest that similar engineering and transplantation of human autologous tissues in patients are clinically feasible. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- W J Landis
- Goodyear Polymer Center, Department of Polymer Science, University of Akron, Akron, OH, USA
| | - S Chubinskaya
- Departments of Biochemistry, Orthopaedic Surgery and Medicine, Rush University Medical Center, Chicago, IL, USA
| | - T Tokui
- Department of Plastic and Reconstructive Surgery, Kinki University Medical School, Osaka-Sayama, Japan
| | - Y Wada
- Department of Plastic and Reconstructive Surgery, Kinki University Medical School, Osaka-Sayama, Japan
| | - N Isogai
- Department of Plastic and Reconstructive Surgery, Kinki University Medical School, Osaka-Sayama, Japan
| | - R Jacquet
- Goodyear Polymer Center, Department of Polymer Science, University of Akron, Akron, OH, USA
| |
Collapse
|
32
|
Waki T, Lee SY, Niikura T, Iwakura T, Dogaki Y, Okumachi E, Oe K, Kuroda R, Kurosaka M. Profiling microRNA expression during fracture healing. BMC Musculoskelet Disord 2016; 17:83. [PMID: 26879131 PMCID: PMC4754871 DOI: 10.1186/s12891-016-0931-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 02/06/2016] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND The discovery of microRNA (miRNA) has revealed a novel type of regulatory control for gene expression. Increasing evidence suggests that miRNA regulates chondrocyte, osteoblast, and osteoclast differentiation and function, indicating miRNA as key regulators of bone formation, resorption, remodeling, and repair. We hypothesized that the functions of certain miRNAs and changes to their expression pattern may play crucial roles during the process of fracture healing. METHODS Standard healing fractures and unhealing fractures produced by periosteal cauterization at the fracture site were created in femurs of seventy rats, with half assigned to the standard healing fracture group and half assigned to the nonunion group. At post-fracture days 3, 7, 10, 14, 21, and 28, total RNA including miRNA was extracted from the newly generated tissue at the fracture site. Microarray analysis was performed with miRNA samples from each group on post-fracture day 14. For further analysis, we selected highly up-regulated five miRNAs in the standard healing fracture group from the microarray data. Real-time PCR was performed with miRNA samples at each time point above mentioned to compare the expression levels of the selected miRNAs between standard healing fractures and unhealing fractures and investigate their time-course changes. RESULTS Microarray and real-time polymerase chain reaction (PCR) analyses on day 14 revealed that five miRNAs, miR-140-3p, miR-140-5p, miR-181a-5p, miR-181d-5p, and miR-451a, were significantly highly expressed in standard healing fractures compared with unhealing fractures. Real-time PCR analysis further revealed that in standard healing fractures, the expression of all five of these miRNAs peaked on day 14 and declined thereafter. CONCLUSION Our results suggest that the five miRNAs identified using microarray and real-time PCR analyses may play important roles during fracture healing. These findings provide valuable information to further understand the molecular mechanism of fracture healing and may lead to the development of miRNA-based tissue engineering strategies to promote fracture healing.
Collapse
Affiliation(s)
- Takahiro Waki
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Sang Yang Lee
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Takahiro Niikura
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Takashi Iwakura
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Yoshihiro Dogaki
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Etsuko Okumachi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Keisuke Oe
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Masahiro Kurosaka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| |
Collapse
|
33
|
Li R, Yan Z, Ye J, Huang H, Wang Z, Wei Q, Wang J, Zhao L, Lu S, Wang X, Tang S, Fan J, Zhang F, Zou Y, Song D, Liao J, Lu M, Liu F, Shi LL, Athiviraham A, Lee MJ, He TC, Zhang Z. The Prodomain-Containing BMP9 Produced from a Stable Line Effectively Regulates the Differentiation of Mesenchymal Stem Cells. Int J Med Sci 2016; 13:8-18. [PMID: 26816490 PMCID: PMC4716815 DOI: 10.7150/ijms.13333] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 11/09/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND BMPs play important roles in regulating stem cell proliferation and differentiation. Using adenovirus-mediated expression of the 14 types of BMPs we demonstrated that BMP9 is one of the most potent BMPs in inducing osteogenic differentiation of mesenchymal stem cells (MSCs), which was undetected in the early studies using recombinant BMP9 proteins. Endogenous BMPs are expressed as a precursor protein that contains an N-terminal signal peptide, a prodomain and a C-terminal mature peptide. Most commercially available recombinant BMP9 proteins are purified from the cells expressing the mature peptide. It is unclear how effectively these recombinant BMP9 proteins functionally recapitulate endogenous BMP9. METHODS A stable cell line expressing the full coding region of mouse BMP9 was established in HEK-293 cells by using the piggyBac transposon system. The biological activities and stability of the conditioned medium generated from the stable line were analyzed. RESULTS The stable HEK-293 line expresses a high level of mouse BMP9. BMP9 conditioned medium (BMP9-cm) was shown to effectively induce osteogenic differentiation of MSCs, to activate BMP-R specific Smad signaling, and to up-regulate downstream target genes in MSCs. The biological activity of BMP9-cm is at least comparable with that induced by AdBMP9 in vitro. Furthermore, BMP9-cm exhibits an excellent stability profile as its biological activity is not affected by long-term storage at -80ºC, repeated thawing cycles, and extended storage at 4ºC. CONCLUSIONS We have established a producer line that stably expresses a high level of active BMP9 protein. Such producer line should be a valuable resource for generating biologically active BMP9 protein for studying BMP9 signaling mechanism and functions.
Collapse
Affiliation(s)
- Ruifang Li
- 1. Department of Neurology, Hubei Zhongshan Hospital, Wuhan, China; 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Zhengjian Yan
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 3. Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Jixing Ye
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 4. Department of Biomedical Engineering, School of Bioengineering, Chongqing University, Chongqing, China
| | - He Huang
- 5. Ben May Department for Cancer Research, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Zhongliang Wang
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 3. Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Qiang Wei
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 3. Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Jing Wang
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 3. Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Lianggong Zhao
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 6. Department of Orthopaedic Surgery, the Second Affiliated Hospital of Lanzhou University, Lanzhou, China
| | - Shun Lu
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 7. Department of Orthopaedic Surgery, Shandong Provincial Hospital and Shandong University School of Medicine, Jinan, China
| | - Xin Wang
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 8. Department of Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Shengli Tang
- 1. Department of Neurology, Hubei Zhongshan Hospital, Wuhan, China; 9. Department of General Surgery, the Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiaming Fan
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 3. Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Fugui Zhang
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 3. Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Yulong Zou
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 3. Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Dongzhe Song
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 8. Department of Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Junyi Liao
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 3. Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Minpeng Lu
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 3. Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Feng Liu
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 3. Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Lewis L Shi
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Aravind Athiviraham
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Michael J Lee
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Tong-Chuan He
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Zhonglin Zhang
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 9. Department of General Surgery, the Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
34
|
Khallaf FG, Kehinde EO, Hussein S. Bone Healing and Hormonal Bioassay in Patients with Long-Bone Fractures and Concomitant Head Injury. Med Princ Pract 2016; 25:336-42. [PMID: 26954461 PMCID: PMC5588422 DOI: 10.1159/000445250] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 03/07/2016] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE The aim of this study is to investigate healing of fractures in patients with concomitant head injuries and to measure blood hormone levels to elucidate the mechanism of a possible accelerated osteogenesis. MATERIALS AND METHODS One hundred and sixty-two patients were included in this study and divided into 3 cohorts: group A with head injuries only (n = 52); group B with head injuries as well as long-bone fractures (n = 50); group C with long-bone fractures only (n = 60). Fracture-healing parameters including time of appearance and thickness of the bridging callus, and blood hormonal assays were measured and compared using Student's t test. RESULTS The mean time to healing was significantly lower in cohort B (6.9 ± 2.9 weeks) than C (22.4 ± 8.7 weeks; p = 0.001). The mean thickness of the healing callus was significantly higher in cohort B (26.3 ± 9.7 mm) than C (8.1 ± 5.9 mm; p = 0.002). The mean healing rate was also higher in cohort B (4.5 ± 2.3 mm/week) than C (0.38 ± 0.21 mm/week; p = 0.001). Blood hormonal assays in group B showed higher values of parathyroid hormone and growth hormone than in group C. However, adrenaline and noradrenaline values were lower in group B than in group C at all measured time intervals, and correspondingly leptin was lower in all groups (p = 0.001). Corticosteroid values were normal in group B compared to slightly higher values in group C, also at all measured time intervals. CONCLUSION In this study, healing of fractures in patients with concomitant head injuries was accelerated, thereby indicating an involvement of a combined neurohormonal mechanism.
Collapse
Affiliation(s)
- Fathy G. Khallaf
- Department of Orthopaedic Surgery, Jahra Hospital, Kuwait, Jabriya
- *Dr. Fathy G. Khallaf, FRCS Glasgow, Department of Orthopaedic Surgery, Jahra Hospital, Ministry of Health, Kuwait (Kuwait), E-Mail
| | - Elijah O. Kehinde
- Department of Surgery, Faculty of Medicine, Kuwait University, Jabriya, Kuwait
| | - Sundus Hussein
- Department of Pathology, Mubarak Al-Kabeer Hospital, Jabriya, Kuwait
| |
Collapse
|
35
|
Reduction of Adipose Tissue Formation by the Controlled Release of BMP-2 Using a Hydroxyapatite-Coated Collagen Carrier System for Sinus-Augmentation/Extraction-Socket Grafting. MATERIALS 2015; 8:7634-7649. [PMID: 28793666 PMCID: PMC5458903 DOI: 10.3390/ma8115411] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 10/29/2015] [Accepted: 11/04/2015] [Indexed: 11/28/2022]
Abstract
The effects of hydroxyapatite (HA)-coating onto collagen carriers for application of recombinant human bone morphogenetic protein 2 (rhBMP-2) on cell differentiation in vitro, and on in vivo healing patterns after sinus-augmentation and alveolar socket-grafting were evaluated. In vitro induction of osteogenic/adipogenic differentiation was compared between the culture media with rhBMP-2 solution and with the released rhBMP-2 from the control collagen and from the HA-coated collagen. Demineralized bovine bone and collagen/HA-coated collagen were grafted with/without rhBMP-2 in sinus-augmentation and tooth-extraction-socket models. Adipogenic induction by rhBMP-2 released from HA-coated collagen was significantly reduced compared to collagen. In the sinus-augmentation model, sites that received rhBMP-2 exhibited large amounts of vascular tissue formation at two weeks and increased adipose tissue formation at eight weeks; this could be significantly reduced by using HA-coated collagen as a carrier for rhBMP-2. In extraction-socket grafting, dimensional reduction of alveolar ridge was significantly decreased at sites received rhBMP-2 compared to control sites, but adipose tissue was increased within the regenerated socket area. In conclusion, HA-coated collagen carrier for Escherichia coli-derived rhBMP-2 (ErhBMP-2) may reduce in vitro induction of adipogenic differentiation and in vivo adipose bone marrow tissue formation in bone tissue engineering by ErhBMP-2.
Collapse
|
36
|
Effects of low-level laser therapy on the expression of osteogenic genes during the initial stages of bone healing in rats: a microarray analysis. Lasers Med Sci 2015; 30:2325-33. [DOI: 10.1007/s10103-015-1807-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 09/10/2015] [Indexed: 12/22/2022]
|
37
|
Sun H, Yang HL. Calcium phosphate scaffolds combined with bone morphogenetic proteins or mesenchymal stem cells in bone tissue engineering. Chin Med J (Engl) 2015; 128:1121-7. [PMID: 25881610 PMCID: PMC4832956 DOI: 10.4103/0366-6999.155121] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Objective: The purpose of this study was to review the current status of calcium phosphate (CaP) scaffolds combined with bone morphogenetic proteins (BMPs) or mesenchymal stem cells (MSCs) in the field of bone tissue engineering (BTE). Date Sources: Data cited in this review were obtained primarily from PubMed and Medline in publications from 1979 to 2014, with highly regarded older publications also included. The terms BTE, CaP, BMPs, and MSC were used for the literature search. Study Selection: Reviews focused on relevant aspects and original articles reporting in vitro and/or in vivo results concerning the efficiency of CaP/BMPs or CaP/MSCs composites were retrieved, reviewed, analyzed, and summarized. Results: An ideal BTE product contains three elements: Scaffold, growth factors, and stem cells. CaP-based scaffolds are popular because of their outstanding biocompatibility, bioactivity, and osteoconductivity. However, they lack stiffness and osteoinductivity. To solve this problem, composite scaffolds of CaP with BMPs have been developed. New bone formation by CaP/BMP composites can reach levels similar to those of autografts. CaP scaffolds are compatible with MSCs and CaP/MSC composites exhibit excellent osteogenesis and stiffness. In addition, a CaP/MSC/BMP scaffold can repair bone defects more effectively than an autograft. Conclusions: Novel BTE products possess remarkable osteoconduction and osteoinduction capacities, and exhibit balanced degradation with osteogenesis. Further work should yield safe, viable, and efficient materials for the repair of bone lesions.
Collapse
Affiliation(s)
| | - Hui-Lin Yang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Jiangsu 215006, China
| |
Collapse
|
38
|
Houdek MT, Wyles CC, Sierra RJ. Osteonecrosis of the femoral head: treatment with ancillary growth factors. Curr Rev Musculoskelet Med 2015; 8:233-9. [PMID: 25985987 PMCID: PMC4596200 DOI: 10.1007/s12178-015-9281-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Osteonecrosis (ON) of the femoral head, also known as avascular necrosis (AVN) of the femoral head, is a progressive disease that predominantly affects younger patients. During early stage of ON, decompression of the femoral head has been commonly used to improve pain. The decompression has been augmented with nonvascularized or vascularized bone grafts, mesenchymal stems cells, and growth factors. The use of adjuvant growth factors to supplement the core decompression has mainly been limited to animal models in an attempt to regenerate the necrotic lesion of ON. Factors utilized include bone morphogenetic proteins, vascular endothelial growth factors, hepatocyte growth factors, fibroblast growth factors, granulocyte colony-stimulating factors, and stem cells factors. In animal models, the use of these factors has been shown to increase bone formation and angiogenesis. Although promising, the use of these growth factors and cell-based therapies clinically remains limited.
Collapse
Affiliation(s)
- Matthew T. Houdek
- />Department of Orthopedic Surgery, Mayo Clinic, 200 First St. SW, Rochester, MN 55905 USA
| | - Cody C. Wyles
- />Mayo Clinic Medical School, 200 First St. SW, Rochester, MN 55909 USA
| | - Rafael J. Sierra
- />Department of Orthopedic Surgery, Mayo Clinic, 200 First St. SW, Rochester, MN 55905 USA
| |
Collapse
|
39
|
Visser R, Bodnarova K, Arrabal PM, Cifuentes M, Becerra J. Combining bone morphogenetic proteins-2 and -6 has additive effects on osteoblastic differentiation in vitro and accelerates bone formation in vivo. J Biomed Mater Res A 2015; 104:178-85. [PMID: 26238281 DOI: 10.1002/jbm.a.35546] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 07/16/2015] [Accepted: 07/31/2015] [Indexed: 11/11/2022]
Abstract
While only two members of the bone morphogenetic protein subfamily (BMP-2 and -7) are approved to be used in combination with collagen type I in orthopaedic surgery, other BMPs are known to also be highly osteoinductive. Although all the osteogenic BMPs signal through Smad-1/-5/-8 phosphorylation, they show different preferences for the available BMP receptors. In this work we studied the effect of combining two osteogenic BMPs (-2 and -6), which belong to different groups within the subfamily and have different affinities to the existing BMP receptors. Both the growth and in vitro differentiation of MC3T3-E1 mouse preosteoblasts and rat bone marrow-derived mesenchymal stem cells (MSCs) were studied, as well as in vivo ectopic bone formation when the BMPs were intramuscularly implanted in rats with collagen type I sponges as carriers. The results show that these two growth factors have additive effects on the osteoblastic differentiation of cells in vitro and that their combination might be helpful to accelerate in vivo osteogenesis while reducing the amount of each individual BMP used.
Collapse
Affiliation(s)
- Rick Visser
- Department of Cell Biology, Genetics and Physiology, University of Malaga, IBIMA, BIONAND, Andalusian Center for Nanomedicine and Biotechnology, Malaga, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Malaga, Spain
| | - Kristina Bodnarova
- Department of Genetics, Faculty of Natural Sciences, Comenius University, Mlynska Dolina, 842 15 Bratislava, Slovakia
| | - Pilar M Arrabal
- Department of Cell Biology, Genetics and Physiology, University of Malaga, IBIMA, BIONAND, Andalusian Center for Nanomedicine and Biotechnology, Malaga, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Malaga, Spain
| | - Manuel Cifuentes
- Department of Cell Biology, Genetics and Physiology, University of Malaga, IBIMA, BIONAND, Andalusian Center for Nanomedicine and Biotechnology, Malaga, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Malaga, Spain
| | - Jose Becerra
- Department of Cell Biology, Genetics and Physiology, University of Malaga, IBIMA, BIONAND, Andalusian Center for Nanomedicine and Biotechnology, Malaga, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Malaga, Spain
| |
Collapse
|
40
|
Huang W, Hasegawa T, Imai Y, Takeda D, Akashi M, Komori T. Low-intensity pulsed ultrasound enhances bone morphogenetic protein expression of human mandibular fracture haematoma-derived cells. Int J Oral Maxillofac Surg 2015; 44:929-35. [DOI: 10.1016/j.ijom.2015.03.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 02/16/2015] [Accepted: 03/02/2015] [Indexed: 01/28/2023]
|
41
|
Ollivier M, Gay AM, Cerlier A, Lunebourg A, Argenson JN, Parratte S. Can we achieve bone healing using the diamond concept without bone grafting for recalcitrant tibial nonunions? Injury 2015; 46:1383-8. [PMID: 25933808 DOI: 10.1016/j.injury.2015.03.036] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 03/17/2015] [Accepted: 03/30/2015] [Indexed: 02/02/2023]
Abstract
The purpose of this study was to evaluate the efficacy and safety of a combination of recombinant human bone morphogenetic protein 7 (rhBMP-7) and resorbable calcium phosphate bone substitute (rCPBS) as a salvage solution for recalcitrant tibial fracture nonunions. Twenty consecutive patients, 16 male and four female, with a mean age of 46.8±15.7 years (21-78) and a mean body mass index (BMI) of 24.2±5.3kgm(-2) (21.5-28.5), suffering from 20 recalcitrant tibial fracture nonunions were included. The mean number of operations performed prior to the procedure was 3.3, with homolateral iliac crest bone grafts being used for all of the patients. All patients were treated with a procedure including debridement and decortications of the bone ends, nonunion fixation with a locking plate, and filling of the bony defect with a combined graft of rhBMP-7 (as osteoinductor) with an rCPBS (as scaffold). The mean follow-up was 14±2.7 months. Both clinical and radiological union occurred in 18 cases, within a mean time of 4.7±3.2 months. A recurrence of deep infection was diagnosed for one of the non-consolidated patients. No specific complication of rCPBS or rhBMP-7 was encountered. This study supports the view that the application of rCPBS combined with rhBMP-7, without any bone grafting, is safe and efficient in the treatment of recalcitrant bone union.
Collapse
Affiliation(s)
- M Ollivier
- Department of Orthopedic Surgery, Institute of Movement, UMR 7287: Aix-Marseille University, 270 Boulevard Sainte Marguerite, BP 29, 13274 Marseille, France.
| | - A M Gay
- Department of Hand Surgery and Limb Reconstruction, Aix-Marseille University, Timone Hospital, 264 Rue Saint Pierre, 13385 Marseille, France
| | - A Cerlier
- Department of Hand Surgery and Limb Reconstruction, Aix-Marseille University, Timone Hospital, 264 Rue Saint Pierre, 13385 Marseille, France
| | - A Lunebourg
- Department of Orthopedic Surgery, Institute of Movement, UMR 7287: Aix-Marseille University, 270 Boulevard Sainte Marguerite, BP 29, 13274 Marseille, France
| | - J N Argenson
- Department of Orthopedic Surgery, Institute of Movement, UMR 7287: Aix-Marseille University, 270 Boulevard Sainte Marguerite, BP 29, 13274 Marseille, France
| | - S Parratte
- Department of Orthopedic Surgery, Institute of Movement, UMR 7287: Aix-Marseille University, 270 Boulevard Sainte Marguerite, BP 29, 13274 Marseille, France
| |
Collapse
|
42
|
Sung NY, Choi JI. Enhanced release of bone morphogenetic proteins from demineralized bone matrix by gamma irradiation. Radiat Phys Chem Oxf Engl 1993 2015. [DOI: 10.1016/j.radphyschem.2015.02.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
43
|
Wang L, Park P, La Marca F, Than KD, Lin CY. BMP-2 inhibits tumor-initiating ability in human renal cancer stem cells and induces bone formation. J Cancer Res Clin Oncol 2015; 141:1013-24. [PMID: 25431339 DOI: 10.1007/s00432-014-1883-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 11/22/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE We have previously shown that BMP-2 induces bone formation and inhibits tumorigenicity of cancer stem cells (CSCs) in a human osteosarcoma OS99-1 cell line. In this study, we sought to determine whether BMP-2 can similarly induce bone formation and inhibit the tumorigenicity of renal CSCs identified based on aldehyde dehydrogenase (ALDH) activity in renal cell carcinoma (RCC) cell lines and primary tumors. METHODS Using a xenograft model in which cells from human RCC cell lines ACHN, Caki-2, and primary tumors were grown in NOD/SCID mice, renal CSCs were identified as a subset of ALDH(br) cells. The ALDH(br) cells possessed a greater colony-forming efficiency, higher proliferative output, increased expression of stem cell marker genes Oct3/4A, Nanog, renal embryonic marker Pax-2, and greater tumorigenicity compared to cells with low ALDH activity (ALDH(lo) cells), generating new tumors with as few as 25 cells in mice. RESULTS In vitro, BMP-2 was found to inhibit the ALDH(br) cell growth, down-regulate the expression of embryonic stem cell markers, and up-regulate the transcription of osteogenic markers. In vivo, all animals receiving a low number of ALDH(br) cells (5 × 10(3)) from ACHN, Caki-2, and primary tumor xenografts treated with 30 µg BMP-2 per animal showed limited tumor growth with significant bone formation, while untreated cells developed large tumor masses without bone formation. CONCLUSIONS These results suggest that BMP-2 inhibits the tumor-initiating ability of renal CSCs and induces osseous bone formation. BMP-2 may therefore provide a beneficial strategy for human RCC treatment by targeting the CSC-enriched population.
Collapse
Affiliation(s)
- Lin Wang
- Spine Research Laboratory, Department of Neurosurgery, University of Michigan Medical School, 1500 E. Medical Center Drive, Room 3552 TC, Ann Arbor, MI, 48109-5338, USA
| | | | | | | | | |
Collapse
|
44
|
Takahashi K, Ogura N, Tomoki R, Eda T, Okada H, Kato R, Iwai S, Ito K, Kuyama K, Kondoh T. Applicability of human dental follicle cells to bone regeneration without dexamethasone: an in vivo pilot study. Int J Oral Maxillofac Surg 2015; 44:664-9. [DOI: 10.1016/j.ijom.2014.11.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 10/27/2014] [Accepted: 11/07/2014] [Indexed: 11/24/2022]
|
45
|
Wehland M, Aleshcheva G, Schulz H, Saar K, Hübner N, Hemmersbach R, Braun M, Ma X, Frett T, Warnke E, Riwaldt S, Pietsch J, Corydon TJ, Infanger M, Grimm D. Differential gene expression of human chondrocytes cultured under short-term altered gravity conditions during parabolic flight maneuvers. Cell Commun Signal 2015; 13:18. [PMID: 25889719 PMCID: PMC4369370 DOI: 10.1186/s12964-015-0095-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 03/02/2015] [Indexed: 01/06/2023] Open
Abstract
Background Chondrocytes are the main cellular component of articular cartilage. In healthy tissue, they are embedded in a strong but elastic extracelluar matrix providing resistance against mechanical forces and friction for the joints. Osteoarthritic cartilage, however, disrupted by heavy strain, has only very limited potential to heal. One future possibility to replace damaged cartilage might be the scaffold-free growth of chondrocytes in microgravity to form 3D aggregates. Results To prepare for this, we have conducted experiments during the 20th DLR parabolic flight campaign, where we fixed the cells after the first (1P) and the 31st parabola (31P). Furthermore, we subjected chondrocytes to isolated vibration and hypergravity conditions. Microarray and quantitative real time PCR analyses revealed that hypergravity regulated genes connected to cartilage integrity (BMP4, MMP3, MMP10, EDN1, WNT5A, BIRC3). Vibration was clearly detrimental to cartilage (upregulated inflammatory IL6 and IL8, downregulated growth factors EGF, VEGF, FGF17). The viability of the cells was not affected by the parabolic flight, but showed a significantly increased expression of anti-apoptotic genes after 31 parabolas. The IL-6 release of chondrocytes cultured under conditions of vibration was not changed, but hypergravity (1.8 g) induced a clear elevation of IL-6 protein in the supernatant compared with corresponding control samples. Conclusion Taken together, this study provided new insights into the growth behavior of chondrocytes under short-term microgravity. Electronic supplementary material The online version of this article (doi:10.1186/s12964-015-0095-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Markus Wehland
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke University, 39120, Magdeburg, Germany.
| | - Ganna Aleshcheva
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke University, 39120, Magdeburg, Germany.
| | - Herbert Schulz
- Max-Delbrück-Center for Molecular Medicine, 13092, Berlin, Germany. .,University of Cologne, Cologne Center for Genomics (CCG), 50931, Cologne, Germany.
| | - Katrin Saar
- Max-Delbrück-Center for Molecular Medicine, 13092, Berlin, Germany.
| | - Norbert Hübner
- Max-Delbrück-Center for Molecular Medicine, 13092, Berlin, Germany.
| | - Ruth Hemmersbach
- DLR German Aerospace Center, Biomedical Research, Gravitational Biology, 51147, Köln, Germany.
| | - Markus Braun
- Institute for Molecular Physiology and Biotechnology of Plants (IMBIO), University of Bonn, Gravitational Biology Group, 53115, Bonn, Germany.
| | - Xiao Ma
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, DK-8000, Aarhus C, Denmark.
| | - Timo Frett
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Biomedical Research, 51147, Köln, Germany.
| | - Elisabeth Warnke
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke University, 39120, Magdeburg, Germany.
| | - Stefan Riwaldt
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke University, 39120, Magdeburg, Germany.
| | - Jessica Pietsch
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke University, 39120, Magdeburg, Germany.
| | - Thomas Juhl Corydon
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, DK-8000, Aarhus C, Denmark.
| | - Manfred Infanger
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke University, 39120, Magdeburg, Germany.
| | - Daniela Grimm
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, DK-8000, Aarhus C, Denmark.
| |
Collapse
|
46
|
Filho GS, Caballé-Serrano J, Sawada K, Bosshardt DD, Bianchini MA, Buser D, Gruber R. Conditioned medium of demineralized freeze-dried bone activates gene expression in periodontal fibroblasts in vitro. J Periodontol 2015; 86:827-34. [PMID: 25786563 DOI: 10.1902/jop.2015.140676] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Demineralized bone matrix (DBM) is used for the treatment of osseous defects. Conditioned medium from native bone chips can activate transforming growth factor (TGF)-β signaling in mesenchymal cells. The aim of this study is to determine whether processing of native bone into DBM affects the activity of the conditioned medium. METHODS Porcine cortical bone blocks were subjected to defatting, different concentrations of hydrochloric acid, and various temperatures. DBM was lyophilized, ground, and placed into culture medium. Human gingiva and periodontal fibroblasts were exposed to the respective conditioned medium obtained from DBM (DBCM). Changes in the expression of TGF-β target genes were determined. RESULTS DBCM altered the expression of TGF-β target genes (e.g., adrenomedullin, pentraxin 3, KN motif and ankyrin repeat domains 4, interleukin 11, NADPH oxidase 4, and BTB [POZ] domain containing 11) by at least five-fold. The response was observed in fibroblasts from both sources. Defatting lowered the activity of DBCM. The TGF-β receptor type I kinase inhibitor SB431542 [4-(4-(benzo[d][1,3]dioxol-5-yl)-5-(pyridin-2-yl)-1H-imidazol-2-yl)benzamide] but not the inhibitor of bone morphogenetic protein receptor dorsomorphin, blocked the effects of DBCM on gene expression. Moreover, conditioned medium obtained from commercial human DBM modulated the expression of TGF-β target genes. CONCLUSION The findings suggest that the DBCM can activate TGF-β signaling in oral fibroblasts.
Collapse
Affiliation(s)
- Guenther Schuldt Filho
- *Laboratory of Oral Cell Biology, School of Dental Medicine, University of Bern, Bern, Switzerland.,†Department of Oral Surgery and Stomatology, School of Dental Medicine, University of Bern.,‡Department of Implant Dentistry, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Jordi Caballé-Serrano
- *Laboratory of Oral Cell Biology, School of Dental Medicine, University of Bern, Bern, Switzerland.,†Department of Oral Surgery and Stomatology, School of Dental Medicine, University of Bern.,§Department of Oral and Maxillofacial Surgery, College of Dentistry, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Kosaku Sawada
- *Laboratory of Oral Cell Biology, School of Dental Medicine, University of Bern, Bern, Switzerland.,‖Department of Cranio-Maxillofacial Surgery, Inselspital, University of Bern
| | - Dieter D Bosshardt
- †Department of Oral Surgery and Stomatology, School of Dental Medicine, University of Bern.,¶Robert K. Schenk Laboratory of Oral Histology, School of Dental Medicine, University of Bern
| | - Marco Aurélio Bianchini
- ‡Department of Implant Dentistry, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Daniel Buser
- †Department of Oral Surgery and Stomatology, School of Dental Medicine, University of Bern
| | - Reinhard Gruber
- *Laboratory of Oral Cell Biology, School of Dental Medicine, University of Bern, Bern, Switzerland.,†Department of Oral Surgery and Stomatology, School of Dental Medicine, University of Bern.,#Department of Oral Biology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
47
|
Seib FP, Berry JE, Shiozawa Y, Taichman RS, Kaplan DL. Tissue engineering a surrogate niche for metastatic cancer cells. Biomaterials 2015; 51:313-319. [PMID: 25771021 DOI: 10.1016/j.biomaterials.2015.01.076] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 01/23/2015] [Accepted: 01/27/2015] [Indexed: 12/18/2022]
Abstract
In breast and prostate cancer patients, the bone marrow is a preferred site of metastasis. We hypothesized that we could use tissue-engineering strategies to lure metastasizing cancer cells to tissue-engineered bone marrow. First, we generated highly porous 3D silk scaffolds that were biocompatible and amenable to bone morphogenetic protein 2 functionalization. Control and functionalized silk scaffolds were subcutaneously implanted in mice and bone marrow development was followed. Only functionalized scaffolds developed cancellous bone and red bone marrow, which appeared as early as two weeks post-implantation and further developed over the 16-week study period. This tissue-engineered bone marrow microenvironment could be readily manipulated in situ to understand the biology of bone metastasis. To test the ability of functionalized scaffolds to serve as a surrogate niche for metastasis, human breast cancer cells were injected into the mammary fat pads of mice. The treatment of animals with scaffolds had no significant effect on primary tumor growth. However, extensive metastasis was observed in functionalized scaffolds, and the highest levels for scaffolds that were in situ manipulated with receptor activator of nuclear factor kappa-B ligand (RANKL). We also applied this tissue-engineered bone marrow model in a prostate cancer and experimental metastasis setting. In summary, we were able to use tissue-engineered bone marrow to serve as a target or "trap" for metastasizing cancer cells.
Collapse
Affiliation(s)
- F Philipp Seib
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Janice E Berry
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Yusuke Shiozawa
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Russell S Taichman
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - David L Kaplan
- Tufts University, Department of Biomedical Engineering, 4 Colby Street Medford, MA 02155, USA.
| |
Collapse
|
48
|
Combined use of low-intensity pulsed ultrasound and rhBMP-2 to enhance bone formation in a rat model of critical size defect. J Orthop Trauma 2014; 28:605-11. [PMID: 24464096 PMCID: PMC4108582 DOI: 10.1097/bot.0000000000000067] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Bone repair is regulated by biological factors and the local mechanical environment. We hypothesize that the combined use of low-intensity pulsed ultrasound (LIPUS) and recombinant human bone morphogenetic protein-2 (rhBMP-2) will synergistically or additively enhance bone regeneration in a model simulating the more difficult scenarios in orthopaedic traumatology. METHODS Femoral defects in rats were replaced with absorbable collagen sponges carrying rhBMP-2 (0, 1.2, 6, or 12 μg; n = 30). Each group was divided equally to receive daily treatment of either LIPUS or sham stimulation. At 4 weeks, new bone formation was assessed using quantitative (radiography and microcomputed tomography), qualitative (histology), and functional (biomechanical) end points. RESULTS LIPUS with 1.2 μg of rhBMP-2 significantly improved the radiographic healing as compared with its sham control starting as early as 2 weeks. Quantitatively, the use of LIPUS with 6 μg of rhBMP-2 significantly increased the bone volume. However, using LIPUS with 12 μg of rhBMP-2 indicated a reduction in callus size, without compromising the bone volume, which was also observable histologically, showing organized lamellar bone and repopulated marrow in the original defect region. Histologically, 1.2 μg of rhBMP-2 alone showed the presence of uncalcified cartilage in the defect, which was reduced with LIPUS treatment. Biomechanically, LIPUS treatment significantly increased the peak torsion and stiffness in the 6- and 12 μg rhBMP-2 groups. CONCLUSIONS LIPUS enhances rhBMP-2-induced bone formation at lower doses (1.2 and 6 μg) and callus maturation at 12-μg dose delivered on absorbable collagen sponge for bone repair in a rat critical-sized femoral segmental defect.
Collapse
|
49
|
Kfuri M, de Freitas RL, Batista BB, Salim R, Castiglia MT, Tavares RA, Araújo PH. Updates in biological therapies for knee injuries: bone. Curr Rev Musculoskelet Med 2014; 7:220-7. [PMID: 25030275 PMCID: PMC4596166 DOI: 10.1007/s12178-014-9225-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Bone is a unique tissue because of its mechanical properties, ability for self-repair, and enrollment in different metabolic processes such as calcium homeostasis and hematopoietic cell production. Bone barely tolerates deformation and tends to fail when overloaded. Fracture healing is a complex process that in particular cases is impaired. Osteoprogenitor cells proliferation, growth factors, and a sound tridimensional scaffold at fracture site are key elements for new bone formation and deposition. Mechanical stability and ample vascularity are also of great importance on providing a proper environment for bone healing. From mesenchymal stem cells delivery to custom-made synthetic scaffolds, many are the biological attempts to enhance bone healing. Impaired fracture healing represents a real burden to contemporary society. Sound basic science knowledge has contributed to newer approaches aimed to accelerate and improve the quality of bone healing.
Collapse
Affiliation(s)
- Mauricio Kfuri
- Departamento de Biomecânica, Medicina e Reabilitação do Aparelho Locomotor - Hospital das Clinicas - Campus USP Av. Bandeirantes 3900 - 11o andar, 14048-900, Ribeirão Preto, SP, Brazil,
| | | | | | | | | | | | | |
Collapse
|
50
|
BMP type I receptor inhibition attenuates endothelial dysfunction in mice with chronic kidney disease. Kidney Int 2014; 87:128-36. [PMID: 24963916 DOI: 10.1038/ki.2014.223] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 04/12/2014] [Accepted: 05/08/2014] [Indexed: 01/10/2023]
Abstract
The molecular mechanisms of endothelial dysfunction and vascular calcification have been considered independently and potential links are currently unknown in chronic kidney disease (CKD). Bone morphogenetic protein (BMP) receptor signaling mediates calcification of atherosclerotic plaques. Here we tested whether BMP receptor signaling contributes to endothelial dysfunction, as well as to osteogenic differentiation of vascular smooth muscle cells (VSMCs), in a model of short-term CKD. In C57BL/6 mice, subtotal nephrectomy activated BMP receptor and increased phosphatase-and-tensin homolog (PTEN) protein in the endothelial cells and medial VSMCs without vascular remodeling in the aorta. In the endothelial cells, PTEN induction led to inhibition of the Akt-endothelial nitric oxide synthase (eNOS) pathway and endothelial dysfunction. In VSMCs, the PTEN increase induced early osteogenic differentiation. CKD-induced inhibition of eNOS phosphorylation and the resultant endothelial dysfunction were inhibited in mice with endothelial cell-specific PTEN ablation. Knockout of the BMP type I receptor abolished endothelial dysfunction, the inhibition of eNOS phosphorylation, and VSMC osteogenic differentiation in mice with CKD. A small molecule inhibitor of BMP type I receptor, LDN-193189, prevented endothelial dysfunction and osteogenic differentiation in CKD mice. Thus, BMP receptor activation is a mechanism for endothelial dysfunction in addition to vascular osteogenic differentiation in a short-term CKD model. PTEN may be key in linking BMP receptor activation and endothelial dysfunction in CKD.
Collapse
|