1
|
Wang J, Yang F, Chen R, Yang X, Wang J, Zhang H. Hydrogel Composite Incorporating Deferoxamine-Loaded Gelatin-Based Microspheres Enhance Angiogenesis Ability of Dental Pulp Stem Cells. ACS OMEGA 2025; 10:12579-12589. [PMID: 40191326 PMCID: PMC11966253 DOI: 10.1021/acsomega.5c00445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/03/2025] [Accepted: 03/14/2025] [Indexed: 04/09/2025]
Abstract
Fast reconstruction of the pulpal vasculature is crucial for effective pulp regeneration. Dental pulp stem cells (DPSCs) are promising candidates for pulp regeneration because of their potential for multilineage differentiation and vasculogenic properties. Deferoxamine (DFO) has been shown to stimulate angiogenesis during wound healing and bone regeneration; however, the effects of DFO on the angiogenic potential of DPSCs remain unknown. Moreover, its usefulness is restricted by a limited half-life and challenges in achieving localized tissue enrichment. This study aimed to develop a sustained-release injectable hydrogel composite as a drug delivery system and to investigate its influence on DPSCs. Herein, gelatin-based microspheres (GMSs) were loaded with DFO, and temperature-sensitive injectable hydrogels incorporating collagen and chitosan were synthesized to enable controlled DFO release. The experimental findings demonstrated that the DFO-loaded GMSs (DFO-GMSs) hydrogel composite possessed favorable physical properties and biocompatibility, enabling sustained DFO delivery for up to 15 days. DFO effectively stimulated DPSC migration, promoted the secretion of angiogenesis-related factors, and induced tube formation in vitro. These results suggest that the DFO-GMSs hydrogel composite significantly increased the migration and angiogenic potential of DPSCs, highlighting its promise for tissue regeneration applications.
Collapse
Affiliation(s)
- Jie Wang
- College
and Hospital of Stomatology, Anhui Medical
University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei230032, China
| | - Fan Yang
- College
and Hospital of Stomatology, Anhui Medical
University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei230032, China
| | - Ruting Chen
- College
and Hospital of Stomatology, Anhui Medical
University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei230032, China
- Department
of Stomatology, Yangjiang People’s
Hospital, Affiliated Yangjiang Hospital of Guangdong Medical University, Yangjiang529500, China
| | - Xinyue Yang
- College
and Hospital of Stomatology, Anhui Medical
University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei230032, China
| | - Jingjing Wang
- College
and Hospital of Stomatology, Anhui Medical
University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei230032, China
| | - Hongyan Zhang
- College
and Hospital of Stomatology, Anhui Medical
University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei230032, China
| |
Collapse
|
2
|
Fan Y, Leape CP, Hugard S, McCanne M, Thomson A, Wojtkiewicz GR, Weaver MJ, Collins JE, Randolph M, Oral E. A longitudinal rat model for assessing postoperative recovery and bone healing following tibial osteotomy and plate fixation. BMC Musculoskelet Disord 2023; 24:854. [PMID: 37907937 PMCID: PMC10617055 DOI: 10.1186/s12891-023-06942-5] [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: 11/03/2022] [Accepted: 10/07/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND Rodent models are commonly employed to validate preclinical disease models through the evaluation of postoperative behavior and allodynia. Our study investigates the dynamic interplay between pain and functional recovery in the context of traumatic osteotomy and surgical repair. Specifically, we established a rat model of tibial osteotomy, followed by internal fixation using a 5-hole Y-plate with 4 screws, to explore the hypothesis that histological bone healing is closely associated with functional recovery. OBJECTIVE Our primary objective was to assess the correlation between bone healing and functional outcomes in a rat model of tibial osteotomy and plate fixation. METHODS Seventeen male Sprague-Dawley rats underwent a metaphyseal transverse osteotomy of the proximal tibia, simulating a fracture-like injury. The resultant bone defect was meticulously repaired by realigning and stabilizing the bone surfaces with the Y-plate. To comprehensively assess recovery and healing, we performed quantitative and qualitative evaluations at 2, 4, 6, and 8 weeks post-surgery. Evaluation methods included micro-CT imaging, X-ray analysis, and histological examination to monitor bone defect healing. Concurrently, we employed video recording and gait analysis to evaluate functional recovery, encompassing parameters such as temporal symmetry, hindlimb duty factor imbalance, phase dispersion, and toe spread. RESULTS Our findings revealed complete healing of the bone defect at 8 weeks, as confirmed by micro-CT and histological assessments. Specifically, micro-CT data showed a decline in fracture volume over time, indicating progressive healing. Histological examination demonstrated the formation of new trabecular bone and the resolution of inflammation. Importantly, specific gait analysis parameters exhibited longitudinal changes consistent with bone healing. Hindlimb duty factor imbalance, hindlimb temporal symmetry, and phase dispersion correlated strongly with the healing process, emphasizing the direct link between bone healing and functional outcomes. CONCLUSIONS The establishment of this tibia osteotomy model underscores the association between bone healing and functional outcomes, emphasizing the feasibility of monitoring postoperative recovery using endpoint measurements. Our overarching objective is to employ this model for assessing the local efficacy of drug delivery devices in ameliorating post-surgical pain and enhancing functional recovery.
Collapse
Affiliation(s)
- Yingfang Fan
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA, USA
- Department of Orthopaedic Surgery, Harvard Medical School, 55 Fruit St. GRJ 1231, Boston, MA, 02114, USA
| | - Charlotte P Leape
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA, USA
| | - Shannon Hugard
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA, USA
| | - Madeline McCanne
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA, USA
| | - Andrew Thomson
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA, USA
- Boston College, Boston, MA, USA
| | | | - Michael J Weaver
- Department of Orthopaedic Surgery, Harvard Medical School, 55 Fruit St. GRJ 1231, Boston, MA, 02114, USA
- Department of Orthopaedic Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Jamie E Collins
- Department of Orthopaedic Surgery, Harvard Medical School, 55 Fruit St. GRJ 1231, Boston, MA, 02114, USA
- Department of Orthopaedic Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Mark Randolph
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA, USA
- Department of Orthopaedic Surgery, Harvard Medical School, 55 Fruit St. GRJ 1231, Boston, MA, 02114, USA
| | - Ebru Oral
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA, USA.
- Department of Orthopaedic Surgery, Harvard Medical School, 55 Fruit St. GRJ 1231, Boston, MA, 02114, USA.
| |
Collapse
|
3
|
Hou J, Ding Z, Zheng X, Shen Y, Lu Q, Kaplan DL. Tough Porous Silk Nanofiber-Derived Cryogels with Osteogenic and Angiogenic Capacity for Bone Repair. Adv Healthc Mater 2023:e2203050. [PMID: 36841910 DOI: 10.1002/adhm.202203050] [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: 11/24/2022] [Revised: 01/30/2023] [Indexed: 02/27/2023]
Abstract
Tough porous cryogels with angiogenesis and osteogenesis features remain a design challenge for utility in bone regeneration. Here, building off of the recent efforts to generate tough silk nanofiber-derived cryogels with osteogenic activity, deferoxamine (DFO) is loaded in silk nanofiber-derived cryogels to introduce angiogenic capacity. Both the mechanical cues (stiffness) and the sustained release of DFO from the gels are controlled by tuning the concentration of silk nanofibers in the system, achieving a modulus above 400 kPa and slow release of the DFO over 60 days. The modulus of the cryogels and the released DFO induce osteogenic and angiogenic activity, which facilitates bone regeneration in vivo in femur defects in rat, resulting in faster regeneration of vascularized bone tissue. The tunable physical and chemical cues derived from these nanofibrous-microporous structures support the potential for silk cryogels in bone tissue regeneration.
Collapse
Affiliation(s)
- Jianwen Hou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, P. R. China.,Department of Trauma Orthopedics, The Second People's Hospital of Lianyungang Affiliated to Bengbu Medical College, Lianyungang, 222023, P. R. China
| | - Zhaozhao Ding
- State Key Laboratory of Radiation Medicine and Radiation Protection, Institutes for Translational Medicine, Soochow University, Suzhou, 215123, P. R. China
| | - Xin Zheng
- Department of Orthopedics, Taizhou Municipal Hospital, Taizhou, 318000, P. R. China
| | - Yixin Shen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, P. R. China
| | - Qiang Lu
- State Key Laboratory of Radiation Medicine and Radiation Protection, Institutes for Translational Medicine, Soochow University, Suzhou, 215123, P. R. China
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| |
Collapse
|
4
|
Novel Techniques and Future Perspective for Investigating Critical-Size Bone Defects. Bioengineering (Basel) 2022; 9:bioengineering9040171. [PMID: 35447731 PMCID: PMC9027954 DOI: 10.3390/bioengineering9040171] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 01/31/2023] Open
Abstract
A critical-size bone defect is a challenging clinical problem in which a gap between bone ends will not heal and will become a nonunion. The current treatment is to harvest and transplant an autologous bone graft to facilitate bone bridging. To develop less invasive but equally effective treatment options, one needs to first have a comprehensive understanding of the bone healing process. Therefore, it is imperative to leverage the most advanced technologies to elucidate the fundamental concepts of the bone healing process and develop innovative therapeutic strategies to bridge the nonunion gap. In this review, we first discuss the current animal models to study critical-size bone defects. Then, we focus on four novel analytic techniques and discuss their strengths and limitations. These four technologies are mass cytometry (CyTOF) for enhanced cellular analysis, imaging mass cytometry (IMC) for enhanced tissue special imaging, single-cell RNA sequencing (scRNA-seq) for detailed transcriptome analysis, and Luminex assays for comprehensive protein secretome analysis. With this new understanding of the healing of critical-size bone defects, novel methods of diagnosis and treatment will emerge.
Collapse
|
5
|
Wixted J, Challa S, Nazarian A. Enhancing fracture repair: cell-based approaches. OTA Int 2022; 5:e168. [PMID: 35282391 PMCID: PMC8900459 DOI: 10.1097/oi9.0000000000000168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/16/2021] [Accepted: 11/24/2021] [Indexed: 05/24/2023]
Abstract
Fracture repair is based both on the macrolevel modulation of fracture fragments and the subsequent cellular activity. Surgeons have also long recognized other influences on cellular behavior: the effect of the fracture or subsequent surgery on the available pool of cells present locally in the periosteum, the interrelated effects of fragment displacement, and construct stiffness on healing potential, patient pathophysiology and systemic disease conditions (such as diabetes), and external regulators of the skeletal repair (such as smoking or effect of medications). A wide variety of approaches have been applied to enhancing fracture repair by manipulation of cellular biology. Many of these approaches reflect our growing understanding of the cellular physiology that underlies skeletal regeneration. This review focuses on approaches to manipulating cell lineages, influencing paracrine and autocrine cell signaling, or applying other strategies to influence cell surface receptors and subsequent behavior. Scientists continue to evolve new approaches to pharmacologically enhancing the fracture repair process.
Collapse
Affiliation(s)
- John Wixted
- Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center
| | - Sravya Challa
- Harvard Combined Orthopedic Residency Program, Boston, Massachusetts
| | - Ara Nazarian
- Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center
| |
Collapse
|
6
|
Eldeeb AE, Salah S, Mabrouk M, Amer MS, Elkasabgy NA. Dual-Drug Delivery via Zein In Situ Forming Implants Augmented with Titanium-Doped Bioactive Glass for Bone Regeneration: Preparation, In Vitro Characterization, and In Vivo Evaluation. Pharmaceutics 2022; 14:274. [PMID: 35214007 PMCID: PMC8876030 DOI: 10.3390/pharmaceutics14020274] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/10/2022] [Accepted: 01/18/2022] [Indexed: 12/28/2022] Open
Abstract
In situ forming implants (IFIs) are non-surgical approach using biodegradable polymers to treat bone fractures. The study aimed at preparing dual-drug-loaded IFIs to deliver pitavastatin (osteogenic drug) and tedizolid (antibiotic) using zein as the implant matrix via solvent-induced phase inversion method. At first, several investigations were done on pitavastatin-loaded zein IFIs, where three concentrations of zein were used (10, 20, and 30% w/v). IFIs were evaluated for their solidification time, rheological properties, injectability, and in vitro release. IFIs containing bioactive glass nanoparticles were prepared by the addition of non-doped bioactive glass nanoparticles (BGT0; 1, 3, 5, and 10% w/v) or titanium-doped bioactive glass nanoparticles (BGT5; 1% w/v) to the selected concentration of zein (30% w/v) and then evaluated. The optimized dual-medicated implant (D-ZIFI 1) containing pitavastatin, tedizolid, sodium hyaluronate (3% w/v), and BGT5 (1% w/v) was prepared and compared to IFI lacking both sodium hyaluronate and BGT5 (D-ZIFI 2). D-ZIFI 1 and 2 sustained the release profiles of both drugs for 28 days. SEM images proved the interconnected porous structure of D-ZIFI 1 due to sodium hyaluronate. In vivo studies on surgically induced bone defects in Sprague-Dawley rats signified the proper accelerated bone healing ability of D-ZIFI 1 over D-ZIFI 2. Results presented D-ZIFI 1 as a promising, effective, non-surgical approach for bone healing.
Collapse
Affiliation(s)
- Alaa Emad Eldeeb
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt; (S.S.); (N.A.E.)
| | - Salwa Salah
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt; (S.S.); (N.A.E.)
| | - Mostafa Mabrouk
- Refractories, Ceramics and Building Materials Department, National Research Centre, Giza 12622, Egypt;
| | - Mohammed S. Amer
- Department of Surgery, Anaesthesiology and Radiology, Faculty of Veterinary Medicine, Cairo University, Cairo 12211, Egypt;
| | - Nermeen A. Elkasabgy
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt; (S.S.); (N.A.E.)
| |
Collapse
|
7
|
Hernández-Flores C, Delgado A, Domínguez-Hernández VM, Valdez-Mijares R, Araujo-Monsalvo VM. A castor oil plant (Ricinus communis)-derived implant improves the biomechanical properties of noncritical bone defects. Acta Cir Bras 2021; 36:e360202. [PMID: 33624719 PMCID: PMC7902079 DOI: 10.1590/acb360202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/17/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose The biomechanical properties of the polyurethanes implant material derived
from castor oil plant (Ricinus communis) were evaluated in a noncritical
bone defect model in rat tibia. Methods After three weeks of the implant application, the tibias were tested by means
of the biomechanical three-point flexion test and resistance, rigidity,
energy at maximum load and maximum energy were evaluated. Nonparametric
statistical analysis was performed. Results It was found that the group that received the implant behaved the same as the
intact control group and also showed a significant increase in maximum load
compared to the spontaneous repair group. Conclusions Our results indicate that the tibias with the implant material in a
noncritical bone defect recover normal biomechanical parameters in less time
than spontaneously.
Collapse
|
8
|
Jing X, Du T, Yang X, Zhang W, Wang G, Liu X, Li T, Jiang Z. Desferoxamine protects against glucocorticoid-induced osteonecrosis of the femoral head via activating HIF-1α expression. J Cell Physiol 2020; 235:9864-9875. [PMID: 32437020 DOI: 10.1002/jcp.29799] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/07/2020] [Indexed: 12/21/2022]
Abstract
Glucocorticoid-induced osteonecrosis of the femoral head (GIOFH) is one of the most common complications of glucocorticoid administration. By chelating Fe2+ , desferoxamine (DFO) was reported to be able to activate the HIF-1α/VEGF pathway and promote angiogenesis. In the present study, we examined whether DFO administration could promote angiogenesis and bone repair in GIOFH. GIOFH was induced in rats by methylprednisolone in combination with lipopolysaccharide. Bone repair was assessed by histologic analysis and microcomputed tomography (micro-CT). Vascularization was assessed by Microfil perfusion and micro-CT analysis. Immunohistochemical staining was performed to analyze the expression of HIF-1α, VEGF, and CD31. Our in vivo study revealed that DFO increased HIF-1α/VEGF expression and promoted angiogenesis and osteogenesis in GIOFH. Moreover, our in vitro study revealed that DFO restored dexamethone-induced HIF-1α downregulation and angiogenesis inhibition. Besides, our in vitro study also demonstrated that DFO could protect bone marrow-derived stem cells from dexamethone-induced apoptosis and mitochondrial dysfunction by promoting mitophagy and mitochondrial fission. In summary, our data provided useful information for the development of novel therapeutics for management of GIOFH.
Collapse
Affiliation(s)
- Xingzhi Jing
- Department of Spine Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Ting Du
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Xiaoxia Yang
- Department of Spine Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Weimin Zhang
- Department of Spine Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Guodong Wang
- Department of Spine Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiaoyang Liu
- Department of Spine Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Tao Li
- Department of Spine Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhensong Jiang
- Department of Spine Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
9
|
In-situ forming chitosan implant-loaded with raloxifene hydrochloride and bioactive glass nanoparticles for treatment of bone injuries: Formulation and biological evaluation in animal model. Int J Pharm 2020; 580:119213. [PMID: 32165229 DOI: 10.1016/j.ijpharm.2020.119213] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 03/01/2020] [Accepted: 03/07/2020] [Indexed: 01/19/2023]
Abstract
In-situ forming implants receive great attention for repairing serious bone injuries. The aim of the present study was to prepare novel chitosan in-situ forming implants (CIFI) loaded with bioactive glass nanoparticles and/or raloxifene hydrochloride (RLX). Incorporating raloxifene hydrochloride (RLX) as a selective estrogen receptor modulator was essential to make use of its anti-resorptive properties. The prepared formulae were tested for their in-vitro gelation time, drug release, injectability, rheological properties, erosion rate and morphological properties. Results revealed that the formulation composed of 1% (w/v) chitosan with 2% (w/v) NaHCO3 and 1% (w/v) bioactive glass nanoparticles (CIFI-BG) possessed the most sustained drug release profile which extended over four months with low burst release effect compared to the same formulation lacking bioactive glass nanoparticles (CIFI). Selected formulations were tested for their ability to enhance bone regeneration in induced puncture in rate tibia. Results declared that these formulations were able to enhance bone regeneration after 12 weeks in comparison to the untreated tibial punctures and that containing bioactive glass could be considered as novel approach for treatment of serious bone injuries which require long term treatment and internal mechanical bone support during healing.
Collapse
|
10
|
Elkasabgy NA, Abdel-Salam FS, Mahmoud AA, Basalious EB, Amer MS, Mostafa AA, Elkheshen SA. Long lasting in-situ forming implant loaded with raloxifene HCl: An injectable delivery system for treatment of bone injuries. Int J Pharm 2019; 571:118703. [PMID: 31536761 DOI: 10.1016/j.ijpharm.2019.118703] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/13/2019] [Accepted: 09/14/2019] [Indexed: 12/16/2022]
Abstract
Bone injury is very serious in elder people or osteoporotic patients. In-situ forming implants (IFI) for bone rebuilding are usually poly-lactic-co-glycolic acid (PLGA)-based, which have a burst release effect. This study aimed to prepare novel liquid lipid-based PLGA-IFI loaded with raloxifene hydrochloride for prolonged non-surgical treatment of bone injuries by applying solvent-induced phase inversion technique. Labrasol® and Maisine® were added to the selected IFI forming long lasting lipid-based IFI (LLL-IFI). The formulations were characterized by analysing their in-vitro drug release, solidification time, injectability, rheological properties, and DSC in addition to their morphological properties. Results revealed that the LLL-IFI composed of 10%w/v PLGA with a lactide to glycolide ratio of 75:25 with ester terminal and 10% Maisine® possessed the most sustained drug release and lowest burst effect, as well as delayed pore formation compared to its counterpart lacking Maisine®. The selected LLL-IFI and PLGA-IFI formulations were tested for their capability to enhance bone regeneration in bone injuries induced in rats. Both formulations succeeded in healing the bones completely with the superiority of LLL-IFI in the formation of well-organized bone structures lacking fibrous tissues. The results suggest that LLL-IFI and PLGA-IFI are innovative approaches for treating critical and non-critical sized bone injuries.
Collapse
Affiliation(s)
- Nermeen A Elkasabgy
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, Egypt
| | | | - Azza A Mahmoud
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmaceutical Sciences and Pharmaceutical Industries, Future University in Egypt, Cairo, Egypt; Department of Pharmaceutical Technology, Pharmaceutical and Drug Industries Research Division, National Research Center, Dokki, Cairo, Egypt
| | - Emad B Basalious
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, Egypt
| | - Mohammed S Amer
- Department of Surgery, Anaesthesiology and Radiology, Faculty of Veterinary Medicine, Cairo University, Egypt
| | - Amany A Mostafa
- Refractories, Ceramics and Building Materials Department, Inorganic Chemical Industries and Mineral Resources Division, Nanomedicine and Tissue Engineering Lab, National Research Centre, Dokki, Cairo, Egypt
| | - Seham A Elkheshen
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmaceutical Sciences and Pharmaceutical Industries, Future University in Egypt, Cairo, Egypt
| |
Collapse
|
11
|
Wang Q, Zhao G, Xing Z, Zhan J, Ma J. Comparative evaluation of the osteogenic capacity of human mesenchymal stem cells from bone marrow and umbilical cord tissue. Exp Ther Med 2018; 17:764-772. [PMID: 30651861 DOI: 10.3892/etm.2018.6975] [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] [Received: 04/23/2018] [Accepted: 09/27/2018] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have been extensively investigated in the field of regenerative medicine. Human bone MSCs (BMSCs) have become a common type of seed cell for bone tissue engineering. However, the viability and cell number of BMSCs are negatively correlated with donor age, and as the extraction process is painful, this method has not been widely used. As human umbilical cord MSCs (UCMSCs) may be harvested inexpensively and inexhaustibly, the present study evaluated and compared the regenerative potential of UCMSCs and BMSCs to determine whether UCMSCs may be used as a novel cell type for bone regeneration. In the present study, the proliferation and osteogenic capacity of BMSCs and UCMSCs was compared in vitro. BMSCs and UCMSCs were respectively combined with biofunctionalized macroporous calcium phosphate cement, and their bone regenerative potentials were determined by investigating their capacity for ectopic bone formation in a nude mouse model as well as their efficacy in a rat model of tibia bone defect. The extent of bone regeneration was examined by X-ray, histological and immunohistochemical analyses. The results revealed that UCMSCs exhibited a good osteogenic differentiation potential, similarly to that of BMSCs, and that UCMSCs were able to contribute to the regeneration of bone and blood vessels. Furthermore, no significant differences were identified between BMSCs and UCMSCs in terms of their bone regenerative effect.
Collapse
Affiliation(s)
- Qian Wang
- Institute of Biomedical Science, Tianjin Kang Ting Biological Engineering Co., Ltd., Tianjin 300385, P.R. China
| | - Gang Zhao
- Institute of Biomedical Science, Tianjin Kang Ting Biological Engineering Co., Ltd., Tianjin 300385, P.R. China
| | - Zijun Xing
- Institute of Biomedical Science, Tianjin Kang Ting Biological Engineering Co., Ltd., Tianjin 300385, P.R. China
| | - Juming Zhan
- Institute of Biomedical Science, Tianjin Kang Ting Biological Engineering Co., Ltd., Tianjin 300385, P.R. China
| | - Jie Ma
- Institute of Biomedical Science, Tianjin Kang Ting Biological Engineering Co., Ltd., Tianjin 300385, P.R. China
| |
Collapse
|
12
|
Li H, Luo B, Wen W, Zhou C, Tian L, Ramakrishna S. Deferoxamine immobilized poly(D,L-lactide) membrane via polydopamine adhesive coating: The influence on mouse embryo osteoblast precursor cells and human umbilical vein endothelial cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 70:701-709. [DOI: 10.1016/j.msec.2016.09.043] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 08/20/2016] [Accepted: 09/21/2016] [Indexed: 01/05/2023]
|
13
|
Li L, Qu Y, Jin X, Guo XQ, Wang Y, Qi L, Yang J, Zhang P, Li LZ. Protective effect of salidroside against bone loss via hypoxia-inducible factor-1α pathway-induced angiogenesis. Sci Rep 2016; 6:32131. [PMID: 27558909 PMCID: PMC4997314 DOI: 10.1038/srep32131] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 08/03/2016] [Indexed: 12/02/2022] Open
Abstract
Hypoxia-inducible factor (HIF)-1α plays a critical role in coupling angiogenesis with osteogenesis during bone development and regeneration. Salidroside (SAL) has shown anti-hypoxic effects in vitro and in vivo. However, the possible roles of SAL in the prevention of hypoxia-induced osteoporosis have remained unknown. Two osteoblast cell lines, MG-63 and ROB, were employed to evaluate the effects of SAL on cell viability, apoptosis, differentiation and mineralization in vitro. Rats subjected to ovariectomy-induced bone loss were treated with SAL in vivo. Our results showed that pre-treatment with SAL markedly attenuated the hypoxia-induced reductions in cell viability, apoptosis, differentiation and mineralization. SAL down-regulated HIF-1α expression and inhibited its translocation; however, SAL increased its transcriptional activity and, consequently, up-regulated vascular endothelial growth factor (VEGF). In vivo studies further demonstrated that SAL caused decreases in the mineral, alkaline phosphatase (ALP), and BGP concentrations in the blood of ovariectomized (OVX) rats. Moreover, SAL improved the trabecular bone microarchitecture and increased bone mineral density in the distal femur. Additionally, SAL administration partially ameliorated this hypoxia via the HIF-1α-VEGF signalling pathway. Our results indicate that SAL prevents bone loss by enhancing angiogenesis and osteogenesis and that these effects are associated with the activation of HIF-1α signalling.
Collapse
Affiliation(s)
- Ling Li
- Tianjin Key Laboratory for Prevention and Control of Occupational and Environmental Hazard, Tianjin, People's Republic of China.,Department of Pharmacology, Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Ye Qu
- Department of Pathogenic Biology and Immunology, Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Xin Jin
- Department of Pharmacology, Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Xiao Qin Guo
- Department of Pathogenic Biology and Immunology, Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Yue Wang
- Tianjin Key Laboratory for Prevention and Control of Occupational and Environmental Hazard, Tianjin, People's Republic of China.,Department of Pathogenic Biology and Immunology, Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Lin Qi
- Department of Pathogenic Biology and Immunology, Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Jing Yang
- Department of Pathogenic Biology and Immunology, Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Peng Zhang
- Department of Orthopaedics, Affiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Ling Zhi Li
- Tianjin Key Laboratory for Prevention and Control of Occupational and Environmental Hazard, Tianjin, People's Republic of China.,Department of Pharmaceutical Chemistry, Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| |
Collapse
|
14
|
Jia P, Chen H, Kang H, Qi J, Zhao P, Jiang M, Guo L, Zhou Q, Qian ND, Zhou HB, Xu YJ, Fan Y, Deng LF. Deferoxamine released from poly(lactic-co-glycolic acid) promotes healing of osteoporotic bone defect via enhanced angiogenesis and osteogenesis. J Biomed Mater Res A 2016; 104:2515-27. [PMID: 27227768 DOI: 10.1002/jbm.a.35793] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/18/2016] [Accepted: 05/24/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Peng Jia
- Department of Orthopaedics; San Xiang Road 1055, The Second Affiliated Hospital of Soochow University; Suzhou Jiangsu Province 215004 China
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese Western Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine; Rui Jin Er Road 197 Shanghai 200020 China
| | - Hao Chen
- Department of Orthopaedics; Shanghai Jiao Tong University School of Medicine, Shanghai Ren Ji Hospital; Pu Jian Road 160 Shanghai 200120 China
| | - Hui Kang
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese Western Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine; Rui Jin Er Road 197 Shanghai 200020 China
| | - Jin Qi
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese Western Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine; Rui Jin Er Road 197 Shanghai 200020 China
| | - Peng Zhao
- Nursing Department; The Second Affiliated Hospital of Soochow University; San Xiang Road 1055 Suzhou Jiangsu Province China 215004
| | - Min Jiang
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese Western Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine; Rui Jin Er Road 197 Shanghai 200020 China
| | - Lei Guo
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese Western Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine; Rui Jin Er Road 197 Shanghai 200020 China
| | - Qi Zhou
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese Western Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine; Rui Jin Er Road 197 Shanghai 200020 China
| | - Nian Dong Qian
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese Western Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine; Rui Jin Er Road 197 Shanghai 200020 China
| | - Han Bing Zhou
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese Western Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine; Rui Jin Er Road 197 Shanghai 200020 China
| | - You Jia Xu
- Department of Orthopaedics; San Xiang Road 1055, The Second Affiliated Hospital of Soochow University; Suzhou Jiangsu Province 215004 China
| | - Yongqian Fan
- Department of Orthopaedics; Huadong Hospital Affiliated Fudan University; Yan'an Western Road 221 Shanghai 200040 China
| | - Lian Fu Deng
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese Western Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine; Rui Jin Er Road 197 Shanghai 200020 China
| |
Collapse
|
15
|
Rassu G, Salis A, Porcu EP, Giunchedi P, Roldo M, Gavini E. Composite chitosan/alginate hydrogel for controlled release of deferoxamine: A system to potentially treat iron dysregulation diseases. Carbohydr Polym 2016; 136:1338-47. [DOI: 10.1016/j.carbpol.2015.10.048] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 09/28/2015] [Accepted: 10/13/2015] [Indexed: 10/22/2022]
|
16
|
Matsumoto T, Sato S. Stimulating angiogenesis mitigates the unloading-induced reduction in osteogenesis in early-stage bone repair in rats. Physiol Rep 2015; 3:e12335. [PMID: 25780087 PMCID: PMC4393168 DOI: 10.14814/phy2.12335] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 02/17/2015] [Accepted: 02/17/2015] [Indexed: 11/24/2022] Open
Abstract
Accelerating fracture healing during bed rest allows early mobilization and avoids prolonged fracture healing times. We tested the hypothesis that stimulating angiogenesis with deferoxamine (DFO) mitigates the unloading-induced reduction in early-stage bone repair. Rats aged 12 weeks were subjected to cortical drilling on their tibial diaphysis under anesthesia and treated with hindlimb unloading (HU), HU and DFO administration (DFOHU), or weight bearing (WB) for 5 or 10 days (HU5/10, DFOHU5/10, WB5/10; n = 8 per groups) until sacrifice for vascular casting with a zirconium dioxide-based contrast agent. Taking advantage of its absorption discontinuity at the K-absorption edge, vascular and bone images in the drill-hole defects were acquired by synchrotron radiation subtraction CT. Bone repair was reduced in HU rats. The bone volume fraction (B.Vf) was 88% smaller in HU5 and 42% smaller in HU10 than in WB5/10. The bone segment densities (B.Seg) were 97% smaller in HU5 and 141% larger in HU10 than in WB5/10, and bone thickness (B.Th) was 38% smaller in HU10 than in WB10. The vascular volume fraction (V.Vf) was 35% and the mean vessel diameter (V.D) was 13% smaller in HU10 than in WB10. When compared according to categorized vessel sizes, V.Vf in the diameter ranges 20-30, 30-40, and >40 μm were smaller in HU10 than in WB10, and V.Seg in the diameter range >40 μm was smaller in HU10 than in WB10. In contrast, there was no difference in B.Vf between DFOHU5/10 and WB5/10 and in V.Vf between DFOHU10 and WB10, though B.Seg remained 86% smaller in DFOHU5 and 94% larger in DFOHU10 than in WB5/10, and B.Th and V.D were 23% and 14% lower in DFOHU10 than in WB10. Vessel size-specific V.Vf in the diameter ranges 10-20 and 20-30 μm was larger in DFOHU5 than in HU5. In conclusion, the enhanced angiogenic ingrowth mitigates the reduction in bone repair during mechanical unloading.
Collapse
Affiliation(s)
- Takeshi Matsumoto
- Bioengineering Division, Osaka University Graduate School of Engineering Science, Toyonaka, Japan
| | - Shota Sato
- Bioengineering Division, Osaka University Graduate School of Engineering Science, Toyonaka, Japan
| |
Collapse
|
17
|
Peng J, Lai ZG, Fang ZL, Xing S, Hui K, Hao C, Jin Q, Qi Z, Shen WJ, Dong QN, Bing ZH, Fu DL. Dimethyloxalylglycine prevents bone loss in ovariectomized C57BL/6J mice through enhanced angiogenesis and osteogenesis. PLoS One 2014; 9:e112744. [PMID: 25394221 PMCID: PMC4231053 DOI: 10.1371/journal.pone.0112744] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/12/2014] [Indexed: 12/16/2022] Open
Abstract
Hypoxia-inducible factor 1-α (HIF-1α) plays a critical role in angiogenesis-osteogenesis coupling during bone development and bone regeneration. Previous studies have shown that 17β-estradiol activates the HIF-1α signaling pathway and that mice with conditional activation of the HIF-1α signaling pathway in osteoblasts are protected from ovariectomy (OVX)-induced bone loss. In addition, it has been shown that hypoxia facilitates the osteogenic differentiation of mesenchymal stem cells (MSCs) and modulates Wnt/β-catenin signaling. Therefore, we hypothesized that activation of the HIF-1α signaling pathway by hypoxia-mimicking agents would prevent bone loss due to estrogen deficiency. In this study, we confirmed the effect of dimethyloxalylglycine (DMOG), a hypoxia-mimicking agent, on the HIF-1α signaling pathway and investigated the effect of DMOG on MSC osteogenic differentiation and the Wnt/β-catenin signaling pathway. We then investigated the effect of DMOG treatment on OVX-induced bone loss. Female C57BL/6J mice were divided into sham, OVX, OVX+L-DMOG (5 mg/kg/day), and OVX+H-DMOG (20 mg/kg/day) groups. At sacrifice, static and dynamic bone histomorphometry were performed with micro computed tomography (micro-CT) and undecalcified sections, respectively. Bone strength was assessed with the three-point bending test, and femur vessels were reconstructed and analyzed by micro-CT. Serum vascular endothelial growth factor (VEGF), osteocalcin, and C-terminal telopeptides of collagen type(CTX) were measured by ELISA. Tartrate-resistant acid phosphatase staining was used to assess osteoclast formation. Alterations in the HIF-1α and Wnt/β-catenin signaling pathways in the bone were detected by western blot. Our results showed that DMOG activated the HIF-1α signaling pathway, which further activated the Wnt/β-catenin signaling pathway and enhanced MSC osteogenic differentiation. The micro-CT results showed that DMOG treatment improved trabecular bone density and restored the bone microarchitecture and blood vessels in OVX mice. Bone strength was also partly restored in DMOG-treated OVX mice. Dynamic bone histomorphometric analysis of the femur metaphysic revealed that DMOG increased the mineralizing surface, mineral apposition rate, and bone formation rate. The serum levels of VEGF and osteocalcin were higher in DMOG-treated OVX mice. However, there were no significant differences in serum CTX or in the number of tartrate-resistant acid phosphatase-stained cells between DMOG-treated OVX mice and OVX mice. Western blot results showed that DMOG administration partly rescued the decrease in HIF-1α and β-catenin expression following ovariectomy. Collectively, these results indicate that DMOG prevents bone loss due to ovariectomy in C57BL/6J mice by enhancing angiogenesis and osteogenesis, which are associated with activated HIF-1α and Wnt/β-catenin signaling pathways.
Collapse
Affiliation(s)
- Jia Peng
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Zuo Gui Lai
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
- Department of Orthopaedics, Qian Fo Shan Hospital, Shang Dong University, Ji Nan, China
| | - Zhang Lian Fang
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shen Xing
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Kang Hui
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Hao
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Jin
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Zhou Qi
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Wang Jin Shen
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Nian Dong
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Zhou Han Bing
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Deng Lian Fu
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
- * E-mail:
| |
Collapse
|
18
|
Vinzenz P, Schröckmair S, Gruber R, Agis H. Bone substitute materials supplemented with prolyl hydroxylase inhibitors decrease osteoclastogenesis in vitro. J Biomed Mater Res B Appl Biomater 2014; 103:1198-203. [PMID: 25312707 DOI: 10.1002/jbm.b.33295] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 08/14/2014] [Accepted: 09/12/2014] [Indexed: 11/09/2022]
Abstract
BACKGROUND AND OBJECTIVE Inhibition of prolyl hydroxylases stimulates bone regeneration. Consequently, bone substitute materials were developed that release prolyl hydroxylase inhibitors. However, the impact of prolyl hydroxylase inhibitors released from these carriers on osteoclastogenesis is not clear. We therefore assessed the effect of bone substitute materials that release prolyl hydroxylase inhibitors on osteoclastogenesis. MATERIAL AND METHODS Dimethyloxalylglycine, desferrioxamine, and l-mimosine were lyophilized onto bovine bone mineral and hydroxyapatite, and supernatants were generated. Osteoclastogenesis was induced in murine bone marrow cultures in the presence of the supernatants from bone substitute materials. The formation of tartrate-resistant acid phosphatase (TRAP)-positive multinucleated cells and TRAP activity were determined. To test for possible effects on osteoclast progenitor cells, we measured the effect of the supernatants on proliferation and viability. In addition, experiments were performed where prolyl hydroxylase inhibitors were directly added to the bone marrow cultures. RESULTS We found that prolyl hydroxylase inhibitors released within the first hours from bone substitute materials reduce the number and activity of TRAP-positive multinucleated cells. In line with this, addition of prolyl hydroxylase inhibitors directly to the bone marrow cultures dose-dependently reduced the number of TRAP-positive multinucleated cells and the overall resorption activity. Moreover, the released prolyl hydroxylase inhibitors decreased proliferation but not viability of osteoclast progenitor cells. CONCLUSION Our results show that prolyl hydroxylase inhibitors released from bone substitute materials decrease osteoclastogenesis in murine bone marrow cultures.
Collapse
Affiliation(s)
- Philipp Vinzenz
- Department of Oral Surgery, Medical University of Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Stefan Schröckmair
- Department of Oral Surgery, Medical University of Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Reinhard Gruber
- Department of Oral Surgery, Medical University of Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Laboratory of Oral Cell Biology, School of Dental Medicine, University of Bern, Bern, Switzerland
| | - Hermann Agis
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Department of Conservative Dentistry and Periodontology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|